Journal list menu

Volume 276, Issue 12 p. 3256-3268
Free Access

Thiaminylated adenine nucleotides. Chemical synthesis, structural characterization and natural occurrence

Michel Frédérich

Michel Frédérich

Laboratory of Pharmacognosy, Université de Liège, Belgium

These authors contributed equally to this work

Search for more papers by this author
David Delvaux

David Delvaux

GIGA-Neurosciences, Université de Liège, Belgium

These authors contributed equally to this work

Search for more papers by this author
Tiziana Gigliobianco

Tiziana Gigliobianco

GIGA-Neurosciences, Université de Liège, Belgium

Search for more papers by this author
Marjorie Gangolf

Marjorie Gangolf

GIGA-Neurosciences, Université de Liège, Belgium

Search for more papers by this author
Georges Dive

Georges Dive

Center for Protein Engineering, Université de Liège, Belgium

Search for more papers by this author
Gabriel Mazzucchelli

Gabriel Mazzucchelli

Physical Chemistry, GIGA-Research, Université de Liège, Belgium

Search for more papers by this author
Benjamin Elias

Benjamin Elias

Organic and Medicinal Chemistry, Université catholique de Louvain, Louvain-la-Neuve, Belgium

Search for more papers by this author
Edwin De Pauw

Edwin De Pauw

Physical Chemistry, GIGA-Research, Université de Liège, Belgium

Search for more papers by this author
Luc Angenot

Luc Angenot

Laboratory of Pharmacognosy, Université de Liège, Belgium

Search for more papers by this author
Pierre Wins

Pierre Wins

GIGA-Neurosciences, Université de Liège, Belgium

Search for more papers by this author
Lucien Bettendorff

Lucien Bettendorff

GIGA-Neurosciences, Université de Liège, Belgium

Search for more papers by this author
First published: 21 May 2009
Citations: 36
L. Bettendorff, GIGA-Neurosciences, University of Liège, Bât. B36, Tour de Pathologie 2, étage +1, Avenue de l’Hôpital, 1, B-4000 Liège 1 (Sart-Tilman), Belgium
Fax: +32 4 366 59 53
Tel: +32 4 366 59 67
E-mail: [email protected]

Abstract

Thiamine and its three phosphorylated derivatives (mono-, di- and triphosphate) occur naturally in most cells. Recently, we reported the presence of a fourth thiamine derivative, adenosine thiamine triphosphate, produced in Escherichia coli in response to carbon starvation. Here, we show that the chemical synthesis of adenosine thiamine triphosphate leads to another new compound, adenosine thiamine diphosphate, as a side product. The structure of both compounds was confirmed by MS analysis and 1H-, 13C- and 31P-NMR, and some of their chemical properties were determined. Our results show an upfield shifting of the C-2 proton of the thiazolium ring in adenosine thiamine derivatives compared with conventional thiamine phosphate derivatives. This modification of the electronic environment of the C-2 proton might be explained by a through-space interaction with the adenosine moiety, suggesting U-shaped folding of adenosine thiamine derivatives. Such a structure in which the C-2 proton is embedded in a closed conformation can be located using molecular modeling as an energy minimum. In E. coli, adenosine thiamine triphosphate may account for 15% of the total thiamine under energy stress. It is less abundant in eukaryotic organisms, but is consistently found in mammalian tissues and some cell lines. Using HPLC, we show for the first time that adenosine thiamine diphosphate may also occur in small amounts in E. coli and in vertebrate liver. The discovery of two natural thiamine adenine compounds further highlights the complexity and diversity of thiamine biochemistry, which is not restricted to the cofactor role of thiamine diphosphate.

Abbreviations

  • AThDP
  • adenosine thiamine diphosphate
  • AThTP
  • adenosine thiamine triphosphate
  • Pi
  • inorganic phosphate
  • Thc
  • thiochrome
  • ThDP
  • thiamine diphosphate
  • ThMP
  • thiamine monophosphate
  • ThTP
  • thiamine triphosphate
  • ThTPase
  • thiamine triphosphatase
  • Thiamine (vitamin B1) is an essential compound for all known life forms. In most cell types, the well-characterized cofactor thiamine diphosphate (ThDP) is the major thiamine compound. Thiamine monophosphate (ThMP), for which no physiological function has been determined thus far, and unphosphorylated thiamine account for only a few percent of the total thiamine content. Thiamine triphosphate (ThTP) is generally a minor compound (≤ 1% of total thiamine) but it is present in most organisms studied to date [1]. Its role remains enigmatic, but it has been found that ThTP phosphorylates certain proteins in electric organs and brain [2]. This might be part of a new cellular signaling pathway. In Escherichia coli, ThTP is synthesized in response to amino acid starvation in the presence of glucose [3,4]. Under special conditions of stress (very low intracellular ATP, but glucose present), E. coli may produce very high amounts of ThTP (60% of total thiamine) [4]. However, the mechanism of its synthesis remains unknown.

    Recently, we discovered the existence of a fourth natural thiamine derivative, adenosine thiamine triphosphate (AThTP) or thiaminylated ATP (Fig. 1). This compound has been found in a variety of organisms from bacteria to mammals [5]. Like ThTP, AThTP is generally a minor compound, but in E. coli, it may be produced in higher amounts (up to 15% of total thiamine) in response to carbon starvation. It seems likely that in bacteria, ThTP and AThTP act as signals (or alarmones) in response to different conditions of cellular stress. Some data were recently obtained concerning the metabolism of AThTP in E. coli. Its synthesis appears to be catalyzed by a soluble ThDP adenylyl transferase according to the reaction inline image. This enzyme seems to be a high molecular mass (355 kDa) multisubunit complex requiring Mg2+ ions for activity [6].

    Details are in the caption following the image

    Expanded structural formulas of adenosine thiamine diphosphate (AThDP, thiaminylated ADP) and adenosine thiamine triphosphate (AThTP, thiaminylated ATP).

    In a previous report [5], we showed that AThTP could be chemically synthesized by condensation of ThDP and AMP in the presence of N,N′-dicyclohexylcarbodiimide. Using this procedure, we found that the mixture obtained after synthesis contained, in addition to AThTP, a new compound that was identified as adenosine thiamine diphosphate (thiaminylated ADP; AThDP) (Fig. 1). As for AThTP, there was no previous mention of AThDP in the scientific literature, but the existence of this compound has been reported in at least two patents [7,8]. In the Kyowa Hakko Kogyo Co, Ltd patent [7] it was claimed that some bacteria, such as Corynebacterium glutamicum, are able, in the presence of adequate precursors (adenine, adenosine, thiamine, ThMP), to accumulate large amounts of AThDP (erroneously called thiamine adenine dinucleotide in the patent) in the extracellular medium. A method for the chemical synthesis of AThDP, using P2-diphenyl S-benzoylthiamine o-diphosphate as precursor, has also been described [8]. It is therefore of interest to better characterize these compounds.

    Here, we report the chemical synthesis of AThTP and AThDP, their purification and their physicochemical characterization using positive ESI-MS, 1H-, 13C- and 31P-NMR (Table 1), as well as molecular modeling. We also show that the two compounds can be detected in E. coli under different culture conditions. Furthermore, significant amounts of both compounds are also detectable in eukaryotic cells, including several mammalian tissues and cultured cells. Thus, thiamine adenine nucleotides may be more widespread than initially thought and may have physiological roles both in prokaryotes and eukaryotes.

    Table 1. 1H-, 13C- and 31P-NMR data of thiamine diphosphate (ThDP), adenosine thiamine diphosphate (AThDP), thiamine triphosphate (ThTP) and adenosine thiamine triphosphate (AThTP) recorded at 500/125/202.5 MHz in D2O at pH 7.4 and 25 °C. TMS and H3PO4 were used as references. ThDP was a commercial preparation (Sigma-Aldrich), and ThTP, AThDP and AThTP were synthesized as described in Experimental procedures. nd, not determined; s, singlet; bs, broadened singlet; d, doublet; dd, doublet of doublet; t, triplet; m, multiplet.
    ThDP AThDP ThTP AThTP
    Position 1H 31P 13C 1H 31P 13C 1H 31P 13C 1H 31P 13C
    2 9.55 (s) 154.50 9.18 (s) nd 9.55 (s) 155.68 9.14 (s) 153.25
    4 143.21 143.13 143.14 143.18
    5 135.58 135.02 135.8 134.92
    6 5.49 (s) 50.16 5.19 (s) 50.87 5.38 (s) 49.84 5.17 (s) 50.93
    7 105.86 103.70 106.32 103.66
    8 162.61 161.43 162.85 161.42
    10 164.68 168.90 163.22 169.24
    12 7.93 (s) 147.62 7.93 (s) 157.03 7.84 (s) 144.31 7.90 (s) 156.80
    13 2.52 (s) 11.10 2.49 (s) 11.16 2.44 (s) 11.05 2.48 (s) 11.20
    14 3.28 (t, 5.4 Hz) 27.56 (d, 8.2 Hz) 3.14 (t, 5.4 Hz) 27.41 (nd) 3.22 (t, 5.2 Hz) 27.52 (d, 8.2 Hz) 3.15 (dd, 4.8 and 5.1 Hz) 27.38 (d, 7.8 Hz)
    15 4.15 (m) 64.64 (d, 5.6 Hz) 4.08 (m) 64.78 (d, 6 Hz) 4.11 (m) 64.92 (d, 5.3 Hz) 4.10 (dd, 5.1 and 5.8 Hz) 64.80 (d, 5.5Hz)
    16 2.55 (s) 21.72 2.38 (s) 23.93 2.52 (s) 21.20 2.32 (s) 23.81
    1′ 6.02 (d, 5.6 Hz) 86.55 6.01 (d, 5.9 Hz) 86.61
    2′ 4.75 (m) 73.69 4.69 (t, 5.6 Hz) 74.11
    3′ 4.44 (t, 4.7 Hz) 70.27 4.46 (t, 4.9 Hz) 70.26
    4′ 4.33 (bs) 83.80 (d, 10.1 Hz) 4.32 (bs) 83.82 (d, 9.2 Hz)
    5′ 4.14 (m) 65.39 (d, 5.3 Hz) 4.17 (m) 65.26 (d, 5.2 Hz)
    2″ 8.09 (s) 152.73 8.08 (s) 152.62
    4″ 149.20 148.75
    5″ 118.69 118.13
    6″ nd 155.11
    8″ 8.43 (s) 139.50 8.41 (s) 139.89
    P-1 −11.40 (d, 20.2 Hz) −13.30 (s) −11.30 (d, 19.5 Hz) −13.13 (d, 18.7 Hz)
    P-2 −10,79 (d, 20.2 Hz) −13.30 (s) −23.20 (bs) −24.70 (t, 18.7 Hz)
    P-3 −11.90 (d, 19.5 Hz) −12.91 (d, 18.7 Hz)

    Results

    Chemical synthesis and purification of AThDP and AThTP

    We have previously shown that the condensation of ThDP and AMP by N,N′-dicyclohexylcarbodiimide leads to the synthesis of AThTP [5]. Here, this reaction has been further characterized with respect to the kinetics and composition of the reaction medium. In particular, we show that other side products, which have been unambiguously identified, are also formed during synthesis. Indeed, as shown in Fig. 2A, synthesis of AThTP (peak 5) is accompanied by the appearance of two other compounds in the reaction medium: AThDP (peak 3) and ThTP (peak 4). The small ThMP peak (peak 1) is essentially a contamination present in the commercially available ThDP used as the precursor (peak 2). However, AThTP synthesis proceeds through an optimum and after 3 h an accumulation of ThTP and AThDP is observed (Fig. 2C), although the amount of AThTP is much lower.

    Details are in the caption following the image

    Composition of the reaction medium during the condensation of ThDP or ThMP with AMP in the presence of N,N′-dicyclohexylcarbodiimide. (A) Chromatographic separation of the reaction mixture using the substrates ThDP and 5′-AMP after 90 min at room temperature (1, ThMP; 2, ThDP; 3, ThTP; 4, ThTP; 5, AThTP). (B) Chromatographic separation of the reaction mixture using the substrates ThMP and 5′-AMP after 90 min at room temperature. (C) Composition of the reaction mixture as a function of time for the condensation of ThDP and AMP in the presence of N,N′-dicyclohexylcarbodiimide (mean ± SD, n = 4, error bars are also given). In all cases, 0.7 mmol of each precursor (5′-AMP, ThMP, ThDP) were dissolved in 0.7 mL tributylamine and 750 μL H2O. To start the synthesis, 5 μL of this mixture were diluted with 1 mL of a mixture containing 500 μL dimethylsulfoxide, 450 μL pyridine and 0.15 g N,N′-dicyclohexylcarbodiimide (in 50 μL pyridine). Aliquots were taken at different time intervals, diluted 2000 times and analyzed by HPLC after derivatization to thiochrome derivatives.

    The presence of AThDP was further confirmed by the condensation of ThMP and AMP in the presence of N,N′-dicyclohexylcarbodiimide (Fig. 2B), which mostly leads to the formation of AThDP. However, the yield of AThDP synthesis according to this latter synthetic procedure is low: after 2 h, < 10% of the ThMP is converted to AThDP. Therefore, we routinely synthesized both compounds by condensing ThDP and AMP in the presence of N,N′-dicyclohexylcarbodiimide for 2 h. To purify AThDP and AThTP, large-scale synthesis was performed using an (AMP)/(ThDP) ratio of 1.5 rather than 1, because this resulted in higher yields of AThTP. After 2 h, thiamine derivatives were precipitated with diethyl ether and dissolved in water. ThTP, AThDP and AThTP were purified using several chromatographic steps. All thiamine phosphate derivatives, except ThTP [9], were retained on a AG 50W-X8 cation-exchange resin and eluted with ammonium acetate (0.2 m; pH 7.0). After lyophilization, the residue was dissolved in water and layered on a column filled with the anion-exchange resin AG-X1 equilibrated in water (Fig. 3). AThDP was eluted in 0.25 m ammonium acetate (pH 7.0) followed by 0.5 m ammonium acetate for the elution of AThTP. Both compounds were further purified on a Polaris C18 HPLC column. The total yield was 5.3% for AThDP and 2.7% for AThTP (Table 2). The purity of the two preparations was checked by HPLC using UV and, after derivatization, fluorescence detection (Fig. 4).

    Details are in the caption following the image

    Separation of thiamine derivatives on an AG-X1 resin equilibrated in water. The arrows indicate the addition of ammonium acetate (pH 5.0) at 0.25 and 0.5 m, respectively. The concentrations of the different thiamine compounds were measured by HPLC after derivatization to thiochrome derivatives.

    Table 2. Purification of chemically synthesized adenosine thiamine diphosphate (AThDP) and adenosine thiamine triphosphate (AThTP).
    AThDP AThTP
    μmol % μmol %
    Synthesis 720 100 802 100
    AG 50W-X8 610 85 644 80
    AG-X1 83 13.6 48 5.9
    Polaris C18 38 5.3 22 2.7
    Details are in the caption following the image

    Analysis of chemically synthesized AThDP (A,C) and AThTP (B,D) by HPLC. The AThDP and AThTP preparations were analyzed on a Polaris C18 HPLC column by UV detection (254 nm) (A,B) and on a PRP-1 column by fluorescence detection after derivatization to thiochrome derivatives (C,D) as described in Experimental procedures.

    Physicochemical characterization of chemically synthesized AThDP and AThTP using MS, NMR, fluorometry and molecular modeling

    Both fractions were analyzed by positive ESI-MS (Fig. 5). As expected, the AThTP fraction contained a major cation with a m/z ratio of 754.1, as described previously [5]. In the AThDP fraction, the major peak had a m/z ratio of 674.1, as expected for AThDP (crude formula C22H30N9O10P2S+, parent ion M+) with an average molecular mass of 674.5 Da (exact monoisotopic mass 674.1 Da). As for AThTP [5], ESI-MS/MS fragmentation of AThDP gave three main peaks: m/z 553.1 (a fragmentation product of AThDP obtained by loss of the pyrimidinium moiety, M+– 121 – pyrimidinium), m/z 348.1 (corresponding to AMP) and m/z 257.1. We were unable to assign the latter ion, which is obtained after fragmentation of both AThTP and AThDP and probably results from a molecular rearrangement.

    Details are in the caption following the image

    Positive-ion ESI MS of chemically synthesized AThTP (A) and AThDP (B). The compounds were diluted at a concentration of 150 μm in H2O/acetonitrile (50:50 v/v). The second major peak of m/z 696.1 in (B) represents the Na adduct of AThDP.

    NMR data for AThTP and AThDP, together with those for ThTP and ThDP, are listed in Table 1. They are clearly in accordance with the presence in AThDP and AThTP of a thiamine and an adenine moiety, as compared with thiamine and adenosine NMR data. The presence of three linked phosphates in AThTP is confirmed by three phosphorous signals in the 31P-NMR spectrum (two doublets and one triplet, as expected). Oddly, in AThDP, the two phosphates seemed to be equivalent, as only one signal was detected on the spectrum. However, the possibility that the molecule is adenosine thiamine monophosphate could be excluded on the basis of the molecular mass (Fig. 5). The linkage (C-15-triphosphate-C-5′) between the thiamine moiety and the adenine moiety of the molecule was proven by the presence of 13C–31P coupling constants for C-14 and C-15 and for C-5′ and C-4′.

    We place special emphasis on the C-2 proton of the thiazolium ring which is required for the catalytic activity of ThDP [10]. This proton is particularly labile and is completely exchanged with deuterium within a few minutes [11]. The experimental shift was 9.61, 9.55 and 9.55 p.p.m. respectively for ThMP, ThDP and ThTP (pH 7.4), values higher than expected for aromatic protons (in general 7.5–8.5 p.p.m.). In AThDP and AThTP, we saw a decrease in the shift (9.14/9.18 p.p.m.) compared with ThMP, ThDP or ThTP, indicating a modification of the electronic environment of the C-2 proton, probably as a consequence of a through-space interaction with the adenine moiety. This would suggest a U-shaped folding of AThDP and AThTP.

    Molecular modeling was applied on a model of the free (without influence of the environment) molecules without any counterion. The phosphate groups are neutralized by hydrogens and the whole system bears a positive charge because of the thiazolium fragment. Calculations showed that a U-folded conformation is energetically accessible for both di- and triphosphorylated derivatives. A possible structure for each derivative is shown in Fig. 6. In this conformation, the C-2 proton is embedded in the closed environment formed by the aromatic adenine and aminopyrimidine rings. Such a folded structure for adenylated thiamine derivatives is not in favor of a cofactor role that requires a highly reactive C-2 proton [10].

    Details are in the caption following the image

    Proposed 3D structures of free (no influence of the environment) AThDP (A) and AThTP (B). The phosphate groups are neutralized by hydrogens and the whole system carries a positive charge caused by the thiazolium fragment. The calculations were performed using the B3LYP functional [33] with the polarized double ζ basis set 6-31G(d) [34] and the gaussian 03 suite of programs [35]. The structures shown represent true energy minima.

    Like free thiamine, AThDP and AThTP can be readily oxidized to highly fluorescent thiochrome (Thc) derivatives. AThcDP and AThcTP gave practically identical fluorescence spectra with an optimum of 353 nm for excitation and 439 nm for emission (Fig. 7). However, when we compared the fluorescence properties of AThcDP and AThcTP with those of nonadenylated thiochromes (Thc, ThcMP, ThcDP and ThcTP, which have roughly the same fluorescence) [12,13], we found some interesting differences. First, the optimum emission wavelength was slightly lower for AThcDP and AThcTP than for thiochrome (439 versus 443 nm; Fig. 7C). More importantly, we observed that AThcDP and AThcTP solutions gave peaks with areas approximately twice as large as thiochrome solutions of the same molarity. These differences were confirmed by comparing the fluorescence of thiochromes obtained before and after the enzymatic hydrolysis of AThTP and AThDP. We have previously shown that complete hydrolysis of AThTP by bacterial membranes yields ThMP as the sole thiamine-containing product [5]. When we incubated synthetic AThDP with a membrane fraction obtained by centrifuging sonicated E. coli, we also observed hydrolysis of this compound with ThMP as product. In these experiments we found that after derivatization, the fluorescence ratios AThcDP/ThcMP and AThcTP/ThcMP were, respectively, 2.1 ± 0.1 and 2.4 ± 0.3, in agreement with a higher fluorescence for adenine thiochrome derivatives than other thiochrome derivatives. The higher fluorescence of adenylated thiochrome derivatives may be caused by either a higher quantum yield for the latter compounds or higher self-quenching in nonadenylated thiochromes. The first possibility seems unlikely because an interaction between adenosine and thiamine moieties, as suggested above, would probably lead to decreased, rather than increased fluorescence. A more probable explanation would be self-quenching in thiochrome, ThcMP, ThcDP and ThcTP, caused by stacking of the molecules; this is possible because of the planar structure of the conjugated thiochrome part. In adenosine thiamine derivatives, because of the U-shaped structure, such stacking would be unlikely to occur.

    Details are in the caption following the image

    Derivatization reaction of thiamine derivatives to thiochrome derivatives (A) and fluorescence excitation (B) and emission (C) spectra of thiochrome derivatives of thiamine, AThDP and AThTP.

    Is AThDP a natural compound?

    AThTP has only very recently been shown to occur naturally in bacteria where it accumulates during carbon starvation [5]. Concerning AThDP, to date, there is no reference to the compound in the scientific literature. However, it was mentioned in at least two patents in 1969 and 1970 [7,8], but no further data have become available since then. It was claimed [7] that some bacteria (Corynebacterium ammoniagenes or C. glutamicum) are able to synthesize AThDP in the presence of suitable precursors (thiamine, ThMP, adenine, adenosine) added to the medium. Under these conditions, the inventors reported that the bacteria accumulated large amounts of AThDP (0.5–1 mg·mL−1) in the culture medium. It was not clear whether AThDP was synthesized inside the bacteria and then excreted or whether it was synthesized in the periplasmic space and then diffused into the fermentation liquor. We repeated these experiments with C. glutamicum and E. coli, but we did not observe any accumulation of AThDP inside or outside the bacteria. Because of the poor description of the methods used in the patent and the lack of any description of the compound synthesized, it is difficult to draw a conclusion concerning the reasons for our failure to reproduce these results. We were also unable to find any mention of AThDP in subsequent patents and any reference to this compound in the peer-reviewed literature.

    However, in E. coli, we observed a transient appearance of AThDP, when the bacteria grown overnight were diluted in Luria–Bertani medium (Fig. 8A). The amounts observed were quite variable, ranging from a few pmol·mg−1 of protein to ∼ 50 pmol·mg−1 of protein, representing a maximum of 2–3% of total thiamine. For comparison, much larger amounts of AThTP could be observed in E. coli under conditions of carbon starvation, i.e. when the bacteria were transferred to a minimal medium without a carbon source. Under these conditions, AThTP slowly accumulates and, after a few hours, reaches a maximum corresponding to ∼ 10–15% of total thiamine [5]. Concerning AThDP, to date, we have no evidence that its appearance might be linked to some kind of cellular stress.

    Details are in the caption following the image

    Occurrence of adenylated thiamine compounds in several organisms: E. coli (A), mouse liver (B), quail liver (C) and cultured 3T3 fibroblasts (D). Bacteria were grown overnight in Luria–Bertani medium and diluted to an absorbance of 0.2–0.4. The sample was taken after 1 h. Mice and quails were decapitated and the livers homogenized in 5 vol. of 12% trichloroacetic acid. Thiamine derivatives were determined by HPLC on a PRP-1 column after transformation to thiochrome derivatives as described in Experimental procedures. The arrows indicate the expected elution times, when the signal was too small to be quantified. 1, ThMP; 2, thiamine; 3, ThDP; 4, AThDP; 5, ThTP; 6, AThTP.

    We then looked for the presence of adenylated thiamine compounds in eukaryotes. We have previously shown that AThTP may be detected in small amounts in yeast, the roots of plants and in several organs in the rat [5]. In the mouse, significant amounts of AThDP were found in the liver (Fig. 8B), although it was below the limits of detection in the brain, heart, kidney and skeletal muscle (Table 3). We also found very small amounts (near the detection limit) of AThDP in quail liver (Fig. 8C), but not in other quail tissue (brain, heart, skeletal muscle). In contrast to mouse tissues, AThTP was never observed in any quail tissues. ThTP, however, was found in relatively high amounts in quail brain (4.6% of total thiamine) and skeletal muscle (1.9% of total thiamine) [1], in small amounts in quail heart (0.15% of total thiamine, this study, not shown), and was hardly detectable in quail liver (≤ 0.1% of total thiamine) (Fig. 8C).

    Table 3. Thiamine derivatives in mouse tissues. The results are expressed as mean ± SD (n = 3). AThDP, adenosine thiamine diphosphate; AThTP, adenosine thiamine triphosphate; ThDP, thiamine diphosphate; ThMP, thiamine monophosphate; ThTP, thiamine triphosphate; nd, not detected.
    Tissue Thiamine ThMP ThDP AThDP ThTP AThTP
    pmol·mg−1 of protein
    Brain 3.4 ± 1.2 16.1 ± 2.3 60 ± 13 nd 0.07 ± 0.02 0.3 ± 0.1
    Skeletal muscle 2 ± 1 3.4 ± 0.5 9 ± 3 nd 0.15 ± 0.03 0.2 ± 0.1
    Heart 1.7 ± 0.2 31 ± 13 361 ± 63 nd 0.2 ± 0.1 0.4 ± 0.1
    Kidney 7 ± 2 50 ± 31 416 ± 54 nd 1.2 ± 0.5 0.2 ± 0.1
    Liver 53 ± 36 252 ± 170 798 ± 257 0.9 ± 0.8 0.9 ± 0.4 1.2 ± 0.2

    In cultured mammalian cells, we found significant amounts of AThTP in 3T3 mouse fibroblasts (Fig. 8D and Table 4), but these cells contained no detectable amounts of AThDP or ThTP. In contrast to 3T3 fibroblasts, Neuro2a neuroblastoma cells contained significant amounts of ThTP but no AThTP. AThDP was not found in any of these cell lines, although it seemed that the commercially available Dulbecco’s modified Eagle’s medium contained a small amount (< 0.01% of thiamine) of this compound.

    Table 4. Thiamine derivatives in several eukaryotic cells lines. The results are expressed as mean ± SD (n is indicated in parentheses). AThDP, adenosine thiamine diphosphate; AThTP, adenosine thiamine triphosphate; ThDP, thiamine diphosphate; ThMP, thiamine monophosphate; ThTP, thiamine triphosphate; nd, not detected.
    Cell line Thiamine ThMP ThDP AThDP ThTP AThTP
    pmol·mg−1 of protein
    Neuro2a (mouse) (4) 22 ± 9 28 ± 10 293 ± 146 nd 2.5 ± 0.2 0.4 ± 0.5
    3T3 (mouse) (6) 80 ± 30 2 ± 1 94 ± 14 nd nd 2.1 ± 0.3
    LN-18 (human) (7) 64 ± 8 4 ± 1 48 ± 1 nd nd 3 ± 1

    Discussion

    In a recent study [5], we reported the presence in E. coli of a new type of nucleotide containing a vitamin part, i.e. AThTP or thiaminylated ATP. We called this compound adenosine thiamine triphosphate to emphasize its close metabolic relationship with thiamine metabolism. Indeed, the intracellular concentrations of these derivatives are orders of magnitude lower than those of conventional adenine nucleotides such as AMP, ADP, ATP or NAD+.

    AThTP was synthesized chemically [5], using a method previously published for the synthesis of ThTP and nucleoside triphosphates [9]. In this study, the reaction was optimized and we found that, along with AThTP, some side products were also synthesized. These were mainly ThTP and another adenosine-containing thiamine compound that was identified as AThDP by MS analysis and 1H-, 13C- and 31P-NMR. The mechanism by which the side products AThDP and ThTP are formed from ThDP and AMP in the presence of excess N,N′-dicyclohexylcarbodiimide (Fig. 2A) is unclear. Our results suggest that, at least in solution, both adenine thiamine compounds should adopt a U-folded structure leading to a through-space interaction between the adenine and thiamine rings.

    The important question, however, is whether the diphosphate analog exists as a natural compound. Here we show that AThDP can indeed be detected in some cell types, both prokaryotic and eukaryotic (in particular liver). This suggests that thiaminylated adenine nucleotides might represent a new family of signaling molecules. These findings are reminiscent of the earlier discovery of diadenosine oligophosphates, which were thought to be a novel class of signaling molecules [14–16]. In prokaryotes, diadenosine tetraphosphate and other members of this family were considered as pleiotropic alarmones produced in response to heat shock or oxidative stress [17]. Our previous results [5] suggested that in E. coli, AThTP is a kind of alarmone produced in response to carbon starvation. The enzymatic synthesis of AThTP requires a new type of enzyme (a ThDP adenylyl transferase) that we partially characterized [6], whereas the synthesis of diadenosine oligophosphates is catalyzed by a completely different mechanism involving aminoacyl-tRNA synthetase [18].

    The finding that vertebrate tissues (especially the liver) contain adenylated thiamine compounds may lead us to re-examine and in some cases question the validity of earlier reports concerning the exact ThTP content of some tissues or the enzymatic mechanisms of ThTP synthesis. For example, several authors [19–22] have claimed that the rat liver had a ThTP content several times higher than the brain. From our data (see Fig. 8B and Table 3), we suspect that peaks corresponding to AThDP and AThTP may have been mistakenly been considered as indicating the presence of ThTP. This would be particularly true in chromatographic methods in which ThTP is eluted first, close to the void volume of the column, increasing the chance of overlap with other compounds such as the here-described adenylated thiamine derivatives. Note that, whereas in mice brain, ThTP is hardly detectable and a significant AThTP peak is observed, the reverse is true in rat brain [5].

    Likewise, synthesis of ‘ThTP’ by soluble enzyme preparations from rat liver [23] and yeast [24] has been reported but, in our laboratory, no synthesis of ThTP was ever observed with soluble preparations, except, unspecifically, with adenylate kinase [4], as reported previously by Kawasaki and coworkers [25,26]. The reason for the discrepancies may be that the authors who described a soluble ThDP kinase [23,24] actually measured the appearance of adenylated thiamine derivatives but not authentic ThTP. Indeed, we recently reported that AThTP synthesis was catalyzed by a soluble enzyme from E. coli or pig brain [6], whereas the synthesis of ThTP seems to require the presence of intact cells or organelles [27].

    In higher organisms, the mechanism of synthesis and degradation of AThDP and AThTP, as well as the possible roles of those compounds, will require further investigation, but our findings emphasize the complexity of thiamine metabolism and further illustrate the concept that the biological role of thiamine derivatives is far from being restricted to the coenzyme role of ThDP [28–31].

    Experimental procedures

    Determination of thiamine compounds by HPLC

    Thiamine compounds, including AThTP and AThDP, were determined by HPLC using a PRP-1 column, as described previously, after transformation to fluorescent thiochrome derivatives [5,32]. Prior to analysis, an 80-μL aliquot was oxidized with 50 μL of 4.3 mm potassium ferricyanide in 15% NaOH. AThTP and AThDP were also quantified using UV detection (254 nm, 535 HPLC detector; Bio-Tek Instruments, Winooski, VT, USA) after separation on a 5-μm Polaris C18 column (150 × 4.6 mm; Varian Benelux, Middelburg, the Netherlands). The mobile phase was composed of 50 mm ammonium acetate adjusted to pH 7.0 and 5% methanol. The flow rate was 1 mL·min−1. All solutions were prepared using milli-Q water (Millipore S.A./N.V., Brussels, Belgium) and all the solvents used for HPLC were of HPLC grade (Biosolve, Valkenswaard, the Netherlands).

    Chemical synthesis and purification of AThDP and AThTP

    AThTP was synthesized by modification of a previously published method [9] for the synthesis of ThTP and nucleoside triphosphates. All products and solvents were from Sigma-Aldrich NV/SA (Bornem, Belgium). Preliminary tests were made using either 0.7 mmol ThDP (acid form) and 0.7 mmol 5′-AMP (acid form) or 0.7 mmol ThMP (acid form) and 0.7 mmol 5′-AMP. The compounds were dissolved in 700 μL tributylamine and 750 μL H2O and mixed until a translucent, slightly viscous, solution was obtained. We diluted 5 μL of this mixture in 500 μL dimethylsulfoxide mixed with 450 μL pyridine and finally added 0.15 g N,N′-dicyclohexylcarbodiimide (dissolved in 50 μL pyridine) to start the synthesis. The reaction was allowed to proceed at room temperature and aliquots were taken at different time intervals, diluted 2000 times in water and analyzed by HPLC. Three main compounds (ThTP, AThDP and AThTP) appeared in the mixture.

    For purification of the compounds, the synthesis was made on a larger scale: 2.25 mmol ThDP (acid form), 3.5 mmol 5′-AMP (acid form), 3.5 mL (14.5 mmol) tributylamine and 3 mL H2O were mixed and dissolved in 500 mL dimethylsulfoxide and 445 mL pyridine. Finally, 45 g N,N′-dicyclohexylcarbodiimide (dissolved in 15 mL pyridine) was added and the mixture was incubated for 2 h at room temperature. Addition of 3 L diethyl ether to the mixture led to the precipitation of synthesized compounds. The suspension was centrifuged (1000 g, 10 min) and the precipitate was dissolved in 40 mL H2O. This solution was applied on a column (8 × 2.5 cm) filled with AG 50W-X8 cation-exchange resin (H+ form; Bio-Rad Laboratories, Nazareth Eke, Belgium) equilibrated in water (pH 4.5 with HCl). The column was washed with 100 mL H2O and 8-mL fractions were collected (flow rate 2 mL·min−1). During this step, ThTP was eluted [9]. All other thiamine derivatives were eluted with 480 mL (60 × 8 mL fractions) ammonium acetate (0.2 m, pH 7.0). Fractions 20–60 were pooled (320 mL) and lyophylized. The powder was dissolved in 25 mL H2O and layered on a column (8 × 2.5 mL) filled with AG-X1 resin (Cl form; Bio-Rad). The resin was washed with 120 mL H2O during which the yellow form was eluted. Residual ThDP and some AThDP were also eluted at this stage. AThDP was eluted with 250 mL ammonium acetate (0.25 m, pH 7.0). The fractions containing AThDP were pooled and lyophilized. AThTP was eluted with 500 mL ammonium acetate (0.5 m, pH 7.0) and lyophilized. The residue was dissolved in 3 mL H2O and filtered on a Millex-GP filter unit (0.22 μm, dia. 25 mm; Millipore). Aliquots of 100 μL of the pool were then purified on a Polaris C18 HPLC column. The mobile phase consisted of 50 mm ammonium acetate and 5% methanol in water and the flow rate was 1 mL·min−1. AThTP was eluted with a retention time of 7 min, and AThDP was eluted after 14 min. The peaks were collected, lyophilized and used for MS analysis and NMR.

    Identification of AThTP and AThDP by ESI tandem MS

    Experiments were performed on a Micromass Q-TOF Ultima Global apparatus (Waters Corp., Zellik, Belgium) operated in nano-ESI positive ion mode. The synthesized compounds were injected at a concentration of 150 μm in 50% water/50% acetonitrile. The source parameters were: capillary voltage, 1.8 kV; cone voltage, 100 V; RF lens 1, 90 V; source temperature, 80 °C; collision energy, 6 eV. The fragmentation pattern of the m/z 674.1 was obtained with 30 V acceleration voltage.

    Characterization of AThDP and AThTP by 1H-NMR, 13C-NMR and 31P-NMR

    One-dimensional 1H-NMR, 13C-NMR and 31P-NMR spectra were recorded at 25 °C (pH 7.4) on a Bruker Avance 500 spectrometer (Bruker Belgium S.A./N.V., Brussels, Belgium) operating at a proton NMR frequency of 500.13 MHz, using a 5-mm probe and a simple pulse-acquire sequence (30° pulses for 1H and 31P and 90° pulse for 13C). Several 2D spectra were also recorded using standard Bruker parameters. NMR data for ThDP, AThDP, ThTP and AThTP are presented in Table 1.

    Determination of fluorescence characteristics of thiochrome derivatives of AThDP and AThTP

    Fluorescence excitation (emission set at 439 nm) and emission (excitation set at 353 nm) spectra were taken using a SFM 25 fluorescence spectrophotometer (Kontron Instruments, Milan, Italy). One milliliter of a 10 μm thiamine, AThDP or AThTP solution was mixed with 500 μL of 4.3 mm potassium ferricyanide in 15% NaOH and the spectra were taken immediately. AThDP and AThTP could be hydrolyzed by a crude membrane preparation from E. coli as described earlier [5].

    Molecular modeling

    All calculations were performed at the quantum chemistry level using the B3LYP functional [33] with the polarized double ζ basis set 6-31G(d) [34] and the gaussian 03 suite of programs (Gaussian Inc, Wallingford, CT, USA, 2004). All the degrees of freedom of the geometry have been fully optimized. Several extended and folded conformations have been generated and located as true energy minima.

    Bacterial cultures

    E. coli (strain BL 21) were grown in Luria–Bertani medium as previously described [5]. C. glutamicum were from the Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH (DSM-No. 1412, Braunschweig, Germany) and grown in MMTG medium (glucose 5 g·L−1; tryptone 10 g·L−1; yeast extract 10 g·L−1, NaCl 5 g·L−1 and methionine 0.2 g·L−1) at 30 °C (250 r.p.m.). The bacteria were then incubated in a medium containing glucose (100 g·L−1), urea (6 g·L−1), K2HPO4 (10 g·L−1), MgSO4·7H2O (10 g·L−1), CaCl2·2H2O (0.1 g·L−1), yeast extract (10 g·L−1) and biotin (30 μg·L−1) as described in Sankyo Company, Ltd [8]. After 48 h at 30 °C, ThMP (2 g·L−1) and adenosine (2 g·L−1) were added and thiamine derivatives were determined by HPLC in the fermentation liquor after 24 h after protein precipitation by 12% trichloroacetic acid. Similar experiments were performed with E. coli in Luria–Bertani medium with either thiamine, ThMP, adenine or adenosine as substrates for AThDP synthesis.

    Eukaryotic cell culture

    Different established cell lines (mouse fibroblast 3T3, human malignant glioma cell line LN-18 and mouse neuroblastoma cell line Neuro2a) were grown at 37 °C in a humidified atmosphere of 95% air, 5% CO2, in 10-cm Petri dishes in 10 mL of Dulbecco’s modified Eagle’s medium (Neuro2a, 3T3) or RPMI (LN-18) supplemented with fetal bovine serum (10%) and penicillin (100 U·mL−1). Cells were subcultured to a fresh culture dish every 2–3 days. At the third day of culture, the medium was discarded and the cells were detached with trypsin (3T3 and LN-18). After centrifugation at 4000 g for 4 min, the pellets were resuspended in 200 μL of trichloroacetic acid 12%. After centrifugation (5000 g, 3 min), the trichloroacetic acid was extracted with diethyl ether. The pellets obtained were dissolved in 0.8 m NaOH for the protein assay.

    Presence of AThDP and AThTP in mouse and quail tissues

    Mice (Mus musculus, C57BL6/129SvJ mixed genetic background) and quails (Coturnix japonica japonica) were decapitated and tissue extracts were prepared as previously described [1]. All animal experiments were made in accordance with the directives of the committee for animal care and use of the University of Liège, in accordance with the European Communities Council Directive of 24 November 1986 (86/609/EEC).

    Acknowledgements

    The authors thank the Fonds de la Recherche Fondamentale Collective (FRFC) for grant 2.4558.04. G. Dive thanks the FRS-FNRS for the financial support of the high performance computing systems installed in Liège and Louvain-la-Neuve. E. de Pauw acknowledges support from the FRS-FNRS for funding of the mass spectrometry facility. G. Mazzucchelly, G. Dive, M. Frédérich and L. Bettendorff are respectively scientific research worker, research associate, senior research associate and research director at the Fonds de la Recherche Scientifique (FRS-FNRS). M. Gangolf and D. Delvaux are research fellows of respectively the FRS-FNRS and the Fonds pour la Formation à la Recherche dans l’Industrie et dans l’Agriculture (FRIA). The quails were a gift from Professor J. Balthazart (Behavioral Neuroendocrinology, GIGA-Neurosciences).