Elsevier

Biochimie

Volume 183, April 2021, Pages 108-125
Biochimie

Thiolatocobalamins repair the activity of pathogenic variants of the human cobalamin processing enzyme CblC

https://doi.org/10.1016/j.biochi.2020.10.006Get rights and content

Highlights

  • Two new thiolatocobalamins, CyaCbl and MpgCbl, were synthesized and characterized.

  • CyaCbl and MpgCbl bound to human CblC in a base-off configuration.

  • CyaCbl and MpgCbl repaired the enzymatic activity of pathogenic variants of CblC.

  • The novel thiolatocobalamins underwent spontaneous dethiolation by CblC.

Abstract

Thiolatocobalamins are a class of cobalamins comprised of naturally occurring and synthetic ligands. Glutathionylcobalamin (GSCbl) occurs naturally in mammalian cells, and also as an intermediate in the glutathione-dependent dealkylation of methylcobalamin (MeCbl) to form cob(I)alamin by pure recombinant CblC from C. elegans. Glutathione-driven deglutathionylation of GSCbl was demonstrated both in mammalian as well as in C. elegans CblC. Dethiolation is orders of magnitude faster than dealkylation of Co–C bonded cobalamins, which motivated us to investigate two synthetic thiolatocobalamins as substrates to repair the enzymatic activity of pathogenic CblC variants in humans. We report the synthesis and kinetic characterization of cysteaminylcobalamin (CyaCbl) and 2-mercaptopropionylglycinocobalamin (MpgCbl). Both CyaCbl and MpgCbl were obtained in high purity (90–95%) and yield (78–85%). UV–visible spectral properties agreed with those reported for other thiolatocobalamins with absorbance maxima observed at 372 nm and 532 nm. Both CyaCbl and MpgCbl bound to wild type human recombinant CblC inducing spectral blue-shifts characteristic of the respective base-on to base-off transitions. Addition of excess glutathione (GSH) resulted in rapid elimination of the β-ligand to give aquacobalamin (H2OCbl) as the reaction product under aerobic conditions. Further, CyaCbl and MpgCbl underwent spontaneous dethiolation thereby repairing the loss of activity of pathogenic variants of human CblC, namely R161G and R161Q. We posit that thiolatocobalamins could be exploited therapeutically for the treatment of inborn errors of metabolism that impair processing of dietary and supplemental cobalamin forms. While these disorders are targets for newborn screening in some countries, there is currently no effective treatment available to patients.

Introduction

Cobalamin (Cbl) is an essential micronutrient that is exclusively synthesized by some groups of bacteria and archaea [1]. Structurally, cobalamins are tetrapyrroles coordinating a cobalt center. The corrin ring presents seven acetamide and propionamide side chains and ligand 5-dimethylbenzimidazole via a phosphoester linkage (Fig. 1). The Co center can be coordinated axially by a 5-dimethylbenzimidazole moiety in the 5th coordination position (α-ligand) and by a number of possible chemical groups in the 6th coordination position (β-ligand). Based on whether the 5-dimethylbenzimidazole moiety is bound to the cobalt center via a N-atom or unbound, cobalamins exist in two configurations, namely, base-on and base-off, respectively. The upper or 6th coordination position can be occupied by a variety of biologically relevant as well as synthetic ligands via Co–C, Co–N, Co–O and Co–S bonds [[2], [3], [4], [5], [6]]. Likewise, biologically relevant Cbl forms exploit all three possible oxidation states of the cobalt center to give cob (III)alamin, cob(II)alamin and cob(I)alamin [5,7]. The interaction of Cbls with sulfur compounds represents an important part of their redox and coordination chemistry; for example, reactions with thiols not only produce stable cob(III)alamin complexes, but also increase reactivity of the corresponding cob(II)alamin species [5,7].

Humans obtain B12 consumption of animal food sources. Because B12 does not occur in the plant kingdom, individuals who adhere to plant-based nutrition must rely on supplemental forms of B12. Cobalamin is required to drive the enzymatic reactions of cytosolic methionine synthase (MS) and of mitochondrial methylmalonyl-CoA mutase (MUT) [8]. In the catalytic cycle of MS, homocysteine is methylated by methylcobalamin (MeCbl) to form the amino acid methionine. Cob(I)alamin, the other product of this reaction, is then remethylated by N5-methyltetrahydrofolate, to regenerate MeCbl and tetrahydrofolate. Occasional unwanted oxidation of cob(I)alamin to form cob(II)alamin is repaired by the enzyme methionine synthase reductase, thus returning the Cbl moiety into the catalytic cycle of MS as cob(I)alamin. The reaction driven by MUT comprises cycling of adenosylcobalamin (AdoCbl) and cob(II)alamin for the isomerization of methylmalonyl-CoA into succinyl-CoA. Failed reduction of cob(II)alamin to cob(I)alamin leads to the formation of aquacobalamin (H2OCbl). Prior to delivery to MS and MUT dietary cobalamins undergo chemical processing by the processing enzyme CblC (a.k.a. MMACHC) [8,9] and partition into cytosol and mitochondrion orchestrated by interactions of CblC with the adaptor protein CblD (MMADHC) [[10], [11], [12]]. Evidence for multi-partite protein complexes involving CblC, CblD and recipient protein MS has also been reported [13].

As expected from the above described metabolism of vitamin B12, the major Cbl forms found in foods are MeCbl, AdoCbl and H2OCbl [[14], [15], [16], [17]]. Cyanocobalamin (CNCbl), the most inexpensive commercial form of vitamin B12 occurs in ocean water [18] but it is absent in human tissue and fluids unless previously taken as an oral supplement or by intramuscular injection. Mutations in the cblC gene (MMACHC) disrupt the cellular processing of dietary cobalamins. This pathology is known as the cblC disease and is characterized by combined homocystinuria with methylmalonic aciduria. The cblC disease is the most common inherited disorder of intracellular cobalamin metabolism. Current treatment with hydroxycobalamin does unfortunately not prevent from the development of clinical symptoms. The cblC disorder has a spectrum of severity with two distinct phenotypic forms: early and late onset. Early-onset patients present with intrauterine growth retardation (IUGR), microcephaly, failure to thrive, developmental delay, hypotonia, progressive retinopathy, and maculopathy [19]. Late-onset CblC patients usually present with extrapyramidal and neuropsychiatric symptoms in any decade of life [20]. In some countries cblC disease is also part of newborn screening programs. An intrauterine treatment is discussed in order to prevent the early onset phenotype [21].

Early work suggested that the thiolatocobalamin glutathionylcobalamin (GSCbl), and sulfitocobalamin (SO3Cbl) also occur naturally, and that they may represent reaction intermediates in the intracellular processing of dietary cobalamins [22]. Comprehensive profiling of cobalamins in mammalian fluids and cells using [57Co]-cyanocobalamin as a precursor demonstrated that GSCbl and SO3Cbl are indeed part of the endogenous cobalamin pool [14,23]. Work performed with the cobalamin processing enzyme CblC later demonstrated that glutathione (GSH) serves as the nucleophile for the dealkylation of MeCbl and AdoCbl as well as for the decyanation of CNCbl, but these reactions occur without formation of a GSCbl intermediate [24,25]. The enzyme CblC catalyzes a reaction that is from the point of view of pH considerations and redox potentials, otherwise unattainable under the conditions of the biological milieu. Firstly, dealkylation of MeCbl and AdoCbl requires weakening of the Co–C bond, which is achieved by induction of the base-off configuration of the α-ligand thus leading to a trans-weakening effect on the β-ligand [26,27]. Because the pKa for the base-on to base-off transitions of biologically relevant Cbls MeCbl, AdoCbl and H2OCbl are 2.90 [28], 3.50 [5], and −2.40 [29], respectively, this change in configuration requires enzyme-mediated distortion of the α-ligand. Secondly, disruption of the Co–C bond most typically requires reduction of the cobalt center, which in biologically relevant cobalamins means a thermodynamic barrier of approximately 1 V [5]. This requires protein-mediated reduction as none of the known intracellular reductants could reach this thermodynamic threshold. In humans, CblC is responsible for the induction of the base-off conformation of cobalamins and glutathione-mediated rupture of the Co–C via nucleophilic attack and reductive elimination [24]. Recent mechanistic studies with non-mammalian variants of CblC demonstrated that GSCbl does form as a stable reaction intermediate in reactions of MeCbl dealkylation by GSH [30]. This suggests that the reactivity of cobalamins with glutathione to form the stable thiolatocobalamin GSCbl has changed through evolution to adjust to nutritional demands or perhaps, to environmental availability of the micronutrient. In comparison to alkylcobalamins, the reduction of thiolatocobalamins requires large negative potentials [31]. At the same time, the Co(III)–S bond is more reactive than the Co(III)–C bond; for example, GSCbl can be readily reduced by selenocysteine [32] whereas its reaction with MeCbl is substantially slower (unpublished). Conversion of the thiolatocobalamins from the base-on to the base-off configuration might destabilize the Co(III)–S bond toward the Co(II)–S state, as suggested by the lower stability of the nucleotide-free Cbl analogue glutathionylcobamide compared to GSCbl [33].

Dethiolation of GSCbl by CblC in the presence of GSH yields the catalytically active cob(II)alamin and cob(I)alamin when reactions are performed under anaerobic conditions reactions [[34], [35], [36]]. Based on these findings, we hypothesized that the Co–S bond enables a more facile conversion of base-on to base-off conformation of the Cbl substrate, thereby decreasing thermodynamic barriers of redox potential that are necessary for the removal of the β-axial ligand. Herein, we describe the synthesis and spectroscopic characterization of two novel thiolatocobalamins, CyaCbl and MpgCbl. The two cobalamins were synthesized in pure form and exhibited spectral properties similar to that of other thiolatocobalamins. CyaCbl and MpgCbl bound to human CblC inducing their respective base-off configurations, and served as substrates for dethiolation in reactions driven with GSH under physiologically relevant conditions. Under aerobic conditions, this reaction yielded H2OCbl as the only cobalamin species. The new thiolatocobalamins CyaCbl and MpgCbl repaired the activity of human pathogenic variants R161G and R161Q, responsible for early and late onset cblC disease in humans, respectively. Our study enriches the repertoire of thiolatocobalamins with biological activity that are suitable for the repair of pathogenic variants of CblC that lead to combined homocystinuria and methylmalonic aciduria in humans. These new compounds may represent an effective treatment alternative.

Section snippets

General procedures and chemicals

Hydroxocobalamin hydrochloride (Sigma, FW 1382.82, product Nr. 95200-1G), cysteamine (Sigma, FW 113.61, product Nr. 30078-25G) and N-(2-mercaptopropionyl)-glycine (Mpg, also known as Tiopronin, FW 163.19, Sigma product Nr. M6635-5G) were used without further purification. Water was purified using a PureLab classic water purification system from ELGA. Milli Q water was used for all reactions. Water (product Nr. 39253) and methanol (product Nr. 34966) for mass spectrometry were purchased from

MpgCbl

MpgCbl was synthesized by addition of an aqueous solution of Mpg (1.9 mol equiv., phosphate buffer, pH 7.2) to an aqueous solution of H2OCbl, in accordance to a previously reported procedure to synthesize other thiolatocobalamins [38]. Formation of MpgCbl was followed by UV–visible spectroscopy, as H2OCbl and thiolatocobalamins have clearly different UV–visible spectra.

CyaCbl

Preliminary tests showed that Cya and H2OCbl react much faster under mild acidic conditions, a finding that is in agreement

Discussion

Evidence of the formation of complexes between thiols and aquacobalamin date back to the early 1960’s [48,49]. The reaction of cysteamine with aquacobalamin was first described by Cavallini et al. [41]. Paucity in further investigation of this reaction partly motivated us to explore the biological properties and potential pharmacological utility of cysteaminylcobalamin and the new derivative mercaptopropionylglycylcobalamin. Several studies on the reaction mechanism of the CblC protein

Conclusions

We synthesized two novel thiolatocobalamins that are stable under physiologically relevant conditions. The two novel thiolatocobalamins bound to wild type and pathogenic variants of human CblC inducing blue-shifted spectra consistent with the formation of base-off cobalamin species. Both GSCbl and new thiolatocobalamins CyaCbl and MpgCbl support dethiolation reactions by human wild type CblC and repair the enzymatic activity of pathogenic variants R161G and R161Q. Furthermore, MpgCbl and CyaCbl

Author contributions

V.W., S.M., A.J.E., S.B., M.K., S.T. and L.H. performed experiments and analyzed data. I.D., S.V.M., D.W.J., U.S. and L.H. analyzed data and critically reviewed the manuscript. V.W. wrote the first draft of the manuscript. I.A.D., S.V.M., D.W.J, U.S. and L.H. wrote and edited the manuscript. L.H. conceived the study and supervised all facets of the experimental work.

Funding

This study was supported with intramural funds from the Department of General Pediatrics, Adolescent Medicine and Neonatology, Faculty of Medicine, Medical Center, University of Freiburg, Germany.

Availability of data and materials

The data and materials used in this study are available from the corresponding author on reasonable request.

Ethical approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Disclosures

The novel cobalamins CyaCbl and MpgCbl are part of a patent application (L.H., filed by University of Freiburg, International Application Nr. PCT/EP2018/063597, Publication Nr. WO/2018/215578).

Declaration of competing interest

The authors declare no conflicts of interest.

Acknowledgements

The authors are grateful to Katharina Klotz for excellent technical support.

References (61)

  • Z. Li et al.

    Glutathione-dependent one-electron transfer reactions catalyzed by a B(1)(2) trafficking protein

    J. Biol. Chem.

    (2014)
  • Z. Li et al.

    Coordination chemistry controls the thiol oxidase activity of the B12-trafficking protein CblC

    J. Biol. Chem.

    (2017)
  • J. Jeong et al.

    Processing of glutathionylcobalamin by a bovine B12 trafficking chaperone bCblC involved in intracellular B12 metabolism

    Biochem. Biophys. Res. Commun.

    (2014)
  • L. Hannibal et al.

    Influence of heme-thiolate in shaping the catalytic properties of a bacterial nitric-oxide synthase

    J. Biol. Chem.

    (2011)
  • H.A. Barker et al.

    Isolation and properties of crystalline cobamide coenzymes containing benzimidazole or 5, 6-dimethylbenzimidazole

    J. Biol. Chem.

    (1960)
  • D. Cavallini et al.

    The inability of thiols to reduce cobalamins in the absence of a metalion

    FEBS Lett.

    (1968)
  • C. Gherasim et al.

    Pathogenic mutations differentially affect the catalytic activities of the human B12-processing chaperone CblC and increase futile redox cycling

    J. Biol. Chem.

    (2015)
  • N.E. Brasch et al.

    Synthesis and characterization of isolable thiolatocobalamin complexes relevant to coenzyme B12-dependent ribonucleoside triphosphate reductase

    J. Inorg. Biochem.

    (1999)
  • D. Cavallini et al.

    Spectrophotometric determination of deoxyadenosylcobalamin [CoB12] in the presence of B12 compounds

    Methods Enzymol.

    (1971)
  • D.S. Froese et al.

    Mechanism of vitamin B12-responsiveness in cblC methylmalonic aciduria with homocystinuria

    Mol. Genet. Metabol.

    (2009)
  • D.S. Froese et al.

    Thermolability of mutant MMACHC protein in the vitamin B12-responsive cblC disorder

    Mol. Genet. Metabol.

    (2010)
  • K.H. Weisgraber et al.

    Human E apoprotein heterogeneity. Cysteine-arginine interchanges in the amino acid sequence of the apo-E isoforms

    J. Biol. Chem.

    (1981)
  • J.R. Roth et al.

    Cobalamin (coenzyme B12): synthesis and biological significance

    Annu. Rev. Microbiol.

    (1996)
  • L. Randaccio et al.

    Crystal chemistry of cobalamins. Structural characterization of the Co-S bond in cobalamins

    Inorg. Chem.

    (1999)
  • L. Randaccio et al.

    Vitamin B12: unique metalorganic compounds and the most complex vitamins

    Molecules

    (2010)
  • E.M. Scheuring et al.

    Sulfur-containing cobalamins: X-ray absorption spectroscopic characterization

    Biochemistry

    (1994)
  • L. Hannibal et al.

    Intracellular processing and utilization of cobalamins

  • L. Hannibal et al.

    Proteomics of vitamin B12 processing

    Clin. Chem. Lab. Med.

    (2013)
  • J. Jusufi et al.

    Characterization of functional domains of the cblD (MMADHC) gene product

    J. Inherit. Metab. Dis.

    (2014)
  • P. Gimsing et al.
  • Cited by (8)

    • Versatile enzymology and heterogeneous phenotypes in cobalamin complementation type C disease

      2022, iScience
      Citation Excerpt :

      These findings have generated renewed interest in the use of thiolatocobalamins as possible drug candidates in diseases involving a defective cobalamin metabolism. In recent studies on the enzymatic activity of MMACHC and pathogenic mutants of MMACHC using synthetic thiolatocobalamins as substrates, a 4-fold increase in rates for β-deligation in comparison with MeCbl has been reported (Wingert et al., 2021). Additionally, these synthetic thiolatocobalamins featuring a reductant β-ligand joined to the cobalamin scaffold, undergo spontaneous β-deligation when MMACHC-bound in the absence of GSH.

    • Intracellular processing of vitamin B<inf>12</inf> by MMACHC (CblC)

      2022, Vitamins and Hormones
      Citation Excerpt :

      Besides decyanation of CNCbl and dealkylation of MeCbl, AdoCbl and synthetic alkylcobalamins, CblC has been remarkable versatile toward other forms of B12 with β-axial ligands that include CoS (thiolatocobalamins) (Pezacka et al., 1990) and CoN bonding (nitrocobalamin) (Mascarenhas, Li, Gherasim, Ruetz, & Banerjee, 2020). Thiolatocobalamin dethiolation was first demonstrated in early studies with cell extracts by Pezacka et al. (1990) and in more recent work with bovine recombinant CblC (Jeong, Park, Park, & Kim, 2014) and human recombinant CblC (Wingert et al., 2021). In vitro studies with the recombinant CblC protein showed that dethiolation is orders of magnitude faster than the dealkylation of alkylcobalamins, and requires lower concentration of co-substrate GSH (Jeong et al., 2014; Wingert et al., 2021).

    View all citing articles on Scopus
    View full text