Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(11): e2220677120, 2023 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-36888659

RESUMEN

Control over transition metal redox state is essential for metalloprotein function and can be achieved via coordination chemistry and/or sequestration from bulk solvent. Human methylmalonyl-Coenzyme A (CoA) mutase (MCM) catalyzes the isomerization of methylmalonyl-CoA to succinyl-CoA using 5'-deoxyadenosylcobalamin (AdoCbl) as a metallocofactor. During catalysis, the occasional escape of the 5'-deoxyadenosine (dAdo) moiety leaves the cob(II)alamin intermediate stranded and prone to hyperoxidation to hydroxocobalamin, which is recalcitrant to repair. In this study, we have identified the use of bivalent molecular mimicry by ADP, coopting the 5'-deoxyadenosine and diphosphate moieties in the cofactor and substrate, respectively, to protect against cob(II)alamin overoxidation on MCM. Crystallographic and electron paramagnetic resonance (EPR) data reveal that ADP exerts control over the metal oxidation state by inducing a conformational change that seals off solvent access, rather than by switching five-coordinate cob(II)alamin to the more air stable four-coordinate state. Subsequent binding of methylmalonyl-CoA (or CoA) promotes cob(II)alamin off-loading from MCM to adenosyltransferase for repair. This study identifies an unconventional strategy for controlling metal redox state by an abundant metabolite to plug active site access, which is key to preserving and recycling a rare, but essential, metal cofactor.


Asunto(s)
Imitación Molecular , Vitamina B 12 , Humanos , Oxidación-Reducción , Adenosina Difosfato/metabolismo , Vitamina B 12/metabolismo , Metilmalonil-CoA Mutasa/química , Metilmalonil-CoA Mutasa/metabolismo
2.
J Am Chem Soc ; 2023 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-37916782

RESUMEN

Cobalt-sulfur (Co-S) coordination is labile to both oxidation and reduction chemistry and is rarely seen in nature. Cobalamin (or vitamin B12) is an essential cobalt-containing organometallic cofactor in mammals and is escorted via an intricate network of chaperones to a single cytoplasmic target, methionine synthase. In this study, we report that the human cobalamin trafficking protein, MMADHC, exploits the chemical lability of Co-S coordination for cofactor off-loading onto methionine synthase. Cys-261 on MMADHC serves as the ß-axial ligand to cobalamin. Complex formation between MMADHC and methionine synthase is signaled by loss of the lower axial nitrogen ligand, leading to five-coordinate thiolato-cobalamin. Nucleophilic displacement by the vicinal thiolate, Cys-262, completes cofactor transfer to methionine synthase and release of a cysteine disulfide-containing MMADHC. The physiological relevance of this mechanism is supported by clinical variants of MMADHC, which impair cofactor binding and off-loading, explaining the molecular basis of the associated homocystinuria.

3.
Inorg Chem ; 62(32): 12630-12633, 2023 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-37526260

RESUMEN

Cobalamin (or vitamin B12)-dependent enzymes and trafficking chaperones exploit redox-linked coordination chemistry to control the cofactor reactivity during catalysis and translocation. As the cobalt oxidation state decreases from 3+ to 1+, the preferred cobalamin geometry changes from six- to four-coordinate (4-c). In this study, we reveal the sizable thermodynamic gain that accrues for human adenosine triphosphate (ATP):cob(I)alamin adenosyltransferase (or MMAB) by enforcing an unfavorable 4-c cob(II)alamin geometry. MMAB-bound cob(II)alamin is reduced to the supernucleophilic cob(I)alamin intermediate during the synthesis of 5'-deoxyadenosylcobalamin. Herein, we report the first experimentally determined reduction potential for 4-c cob(II)alamin (-325 ± 9 mV), which is 180 mV more positive than for the five-coordinate (5-c) water-liganded species. The redox potential of MMAB-bound cob(II)alamin is within the range of adrenodoxin, which we demonstrate functions as an electron donor. We also show that stabilization of 5-c cob(II)alamin by a subset of MMAB patient variants compromises the reduction by adrenodoxin, explaining the underlying pathogenic mechanism.


Asunto(s)
Adenosina Trifosfato , Adrenodoxina , Humanos , Vitamina B 12
4.
Proc Natl Acad Sci U S A ; 117(48): 30412-30422, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33199623

RESUMEN

Cobalamin is a complex organometallic cofactor that is processed and targeted via a network of chaperones to its dependent enzymes. AdoCbl (5'-deoxyadenosylcobalamin) is synthesized from cob(II)alamin in a reductive adenosylation reaction catalyzed by adenosyltransferase (ATR), which also serves as an escort, delivering AdoCbl to methylmalonyl-CoA mutase (MCM). The mechanism by which ATR signals that its cofactor cargo is ready (AdoCbl) or not [cob(II)alamin] for transfer to MCM, is not known. In this study, we have obtained crystallographic snapshots that reveal ligand-induced ordering of the N terminus of Mycobacterium tuberculosis ATR, which organizes a dynamic cobalamin binding site and exerts exquisite control over coordination geometry, reactivity, and solvent accessibility. Cob(II)alamin binds with its dimethylbenzimidazole tail splayed into a side pocket and its corrin ring buried. The cosubstrate, ATP, enforces a four-coordinate cob(II)alamin geometry, facilitating the unfavorable reduction to cob(I)alamin. The binding mode for AdoCbl is notably different from that of cob(II)alamin, with the dimethylbenzimidazole tail tucked under the corrin ring, displacing the N terminus of ATR, which is disordered. In this solvent-exposed conformation, AdoCbl undergoes facile transfer to MCM. The importance of the tail in cofactor handover from ATR to MCM is revealed by the failure of 5'-deoxyadenosylcobinamide, lacking the tail, to transfer. In the absence of MCM, ATR induces a sacrificial cobalt-carbon bond homolysis reaction in an unusual reversal of the heterolytic chemistry that was deployed to make the same bond. The data support an important role for the dimethylbenzimidazole tail in moving the cobalamin cofactor between active sites.


Asunto(s)
Transferasas Alquil y Aril/química , Transferasas Alquil y Aril/metabolismo , Cobamidas/química , Cobamidas/metabolismo , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Adenosina Trifosfato/química , Adenosina Trifosfato/metabolismo , Sitios de Unión , Catálisis , Dominio Catalítico , Cinética , Modelos Biológicos , Conformación Molecular , Complejos Multiproteicos , Unión Proteica , Relación Estructura-Actividad
5.
J Biol Chem ; 297(6): 101373, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34757128

RESUMEN

Human ATP:cob(I)alamin adenosyltransferase (ATR) is a mitochondrial enzyme that catalyzes an adenosyl transfer to cob(I)alamin, synthesizing 5'-deoxyadenosylcobalamin (AdoCbl) or coenzyme B12. ATR is also a chaperone that escorts AdoCbl, transferring it to methylmalonyl-CoA mutase, which is important in propionate metabolism. Mutations in ATR lead to methylmalonic aciduria type B, an inborn error of B12 metabolism. Our previous studies have furnished insights into how ATR protein dynamics influence redox-linked cobalt coordination chemistry, controlling its catalytic versus chaperone functions. In this study, we have characterized three patient mutations at two conserved active site residues in human ATR, R190C/H, and E193K and obtained crystal structures of R190C and E193K variants, which display only subtle structural changes. All three mutations were found to weaken affinities for the cob(II)alamin substrate and the AdoCbl product and increase KM(ATP). 31P NMR studies show that binding of the triphosphate product, formed during the adenosylation reaction, is also weakened. However, although the kcat of this reaction is significantly diminished for the R190C/H mutants, it is comparable with the WT enzyme for the E193K variant, revealing the catalytic importance of Arg-190. Furthermore, although the E193K mutation selectively impairs the chaperone function by promoting product release into solution, its catalytic function might be unaffected at physiological ATP concentrations. In contrast, the R190C/H mutations affect both the catalytic and chaperoning activities of ATR. Because the E193K mutation spares the catalytic activity of ATR, our data suggest that the patients carrying this mutation are more likely to be responsive to cobalamin therapy.


Asunto(s)
Adenosina Trifosfato/metabolismo , Transferasas Alquil y Aril/metabolismo , Chaperonas Moleculares/metabolismo , Mutación , Transferasas Alquil y Aril/química , Catálisis , Dominio Catalítico , Espectroscopía de Resonancia por Spin del Electrón , Humanos , Cinética , Unión Proteica
6.
J Biol Chem ; 295(28): 9630-9640, 2020 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-32457044

RESUMEN

In humans, cobalamin or vitamin B12 is delivered to two target enzymes via a complex intracellular trafficking pathway comprising transporters and chaperones. CblC (or MMACHC) is a processing chaperone that catalyzes an early step in this trafficking pathway. CblC removes the upper axial ligand of cobalamin derivatives, forming an intermediate in the pathway that is subsequently converted to the active cofactor derivatives. Mutations in the cblC gene lead to methylmalonic aciduria and homocystinuria. Here, we report that nitrosylcobalamin (NOCbl), which was developed as an antiproliferative reagent, and is purported to cause cell death by virtue of releasing nitric oxide, is highly unstable in air and is rapidly oxidized to nitrocobalamin (NO2Cbl). We demonstrate that CblC catalyzes the GSH-dependent denitration of NO2Cbl forming 5-coordinate cob(II)alamin, which had one of two fates. It could be oxidized to aquo-cob(III)alamin or enter a futile thiol oxidase cycle forming GSH disulfide. Arg-161 in the active site of CblC suppressed the NO2Cbl-dependent thiol oxidase activity, whereas the disease-associated R161G variant stabilized cob(II)alamin and promoted futile cycling. We also report that CblC exhibits nitrite reductase activity, converting cob(I)alamin and nitrite to NOCbl. Finally, the denitration activity of CblC supported cell proliferation in the presence of NO2Cbl, which can serve as a cobalamin source. The newly described nitrite reductase and denitration activities of CblC extend its catalytic versatility, adding to its known decyanation and dealkylation activities. In summary, upon exposure to air, NOCbl is rapidly converted to NO2Cbl, which is a substrate for the B12 trafficking enzyme CblC.


Asunto(s)
Nitrito Reductasas , Oxidorreductasas , Vitamina B 12/análogos & derivados , Transporte Biológico Activo , Catálisis , Células HT29 , Humanos , Nitrito Reductasas/química , Nitrito Reductasas/metabolismo , Oxidorreductasas/química , Oxidorreductasas/metabolismo , Unión Proteica , Vitamina B 12/química , Vitamina B 12/metabolismo
7.
Inorg Chem ; 59(21): 16065-16072, 2020 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-33074687

RESUMEN

CblC is a chaperone that catalyzes removal of the ß-axial ligand of cobalamin (or B12), generating cob(II)alamin in an early step in the cofactor trafficking pathway. Cob(II)alamin is subsequently partitioned to support cellular needs for the synthesis of active cobalamin cofactor derivatives. In addition to the ß-ligand transferase activity, the Caenorhabdiitis elegans CblC (ceCblC) and clinical R161G/Q variants of the human protein exhibit robust thiol oxidase activity, converting glutathione to glutathione disulfide while concomitantly reducing O2 to H2O2. The chemical efficiency of the thiol oxidase side reaction during ceCblC-catalyzed dealkylation of alkylcobalamins is noteworthy in that it effectively scrubs ambient oxygen from the reaction mixture, leading to air stabilization of the highly reactive cob(I)alamin product. In this study, we report that the enhanced thiol oxidase activity of ceCblC requires the presence of KCl, which explains how the wasteful thiol oxidase activity is potentially curtailed inside cells where the chloride concentration is low. We have captured an unusual chlorocob(II)alamin intermediate that is formed in the presence of potassium chloride, a common component of the reaction buffer, and have characterized it by electron paramagnetic resonance, magnetic circular dichroism, and computational analyses. The ability to form a chlorocob(II)alamin intermediate could represent an evolutionary vestige in ceCblC, which is structurally related to bacterial B12-dependent reductive dehalogenases that have been proposed to form halogen cob(II)alamin intermediates in their catalytic cycle.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Glutatión Transferasa/metabolismo , Oxidorreductasas/metabolismo , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Vitamina B 12/biosíntesis , Biocatálisis , Proteínas de Caenorhabditis elegans/química , Glutatión Transferasa/química , Modelos Moleculares , Proteínas Proto-Oncogénicas c-cbl/química , Vitamina B 12/química
8.
J Biol Chem ; 292(16): 6512-6528, 2017 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-28246171

RESUMEN

Hydrogen sulfide is a critical signaling molecule, but high concentrations cause cellular toxicity. A four-enzyme pathway in the mitochondrion detoxifies H2S by converting it to thiosulfate and sulfate. Recent studies have shown that globins like hemoglobin and myoglobin can also oxidize H2S to thiosulfate and hydropolysulfides. Neuroglobin, a globin enriched in the brain, was reported to bind H2S tightly and was postulated to play a role in modulating neuronal sensitivity to H2S in conditions such as stroke. However, the H2S reactivity of the coordinately saturated heme in neuroglobin is expected a priori to be substantially lower than that of the 5-coordinate hemes present in myoglobin and hemoglobin. To resolve this discrepancy, we explored the role of the distal histidine residue in muting the reactivity of human neuroglobin toward H2S. Ferric neuroglobin is slowly reduced by H2S and catalyzes its inefficient oxidative conversion to thiosulfate. Mutation of the distal His64 residue to alanine promotes rapid binding of H2S and its efficient conversion to oxidized products. X-ray absorption, EPR, and resonance Raman spectroscopy highlight the chemically different reaction options influenced by the distal histidine ligand. This study provides mechanistic insights into how the distal heme ligand in neuroglobin caps its reactivity toward H2S and identifies by cryo-mass spectrometry a range of sulfide oxidation products with 2-6 catenated sulfur atoms with or without oxygen insertion, which accumulate in the absence of the His64 ligand.


Asunto(s)
Globinas/química , Sulfuro de Hidrógeno/química , Proteínas del Tejido Nervioso/química , Catálisis , Cristalografía por Rayos X , Cisteína/química , Espectroscopía de Resonancia por Spin del Electrón , Hemo/química , Hemoglobinas/química , Histidina/química , Humanos , Concentración de Iones de Hidrógeno , Cinética , Ligandos , Espectrometría de Masas , Mutación , Mioglobina/química , Neuroglobina , Oxígeno/química , Conformación Proteica , Espectrometría de Masa por Ionización de Electrospray , Espectrometría Raman , Sulfuros/química , Tiosulfatos/química , Trombina/química
9.
J Biol Chem ; 292(23): 9733-9744, 2017 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-28442570

RESUMEN

The cobalamin or B12 cofactor supports sulfur and one-carbon metabolism and the catabolism of odd-chain fatty acids, branched-chain amino acids, and cholesterol. CblC is a B12-processing enzyme involved in an early cytoplasmic step in the cofactor-trafficking pathway. It catalyzes the glutathione (GSH)-dependent dealkylation of alkylcobalamins and the reductive decyanation of cyanocobalamin. CblC from Caenorhabditis elegans (ceCblC) also exhibits a robust thiol oxidase activity, converting reduced GSH to oxidized GSSG with concomitant scrubbing of ambient dissolved O2 The mechanism of thiol oxidation catalyzed by ceCblC is not known. In this study, we demonstrate that novel coordination chemistry accessible to ceCblC-bound cobalamin supports its thiol oxidase activity via a glutathionyl-cobalamin intermediate. Deglutathionylation of glutathionyl-cobalamin by a second molecule of GSH yields GSSG. The crystal structure of ceCblC provides insights into how architectural differences at the α- and ß-faces of cobalamin promote the thiol oxidase activity of ceCblC but mute it in wild-type human CblC. The R161G and R161Q mutations in human CblC unmask its latent thiol oxidase activity and are correlated with increased cellular oxidative stress disease. In summary, we have uncovered key architectural features in the cobalamin-binding pocket that support unusual cob(II)alamin coordination chemistry and enable the thiol oxidase activity of ceCblC.


Asunto(s)
Proteínas de Caenorhabditis elegans/química , Caenorhabditis elegans/enzimología , Proteínas Portadoras/química , Cobamidas/química , Estrés Oxidativo , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Cobamidas/genética , Cobamidas/metabolismo , Humanos , Mutación Missense , Oxidorreductasas , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro
10.
J Am Chem Soc ; 140(41): 13205-13208, 2018 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-30282455

RESUMEN

A sophisticated intracellular trafficking pathway in humans is used to tailor vitamin B12 into its active cofactor forms, and to deliver it to two known B12-dependent enzymes. Herein, we report an unexpected strategy for cellular retention of B12, an essential and reactive cofactor. If methylmalonyl-CoA mutase is unavailable to accept the coenzyme B12 product of adenosyltransferase, the latter catalyzes homolytic scission of the cobalt-carbon bond in an unconventional reversal of the nucleophilic displacement reaction that was used to make it. The resulting homolysis product binds more tightly to adenosyltransferase than does coenzyme B12, facilitating cofactor retention. We have trapped, and characterized spectroscopically, an intermediate in which the cobalt-carbon bond is weakened prior to being broken. The physiological relevance of this sacrificial catalytic activity for cofactor retention is supported by the significantly lower coenzyme B12 concentration in patients with dysfunctional methylmalonyl-CoA mutase but normal adenosyltransferase activity.


Asunto(s)
Cobamidas/metabolismo , Transferasas Alquil y Aril/química , Transferasas Alquil y Aril/metabolismo , Carbono/química , Dominio Catalítico , Cobalto/química , Cobamidas/química , Fibroblastos/metabolismo , Humanos , Metilmalonil-CoA Mutasa/metabolismo , Estructura Molecular
11.
PLoS Pathog ; 12(10): e1005949, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27760199

RESUMEN

The methylfolate trap, a metabolic blockage associated with anemia, neural tube defects, Alzheimer's dementia, cardiovascular diseases, and cancer, was discovered in the 1960s, linking the metabolism of folate, vitamin B12, methionine and homocysteine. However, the existence or physiological significance of this phenomenon has been unknown in bacteria, which synthesize folate de novo. Here we identify the methylfolate trap as a novel determinant of the bacterial intrinsic death by sulfonamides, antibiotics that block de novo folate synthesis. Genetic mutagenesis, chemical complementation, and metabolomic profiling revealed trap-mediated metabolic imbalances, which induced thymineless death, a phenomenon in which rapidly growing cells succumb to thymine starvation. Restriction of B12 bioavailability, required for preventing trap formation, using an "antivitamin B12" molecule, sensitized intracellular bacteria to sulfonamides. Since boosting the bactericidal activity of sulfonamides through methylfolate trap induction can be achieved in Gram-negative bacteria and mycobacteria, it represents a novel strategy to render these pathogens more susceptible to existing sulfonamides.


Asunto(s)
Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Bacterias/metabolismo , Farmacorresistencia Microbiana/fisiología , Ácido Fólico/metabolismo , Homocisteína/metabolismo , Metionina/metabolismo , Pruebas de Sensibilidad Microbiana , Vitamina B 12/metabolismo
12.
Chemistry ; 23(41): 9726-9731, 2017 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-28557054

RESUMEN

Organometallic aryl-cobalamins are B12 -derivatives featuring properties of potential 'B12 antivitamins'. Herein, we describe a new method for the preparation of aryl-cobalamins using versatile diaryliodonium salts as arylation agents. Formate or sodium borohydride reduction of aquocobalamin in presence of diphenyliodonium chloride furnished Coß -phenyl-cobalamin PhCbl in a roughly 3:1 to 1:1 ratio with its coordination isomer αPhCbl, a first representative 'base-off' Coα -aryl-cobalamin. The new structures were secured by detailed spectroscopic analysis, supplemented by an X-ray crystal structure analysis of PhCbl. Both types of coordination isomers of the aryl-cobalamins promise to be useful molecular tools in biomedical and biological studies.


Asunto(s)
Compuestos de Bifenilo/química , Cobalto/química , Compuestos Onio/química , Dicroismo Circular , Cristalografía por Rayos X , Conformación Molecular , Espectrofotometría , Estereoisomerismo , Vitamina B 12/síntesis química , Vitamina B 12/química
13.
Angew Chem Int Ed Engl ; 56(26): 7387-7392, 2017 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-28544088

RESUMEN

B12 antivitamins are important and robust tools for investigating the biological roles of vitamin B12 . Here, the potential antivitamin B12 2,4-difluorophenylethynylcobalamin (F2PhEtyCbl) was prepared, and its 3D structure was studied in solution and in the crystal. Chemically inert F2PhEtyCbl resisted thermolysis of its Co-C bond at 100 °C, was stable in bright daylight, and also remained intact upon prolonged storage in aqueous solution at room temperature. It binds to the human B12 -processing enzyme CblC with high affinity (KD =130 nm) in the presence of the cosubstrate glutathione (GSH). F2PhEtyCbl withstood tailoring by CblC, and it also stabilized the ternary complex with GSH. The crystal structure of this inactivated assembly provides first insight into the binding interactions between an antivitamin B12 and CblC, as well as into the organization of GSH and a base-off cobalamin in the active site of this enzyme.


Asunto(s)
Glutatión/química , Vitamina B 12/antagonistas & inhibidores , Dominio Catalítico , Cristalografía por Rayos X , Flúor/química , Humanos , Hidrólisis , Cinética , Modelos Moleculares , Estructura Molecular , Análisis Espectral/métodos , Especificidad por Sustrato , Temperatura , Vitamina B 12/química , Vitamina B 12/farmacología
15.
J Biol Chem ; 290(18): 11393-402, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25809485

RESUMEN

Human CblC catalyzes the elimination of the upper axial ligand in cobalamin or B12 derivatives entering the cell from circulation. This processing step is critical for assimilation of dietary cobalamin into the active cofactor forms that support the B12-dependent enzymes, methionine synthase and methylmalonyl-CoA mutase. Using a modified nitroreductase scaffold tailored to bind cobalamin and glutathione, CblC exhibits versatility in the mechanism by which it removes cyano versus alkyl ligands in cobalamin. In this study, we have characterized the effects of two pathogenic missense mutations at the same residue, R161G and R161Q, which are associated with early and late onset of the CblC disorder, respectively. We find that the R161Q and R161G CblC mutants display lower protein stability and decreased dealkylation but not decyanation activity, suggesting that cyanocobalamin might be therapeutically useful for patients carrying mutations at Arg-161. The mutant proteins also exhibit impaired glutathione binding. In the presence of physiologically relevant glutathione concentrations, stabilization of the cob(II)alamin derivative is observed, which occurs at the expense of increased oxidation of glutathione. Futile redox cycling, which is suppressed in wild-type human CblC, explains the reported increase in oxidative stress levels associated with the CblC disorder.


Asunto(s)
Biocatálisis , Errores Innatos del Metabolismo/genética , Mutación Missense , Proteínas Proto-Oncogénicas c-cbl/genética , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Vitamina B 12/metabolismo , Alquilación , Arginina/metabolismo , Glutatión/farmacología , Humanos , Modelos Moleculares , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Nitrilos/metabolismo , Oxidación-Reducción/efectos de los fármacos , Estabilidad Proteica , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-cbl/química , Especies Reactivas de Oxígeno/metabolismo , Vitamina B 12/análogos & derivados , Vitamina B 12/química
16.
J Am Chem Soc ; 138(43): 14250-14256, 2016 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-27797190

RESUMEN

Cobalamins are of widespread importance in biology. Both of the cofactors essential for human metabolism, the organocobalamins coenzyme B12 and methylcobalamin, are highly photolabile, as are other alkylcobalamins. The alkynylcobalamin phenylethynylcobalamin (PhEtyCbl) and the arylcobalamin 4-ethylphenylcobalamin (EtPhCbl) with "atypical" Co-C-bonds to unsaturated carbons, were recently designed as metabolically inert cobalamins, classified as "antivitamins B12". The further development of an ideal light-activated or "conditional" antivitamin B12 would require it to be readily converted by light into an active B12 vitamin form. Very photolabile "antivitamins B12" would also represent particularly useful scaffolds for therapeutic light-activated reagents. Here, the photoactive arylcobalamin EtPhCbl and the remarkably photostable alkynylcobalamin PhEtyCbl are examined using femtosecond to picosecond UV-visible transient absorption spectroscopy. PhEtyCbl undergoes internal conversion to the ground state with near unit quantum yield on a time scale < 100 ps and an activation energy of 12.6 ± 1.4 kJ/mol. The arylcobalamin EtPhCbl forms an excited state with a ca. 247 ps lifetime. This excited state branches between internal conversion to the ground state and formation of a long-lived base-off species with a quantum yield of ∼9%. Anaerobic steady state photolysis of "light-sensitive" EtPhCbl results in the formation of cob(II)alamin, but only with quantum yield <1%. Hence, our studies suggest that suitably modified arylcobalamins may be a rational basis for the design of photoresponsive "antivitamins B12".


Asunto(s)
Absorción Fisicoquímica , Alquinos/química , Cobamidas/química , Diseño de Fármacos , Procesos Fotoquímicos , Cobamidas/metabolismo , Modelos Moleculares , Conformación Molecular
17.
Environ Microbiol ; 16(11): 3361-9, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24433392

RESUMEN

Corrinoid-dependent reductive dehalogenation is mediated by phylogenetically diverse anaerobic bacteria that either synthesize corrinoids de novo or are dependent on corrinoid salvaging from the environment. The tetrachloroethene (PCE) reductive dehalogenase (PceA) of the Gram-negative Epsilonproteobacterium Sulfurospirillum multivorans harbours a norpseudo-B12 as corrinoid cofactor. Norpseudo-B12 differs from coenzyme B12 in the nucleotide loop structure. Adenine instead of 5,6-dimethylbenzimidazole (DMB) serves as lower ligand base of the central cobalt ion, and the nucleotide loop of norpseudo-B12 lacks a methyl group at position 176. In this study, S. multivorans was grown anaerobically with PCE in the presence of DMB. At a DMB concentration of 25 µM, the adenine moiety in the nucleotide loop of norpseudo-B12 was quantitatively replaced by DMB. The formation of the DMB-containing nor-B12 severely affected PCE-dependent growth and the PceA activity. In DMB-treated cells processing of the cytoplasmic PceA precursor was impeded, a result pointing to retarded cofactor incorporation. PceA enriched from cells cultivated with DMB contained nor-B12 . Nor-B12 purified from cells grown in the presence of DMB mediated the abiotic reductive dehalogenation of trichloroacetate to dichloroacetate at a 25-fold lower rate in comparison with norpseudo-B12 , a fact underpinning the relevance of norpseudo-B12 as efficient catalyst for reductive dehalogenation in general.


Asunto(s)
Bencimidazoles/metabolismo , Epsilonproteobacteria/enzimología , Oxidorreductasas/metabolismo , Cobamidas/biosíntesis , Cobamidas/química , Corrinoides/biosíntesis , Epsilonproteobacteria/crecimiento & desarrollo
18.
Angew Chem Int Ed Engl ; 52(9): 2606-10, 2013 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-23404623

RESUMEN

Locked B(12): 4-Ethylphenylcobalamin, a novel organometallic arylcobalamin, has been synthesized in a radical reaction. This vitamin B(12) antimetabolite features a strong Co-C bond, and represents a "locked" form of vitamin B(12) . It may be used in animal studies to induce functional vitamin B(12) deficiency artificially to help clarify still controversial issues related to the pathophysiology of vitamin B(12) deficiency.


Asunto(s)
Compuestos Organometálicos/síntesis química , Vitamina B 12/análogos & derivados , Cristalografía por Rayos X , Modelos Moleculares , Compuestos Organometálicos/química , Vitamina B 12/síntesis química , Vitamina B 12/química
19.
Angew Chem Int Ed Engl ; 52(43): 11406-9, 2013 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-24030966

RESUMEN

Don't take this antivitamin! 2-Phenylethynylcobalamin was prepared in a newly developed radical reaction using cob(II)alamin and 1-iodo-2-phenylethyne. It has an exceptionally short organometallic bond and is a remarkably light-stable and heat-resistant organometallic cobalamin. It is bound well by two important proteins of the human B12 transport system and has properties that are as expected for a new type of an "antivitamin B12 ".


Asunto(s)
Vitamina B 12/química , Vitamina B 12/síntesis química , Alquinos , Catálisis , Humanos , Fotoquímica , Vitamina B 12/análogos & derivados
20.
Nat Commun ; 14(1): 4332, 2023 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-37468522

RESUMEN

G-proteins function as molecular switches to power cofactor translocation and confer fidelity in metal trafficking. The G-protein, MMAA, together with MMAB, an adenosyltransferase, orchestrate cofactor delivery and repair of B12-dependent human methylmalonyl-CoA mutase (MMUT). The mechanism by which the complex assembles and moves a >1300 Da cargo, or fails in disease, are poorly understood. Herein, we report the crystal structure of the human MMUT-MMAA nano-assembly, which reveals a dramatic 180° rotation of the B12 domain, exposing it to solvent. The complex, stabilized by MMAA wedging between two MMUT domains, leads to ordering of the switch I and III loops, revealing the molecular basis of mutase-dependent GTPase activation. The structure explains the biochemical penalties incurred by methylmalonic aciduria-causing mutations that reside at the MMAA-MMUT interfaces we identify here.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos , Transferasas Intramoleculares , Humanos , Metilmalonil-CoA Mutasa/genética , Metilmalonil-CoA Mutasa/metabolismo , Mutación , Errores Innatos del Metabolismo de los Aminoácidos/genética , Proteínas de Unión al GTP/genética , GTP Fosfohidrolasas/metabolismo , Transferasas Intramoleculares/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda