Alanine racemase

Last updated
alanine racemase
3hur.jpg
Alanine racemase homotetramer, Oenococcus oeni
Identifiers
EC no. 5.1.1.1
CAS no. 9024-06-0
Databases
IntEnz IntEnz view
BRENDA BRENDA entry
ExPASy NiceZyme view
KEGG KEGG entry
MetaCyc metabolic pathway
PRIAM profile
PDB structures RCSB PDB PDBe PDBsum
Gene Ontology AmiGO / QuickGO
Search
PMC articles
PubMed articles
NCBI proteins
Ala_racemase_N
PDB 1rcq EBI.jpg
the 1.45Å a crystal structure of alanine racemase from a pathogenic bacterium, pseudomonas aeruginosa, contains both internal and external aldimine forms
Identifiers
SymbolAla_racemase_N
Pfam PF01168
Pfam clan CL0036
InterPro IPR001608
PROSITE PDOC00332
SCOP2 1sft / SCOPe / SUPFAM
Available protein structures:
Pfam   structures / ECOD  
PDB RCSB PDB; PDBe; PDBj
PDBsum structure summary
Ala_racemase_C
Identifiers
SymbolAla_racemase_C
Pfam PF00842
InterPro IPR011079
PROSITE PDOC00332
SCOP2 1sft / SCOPe / SUPFAM
Available protein structures:
Pfam   structures / ECOD  
PDB RCSB PDB; PDBe; PDBj
PDBsum structure summary

In enzymology, an alanine racemase (EC 5.1.1.1) is an enzyme that catalyzes the chemical reaction

Contents

L-alanine D-alanine

Hence, this enzyme has one substrate, L-alanine, and one product, D-alanine.

This enzyme belongs to the family of isomerases, specifically those racemases and epimerases acting on amino acids and derivatives. The systematic name of this enzyme class is alanine racemase. This enzyme is also called L-alanine racemase. This enzyme participates in alanine and aspartate metabolism and D-alanine metabolism. It employs one cofactor, pyridoxal phosphate. At least two compounds, 3-Fluoro-D-alanine and D-Cycloserine are known to inhibit this enzyme.

The D-alanine produced by alanine racemase is used for peptidoglycan biosynthesis. Peptidoglycan is found in the cell walls of all bacteria, including many which are harmful to humans. The enzyme is absent in higher eukaryotes but found everywhere in prokaryotes, making alanine racemase a great target for antimicrobial drug development. [1] Alanine racemase can be found in some invertebrates. [2]

Bacteria can have one (alr gene) or two alanine racemase genes. Bacterial species with two genes for alanine racemase have one that is continually expressed and one that is inducible, which makes it difficult to target both genes for drug studies. However, knockout studies have shown that without the alr gene being expressed, the bacteria would need an external source of D-alanine in order to survive. Therefore, the alr gene is a feasible target for antimicrobial drugs. [1]

Structural studies

To catalyze the interconversion of D and L alanine, Alanine racemase must position residues capable of exchanging protons on either side of the alpha carbon of alanine. Structural studies of enzyme-inhibitor complexes suggest that Tyrosine 265 and Lysine 39 are these residues. The alpha-proton of the L-enantiomer is oriented toward Tyr265 and the alpha proton of the D-enantiomer is oriented toward Lys39 (Figure 1).

Figure 1. Active site of Alanine Racemase. Tyrosine-265 and Lysine-39 are displayed with their distances to the alpha-carbon of alanine, which is colored green and attached to PLP. Alanine Racemase Active Site.PNG
Figure 1. Active site of Alanine Racemase. Tyrosine-265 and Lysine-39 are displayed with their distances to the alpha-carbon of alanine, which is colored green and attached to PLP.

The distance between the enzyme residues and the enantiomers is 3.5 Å and 3.6 Å respectively. [3] Structural studies of enzyme complexes with a synthetic L-alanine analog, a tight binding inhibitor [4] and propionate [5] further validate that Tyr265 and Lys39 are catalytic bases for the reaction,. [4] [6]

The PLP-L-Ala and PLP-D-Ala complexes are almost superimposability. [3] The regions that do not overlap are the arms connected the pyridine ring of PLP and the alpha carbon of alanine. An interaction between both the phosphate oxygen and pyridine nitrogen atoms to the 5’phosphopyridoxyl region of PLP-Ala probably creates tight binding to the enzyme. [3]

Figure 2. Surface Diagram of Alanine Racemase. The two monomers are colored in blue and green. The two reaction sites are colored in red. Alanine Racemase.PNG
Figure 2. Surface Diagram of Alanine Racemase. The two monomers are colored in blue and green. The two reaction sites are colored in red.

The structure of alanine racemase from Bacillus stearothermophilus (Geobacillus stearothermophilus) was determined by X-ray crystallography to a resolution of 1.9 A. [6] The alanine racemase monomer is composed of two domains, an eight-stranded alpha/beta barrel at the N terminus, and a C-terminal domain essentially composed of beta-strand. A model of the two domain structure is shown in Figure 2. The N-terminal domain is also found in the PROSC (proline synthetase co-transcribed bacterial homolog) family of proteins, which are not known to have alanine racemase activity. The pyridoxal 5'-phosphate (PLP) cofactor lies in and above the mouth of the alpha/beta barrel and is covalently linked via an aldimine linkage to a lysine residue, which is at the C terminus of the first beta-strand of the alpha/beta barrel.

Proposed Mechanism

Reaction mechanisms are difficult to fully prove by experiment. The traditional mechanism attributed to an alanine racemase reaction is that of a two-base mechanism with a PLP-stabilized carbanion intermediate. PLP is used as an electron sink stabilize the negative charge resulting from the deprotonation of the alpha carbon. The two based mechanism favors reaction specificity compared to a one base mechanism. The second catalytic residue is pre-positioned to donate a proton quickly after a carbanionic intermediate is formed and thus reduces the chance of alternative reactions occurring. There are two potential conflicts with this traditional mechanism, as identified by Watanabe et al. First, Arg219 forms a hydrogen bond with pyridine nitrogen of PLP. [6] The arginine group has a pKa of about 12.6 and is therefore unlikely to protonate the pyridine. Normally in PLP reactions an acidic amino acid residue such as a carboxylic acid group, with a pKa of about 5, protonates the pyridine ring. [7] The protonation of the pyridine nitrogen allows the nitrogen to accept additional negative charge. Therefore, due to the Arg219, the PLP-stabilized carbanion intermediate is less likely to form. Another problem identified was the need for another basic residue to return Lys39 and Tyr265 back to their protonated and unprotonated forms for L-alanine and vice versa for D-alanine. Watanabe et al. found no amino acid residues or water molecules, other than the carboxylate group of PLP-Ala, to be close enough (within 4.5A) to protonate or deprotonate Lys or Tyr. This is shown in Figure 3. [3]

Figure 3. Schematic diagram of the distance between Lys39, Tyr 265, and PLP-L-Ala in the active site. All interactions shown are under 4.5 and therefore are capable of hydrogen bonding. Adapted from Watanabe et al. Distance Between Lys39, Tyr 265, and PLP-L-Ala in the Active Site.PNG
Figure 3. Schematic diagram of the distance between Lys39, Tyr 265, and PLP-L-Ala in the active site. All interactions shown are under 4.5 and therefore are capable of hydrogen bonding. Adapted from Watanabe et al.

Based on the crystal structures of N-(5’-phosphopyridoxyl) L- alanine (PKP-L-Ala ( and N-(5’-phosphopyridoxyl) D-alanine (PLP-D-Ala)

Watanabe et al. proposed an alternative mechanism in 2002, as seen in the figure 4. In this mechanism the carboxylate oxygen atoms of PLP-Ala directly participates in catalysis by mediating proton transfer between Lys39 and Tyr265. The crystallization structure identified that the carboxylate oxygen of PLP-L-Ala to the OH of Tyr265 was only 3.6A and the carboxylate oxygen of PLP-L-Ala to the nitrogen of Lys39 was only 3.5A. Therefore, both were close enough to cause a reaction.

Figure 4: The mechanism is based on x-ray structures of PLP-L-Ala and PLP-D-Ala and molecular orbital calculations done by Watanabe (4). Alanine Racemase Mechanism.PNG
Figure 4: The mechanism is based on x-ray structures of PLP-L-Ala and PLP-D-Ala and molecular orbital calculations done by Watanabe (4).

This mechanism is supported by mutations of Arg219. Mutations changing Arg219 to a carboxylate result in a quinonoid intermediate being detected whereas none was detected with arginine. [8] The arginine intermediate has much more free energy, is more unstable, than the acidic residue mutants. [7] The destabilization of the intermediate promotes specificity of the reaction,. [8] [9]

Related Research Articles

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

Histidine (symbol His or H) is an essential amino acid that is used in the biosynthesis of proteins. It contains an α-amino group (which is in the protonated –NH3+ form under biological conditions), a carboxylic acid group (which is in the deprotonated –COO form under biological conditions), and an imidazole side chain (which is partially protonated), classifying it as a positively charged amino acid at physiological pH. Initially thought essential only for infants, it has now been shown in longer-term studies to be essential for adults also. It is encoded by the codons CAU and CAC.

<span class="mw-page-title-main">Aminolevulinic acid synthase</span> Class of enzymes

Aminolevulinic acid synthase (ALA synthase, ALAS, or delta-aminolevulinic acid synthase) is an enzyme (EC 2.3.1.37) that catalyzes the synthesis of δ-aminolevulinic acid (ALA) the first common precursor in the biosynthesis of all tetrapyrroles such as hemes, cobalamins and chlorophylls. The reaction is as follows:

<span class="mw-page-title-main">Pyridoxal phosphate</span> Active form of vitamin B6

Pyridoxal phosphate (PLP, pyridoxal 5'-phosphate, P5P), the active form of vitamin B6, is a coenzyme in a variety of enzymatic reactions. The International Union of Biochemistry and Molecular Biology has catalogued more than 140 PLP-dependent activities, corresponding to ~4% of all classified activities. The versatility of PLP arises from its ability to covalently bind the substrate, and then to act as an electrophilic catalyst, thereby stabilizing different types of carbanionic reaction intermediates.

<span class="mw-page-title-main">Aspartate transaminase</span> Enzyme involved in amino acid metabolism

Aspartate transaminase (AST) or aspartate aminotransferase, also known as AspAT/ASAT/AAT or (serum) glutamic oxaloacetic transaminase, is a pyridoxal phosphate (PLP)-dependent transaminase enzyme that was first described by Arthur Karmen and colleagues in 1954. AST catalyzes the reversible transfer of an α-amino group between aspartate and glutamate and, as such, is an important enzyme in amino acid metabolism. AST is found in the liver, heart, skeletal muscle, kidneys, brain, red blood cells and gall bladder. Serum AST level, serum ALT level, and their ratio are commonly measured clinically as biomarkers for liver health. The tests are part of blood panels.

<span class="mw-page-title-main">Malate dehydrogenase</span> Class of enzymes

Malate dehydrogenase (EC 1.1.1.37) (MDH) is an enzyme that reversibly catalyzes the oxidation of malate to oxaloacetate using the reduction of NAD+ to NADH. This reaction is part of many metabolic pathways, including the citric acid cycle. Other malate dehydrogenases, which have other EC numbers and catalyze other reactions oxidizing malate, have qualified names like malate dehydrogenase (NADP+).

<span class="mw-page-title-main">Isocitrate dehydrogenase</span> Class of enzymes

Isocitrate dehydrogenase (IDH) (EC 1.1.1.42) and (EC 1.1.1.41) is an enzyme that catalyzes the oxidative decarboxylation of isocitrate, producing alpha-ketoglutarate (α-ketoglutarate) and CO2. This is a two-step process, which involves oxidation of isocitrate (a secondary alcohol) to oxalosuccinate (a ketone), followed by the decarboxylation of the carboxyl group beta to the ketone, forming alpha-ketoglutarate. In humans, IDH exists in three isoforms: IDH3 catalyzes the third step of the citric acid cycle while converting NAD+ to NADH in the mitochondria. The isoforms IDH1 and IDH2 catalyze the same reaction outside the context of the citric acid cycle and use NADP+ as a cofactor instead of NAD+. They localize to the cytosol as well as the mitochondrion and peroxisome.

<span class="mw-page-title-main">Muconate lactonizing enzyme</span>

Muconate lactonizing enzymes are involved in the breakdown of lignin-derived aromatics, catechol and protocatechuate, to citric acid cycle intermediates as a part of the β-ketoadipate pathway in soil microbes. Some bacterial species are also capable of dehalogenating chloroaromatic compounds by the action of chloromuconate lactonizing enzymes. MLEs consist of several strands which have variable reaction favorable parts therefore the configuration of the strands affect its ability to accept protons. The bacterial MLEs belong to the enolase superfamily, several structures from which are known. MLEs have an identifying structure made up of two proteins and two Magnesium ions as well as various classes depending on whether it is bacterial or eukaryotic. The reaction mechanism that MLEs undergo are the reverse of beta-elimination in which the enolate alpha-carbon is protonated. MLEs can undergo mutations caused by a deletion of catB structural genes which can cause some bacteria to lose its functions such as the ability to grow. Additional mutations to MLEs can cause its structure and function to alter and could cause the conformation to change therefore making it an inactive enzyme that is unable to bind its substrate. There is another enzyme called Mandelate Racemase that is very similar to MLEs in the structural way as well as them both being a part of the enolase superfamily. They both have the same end product even though they undergo different chemical reactions in order to reach the end product.

<span class="mw-page-title-main">Carboxypeptidase</span>

A carboxypeptidase is a protease enzyme that hydrolyzes (cleaves) a peptide bond at the carboxy-terminal (C-terminal) end of a protein or peptide. This is in contrast to an aminopeptidases, which cleave peptide bonds at the N-terminus of proteins. Humans, animals, bacteria and plants contain several types of carboxypeptidases that have diverse functions ranging from catabolism to protein maturation. At least two mechanisms have been discussed.

<span class="mw-page-title-main">Serine hydroxymethyltransferase</span>

Serine hydroxymethyltransferase (SHMT) is a pyridoxal phosphate (PLP) (Vitamin B6) dependent enzyme (EC 2.1.2.1) which plays an important role in cellular one-carbon pathways by catalyzing the reversible, simultaneous conversions of L-serine to glycine and tetrahydrofolate (THF) to 5,10-Methylenetetrahydrofolate (5,10-CH2-THF). This reaction provides the largest part of the one-carbon units available to the cell.

<span class="mw-page-title-main">Methylmalonyl CoA epimerase</span>

Methylmalonyl CoA epimerase is an enzyme involved in fatty acid catabolism that is encoded in human by the "MCEE" gene located on chromosome 2. It is routinely and incorrectly labeled as "methylmalonyl-CoA racemase". It is not a racemase because the CoA moiety has 5 other stereocenters.

<span class="mw-page-title-main">Cystathionine gamma-lyase</span> Protein-coding gene in the species Homo sapiens

The enzyme cystathionine γ-lyase (EC 4.4.1.1, CTH or CSE; also cystathionase; systematic name L-cystathionine cysteine-lyase (deaminating; 2-oxobutanoate-forming)) breaks down cystathionine into cysteine, 2-oxobutanoate (α-ketobutyrate), and ammonia:

In enzymology, an aspartate racemase is an enzyme that catalyzes the following chemical reaction:

<span class="mw-page-title-main">D-lysine 5,6-aminomutase</span>

In enzymology, D-lysine 5,6-aminomutase is an enzyme that catalyzes the chemical reaction

<span class="mw-page-title-main">Cystathionine beta-lyase</span> Enzyme

Cystathionine beta-lyase, also commonly referred to as CBL or β-cystathionase, is an enzyme that primarily catalyzes the following α,β-elimination reaction

<span class="mw-page-title-main">Threonine ammonia-lyase</span>

Threonine ammonia-lyase (EC 4.3.1.19, systematic name L-threonine ammonia-lyase (2-oxobutanoate-forming), also commonly referred to as threonine deaminase or threonine dehydratase, is an enzyme responsible for catalyzing the conversion of L-threonine into α-ketobutyrate and ammonia:

<span class="mw-page-title-main">Arginine decarboxylase</span>

The enzyme Acid-Induced Arginine Decarboxylase (AdiA), also commonly referred to as arginine decarboxylase, catalyzes the conversion of L-arginine into agmatine and carbon dioxide. The process consumes a proton in the decarboxylation and employs a pyridoxal-5'-phosphate (PLP) cofactor, similar to other enzymes involved in amino acid metabolism, such as ornithine decarboxylase and glutamine decarboxylase. It is found in bacteria and virus, though most research has so far focused on forms of the enzyme in bacteria. During the AdiA catalyzed decarboxylation of arginine, the necessary proton is consumed from the cell cytoplasm which helps to prevent the over-accumulation of protons inside the cell and serves to increase the intracellular pH. Arginine decarboxylase is part of an enzymatic system in Escherichia coli, Salmonella Typhimurium, and methane-producing bacteria Methanococcus jannaschii that makes these organisms acid resistant and allows them to survive under highly acidic medium.

<span class="mw-page-title-main">Diaminopimelate decarboxylase</span>

The enzyme diaminopimelate decarboxylase (EC 4.1.1.20) catalyzes the cleavage of carbon-carbon bonds in meso 2,6 diaminoheptanedioate to produce CO2 and L-lysine, the essential amino acid. It employs the cofactor pyridoxal phosphate, also known as PLP, which participates in numerous enzymatic transamination, decarboxylation and deamination reactions.

<span class="mw-page-title-main">Serine C-palmitoyltransferase</span>

In enzymology, a serine C-palmitoyltransferase (EC 2.3.1.50) is an enzyme that catalyzes the chemical reaction:

In enzymology, a D-amino-acid transaminase is an enzyme that catalyzes the chemical reaction:

Glutamate 2,3-aminomutase is an enzyme that belongs to the radical s-adenosyl methionine (SAM) superfamily. Radical SAM enzymes facilitate the reductive cleavage of S-adenosylmethionine (SAM) through the use of radical chemistry and an iron-sulfur cluster. This enzyme family is implicated in the biosynthesis of DNA precursors, vitamin, cofactor, antibiotic and herbicides and in biodegradation pathways. In particular, glutamate 2,3 aminomutase is involved in the conversion of L-alpha-glutamate to L-beta-glutamate in Clostridium difficile. The generalized reaction is shown below:

References

  1. 1 2 Milligan Daniel L.; et al. (2007). "The Alanine Racemase of Mycobacterium smegmatis Is Essential for Growth in the Absence of D-Alanine". Journal of Bacteriology. 189 (22): 8381–8386. doi:10.1128/jb.01201-07. PMC   2168708 . PMID   17827284.
  2. Abe, H; Yoshikawa, N; Sarower, M. G.; Okada, S (2005). "Physiological function and metabolism of free D-alanine in aquatic animals". Biological & Pharmaceutical Bulletin. 28 (9): 1571–7. doi: 10.1248/bpb.28.1571 . PMID   16141518.
  3. 1 2 3 4 Watanabe, A., Yoshimura, T., Mikami, B., Hayashi, H., Kagamiyama, H., Esaki, N. (2002) Reaction Mechanism of Alanine Racemase from Bacillus stearothermophilus: X-ray crystallographic studies of the enzyme bound within -(5’-phosphopyridoxyl)alanine Journal of Biological Chemistry 277, 19166-19172.
  4. 1 2 Stamper, G. F., Morollo, A. A., and Ringe, D. (1998) Biochemistry 37, 10438 –10445
  5. Morollo, A. A., Petsko, G. A., and Ringe, D. (1999) Biochemistry 38, 3293–3301
  6. 1 2 3 Shaw, J. P., Petsko, G. A., and Ringe, D. (1997) Determination of the Structure of Alanine racemase from Bacillus stearothermophilus at 1.9-A Resolution Biochemistry 36, 1329–1342
  7. 1 2 Toney, Michael D. (2004) Reaction specificity in pyridoxal phosphate enzymes, Archives of Biochemistry and Biophysics 433, 279-287.
  8. 1 2 Sun S., Toney, M.D. (1998) Evidence for a Two-Base Mechanism Involving Tyrosine-265 from Arginine-219 Mutants of Alanine Racemase Biochemistry 38, 4058-4065
  9. Rubinstein, A., Major, D. T. (2010) Understanding Catalytic Specificity in Alanine Racemase from Quantum Mechanical Molecular Mechanical Simulations of the Arginine 210 Mutant Biochemistry 49, 3957-3963.

Further reading

This article incorporates text from the public domain Pfam and InterPro: IPR011079