Malate dehydrogenase

Last updated
Malate dehydrogenase
Malate dehydrogenase structure.png
Structure of the protein with attached cofactors
Identifiers
EC no. 1.1.1.37
CAS no. 9001-64-3
Databases
IntEnz IntEnz view
BRENDA BRENDA entry
ExPASy NiceZyme view
KEGG KEGG entry
MetaCyc metabolic pathway
PRIAM profile
PDB structures RCSB PDB PDBe PDBsum
Search
PMC articles
PubMed articles
NCBI proteins

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+).

Contents

Isozymes

Several isozymes of malate dehydrogenase exist. There are two main isoforms in eukaryotic cells. [1] One is found in the mitochondrial matrix, participating as a key enzyme in the citric acid cycle that catalyzes the oxidation of malate. The other is found in the cytoplasm, assisting the malate-aspartate shuttle with exchanging reducing equivalents so that malate can pass through the mitochondrial membrane to be transformed into oxaloacetate for further cellular processes. [2]

Humans and most other mammals express the following two malate dehydrogenases:

Protein families

3-D crystal structure of the mobile loop region in malate dehydrogenase in the closed and open conformation. The MDH closed conformation is shown in pink (indicated by the pink arrow) while the open conformation is shown in cyan (indicated by the cyan arrow). Malate dehydrogenase Mobile Loop Region.png
3-D crystal structure of the mobile loop region in malate dehydrogenase in the closed and open conformation. The MDH closed conformation is shown in pink (indicated by the pink arrow) while the open conformation is shown in cyan (indicated by the cyan arrow).

The malate dehydrogenase family contains L-lactate dehydrogenase and L-2-hydroxyisocaproate dehydrogenases. L-lactate dehydrogenases catalyzes the conversion of L-lactate to pyruvate, the last step in anaerobic glycolysis. The N-terminus is a Rossmann NAD-binding fold and the C-terminus is an unusual alpha+beta fold. [3] [4]

Evolution and structure

In most organisms, malate dehydrogenase (MDH) exists as a homodimeric molecule and is closely related to lactate dehydrogenase (LDH) in structure. It is a large protein molecule with subunits weighing between 30 and 35 kDa. [5] Based on the amino acid sequences, it seems that MDH has diverged into two main phylogenetic groups that closely resemble either mitochondrial isozymes or cytoplasmic/chloroplast isozymes. [6] Because the sequence identity of malate dehydrogenase in the mitochondria is more closely related to its prokaryotic ancestors in comparison to the cytoplasmic isozyme, the theory that mitochondria and chloroplasts were developed through endosymbiosis is plausible. [7] The amino acid sequences of archaeal MDH are more similar to that of LDH than that of MDH of other organisms. This indicates that there is a possible evolutionary linkage between lactate dehydrogenase and malate dehydrogenase. [8]

Each subunit of the malate dehydrogenase dimer has two distinct domains that vary in structure and functionality. A parallel β-sheet structure makes up the NAD+ binding domain, while four β-sheets and one α-helix comprise the central NAD+ binding site. The subunits are held together through extensive hydrogen-bonding and hydrophobic interactions. [9]

Malate dehydrogenase has also been shown to have a mobile loop region that plays a crucial role in the enzyme's catalytic activity. Studies have shown that conformational change of this loop region from the open conformation to the closed conformation after binding of substrate enhances MDH catalysis through shielding of substrate and catalytic amino acids from solvent. Studies have also indicated that this loop region is highly conserved in malate dehydrogenase. [6]

Mechanism

Active site of malate dehydrogenase Malate dehydrogenase active site.svg
Active site of malate dehydrogenase

The active site of malate dehydrogenase is a hydrophobic cavity within the protein complex that has specific binding sites for the substrate and its coenzyme, NAD+. In its active state, MDH undergoes a conformational change that encloses the substrate to minimize solvent exposure and to position key residues in closer proximity to the substrate. [6] The three residues in particular that comprise a catalytic triad are histidine (His-195), aspartate (Asp-168), both of which work together as a proton transfer system, and arginines (Arg-102, Arg-109, Arg-171), which secure the substrate. [10]

Mechanistically, malate dehydrogenase catalyzes the oxidation of the hydroxyl group of malate by utilizing NAD+ as an electron acceptor. This oxidation step results in the elimination of a proton and a hydride ion from the substrate. NAD+ receives the hydride ion (specifically, the hydride ion is transferred to the nicotinamide ring of the NAD+) and becomes reduced to NADH while concomitantly, the His-195 residue on the enzyme accepts the proton. [11] The positively charged His-195 residue, which is involved in base catalysis of the substrate, is stabilized by the adjacent, negatively charged Asp-168 residue. This electrostatic stabilization helps facilitate the transfer of the proton. [1] Arg-102, Arg-109, and Arg-171 (which are protonated, and thus positively charged) participate in electrostatic catalysis and help to bind the negatively charged carboxylates on the substrate. Additionally, the Arginine residues on the enzyme provide additional substrate specificity and binding through hydrogen bonding between the guanidinium side chain of the Arginine amino acid residues and the carboxylates of the substrate. [12]

Studies have also identified a mobile loop in malate dehydrogenase that participates in the catalytic activity of the enzyme. The loop undergoes a conformational change to shield the substrate and catalytic amino acids from the solvent in response to the binding of the malate dehydrogenase:coenzyme complex to substrate. This flipping of the loop to the up position to cover the active site also promotes enhanced interaction of the catalytically important amino residues on the enzyme with the substrate. Additionally, the movement of the loop has been shown to correlate with the rate determining step of the enzyme. [13]

Function

Reaction

General reaction showing malate dehydrogenase catalyzed oxidation of malate through reduction of NAD . General Malate Dehydrogenase Catalyzed Reaction.jpg
General reaction showing malate dehydrogenase catalyzed oxidation of malate through reduction of NAD .

Malate dehydrogenases catalyzes the interconversion of malate to oxaloacetate. In the citric acid cycle, malate dehydrogenase is responsible for catalyzing the regeneration of oxaloacetate This reaction occurs through the oxidation of hydroxyl group on malate and reduction of NAD+. The mechanism of the transfer of the hydride ion to NAD+ is carried out in a similar mechanism seen in lactate dehydrogenase and alcohol dehydrogenase. The ΔG'° of malate dehydrogenase is +29.7 kJ/mol and the ΔG (in the cell) is 0 kJ/mol. [11]

Other pathways

Malate dehydrogenase is also involved in gluconeogenesis, the synthesis of glucose from smaller molecules. Pyruvate in the mitochondria is acted upon by pyruvate carboxylase to form oxaloacetate, a citric acid cycle intermediate. In order to get the oxaloacetate out of the mitochondria, malate dehydrogenase reduces it to malate, and it then traverses the inner mitochondrial membrane. Once in the cytosol, the malate is oxidized back to oxaloacetate by cytosolic malate dehydrogenase. Finally, phosphoenolpyruvate carboxykinase (PEPCK) converts oxaloacetate to phosphoenolpyruvate (PEP). [14]

Kinetics

Kinetic studies show that malate dehydrogenase enzymatic activity is ordered. The cofactor NAD+/NADH is bound to the enzyme before the substrate. [15] The Km value for malate, i.e., the concentration at which the enzyme activity is half-maximal, is 2 mM. The Kcat value is 259.2 s−1. [16]

Effect of pH on catalytic activity

Additionally, pH levels control specificity of substrate binding by malate dehydrogenase due to proton transfer in the catalytic mechanism. [17] A histidine moiety with a pK value of 7.5 has been suggested to play a role in the pH-dependency of the enzyme. Studies have indicated that the binding of the enol form oxaloacetate with the malate dehydrogenase:NADH complex forms much more rapidly at higher pH values. [12] Additionally, L-malate binding to malate dehydrogenase is promoted at alkaline conditions. Consequently, the non-protonated form malate dehydrogenase binds preferentially to L-malate and the enol form of oxaloacetate. In contrast, D-malate, hydroxymalonate, and the keto form of oxaloacetate have been found to bind exclusively to the protonated form of the enzyme. Specifically, when the histidine is protonated, the His residue can form a hydrogen bond with the substrate's carbonyl oxygen, which shifts electron density away from the oxygen and makes it more susceptible to nucleophilic attack by hydride. This promotes the binding of malate dehydrogenase to these substrates. As a result, at lower pH values malate dehydrogenase binds preferentially to D-malate, hydroxymalonate, and keto-oxaloacetate. [18]

Allosteric regulation

Because malate dehydrogenase is closely tied to the citric acid cycle, studies have proposed and experimentally demonstrated that citrate is an allosteric regulator of malate dehydrogenase depending on the concentrations of L-malate and NAD+. This may be due to deviations observed in the kinetic behavior of malate dehydrogenase at high oxaloacetate and L-malate concentrations. Experiments have shown that Citrate can both allosterically activate and inhibit the enzymatic activity of malate dehydrogenase. Citrate has been shown to inhibit the oxidation of L-malate when there are low levels of L-malate and NAD+. However, in the presence of high levels of malate and NAD+, citrate can stimulate the production of oxaloacetate. Although malate dehydrogenase is typically considered a reversible enzyme, it is believed that there is an allosteric regulatory site on the enzyme where citrate can bind to and drive the reaction equilibrium in either direction. [19]

Glutamate has also been shown to inhibit malate dehydrogenase activity. Furthermore, it has been shown that alpha ketoglutarate dehydrogenase can interact with mitochondrial aspartate aminotransferase to form a complex, which can then bind to malate dehydrogenase, forming a ternary complex that reverses inhibitory action on malate dehydrogenase enzymatic activity by glutamate. Additionally, the formation of this complex enables glutamate to react with aminotransferase without interfering activity of malate dehydrogenase. The formation of this ternary complex also facilitates the release of oxaloacetate from malate dehydrogenase to aminotransferase. Kinetically, the binding of malate dehydrogenase to the binary complex of alpha ketoglutarate dehydrogenase and aminotrannferase has been shown to increase reaction rate of malate dehydrogenase because the Km of malate dehydrogenase is decreased when it is bound as part of this complex. [20]

Interactive pathway map

Click on genes, proteins and metabolites below to link to respective articles. [§ 1]

[[File:
WP534.png go to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to article
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
WP534.png go to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to articlego to article
|alt=Glycolysis and Gluconeogenesis edit]]
Glycolysis and Gluconeogenesis edit
  1. The interactive pathway map can be edited at WikiPathways: "GlycolysisGluconeogenesis_WP534".

Related Research Articles

<span class="mw-page-title-main">Adenosine triphosphate</span> Energy-carrying molecule in living cells

Adenosine triphosphate (ATP) is an organic compound that provides energy to drive and support many processes in living cells, such as muscle contraction, nerve impulse propagation, condensate dissolution, and chemical synthesis. Found in all known forms of life, ATP is often referred to as the "molecular unit of currency" of intracellular energy transfer. When consumed in metabolic processes, it converts either to adenosine diphosphate (ADP) or to adenosine monophosphate (AMP). Other processes regenerate ATP. It is also a precursor to DNA and RNA, and is used as a coenzyme. A human adult processes around 50 kg of ATP daily.

<span class="mw-page-title-main">Citric acid cycle</span> Chemical reactions to release energy in cells

The citric acid cycle —also known as the Krebs cycle, Szent-Györgyi-Krebs cycle or the TCA cycle (tricarboxylic acid cycle)—is a series of chemical reactions to release stored energy through the oxidation of acetyl-CoA derived from carbohydrates, fats, and proteins. The Krebs cycle is used by organisms that respire (as opposed to organisms that ferment) to generate energy, either by anaerobic respiration or aerobic respiration. In addition, the cycle provides precursors of certain amino acids, as well as the reducing agent NADH, that are used in numerous other reactions. Its central importance to many biochemical pathways suggests that it was one of the earliest components of metabolism. Even though it is branded as a 'cycle', it is not necessary for metabolites to follow only one specific route; at least three alternative segments of the citric acid cycle have been recognized.

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

Acetyl-CoA is a molecule that participates in many biochemical reactions in protein, carbohydrate and lipid metabolism. Its main function is to deliver the acetyl group to the citric acid cycle to be oxidized for energy production. Coenzyme A consists of a β-mercaptoethylamine group linked to the vitamin pantothenic acid (B5) through an amide linkage and 3'-phosphorylated ADP. The acetyl group of acetyl-CoA is linked to the sulfhydryl substituent of the β-mercaptoethylamine group. This thioester linkage is a "high energy" bond, which is particularly reactive. Hydrolysis of the thioester bond is exergonic (−31.5 kJ/mol).

Gluconeogenesis (GNG) is a metabolic pathway that results in the generation of glucose from certain non-carbohydrate carbon substrates. It is a ubiquitous process, present in plants, animals, fungi, bacteria, and other microorganisms. In vertebrates, gluconeogenesis occurs mainly in the liver and, to a lesser extent, in the cortex of the kidneys. It is one of two primary mechanisms – the other being degradation of glycogen (glycogenolysis) – used by humans and many other animals to maintain blood sugar levels, avoiding low levels (hypoglycemia). In ruminants, because dietary carbohydrates tend to be metabolized by rumen organisms, gluconeogenesis occurs regardless of fasting, low-carbohydrate diets, exercise, etc. In many other animals, the process occurs during periods of fasting, starvation, low-carbohydrate diets, or intense exercise.

<span class="mw-page-title-main">Pyruvate dehydrogenase complex</span> Three-enzyme complex responsible for pyruvate decarboxylation

Pyruvate dehydrogenase complex (PDC) is a complex of three enzymes that converts pyruvate into acetyl-CoA by a process called pyruvate decarboxylation. Acetyl-CoA may then be used in the citric acid cycle to carry out cellular respiration, and this complex links the glycolysis metabolic pathway to the citric acid cycle. Pyruvate decarboxylation is also known as the "pyruvate dehydrogenase reaction" because it also involves the oxidation of pyruvate.

<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">Oxaloacetic acid</span> Organic compound

Oxaloacetic acid (also known as oxalacetic acid or OAA) is a crystalline organic compound with the chemical formula HO2CC(O)CH2CO2H. Oxaloacetic acid, in the form of its conjugate base oxaloacetate, is a metabolic intermediate in many processes that occur in animals. It takes part in gluconeogenesis, the urea cycle, the glyoxylate cycle, amino acid synthesis, fatty acid synthesis and the citric acid cycle.

<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">Mitochondrial matrix</span> Space within the inner membrane of the mitochondrion

In the mitochondrion, the matrix is the space within the inner membrane. The word "matrix" stems from the fact that this space is viscous, compared to the relatively aqueous cytoplasm. The mitochondrial matrix contains the mitochondrial DNA, ribosomes, soluble enzymes, small organic molecules, nucleotide cofactors, and inorganic ions.[1] The enzymes in the matrix facilitate reactions responsible for the production of ATP, such as the citric acid cycle, oxidative phosphorylation, oxidation of pyruvate, and the beta oxidation of fatty acids.

<span class="mw-page-title-main">Pyruvate carboxylase</span> Enzyme

Pyruvate carboxylase (PC) encoded by the gene PC is an enzyme of the ligase class that catalyzes the physiologically irreversible carboxylation of pyruvate to form oxaloacetate (OAA).

<span class="mw-page-title-main">Citrate synthase</span> Enzyme found in humans

The enzyme citrate synthase E.C. 2.3.3.1 ] exists in nearly all living cells and stands as a pace-making enzyme in the first step of the citric acid cycle. Citrate synthase is localized within eukaryotic cells in the mitochondrial matrix, but is encoded by nuclear DNA rather than mitochondrial. It is synthesized using cytoplasmic ribosomes, then transported into the mitochondrial matrix.

<span class="mw-page-title-main">Fumarase</span> Type of enzyme

Fumarase is an enzyme that catalyzes the reversible hydration/dehydration of fumarate to malate. Fumarase comes in two forms: mitochondrial and cytosolic. The mitochondrial isoenzyme is involved in the Krebs cycle and the cytosolic isoenzyme is involved in the metabolism of amino acids and fumarate. Subcellular localization is established by the presence of a signal sequence on the amino terminus in the mitochondrial form, while subcellular localization in the cytosolic form is established by the absence of the signal sequence found in the mitochondrial variety.

<span class="mw-page-title-main">Phosphoenolpyruvate carboxylase</span> Class of enzymes

Phosphoenolpyruvate carboxylase (also known as PEP carboxylase, PEPCase, or PEPC; EC 4.1.1.31, PDB ID: 3ZGE) is an enzyme in the family of carboxy-lyases found in plants and some bacteria that catalyzes the addition of bicarbonate (HCO3) to phosphoenolpyruvate (PEP) to form the four-carbon compound oxaloacetate and inorganic phosphate:

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

Pyruvate dehydrogenase is an enzyme that catalyzes the reaction of pyruvate and a lipoamide to give the acetylated dihydrolipoamide and carbon dioxide. The conversion requires the coenzyme thiamine pyrophosphate.

Malate dehydrogenase (oxaloacetate-decarboxylating) (NADP<sup>+</sup>) Enzyme

Malate dehydrogenase (oxaloacetate-decarboxylating) (NADP+) (EC 1.1.1.40) or NADP-malic enzyme (NADP-ME) is an enzyme that catalyzes the chemical reaction in the presence of a bivalent metal ion:

Octopine dehydrogenase (N2-(D-1-carboxyethyl)-L-arginine:NAD+ oxidoreductase, OcDH, ODH) is a dehydrogenase enzyme in the opine dehydrogenase family that helps maintain redox balance under anaerobic conditions. It is found largely in aquatic invertebrates, especially mollusks, sipunculids, and coelenterates, and plays a role analogous to lactate dehydrogenase (found largely in vertebrates) . In the presence of NADH, OcDH catalyzes the reductive condensation of an α-keto acid with an amino acid to form N-carboxyalkyl-amino acids (opines). The purpose of this reaction is to reoxidize glycolytically formed NADH to NAD+, replenishing this important reductant used in glycolysis and allowing for the continued production of ATP in the absence of oxygen.

Glutaminolysis (glutamine + -lysis) is a series of biochemical reactions by which the amino acid glutamine is lysed to glutamate, aspartate, CO2, pyruvate, lactate, alanine and citrate.

<span class="mw-page-title-main">Inosine-5′-monophosphate dehydrogenase</span> Class of enzymes

Inosine-5′-monophosphate dehydrogenase (IMPDH) is a purine biosynthetic enzyme that catalyzes the nicotinamide adenine dinucleotide (NAD+)-dependent oxidation of inosine monophosphate (IMP) to xanthosine monophosphate (XMP), the first committed and rate-limiting step towards the de novo biosynthesis of guanine nucleotides from IMP. IMPDH is a regulator of the intracellular guanine nucleotide pool, and is therefore important for DNA and RNA synthesis, signal transduction, energy transfer, glycoprotein synthesis, as well as other process that are involved in cellular proliferation.

<span class="mw-page-title-main">Malate dehydrogenase 2</span> Enzyme that oxidizes malate to oxaloacetate in Krebs cycle

Malate dehydrogenase, mitochondrial also known as malate dehydrogenase 2 is an enzyme that in humans is encoded by the MDH2 gene.

<span class="mw-page-title-main">Citrate–malate shuttle</span>

The citrate–malate shuttle is a series of chemical reactions – commonly referred to as a biochemical cycle or system – that transports acetyl-CoA in the mitochondrial matrix across the inner and outer mitochondrial membrane for fatty acid synthesis. Mitochondria is enclosed in a double membrane. As the inner mitochondrial membrane is impermeable to acetyl-CoA, the shuttle system is essential to fatty acid synthesis in the cytosol. It plays an important role in the generation of lipids in the liver.

References

  1. 1 2 Minárik P, Tomásková N, Kollárová M, Antalík M (September 2002). "Malate dehydrogenases--structure and function". General Physiology and Biophysics. 21 (3): 257–65. PMID   12537350.
  2. Musrati RA, Kollárová M, Mernik N, Mikulásová D (September 1998). "Malate dehydrogenase: distribution, function and properties". General Physiology and Biophysics. 17 (3): 193–210. PMID   9834842.
  3. Chapman AD, Cortés A, Dafforn TR, Clarke AR, Brady RL (January 1999). "Structural basis of substrate specificity in malate dehydrogenases: crystal structure of a ternary complex of porcine cytoplasmic malate dehydrogenase, alpha-ketomalonate and tetrahydoNAD". Journal of Molecular Biology. 285 (2): 703–12. doi: 10.1006/jmbi.1998.2357 . PMID   10075524.
  4. Madern D (June 2002). "Molecular evolution within the L-malate and L-lactate dehydrogenase super-family". Journal of Molecular Evolution. 54 (6): 825–40. Bibcode:2002JMolE..54..825M. doi:10.1007/s00239-001-0088-8. PMID   12029364. S2CID   469660.
  5. Banaszak LJ, Bradshaw RA (1975). "Malate dehydrogenase". In Boyer PD (ed.). The Enzymes. Vol. 11 (3rd ed.). New York: Academic Press. pp. 369–396.
  6. 1 2 3 Goward CR, Nicholls DJ (October 1994). "Malate dehydrogenase: a model for structure, evolution, and catalysis". Protein Science. 3 (10): 1883–8. doi:10.1002/pro.5560031027. PMC   2142602 . PMID   7849603.
  7. McAlister-Henn L (May 1988). "Evolutionary relationships among the malate dehydrogenases". Trends in Biochemical Sciences. 13 (5): 178–81. doi:10.1016/0968-0004(88)90146-6. PMID   3076279.
  8. Cendrin F, Chroboczek J, Zaccai G, Eisenberg H, Mevarech M (April 1993). "Cloning, sequencing, and expression in Escherichia coli of the gene coding for malate dehydrogenase of the extremely halophilic archaebacterium Haloarcula marismortui". Biochemistry. 32 (16): 4308–13. doi:10.1021/bi00067a020. PMID   8476859.
  9. Hall MD, Levitt DG, Banaszak LJ (August 1992). "Crystal structure of Escherichia coli malate dehydrogenase. A complex of the apoenzyme and citrate at 1.87 A resolution". Journal of Molecular Biology. 226 (3): 867–82. doi:10.1016/0022-2836(92)90637-Y. PMID   1507230.
  10. Lamzin VS, Dauter Z, Wilson KS (May 1994). "Dehydrogenation through the looking-glass". Nature Structural Biology. 1 (5): 281–2. doi:10.1038/nsb0594-281. PMID   7664032. S2CID   26167967.
  11. 1 2 Voet D, Voet JG, Pratt CW (2015). Fundamentals of Biochemistry: Life at the Molecular Level (4th ed.). Hoboken, NJ: Wiley. pp. 574–5. ISBN   978-0-470-54784-7.
  12. 1 2 Bernstein LH, Everse J (December 1978). "Studies on the mechanism of the malate dehydrogenase reaction" (PDF). The Journal of Biological Chemistry. 253 (24): 8702–7. doi: 10.1016/S0021-9258(17)34234-5 . PMID   31361.
  13. Waldman AD, Hart KW, Clarke AR, Wigley DB, Barstow DA, Atkinson T, Chia WN, Holbrook JJ (January 1988). "The use of genetically engineered tryptophan to identify the movement of a domain of B. stearothermophilus lactate dehydrogenase with the process which limits the steady-state turnover of the enzyme". Biochemical and Biophysical Research Communications. 150 (2): 752–9. doi:10.1016/0006-291X(88)90455-X. PMID   3422557.
  14. Hung GC, Brown CR, Wolfe AB, Liu J, Chiang HL (November 2004). "Degradation of the gluconeogenic enzymes fructose-1,6-bisphosphatase and malate dehydrogenase is mediated by distinct proteolytic pathways and signaling events". The Journal of Biological Chemistry. 279 (47): 49138–50. doi: 10.1074/jbc.M404544200 . PMID   15358789.
  15. Shows TB, Chapman VM, Ruddle FH (December 1970). "Mitochondrial malate dehydrogenase and malic enzyme: Mendelian inherited electrophoretic variants in the mouse". Biochemical Genetics. 4 (6): 707–18. doi:10.1007/BF00486384. PMID   5496232. S2CID   35435579.
  16. Wood DC, Jurgensen SR, Geesin JC, Harrison JH (March 1981). "Subunit interactions in mitochondrial malate dehydrogenase. Kinetics and mechanism of reassociation". The Journal of Biological Chemistry. 256 (5): 2377–82. doi: 10.1016/S0021-9258(19)69790-5 . PMID   7462244.
  17. Dasika SK, Vinnakota KC, Beard DA (January 2015). "Determination of the catalytic mechanism for mitochondrial malate dehydrogenase". Biophysical Journal. 108 (2): 408–19. doi:10.1016/j.bpj.2014.11.3467. PMC   4302198 . PMID   25606688.
  18. Lodola A, Shore JD, Parker DM, Holbrook J (December 1978). "Malate dehydrogenase of the cytosol. A kinetic investigation of the reaction mechanism and a comparison with lactate dehydrogenase". The Biochemical Journal. 175 (3): 987–98. doi:10.1042/bj1750987. PMC   1186162 . PMID   217361.
  19. Gelpí JL, Dordal A, Montserrat J, Mazo A, Cortés A (April 1992). "Kinetic studies of the regulation of mitochondrial malate dehydrogenase by citrate". The Biochemical Journal. 283 ( Pt 1) (Pt 1): 289–97. doi:10.1042/bj2830289. PMC   1131027 . PMID   1567375.
  20. Fahien LA, Kmiotek EH, MacDonald MJ, Fibich B, Mandic M (August 1988). "Regulation of malate dehydrogenase activity by glutamate, citrate, alpha-ketoglutarate, and multienzyme interaction" (PDF). The Journal of Biological Chemistry. 263 (22): 10687–97. doi: 10.1016/S0021-9258(18)38026-8 . PMID   2899080.

Further reading