2,4 Dienoyl-CoA reductase

Last updated
2,4-dienoyl CoA reductase 1, mitochondrial
1w6u.jpg
DECR1, homotetramer, Human
Identifiers
SymbolDECR1
Alt. symbolsDECR
NCBI gene 1666
HGNC 2753
OMIM 222745
PDB 1w6u
RefSeq NM_001359
UniProt Q16698
Other data
EC number 1.3.1.34
Locus Chr. 8 q21.3
Search for
Structures Swiss-model
Domains InterPro

2,4 Dienoyl-CoA reductase also known as DECR1 is an enzyme which in humans is encoded by the DECR1 gene which resides on chromosome 8. This enzyme catalyzes the following reactions [1] [2] [3]

Contents

Dienoyl-CoA reductase reaction cis-trans.svg

DECR1 participates in the beta oxidation and metabolism of polyunsaturated fatty enoyl-CoA esters. Specifically, it catalyzes the reduction of 2,4 dienoyl-CoA thioesters of varying length by NADPH cofactor to 3-trans-enoyl-CoA of equivalent length. Unlike the breakdown of saturated fat, cis and trans polyunsaturated fatty acid degradation requires three additional enzymes to generate a product compatible with the standard beta oxidation pathway. DECR is the second such enzyme (the others being enoyl CoA isomerase and dienoyl CoA isomerase) and is the rate limiting step in this auxiliary flow. DECR is capable of reducing both 2-trans,4-cis-dienoyl-CoA and 2-trans,4-trans-dienoyl-CoA thioesters [4] with equal efficiency. [5] This is unusual, since most enzymes are highly stereoselective or stereospecific. [6] There is no clear explanation for DECR's of lack of stereospecificity. [5]

Structure

Crystallization of DECR with 2,4 Hexadienoyl-CoA and NADPH (not shown). Key residues in the enzyme active site orient the substrate for hydride transfer through a network of hydrogen bonds. 2,4 Hexadienoyl-CoA with DECR1.png
Crystallization of DECR with 2,4 Hexadienoyl-CoA and NADPH (not shown). Key residues in the enzyme active site orient the substrate for hydride transfer through a network of hydrogen bonds.

Eukaryotic DECR exists in both the mitochondria (mDECR) and the peroxisome (pDECR, coded by gene DECR2). The enzymes from each organelle are homologous and part of the short-chain dehydrogenase/reductase SDR super-family. mDECR is 124 kDa consisting of 335 amino acids before post-translational modification. [2] The secondary structure shares many of the motifs of SDR, including a Rossmann fold for strong NADPH binding. The protein exists as a homotetramer in physiological environment, but has been shown to also form monomers and dimers in solution. [8]

Crystallization of mDECR [7] shows the enzyme provides a network of hydrogen bonds from key residues in the active site to NADPH and the 2,4-dienoyl-CoA which positions the hydride at 3.4 Å to the Cδ, compared with 4.0 Å to the Cβ (not shown). The enolate intermediate discussed earlier is stabilized by residues additional hydrogen bonds to Tyr166 and Asn148. Lys214 and Ser210 (conserved residues in all SDR enzymes) are thought to increase the pKa of Tyr166 and stabilize the transition state. [7] Additionally, at one end of the active site there is a flexible loop that provides sufficient room for long carbon chains. This likely gives the enzyme flexibility to process fatty acid chains of various lengths. Substrate length for mDECR catalysis is thought to be limited at 20 carbons, at which this very long chain fatty acid is first partially oxidized by pDECR in the peroxisome. [9]

Enzyme mechanism

Eukaryotic DECR

2,4 Dienoyl-CoA thioester reduction by NADPH to 3-Enoyl CoA occurs by a two-step sequential mechanism via an enolate intermediate. [10] DECR binds NADPH and the fatty acid thioester and positions them for specific hydride transfer to the Cδ on the hydrocarbon chain. The electrons from the Cγ-Cδ double bond move over to the Cβ-Cγ position, and those from the Cα-Cβ form an enolate. In the final step, a proton is abstracted from the water [11] to the Cα and the thioester is reformed, resulting in a single Cβ-Cγ trans double bond. Since the final proton comes from water, the pH has a significant effect on the catalytic rate with the enzyme demonstrating maximal activity at ~6.0. A decrease in activity at pH < 6.0 can be explained by de-protonation of titratable residues that affect protein folding or substrate binding. Mutant proteins with modifications at key acidic amino acids (E154, E227, E276, D300, D117) show order of magnitude increases in Km and/or decreases in Vmax. [8]

Proposed mechanism of 2,4-Trans dienoyl-CoA reduction by NADPH in mammalian DECR. The mechanism proceeds stepwise through an enolate intermediate. DECR Mechansim.png
Proposed mechanism of 2,4-Trans dienoyl-CoA reduction by NADPH in mammalian DECR. The mechanism proceeds stepwise through an enolate intermediate.

Prokaryotic DECR

2,4 Dienoyl-CoA Reductase from Escherichia coli shares very similar kinetic properties to that of eukaryotes, but differs significantly in both structure and mechanism. In addition to NADPH, E. Coli DECR requires a set of FAD, FMN and iron–sulfur cluster molecules to complete the electron transfer. [12] A further distinction is E. Coli DECR produces the final 2-trans-enoyl-CoA without the need for Enoyl CoA Isomerase. [11] The active site contains accurately positioned Tyr166 that donates a proton to the Cγ after hydride attack at the Cδ, completing the reduction in a single concerted step. [13] Surprisingly, mutation of the Tyr166 does not eliminate enzyme activity but instead changes the product to 3-trans-enoyl-CoA. The current explanation is that Glu164, an acidic residue in the active site, acts as a proton donor to Cα when Tyr166 is not present. [14]

Function

DECR is one of three auxiliary enzymes involved in a rate-limiting step of unsaturated fatty acid oxidation in mitochondria. In particular, this enzyme contributes to breaking the double bonds at all even-numbered positions, and some double bonds at odd-numbered position. [8] The structure of the ternary complex of pDCR (peroxisomal 2,4-dienoyl CoA reductases) with NADP and its substrate provides essential and unique insights into the mechanism of catalysis. [15] Unlike other members belonging to the SDR family, catalysis by pDCR does not involve a tyrosine-serine pair. [8] Instead, a catalytically critical aspartate, together with an invariant lysine, polarizes a water molecule to donate a proton for the formation of the product. [9] Although pDCR can use 2,4-hexadienoyl CoA as a substrate, the affinities for short chain fatty acids are lower. Analysis of the hinge movement of DCRs from the mitochondrion and peroxisomes sheds light on the reason behind the unique ability of the peroxisome to shorten very long chain fatty acids. [16]

Clinical significance

Mutations in the DECR1 gene may result in 2,4 Dienoyl-CoA reductase deficiency, [17] a rare but lethal disorder.

Due to its role in fatty acid oxidation, DECR may serve as a therapeutic target for treating non-insulin dependent diabetes mellitus (NIDDM), which features hyperglycemia due to increased fatty acid oxidation. [8]

In knockout mice studies, DECR1−/− subjects accumulate significant concentrations of mono and polyunsaturated fatty acids in the liver during fasting (such as oleic acid, palmitoleic acid, linoleic acid, and linolenic acid). Mutant subjects were also found to have poor tolerance to cold, decrease in diurnal activity, and an overall reduction in adaptation to metabolic stressors. [18]

See also

Related Research Articles

<span class="mw-page-title-main">Dihydrofolate reductase</span> Mammalian protein found in Homo sapiens

Dihydrofolate reductase, or DHFR, is an enzyme that reduces dihydrofolic acid to tetrahydrofolic acid, using NADPH as an electron donor, which can be converted to the kinds of tetrahydrofolate cofactors used in 1-carbon transfer chemistry. In humans, the DHFR enzyme is encoded by the DHFR gene. It is found in the q14.1 region of chromosome 5.

<span class="mw-page-title-main">Enoyl CoA isomerase</span>

Enoyl-CoA-(∆) isomerase (EC 5.3.3.8, also known as dodecenoyl-CoA- isomerase, 3,2-trans-enoyl-CoA isomerase, ∆3 ,∆2 -enoyl-CoA isomerase, or acetylene-allene isomerase, is an enzyme that catalyzes the conversion of cis- or trans-double bonds of coenzyme A bound fatty acids at gamma-carbon to trans double bonds at beta-carbon as below:

<span class="mw-page-title-main">Nicotinamide adenine dinucleotide phosphate</span> Chemical compound

Nicotinamide adenine dinucleotide phosphate, abbreviated NADP+ or, in older notation, TPN (triphosphopyridine nucleotide), is a cofactor used in anabolic reactions, such as the Calvin cycle and lipid and nucleic acid syntheses, which require NADPH as a reducing agent ('hydrogen source'). NADPH is the reduced form, whereas NADP+ is the oxidized form. NADP+ is used by all forms of cellular life.

Fatty acid metabolism consists of various metabolic processes involving or closely related to fatty acids, a family of molecules classified within the lipid macronutrient category. These processes can mainly be divided into (1) catabolic processes that generate energy and (2) anabolic processes where they serve as building blocks for other compounds.

In biochemistry and metabolism, beta oxidation (also β-oxidation) is the catabolic process by which fatty acid molecules are broken down in the cytosol in prokaryotes and in the mitochondria in eukaryotes to generate acetyl-CoA. Acetyl-CoA enters the citric acid cycle, generating NADH and FADH2, which are electron carriers used in the electron transport chain. It is named as such because the beta carbon of the fatty acid chain undergoes oxidation and is converted to a carbonyl group to start the cycle all over again. Beta-oxidation is primarily facilitated by the mitochondrial trifunctional protein, an enzyme complex associated with the inner mitochondrial membrane, although very long chain fatty acids are oxidized in peroxisomes.

Acyl-CoA dehydrogenases (ACADs) are a class of enzymes that function to catalyze the initial step in each cycle of fatty acid β-oxidation in the mitochondria of cells. Their action results in the introduction of a trans double-bond between C2 (α) and C3 (β) of the acyl-CoA thioester substrate. Flavin adenine dinucleotide (FAD) is a required co-factor in addition to the presence of an active site glutamate in order for the enzyme to function.

<span class="mw-page-title-main">Enoyl-CoA hydratase</span>

Enoyl-CoA hydratase (ECH) or crotonase is an enzyme EC 4.2.1.17 that hydrates the double bond between the second and third carbons on 2-trans/cis-enoyl-CoA:

In biochemistry, fatty acid synthesis is the creation of fatty acids from acetyl-CoA and NADPH through the action of enzymes called fatty acid synthases. This process takes place in the cytoplasm of the cell. Most of the acetyl-CoA which is converted into fatty acids is derived from carbohydrates via the glycolytic pathway. The glycolytic pathway also provides the glycerol with which three fatty acids can combine to form triglycerides, the final product of the lipogenic process. When only two fatty acids combine with glycerol and the third alcohol group is phosphorylated with a group such as phosphatidylcholine, a phospholipid is formed. Phospholipids form the bulk of the lipid bilayers that make up cell membranes and surrounds the organelles within the cells. In addition to cytosolic fatty acid synthesis, there is also mitochondrial fatty acid synthesis (mtFASII), in which malonyl-CoA is formed from malonic acid with the help of malonyl-CoA synthetase (ACSF3), which then becomes the final product octanoyl-ACP (C8) via further intermediate steps.

<span class="mw-page-title-main">Beta-ketoacyl-ACP synthase</span> Enzyme

In molecular biology, Beta-ketoacyl-ACP synthase EC 2.3.1.41, is an enzyme involved in fatty acid synthesis. It typically uses malonyl-CoA as a carbon source to elongate ACP-bound acyl species, resulting in the formation of ACP-bound β-ketoacyl species such as acetoacetyl-ACP.

D-Bifunctional protein deficiency is an autosomal recessive peroxisomal fatty acid oxidation disorder. Peroxisomal disorders are usually caused by a combination of peroxisomal assembly defects or by deficiencies of specific peroxisomal enzymes. The peroxisome is an organelle in the cell similar to the lysosome that functions to detoxify the cell. Peroxisomes contain many different enzymes, such as catalase, and their main function is to neutralize free radicals and detoxify drugs. For this reason peroxisomes are ubiquitous in the liver and kidney. D-BP deficiency is the most severe peroxisomal disorder, often resembling Zellweger syndrome.

The crotonase family comprises mechanistically diverse proteins that share a conserved trimeric quaternary structure, the core of which consists of 4 turns of a (beta/beta/alpha)n superhelix.

<span class="mw-page-title-main">3-oxoacyl-(acyl-carrier-protein) reductase</span> Enzyme

In enzymology, a 3-oxoacyl-[acyl-carrier-protein] reductase (EC 1.1.1.100) is an enzyme that catalyzes the chemical reaction

<span class="mw-page-title-main">Acyl-CoA dehydrogenase (NADP+)</span> Class of enzymes

In enzymology, an acyl-CoA dehydrogenase (NADP+) (EC 1.3.1.8) is an enzyme that catalyzes the chemical reaction

In enzymology, a cis-2-enoyl-CoA reductase (NADPH) (EC 1.3.1.37) is an enzyme that catalyzes the chemical reaction

In enzymology, an enoyl-[acyl-carrier-protein] reductase (NADPH, B-specific) (EC 1.3.1.10) is an enzyme that catalyzes the chemical reaction

In enzymology, a trans-2-enoyl-CoA reductase (NADPH) (EC 1.3.1.38) is an enzyme that catalyzes the chemical reaction

In enzymology, a trans-2-decenoyl-[acyl-carrier protein] isomerase is an enzyme that catalyzes the chemical reaction

Crotonyl-CoA reductase (EC 1.3.1.86, butyryl-CoA dehydrogenase, butyryl dehydrogenase, unsaturated acyl-CoA reductase, ethylene reductase, enoyl-coenzyme A reductase, unsaturated acyl coenzyme A reductase, butyryl coenzyme A dehydrogenase, short-chain acyl CoA dehydrogenase, short-chain acyl-coenzyme A dehydrogenase, 3-hydroxyacyl CoA reductase, butanoyl-CoA:(acceptor) 2,3-oxidoreductase, CCR) is an enzyme with systematic name butanoyl-CoA:NADP+ 2,3-oxidoreductase. This enzyme catalyses the following chemical reaction

Very-long-chain enoyl-CoA reductase (EC 1.3.1.93, TSC13 (gene name), CER10 (gene)) is an enzyme with systematic name very-long-chain acyl-CoA:NADP+ oxidoreductase. This enzyme catalyses the following chemical reaction

3-hydroxydecanoyl-(acyl-carrier-protein) dehydratase (EC 4.2.1.60, D-3-hydroxydecanoyl-[acyl-carrier protein] dehydratase, 3-hydroxydecanoyl-acyl carrier protein dehydrase, 3-hydroxydecanoyl-acyl carrier protein dehydratase, β-hydroxydecanoyl thioester dehydrase, β-hydroxydecanoate dehydrase, beta-hydroxydecanoyl thiol ester dehydrase, FabA, β-hydroxyacyl-acyl carrier protein dehydratase, HDDase, β-hydroxyacyl-ACP dehydrase, (3R)-3-hydroxydecanoyl-[acyl-carrier-protein] hydro-lyase) is an enzyme with systematic name (3R)-3-hydroxydecanoyl-(acyl-carrier protein) hydro-lyase. This enzyme catalyses the following chemical reaction

References

  1. "Entrez Gene: 2,4-dienoyl CoA reductase 1, mitochondrial".
  2. 1 2 Koivuranta KT, Hakkola EH, Hiltunen JK (December 1994). "Isolation and characterization of cDNA for human 120 kDa mitochondrial 2,4-dienoyl-coenzyme A reductase". The Biochemical Journal. 304 (3): 787–792. doi:10.1042/bj3040787. PMC   1137403 . PMID   7818482.
  3. Helander HM, Koivuranta KT, Horelli-Kuitunen N, Palvimo JJ, Palotie A, Hiltunen JK (November 1997). "Molecular cloning and characterization of the human mitochondrial 2,4-dienoyl-CoA reductase gene (DECR)". Genomics. 46 (1): 112–119. doi:10.1006/geno.1997.5004. PMID   9403065.
  4. Cuebas D, Schulz H (December 1982). "Evidence for a modified pathway of linoleate degradation. Metabolism of 2,4-decadienoyl coenzyme A". The Journal of Biological Chemistry. 257 (23): 14140–14144. doi: 10.1016/S0021-9258(19)45356-8 . PMID   7142199.
  5. 1 2 Liang X, Thorpe C, Schulz H (August 2000). "2,4-Dienoyl-CoA reductase from Escherichia coli is a novel iron-sulfur flavoprotein that functions in fatty acid beta-oxidation". Archives of Biochemistry and Biophysics. 380 (2): 373–379. doi:10.1006/abbi.2000.1941. PMID   10933894.
  6. Hanson, Kenneth R.; Rose, Irwin A. (1975-01-01). "Interpretations of enzyme reaction stereospecificity". Accounts of Chemical Research. 8 (1): 1–10. doi:10.1021/ar50085a001. ISSN   0001-4842.
  7. 1 2 3 PDB: 1w6u ; Alphey MS, Yu W, Byres E, Li D, Hunter WN (January 2005). "Structure and reactivity of human mitochondrial 2,4-dienoyl-CoA reductase: enzyme-ligand interactions in a distinctive short-chain reductase active site". The Journal of Biological Chemistry. 280 (4): 3068–3077. doi: 10.1074/jbc.M411069200 . PMID   15531764.
  8. 1 2 3 4 5 Yu W, Chu X, Chen G, Li D (February 2005). "Studies of human mitochondrial 2,4-dienoyl-CoA reductase". Archives of Biochemistry and Biophysics. 434 (1): 195–200. doi:10.1016/j.abb.2004.10.018. PMID   15629123.
  9. 1 2 Hua T, Wu D, Ding W, Wang J, Shaw N, Liu ZJ (August 2012). "Studies of human 2,4-dienoyl CoA reductase shed new light on peroxisomal β-oxidation of unsaturated fatty acids". The Journal of Biological Chemistry. 287 (34): 28956–28965. doi: 10.1074/jbc.M112.385351 . PMC   3436514 . PMID   22745130.
  10. Fillgrove KL, Anderson VE (October 2001). "The mechanism of dienoyl-CoA reduction by 2,4-dienoyl-CoA reductase is stepwise: observation of a dienolate intermediate". Biochemistry. 40 (41): 12412–12421. doi:10.1021/bi0111606. PMID   11591162.
  11. 1 2 Mizugaki M, Kimura C, Nishimaki T, Kawaguchi A, Okuda S, Yamanaka H (August 1983). "Studies on the metabolism of unsaturated fatty acids. XII. Reaction catalyzed by 2,4-dienoyl-CoA reductase of Escherichia coli". Journal of Biochemistry. 94 (2): 409–413. doi:10.1093/oxfordjournals.jbchem.a134370. PMID   6355075.
  12. Liang X, Thorpe C, Schulz H (August 2000). "2,4-Dienoyl-CoA reductase from Escherichia coli is a novel iron-sulfur flavoprotein that functions in fatty acid beta-oxidation". Archives of Biochemistry and Biophysics. 380 (2): 373–379. doi:10.1006/abbi.2000.1941. PMID   10933894.
  13. Hubbard PA, Liang X, Schulz H, Kim JJ (September 2003). "The crystal structure and reaction mechanism of Escherichia coli 2,4-dienoyl-CoA reductase". The Journal of Biological Chemistry. 278 (39): 37553–37560. doi: 10.1074/jbc.M304642200 . PMID   12840019.
  14. Tu X, Hubbard PA, Kim JJ, Schulz H (January 2008). "Two distinct proton donors at the active site of Escherichia coli 2,4-dienoyl-CoA reductase are responsible for the formation of different products". Biochemistry. 47 (4): 1167–1175. doi:10.1021/bi701235t. PMID   18171025.
  15. Ylianttila MS, Pursiainen NV, Haapalainen AM, Juffer AH, Poirier Y, Hiltunen JK, Glumoff T (May 2006). "Crystal structure of yeast peroxisomal multifunctional enzyme: structural basis for substrate specificity of (3R)-hydroxyacyl-CoA dehydrogenase units". Journal of Molecular Biology. 358 (5): 1286–1295. doi:10.1016/j.jmb.2006.03.001. PMID   16574148.
  16. Emekli U, Schneidman-Duhovny D, Wolfson HJ, Nussinov R, Haliloglu T (March 2008). "HingeProt: automated prediction of hinges in protein structures". Proteins. 70 (4): 1219–1227. doi: 10.1002/prot.21613 . PMID   17847101. S2CID   26975077.
  17. Roe CR, Millington DS, Norwood DL, Kodo N, Sprecher H, Mohammed BS, et al. (May 1990). "2,4-Dienoyl-coenzyme A reductase deficiency: a possible new disorder of fatty acid oxidation". The Journal of Clinical Investigation. 85 (5): 1703–1707. doi:10.1172/JCI114624. PMC   296625 . PMID   2332510.
  18. Miinalainen IJ, Schmitz W, Huotari A, Autio KJ, Soininen R, Ver Loren van Themaat E, et al. (July 2009). "Mitochondrial 2,4-dienoyl-CoA reductase deficiency in mice results in severe hypoglycemia with stress intolerance and unimpaired ketogenesis". PLOS Genetics. 5 (7): e1000543. doi: 10.1371/journal.pgen.1000543 . PMC   2697383 . PMID   19578400.