Carnitine O-octanoyltransferase

Last updated
Carnitine O-octanoyltransferase
CROT structure.png
Identifiers
EC no. 2.3.1.137
CAS no. 39369-19-2
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

Carnitine O-octanoyltransferase (CROT or COT) is a member of the transferase family, more specifically a carnitine acyltransferase, a type of enzyme which catalyzes the transfer of acyl groups from acyl-CoAs to carnitine, generating CoA and an acyl-carnitine. (EC 2.3.1.137) The systematic name of this enzyme is octanoyl-CoA:L-carnitine O-octanoyltransferase. Other names in common use include medium-chain/long-chain carnitine acyltransferase, carnitine medium-chain acyltransferase, easily solubilized mitochondrial carnitine palmitoyltransferase, and overt mitochondrial carnitine palmitoyltransferase. Specifically, CROT catalyzes the chemical reaction:

Contents

octanoyl-CoA + L-carnitine CoA + L-octanoylcarnitine

Thus, the two substrates of this enzyme are octanoyl-CoA and L-carnitine and its two products are CoA and L-octanoylcarnitine. [1]

This reaction is easily chemically reversible, and does not require any energy input, as both fatty acyl-CoAs and fatty acylcarnitines are considered chemically “activated” forms of fatty acyl groups. [2]

This enzyme belongs to the family of transferases, specifically those acyltransferases transferring groups other than aminoacyl groups.

Structure

Active site His327 His327 CROT.png
Active site His327

CROT is 612 amino acids long, [3] with a molecular weight of about 70 kDa. [2] In terms of broad overall structural features, CROT has 20 α-helices and 16 β-strands, and can be divided into two overall domains, named N and C. [3]

As of late 2007, 4 structures have been solved for this class of enzymes, with PDB accession codes 1XL7, 1XL8, 1XMC, and 1XMD.

CROT complex with carnitine.png
CROT in complex with carnitine
CROT active site pocket.png
Another representation of the CROT active site pocket in complex with carnitine

The significant catalytic residue within all carnitine acyltransferases, including CROT, has been found to be a histidine residue, confirmed by site-directed mutagenesis studies. In CROT, this residue is at position 327. This residue, including the rest of the active site, is found at the interface between the N and C domains. The active site stabilizes carnitine by an intricate network of hydrogen-bonding residues, along with a key water molecule. The longer acyl chain is stabilized by hydrophobic residues arrayed in an approximately-cylindrical fashion. [3]

As may be expected from members of the same enzymatic family, there is strong similarity between the structures of carnitine acetyltransferase (CRAT) and CROT, as these enzymes have 36% sequence homology. [3] A key difference between these enzymes that may explain their selectivities between short and medium-chain acyl-CoAs hinges on a glycine residue which is present in the acyl binding site in CROT, Gly-553. In CRAT, however, the residue in the same position in the acyl binding site is a methionine residue, Met-564. [3] These residues has been shown to serve as a substrate “gatekeeper” in both CRAT and CROT. M564G CRAT mutants have been shown to accept a wider variety of acyl-CoA substrates. [4] Similarly, G553M CROT mutants show marked inactivity with octanoyl-CoA, while maintaining activity with short-chain acyl-CoAs. [3]

Function

One function of CROT is to supply acetyl-CoA to glucose-starved cells. In the absence of carnitine acetyltransferase (CRAT), acyltransferases such as CROT can catalyze the acetyl group transfer from acetylcarnitine to coenzyme A. Rescue experiments with CROT gene knockout cells have shown that peroxisomal CROT can mediate acetyl-CoA production under glucose-limited conditions. The peroxisome can then export these products into the cytosol. [5]

Localization

Though CROT is distributed on both sides of microsomal vesicles, it has also been found that the bulk of CROT activity in murine liver is in the cytoplasm face of the vesicles and endoplasmic reticulum. CROT may play a role in converting peroxisomal medium-chain acylcarnitine derivatives to medium-chain acyl-CoA derivatives. These can then feed into a variety of biosynthetic pathways for elongations and other modifications. [6]

In addition, CROT is inhibited by trypsin in a dose-dependent manner. A maximum of 60% inhibition was observed in purified CROT, similar to what was seen with carnitine palmitoyltransferase (CPT). CROT activity also appears to be inhibited to the same extent in both permeable and sealed microsomal membranes. [6]

CROT is thought to be peroxisomally-located. It was found that administration of di(2-ethylhexyl)phthalate (DEHP), a peroxisomal proliferator, to Wistar rats led to an increase in the expression of CROT by a factor of 14.1. This was as a result of increased translation of CROT mRNA, along with decreased degradation by a factor of 1.5. [7]

CROT activity has also been reported in mouse liver, kidney, adipocyte, mammary gland, skeletal muscle, and heart tissues. It was found that CROT activity in the kidney was mostly overt, while in the liver and heart it was mainly latent. Interestingly, the trend for a related enzyme, carnitine palmitoyl transferase (CPT), the opposite trend was found. [8]

Substrates

Carnitine octanoyltransferase reaction.png
Canonical CROT reaction
Catalytic promiscuity of carnitine octanoyltransferase.png
Other acyl-CoAs which CROT can take as substrates

While CROT's canonical substrate is octanoyl-CoA, CROT is also known to be able to catalyze the deacylation of numerous acyl-CoAs, such as acetyl-CoA, propionyl-CoA, butyryl-CoA, and hexanoyl-CoA. [5] CROT can also take branched-chain fatty acyl-CoAs as substrates, such as 4,8-dimethylnonanoyl-CoA, which is derived from the metabolism of pristanic acid in the peroxisome. [9]

Regulation

Because CROT activity has a role in beta-oxidation of fatty acids and ketone body synthesis, it is an important point of regulation. One known inhibitor of CROT is malonyl-CoA, which inhibits CROT non-linearly. Complex kinetic behavior is observed when malonyl-CoA is incubated with purified CROT.

A decrease in pH can also enhance malonyl-CoA inhibition of CROT. Some studies have indicated that when the pH of assaying conditions was decreased from 7.4 to 6.8, inhibition could increase by 20-30%. Further, the Ki for malonyl-CoA in CROT decreases from 106 uM to 35 uM over this drop. This change is not seen for palmitoyl-CoA and decanoyl-CoA. However, the degree of inhibition by malonyl-CoA is similar to that observed with other short-chain acyl-CoA esters, such as glutaryl-CoA, hydroxymethylglutaryl-CoA, and methylmalonyl-CoA.

The ionization state of malonyl-CoA does not change significantly over the pH range 7.4-6.8. The change in sensitivity to inhibitors may be due to the CROT active-site His-327 residue. Malonyl-CoA is also found at a lower concentration in the cell (1-6 uM) than its Ki.Thus, its inhibition of CROT may not be physiologically significant under homeostatic conditions. [10]

Related Research Articles

<span class="mw-page-title-main">Carnitine</span> Amino acid active in mitochondria

Carnitine is a quaternary ammonium compound involved in metabolism in most mammals, plants, and some bacteria. In support of energy metabolism, carnitine transports long-chain fatty acids from the cytosol into mitochondria to be oxidized for free energy production, and also participates in removing products of metabolism from cells. Given its key metabolic roles, carnitine is concentrated in tissues like skeletal and cardiac muscle that metabolize fatty acids as an energy source. Generally individuals, including strict vegetarians, synthesize enough L-carnitine in vivo.

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.

<span class="mw-page-title-main">Beta oxidation</span> Process of fatty acid breakdown

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, which enters the citric acid cycle, and NADH and FADH2, which are co-enzymes used in the electron transport chain. It is named as such because the beta carbon of the fatty acid undergoes oxidation to a carbonyl group. 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.

<span class="mw-page-title-main">Inborn error of lipid metabolism</span> Medical condition

Numerous genetic disorders are caused by errors in fatty acid metabolism. These disorders may be described as fatty oxidation disorders or as a lipid storage disorders, and are any one of several inborn errors of metabolism that result from enzyme defects affecting the ability of the body to oxidize fatty acids in order to produce energy within muscles, liver, and other cell types.

<span class="mw-page-title-main">Acetyl-CoA carboxylase</span> Enzyme that regulates the metabolism of fatty acids

Acetyl-CoA carboxylase (ACC) is a biotin-dependent enzyme that catalyzes the irreversible carboxylation of acetyl-CoA to produce malonyl-CoA through its two catalytic activities, biotin carboxylase (BC) and carboxyltransferase (CT). ACC is a multi-subunit enzyme in most prokaryotes and in the chloroplasts of most plants and algae, whereas it is a large, multi-domain enzyme in the cytoplasm of most eukaryotes. The most important function of ACC is to provide the malonyl-CoA substrate for the biosynthesis of fatty acids. The activity of ACC can be controlled at the transcriptional level as well as by small molecule modulators and covalent modification. The human genome contains the genes for two different ACCs—ACACA and ACACB.

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

Malonyl-CoA is a coenzyme A derivative of malonic acid.

<span class="mw-page-title-main">Carnitine palmitoyltransferase II deficiency</span> Medical condition

Carnitine palmitoyltransferase II deficiency, sometimes shortened to CPT-II or CPT2, is an autosomal recessively inherited genetic metabolic disorder characterized by an enzymatic defect that prevents long-chain fatty acids from being transported into the mitochondria for utilization as an energy source. The disorder presents in one of three clinical forms: lethal neonatal, severe infantile hepatocardiomuscular and myopathic.

<span class="mw-page-title-main">Carnitine O-palmitoyltransferase</span>

Carnitine O-palmitoyltransferase is a mitochondrial transferase enzyme involved in the metabolism of palmitoylcarnitine into palmitoyl-CoA. A related transferase is carnitine acyltransferase.

<span class="mw-page-title-main">Malonyl-CoA decarboxylase</span> Class of enzymes

Malonyl-CoA decarboxylase, is found in bacteria and humans and has important roles in regulating fatty acid metabolism and food intake, and it is an attractive target for drug discovery. It is an enzyme associated with Malonyl-CoA decarboxylase deficiency. In humans, it is encoded by the MLYCD gene.

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

Acyl-CoA is a group of coenzymes that metabolize fatty acids. Acyl-CoA's are susceptible to beta oxidation, forming, ultimately, acetyl-CoA. The acetyl-CoA enters the citric acid cycle, eventually forming several equivalents of ATP. In this way, fats are converted to ATP, the universal biochemical energy carrier.

Fatty acid degradation is the process in which fatty acids are broken down into their metabolites, in the end generating acetyl-CoA, the entry molecule for the citric acid cycle, the main energy supply of living organisms, including bacteria and animals. It includes three major steps:

<span class="mw-page-title-main">Long-chain-fatty-acid—CoA ligase</span> Class of enzymes

The long chain fatty acyl-CoA ligase is an enzyme of the ligase family that activates the oxidation of complex fatty acids. Long chain fatty acyl-CoA synthetase catalyzes the formation of fatty acyl-CoA by a two-step process proceeding through an adenylated intermediate. The enzyme catalyzes the following reaction,

<span class="mw-page-title-main">Carnitine palmitoyltransferase I</span> Protein-coding gene in the species Homo sapiens

Carnitine palmitoyltransferase I (CPT1) also known as carnitine acyltransferase I, CPTI, CAT1, CoA:carnitine acyl transferase (CCAT), or palmitoylCoA transferase I, is a mitochondrial enzyme responsible for the formation of acyl carnitines by catalyzing the transfer of the acyl group of a long-chain fatty acyl-CoA from coenzyme A to l-carnitine. The product is often Palmitoylcarnitine, but other fatty acids may also be substrates. It is part of a family of enzymes called carnitine acyltransferases. This "preparation" allows for subsequent movement of the acyl carnitine from the cytosol into the intermembrane space of mitochondria.

<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.

Pantothenate kinase (EC 2.7.1.33, PanK; CoaA) is the first enzyme in the Coenzyme A (CoA) biosynthetic pathway. It phosphorylates pantothenate (vitamin B5) to form 4'-phosphopantothenate at the expense of a molecule of adenosine triphosphate (ATP). It is the rate-limiting step in the biosynthesis of CoA.

Palmitoyl-CoA hydrolase (EC 3.1.2.2) is an enzyme in the family of hydrolases that specifically acts on thioester bonds. It catalyzes the hydrolysis of long chain fatty acyl thioesters of acyl carrier protein or coenzyme A to form free fatty acid and the corresponding thiol:

In enzymology, a [acyl-carrier-protein] S-acetyltransferase is an enzyme that catalyzes the reversible chemical reaction

<span class="mw-page-title-main">Carnitine O-acetyltransferase</span> Enzyme

Carnitine O-acetyltransferase also called carnitine acetyltransferase is an enzyme that encoded by the CRAT gene that catalyzes the chemical reaction

<span class="mw-page-title-main">Fatty-acyl-CoA synthase</span>

Fatty-acyl-CoA Synthase, or more commonly known as yeast fatty acid synthase, is an enzyme complex responsible for fatty acid biosynthesis, and is of Type I Fatty Acid Synthesis (FAS). Yeast fatty acid synthase plays a pivotal role in fatty acid synthesis. It is a 2.6 MDa barrel shaped complex and is composed of two, unique multi-functional subunits: alpha and beta. Together, the alpha and beta units are arranged in an α6β6 structure. The catalytic activities of this enzyme complex involves a coordination system of enzymatic reactions between the alpha and beta subunits. The enzyme complex therefore consists of six functional centers for fatty acid synthesis.

<span class="mw-page-title-main">CROT (gene)</span> Protein-coding gene in the species Homo sapiens

Peroxisomal carnitine O-octanoyltransferase is an enzyme that in humans is encoded by the CROT gene.

References

  1. Farrell, S. O.; Fiol, C. J.; Reddy, J. K.; Bieber, L. L. (1984-11-10). "Properties of purified carnitine acyltransferases of mouse liver peroxisomes". The Journal of Biological Chemistry. 259 (21): 13089–13095. doi: 10.1016/S0021-9258(18)90661-7 . ISSN   0021-9258. PMID   6436243.
  2. 1 2 Jogl, Gerwald; Hsiao, Yu-Shan; Tong, Liang (November 2004). "Structure and function of carnitine acyltransferases". Annals of the New York Academy of Sciences. 1033 (1): 17–29. Bibcode:2004NYASA1033...17J. doi:10.1196/annals.1320.002. ISSN   0077-8923. PMID   15591000. S2CID   24466239.
  3. 1 2 3 4 5 6 Jogl, Gerwald; Hsiao, Yu-Shan; Tong, Liang (2005-01-07). "Crystal structure of mouse carnitine octanoyltransferase and molecular determinants of substrate selectivity". The Journal of Biological Chemistry. 280 (1): 738–744. doi: 10.1074/jbc.M409894200 . ISSN   0021-9258. PMID   15492013.
  4. Hsiao, Yu-Shan; Jogl, Gerwald; Tong, Liang (2004-07-23). "Structural and biochemical studies of the substrate selectivity of carnitine acetyltransferase". The Journal of Biological Chemistry. 279 (30): 31584–31589. doi: 10.1074/jbc.M403484200 . ISSN   0021-9258. PMID   15155726.
  5. 1 2 Hsu, Jake; Fatuzzo, Nina; Weng, Nielson; Michno, Wojciech; Dong, Wentao; Kienle, Maryline; Dai, Yuqin; Pasca, Anca; Abu-Remaileh, Monther; Rasgon, Natalie; Bigio, Benedetta; Nasca, Carla; Khosla, Chaitan (February 2023). "Carnitine octanoyltransferase is important for the assimilation of exogenous acetyl-L-carnitine into acetyl-CoA in mammalian cells". The Journal of Biological Chemistry. 299 (2): 102848. doi: 10.1016/j.jbc.2022.102848 . ISSN   1083-351X. PMC   9898754 . PMID   36587768.
  6. 1 2 Valkner, K. J.; Bieber, L. L. (1982-07-14). "The sidedness of carnitine acetyltransferase and carnitine octanoyltransferase of rat liver endoplasmic reticulum". Biochimica et Biophysica Acta (BBA) - Biomembranes. 689 (1): 73–79. doi:10.1016/0005-2736(82)90190-0. ISSN   0006-3002. PMID   7104352.
  7. Ozasa, H.; Miyazawa, S.; Osumi, T. (August 1983). "Biosynthesis of carnitine octanoyltransferase and carnitine palmitoyltransferase". Journal of Biochemistry. 94 (2): 543–549. doi:10.1093/oxfordjournals.jbchem.a134385. ISSN   0021-924X. PMID   6630174.
  8. Saggerson, E. D.; Carpenter, C. A. (1981-07-06). "Carnitine palmitoyltransferase and carnitine octanoyltransferase activities in liver, kidney cortex, adipocyte, lactating mammary gland, skeletal muscle and heart". FEBS Letters. 129 (2): 229–232. doi: 10.1016/0014-5793(81)80171-8 . ISSN   0014-5793. PMID   7286216. S2CID   2011527.
  9. Ferdinandusse, S.; Mulders, J.; IJlst, L.; Denis, S.; Dacremont, G.; Waterham, H. R.; Wanders, R. J. (1999-09-16). "Molecular cloning and expression of human carnitine octanoyltransferase: evidence for its role in the peroxisomal beta-oxidation of branched-chain fatty acids". Biochemical and Biophysical Research Communications. 263 (1): 213–218. doi:10.1006/bbrc.1999.1340. ISSN   0006-291X. PMID   10486279.
  10. A'Bháird, N. N.; Ramsay, R. R. (1992-09-01). "Malonyl-CoA inhibition of peroxisomal carnitine octanoyltransferase". The Biochemical Journal. 286 ( Pt 2) (Pt 2): 637–640. doi:10.1042/bj2860637. ISSN   0264-6021. PMC   1132947 . PMID   1530596.