Download as pdf or txt
Download as pdf or txt
You are on page 1of 24

Licenciado para - Paulo Sérgio Martins dos Santos - 09609690718 - Protegido por Eduzz.

com

Current Topic

pubs.acs.org/biochemistry

Cholesterol Biosynthesis: A Mechanistic Overview


Nuno M. F. S. A. Cerqueira, Eduardo F. Oliveira, Diana S. Gesto, Diogo Santos-Martins, Cátia Moreira,
Hari N. Moorthy, Maria J. Ramos, and P. A. Fernandes*
UCIBO-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, 4169-007 Porto,
Portugal

ABSTRACT: Cholesterol is an essential component of cell membranes and


the precursor for the synthesis of steroid hormones and bile acids. The
synthesis of this molecule occurs partially in a membranous world (especially
Downloaded from pubs.acs.org by WESTERN SYDNEY UNIV on 01/09/19. For personal use only.

the last steps), where the enzymes, substrates, and products involved tend to be
extremely hydrophobic. The importance of cholesterol has increased in the past
half-century because of its association with cardiovascular diseases, which are
considered one of the leading causes of death worldwide. In light of the current
need for new drugs capable of controlling the levels of cholesterol in the
bloodstream, it is important to understand how cholesterol is synthesized in the organism and identify the main enzymes
involved in this process. Taking this into account, this review presents a detailed description of several enzymes involved in the
biosynthesis of cholesterol. In this regard, the structure and catalytic mechanism of the enzymes involved in cholesterol
biosynthesis, from the initial two-carbon acetyl-CoA building block, will be reviewed and their current pharmacological
importance discussed. We believe that this review may contribute to a deeper level of understanding of cholesterol metabolism
Biochemistry 2016.55:5483-5506.

and that it will serve as a useful resource for future studies of the cholesterol biosynthesis pathway.

C holesterol is the major sterol present in animal tissues. This


molecule is almost planar and rigid and contains a steroid
nucleus of four fused rings, three of which have six carbons and a
hormones that are produced by the adrenal gland and by the male
and female sex glands.
Over the years, cholesterol has gained a bad reputation in the
fourth that has five. The molecule is amphiphilic, having a world of health and nutrition, especially because of its association
hydrophobic hydrocarbon body and a hydrophilic hydroxyl with cardiovascular diseases. According to the World Health
headgroup. Organization (WHO), from the top 10 leading causes of death
In mammals, cholesterol plays a vital role in life, being an worldwide in 2008, ischemic heart disease was number one,
essential component for the normal functioning of cells. Its roles accounting for 12.8% of deaths, followed by stroke and other
range from being a component in cell membranes to being a cerebrovascular disease as number two (10.8%). Because both of
precursor of several steroid hormones. these diseases are associated with high levels of cholesterol in the
Cell membranes are very complex systems, which separate the blood (hypercholesterolemia), it is easy to understand why this
molecule has acquired such a bad name.
cytosol of the cells from the external medium or from the
Despite its association with several heart conditions,
medium inside cell compartments (like lysosome or perox-
cholesterol should not be perceived as a bad compound that
isomes) while still allowing for the transfer of compounds from
needs to be avoided at all costs. In fact, life as we know it would
the inside to the outside and vice versa, as well as performing not be possible without cholesterol.
other important functions. Still, in a simplistic manner, they can In terms of composition, it is possible to distinguish between
be seen essentially as a double layer of phospholipids. good and bad cholesterol. Because it is a lipid, cholesterol cannot
Cholesterol is normally present in membranes, and its be dissolved in the bloodstream, which is water-based. To solve
importance can be easily understood just by comparing the this problem, the body packages cholesterol and other fats into
difference in the fluidity of membranes with different cholesterol protein-covered molecular assemblies called lipoproteins that do
concentrations. The polar headgroup of cholesterol interacts mix easily with blood. The proteins that are used in this assembly
with the similar headgroup of phospholipids, while the nonpolar are known as apolipoproteins. When the proportion of protein to
group interacts with their hydrophobic tails. Because cholesterol lipids (cholesterol and others) in the lipoprotein is high, it is
is somewhat rigid (more rigid than phospholipids), membranes known as high-density lipoprotein (HDL), or good cholesterol.
with higher cholesterol content will tend to be more rigid and On the other hand, when this proportion is small, meaning that
packed, while those with less cholesterol will be more fluid. there is a higher lipidic content, it is called low-density
Cholesterol is also important in other membrane processes, such lipoprotein (LDL), or bad cholesterol.
as endocytosis.
In addition to being a structural component of membranes, Received: April 12, 2016
cholesterol also serves as a precursor for the biosynthesis of Revised: September 7, 2016
several compounds like bile acids, vitamin D, and several steroid Published: September 7, 2016

© 2016 American Chemical Society 5483 DOI: 10.1021/acs.biochem.6b00342


Biochemistry 2016, 55, 5483−5506
Licenciado para - Paulo Sérgio Martins dos Santos - 09609690718 - Protegido por Eduzz.com

Biochemistry Current Topic

Figure 1. Diagram describing most of the enzymes involved in cholesterol biosynthesis. Each enzyme is identified with a different letter that corresponds
to the header of the following sections, where each enzyme is described in more detail. When this letter is followed by an “s”, it means that multiple
enzymes are involved in that step.

High levels of cholesterol are especially dangerous when the cerebrovascular diseases.1,2 Atherosclerosis is nothing more
concentration of LDL in the blood is high and that of HDL is low. than the accumulation of fatty materials, such as cholesterol, in
High levels of LDL may lead to atherosclerosis, which is one of the blood vessels. It is a complex process, which involves a
the main causes of both ischemic heart disease and chronic inflammatory response to oxidized LDL on the walls of
5484 DOI: 10.1021/acs.biochem.6b00342
Biochemistry 2016, 55, 5483−5506
Licenciado para - Paulo Sérgio Martins dos Santos - 09609690718 - Protegido por Eduzz.com

Biochemistry Current Topic

Figure 2. (A) Structure of thiolase II from Zoogloea ramigera [Protein Data Bank (PDB) entry 1dm38] and the active site with a reaction intermediate
(the enzyme is acetylated at Cys89, and a molecule of acetyl-CoA is found in the active-site pocket). (B) Proposed catalytic mechanism.17

arteries. LDL is very rich in cholesterol and cholesteryl esters, considerable ingestion of cholesterol-rich foods, exceeds the
which, when oxidized, are toxic to the cells on the walls of amount needed for the production of steroids, bile acids, and
arteries, triggering an inflammatory response. This leads to a membranes.3
pathogenic accumulation of cholesterol in blood vessels and the Hypercholesterolemia is currently treated with a combination
formation of atherosclerotic plaques, resulting in the constriction of dietary and pharmaceutical therapies.4,5 Often, more than a
of blood vessels. Atherosclerosis occurs when the amount of single pharmaceutical agent and a dietary regimen are necessary
cholesterol in the blood, due to either unregulated synthesis or to decrease total cholesterol and LDL levels to the desired level.
5485 DOI: 10.1021/acs.biochem.6b00342
Biochemistry 2016, 55, 5483−5506
Licenciado para - Paulo Sérgio Martins dos Santos - 09609690718 - Protegido por Eduzz.com

Biochemistry Current Topic

Drugs such as bile acid sequestrates, niacin, and statins are fatty acids or pyruvate) in the mitochondria or synthesized from
commonly used to treat hypercholesterolemia and atheroscle- the cytosolic acetate derived from cytoplasmic oxidation of
rosis. The application of several of these compounds is, however, ethanol, which is initiated by cytoplasmic alcohol dehydrogenase.
limited by the numerous side effects that can be experienced by Thiolases are ubiquitous enzymes that have key roles in many
patients. Thus, the need for therapeutic agents that decrease vital biochemical pathways.7 The members of this family of
cholesterol levels still exists. enzymes can be divided into two broad categories: degradative
In light of the current need for new drugs capable of thiolases [3-ketoacyl-CoA thiolase (EC 2.3.1.16)] and bio-
controlling the levels of cholesterol in the bloodstream, it is synthetic thiolases [acetoacetyl-CoA thiolase (EC 2.3.1.9)]. 3-
important to understand how the organism synthesizes this Ketoacyl-CoA thiolases have broad chain-length specificity for
molecule and identify the main enzymes involved in this process. their substrates and are involved in degradative pathways such as
Taking this into account, we will describe the most important fatty acid β-oxidation.8 Acetoacetyl-CoA thiolases are specific for
enzymes participating in cholesterol biosynthesis in this review the thiolysis of acetoacetyl-CoA and are involved in the first step
and pay special attention to those that are current drug targets.


of many biosynthetic pathways, including those that generate
cholesterol, steroid hormones, and ketone body energy storage
ENZYMES INVOLVED IN THE CHOLESTEROL molecules.9 There are two types of acetoacetyl-CoA thiolases,
PATHWAY mitochondrial ACAT1 and cytosolic ACAT2. Kovacs et al.
All cholesterol present in our bodies arises from two different suggest a possible distribution of ACAT1 between peroxisomes
sources. It can be either synthesized de novo within our cells or and mitochondria, as experimental evidence supports the
obtained through ingestion of certain foods. Although many formation of acetoacetyl-CoA in peroxisomes.10
people regularly include these foods in their diet, there is no need The physiological importance of thiolases can be illustrated by
to ingest them for the sole purpose of obtaining cholesterol the severe, and usually lethal, phenotypes of patients with
because our own cells are capable of producing enough of this deficient thiolases,11,12 particularly in the case of defective
molecule for our bodily requirements.6 Nonetheless, regardless biosynthetic thiolases.13,14 Currently, inhibitors against ACATs
of whether there is dietary intake of cholesterol, its levels are have been used to develop new drugs against African sleeping
maintained through regulation of its synthesis and absorption, sickness and other diseases.15,16
which means that when small quantities of cholesterol are Most enzymes of the thiolase family are dimers. The active site
ingested, absorption and synthesis will be upregulated. Likewise, of the biosynthetic thiolases is located in a shallow cleft at the
if the level of dietary intake is high, its rate of excretion will be protein surface. Enzyme kinetics, active-site labeling, and site-
increased and its rate of synthesis will be decreased. directed mutagenesis experiments allowed the identification of
The biosynthesis of cholesterol is a complex process, heavily three important residues, Cys89, His348, and Cys378, which
regulated at several points throughout its progression. Some of have been found to be important for the catalytic activity of the
the intermediaries can be diverted and used as precursors in the enzyme (Figure 2A). These residues are part of an extensive
biosynthesis of other compounds or perform themselves certain hydrogen bond network, which stretches from the active site to
functions on the body. This process requires numerous enzymes, the enzyme backside, suggesting that this hydrogen bond
some of which are among the most regulated enzymes currently network is important for the stabilization of different protonation
known. states of the catalytic residues.8
The first step in the synthesis of cholesterol is the formation of
On the basis of these studies, a “ping-pong” reaction
mevalonate from acetate. It begins with the condensation of two
mechanism consisting of two steps has been proposed17 (Figure
acetyl-coenzyme A (acetyl-CoA) molecules to form acetoacetyl-
2B). The first step of the mechanism involves the deprotonation
CoA, a process catalyzed by the enzyme thiolase. Next, HMG-
of Cys89 by His348 (Figure 2B, step 2).18 Simultaneously, the
CoA synthase catalyzes the reaction between acetoacetyl-CoA
and another molecule of acetyl-CoA to form HMG-CoA. The nucleophilic Cys89 attacks the acyl-CoA substrate, leading to the
final step in the synthesis of mevalonate is accomplished by formation of a covalent acyl-CoA tetrahedral intermediate
HMG-CoA reductase. This step is not only the committed step (Figure 2B, step 2). It has then been proposed that afterward
of the whole process but also the rate-limiting one. Cys378 becomes negatively charged, following the donation of
The subsequent step in the biosynthesis of cholesterol its proton to the leaving group of the acetylation reaction, CoA
comprises the conversion of mevalonate into two activated (Figure 2B, step 3). Subsequently, in the second half of the
isoprenes (isopentanyl 5-pyrophosphate and dimethylallyl synthesis reaction, Cys378 acts as a catalytic base that
pyrophosphate). Following a series of successive condensations deprotonates C2 of a new acetyl-CoA molecule, resulting in
of activated isoprenes, a 30-carbon molecule, squalene, is formed. the formation of an enolate intermediate (Figure 2B, step 4). The
Squalene is the biochemical precursor of all steroids, and despite following steps of the catalytic mechanisms are the reverse of
it being a linear compound, its structure can still be linked to that steps 2 and 3 and involve the formation of a second covalent
of cyclic steroids. To form cholesterol, squalene has to endure a tetrahedral intermediate (Figure 2B, step 5), the transfer of a
succession of changes, being initially converted to lanosterol, a proton from His348 to Cys89, and the subsequent release of the
four-ring compound, which is finally transformed into product of the reaction (Figure 2B, steps 6 and 7). During the
cholesterol after several sequential reactions. catalytic process, the formation of the negatively charged
Figure 1 describes the overall process by which cholesterol is tetrahedral intermediates stabilized the oxyanion hole, which is
synthesized, from acetyl-CoA to lanosterol. Each of these composed of the Asn316-Wat82 dyad and His348.
reactions will be described in more detail below. B. HMG-CoA Synthase. HMG-CoA synthase (HMG-CoA-
A. Thiolase. Thiolases, also known as acetyl-coenzyme A S, EC 2.3.3.10) is a 42 kDa homodimeric protein that catalyzes
acetyltransferases (ACAT), are the enzymes responsible for the the irreversible condensation of acetyl-CoA and acetoacetyl-CoA
conversion of 2 units of acetyl-CoA into acetoacetyl-CoA. The to form 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) and
acetyl-CoA is either obtained from an oxidation reaction (e.g., CoA.
5486 DOI: 10.1021/acs.biochem.6b00342
Biochemistry 2016, 55, 5483−5506
Licenciado para - Paulo Sérgio Martins dos Santos - 09609690718 - Protegido por Eduzz.com

Biochemistry Current Topic

Figure 3. (A) Structure of cHMG-CoA synthase (PDB entry 2P8U24) and the active site with the product of the reaction and Cys117 acetylated. (B)
Proposed catalytic mechanism of the enzyme.24−27

HMG-CoA-S is found in eukaryotes, archaea, and certain isoprenoid pathway, which yields cholesterol and other
bacteria.19 Cells of higher eukaryotes have three forms of HMG- important products, such as ubiquinone, dolichol, isopentenyl
CoA synthase: the cytosolic, the mitochondrial, and the adenosine, and farnesyl groups. Mitochondrial HMG-CoA-S
peroxisomal.20−22 Cytosolic and peroxisomal HMG-CoA-S (mHMG-CoA-S) participates mainly in the ketogenesis pathway
(cHMG-CoA-S and pHMG-CoA-S, respectively) catalyze the for acetoacetate biosynthesis. Acetoacetate is then transformed
second step in the synthesis of cholesterol. This starts the into hydroxybutyrate and acetone, often known as ketone bodies.
5487 DOI: 10.1021/acs.biochem.6b00342
Biochemistry 2016, 55, 5483−5506
Licenciado para - Paulo Sérgio Martins dos Santos - 09609690718 - Protegido por Eduzz.com

Biochemistry Current Topic

Figure 4. (A) Structure and active site of HMG-CoA-R (PDB entry 1dq935). (B) Currently accepted catalytic mechanism of HMG-CoA-R.40−44,56

In bacteria, isoprenoid precursors are generally synthesized via an between the upper and lower regions defines a 15 Å deep narrow
alternative non-mevalonate pathway. However, a number of tunnel that forms the active site in each monomer (Figure 3A).
Gram-positive pathogens use a mevalonate pathway involving a Both substrates, acetyl-CoA and acetoacetyl-CoA, fit in the
bacterial HMG-CoA-S isoform that is parallel to that found in active-site tunnel with the nucleotide part pointing toward the
eukaryotes.23 protein surface. The pantetheine part of the substrate is located
The main structure of HMG-CoA-S can be divided into two in the center of the tunnel that is enclosed by a hydrophobic
main regions: the larger upper region that contains a conserved sleeve, and the CoA thiol group is buried in the bottom of the
αβαβα structure from the thiolase fold and the smaller lower active-site tunnel, where the catalytic triad, formed by Glu95,
region, which is unique among HMG-CoA-S enzymes and Cys129, and His264, is located24(Figure 3A). Cys129 is placed in
consists of several α-helices and β-strands.24 The interface the deepest portion of the active site and is responsible for the
5488 DOI: 10.1021/acs.biochem.6b00342
Biochemistry 2016, 55, 5483−5506
Licenciado para - Paulo Sérgio Martins dos Santos - 09609690718 - Protegido por Eduzz.com

Biochemistry Current Topic

nucleophilic attack on the substrate. The main function of HMG-CoA.10,33 For this reason, the enzyme commission
His264 and Glu95 is to remove the proton from Cys129 and turn designated this enzyme as EC 1.1.1.34 for the NADPH-
it into a stronger nucleophile. Mutations on any of these residues dependent enzyme, whereas EC 1.1.1.88 links to the NADH-
inactivate the enzyme.25−27 dependent enzyme.
From a mechanistic point of view, the condensation reaction From sequence analysis, it was possible to divide different
catalyzed by HMG-CoA-S is similar to that of thiolase. However, forms of HMG-CoA-R into two main classes. Class I enzymes
the mechanism diverges at the second (condensation) step of the comprise those found in eukaryotes and the majority of archaea,
reaction, where the methyl group of the acetylated enzyme is while class II enzymes are found in prokaryotes and some
activated and attacks the incoming β-keto thioester, whereas the archaea.
members of the thiolase family activate a carbon of the second Class I HMG-CoA-R contains a transmembrane domain
substrate to attack the enzyme-bound thioester.28 On the basis of (lacking in archaeal enzymes) and a C-terminal catalytic region.34
these mutagenic studies, other kinetic studies,27,29 and cocrystal- The catalytic region of these enzymes is well-conserved, with
lized X-ray structures with bound intermediates,24,28,30 one can sequence identities of ∼60%, and can be subdivided into three
propose the mechanism of HMG-CoA synthase that is domains: an N-domain (N-terminal), a large L-domain, and a
summarized in Figure 3B. small S-domain (inserted within the L-domain). The L-domain
The first step of the catalytic process involves the activation of binds the substrate, while the S-domain binds NADPH. The
Cys129, through the action of His264, which abstracts a proton membrane region is diverse in sequence and length and exhibits
from that residue (Figure 3B, step 2). The negatively charged functional differences. For instance, plants have two membrane
cysteine promptly attacks the acetyl-CoA and forms a thioester domains, yeast seven, and mammals eight.35 Class II HMG-CoA-
acyl−enzyme intermediate at the same time that a reduced CoA R lacks the membrane domain and therefore is found dissolved in
is generated. The next step involves a Claisen-like condensation the cytosol. Their catalytic region is structurally related to the one
of the second substrate of the reaction, acetoacetyl-CoA, with the found in class I enzymes but consists of only two domains: a large
thioester acyl−enzyme intermediate to form HMG-CoA, while it L-domain and a small S-domain (inserted within the L-domain).
retains the thioester bond to the enzyme (Figure 3B, step 3). As with class I enzymes, the L-domain binds the substrate,
This reaction is favored by the proximity of Glu95, which whereas the S-domain binds NADH (instead of NADPH in class
abstracts a proton from the methyl group of the thioester acyl− I).
enzyme intermediate, turning it into a better nucleophile, and by HMG-CoA-R from both classes has a dimeric active site
His264, which gives a proton to one of the carbonyl groups of (Figure 4A), with residues contributed by both a monomer and a
acetoacetyl-CoA, turning it into a better electrophile.24,31 The nucleotide binding motif that is found in many enzymes that use
last two steps of the catalytic process involve the hydrolysis of the the dinucleotides NADH and NADPH for catalysis.36 The core
acetylcysteine bond (Figure 3B, steps 4 and 5), which yields the regions containing the catalytic domains of the two enzymes have
free HMG-CoA. At the end of this step, Cys129 becomes similar folds. Despite the differences in amino acid sequence and
reduced, and it is suggested that Glu95 loses a proton to the overall architecture, functionally similar residues participate in
solvent, thus making the active site ready for a new catalytic the binding of coenzyme A, and the position and orientation of
cycle.31 four key catalytic residues (glutamate, lysine, aspartate, and
HMG-CoA-S is currently a potential drug target. Inhibitors of histidine) are conserved in both classes of HMG-CoA-R.
this enzyme can be used to regulate the serum cholesterol level or Both classes of HMG-CoA-R are catalytically competent and
even in the development of novel antibiotics against certain have similar reaction mechanisms and kinetic parameters.35,37−39
pathogens. This last area of research is in line with the discovery The currently available proposal for the catalytic mechanism of
that many human pathogenic Gram-positive bacteria can HMG-CoA-R is based on kinetic and labeling experiments, site-
produce isopentoyl disphosphate (IPP) only through the directed mutagenesis, protein sequencing, and the mechanism of
mevalonate pathway, which is a necessary pathway for the dehydrogenases, which catalyze a similar reaction.40−45
bacteria to live. If the differences between the prokaryotic and The overall reaction catalyzed by these enzymes can be divided
eukaryotic HMG-CoA-S forms are taken into account, this into three main stages (Figure 4B). Once the HMG-CoA and
suggests that the inhibition of the mevalonate pathway in bacteria NADH (or NADPH) bind to the active site, the thioester of
may not affect the HMG-CoA-S enzymes of the host body. This HMG-CoA-R is reduced and the generated intermediate is
could thus lead to a new generation of antibacterial drugs that stabilized by the protonated Glu559 and Lys691 (Figure 4B,
have a significant therapeutic advantage over the commonly used steps 1 and 2). By the end of this reaction, a mevaldyl-CoA
broad-spectrum antibiotics.32 hemithioacetal intermediate is generated, and the NAD+ (or
mHMG-CoA-S also plays an important role in diabetes. The NADP+) cofactor dissociates from the active site. In the second
reaction catalyzed by this enzyme is overactivated in patients with stage of the mechanism, Glu559 assists the base-catalyzed
diabetes mellitus type I and if left untreated, because of decomposition of the hemithioacetal to produce mevaldehyde
prolonged insulin deficiency and the exhaustion of substrates and the CoA thiolate anion, which is promptly protonated by the
for gluconeogenesis, results in the shunting of excess acetyl-CoA cationic His752 (Figure 4B, step 3). In the third stage of the
into the ketone synthesis pathway and the development of catalytic mechanism, the mevaldehyde is reduced by a second
diabetic ketoacidosis. This means that under such a prognostic, NADH (or NADPH) molecule and the intermediate is again
inhibitors of this enzyme could help in the reversion of the stabilized by the protonated Glu559 and Lys691 (Figure 4B, step
process and decrease the levels of ketonic compounds in blood 3). Subsequently, the product mevalonate, CoASH, and the
serum. oxidized cofactor (NAD+ or NADP+) are released, and the
C. HMG-CoA Reductase. 3-Hydroxy-3-methyl-glutaryl- enzyme is ready for a new turnover (Figure 4B, step 5). During
CoA reductase or HMG-CoA reductase (HMG-CoA-R) the full catalytic process, Asp690 established an important
catalyzes the NADP-dependent (in mammals) or NAD- hydrogen bond with Lys691 that has been shown to be important
dependent (in prokaryotes) synthesis of mevalonate from for the catalytic process.46
5489 DOI: 10.1021/acs.biochem.6b00342
Biochemistry 2016, 55, 5483−5506
Licenciado para - Paulo Sérgio Martins dos Santos - 09609690718 - Protegido por Eduzz.com

Biochemistry Current Topic

Figure 5. (A) Structure and active-site topology of mevalonate kinase (PDB entry 1kvk71). (B) Schematic proposal for the catalytic mechanism of
mevalonate kinase.76

The reaction catalyzed by HMG-CoA-R is the point of Because of its importance, this enzyme is also a main target for
feedback control for the mevalonate pathway, which is medical intervention. Currently, this enzyme is the target of a
responsible for the biosynthesis of many important isoprenoid class of drugs called statins, which are used to treat hyper-
compounds in humans. This enzyme is also one of the most cholesterolemia and reduce the risk of cardiovascular disease.
Additionally, this enzyme has been a target for the development
regulated enzymes in our bodies.47 Dietary cholesterol exerts
of new antimicrobial agents, which take advantage of the
feedback control mostly at a translational level,48−50 but evolutionary divergence between the HMG-CoA-R of patho-
regulation mechanisms at the transcription level,51,52 enzyme genic bacteria (from class II) and that of eukaryotes (class I).
degradation,53,54 and modulation of enzyme activity55 are also Statins are potent competitive inhibitors of HMG-CoA-R57
known. and can be divided into two different types based on their
5490 DOI: 10.1021/acs.biochem.6b00342
Biochemistry 2016, 55, 5483−5506
Licenciado para - Paulo Sérgio Martins dos Santos - 09609690718 - Protegido por Eduzz.com

Biochemistry Current Topic

Figure 6. Structure of phosphomevalonate kinase (PMK) and some important active-site residues that have been identified by experimental means
(PDB entry 3CH480).

structure.58 Type I statins (lovastatin, pravastatin, and kinase (PMK), and mevalonate 5-diphosphate decarboxylase
simvastatin) are natural fungal products, whereas type II statins (MDD). According to Hogenboom and co-authors, the enzymes
are fully synthetic, characterized by the presence of larger MK, PMK, and MDD are cytosolic enzymes,66−68 but this
hydrophobic regions and attached fluorophenyl groups.59 The contradicts recent studies by Kovacs and co-authors that
ability of statins to inhibit HMG-CoA-R arises from their HMG- confirmed their previous findings of peroxisomal localization of
like moiety, which competes with HMG-CoA to bind to the the three enzymes using stable isotopic techniques and human
HMG binding site of the enzyme (see PDB entries 1HW860 and cells.10,69,70
1HMK60 in which HMG-Coa-R is bound to mevastatin and D1. Mevalonate Kinase. Mevalonate kinases (EC 2.7.1.36)
atorvastatin, respectively). Although the hydrophobic part of catalyze the transfer of the γ-phosphoryl group of ATP to the C5
statins is normally very different from that of the coenzyme A hydroxyl oxygen of mevalonic acid to form mevalonate 5-
portion of the substrate, these nonpolar groups also contribute to phosphate, a key intermediate in the biosynthetic pathway for
block the access of HMG-CoA to the active site. The affinity of isoprenoids and sterols from acetate.71,72
the enzyme for statins is slightly higher than its affinity for the The enzyme MK was discovered in the late 1950s in yeast,73
substrate.61 but it suffered from more than three decades of neglect, as
HMG-CoA-R can also be inhibited at a translational level. The research on the isoprenoid pathway was mainly focused on
oxylanosterol 15-oxa-32-vinyl lanost-8-ene-3b,32-diol is one of HMG-CoA-R. The interest in MK has been revived, however,
the inhibitors that works at this level, and it is a potent because this enzyme was found to be involved in the synthesis of
hypocholesterolemic agent.62 diverse non-sterol isoprenoid metabolites that participate in
New antimicrobial agents that target HMG-CoA-R of numerous cellular functions, e.g., protein prenylation, protein
pathogenic bacteria (class II) are also a hot topic of research in glycosylation, and cell cycle regulation. This enzyme is also
this field, in part because of the increasingly problematic broad currently being studied as a promising anticancer target because
antimicrobial resistance among Gram-negative bacteria to the certain types of cancer are particularly sensitive to mevalonate
current available drugs.63 Very few novel pharmacologic agents pathway inhibition.74 In addition, the significance of MK has
are in the research and development pipeline, and several recent been further highlighted by the implication of the enzyme in
studies have shown that some statins exhibit antibacterial human inherited diseases, such as mevalonic aciduria and
action.64,65 hyperimmunoglobulinemia D/periodic fever syndrome.
D. ATP-Dependent Enzymes Involved in the Choles- MK can be found in eukaryotes, archaebacteria, and some
terol Pathway. In animal cells, the cholesterol biosynthetic eubacteria. The mammalian enzyme is reported to be a
pathway contains a unique series of three sequential ATP- homodimer with a subunit molecular mass of 42 kDa.75
dependent enzymes that convert mevalonate to isopentenyl Kinetic studies suggest that the enzyme catalyzes an ordered
diphosphate: mevalonate kinase (MK), phosphomevalonate sequential reaction, with mevalonic acid binding prior to the
5491 DOI: 10.1021/acs.biochem.6b00342
Biochemistry 2016, 55, 5483−5506
Licenciado para - Paulo Sérgio Martins dos Santos - 09609690718 - Protegido por Eduzz.com

Biochemistry Current Topic

Figure 7. (A) Structure and active site of MDD (PDB entry 3D4J88). (B) Proposed catalytic mechanism of MDD.88−90

enzyme in relation to ATP and the phosphomevalonate being the C5 OH group of mevalonic acid.75 Ser146 and Asp204, located in
first product of the reaction.77 Site-specific mutagenesis studies a conserved glycine-rich region, have been implicated in the
have been employed to identify catalytic residues and to binding of Mg-ATP.78 Lys13 has been shown to be involved in
investigate the mechanism of the enzyme. Asp204 has been the binding of ATP as well as in facilitating catalysis79(Figure
suggested as the catalytic base that abstracts the proton from the 5A).
5492 DOI: 10.1021/acs.biochem.6b00342
Biochemistry 2016, 55, 5483−5506
Licenciado para - Paulo Sérgio Martins dos Santos - 09609690718 - Protegido por Eduzz.com

Biochemistry Current Topic

Figure 8. (A) Structure and active site of the enzyme isopentenyl pyrophosphate isomerase (PDB entry 1Q5495). (B) Proposed catalytic mechanism for
IPP isomerase.95

The reaction mechanism has not yet been deeply studied. The crystal structure of the human phosphomevalonate kinase
However, the reaction is likely to be initiated by the nucleophilic has 192 amino acid residues and a predicted molecular mass of
attack of the 5′-OH of mevalonate on the γ-phosphorus of ATP. 21.8 kDa. Despite its importance as the biosynthetic source of a
At the same time, one proton is transferred from mevalonate to diverse class of metabolites, the mechanism of PMK has not been
Lys13 and another from Lys13 to the phosphate group (Figure completely characterized. The only PDB structure of PMK that is
5B, step 1). On the basis of mutation data available for human currently available (Figure 6, PDB entry 3CH480) reveals a
MK, it is likely that Lys13 or even Asp204 may function as a positively charged cavity on the protein surface that is believed to
general base in this process and abstract a proton from the 5′-OH be essential for the binding of the phosphate group and hydroxyl
of mevalonate, thereby facilitating the nucleophilic attack of groups of the substrate. This cavity includes residues Lys48,
mevalonate.76 The Mg2+ is coordinated by Glu193 and Asp204 Arg73, Arg84, Arg110, Arg111, and Lys73.81,82 Site-directed
(being stabilized by the nearby Ser146), which are thought to be mutagenesis studies have also identified four additional amino
responsible for the activation of the γ-phosphate. acid residues, Lys17, Arg18, Lys19, and Lys22, that, if mutated,
Currently, most of the inhibitors targeting MK have in their decrease the rate of catalysis up to 10000-fold and are believed to
structure phosphate groups. For example, farnesyl diphosphate be part of the ATP binding motif.83−85 This means that these
and related compounds are potent and competitive inhibitors of residues are directly involved in the catalysis and therefore in the
MK. They bind in the active site in a manner similar to that is binding and transfer of the γ-phosphoryl group of ATP to
observed with ATP, through the same type of interactions that mevalonate 5-phosphate.
were described above (see PDB entry 2r4272). At present, no cocrystallized structure of PMK with the
D2. Phosphomevalonate Kinase. Phosphomevalonate kin- substrate, reaction intermediate, or inhibitor is available. Thus,
ase (EC 2.7.4.2) catalyzes the transfer of a second γ-phosphoryl no absolute conclusions about the catalytic mechanism can be
group from ATP to mevalonate 5-phosphate (MVAPP), which inferred. The current understanding of the mechanism considers
results in the formation of ADP and mevalonate 5-diphosphate. that bringing two phosphate groups into proximity to react is
5493 DOI: 10.1021/acs.biochem.6b00342
Biochemistry 2016, 55, 5483−5506
Licenciado para - Paulo Sérgio Martins dos Santos - 09609690718 - Protegido por Eduzz.com

Biochemistry Current Topic

especially challenging, given the high negative charge density on ase, IPP isomerase (EC 5.3.3.2)] catalyzes the conversion of the
the four phosphate groups in the active site. Therefore, only a relatively unreactive IPP to the more reactive electrophile
very specific and highly positively charged active site, like the one dimethylallyl pyrophosphate (DMAPP). Both reactants and
found in PMK, can catalyze this sort of reaction. Furthermore, products of this reaction are substrates for the successive reaction
the possible involvement of one or more magnesium ions should that results in the synthesis of farnesyl diphosphate (FPP) and,
not be discarded, as they could help the binding of the phosphate ultimately, cholesterol. This isomerization is therefore a key step
groups as well as the catalysis, like what is observed in the in the biosynthesis of isoprenoids through the mevalonate
previously described kinases. pathway.
D3. Diphosphomevalonate Decarboxylase. Diphosphome- Crystallographic studies have shown that the active form of
valonate decarboxylase, also known as mevalonate diphosphate IPP isomerase is a monomer with alternating α-helices and β-
decarboxylase (MDD, EC 4.1.1.33), catalyzes the final step of the sheets93,94 (Figure 8A). The active site of IPP isomerase is deeply
mevalonate pathway, i.e., the divalent cation-dependent buried within the enzyme and consists of a glutamic acid residue
decarboxylation of MVAPP to isopentenyl diphosphate [or (Glu116) and a cysteine residue (Cys67) that interact with
isopentenyl pyrophosphate (IPP)], with concurrent hydrolysis opposite sides of the IPP substrate, consistent with the
of ATP to form ADP and inorganic phosphate. This reaction is antarafacial stereochemistry of isomerization. The enzyme also
required for the production of polyisoprenoids and sterols from contains two metal sites: a first site occupied by Mn2+, which is
acetyl-CoA. coordinated with three histidine and two glutamate residues, and
Inhibition of this enzyme effectively diminishes the rate of a second site containing Mg2+, which is coordinated with two
biosynthesis of cholesterol,86,60 and low MDD activity correlates pyrophosphate oxygens, the carbonyl group of the highly
with decreased cholesterol levels in a hypertensive rat strain.87 conserved residue Ala67, a carboxylate oxygen of Glu87, and
Genes encoding this enzyme have been detected in archaebac- two water molecules.
teria, some eubacteria, protozoa, plants, fungi, and animals. The mechanism through which the enzyme undergoes the
The X-ray structure of human MDD shows that it consists of initial protonation step (Figure 8B, step 1) has not been
two domains (Figure 7A) and has many similarities to the conclusively established. Recent evidence suggests that Glu116 is
bacterial, protozoan, and yeast structures.88,91 This structure involved in the protonating step of the C3−C4 double bond,
revealed the presence of tightly bound PO4− and SO4− anions, forming a carbocation.95 The thiolate of Cys67 then removes a
suggesting that they might be important for the catalytic process. proton from C2, resulting in the isomerization of IPP into
Site-directed mutagenesis studies also revealed four conserved DMAPP (Figure 8B, step 2). During this process, the two metal
residues that are important for catalysis, Asp17, Ser127, Arg161, centers have an active role in catalysis. The Mg2+ is required for
and Asp305.88−90 Asp17 and Arg161 are believed to be part of the stabilization of the phosphate groups during the catalytic
the phosphoryl acceptor binding site. Asp305 and Ser127 are process, whereas the Mn2+ is actively involved in catalysis. It is
suggested to be actively involved in the base catalysis that takes also proposed that the reaction is facilitated by the conversion of
place in the active site. The correct position of the magnesium the carboxyl group of Glu116 to a carboxylate, resulting in an
ion in the active site has still not been fully established. On the electro-neutral Mn2+ with a bis-carboxylate complex. The water
basis of these studies, the catalytic mechanism of MDD has been molecules present in the active site are also proposed to be
proposed to be that illustrated in Figure 7B. involved in the catalysis, but the mechanism for how this happens
Once the substrate, MVAPP, binds in the active site, it is remains unknown.96,97
thought that it is positioned so that the terminal phosphate is F. Farnesyl Diphosphate Synthase. Farnesyl diphosphate
located near the sulfate/phosphate anions that were detected in synthase also known as farnesyl pyrophosphate synthase (FDPS,
the X-ray structure (Figure 7B, step 1). The C3 hydroxyl group of EC 2.5.1.10) is a Mg2+-dependent homodimeric enzyme,
MVAPP stands in juxtaposition to the catalytic Asp305, and the localized in peroxisomes, which catalyzes a chain elongation
ATP’s γ-phosphoryl group is oriented to facilitate the transfer of reaction and controls the first branching point of the mevalonate
the γ-phosphoryl to MVAPP, with the help of Ser127. The pathway.
proposed general base catalyst, Asp305, is believed to Chain elongation enzymes can be divided into two genetically
deprotonate MVAPP’s C3 hydroxyl in the first step of the different families depending on whether the stereochemistry of
catalytic process to facilitate the attack on the γ-phosphoryl of the newly formed double bond during each cycle of chain
ATP (Figure 7B, step 2). This reaction spurs the release of ADP elongation is E or Z. FDPS is a member of the E-double bond
and the formation of the 3-phosphoMVAPP intermediate.88 The family and catalyzes the sequential addition of IPP and DMAPP
next step of the catalytic process involves the dissociation of to form geranyl diphosphate (GPP) and then FPP.
phosphate, which generates a carbocation intermediate at C3 FDPS is essential for the post-translational prenylation of all
(Figure 7B, step 3). The positively charged carbon at position 3 small GTPase proteins that play a crucial role in cell signaling, cell
creates an electron sink that accelerates the decarboxylation proliferation, and osteoclast-mediated bone resorption. Inhib-
process with the help of Arg161. At the end of the reaction, ition of FPP production can decrease the activity of mutated H-
carbon dioxide is generated concomitant with the formation of Ras, K-Ras, and N-Ras proteins that function as major drivers of
IPP (Figure 7B, step 4). tumor growth in many cancers.98,99 Thus, the clinical benefits of
Inhibitors of MDD, in particular inhibitors of the bacterial human FDPS inhibition include both a decrease in the level of
enzyme form, are currently under development to be used as prenylation of mutated Ras proteins, leading to a decrease in the
antimicrobial agents. Several substrate analogues, such as rate of cellular growth and/or survival, and alleviation of tumor-
diphosphoglycolylproline (see PDB entry 4du892) and 6- associated bone destruction via inhibition of osteoclast
fluoromevalonate diphosphate, were shown to be competitive activity.100,101 Additionally, blocking the catalytic activity of
inhibitors for the substrate in the bacterial enzyme.92 human FDPS impacts both the downstream and upstream levels
E. Isopentenyl-diphosphate δ-Isomerase. Isopentenyl- of isoprenoids in the mevalonate pathway, leading to numerous
diphosphate δ-isomerase [or isopentenyl pyrophosphate isomer- cellular changes, including the inhibition of cholesterol biosyn-
5494 DOI: 10.1021/acs.biochem.6b00342
Biochemistry 2016, 55, 5483−5506
Licenciado para - Paulo Sérgio Martins dos Santos - 09609690718 - Protegido por Eduzz.com

Biochemistry Current Topic

Figure 9. (A) Monomeric structure and active site of farnesyl diphosphate synthase, FDPS (PDB entry 1RQJ110). (B) Catalytic mechanism of farnesyl
diphosphate synthase,107,108 FDPS. R = Me or 3-isopenteny, depending on whether FDPS catalyzes the first or second reaction.

thesis.102,103 Currently, FPPS inhibitors are mainly used in the magnesium cations, whose chelation with the substrate is
treatment of a number of bone disorders, such as Paget’s disease, required for enzyme activity.
hypercalcemia, metastatic osteolysis, and osteoporosis.104 When the crystallographic and kinetic evidence was taken into
The available X-ray structure for the human FDPS shows that account, two types of catalytic mechanisms were proposed for
the protein is a homodimer (Figure 9A).105,106 Each subunit is FDPS: those in which condensation is initiated by heterolytic
folded in a single domain whose central feature is a core cleavage of the carbon−oxygen bond of the allylic pyrophos-
composed of 10 helices that surround a large deep cleft identified phate, yielding a cationic intermediate, and those in which the
as the substrate binding pocket. The active site is located in the formation of the C1−C4 bond between the two substrates and
bottom of the cleft and comprises two conserved aspartate-rich rupture of the C1−oxygen bond is simultaneous through a
motifs.107 One includes residues Asp117, Asp118, Ile119, transition state (TS) with a carbocation character.
Met120, and Asp121 and the other residues Asp227, Asp258, Both mechanisms suggest the involvement of a dissociative
Tyr259, Leu260, and Asp261. The aspartate-rich motifs are electrophilic alkylation (Figure 9B). During the reaction, the
positioned so that the side chain carboxylate groups point into bond between C1 and its neighboring oxygen in the diphosphate
the cleft from opposite sides.108 The Asp residues in the two (in DMAPP or GPP) is cleaved to generate a resonance-
aspartate-rich motifs, except the last one in the second motif, are stabilized allylic cation, which then alkylates the double bond in
important for catalysis, while the remaining residues of the IPP to produce a tertiary carbocation. Whether this occurs in two
second motif are essential for substrate binding. These results are independent steps or in a concerted manner is still a matter of
consistent with the cocrystal structure of avian FPP in complex some controversy, but recent quantum mechanics/molecular
with GPP and IPP.109 The active site also lodges three mechanics (QM/MM) studies suggest that the latter type of
5495 DOI: 10.1021/acs.biochem.6b00342
Biochemistry 2016, 55, 5483−5506
Licenciado para - Paulo Sérgio Martins dos Santos - 09609690718 - Protegido por Eduzz.com

Biochemistry Current Topic

Figure 10. (A) Structure and active site of squalene synthase (SQS). The image of the SQS active site was built through the superimposition of two X-ray
structures: 3W7F (enzyme analogous to SQS, carotenoid dehydrosqualene synthase, which catalyzes a similar reaction)116 and 1EZF112 (human SQS).
The amino acid residues represented in panel A are from the human SQS. Only the FPP was retrieved from PDB entry 3W7F. (B) Proposed catalytic
mechanism for SQS.112,116

mechanism is more favorable.111 It is also suggested that the main responsible for the production of a metabolite that is solely
chain carbonyl oxygen of Lys202 and the side chain oxygens of committed to cholesterol synthesis.
Thr203 and Gln241 are important for stabilizing the allylic cation SQS is located in the membrane of the endoplasmic reticulum.
transition state (Figure 9A). It is anchored to it by a short C-terminal membrane-spanning
The next step involves the elimination of a proton from C2 of domain, whereas the N-terminal catalytic domain of the enzyme
the IPP unit to give rise to a new E-double bond between C2 and protrudes into the cytosol.
C3. Hosfield et al. suggest that the non-metal-ligated Mammalian forms of SQS are approximately 47 kDa and
pyrophosphate oxygen is the catalytic base that deprotonates consist of approximately 416 amino acids. The crystal structure
the condensed intermediate to generate the C5-extended of human SQS was determined in 2000112 and revealed that the
protein was composed entirely of α-helices. The active site of this
isoprenoid reaction product and that Arg116 and Lys258
enzyme is located in a large channel that is found in the middle of
might be important for such reaction.110 The new allylic
the protein. One end of the channel is open to the cytosol, from
diphosphate is then generated, and it is one isoprene unit longer
where the substrates are obtained (soluble allylic compound
than the initial substrate. The role of the magnesium cations containing 15 carbon atoms). The other end forms a hydro-
present in the active site is believed to be the stabilization of the phobic pocket that allows the product of the catalytic process to
pyrophosphate groups and, at the same time, the enhancement of reach the membrane environment (squalene, an insoluble
the rate of the reaction. compound with 30 carbon atoms) (Figure 10A).
G. Squalene Synthase. Squalene synthase or farnesyl- The detailed mechanism by which SQS operates has not been
diphosphate:farnesyl-diphosphate farnesyl transferase (SQS, EC fully resolved. The uniqueness of the head-to-head coupling of
2.5.1.21) catalyzes an unusual head-to-head reductive dimeriza- two FPP molecules to form squalene via a stable cyclo-
tion of two molecules of FPP to form squalene. This enzyme is of propylcarbinyl diphosphate intermediate has elicited much
particular importance as it is the first enzyme in the pathway mechanistic speculation over the years.113,114 To shed some
5496 DOI: 10.1021/acs.biochem.6b00342
Biochemistry 2016, 55, 5483−5506
Licenciado para - Paulo Sérgio Martins dos Santos - 09609690718 - Protegido por Eduzz.com

Biochemistry Current Topic

light on the catalytic mechanism of SQS, several site-directed inhibitor of SQS, lapaquistat, went through clinical trials but
mutagenesis experiments on diverse amino acid residues of the failed because of potential hepatic safety issues.123
SQS active site were conducted. The results showed that SQS was also mentioned as an interesting chemotherapeutic
mutations of Phe288 prevent the enzyme from catalyzing the target against Trypanosoma cruzi and Leishmania parasites, which
second reaction, in which the cyclopropyl intermediate is are responsible for large parasitic disease loads and socio-
rearranged to produce squalene.115 On the other hand, economic losses, particularly in developing countries.124 Specific
mutations at Tyr171 make the enzyme inactive. This result chemotherapy of the diseases caused by these protozoa remains
suggests that Tyr171 is required for the first half-reaction, the unsatisfactory, as currently available drugs have limited activity,
cyclopropanation. Because tyrosine is an aromatic residue and as well as frequent toxic side effects and rising drug resistance.
has a hydroxyl group, a process in which Tyr171 loses a proton to However, these parasites have a strict requirement for specific
the leaving pyrophosphate group was proposed. This would turn endogenous sterols (ergosterol and analogues) for survival and
Tyr171 more negative, which in turn would stabilize the growth and cannot use the abundant supply of cholesterol
carbocation intermediate that is generated during the reaction. present in their mammalian host.125,126 Further studies led to the
In 2000 and with the release of the SQS structure,112 this discovery of E5700 and ER-119884, two novel quinuclidine SQS
hypothesis developed greater consensus, as Tyr171 was found to inhibitors under development as cholesterol- and triglyceride-
be in the binding pocket. Also, likely to play a key role in catalysis lowering agents in humans by Eisai Co., which have very potent
are the conserved Tyr73 and Gln212. In addition, three anti-T. cruzi activity in vitro. One of them (E5700) was able to
magnesium ions are important for the catalysis and might play provide full protection against death, and it completely arrested
a major role in the stabilization and orientation of the two FPP development of parasitaemia in a murine model of acute disease
molecules in the active site of SQS. when administered orally. This was the first report of an orally
In 2010, the crystal structure of a very similar enzyme, active SQS inhibitor as an anti-infective agent.127,128 Although
dehydrosqualene synthase (crtM), was crystallized with a these compounds and other aryl-quinuclidines are also potent
substrate analogue, farnesyl thiopyrophosphate (PDB entry inhibitors of mammalian SQS, their selective antiparasitic activity
3W7F116) and the intermediate PSDP (PDB entry 3NPR117). in vitro and in vivo might be explained by the capacity of a host’s
Superimposition of the substrate and intermediate in the SQS cells to compensate for the blockage of de novo cholesterol
structure revealed that one of the substrate molecules remains in synthesis by upregulating the expression of LDL receptors and
the same position during the first half-reaction and keeps the capturing this sterol from the growth medium or serum.129,130 In
pyrophosphate moiety (Figure 10A). contrast, there is no way for the parasite to compensate in this
manner for the quinuclidine-induced blockage of ergosterol
On the basis of the available data, it is suggested that the
biosynthesis, because there are no appreciable amounts of
reaction catalyzed by SQS proceeds in two distinct steps, both of
ergosterol in host cells or growth media.
which involve the formation of carbocationic reaction
H. Squalene Monooxygenase. Squalene monooxygenase
intermediates (Figure 10B). In the first half-reaction, two
(SM, EC 1.14.13.132), also known as squalene epoxidase, is a 64
identical molecules of FPP bind in distinct regions of the active
kDa flavin adenine dinucleotide (FAD)-containing enzyme that
site of SQS. The pyrophosphate group of one of the FPPs
catalyzes the first oxygenation step of cholesterol synthesis. The
(normally designated as donor FPP) is then cleaved, and an reaction requires NADPH and molecular oxygen to oxidize
allylic carbocation intermediate is generated concomitant with squalene to 2,3-oxidosqualene (squalene epoxide).
the release of pyrophosphate (Figure 10B, step 1). The SM is bound to the endoplasmic reticulum of eukaryotic cells
carbocation promptly reacts with the C-2,3 double bond of the and belongs to the flavoprotein monooxygenase family, which
acceptor FPP in a 1′,2,3-prenyltransferase reaction that is catalyzes a wide variety of oxidative reactions.33,131 However,
accompanied by the loss of a proton (Figure 10B, step 2). The there are several differences between squalene monooxygenase
product of this condensation is presqualene pyrophosphate and other known flavin monooxygenases. First, SM catalyzes
(PSPP), a stable cyclopropylcarbinyl pyrophosphate intermedi- epoxidation but not hydroxylation of substrates. Second, it does
ate that remains associated with SQS for the second reaction not contain cytochrome P450 as a prosthetic group and does not
(Figure 10B, step 3). bind NADPH directly. Instead, the electrons that are required for
In the second half-reaction of SQS, PSPP loses the the reaction are passed from NADPH, via cytochrome P450
pyrophosphate group and the resulting cyclopropylcarbinyl reductase, to the loosely bound FAD group of squalene
carbocation undergoes a ring opening and reduction by NADPH monooxygenase. These differences turn SM into an attractive
(through a hydride transfer) to the linear and final product, target for the treatment of hypercholesterolemia, while not
squalene (Figure 10B, steps 4−6). SQS then releases squalene disturbing other flavin monooxygenase-dependent processes.
into the membrane of the endoplasmic reticulum (Figure 10B, SM was first identified in the early 1970s, but studies of the
step 7). human enzyme began only in the 1990s. The mammalian SM
From a metabolic perspective, SQS has a pivotal role as it takes showed limited sequence similarity to that found in yeast.
FPP from the more general mevalonate pathway to commit it to Interestingly, it has also been shown that there are substantial
cholesterol synthesis.118 This stimulated interest in using SQS as differences between the mammalian enzymes from different
a drug target for the treatment of hypercholesterolemia. species,132,133 revealing that they might be specific to each
Currently, strong inhibitors of SQS have been found, such as species. Unfortunately, structural details of these enzymes from
zaragozic acid A,119,120 lapaquistat (recently discontinued from their X-ray diffraction studies are still not available, which
clinical development),121 and DF-461.122 Several authors suggest impedes further analysis.
that the use of SQS in the treatment of hypercholesterolemia The reaction catalyzed by squalene monooxygenase is highly
could have fewer side effects than the classical statin treatments, specific.134 It involves insertion of an oxygen atom across a
which inhibit the whole mevalonate pathway by targeting HMG- carbon−carbon double bond to form an epoxide. Flavoprotein
CoA-R. At the moment this is still speculative, because only one monooxygenases accomplish this oxygenation by forming a
5497 DOI: 10.1021/acs.biochem.6b00342
Biochemistry 2016, 55, 5483−5506
Licenciado para - Paulo Sérgio Martins dos Santos - 09609690718 - Protegido por Eduzz.com

Biochemistry Current Topic

Figure 11. (A) Structure of the enzyme and of the active site of OSC (PDB entry 1w6k). (B) Catalytic mechanism catalyzed by OSC.

flavin hydroperoxide at the enzyme active site, which then oxygenase presumably utilizes this same mechanism but differs
transfers the terminal oxygen atom of the hydroperoxide (OH) from other known flavin monooxygenases in that the oxygen is
to the substrate.135 The remaining “hydroxyflavin” then inserted as an epoxide rather than as a hydroxyl group. Another
reoxidizes, with the release of a water molecule and the thing that differentiates these enzymes is the fact that squalene
concomitant formation of 2,3-oxidosqualene. Squalene mono- monooxygenase contains a loosely bound FAD flavin and obtains
5498 DOI: 10.1021/acs.biochem.6b00342
Biochemistry 2016, 55, 5483−5506
Licenciado para - Paulo Sérgio Martins dos Santos - 09609690718 - Protegido por Eduzz.com

Biochemistry Current Topic

electrons from NADPH-cytochrome P450 reductase, rather than OSC can be divided into three main stages. The mechanism
binding the nicotinamide cofactor NADPH directly.136 starts with the adoption of a preorganized conformation by 2,3-
Although there is a lack of information regarding the structure monoepoxysqualene, followed by protonation of the epoxide
and biochemistry of SM, this enzyme has been extensively ring. A cascade of reactions is then triggered, resulting in an array
exploited for antifungal drug development.137,138 Currently, of ring-forming reactions followed by a series of 1,2-hydride and
several fungal SM inhibitors, such as terbinafine, naftifine, and 1,2-methyl group shifts and a final deprotonation step, from
numerous related compounds, are currently on the market or which results lanosterol (Figure 11B). All reactions proceed with
under investigation. Some of the best compounds are precise stereo- and regiospecificity required for the formation of
Terbinafine (Lamisil) and Naftifine (Naftin), which show good the multi C−C bonds.
specificity for the fungal enzyme without inhibiting human The first stage of the reaction is assisted by Asp455, which
squalene monooxygenase.139 Both are fungicidal, interfering with protonates the epoxide of the substrate (Figure 11B, step 1). The
cell membrane synthesis and preventing growth. acidic character of this residue is enhanced by the neighboring
Because this enzyme catalyzes the second committed (and Cys465 and Cys533 with which it interacts through two
likely rate-limiting) step in cholesterol biosynthesis, it has also hydrogen bonds. The second stage of the mechanism was
become an attractive pharmacotherapeutic target in the treat- initially thought to involve the sequential formation of the four
ment of hypercholesterolemia and resultant cardiovascular rings (named A−D) in a concerted manner.145−153 Corey et al.
diseases. Several preclinical studies suggest an effective proposed that the formation of the A-ring should be concomitant
hypocholesterolemic activity, comparable to or even better with the protonation of the epoxide, in a concerted SN2
than that which can be achieved by HMG-CoA-R inhibition. substitution-like process. This proposal was confirmed very
The most effective inhibitor of mammalian SM known to date recently by QM/MM calculations.154 The same calculations have
is NB-598, developed at Banyu Pharmaceutical Co.140 This also suggested that the formation of the B-ring is concerted with
fungally derived natural compound is a competitive inhibitor of this step, and that the three concomitant reactions make up the
SM in human HepG2 cells and effectively reduces the level of rate-limiting step of the full catalytic process (Figure 11B, step 2).
serum cholesterol in dogs with no apparent adverse effects.141 In Corey and co-workers also proposed that formation of rings C
a comparative study in dogs, NB-598 was more potent than the and D involved discrete carbocation intermediates. In particular,
HMG-CoA-R inhibitor simvastatin, decreasing total and serum they showed that the formation of ring C involves a
LDL cholesterol levels. NB-598 also decreased triacylglycerol cyclopentylcarbinyl carbocation intermediate that subsequently
levels, an effect not observed with the HMG-CoA-R undergoes a ring expansion to form the six-membered C-ring. It
simvastatin.141 No studies have yet been reported in humans. is also proposed that the closure of the D-ring is concerted with
I. 2,3-Oxidosqualene Cyclase-Lanosterol Synthase. 2,3- the formation of the C-ring155,156 (Figure 11B, steps 3′ and 4′).
Oxidosqualene cyclase-lanosterol synthase (OSC, EC 5.4.99.7) The only drawback of this proposal is the formation of ring C,
is a 78 kDa membrane-bound enzyme that catalyzes the which does not follow the Markovnikov rule and, therefore, does
conversion of the acyclic compound 2,3-oxidosqualene (OS) not ensure the formation of a stable carbocation during the
to the cyclic lanosterol. Both oxidosqualene and lanosterol are addition reactions. In 2002, Hess presented another hypothesis
mostly hydrocarbons and thus are not very soluble in water. The for C- and D-ring formation.146 The double bond between C18
enzyme solves this problem by sticking to the membrane in and C19 of squalene might be involved anchimerically in the
peroxisomes. It then can pull oxidosqualene directly out of the cyclopentylcarbinylcyclohexyl ring expansion, meaning that the
membrane and release lanosterol back there. The reaction expansion of the C-ring and formation of the D-ring are
catalyzed by this enzyme is considered to be one of the most concerted. This mechanism avoids the formation of the stable
complex reactions that is catalyzed in the enzymatic world, as it cyclohexyl carbocation intermediate as well as the violation of
involves a sequence of cyclization and 1,2-group rearrangements Markovnikov’s rule (Figure 11B, steps 3−5). Although this
of high-energy carbocations to lanosterol, with complete mechanism is more favorable than the previous one, there is still
structural and stereochemical control of seven chiral centers.142 no certainty about which one actually occurs. The only detail that
OSC is almost exclusively found in eukaryotes, apart from a seems clear is that the formation of rings C and D is controlled by
few prokaryotes that can produce this enzyme.143 Human OSC is a set of intramolecular forces (cation stabilization and hydrogen
a monomeric enzyme that it is located in the membrane of the bonding) that are imposed by some active-site residues to the
endoplasmic reticulum.144 OSC is composed by two α-helix substrate and are located along the active-site tunnel. Muta-
barrel domains (domain 1 and domain 2) connected by loops genesis studies have revealed that Tyr704, Tyr707, Tyr587, and
and three smaller β-sheet structures. The active-site cavity is His234 have an important role in this context.157
located in the middle of the protein, between domains 1 and 2 Once the formation of the D-ring is accomplished, a cation
(Figure 11A).142 Domain 2 contains a region inserted into the intermediate is obtained, which undergoes a series of 1,2-methyl
membrane and forms a tunnel that allows the substrate to enter and hydride shifts (Figure 11B, steps 6−9). The last step of the
the enzyme and reach the active-site cavity. Although the catalytic cycle involves a proton abstraction that is catalyzed by
architecture of the channel is clear in the available X-ray Tyr503 (Figure 11B, step 10). At the end of this step, the product
structure, its role in enzyme−subtract interaction is still far from of the reaction, lanosterol, is obtained and the enzyme is ready for
being understood. a new turnover (Figure 11B, step 11).
The catalytic mechanism of OSC has been a subject of Interest in OSC has grown in recent years, mostly because of
numerous studies in the past four decades.142,145 In spite of these its late position in the cholesterol biosynthesis, which means it
efforts, much remains to be learned about how OSC mediates the can be an optimal target in the fight against hypercholester-
individual cyclization and rearrangement steps. Currently, there olemia. This interest is partially motivated by the adverse effects
is still great speculation about the mechanism of OSC, and of statin that inhibit HMG-CoA-R and may cause myopathy.
several contradictory proposals are present in the literature. In However, until now, no OSC inhibitor has been approved as a
general, all the authors agree that the catalytic mechanism of clinical drug for the treatment of hypercholesterolemia.
5499 DOI: 10.1021/acs.biochem.6b00342
Biochemistry 2016, 55, 5483−5506
Licenciado para - Paulo Sérgio Martins dos Santos - 09609690718 - Protegido por Eduzz.com

Biochemistry Current Topic

Figure 12. Enzymes involved in the catalysis of cholesterol from lanosterol.

However, some OSC inhibitors have already been described and different routes, both of which end with a cholesterol molecule.
cocrystallized in the active center of a close homologue of OSC. They are termed the Bloch pathway and the Kandutsch−Russell
One in particular, named Ro 48-8071, is one of the most popular pathway. The difference between them is that the first uses Δ24-
among OSC inhibitors, being cocrystallized in both the human unsaturated sterols while in the second the intermediates have
OSC enzyme (PDB entry 1w6j142) and the homologous SHC their side chain saturated.
enzyme (PDB entry 3SQC158). It acts on human OSC as a To be transformed into a cholesterol molecule, lanosterol has
competitive inhibitor, preventing the binding of the oxidosqua- to undergo a series enzymatic reactions (Figure 12). First, C14
lene. undergoes a two-step demethylation process, which is catalyzed
J. From Lanosterol to Cholesterol. The conversion of by the enzymes C14α-demethylase and Δ14-reductase. This is
lanosterol into cholesterol is a very complex and multistep followed by two subsequent demethylations at C4, mediated by
pathway, which involves several enzymes. After squalene is C4-demethylase. The next step is the isomerization of the double
transformed into lanosterol, this molecule can follow two bond at Δ8 to Δ7, a reaction catalyzed by Δ8-Δ7-isomerase.
5500 DOI: 10.1021/acs.biochem.6b00342
Biochemistry 2016, 55, 5483−5506
Licenciado para - Paulo Sérgio Martins dos Santos - 09609690718 - Protegido por Eduzz.com

Biochemistry Current Topic

Subsequently, a desaturation occurs between C5 and C6 of statins, important drugs that lower blood cholesterol levels and
(catalyzed by Δ5-desaturase) followed by the reduction of two treat cardiovascular diseases. However, this enzyme acts very
double bonds, the first at Δ7 and the second at Δ24. The enzyme early in the cholesterol synthesis pathway, with nearly 20
Δ24-reductase can convert any intermediate on the Bloch subsequent enzymes needed to produce cholesterol. SM and
pathway into its unsaturated counterpart, although it has SQS are enzymes that are downstream from HMG-CoA-R and
different affinities for them, and the conversion will shift to the located in the final branching point of cholesterol biosynthesis.
Kandutsch−Russell pathway. However, after this route is chosen, The inhibitors for these enzymes are not as well developed as the
it cannot revert to the previous one. There is a great lack of ones for HMG-CoA-R, but currently available studies show very
structural and mechanistic information about this final stage of promising results in reducing levels of cholesterol. In fact,
the cholesterol biosynthetic pathway, and for that reason, it is preclinical studies suggest that the inhibition of SM has
premature to explore it in this review. hypocholesterolemic activity comparable to, or even better

■ CONCLUSIONS AND FUTURE PERSPECTIVES


The complexities of the structure and the biosynthesis of
than, that obtained with HMG-CoA-R inhibition. Studies
involving OSC also show a direct decrease in the level of
lanosterol formation and an inherent decrease in HMG-CoA
cholesterol have spanned decades of research and been the reductase activity. The great advantage of these inhibitors in
subject of research that has been recognized with several Nobel relation to the ones that target HMG-CoA-R is the absence of
prizes. It is perhaps the only molecule whose studies have paved myopathic side effects, one of the major drawbacks of statins.
the way for the discovery of several important biomedical It is also worth mentioning that there are currently other
benefits. After one century of research, significant advances have efficient methods that can be used to treat hypercholesterolemia.
taken place, and our current knowledge regarding the enzymes For example, the bile acid binding sequestrant cholestyramine
involved in steroid hormone biosynthesis has improved has been used for several decades to treat hypercholesterolemia,
substantially. This means that we are now closer to deciphering even before statins became available on the market.165 The
the full mechanistic puzzle behind cholesterol biosynthesis. cholesterol absorption inhibitor ezetimibe can also be used to the
Currently, all the enzymes involved in the biosynthesis of same end, alone or combined with statins.166,167 Recently, it was
cholesterol have already been identified, and the three-dimen- also found that inhibitors of the proprotein convertase subtilisin/
sional structures of many of them have been deciphered and kexin9 (PCSK9) monoclonal antibody can be used effectively to
intensively studied. In this review, all the mechanistic knowledge treat hypercholesterolemia.168,169 The mechanism of action of
regarding the majority of the enzymes involved in the these drugs is still poorly understood, but it is believed that they
biosynthesis of the cholesterol molecule, starting from acetyl- might not interact directly with the enzymes involved in
CoA, was explored and discussed. The structure of each enzyme cholesterol biosynthesis.
involved in this process and a detailed description of their active All of these new compounds and methods hold great promise
site were scrutinized. Particular attention was given to the and may become the next generation of drugs that will be used to
catalytic mechanism of each enzyme, and the chemistry behind reduce low-density lipoprotein cholesterol levels, possibly in a
future not far from today.


the transformations from the reactants into the products was
carefully described. The chemistry of these transformations is
extremely challenging, involving, in the many cases, molecules AUTHOR INFORMATION
that are very hydrophobic. Such a rich chemistry is only possible Corresponding Author
because of the involvement of remarkable enzymes that allow the *E-mail: pafernan@fc.up.pt.
binding of more than one substrate in their active sites, stabilize Funding
uncommon intermediates, manage the full stereochemical N.M.F.S.A.C. is grateful to the Fundacao para a Ciencia e
control of chiral centers, etc. The mechanisms through which Technologia (FCT), Portugal, for program Ciência 2007. This
these enzymes achieve the enormous rate enhancements and work has been funded by FEDER/COMPETE and Fundaçaõ
exquisite specificity are also diverse. For instance, the first three para a Ciência e a Tecnologia Grants PTDC/QUI-QUI/
enzymes involved in cholesterol biosynthesis, ACAT, HMG- 102760/2008, PEst-C/EQB/LA0006/2011, IF/01310/2013,
CoA-S, and HMG-CoA-R, follow a ping-pong type of and EXCL/QEQ-COM/0394/2012.
mechanism involving the formation of tetrahedral intermediates Notes
with active-site residues. The next three enzymes, MK, PMK, and The authors declare no competing financial interest.
MDD, are ATP-dependent and involve the exchange of
phosphoryl groups. The enzymes FDPS and SQS follow a
sequential addition of substrates. SM catalyzes the oxygenation
■ REFERENCES
(1) Gordon, D. J., Probstfield, J. L., Garrison, R. J., Neaton, J. D.,
of a squalene, and OSC isomerizes oxidosqualene to lanosterol. Castelli, W. P., Knoke, J. D., Jacobs, D. R., Jr., Bangdiwala, S., and
In the quest to unravel the catalytic mechanism of the enzymes Tyroler, H. A. (1989) High-density lipoprotein cholesterol and
involved in the cholesterol pathway, theoretical and computa- cardiovascular disease. Four prospective American studies. Circulation
tional methods are beginning to play an important role, because 79, 8−15.
they can predict and tackle the formation of reaction (2) Briel, M., Ferreira-Gonzalez, I., You, J. J., Karanicolas, P. J., Akl, E.
intermediates that are difficult to detect by experimental A., Wu, P., Blechacz, B., Bassler, D., Wei, X. G., Sharman, A., Whitt, I.,
means, as observed in other biological systems.159−164 Some of Alves da Silva, S., Khalid, Z., Nordmann, A. J., Zhou, Q., Walter, S. D.,
Vale, N., Bhatnagar, N., O’Regan, C., Mills, E. J., Bucher, H. C., Montori,
these proposals were validated later by experimental means, V. M., and Guyatt, G. H. (2009) Association between change in high
while others were used to explore new mechanistic var- density lipoprotein cholesterol and cardiovascular disease morbidity and
iants.146,154 mortality: systematic review and meta-regression analysis. BMJ [Br. Med.
From all the enzymes described in cholesterol biosynthesis, J.] 338, b92.
HMG-CoA-R, SQS, and OSC are perhaps the most important, (3) Libby, P. (2002) Inflammation in atherosclerosis. Nature 420,
from a pharmacological point of view. HMG-CoA-R is the target 868−874.

5501 DOI: 10.1021/acs.biochem.6b00342


Biochemistry 2016, 55, 5483−5506
Licenciado para - Paulo Sérgio Martins dos Santos - 09609690718 - Protegido por Eduzz.com

Biochemistry Current Topic

(4) Aarden, E., Van Hoyweghen, I., and Horstman, K. (2011) The (20) Hegardt, F. G. (1999) Mitochondrial 3-hydroxy-3-methylglu-
paradox of public health genomics: Definition and diagnosis of familial taryl-CoA synthase: a control enzyme in ketogenesis. Biochem. J. 338,
hypercholesterolaemia in three European countries. Scand J. Public Healt 569−582.
39, 634−639. (21) Olivier, L. M., and Krisans, S. K. (2000) Peroxisomal protein
(5) Newby, L. K., Kandzari, D., Roe, M. T., Mulgund, J., Bhatt, D. L., targeting and identification of peroxisomal targeting signals in
DeLong, E., Ohman, E. M., Gibler, W. B., and Peterson, E. D. (2005) cholesterol biosynthetic enzymes. Biochim. Biophys. Acta, Mol. Cell
Examining the hypercholesterolemia paradox in acute coronary Biol. Lipids 1529, 89−102.
syndromes: Results from CRUSADE. Circulation 111, E329−E329. (22) Olivier, L. M., Kovacs, W., Masuda, K., Keller, G. A., and Krisans,
(6) Hussain, M. M., Strickland, D. K., and Bakillah, A. (1999) The S. K. (2000) Identification of peroxisomal targeting signals in cholesterol
mammalian low-density lipoprotein receptor family. Annu. Rev. Nutr. 19, biosynthetic enzymes: AA-CoA thiolase, HMG-CoA synthase, MPPD,
141−172. and FPP synthase. J. Lipid Res. 41, 1921−1935.
(7) Thompson, S., Mayerl, F., Peoples, O. P., Masamune, S., Sinskey, A. (23) Sutherlin, A., Hedl, M., Sanchez-Neri, B., Burgner, J. W.,
J., and Walsh, C. T. (1989) Mechanistic studies on beta-ketoacyl thiolase Stauffacher, C. V., and Rodwell, V. W. (2002) Enterococcus faecalis 3-
from Zoogloea ramigera: identification of the active-site nucleophile as hydroxy-3-methylglutaryl coenzyme A synthase, an enzyme of
Cys89, its mutation to Ser89, and kinetic and thermodynamic isopentenyl diphosphate biosynthesis. J. Bacteriol. 184, 4065−4070.
characterization of wild-type and mutant enzymes. Biochemistry 28, (24) Shafqat, N., Turnbull, A., Zschocke, J., Oppermann, U., and Yue,
5735−5742. W. W. (2010) Crystal Structures of Human HMG-CoA Synthase
(8) Modis, Y., and Wierenga, R. K. (2000) Crystallographic analysis of Isoforms Provide Insights into Inherited Ketogenesis Disorders and
the reaction pathway of Zoogloea ramigera biosynthetic thiolase. J. Mol. Inhibitor Design. J. Mol. Biol. 398, 497−506.
Biol. 297, 1171−1182. (25) Misra, I., Narasimhan, C., and Miziorko, H. M. (1993) Avian 3-
(9) Antonenkov, V. D., Van Veldhoven, P. P., Waelkens, E., and Hydroxy-3-Methylglutaryl-Coa Synthase - Characterization of a
Mannaerts, G. P. (1997) Substrate specificities of 3-oxoacyl-CoA Recombinant Cholesterogenic Isozyme and Demonstration of the
thiolase A and sterol carrier protein 2/3-oxoacyl-CoA thiolase purified Requirement for a Sulfhydryl Functionality in Formation of the Acetyl-
from normal rat liver peroxisomes. Sterol carrier protein 2/3-oxoacyl- Enzyme Reaction Intermediate. J. Biol. Chem. 268, 12129−12135.
CoA thiolase is involved in the metabolism of 2-methyl-branched fatty (26) Chun, K. Y., Vinarov, D. A., and Miziorko, H. M. (2000) 3-
acids and bile acid intermediates. J. Biol. Chem. 272, 26023−26031. Hydroxy-3-methylglutaryl-CoA synthase: participation of invariant
(10) Kovacs, W. J., Tape, K. N., Shackelford, J. E., Duan, X. Y., acidic residues in formation of the acetyl-S-enzyme reaction
Kasumov, T., Kelleher, J. K., Brunengraber, H., and Krisans, S. K. (2007) intermediate. Biochemistry 39, 14670−14681.
Localization of the pre-squalene segment of the isoprenoid biosynthetic (27) Miziorko, H. M., and Lane, M. D. (1977) 3-Hydroxy-3-
pathway in mammalian peroxisomes. Histochem. Cell Biol. 127, 273− methylgutaryl-CoA synthase. Participation of acetyl-S-enzyme and
290. enzyme-S-hydroxymethylgutaryl-SCoA intermediates in the reaction.
(11) Tyni, T., Palotie, A., Viinikka, L., Valanne, L., Salo, M. K., von J. Biol. Chem. 252, 1414−1420.
Dobeln, U., Jackson, S., Wanders, R., Venizelos, N., and Pihko, H. (28) Theisen, M. J., Misra, I., Saadat, D., Campobasso, N., Miziorko, H.
(1997) Long-chain 3-hydroxyacyl-coenzyme A dehydrogenase defi- M., and Harrison, D. H. (2004) 3-hydroxy-3-methylglutaryl-CoA
ciency with the G1528C mutation: clinical presentation of thirteen synthase intermediate complex observed in ″real-time″. Proc. Natl.
patients. J. Pediatr. 130, 67−76. Acad. Sci. U. S. A. 101, 16442−16447.
(12) Kamijo, T., Indo, Y., Souri, M., Aoyama, T., Hara, T., Yamamoto, (29) Miziorko, H. M., Clinkenbeard, K. D., Reed, W. D., and Lane, M.
S., Ushikubo, S., Rinaldo, P., Matsuda, I., Komiyama, A., and Hashimoto, D. (1975) 3-Hydroxy-3-methylglutaryl coenzyme A synthase. Evidence
T. (1997) Medium chain 3-ketoacyl-coenzyme A thiolase deficiency: a for an acetyl-S-enzyme intermediate and identification of a cysteinyl
new disorder of mitochondrial fatty acid beta-oxidation. Pediatr. Res. 42, sulfhydryl as the site of acetylation. J. Biol. Chem. 250, 5768−5773.
569−576. (30) Bahnson, B. J. (2004) An atomic-resolution mechanism of 3-
(13) Hartlage, P., Eller, G., Carter, L., Roesel, A., and Hommes, F. hydroxy-3-methylglutaryl-CoA synthase. Proc. Natl. Acad. Sci. U. S. A.
(1986) Mitochondrial acetoacetyl-CoA thiolase deficiency. Biochem. 101, 16399−16400.
Med. Metab. Biol. 36, 198−206. (31) Pojer, F., Ferrer, J. L., Richard, S. B., Nagegowda, D. A., Chye, M.
(14) Fukao, T., Nakamura, H., Song, X. Q., Nakamura, K., Orii, K. E., L., Bach, T. J., and Noel, J. P. (2006) Structural basis for the design of
Kohno, Y., Kano, M., Yamaguchi, S., Hashimoto, T., Orii, T., and Kondo, potent and species-specific inhibitors of 3-hydroxy-3-methylglutaryl
N. (1998) Characterization of N93S, I312T, and A333P missense CoA synthases. Proc. Natl. Acad. Sci. U. S. A. 103, 11491−11496.
mutations in two Japanese families with mitochondrial acetoacetyl-CoA (32) Marrakchi, H., Patel, D., and Rock, C. (2001) Regulation of fatty
thiolase deficiency. Hum. Mutat. 12, 245−254. acid biosynthesis and degradation in Escherichia coli. FASEB J. 15,
(15) Harijan, R. K., Kiema, T. R., Karjalainen, M. P., Janardan, N., A192−A192.
Murthy, M. R., Weiss, M. S., Michels, P. A., and Wierenga, R. K. (2013) (33) Mazein, A., Watterson, S., Hsieh, W. Y., Griffiths, W. J., and
Crystal structures of SCP2-thiolases of Trypanosomatidae, human Ghazal, P. (2013) A comprehensive machine-readable view of the
pathogens causing widespread tropical diseases: the importance for mammalian cholesterol biosynthesis pathway. Biochem. Pharmacol. 86,
catalysis of the cysteine of the unique HDCF loop. Biochem. J. 455, 119− 56−66.
130. (34) Hampton, R., DimsterDenk, D., and Rine, J. (1996) The biology
(16) Arnesen, T., Thompson, P. R., Varhaug, J. E., and Lillehaug, J. R. of HMG-CoA reductase: The pros of contra-regulation. Trends Biochem.
(2008) The Protein Acetyltransferase ARD1: A Novel Cancer Drug Sci. 21, 140−145.
Target? Curr. Cancer Drug Targets 8, 545−553. (35) Istvan, E. S., Palnitkar, M., Buchanan, S. K., and Deisenhofer, J.
(17) Modis, Y., and Wierenga, R. K. (1999) A biosynthetic thiolase in (2000) Crystal structure of the catalytic portion of human HMG-CoA
complex with a reaction intermediate: the crystal structure provides new reductase: insights into regulation of activity and catalysis. EMBO J. 19,
insights into the catalytic mechanism. Structure 7, 1279−1290. 819−830.
(18) Merilainen, G., Poikela, V., Kursula, P., and Wierenga, R. K. (36) Gesto, D. S., Cerqueira, N. M., Ramos, M. J., and Fernandes, P. A.
(2009) The thiolase reaction mechanism: the importance of Asn316 and (2014) Discovery of new druggable sites in the anti-cholesterol target
His348 for stabilizing the enolate intermediate of the Claisen HMG-CoA reductase by computational alanine scanning mutagenesis. J.
condensation. Biochemistry 48, 11011−11025. Mol. Model. 20, 2178.
(19) Bahnson, B. J. (2004) An atomic-resolution mechanism of 3- (37) Tabernero, L., Bochar, D. A., Rodwell, V. W., and Stauffacher, C.
hydroxy-3-methylglutaryl-CoA synthase. Proc. Natl. Acad. Sci. U. S. A. V. (1999) Substrate-induced closure of the flap domain in the ternary
101, 16399−16400. complex structures provides insights into the mechanism of catalysis by

5502 DOI: 10.1021/acs.biochem.6b00342


Biochemistry 2016, 55, 5483−5506
Licenciado para - Paulo Sérgio Martins dos Santos - 09609690718 - Protegido por Eduzz.com

Biochemistry Current Topic

3-hydroxy-3-methylglutaryl-CoA reductase. Proc. Natl. Acad. Sci. U. S. A. (56) Haines, B. E., Wiest, O., and Stauffacher, C. V. (2013) The
96, 7167−7171. Increasingly Complex Mechanism of HMG-CoA Reductase. Acc. Chem.
(38) Rodwell, V. W., Beach, M. J., Bischoff, K. M., Bochar, D. A., Res. 46, 2416−2426.
Darnay, B. G., Friesen, J. A., Gill, J. F., Hedl, M., Jordan-Starck, T., (57) Brown, M. S., Faust, J. R., Goldstein, J. L., Kaneko, I., and Endo, A.
Kennelly, P. J., Kim, D., and Wang, Y. L. (2000) 3-hydroxy-3- (1978) Induction of 3-hydroxy-3-methylglutaryl coenzyme A reductase
methylglutaryl-CoA reductase. Methods Enzymol. 324, 259−280. activity in human fibroblasts incubated with compactin (ML-236B), a
(39) Istvan, E. S., and Deisenhofer, J. (2000) The structure of the competitive inhibitor of the reductase. J. Biol. Chem. 253, 1121−1128.
catalytic portion of human HMG-CoA reductase. Biochim. Biophys. Acta, (58) Istvan, E. S., and Deisenhofer, J. (2001) Structural mechanism for
Mol. Cell Biol. Lipids 1529, 9−18. statin inhibition of HMG-CoA reductase. Science 292, 1160−1164.
(40) Veloso, D., Cleland, W. W., and Porter, J. W. (1981) Ph Properties (59) Tobert, J. A. (2003) Lovastatin and beyond: the history of the
and Chemical Mechanism of Action of 3-Hydroxy-3-Methylglutaryl HMG-CoA reductase inhibitors. Nat. Rev. Drug Discovery 2, 517−526.
Coenzyme-a Reductase. Biochemistry 20, 887−894. (60) Istvan, E. S., and Deisenhofer, J. (2001) Structural mechanism for
(41) Wang, Y. L., Darnay, B. G., and Rodwell, V. W. (1990) statin inhibition of HMG-CoA reductase. Science 292, 1160−1164.
Identification of the Principal Catalytically Important Acidic Residue of (61) Singh, N., Tamariz, J., Chamorro, G., and Medina-Franco, J. L.
3-Hydroxy-3-Methylglutaryl Coenzyme-a Reductase. J. Biol. Chem. 265, (2009) Inhibitors of HMG-CoA Reductase: Current and Future
Prospects. Mini-Rev. Med. Chem. 9, 1272−1283.
21634−21641.
(62) Ness, G. C., Lopez, D., Chambers, C. M., Zhao, Z., Beach, D. L.,
(42) Darnay, B. G., Wang, Y. L., and Rodwell, V. W. (1992)
Ko, S. S., and Trzaskos, J. M. (1998) Effects of 15-oxa-32-vinyl-lanost-8-
Identification of the Catalytically Important Histidine of 3-Hydroxy-3-
ene-3 beta,32 diol on the expression of 3-hydroxy-3-methylglutaryl
Methylglutaryl-Coenzyme-a Reductase. J. Biol. Chem. 267, 15064−
coenzyme A reductase and low density lipoprotein receptor in rat liver.
15070. Arch. Biochem. Biophys. 357, 259−264.
(43) Bochar, D. A., Tabernero, L., Stauffacher, C. V., and Rodwell, V. (63) Farmer, A. R., Murray, C. K., Mende, K., Akers, K. S., Zera, W. C.,
W. (1999) Aminoethylcysteine can replace the function of the essential Beckius, M. L., and Yun, H. C. (2013) Effect of HMG-CoA reductase
active site lysine of Pseudomonas mevalonii 3-hydroxy-3-methylglutaryl inhibitors on antimicrobial susceptibilities for Gram-Negative rods. J.
coenzyme A reductase. Biochemistry 38, 8879−8883. Basic Microbiol. 53, 336−339.
(44) Frimpong, K., and Rodwell, V. W. (1994) Catalysis by Syrian- (64) Nalin, D. R. (2008) Comment on: Unexpected antimicrobial
Hamster 3-Hydroxy-3-Methylglutaryl-Coenzyme-a Reductase - Pro- effect of statins. J. Antimicrob. Chemother. 61, 1400−1400.
posed Roles of Histidine-865, Glutamate-558, and Aspartate-766. J. Biol. (65) Jerwood, S., and Cohen, J. (2007) Unexpected antimicrobial
Chem. 269, 11478−11483. effect of statins. J. Antimicrob. Chemother. 61, 362−364.
(45) Haines, B. E., Wiest, O., and Stauffacher, C. V. (2013) The (66) Hogenboom, S., Tuyp, J. J. M., Espeel, M., Koster, J., Wanders, R.
Increasingly Complex Mechanism of HMG-CoA Reductase. Acc. Chem. J. A., and Waterham, H. R. (2004) Phosphomevalonate kinase is a
Res. 46, 2416−2426. cytosolic protein in humans. J. Lipid Res. 45, 697−705.
(46) Friesen, J. A., and Rodwell, V. W. (2004) The 3-hydroxy-3- (67) Hogenboom, S., Tuyp, J. J. M., Espeel, M., Koster, J., Wanders, R.
methylglutaryl coenzyme-A (HMG-CoA) reductases. Genome Biol. 5, J. A., and Waterham, H. R. (2004) Human mevalonate pyrophosphate
248. decarboxylase is localized in the cytosol. Mol. Genet. Metab. 81, 216−
(47) Goldstein, J. L., and Brown, M. S. (1990) Regulation of the 224.
mevalonate pathway. Nature 343, 425−430. (68) Hogenboom, S., Tuyp, J. J. M., Espeel, M., Koster, J., Wanders, R.
(48) Chambers, C. M., and Ness, G. C. (1998) Dietary cholesterol J. A., and Waterham, H. R. (2004) Mevalonate kinase is a cytosolic
regulates hepatic 3-hydroxy-3-methylglutaryl coenzyme A reductase enzyme in humans. J. Cell Sci. 117, 631−639.
gene expression in rats primarily at the level of translation. Arch. (69) Olivier, L. M., Chambliss, K. L., Gibson, K. M., and Krisans, S. K.
Biochem. Biophys. 354, 317−322. (1999) Characterization of phosphomevalonate kinase: chromosomal
(49) Ness, G. C. (2015) Physiological feedback regulation of localization, regulation, and subcellular targeting. J. Lipid Res. 40, 672−
cholesterol biosynthesis: Role of translational control of hepatic 679.
HMG-CoA reductase and possible involvement of oxylanosterols. (70) Kovacs, W. J., Olivier, L. M., and Krisans, S. K. (2002) Central role
Biochim. Biophys. Acta, Mol. Cell Biol. Lipids 1851, 667−673. of peroxisomes in isoprenoid biosynthesis. Prog. Lipid Res. 41, 369−391.
(50) Chambers, C. M., and Ness, G. C. (1997) Translational regulation (71) Fu, Z. J., Wang, M., Potter, D., Miziorko, H. M., and Kim, J. J. P.
of hepatic HMG-CoA reductase by dietary cholesterol. Biochem. Biophys. (2002) The structure of a binary complex between a mammalian
Res. Commun. 232, 278−281. mevalonate kinase and ATP - Insights into the reaction mechanism and
(51) Rajavashisth, T. B., Taylor, A. K., Andalibi, A., Svenson, K. L., and human inherited disease. J. Biol. Chem. 277, 18134−18142.
Lusis, A. J. (1989) Identification of a zinc finger protein that binds to the (72) Fu, Z., Voynova, N. E., Herdendorf, T. J., Miziorko, H. M., and
Kim, J. J. P. (2008) Biochemical and structural basis for feedback
sterol regulatory element. Science 245, 640−643.
(52) Osborne, T. F., Gil, G., Goldstein, J. L., and Brown, M. S. (1988) inhibition of mevalonate kinase and isoprenoid metabolism. Biochem-
istry 47, 3715−3724.
Operator constitutive mutation of 3-hydroxy-3-methylglutaryl coen-
(73) Amdur, B. H., Rilling, H., and Bloch, K. (1957) The Enzymatic
zyme A reductase promoter abolishes protein binding to sterol
Conversion of Mevalonic Acid to Squalene. J. Am. Chem. Soc. 79, 2646−
regulatory element. J. Biol. Chem. 263, 3380−3387. 2647.
(53) Faust, J. R., Luskey, K. L., Chin, D. J., Goldstein, J. L., and Brown, (74) Thurnher, M., Nussbaumer, O., and Gruenbacher, G. (2012)
M. S. (1982) Regulation of synthesis and degradation of 3-hydroxy-3- Novel Aspects of Mevalonate Pathway Inhibitors as Antitumor Agents.
methylglutaryl-coenzyme A reductase by low density lipoprotein and Clin. Cancer Res. 18, 3524−3531.
25-hydroxycholesterol in UT-1 cells. Proc. Natl. Acad. Sci. U. S. A. 79, (75) Potter, D., and Miziorko, H. M. (1997) Identification of catalytic
5205−5209. residues in human mevalonate kinase. J. Biol. Chem. 272, 25449−25454.
(54) Keller, R. K., Zhao, Z., Chambers, C., and Ness, G. C. (1996) (76) Berger, S. A., and Evans, P. R. (1992) Site-Directed Mutagenesis
Farnesol Is Not the Nonsterol Regulator Mediating Degradation of Identifies Catalytic Residues in the Active-Site of Escherichia-Coli
HMG-CoA Reductase in Rat Liver. Arch. Biochem. Biophys. 328, 324− Phosphofructokinase. Biochemistry 31, 9237−9242.
330. (77) Beytia, E., Dorsey, J. K., Marr, J., Cleland, W. W., and Porter, J. W.
(55) Beg, Z. H., Stonik, J. A., and Brewer, H. B., Jr. (1984) In vivo (1970) Purification and Mechanism of Action of Hog Liver Mevalonic
modulation of rat liver 3-hydroxy-3-methylglutaryl-coenzyme A Kinase. J. Biol. Chem. 245, 5450−5458.
reductase, reductase kinase, and reductase kinase kinase by (78) Cho, Y. K., Rios, S. E., Kim, J. J. P., and Miziorko, H. M. (2001)
mevalonolactone. Proc. Natl. Acad. Sci. U. S. A. 81, 7293−7297. Investigation of invariant serine/threonine residues in mevalonate

5503 DOI: 10.1021/acs.biochem.6b00342


Biochemistry 2016, 55, 5483−5506
Licenciado para - Paulo Sérgio Martins dos Santos - 09609690718 - Protegido por Eduzz.com

Biochemistry Current Topic

kinase - Tests of the functional significance of a proposed substrate (97) Street, I. P., Christensen, D. J., and Poulter, C. D. (1990)
binding motif and a site implicated in human inherited disease. J. Biol. Hydrogen-Exchange during the Enzyme-Catalyzed Isomerization of
Chem. 276, 12573−12578. Isopentenyl Diphosphate and Dimethylallyl Diphosphate. J. Am. Chem.
(79) Potter, D., Wojnar, J. M., Narasimhan, C., and Miziorko, H. M. Soc. 112, 8577−8578.
(1997) Identification and functional characterization of an active-site (98) Chapman, M. A., Lawrence, M. S., Keats, J. J., Cibulskis, K.,
lysine in mevalonate kinase. J. Biol. Chem. 272, 5741−5746. Sougnez, C., Schinzel, A. C., Harview, C. L., Brunet, J. P., Ahmann, G. J.,
(80) Chang, Q., Yan, X. X., Gu, S. Y., Liu, J. F., and Liang, D. C. (2008) Adli, M., Anderson, K. C., Ardlie, K. G., Auclair, D., Baker, A., Bergsagel,
Crystal structure of human phosphomavelonate kinase at 1.8 angstrom P. L., Bernstein, B. E., Drier, Y., Fonseca, R., Gabriel, S. B., Hofmeister,
resolution. Proteins: Struct., Funct., Genet. 73, 254−258. C. C., Jagannath, S., Jakubowiak, A. J., Krishnan, A., Levy, J., Liefeld, T.,
(81) Herdendorf, T. J., and Miziorko, H. M. (2007) Functional Lonial, S., Mahan, S., Mfuko, B., Monti, S., Perkins, L. M., Onofrio, R.,
evaluation of conserved basic residues in human phosphomevalonate Pugh, T. J., Rajkumar, S. V., Ramos, A. H., Siegel, D. S., Sivachenko, A.,
kinase. Biochemistry 46, 11780−11788. Stewart, A. K., Trudel, S., Vij, R., Voet, D., Winckler, W., Zimmerman,
(82) Olson, A. L., Yao, H. L., Herdendorf, T. J., Miziorko, H. M., T., Carpten, J., Trent, J., Hahn, W. C., Garraway, L. A., Meyerson, M.,
Hannongbua, S., Saparpakorn, P., Cai, S., and Sem, D. S. (2009) Lander, E. S., Getz, G., and Golub, T. R. (2011) Initial genome
Substrate induced structural and dynamics changes in human sequencing and analysis of multiple myeloma. Nature 471, 467−472.
phosphomevalonate kinase and implications for mechanism. Proteins: (99) Coleman, R. E. (2006) Clinical features of metastatic bone disease
Struct., Funct., Genet. 75, 127−138. and risk of skeletal morbidity. Clin. Cancer Res. 12, 6243s−6249s.
(83) Leipe, D. D., Koonin, E. V., and Aravind, L. (2003) Evolution and (100) Fournier, P. G., Stresing, V., Ebetino, F. H., and Clezardin, P.
classification of P-loop kinases and related proteins. J. Mol. Biol. 333, (2010) How Do Bisphosphonates Inhibit Bone Metastasis In Vivo?
781−815. Neoplasia 12, 571−578.
(84) Koonin, E. V. (1993) A Superfamily of Atpases with Diverse (101) Leung, C. Y., Park, J., De Schutter, J. W., Sebag, M., Berghuis, A.
Functions Containing Either Classical or Deviant Atp-Binding Motif M., and Tsantrizos, Y. S. (2013) Thienopyrimidine Bisphosphonate
(Vol 229, Pg 1165, 1993). J. Mol. Biol. 232, 1013−1013. (ThPBP) Inhibitors of the Human Farnesyl Pyrophosphate Synthase:
(85) Herdendorf, T. J., and Miziorko, H. M. (2006) Phosphomeval- Optimization and Characterization of the Mode of Inhibition. J. Med.
onate kinase: Functional investigation of the recombinant human Chem. 56, 7939−7950.
enzyme. Biochemistry 45, 3235−3242. (102) Monkkonen, H., Auriola, S., Lehenkari, P., Kellinsalmi, M.,
(86) Ramachandran, C. K., and Shah, S. N. (1976) Decarboxylation of Hassinen, I. E., Vepsalainen, J., and Monkkonen, J. (2006) A new
Mevalonate Pyrophosphate Is One Rate-Limiting Step in Hepatic
endogenous ATP analog (ApppI) inhibits the mitochondrial adenine
Cholesterol-Synthesis in Suckling and Weaned Rats. Biochem. Biophys.
nucleotide translocase (ANT) and is responsible for the apoptosis
Res. Commun. 69, 42−47.
induced by nitrogen-containing bisphosphonates. Br. J. Pharmacol. 147,
(87) Sawamura, M., Nara, Y., and Yamori, Y. (1992) Liver Mevalonate
437−445.
5-Pyrophosphate Decarboxylase Is Responsible for Reduced Serum-
(103) Mitrofan, L. M., Pelkonen, J., and Monkkonen, J. (2009) The
Cholesterol in Stroke-Prone Spontaneously Hypertensive Rat. J. Biol.
level of ATP analog and isopentenyl pyrophosphate correlates with
Chem. 267, 6051−6055.
(88) Voynova, N. E., Fu, Z. J., Battaile, K. P., Herdendorf, T. J., Kim, J. J. zoledronic acid-induced apoptosis in cancer cells in vitro. Bone 45,
P., and Miziorko, H. M. (2008) Human mevalonate diphosphate 1153−1160.
decarboxylase: Characterization, investigation of the mevalonate (104) Russell, R. G. G. (2011) Bisphosphonates: The first 40 years.
diphosphate binding site, and crystal structure. Arch. Biochem. Biophys. Bone 49, 2−19.
480, 58−67. (105) Szkopinska, A., and Plochocka, D. (2005) Farnesyl diphosphate
(89) Krepkiy, D., and Miziorko, H. M. (2004) Identification of active synthase; regulation of product specificity. Acta Biochim. Pol. 52, 45−55.
site residues in mevalonate diphosphate decarboxylase: Implications for (106) Krisans, S. K., Ericsson, J., Edwards, P. A., and Keller, G. A.
a family of phosphotransferases. Protein Sci. 13, 1875−1881. (1994) Farnesyl-Diphosphate Synthase Is Localized in Peroxisomes. J.
(90) Krepkiy, D. V., and Miziorko, H. M. (2005) Investigation of the Biol. Chem. 269, 14165−14169.
functional contributions of invariant serine residues in yeast mevalonate (107) Song, L. S., and Poulter, C. D. (1994) Yeast Farnesyl-
diphosphate decarboxylase. Biochemistry 44, 2671−2677. Diphosphate Synthase - Site-Directed Mutagenesis of Residues in
(91) Barta, M. L., Skaff, D. A., McWhorter, W. J., Herdendorf, T. J., Highly Conserved Prenyltransferase Domain-I and Domain-Ii. Proc.
Miziorko, H. M., and Geisbrecht, B. V. (2011) Crystal Structures of Natl. Acad. Sci. U. S. A. 91, 3044−3048.
Staphylococcus epidermidis Mevalonate Diphosphate Decarboxylase (108) Poulter, C. D., Mash, E. A., Argyle, J. C., Muscio, O. J., and
Bound to Inhibitory Analogs Reveal New Insight into Substrate Binding Rilling, H. C. (1979) Farnesyl Pyrophosphate Synthetase - Mechanistic
and Catalysis. J. Biol. Chem. 286, 23900−23910. Studies of the 1′-4 Coupling Reaction in the Terpene Biosynthetic-
(92) Barta, M. L., McWhorter, W. J., Miziorko, H. M., and Geisbrecht, Pathway. J. Am. Chem. Soc. 101, 6761−6763.
B. V. (2012) Structural basis for nucleotide binding and reaction (109) Tarshis, L. C., Proteau, P. J., Kellogg, B. A., Sacchettini, J. C., and
catalysis in mevalonate diphosphate decarboxylase. Biochemistry 51, Poulter, C. D. (1996) Regulation of product chain length by isoprenyl
5611−5621. diphosphate synthases. Proc. Natl. Acad. Sci. U. S. A. 93, 15018−15023.
(93) Steinbacher, S., Kaiser, J., Gerhardt, S., Eisenreich, W., Huber, R., (110) Hosfield, D. J., Zhang, Y. M., Dougan, D. R., Broun, A., Tari, L.
Bacher, A., and Rohdich, F. (2003) Crystal structure of the type II W., Swanson, R. V., and Finn, J. (2004) Structural basis for
isopentenyl diphosphate: Dimethylallyl diphosphate isomerase from bisphosphonate-mediated inhibition of isoprenoid biosynthesis. J. Biol.
Bacillus subtilis. J. Mol. Biol. 329, 973−982. Chem. 279, 8526−8529.
(94) Durbecq, V., Sainz, G., Oudjama, Y., Clantin, B., Bompard-Gilles, (111) Sanchez, V. M., Crespo, A., Gutkind, J. S., and Turjanski, A. G.
C., Tricot, C., Caillet, J., Stalon, V., Droogmans, L., and Villeret, V. (2006) Investigation of the catalytic mechanism of farnesyl
(2001) Crystal structure of isopentenyl diphosphate: dimethylallyl pyrophosphate synthase by computer simulation. J. Phys. Chem. B
diphosphate isomerase. EMBO J. 20, 1530−1537. 110, 18052−18057.
(95) Wouters, J., Oudjama, Y., Ghosh, S., Stalon, V., Droogmans, L., (112) Pandit, J., Danley, D. E., Schulte, G. K., Mazzalupo, S., Pauly, T.
and Oldfield, E. (2003) Structure and mechanism of action of A., Hayward, C. M., Hamanaka, E. S., Thompson, J. F., and Harwood, H.
isopentenylpyrophosphate-dimethylallylpyrophosphate isomerase. J. J. (2000) Crystal structure of human squalene synthase - A key enzyme
Am. Chem. Soc. 125, 3198−3199. in cholesterol biosynthesis. J. Biol. Chem. 275, 30610−30617.
(96) Reardon, J. E., and Abeles, R. H. (1986) Mechanism of Action of (113) Poulter, C. D. (1990) Biosynthesis of Non-Head-to-Tail
Isopentenyl Pyrophosphate Isomerase - Evidence for a Carbonium-Ion Terpenes - Formation of 1′-1 and 1′-3 Linkages. Acc. Chem. Res. 23,
Intermediate. Biochemistry 25, 5609−5616. 70−77.

5504 DOI: 10.1021/acs.biochem.6b00342


Biochemistry 2016, 55, 5483−5506
Licenciado para - Paulo Sérgio Martins dos Santos - 09609690718 - Protegido por Eduzz.com

Biochemistry Current Topic

(114) Sandifer, R. M., Thompson, M. D., Gaughan, R. G., and Poulter, (132) Gotteland, J. P., Junquero, D., Oms, P., Delhon, A., and Halazy,
C. D. (1982) Squalene Synthetase - Inhibition by an Ammonium Analog S. (1996) Comparative study of new and known thienyl derivatives of
of a Carbocationic Intermediate in the Conversion of Presqualene ENE-YNE benzylamine as mammalian squalene epoxidase inhibitors.
Pyrophosphate to Squalene. J. Am. Chem. Soc. 104, 7376−7378. Med. Chem. Res. 6, 333−342.
(115) Gu, P., Ishii, Y., Spencer, T. A., and Shechter, I. (1998) Function- (133) Astruc, M., Tabacik, C., Descomps, B., and de Paulet, A. C.
structure studies and identification of three enzyme domains involved in (1977) Squalene Epoxidase and Oxidosqualene Lanosterol-Cyclase
the catalytic activity in rat hepatic squalene synthase. (vol 273, pg 12515, Activities in Cholesterogenic and Non-Cholesterogenic Tissues.
1998). J. Biol. Chem. 273, 17296. Biochim. Biophys. Acta, Lipids Lipid Metab. 487, 204−211.
(116) Liu, C. I., Liu, G. Y., Song, Y. C., Yin, F. L., Hensler, M. E., Jeng, (134) Nagumo, A., Kamei, T., Sakakibara, J., and Ono, T. (1995)
W. Y., Nizet, V., Wang, A. H. J., and Oldfield, E. (2008) A cholesterol Purification and Characterization of Recombinant Squalene Epoxidase.
biosynthesis inhibitor blocks Staphylococcus aureus virulence. Science J. Lipid Res. 36, 1489−1497.
319, 1391−1394. (135) Abe, I., Seki, T., and Noguchi, H. (2000) Potent and selective
(117) Lin, F. Y., Liu, C. I., Liu, Y. L., Zhang, Y. H., Wang, K., Jeng, W. Y., inhibition of squalene epoxidase by synthetic galloyl esters. Biochem.
Ko, T. P., Cao, R., Wang, A. H. J., and Oldfield, E. (2010) Mechanism of Biophys. Res. Commun. 270, 137−140.
action and inhibition of dehydrosqualene synthase. Proc. Natl. Acad. Sci. (136) Laden, B. P., and Porter, T. D. (2001) Inhibition of human
U. S. A. 107, 21337−21342. squalene monooxygenase by tellurium compounds: evidence of
(118) Do, R., Kiss, R. S., Gaudet, D., and Engert, J. C. (2009) Squalene interaction with vicinal sulfhydryls. J. Lipid Res. 42, 235−240.
synthase: a critical enzyme in the cholesterol biosynthesis pathway. Clin. (137) Chugh, A., Ray, A., and Gupta, J. B. (2003) Squalene epoxidase
Genet. 75, 19−29. as hypocholesterolemic drug target revisited. Prog. Lipid Res. 42, 37−50.
(119) Ness, G. C., Zhao, Z. H., and Keller, R. K. (1994) Effect of (138) Belter, A., Skupinska, M., Giel-Pietraszuk, M., Grabarkiewicz, T.,
Squalene Synthase Inhibition on the Expression of Hepatic Cholesterol Rychlewski, L., and Barciszewski, J. (2011) Squalene monooxygenase - a
Biosynthetic-Enzymes, Ldl Receptor, and Cholesterol 7-Alpha Hydrox- target for hypercholesterolemic therapy. Biol. Chem. 392, 1053−1075.
ylase. Arch. Biochem. Biophys. 311, 277−285. (139) Ryder, N. S. (1992) Terbinafine - Mode of Action and Properties
(120) Bergstrom, J. D., Kurtz, M. M., Rew, D. J., Amend, A. M., Karkas, of the Squalene Epoxidase Inhibition. Br. J. Dermatol. 126, 2−7.
J. D., Bostedor, R. G., Bansal, V. S., Dufresne, C., Vanmiddlesworth, F. (140) Horie, M., Tsuchiya, Y., Hayashi, M., Iida, Y., Iwasawa, Y.,
L., Hensens, O. D., Liesch, J. M., Zink, D. L., Wilson, K. E., Onishi, J., Nagata, Y., Sawasaki, Y., Fukuzumi, H., Kitani, K., and Kamei, T. (1990)
Milligan, J. A., Bills, G., Kaplan, L., Omstead, M. N., Jenkins, R. G., Nb-598 - a Potent Competitive Inhibitor of Squalene Epoxidase. J. Biol.
Huang, L., Meinz, M. S., Quinn, L., Burg, R. W., Kong, Y. L., Mochales, Chem. 265, 18075−18078.
(141) Horie, M., Sawasaki, Y., Fukuzumi, H., Watanabe, K., Iizuka, Y.,
S., Mojena, M., Martin, I., Pelaez, F., Diez, M. T., and Alberts, A. W.
Tsuchiya, Y., and Kamei, T. (1991) Hypolipidemic Effects of Nb-598 in
(1993) Zaragozic Acids - a Family of Fungal Metabolites That Are
Dogs. Atherosclerosis 88, 183−192.
Picomolar Competitive Inhibitors of Squalene Synthase. Proc. Natl.
(142) Thoma, R., Schulz-Gasch, T., D’Arcy, B., Benz, J., Aebi, J.,
Acad. Sci. U. S. A. 90, 80−84.
Dehmlow, H., Hennig, M., Stihle, M., and Ruf, A. (2004) Insight into
(121) Stein, E. A., Bays, H., O’Brien, D., Pedicano, J., Piper, E., and
steroid scaffold formation from the structure of human oxidosqualene
Spezzi, A. (2011) Lapaquistat acetate: development of a squalene
cyclase. Nature 432, 118−122.
synthase inhibitor for the treatment of hypercholesterolemia. Circulation (143) Nakano, C., Motegi, A., Sato, T., Onodera, M., and Hoshino, T.
123, 1974−1985. (2007) Sterol biosynthesis by a prokaryote: First in vitro identification
(122) Ichikawa, M., Ohtsuka, M., Ohki, H., Ota, M., Haginoya, N., Itoh, of the genes encoding squalene epoxidase and lanosterol synthase from
M., Shibata, Y., Ishigai, Y., Terayama, K., Kanda, A., Sugita, K., and Usui, Methylococcus capsulatus. Biosci., Biotechnol., Biochem. 71, 2543−2550.
H. (2013) Discovery of DF-461, a Potent Squalene Synthase Inhibitor. (144) Ruf, A., Muller, F., D’Arcy, B., Stihle, M., Kusznir, E., Handschin,
ACS Med. Chem. Lett. 4, 932−936. C., Morand, O. H., and Thoma, R. (2004) The monotopic membrane
(123) Stein, E. A., Bays, H., O’Brien, D., Pedicano, J., Piper, E., and protein human oxidosqualene cyclase is active as monomer. Biochem.
Spezzi, A. (2011) Lapaquistat Acetate Development of a Squalene Biophys. Res. Commun. 315, 247−254.
Synthase Inhibitor for the Treatment of Hypercholesterolemia. (145) Abe, I., Rohmer, M., and Prestwich, G. D. (1993) Enzymatic
Circulation 123, 1974−1985. Cyclization of Squalene and Oxidosqualene to Sterols and Triterpenes.
(124) Urbina, J. A., Concepcion, J. L., Rangel, S., Visbal, G., and Lira, R. Chem. Rev. 93, 2189−2206.
(2002) Squalene synthase as a chemotherapeutic target in Trypanosoma (146) Hess, B. A. (2002) Concomitant C-ring expansion and D-ring
cruzi and Leishmania mexicana. Mol. Biochem. Parasitol. 125, 35−45. formation in lanosterol biosynthesis from squalene without violation of
(125) Urbina, J. A. (2001) Specific treatment of Chagas disease: Markovnikov’s rule. J. Am. Chem. Soc. 124, 10286−10287.
current status and new developments. Curr. Opin. Infect. Dis. 14, 733− (147) van Tamelen, E. E., Willett, J. D., and Clayton, R. B. (1967) On
741. the mechanism of lanosterol biosynthesis from squalene 2,3-oxide. J.
(126) Urbina, J. A. (1997) Lipid biosynthesis pathways as chemo- Am. Chem. Soc. 89, 3371−3373.
therapeutic targets in kinetoplastid parasites. Parasitology 114, S91−S99. (148) van Tamelen, E. E., Willet, J., Schwartz, M., and Nadeau, R.
(127) Urbina, J. A., Concepcion, J. L., Caldera, A., Payares, G., Sanoja, (1966) Nonenzymic Laboratory Cyclization of Squalene 2,3-Oxide. J.
C., Otomo, T., and Hiyoshi, H. (2004) In vitro and in vivo activities of Am. Chem. Soc. 88, 5937−5938.
E5700 and ER-119884, two novel orally active squalene synthase (149) van Tamelen, E. E., Lees, R. G., and Grieder, A. (1974)
inhibitors, against Trypanosoma cruzi. Antimicrob. Agents Chemother. 48, Cyclization of a Terpenoid Diene with Preformed a-B-D Rings and Its
2379−2387. Significance for Mechanism of Terpenoid Terminal Epoxide Cycliza-
(128) Urbina, J. A. (2009) Ergosterol biosynthesis and drug tions. J. Am. Chem. Soc. 96, 2255−2256.
development for Chagas disease. Mem I Oswaldo Cruz 104, 311−318. (150) Vantamelen, E. E., and James, D. R. (1977) Overall Mechanism
(129) McTaggart, F., Brown, G. R., Davidson, R. G., Freeman, S., of Terpenoid Terminal Epoxide Polycyclizations. J. Am. Chem. Soc. 99,
Holdgate, G. A., Mallion, K. B., Mirrlees, D. J., Smith, G. J., and Ward, W. 950−952.
H. J. (1996) Inhibition of squalene synthase of rat liver by novel 3′ (151) Vantamelen, E. E., and Hopla, R. E. (1979) Generation of the
substituted quinuclidines. Biochem. Pharmacol. 51, 1477−1487. Onocerin System by Lanosterol 2,3-Oxidosqualene Cyclase -
(130) Goldstein, J. L., and Brown, M. S. (2001) Molecular medicine - Implications for the Cyclization Process. J. Am. Chem. Soc. 101,
The cholesterol quartet. Science 292, 1310−1312. 6112−6114.
(131) Ono, T., and Bloch, K. (1975) Solubilization and Partial (152) Vantamelen, E. E. (1982) Bioorganic Characterization and
Characterization of Rat-Liver Squalene Epoxidase. J. Biol. Chem. 250, Mechanism of the 2,3-Oxidosqualene-] Lanosterol Conversion. J. Am.
1571−1579. Chem. Soc. 104, 6480−6481.

5505 DOI: 10.1021/acs.biochem.6b00342


Biochemistry 2016, 55, 5483−5506
Licenciado para - Paulo Sérgio Martins dos Santos - 09609690718 - Protegido por Eduzz.com

Biochemistry Current Topic

(153) Wendt, K. U., Schulz, G. E., Corey, E. J., and Liu, D. R. (2000)
Enzyme mechanisms for polycyclic triterpene formation. Angew. Chem.,
Int. Ed. 39, 2812−2833.
(154) Tian, B. X., and Eriksson, L. A. (2012) Catalytic Mechanism and
Product Specificity of Oxidosqualene-Lanosterol Cyclase: A QM/MM
Study. J. Phys. Chem. B 116, 13857−13862.
(155) Corey, E. J., Russey, W. E., and Ortiz de Montellano, P. R. (1966)
2,3-oxidosqualene, an intermediate in the biological synthesis of sterols
from squalene. J. Am. Chem. Soc. 88, 4750−4751.
(156) Jenson, C., and Jorgensen, W. L. (1997) Computational
investigations of carbenium ion reactions relevant to sterol biosynthesis.
J. Am. Chem. Soc. 119, 10846−10854.
(157) Wu, T. K., Wang, T. T., Chang, C. H., Liu, Y. T., and Shie, W. S.
(2008) Importance of Saccharomyces cerevisiae oxidosqualene-
lanosterol cyclase tyrosine 707 residue for chair-boat bicyclic ring
formation and deprotonation reactions. Org. Lett. 10, 4959−4962.
(158) Wendt, K. U., Lenhart, A., and Schulz, G. E. (1999) The
structure of the membrane protein squalene-hopene cyclase at 2.0 A
resolution. J. Mol. Biol. 286, 175−187.
(159) Cerqueira, N. M. F. S. A., Fernandes, P. A., and Ramos, M. J.
(2011) Computational Mechanistic Studies Addressed to the
Transimination Reaction Present in All Pyridoxal 5 ′-Phosphate-
Requiring Enzymes. J. Chem. Theory Comput. 7, 1356−1368.
(160) Gesto, D. S., Cerqueira, N. M., Fernandes, P. A., and Ramos, M.
J. (2013) Unraveling the enigmatic mechanism of L-asparaginase II with
QM/QM calculations. J. Am. Chem. Soc. 135, 7146−7158.
(161) Ngo, H. P., Cerqueira, N. M., Kim, J. K., Hong, M. K., Fernandes,
P. A., Ramos, M. J., and Kang, L. W. (2014) PLP undergoes
conformational changes during the course of an enzymatic reaction.
Acta Crystallogr., Sect. D: Biol. Crystallogr. 70, 596−606.
(162) Oliveira, E. F., Cerqueira, N. M., Fernandes, P. A., and Ramos, M.
J. (2011) Mechanism of formation of the internal aldimine in pyridoxal
5′-phosphate-dependent enzymes. J. Am. Chem. Soc. 133, 15496−15505.
(163) Ramos, M. J., and Fernandes, P. A. (2008) Computational
enzymatic catalysis. Acc. Chem. Res. 41, 689−698.
(164) Sousa, S. F., Fernandes, P. A., and Ramos, M. J. (2012)
Computational enzymatic catalysis–clarifying enzymatic mechanisms
with the help of computers. Phys. Chem. Chem. Phys. 14, 12431−12441.
(165) Wilcox, C., Turner, J., and Green, J. (2014) Systematic review:
the management of chronic diarrhoea due to bile acid malabsorption.
Aliment. Pharmacol. Ther. 39, 923−939.
(166) Cannon, C. P., Blazing, M. A., Giugliano, R. P., McCagg, A.,
White, J. A., Theroux, P., Darius, H., Lewis, B. S., Ophuis, T. O., Jukema,
J. W., De Ferrari, G. M., Ruzyllo, W., De Lucca, P., Im, K., Bohula, E. A.,
Reist, C., Wiviott, S. D., Tershakovec, A. M., Musliner, T. A., Braunwald,
E., and Califf, R. M. (2015) Ezetimibe Added to Statin Therapy after
Acute Coronary Syndromes. N. Engl. J. Med. 372, 2387−2397.
(167) Ness, G. C., Holland, R. C., and Lopez, D. (2006) Selective
compensatory induction of hepatic HMG-CoA reductase in response to
inhibition of cholesterol absorption. Exp. Biol. Med. 231, 559−565.
(168) Li, C., Lin, L., Zhang, W., Zhou, L., Wang, H., Luo, X., Luo, H.,
Cai, Y., and Zeng, C. (2015) Efficiency and safety of proprotein
convertase subtilisin/kexin 9 monoclonal antibody on hypercholester-
olemia: a meta-analysis of 20 randomized controlled trials. J. Am. Heart
Assoc. 4, e001937.
(169) Navarese, E. P., Kolodziejczak, M., Schulze, V., Gurbel, P. A.,
Tantry, U., Lin, Y., Brockmeyer, M., Kandzari, D. E., Kubica, J. M.,
D’Agostino, R. B., Sr., Kubica, J., Volpe, M., Agewall, S., Kereiakes, D. J.,
and Kelm, M. (2015) Effects of Proprotein Convertase Subtilisin/Kexin
Type 9 Antibodies in Adults With Hypercholesterolemia: A Systematic
Review and Meta-analysis. Ann. Intern. Med. 163, 40−51.

5506 DOI: 10.1021/acs.biochem.6b00342


Biochemistry 2016, 55, 5483−5506

You might also like