You are on page 1of 17

Home Lipid Essentials Literature Service Mass Spectrometry Blog

Prostanoids: Prostaglandins, Prostacyclins


and Thromboxanes
The prostanoids are part of the oxylipin family of biologically active lipids derived from the action of
cyclooxygenases or prostaglandin synthases upon the twenty-carbon essential fatty acids or
eicosanoids, mainly arachidonic acid. They can be further subdivided into two main groups, the
prostacyclopentanes, comprising the prostaglandins and prostacyclins, and the thromboxanes, each
of which is involved in some aspect of the inflammatory response. The prostaglandins were first isolated
from semen and named from the prostate gland, thought to be their source, as long ago as the 1930s,
but it was the 1960s before the biosynthetic relationship to specific essential fatty acids was described
and intensive research into their biological properties began. The Nobel Prize for Medicine for 1982 was
awarded to Professors Bengt Samuelsson, John Vane and Sune Bergström for their discoveries in this
field (see the review by Samuelsson cited below). In general, prostaglandins occur at very low levels in
tissues, of the order of nanomolar concentrations, but they have profound biological activities. While
most studies have been concerned with their occurrence and function in mammals, they have also been
detected in birds, ray-finned fishes, marine invertebrates, trypanosomes, blood flukes, and some algae
and yeasts.

1. Nomenclature and Structures of Prostanoids


In structure, prostanoids are best considered as derivatives of a C20 saturated fatty acid, prostanoic
acid, which does not itself occur in nature. A key feature is a five-membered ring encompassing carbons
8 to 12, as illustrated below. The thromboxanes are similar but have heterocyclic oxane structures. They
are all synthesised by specific enzymes, which confer stereospecificity and chirality on every functional
group, and are thus distinct from the isoprostanes, which are produced by non-enzymic means and
have their own web page.
In the approved nomenclature, each prostaglandin is named using the prefix 'PG' followed by a letter A
to K depending on the nature and position of the substituents on the ring. Thus PGA to PGE and PGJ
have a keto group in various positions on the ring, and are further distinguished by the presence or
absence of double bonds or hydroxyl groups in various positions in the ring. PGF has two hydroxyl
groups while PGK has two keto substituents on the ring. PGG and PGH are bicyclic endoperoxides. An
oxygen bridge between carbons 6 and 9 distinguishes prostacyclin (PGI). Thromboxane A (TXA)
contains an unstable bicyclic oxygenated ring structure, while thromboxane B (TXB) has a stable oxane
ring. In addition, all prostaglandins have a hydroxyl group in the S-configuration on carbon 15 and a
trans-double bond at carbon 13 of the alkyl substituent (R2).

Further, a numerical subscript (1 to 3) is used to denote the total number of double bonds in the alkyl
substituents, and a Greek subscript (α or β) is used with prostaglandins of the PGF series to describe
the stereochemistry of the hydroxyl group on carbon 9. This is illustrated for prostaglandins PGE and
PGFα of the 1, 2 and 3 series below, as examples.
The number of double bonds depends on the nature of the fatty acid precursor. Thus, the prostaglandins
PGE1, PGE2 and PGE3 are derived from 8c,11c,14c-eicosatrienoic (dihomo-γ-linolenic), 5c,8c,11c,14c-
eicosatetraenoic (arachidonic) and 5c,8c,11c,14c,17c-eicosapentaenoic acids, respectively. Of these,
PGE2 is the most actively produced, and it is involved in innumerable physiological processes. Dihomo-
prostaglandins derived from adrenic acid (22:4(n-6) have also been detected in cell preparations, but no
such compounds are produced from docosahexaenoic acid (DHA).

2. Biosynthesis of Prostaglandins
Cyclooxygenases: Eicosanoids, including the prostanoids, are not stored within cells but are
synthesised as required in response to hormonal stimuli. The prostaglandins PGE2 and PGF2α were first
isolated and characterized from human seminal fluid in 1963 by Samuelsson, but prostaglandins and
other eicosanoids are now known to be produced in a highly selective manner by most cell types,
depending on the activation state and the physiological condition of the tissues in which they occur. The
first step in their synthesis is the release of the substrate fatty acid, such as arachidonic acid, from the
cellular phospholipids by the action of the enzyme phospholipase A2, and this is discussed in the
Introductory document to this series. Next, the free acids are acted upon by one of two related enzymes,
cyclooxygenase-1 (COX-1) and cyclooxygenase-2 (COX-2), more correctly termed prostaglandin
endoperoxide H synthases-1 and -2 (PGHS-1 and PGHS-2). These are key enzymes that catalyse the
first committed step in the synthesis of prostanoids from fatty acid precursors; COX-1 is always present
in tissues, while COX-2 is induced by appropriate physiological stimuli (cytokines, tumor promoters and
growth factors). The two iso-enzymes have approximately 90% sequence identity, and they are very
similar in structure, but an important difference is that COX-2 has a larger pocket at the active site
because of an isoleucine to valine substitution. The result is that in comparison with COX-1 it can be
more permissive in utilizing fatty acids, such as dihomo-γ-linolenic and eicosapentaenoic acids (and
even linoleic and α-linolenic acids) in addition to arachidonic acid and other lipid substrates (see below).

In humans, COX-1 and COX-2 are homodimers of 576 and 581 amino acids, respectively, and each has
three mannose-containing oligosaccharides linked to it, one of which facilitates protein folding. Each
subunit of the dimer consists of three domains, the epidermal growth factor, the membrane binding
domain, and the substantial catalytic domain, which contains two active sites on either side of a heme
prosthetic group. A fourth oligosaccharide is found only in COX-2 and regulates its degradation. They
are integral membrane proteins of the endoplasmic reticulum and are located on the lumenal side only
of the bilayer (COX-2 localizes to the Golgi in cancer cell lines). In addition, they are present on the
inner and outer membranes of the nuclear envelope.

Both enzymes catalyse the same two reactions. Thus, each carries out a cyclooxygenase reaction in
which two molecules of oxygen are added to arachidonic acid to form a bicyclic endoperoxide with a
further hydroperoxy group in position 15, i.e. to form prostaglandin PGG2. The first reaction occurs at a
hydrophobic channel in the centre of the enzyme, before the hydroperoxide intermediate is transferred
to the heme-containing site on the surface of the enzyme where it is reduced by a peroxidase to form
prostaglandin PGH2.

Although the reactions occur at different sites, they are functionally coupled. The combined reactions are
initiated by the oxidation of the heme group involved in the peroxidase reaction by traces of endogenous
hydroperoxides with formation of a tyrosyl radical. This abstracts the 13‑pro‑S hydrogen from
arachidonic acid and initiates the cyclooxygenase reaction by formation of a carbon-centered radical at
C-11; attack of molecular oxygen at this position leads to the intramolecular rearrangement with
formation of a bicyclic endoperoxide and a further carbon-centered radical at C-15. This radical reacts
with a further molecule of oxygen to form a hydroperoxide, which is reduced to form PGG2 and thence
PGH2 via the peroxidase activity. During the reduction step the tyrosyl radical is regenerated so that
activated COX can carry out multiple turnovers without a need to repeat the activation step. The other
precursor polyunsaturated fatty acids interact with the enzymes in similar ways. As the catalytic tyrosyl
radical can be transferred to an adjacent tyrosyl residue and become inactive after about 300 turnovers,
the enzyme must be re-expressed constantly to generate metabolites. PGH2 is highly reactive and is the
starting point for the biosynthesis of most other prostanoids.

The requirement for two distinct cyclooxygenases is not fully understood. In spite of the structural
homology, separate genes encode COX-1 (on chromosome 9 in humans) and COX-2 (on chromosome
1) and they are regulated independently by different systems. The enzymes differ in their subcellular
localization, substrate specificity and the manner in which they are coupled to upstream and
downstream enzymes. In addition, the catalytic domains differ in structure, so that the
susceptibilities to some inhibitors are not the same. It is now apparent that the two enzymes
have different functional roles. Although it is almost certainly an over-simplification, it is
usually suggested that COX-1 is used for ‘housekeeping’ (homeostatic) purposes,
responding rapidly to circulating hormones, which require constant monitoring and
regulation. It is a constitutive enzyme that produces prostaglandins in the endoplasmic
reticulum, which exit cells and signal through G-protein-linked receptors at the cell surface.
However, there are also suggestions that it functions only at relatively high concentrations of
arachidonic acid, for example during platelet aggregation, cell injury or acute inflammation.
In those tissues where prostaglandins have specialized signalling functions, such as kidney, stomach,
vascular endothelium, and especially blood platelets, COX-1 is expressed at higher concentrations, i.e.
where the enzyme provides precursors for thromboxane synthesis.

In contrast, COX-2 is an inducible enzyme that is not present in unactivated tissues other than lung,
kidney and brain (where COX-2 is constitutive in neurons and radial glia, but not other cell types). It is
expressed under the control of the pro-inflammatory transcription factor NF‑κB in response to a wide
range of extracellular and intracellular stimuli, such as cytokines, growth factors and tumor promoters,
and produces prostanoids that are primarily pathophysiological or that function during defined stages of
cellular development. It is able to utilize much lower concentrations of arachidonic acid and substrates
other than the free acid. COX-2 expression is inducible by bacterial lipopolysaccharides and is
especially important in cells that are involved in inflammation, such as macrophages and monocytes,
and it is believed to be the form of the enzyme that has the main responsibility for the synthesis of those
prostanoids involved in most severe inflammatory states, including cancer, rheumatoid arthritis,
Alzheimer's disease and respiratory disorders, although COX-1 is also important in this context. On the
other hand, COX-2 provides the substrate for synthesis of prostacyclin, which opposes the actions of
thromboxanes (see below). Some COX-2 products may modulate the transcription of certain genes in
the cell nucleus. COX-2 is activated by hydroperoxide concentrations that are approximately tenfold
lower than those that activate COX-1, raising the possibility that under limiting concentrations of
peroxide, COX-2 may be fully active while COX-1 is not. Induction of COX-2 expression is also
regulated by sphingosine-1-phosphate, a further effect of sphingolipids on prostanoid biosynthesis.

Other prostaglandin synthases: PGH2 produced by the COX enzymes is an unstable intermediate
from which all other prostanoids are derived by a variety of different enzymic reactions. Some of these
are illustrated next for arachidonate as the primary precursor. The nature and proportions of the various
enzymes and of the prostanoids produced differ according to cell type. Indeed different forms of some of
the enzymes exist in cells that may be functionally similar, but differ in amino acid sequence, structure
and co-factor requirements. Thus, PGH2 is converted to PGE2 by prostaglandin E synthases, of which
at least three forms exist that are structurally and biologically distinct. The most important of these is a
cytosolic enzyme, which is expressed constitutively in many different types of cell and is linked
functionally to COX-1 to promote immediate PGE2 production. A second membrane-bound enzyme is
induced by inflammatory stimuli and functions in concert with the inducible COX-2. PGD2 is formed in a
similar way from PGH2 by the action of prostaglandin D synthases, which exist in two forms that are
evolutionarily distinct but convergent in their functions; one is located in the central nervous system and
the other in peripheral tissues. In rat peritoneal macrophages, PGD synthase and COX-1 appear to be
functionally coupled.
Levuglandins, such as LGE2 (also termed ‘isoketals’), are
formed from PGH2 by a non-enzymic rearrangement. They
have a very short half-life and react more rapidly than most
lipid oxidation products with the free primary amine groups of
proteins and phosphatidylethanolamine (see below) to form
covalent adducts. Indeed, the reaction is so rapid that the free
levuglandins have never been isolated.

The most common stereochemical form of prostaglandin F2α (PGF2α) is synthesised by two main
routes. For example, it can be produced directly from PGH2 by the action of prostaglandin H-
endoperoxide reductase, requiring NADPH. Interestingly, this enzyme can also utilize PGD2 as a
substrate for the synthesis of the second of the four stereochemical forms of PGF2α, i.e. 9α,11β-PGF2α.
As an alternative, PGF2α is synthesised via PGE2 by the action of an enzyme prostaglandin E 9-
ketoreductase.

The cyclopentenone prostaglandins A and J, with reactive α,β-unsaturated keto groups and high
biological activity, are produced by spontaneous dehydration reactions from PGE and PGD, respectively,
and further modifications can then occur. For example, PGA2 isomerizes to form the highly unstable
PGC2, which rapidly undergoes a secondary isomerization to produce PGB2. Similarly, in the presence
of human serum albumin in vitro, it has been demonstrated that PGD2 is transformed into three
dehydration products, i.e. 15-deoxy-PGD2, Δ12-PGJ2 and 15-deoxy-Δ12,14-PGJ2 (the last two via the
intermediate PGJ2).

PGE2 is the major product of prostaglandin biosynthesis pathways following activation by pro-
inflammatory substances and with its metabolite PGF2α, it is involved in a positive feedback loop to
regulate COX-2 expression. Before they can function, prostanoids that have been newly synthesised
must be transported from the cytosol and cross various membranes by means of active transporter
systems.

Endocannabinoid metabolism: There is a significant difference in the substrate requirements of the


two iso-enzymes. While both utilize unesterified arachidonic acid as substrate, COX-2 can also
metabolize dihomo-γ-linolenic and eicosapentaenoic acids. COX-1 can only utilize free fatty acids, but
COX-2 can react with the endocannabinoid 2-arachidonoylglycerol to form esterified
2‑prostanoylglycerol derivatives, i.e. hydroxy endoperoxides analogous to PGH2, which can be further
metabolized by downstream synthases. Similarly, COX-2 is involved in conversion of anandamide
(arachidonoylethanolamine) and arachidonoylglycine to biologically active ‘prostamides’, though with
lower efficiency. While these may simply serve as precursors of free prostanoids through hydrolysis,
there is increasing evidence that they are new classes of lipid mediators with distinct biological
properties of their own. The amide derivatives especially are relatively long-lived in plasma, and amides
of PGF2α are available as drugs to lower ocular pressure and treat glaucoma. There is evidence for
effects of 2-prostanoylglycerol on calcium mobilization through distinct and novel receptors as well as
activation of the PPARδ receptor. It is subject to hydrolysis by esterases present in blood and some
tissues, and especially the lysophospholipid lipase LYPLA2, which releases the prostaglandin in free
form.

The Role of Aspirin: Both COX iso-enzymes and thence prostaglandin


synthesis are inhibited by non-steroidal anti-inflammatory drugs ('NSAIDs'),
such as aspirin (acetylsalicylic acid - one of the earliest known and most widely
used of all pharmaceuticals) and ibuprofen. Aspirin exerts this inhibition by
binding to the cyclooxygenase site and transferring its acetyl group irreversibly
to a specific serine residue (Ser-530), which then protrudes into the active site
and obstructs the binding of arachidonate. Because of differences in the
structures of the binding sites, COX-1 is completely inhibited by this means, but aspirin acetylation of
COX-2 results in a shift in reaction specificity, converting the enzyme activity from that of a
cyclooxygenase to a lipoxygenase, and resulting in the generation of 15(R)-hydroxy-5,8,11,13-
eicosatetraenoic acid (15(R)-HETE), i.e. with the opposite chirality to that produced in the lipoxygenase
reaction. In addition, some PGD2, but not PGE2, may be formed - again with the 15(R)‑configuration. In
contrast, ibuprofen and all other drugs of this type exert their effects by reversible binding and
competition with arachidonic acid for the active sites. The specific inhibition by aspirin is the reason for
its well-known analgesic, anti-pyretic and anti-inflammatory effects as a pharmaceutical. Via its effect on
cyclooxygenases, it inhibits thromboxane synthesis and thence platelet aggregation, and it is now
recommended in cardiovascular therapy.

However, this does not fully explain aspirin's repertoire of anti-inflammatory effects, and it is now known
to be intimately involved, through an action with COX-2, in the generation of oxygenated lipid mediators
such as the ‘aspirin-triggered’ protectins (resolvins) and the epi-lipoxins, as well as the eicosanoids
with the (R)-configuration, which all exert profound anti-inflammatory effects. This may be the reason for
some of the clinical benefits of aspirin, especially in neuro-inflammation.

Synthesis of COX-2 is inhibited by steroidal anti-inflammatory drugs at the level of transcription. As the
active site of COX-2 is smaller than that of COX-1, it has proved possible to develop a number of drugs
that specifically inhibit the action of COX-2. As well as having analgesic and anti-inflammatory effects,
these are used clinically to prevent cancer of the colon. However, some COX-2 selective inhibitors have
been associated with an increased risk of cardiovascular disease and have been withdrawn from the
market.

Insects: The phospholipids in insects tend to contain very little arachidonic acid, so the starting point in
the biosynthesis of prostaglandins is the release of linoleic acid by the action of phospholipase A2.
Linoleic acid serves as the precursor for arachidonate biosynthesis, and this is acted upon by a specific
peroxidase termed peroxinectin, and not by cyclooxygenases, to produce PGH2, which is then
converted to PGE2 by a PGE2 synthase.

Related enzyme activities: Certain pathogenic fungi and yeasts produce 3-hydroxy-eicosanoids
from host arachidonic acid and they can hijack the host’s COX-2 enzymes to produce 3-hydroxy-
prostaglandins from these that are as active biologically as the normal compounds. Other fungi produce
enzymes with significant homologies to mammalian cyclooxygenases COX-1 and COX-2 and termed
Ppo proteins that synthesise PGH2. In addition, the yeast Candida albicans and related pathogenic fungi
produce PGE2 and other prostanoids in vitro from exogenous arachidonate, but the enzymes with COX-
like activities have not yet been characterized. A prostaglandin H synthase isolated from the red alga
Gracilaria vertniculophylla is very different in structure from its animal counterparts, but it appears to
function in a similar way, although it is not inhibited by non-steroidal anti-inflammatory drugs. Some
pathogenic protozoa including the Chaga's disease agent Trypanosoma cruzi produce COX-like proteins
that are distant evolutionarily from mammalian COX; thromboxane A2 is the main prostanoid found with
a little PGF2α.

3. Prostacyclin and Thromboxane Biosynthesis


Prostacyclin (PGI2) and thromboxanes are also synthesised directly from PGH2 as illustrated below.
Thus, a prostacyclin synthase converts PGH2 (synthesised by COX-2) to PGI2 (half-life 42 seconds),
while a thromboxane A2 synthase catalyses the production of TXA2 from PGH2 (synthesised by COX-1).
These enzymes are related to the cytochrome P450 group of proteins and are located on the cytosolic
face of the endoplasmic reticulum, so the precursor PGH must cross the membrane. PGI and TXA are
the main prostanoids formed in endothelial and smooth muscle cells and in platelets and lung,
respectively. In addition, PGI2 and some other prostanoids can be produced by cell-cell interactions by
using enzymes in adjacent cells, i.e. PGH2 of platelet origin is converted to PGI2 in the vascular
epithelium. Subsequently, PGI2 can be released by endothelial cells to function through a signalling
cascade with G-protein coupled receptors on nearby platelets. Similarly, prostacyclin production by
erythrocytes is at least in part dependent on PGH2 from lymphocytes. COX-2 is the enzyme that
provides PGH2 required for prostacyclin synthesis.
While platelets are able to synthesise thromboxane TXA2 from endothelial PGH2 in vitro, this is not
believed to be a major pathway in vivo. In rat peritoneal macrophages, thromboxane A synthase and
COX-1 appear to be functionally coupled in the endoplasmic reticulum. The thromboxane A2 synthase
also produces 12(S)-hydroxy-5Z,8E,10E-heptadecatrienoic acid (12-HHT) and malondialdehyde from
PGH2 by cleavage of the pentagonal ring in epithelial cells in various tissues but especially the intestine
and skin; relatively large amounts are produced in activated platelets during skin injury. This metabolite
is of special relevance to leukotriene function.

Diepoxide: Thrombin-activated human platelets generate an


eicosanoid in nanogram amounts that has been identified as
8,9‑11,12‑diepoxy-13-hydroxyeicosadienoic acid (8,9-11,12-
DiEp-13-HEDE or DiEpHEDE), which both stimulates and
primes the expression of human neutrophil integrin (it was first
erroneously identified as 8-hydroxy-9,10-dioxolane A3 (DXA3)).
It is believed to have a role in innate immunity and acute
inflammation. COX1 is the key enzyme involved in its biosynthesis from unesterified arachidonic acid.
After synthesis, it is rapidly esterified to position sn-2 of phosphatidylethanolamine in which position sn-1
is occupied by a 16:0, 18:0 or 18:1 vinyl ether or an 18:0 fatty acid; the intact phospholipid remains in
the membrane and has similar biological activity to the free eicosanoid. PGE2/D2 formed in platelets via
COX-1 are esterified in the same way. Similar endoperoxides may be formed in tissues via the co-
occurrence of LOX and cytochrome P450 or peroxygenase enzymes in tissues.

4. Prostanoid Catabolism
Prostanoids function close to the site of synthesis, and they are often deactivated before they are
exported into the circulation as inactive metabolites. Some, such as PGI and TXA, are deactivated
spontaneously, but active enzyme systems also operate, and these function primarily by reaction with
the 15(S)-hydroxyl group as discussed in the Introductory web page. Prostanoids are first transported
from the extracellular fluid to the cytoplasm by the prostaglandin transport protein (PGT) where, for
example, prostaglandin PGE2 is oxidized to 15-keto-PGE2, which was long thought to be biologically
inactive. It is now recognized that inactivation of PGE2 by 15‑hydroxyprostaglandin dehydrogenase is a
vital step in halting tumor cell proliferation, and that the product 15-keto-PGE2 is an electrophilic
molecule that functions in association with the PPARγ (see below) and other proteins to inhibit cell
proliferation. More extensive oxidation of prostanoids eventually yields metabolites such as 11α-
hydroxy-9,15-dioxo-2,3,4,5-tetranor-prostane-1,20-dioic acid or PGE-M, which is the main metabolite of
PGE2 found in urine.

The vinyl ether moiety in prostacyclin is unstable below pH 8.0, and PGI2 is rapidly deactivated non-
enzymatically by a hydrolysis reaction to form 6‑keto-PGF1α. Similarly, TXA2 contains an unstable ether
linkage, and it is deactivated by non-enzymatic hydrolysis to open the bicyclic oxygenated ring and form
inert thromboxane B2 (TXB2). A significant portion of TXB2 then undergoes dehydrogenation at C-11 by
an 11‑dehydroxythromboxane B2 dehydrogenase to form 11‑dehydro-TXB2, a metabolite found in
human blood plasma and urine, which can be monitored to assess COX-1 activity and its responses to
drug treatments.

5. The Functions of Prostanoids


Prostanoids are ubiquitous lipids in animal tissues that coordinate a multitude of physiological and
pathological processes at concentrations down to 10-9g per g of tissue, either within the cells in which
they are formed (autocrine) or in closely adjacent cells (paracrine) (they are deactivated too readily to be
transported far) in response to specific stimuli. They are transported out of cells mainly by members of
the ABC transporter superfamily. Under normal physiological conditions, they have essential
homeostatic functions in the cytoprotection of gastric mucosa, renal physiology, gestation, and
parturition, but they are also implicated in a number of pathological conditions, such as inflammation,
cardiovascular disease and cancer. Different prostanoids can have complementary or opposing
functions depending on tissue or physiological conditions and the correct balance between them can
often be crucial. Such is the complexity of these interactions that an outline only of some of the more
important can be presented here.

Receptors: Prostanoids are sometimes described as local hormones that act in an autocrine fashion
close to the site of their synthesis to coordinate the effects of other hormones in the circulation, although
some can undergo facilitated transport from the cell via specific transporters to exert paracrine actions.
In order to express their activity, they interact with specific cell-surface G-protein-linked receptors
(GPCRs) mainly. These comprise a large protein family with seven trans-membrane domains that sense
molecules outside the cell and activate signal transduction pathways inside the cell and thence the
cellular responses. When a ligand binds to a GPCR, it causes a conformational change, which allows it
to act as a guanine nucleotide exchange factor to activate an
associated G protein. Five classes (and several sub-classes) of
GPCR have been identified in the mouse and man that interact
with prostanoids, and these are specific for PGE2 (designated EP
or four subclasses EP1 to EP4), PGD2 (DP or two subclasses DP1
and DP2), PGF2α (FP), PGI2 (IP) and TXA2 (TP). The immediate
result is an increase or decrease in the rate of generation of
cytosolic second messengers (cAMP or Ca2+), a change in
membrane potential or activation of a specific protein kinase. The
different receptors characterized from diverse cell types tend to
have high, but not absolute, specificity for particular prostanoids
with characteristic functions in each cell.

Certain of the cyclopentanone prostanoids (PGA and PGJ series)


interact at the cell nucleus with peroxisome proliferator-
activated receptors (PPARs) of which there are three, but in this
context PPARγ is especially important. This is a nuclear hormone receptor or ligand-activated
transcription factor regulating the expression of genes involved in adipogenesis, glucose homeostasis
and lipid metabolism. All PPARs heterodimerize with the retinoid X receptor (RXR), which must itself be
activated by binding to 9-cis-retinoic acid, and bind to specific regions on the DNA of target genes.

The picture of prostanoid actions is complicated by the fact that a given prostanoid can have a number
of different biological functions, sometimes opposing, according to the cell type, the nature of the
stimulatory response and the type of receptor. For example, PGE2 can have either pro- or anti-
inflammatory effects depending on its interactions with one of four receptors in different cell types. The
relative activities of the two iso-enzymes COX-1 and COX-2 are also essential to an understanding of
the activity of prostanoids in any given circumstance. However, the complexity of the various interactions
can only be hinted at here.

Inflammation and immune responses: Arguably the best known of the functions of prostaglandins
and thromboxanes in cells is that they modify the inflammatory response, affecting symptoms, such as
pain, fever and swelling. It should be recognized that inflammation is an intrinsically beneficial event that
leads to removal of offending molecules and restoration of tissue structure and function. The main cause
for concern is when acute inflammation fails to resolve and causes excessive tissue damage as occurs
in sepsis, in which a prolonged hyperactive immune response is followed by an immunosuppressive
stage often leading to high mortality rates. In the early days of prostaglandin research, it was evident
that prostaglandins injected into tissues could induce all the symptoms of inflammation. However, it is
now recognized that the interactions are complex, and prostanoids can act both in a pro- and anti-
inflammatory manner according to the nature of the inflammatory stimulus and the specific prostanoid
produced, together with the profile of prostanoid receptors in a given type of cell. For example, EP3
receptors are involved in the development of fever, while EP2 and EP4 function in allergy and bone
resorption.

Under normal conditions, prostanoid levels in cells are low, but during inflammation both the
nature and concentration of prostanoids can change dramatically. For example,
macrophages produce both PGE2 and TXA2, but the ratio changes to an excess of PGE2
with an inflammatory stimulus, for example by enhancement of the release cascade of pro-
inflammatory cytokines. In these actions, prostanoids are best viewed as part of complex
regulatory networks that modulate the actions of immune cells. PGE2 in particular has potent
pro-inflammatory effects and is involved in all the processes leading to the classic signs of
inflammation, including inducing fever and enhancing pain. It can cause the transition to chronic
inflammation by acting as a cytokine amplifier. On the other hand, it has anti-inflammatory properties
also, such as suppressing lymphocyte proliferation and inhibiting the production of certain interleukins
and other cytokines. It inhibits the action of 5-lipoxygenase, which is involved in the synthesis of pro-
inflammatory leukotrienes, and stimulates the activity of the anti-inflammatory lipoxins. Therefore, PGE2
has a role in initiating the inflammatory response and in its eventual resolution. There is a particular
interest in findings that in its pro-inflammatory role PGE2 promotes the growth of colorectal tumors (see
below), and it is also involved in the pathology of rheumatoid arthritis and in respiratory diseases; with
the latter, it can have both positive and negative effects depending upon circumstances. By stimulating
the production of endothelial progenitor cells, PGE2 promotes wound healing, but PGD2 has opposing
effects; thromboxanes are also important in this context (see below). Prostaglandins with
15R‑stereochemistry are anti-inflammatory.

Prostaglandin PGF2α has an important pro-inflammatory function, especially in patients with chronic
inflammatory diseases such as rheumatoid arthritis. In contrast to its protective role in cardiovascular
disease (see below), PGI2 an important mediator of the oedema and pain that accompany acute
inflammation and it is produced rapidly following tissue injury or inflammation. For example, it is the
most abundant prostanoid in synovial fluid in human arthritic knee joints.

The high levels of prostanoids found in inflammation are presumed to be due to the recruitment of
leukocytes and the induction of the COX-2 enzyme (COX-1 appears to have a minor role only), which
then in many tissues produces the pro-inflammatory prostanoids mainly. This explains the interest in
COX-2 inhibitors for treating arthritis and other chronic inflammatory diseases. Simplistically, it is
believed that COX-2 is pro-inflammatory in the early stages of inflammation, but is beneficial at later
stages by generating anti-inflammatory prostanoids. COX-1 derived prostanoids may sustain the
inflammatory response. Inhibition of cyclooxygenases also explains the role of non-steroidal drugs, such
as aspirin, in reducing the symptoms of fever. In the brain, COX-2 is present in neurons and has been
implicated in the progression of Alzheimer's disease.

Immune responses are initiated and coordinated by T lymphocytes, which are influenced by prostanoids
in a variety of ways; they appear to modify their development and maturation. Thus, PGE2 inhibits
lymphocyte activation and proliferation, while TXA2 has opposing effects. PGE2 has beneficial functions
on innate and adaptive immune systems also by regulating immunity and host defense against viral,
fungal and bacterial pathogens, but prostanoids produced by such organisms can prolong the effects of
infection. Again, the actions of COX-2 (and COX-1) may be the key to triggering antigen-specific
inflammation.

Although PGD2 has some pro-inflammatory properties in allergic responses and in brain in the
perception of pain, it is recognized to be a key anti-inflammatory prostanoid that may be involved in the
resolution of inflammation. It is the principal ligand for two receptors, DP1 and DP2, but it is also an
agonist of the thromboxane receptor, TP. Appreciable amounts are found only in the brain and in mast
cells (eosinophils). PGD2 exerts significant anti-inflammatory effects in experimental colitis, and its
synthesis is elevated rapidly in response to tissue injury to countered the pro-inflammatory effects of
PGE2 and other chemotaxins.

Similarly, PGJ2, with Δ12-PGJ2 and the short-lived 15-deoxy-


Δ12,14-PGJ2 (15d-PGJ2), the cyclopentenone-containing J-
series of prostaglandins produced by non-enzymatic
dehydration of PGD2, are now well established as anti-
inflammatory regulators, which function as agonists for PPARγ
as discussed briefly above, although they also activate the DP2
receptor. 15d-PGJ2 enters cells and binds to PPARγ, activating it to form heterodimers with retinoid X
receptor alpha (RXRα), which bind to specific DNA sequences to activate expression of target genes
with functions in lipid metabolism, inflammatory responses and immunity. The J-series of prostaglandins
are involved in the immune response as they are produced in antigen-presenting cells such as activated
T lymphocytes. For example, 15d-PGJ2 functions in the resolution of the inflammatory response by
inducing apoptotic cell death of activated macrophages. As it contains an electrophilic α,β-unsaturated
ketone moiety in its cyclopentenone ring, it can act as an endogenous electrophile, which can undergo
Michael addition with key cellular nucleophiles such as the free cysteine residues of proteins; covalent
modifications of this type may be one mechanism by which it induces many of its biological responses.
Its effects on cancer as an inhibitor of tumorigenesis are discussed below.

Polyunsaturated fatty acids of the omega-3 family are known to have anti-inflammatory properties. One
explanation is that they inhibit the release of arachidonate from membrane phospholipids for eicosanoid
production, or they may compete with arachidonate for the same enzymes of eicosanoid biosynthesis,
for example to produce PGE3 from eicosapentaenoic acid (EPA). The 3-series prostanoids derived from
EPA have very different biological activities from those of the 2-series, and tend to be much less
inflammatory. The protectins, resolvins and maresins ('specialized pro-resolving mediators') must
also be considered in this context as they can be effective in bringing about the resolution of
inflammation. Similarly, prostaglandins derived from dihomo-γ-linolenic acid (20:3(n-6)), i.e. 1-series
prostanoids, have properties distinct from those of the 2-series, and for example, PGE1 has been shown
to suppress inflammation and promote vasodilation.

Cardiovascular effects: Two prostanoids are especially important and have essential but opposing
functions in the maintenance of vascular homeostasis, i.e. thromboxane TXA2 and prostacyclin PGI2
(although PGE2 and PGD2 are also relevant). TXA2 is synthesised mainly in platelets (which express
only COX-1), production being enhanced during platelet activation, and acting via the thromboxane
receptor TPα, it promotes platelet aggregation, vasoconstriction, and smooth muscle proliferation even
though it has a half-life of only 20-30 seconds. This is part of an essential repair mechanism for wound
healing, including damaged vessel walls, and via TP receptor signalling is responsible for timely tissue
regeneration. The thromboxane metabolite 12-HHT is especially important for skin regeneration.
However, when the damage is too great, blood clots can result with the potential to cause strokes or
heart attacks.

In contrast, PGI2 is the main product of


macro-vascular endothelial cells. It is
produced as required and exerts its effects
as a potent vasodilator locally mainly
through a specific IP receptor but also
through the cytosolic nuclear receptor
PPARβ. In addition, it inhibits platelet
aggregation and smooth muscle cell
proliferation and so contributes substantially
to myocardial protection. Both TXA2 and
PGI2 are therefore important mediators of
pathological vascular events including
thrombosis and atherogenesis, and it is
evident that the correct balance between the
two prostanoids is essential to good
cardiovascular health. The ratio of TXA2:PGI2 seems to be more important than the absolute amounts of
these mediators that are produced in vivo. In platelets and certain other cells, PGI2 is believed to
function by elevating cAMP concentrations and activating adenyl cyclase, while TXA2 has the opposite
effect. While prostacyclin can exert acute effects that are evident rapidly after adding prostacyclin to a
system, it can also exert longer term genomic effects by directing gene transcription. Further relevant
factors are increased expression and activation of the TP receptor (for TXA2) in atherosclerotic lesions,
which can directly accelerate atherogenesis and plaque growth.

The cardio-protective effect of a low dose of aspirin (81 mg/d) that has been established by clinical trials
is exerted by the irreversible long-term inhibition of platelet COX-1 and thence of TXA2 biosynthesis for
the lifetime of a platelet in the circulation (aspirin has little effect on PGI synthesis). Indeed, aspirin
appears to be the only COX inhibitor with proven cardio-protective activity and demonstrates the
causality of eicosanoids in the development of cardiovascular pathophysiologies. It may be relevant that
15(R)-PGD2, produced by aspirin-treated COX-2, inhibits aggregation of human platelets. In contrast,
there is some concern that some specific COX-2 inhibitors may have pro-thrombotic effects by inhibiting
prostacyclin synthesis relative to that of thromboxanes. In clinical practice, such potential adverse
effects of these drugs have to be balanced against positive effects in other tissues since only 1-2% of
patients are believed to be at risk. Once more, polyunsaturated fatty acids of the omega-3 family are
believed to have beneficial effects via the action of specialized pro-resolving mediators. As
inflammation promotes atherogenesis and the associated thrombotic events, there is concern that other
inflammatory prostaglandins, such as PGE2, PGD2 or PGF2α, may exert deleterious effects.

Lung: PGE2 produced by COX-2 in lung epithelial cells has robust anti-inflammatory and anti-asthmatic
effects by activating the EP2 receptor. Similarly, PGI2 signalling through the IP receptor inhibits allergic
airway inflammation, and PGI2 analogues are used to treat pulmonary arterial hypertension. The roles of
PGD2 and thromboxane A2 are more complex, but they are believed to be mainly pro-inflammatory in
relation to asthma.

Gastrointestinal system: COX-1 is always present throughout the human gastrointestinal tract, and
produces PGI2 and PGE2, which have protective effects on the gastrointestinal mucosa. Both of these
prostanoids reduce acid secretion from parietal cells, while increasing blood flow and stimulating the
secretion of mucus. In this instance, the non-steroidal anti-inflammatory drugs, such as aspirin, have
negative effects, while the COX-2 inhibitors can be beneficial. On the other hand, these findings are
challenged by studies showing that COX-2 is expressed in the intestinal mucosa, and is induced in
ulceration, for example, when large amounts of prostaglandins are produced that assist in healing.
PGD2 has beneficial effects, as discussed above.

Kidney function: Prostaglandins generated by both COX-1 and COX-2, especially PGE2, assist in the
regulation of kidney function by maintaining vascular tone, blood flow, and salt and water excretion.
PGE2 is required for the regulation of sodium re-absorption, while PGI2 (and possibly PGE2) increases
potassium secretion. In addition, PGI2 with its well-known vasodilatory properties increases renal blood
flow and the flow of fluids through the kidney. These actions are again mediated via specific receptors.

Reproductive system: Prostaglandins produced both by COX-1 and COX-2 are involved in many
aspects of reproduction in females, from ovulation and fertilization through to labour. They are produced
in the fetus and in the placenta as well as in other reproductive tissues. In particular, the synthesis of
PGE2 and PGF2α is increased appreciably during labour, and these prostaglandins are in fact used as
drugs to induce labour. PGF2α is used to induce ovulation in dairy cows and to induce abortions in
women in midtrimester. Increased levels of placental thromboxanes are produced in patients with pre-
eclampsia, a disease state during pregnancy that results in high blood pressure and often kidney failure,
and the effects can often be ameliorated by the administration of aspirin.

Adipose tissue: Prostaglandins have diverse and opposing roles in adipogenesis and adipose tissue
metabolism. For example PGE2 and PGF2α act together to inhibit the differentiation of pre-adipocytes,
while PGD2 promotes adipogenesis by acting as a ligand for PPARγ and suppressing lipolysis via its
receptor DP2. The PGJ2 series activate PPARγ to up-regulate lipid accumulation in adipocytes, while
PGI2 has broad effects upon the regulation of the life cycle of adipocytes and impacts upon terminal
differentiation.

Cancer: COX-2 is over-expressed in many cancers, including those of the breast, colon and prostate.
In particular, PGE2 produced by the enzyme occurs at much higher concentrations in tumor than in
normal tissues, while urinary concentrations of its metabolite PGE-M are considered to be biomarkers
for predicting the risk and prognosis for some cancer types. PGE2 has been shown to promote intestinal
tumor initiation and growth by silencing certain tumor suppressor and DNA repair genes via DNA
methylation. It promotes survival of tumor cells by inhibiting apoptosis and inducing
proliferation, and by increasing cell motility and migration. In addition, via its effect on the
immune system and inflammation, it has adverse effects in relation to the destruction of
tumors. In consequence both the non-steroidal anti-inflammatory drugs, such as aspirin, and
the COX-2 inhibitors have been found to have beneficial effects towards some types of
cancer. Aspirin reduces the risk of gastrointestinal cancers, for example. Also, it is
established that EP1 receptors are involved in chemically induced colon cancer. In contrast,
both pro- and anti-tumorigenic activities have been demonstrated for PGD2 depending on
the experimental model. Similarly, PGE3, derived from the n-3 eicosapentaenoic acid (EPA),
has anti-proliferative activity in various cancers, possibly by interfering with PGE2 activity.

Thromboxane TXA4 is a pro-carcinogenic mediator that affects a number of tumor cell survival
pathways, including cell proliferation, apoptosis and metastasis, and its activity is again balanced by that
of prostacyclin PGI2.

15-Deoxy-Δ12,14-PGJ2 (15d-PGJ2), a potent anti-inflammatory regulator that functions via its interaction
with PPARγ, also regulates adipogenesis and tumorigenesis and is produced by a variety of cells. An
active transport system may carry it to the cells where it is required, and thence it is transported into the
nucleus, where it affects gene transcription. Unlike PGE2, 15d-PGJ2 is a potent anti-tumor agent,
inhibiting tumor growth both in vitro and in vivo in many tissues. It appears to act in a number of ways,
for example directly by inhibiting proliferation and stimulating apoptosis. Also, it can interact indirectly to
inhibit migration of tumor cells, and it can affect surrounding cells to reduce the expression of key
receptors. However, some experimental conditions have been identified in which it exerts contrary
effects. In general, PGE2 and 15d-PGJ2 have profound but opposing effects on tumorigenesis. It is
evident that the prostaglandin synthases that are responsible for their biosynthesis are likely to be key
targets for the development of anticancer drugs.

Stem cells: Signalling by Wnt proteins, a family of proteolipids containing covalently esterified
palmitoleic acid, controls the self-renewal of hematopoietic stem cells and bone marrow repopulation.
Activation of this process requires prostaglandin E2, and it has been suggested that the PGE2/Wnt
interaction is a master regulator of vertebrate regeneration and recovery in stem cells and other organ
systems.

Protein metabolism: γ-Keto aldehydes such as the levuglandins (see above) and
isolevuglandins, the latter produced in an analogous manner to the isoprostanes, have a remarkable
reactivity towards proteins, forming adducts with greatly modified biological functions. Thus, these di-
aldehydes react with lysyl residues on proteins to form first Schiff base adducts and thence pyrrole
derivatives, which are able to form intra- and intermolecular protein-protein cross-links. Pyrrole adducts
are in turn sensitive to oxygen and are further oxidized in vivo to stable lactam and hydroxylactam
products. Protein adducts of this type are not at all easy to analyse, but those in brain have been
correlated with the severity of Alzheimer’s disease, for example. Indeed, levuglandins and
isolevuglandins are believed to be among the most potent neurotoxic products of lipid oxidation.

Levuglandins also react with phosphatidylethanolamine, which are readily oxidized to hydroxy-
lactam derivatives that may be better markers of oxidative injury from a practical standpoint, as they are
more easily analysed.

Insects: Prostaglandins have a wide range of downstream signalling functions in insects as in


vertebrates, including hormone actions in the fat body and effects upon reproduction, fluid secretion, and
the immune response. As a means of increasing their virulence, some insect pathogens target
phospholipase A2 to reduce prostaglandin biosynthesis and bring about immunosuppression.

Parasitic infections: It has been established that a number of parasitic organisms produce
prostaglandins in the same way as their mammalian hosts, and by similar enzymic mechanisms. They
may play a part in the pathogenesis of parasitic diseases.

6. Some Exotic Prostanoids


Marine invertebrates, including sponges, corals, and molluscs, contain a wide range of prostaglandins,
many of which are of the conventional type such as PGE2, PGF2 and so forth. They are presumed to
perform similar functions as in mammals, and are also involved in the regulation of oogenesis and
spermatogenesis, in ion transport and perhaps as defence compounds. One species of coral (Plexaura
homomalla) contains up to 8% of its dry mass as prostanoid esters. However, these have the 15-
hydroxyl group in the (R)- rather than the S-configuration, so can have very different biological
properties from the conventional prostanoids in that they are potent anti-inflammatory agents. In many
marine invertebrates, the prostaglandins exist largely in esterified form or as lactones rather than as the
free acids.

In addition, a number of novel prostanoids have been discovered in corals, some examples of which are
illustrated above, which differ in stereochemistry from the typical prostanoids, or contain acetyl groups,
or are substituted with halogen atoms, such as chlorine or bromine. Little is known of the biochemistry or
function of the "clavulones, bromovulones or punaglandins" in marine organisms, but there is increasing
interest in them because of reported anti-tumor activities.

It is perhaps more surprising that some red algae such as Gracilaria species contain prostaglandins
(PGE2, PGF2α and others) and are known to have a cyclooxygenase gene. Although the function of
these oxylipins in the organisms is not known, they may have a defensive function as they are believed
to be causative toxins for lethal food poisonings that have occurred in Japan.

7. Prostanoid Analysis
Analysis of prostanoids is not a simple task because they occur at such low levels in tissues and
because of their high reactivity. Extraction must be carried out under mild conditions as rapidly as
possible, and solid-phase extraction methods are now available that set the standard. Subsequent
analysis usually involves HPLC linked to mass spectrometry. Chiral chromatography can be used to
distinguish between prostaglandins and isoprostanes. Immunoassays are available that may be suitable
for some clinical applications, but they are not sensitive to minor differences in prostanoid structure.

Suggested Reading
Buczynski, M.W., Dumlao, D.S. and Dennis, E.A. Proteomics. An integrated omics analysis of eicosanoid
biology. J. Lipid Res., 50, 1015-1038 (2009); DOI.
Dennis, E.A. and Norris, P.C. Eicosanoid storm in infection and inflammation. Nature Rev. Immunol., 15, 511-
523 (2015); DOI.
Di Costanzo, F. Di Dato, V., Ianora, A. and Romano, G. Prostaglandins in marine organisms: a review.
Marine Drugs, 17, 428 (2019); DOI
Gabbs, M., Leng, S., Devassy, J.G., Monirujjaman, M. and Aukema, H.M. Advances in our understanding of
oxylipins derived from dietary PUFAs. Adv. Nutr., 6, 513-540 (2015); DOI.
Lagarde, M. and Nicolaou, A. (Editors) Oxygenated metabolism of PUFA: analysis and biological relevance.
Biochim. Biophys. Acta (Volume 1851, Issue 4, Pages 307-518) (2015) - special issue.
Li, J.J., Guo, C.Y. and Wu, J.Y. 15-Deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2), an endogenous ligand of
PPAR-γ: function and mechanism. PPAR Res., 7242030 (2019); DOI.
Maseda, D., Ricciotti, E. and Crofford, L.J. Prostaglandin regulation of T cell biology. Pharmacol. Res., 149,
104456 (2019); DOI
Mitchell, J.A. and Kirkby, N.S Eicosanoids, prostacyclin and cyclooxygenase in the cardiovascular system.
Brit. J. Pharm., 176, 1038-1050 (2019); DOI.
Niu, M.Y. and Keller, N.P. Co-opting oxylipin signals in microbial disease. Cell. Microbiol., 21, e13025 (2019);
DOI.
Peebles, R.S. Prostaglandins in asthma and allergic diseases. Pharmacol. Therapeut., 193, 1-19 (2019);
DOI.
Pluchart, H., Khouri, C., Blaise, S., Roustit, M. and Cracowski, J.L. Targeting the prostacyclin pathway:
beyond pulmonary arterial hypertension. Trends Pharm. Sci., 38, 512-523 (2017); DOI.
Seo, M.-J. and Oh, D.-K. Prostaglandin synthases: molecular characterization and involvement in
prostaglandin biosynthesis. Prog. Lipid Res., 66, 50-68 (2017); DOI.
Smith, M.L. and Murphy, R.C. The eicosanoids: cyclooxygenase, lipoxygenase and epoxygenase pathways.
In: Biochemistry of Lipids, Lipoproteins and Membranes (6th Edition). pp. 260-296 (Edited by N.D. Ridgeway
and R.S. McLeod, Elsevier, Amsterdam) (2016) - see Science Direct.
Smith, W.L., Urade, Y. and Jakobsson, P.-J. Enzymes of the cyclooxygenase pathways of prostanoid
biosynthesis. Chem. Rev., 111, 5821-5865 (2011); DOI.
Stanley, D. and Kim, Y. Prostaglandins and other eicosanoids in insects: biosynthesis and biological actions.
Front. Physiol., 9, 1927 (2019); DOI.
Sugimoto, Y., Inazumi, T. and Tsuchiya, S. Roles of prostaglandin receptors in female reproduction. J.
Biochem., 157, 73-80 (2015); DOI.
Tsuge, K., Inazumi, T., Shimamoto, A. and Sugimoto, Y. Molecular mechanisms underlying prostaglandin E2-
exacerbated inflammation and immune diseases. Int. Immun., 31, 597-606 (2019); DOI.
Wang, D.Z. and DuBois, R.N. Role of prostanoids in gastrointestinal cancer. J. Clin. Invest., 128, 2732-2742
(2018); DOI.
Wang, Y., Armando, A.M., Quehenberger, O., Yan, C. and Dennis, E.A. Comprehensive ultra-performance
liquid chromatographic separation and mass spectrometric analysis of eicosanoid metabolites in human
samples. J. Chromatogr. A, 1359, 60-69 (2014); DOI.
Zhang, M., Li, W. and Li, T. Generation and detection of levuglandins and isolevuglandins in vitro and in vivo.
Molecules, 16, 5333-5348 (2011); DOI.

Updated: February Author: William W.


Credits/disclaimer
11th, 2020 Christie

This Version Copyright © William W. Christie (2020)

You might also like