Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

Molecular and Cellular Endocrinology 248 (2006) 3–8

11␤-Hydroxysteroid dehydrogenase type 1—A role in inflammation?


Karen E. Chapman a,∗ , James S. Gilmour a,b , Agnes E. Coutinho a,b ,
John S. Savill b , Jonathan R. Seckl a
a Endocrinology Unit, Centre for Cardiovascular Sciences, The Queen’s Medical Research Institute, University of Edinburgh,
47 Little France Crescent, Edinburgh EH16 4TJ, UK
b MRC Centre for Inflammation Research, The Queen’s Medical Research Institute,

University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK

Abstract
Glucocorticoids are widely used for their potent anti-inflammatory effects. Endogenous glucocorticoids are immunomodulatory and shape
both adaptive and innate immune responses. Over the past decade, it has become apparent that an important level of control over endogenous
glucocorticoid action is exerted by the 11␤-hydroxysteroid dehydrogenase enzymes. The type 1 enzyme, 11␤-HSD1, reduces inert glucocorticoids
into active forms, thereby increasing intracellular ligand availability to receptors. Although 11␤-HSD1 activity has been shown to play an important
role in the metabolic actions of glucocorticoids, its role in the immune response has, until recently, remained unclear. Here we review recent evidence
pertaining to the role of 11␤-HSD1 in the inflammatory response.
© 2005 Elsevier Ireland Ltd. All rights reserved.

Keywords: Glucocorticoid; 11␤-hydroxysteroid dehydrogenase; Steroid metabolism; Macrophage; Inflammation

1. Perspective coids, those that express 11␤-HSD1 effectively get a “double


whammy” from circulating active glucocorticoids as well as
Glucocorticoids exert potent immunomodulatory effects. At from the local reactivation of intrinsically inert glucocorticoids.
pharmacological levels they are powerful immunosuppressive This provides a mechanism to locally amplify glucocorticoid
and anti-inflammatory agents. Clinically, they are widely pre- levels, thus targeting their effects to where they are most needed.
scribed to treat inflammation, where often they are the most Here, we review recent data demonstrating expression of 11␤-
effective therapy and their utility is limited only by the side HSD1 in leukocytes and its potential anti-inflammatory role.
effects of long term treatment. The first use of glucocorticoids
to treat inflammation in humans was in the late 1940s, by Philip
2. Inflammation and its resolution
Hench, who in collaboration with Edward Kendall, success-
fully used cortisone to treat rheumatoid arthritis, for which
2.1. The inflammatory response as an innate response to
they were awarded a Nobel Prize in 1950 (Hench, 1950). Of
injury or infection
course, to be effective, the intrinsically inert cortisone must
first be converted to the active glucocorticoid, cortisol, by the
The inflammatory response, part of the innate immune sys-
intracellular action of 11␤-hydroxysteroid dehydrogenase type
tem, is a first line host defence mechanism that, upon injury,
1 (11␤-HSD1). Paradoxically, glycyrrhetinic acid, a deriva-
protects against invading pathogens and repairs damaged tissue.
tive of liquorice which inhibits 11␤-HSD, also shows anti-
The classic signs of inflammation include redness, swelling,
inflammatory activity (Finney and Somers, 1958), presumably
heat and pain and result from the release of proinflammatory
through delaying the clearance of cortisol (or corticosterone)
mediators (e.g. tumour necrosis factor-␣; TNF-␣) and increased
from blood by renal 11␤-HSD2. However, whereas cells that
vasodilation and capillary permeability, promoting migration of
express 11␤-HSD2 are resistant to the effects of glucocorti-
leukocytes into injured tissues. Activated neutrophil granulo-
cytes are generally the first cells attracted to the site of inflam-
Abbreviations: 11␤-HSD, 11␤-hydroxysteroid dehydrogenase
mation, rapidly followed by monocyte emigration from blood
∗ Corresponding author. Tel.: +44 131 242 6736. vessels and their subsequent maturation into macrophages. Once
E-mail address: Karen.Chapman@ed.ac.uk (K.E. Chapman). at the inflamed site, neutrophils undergo constitutive apoptosis,

0303-7207/$ – see front matter © 2005 Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.mce.2005.11.036
4 K.E. Chapman et al. / Molecular and Cellular Endocrinology 248 (2006) 3–8

functionally isolating them from the inflammatory environment prevent its potentially lethal overshoot (Munck et al., 1984).
by loss of stimulated chemotaxis, phagocytosis, degranulation Support for this view has come from experiments in adrenalec-
and respiratory burst (Haslett et al., 1991; Whyte et al., 1993), tomised rats, in which glucocorticoid treatment is essential to
whilst at the same time, facilitating safe removal of their poten- survive challenge with horse serum (Dougherty and Schneebeli,
tially histotoxic contents by macrophages (Savill et al., 2002). 1955), bacterial lipopolysaccharide (LPS) or tumour necrosis
factor-␣ (TNF-␣) (Bertini et al., 1988). However, the view that
2.2. The importance of resolution of inflammation glucocorticoid actions are solely immunosuppressive is over-
simplistic; physiological levels exert permissive and stimula-
The inflammatory response is largely beneficial, and acute tory effects as well as suppressive and are better described as
inflammation normally quickly resolves. However, it may immunomodulatory (reviewed in McEwen et al. (1997); Munck
become persistent, leading to chronic disease and irreversible et al. (1984); Yeager et al. (2004)). Glucocorticoids potently alter
organ damage. The hallmark of chronic inflammation is the sus- leukocyte distribution/trafficking, alter cellular differentiation
tained presence and accumulation of activated macrophages, programmes and alter transcription of numerous genes in the
which continue to release proinflammatory mediators and are glucocorticoid sensitive leukocytes; monocytes/macrophages,
thus responsible for much of the tissue damage. Although many neutrophils and granulocytes (Ashwell et al., 2000; McEwen
of the initial events in an inflammatory response have been char- et al., 1997). Notably, glucocorticoids repress transcription
acterised, the mechanisms responsible for its resolution remain of genes encoding proinflammatory mediators – particularly
incompletely understood. It is likely that chronic inflammation, cytokines and their receptors – whilst increasing transcription
such as occurs during rheumatoid arthritis or asthma, results of anti-inflammatory proteins including IL-10 and IL-1 receptor
from an imbalance between the initial proinflammatory events antagonist (Barnes, 1998).
needed for adequate protection against infection and the mecha-
nisms that terminate the inflammatory response, restoring home- 3.2. Glucocorticoids promote the resolution of
ostasis (Haslett, 1999). inflammation

2.3. The role of the macrophage The mechanisms by which glucocorticoids promote reso-
lution of inflammation have received less attention than those
Crucial to the rapid and safe resolution of acute inflamma- associated with suppression of the initial inflammatory response.
tion is macrophage clearance of leukocytes undergoing apop- Recent work from Edinburgh is elucidating some of the mech-
tosis at sites of inflammation (Botto et al., 1998; Savill et al., anisms that may account for the efficacy of glucocorticoids in
2002; Taylor et al., 2000). Unlike phagocytosis of necrotic resolving inflammation. Synthetic glucocorticoids increase the
cells or invading pathogens, the ingestion of apoptotic cells by capacity of individual macrophages to ingest multiple apoptotic
phagocytes does not evoke a proinflammatory response (Fadok cells (Liu et al., 1999). Moreover, exposure of blood mono-
et al., 1998; Meagher et al., 1992). In contrast, active anti- cytes to glucocorticoids, at the beginning of their maturation
inflammatory and immunosuppressive responses are elicited into macrophages, reprogrammes their differentiation into a
following phagocytosis of apoptotic cells (Voll et al., 1997). If highly phagocytic “anti-inflammatory” macrophage phenotype
the removal of apoptotic granulocytes is impaired, they rapidly (Giles et al., 2001; Heasman et al., 2003), a phenomenon that
undergo secondary necrosis, provoking release of proinflamma- can be modulated by local cytokine environment (Heasman
tory mediators from the phagocyte and prolonging the inflam- et al., 2003). In addition, glucocorticoids modify granulocyte
matory response (Botto et al., 1998; Taylor et al., 2000). The apoptosis—accelerating eosinophil apoptosis whilst delaying
macrophage is the principal phagocyte. However, under cer- neutrophil apoptosis (Meagher et al., 1996). Thus, the pro-
tain circumstances, other cell types, e.g. glomerular mesangial resolution effects of glucocorticoids upon inflammation are
cells (Hughes et al., 1997) are able to phagocytose apoptotic complex—mediated by several cell types and dependent upon
cells. Although blood monocytes can bind apoptotic cells, they the local (cytokine) environment.
lack phagocytic capacity (Newman et al., 1982; Savill et al.,
1989) and little is known of the mechanisms that operate early 3.3. Influence of endogenous glucocorticoids and HPA
in the acute inflammatory response to promote the acquisition activity upon the inflammatory response
by maturing monocytes of phagocytic capacity for apoptotic
leukocytes. However, recent data suggest this can be profoundly It has been proposed that the effects of glucocorticoids
influenced by glucocorticoids. follow a bell-shaped dose response curve, with permissive
and stimulatory effects exerted at physiological concentra-
3. Glucocorticoids and the inflammatory response tions and suppressive effects at high pharmacological con-
centrations (Munck and Naray-Fejes-Toth, 1992; Sapolsky et
3.1. Glucocorticoids suppress the initiation of the al., 2000). Thus, whilst pharmacological levels of glucocorti-
inflammatory response coids are immunosuppressive, the effects of physiological levels
are immunomodulatory. Considerable evidence implicates the
It has been hypothesised that endogenous glucocorticoids hypothalamic–pituitary–adrenal (HPA) axis, governing the out-
provide a “brake” during the initial inflammatory response, to put of glucocorticoid from the adrenal gland, in susceptibility to
K.E. Chapman et al. / Molecular and Cellular Endocrinology 248 (2006) 3–8 5

inflammatory and autoimmune disease (Sternberg, 2001). Fis- HSD2. Intracellular glucocorticoid concentrations can there-
cher 344/N rats, which have larger adrenals and mount a greater fore differ from plasma glucocorticoid levels and depend upon
corticosterone response to stress than the genetically related the 11␤-HSD isozyme expressed. The many and varied roles
Lewis/N rat are resistant to the development of arthritis induced of 11␤-HSD1 are now being elucidated—particularly impor-
by streptococcal cell wall, whereas Lewis/N rats are susceptible, tant in this has been the generation of mice deficient in 11␤-
a difference that can be reversed by manipulation of gluco- HSD1 (Kotelevtsev et al., 1997). These mice cannot reduce
corticoid action in these strains (Sternberg et al., 1992). Sim- 11-dehydrocorticosterone to corticosterone (Kotelevtsev et al.,
ilarly, development of experimental allergic encephalomyelitis 1997) and are protected against the adverse metabolic effects of
in rats is strongly influenced by endogenous glucocorticoids glucocorticoids (Kotelevtsev et al., 1997; Morton et al., 2001;
(Mason, 1991). Importantly, neonatal exposure of rats to bacte- Morton et al., 2004) and glucocorticoid-associated cognitive
rial LPS, which programmes lifelong changes in HPA activity, decline (Yau et al., 2001). Although they have normal amounts
also strongly influences susceptibility to inflammatory disease in of adipose tissue, at least on a low fat diet (Morton et al., 2004),
adulthood (Shanks et al., 2000). Increased glucocorticoid (HPA they lack bone marrow adipocytes (Justesen et al., 2004); the
axis) activity shifts the balance of an immune response from a functional relevance of this is currently unclear. However, until
type 1-associated (proinflammatory) cytokine profile to a type very recently, immune function, and particularly the inflamma-
2-associated (anti-inflammatory) cytokine profile (McEwen et tory response, had not been investigated in these mice.
al., 1997). Whilst this is beneficial during normal progression
of an inflammatory/immune response, if the balance is altered 4.1. 11β-HSD1 is expressed in immune cells, including
in favour of the type 2 response prematurely, then a suffi- macrophages
ciently robust response may not be mounted in the early stages
of the inflammatory response, allowing progression to chronic Previous studies with the liquorice-derived 11␤-HSD
inflammation. Thus, initially following injury, it is important to inhibitor glycyrrhetinic acid suggested a role for 11␤-HSDs in
contain adventitious infections and a strong type 1 response is immunity (Finney and Somers, 1958; Hennebold et al., 1997;
required for this. However, an unregulated response is poten- Pompei et al., 1979), although as mentioned above, the anti-
tially lethal. Endogenous glucocorticoids maintain the balance viral and anti-inflammatory effects are probably attributable to
between the early control and elimination of infection follow- 11␤-HSD2 inhibition (Horigome et al., 1999), rather than 11␤-
ing injury and overshoot leading to tissue damage. Perhaps the HSD1 inhibition. Older studies on 11␤-HSD function in lym-
ability to switch from this early phase of infection control, to the phocytes measured 11␤-HSD dehydrogenase activity or failed
later pro-resolution phase of tissue repair is similarly influenced to discriminate between the isozymes (Berliner and Dougherty,
by endogenous glucocorticoids. 1964; Klein et al., 1980)—it remains unclear whether these
data pertain to 11␤-HSD1 or 11␤-HSD2 activity. More recently,
4. 11␤-HSD1 11␤-HSD1 expression has been described in immune cells.
In 2000, 11␤-HSD1 was identified in a subtractive hybridisa-
The enzymatic conversion of cortisone to cortisol by 11␤- tion screen as induced during the differentiation of a human
HSD was described by Amelung et al. (1953), shortly after myelomonocytic cell line (Gingras and Margolin, 2000). Sub-
the award of the 1950 Nobel Prize to Hench. However, it was sequent work by Thieringer showed that although 11␤-HSD1
almost 40 years later that it became clear that there are two is not expressed in circulating human monocytes, it is induced
enzymes responsible for the interconversion of cortisone and upon differentiation to macrophages (Thieringer et al., 2001).
cortisol; 11␤-HSD1 which predominantly reactivates and 11␤- Furthermore, 11␤-HSD1 expression could be induced in mono-
HSD2, which inactivates glucocorticoids in vivo (reviewed in cytes by the “type 2” response cytokines IL-4 and IL-13, an
Seckl and Walker (2001); Tomlinson and Stewart (2001) and effect that was abrogated in the presence of proinflammatory
elsewhere in this volume). Whilst the importance of the liver to IFN-␥ (Thieringer et al., 2001). Somewhat confusingly, in the
the regeneration of blood cortisol from cortisone (or corticos- same study, LPS was shown to increase 11␤-HSD1 expres-
terone from 11-dehydrocorticosterone in rodents) has long been sion in the macrophage-like cell line, THP-1 (Thieringer et al.,
known, the role of 11␤-HSD1 in the local regeneration of glu- 2001). We have confirmed expression of 11␤-HSD1 in human
cocorticoids, within cells, from inert substrate has only recently monocyte-derived macrophages (Gilmour, 2003) and have also
been fully recognised. The 11␤-HSDs play a dual role in glu- demonstrated expression of 11␤-HSD1, but not 11␤-HSD2,
cocorticoid action. Firstly, adrenal secretion of glucocorticoids in mouse macrophages (Gilmour et al., 2005). Our own data
into the circulation is influenced by the balance between hep- in human cells suggest that IL-4 may accelerate macrophage
atic 11␤-HSD1 (reactivation) and renal 11␤-HSD2 (clearance) differentiation, rather than directly induce 11␤-HSD1 activity
activities and secondly, within specific cell types, the enzymes (Gilmour, 2003). Recently, 11␤-HSD1 expression and activ-
gate glucocorticoid access to glucocorticoid receptors. Thus, ity have been described in mouse and human dendritic cells
cells which express 11␤-HSD2 are glucocorticoid resistant, (Freeman et al., 2005; Zhang et al., 2005) as well as mouse T and
whereas cells that express 11␤-HSD1 boost the glucocorticoid B lymphocytes (Zhang et al., 2005). Interestingly, in immature
levels which access receptors, through intracellular regener- human dendritic cells activated by proinflammatory mediators,
ation of the ample circulating inert 11keto-steroid substrate, including TNF-␣ or LPS (innate immune activating signals),
levels of which depend primarily on the action of renal 11␤- expression of 11␤-HSD1 was maintained, whereas maturation
6 K.E. Chapman et al. / Molecular and Cellular Endocrinology 248 (2006) 3–8

induced by CD40 ligation (a signal associated with adaptive ineffective in 11␤-HSD1−/− macrophages (Gilmour et al.,
immune activation) markedly reduced expression (Freeman et 2005). Crucially, during sterile peritonitis, the acquisition of
al., 2005). Thus, 11␤-HSD1 expression in monocyte-derived macrophage phagocytic capacity was delayed in 11␤-HSD1-
cells is exquisitely sensitive to differentiation state and activating deficient mice compared to control mice, and these mice showed
signals. We have recently used a mouse model of acute inflam- delayed clearance of apoptotic neutrophils as experimental peri-
mation, thioglycollate-induced sterile peritonitis, to show a rapid tonitis progressed (Gilmour et al., 2005). Based on these data
increase (within 3 h) in 11␤-HSD1 activity in cells elicited to the and the evidence pertaining to its regulation by proinflammatory
peritoneum following thioglycollate injection (Gilmour et al., mediators, we have proposed that 11␤-HSD1 is a component of
2005), consistent with induction by proinflammatory mediators. mechanisms that are engaged early in the acute inflammatory
Levels remained high in peritoneal macrophages until resolu- response in order to promote later resolution (Gilmour et al.,
tion, when activity returned to that of resident (unstimulated) 2005).
macrophages (Gilmour et al., 2005). A similar role for 11␤-HSD1 in shaping the immune response
has previously been proposed (Rook et al., 2000). Initially,
4.2. Regulation of 11β-HSD1 by inflammatory mediators following M. tuberculosis infection in mice, type 1 (IFN-␥,
IL-2) and proinflammatory (IL-1, TNF-␣) cytokines predom-
Numerous lines of evidence show that proinflammatory inate, associated with an 11␤-HSD equilibrium in lung favour-
mediators, especially TNF-␣ and IL-1, are potent inducers ing reductase. This enhances local glucocorticoid availability,
of 11␤-HSD1 expression and activity in a variety of cell polarising the subsequent response towards type 2 (IL-4), and
types. Mesangial cells, which share some characteristics with decreased IL-1 and TNF-␣ expression. The latter phase of type
macrophages, showed increased activity of 11␤-HSD1 with the 2 cytokine production was associated with a switch in the 11␤-
proinflammatory cytokines, TNF-␣ and IL-1␤ (Escher et al., HSD equilibrium to increased dehydrogenase. Although it is
1997). Both TNF-␣ and IL-1␤ increased 11␤-HSD1 expression not yet clear to what extent these shifts of the 11␤-HSD equi-
in primary cultures of human preadipocytes (Tomlinson et al., librium reflect alterations in local expression of 11␤-HSD1,
2001), a cell type with a lineage closely related to macrophages 11␤-HSD2 or both, it is nevertheless apparent that local (11␤-
(Charriere et al., 2003). During ovulation, a process akin to an HSD-mediated) alterations in glucocorticoid availability may be
inflammatory response with “injury” (ovulation) and “resolu- actively shaping the immune/inflammatory response as it pro-
tion” (luteinising) phases, there is a reciprocal switch in expres- gresses. Further characterisation of the immune/inflammatory
sion of 11␤-HSD1 and 11␤-HSD2 in granulosa cells. Prior to response in 11␤-HSD1−/− mice will illuminate the role of 11␤-
ovulation, 11␤-HSD2 is expressed, rendering granulosa cells HSD1.
glucocorticoid insensitive. By contrast, uniquely 11␤-HSD1 is
expressed in luteinised granulosa cells, where expression is
further increased by IL-1␤, enhancing local production of gluco- 5. Future prospects
corticoid (Tetsuka et al., 1999, 1997). Ovarian surface epithelial
cells also express 11␤-HSD1, and again, expression is increased Macrophages play an essential role in the innate immune
by IL-1 (Yong et al., 2002). Thus, induction of 11␤-HSD1 system. However, activated macrophages are also central to the
expression and activity by proinflammatory-like mediators in pathogenesis of human diseases, including chronic inflamma-
the ovary may serve to minimise inflammatory tissue damage tory conditions and atherogenesis. Given the well known adverse
following ovulatory rupture. A similar reciprocal regulation of cardiometabolic effects of glucocorticoids and the expression
11␤-HSD1 and 2 has been described in human MG-63 osteosar- of 11␤-HSD1 in macrophages, there is clear potential for
coma cells (Cooper et al., 2001) and human aortic smooth macrophage 11␤-HSD1 to influence atherogenesis (reviewed
muscle cells (Cai et al., 2001). In both cases, IL-1␤ or TNF- by Theringer, this volume).
␣ potently down-regulated 11␤-HSD2 expression, concomitant Elucidation of the transcriptional regulation of 11␤-HSD1 in
with up-regulation of 11␤-HSD1 (Cai et al., 2001; Cooper et immune cells, particularly macrophages, will provide a greater
al., 2001). Thus a consistent pattern emerges, with proinflam- understanding of the part the enzyme plays in the inflamma-
matory mediators, particularly TNF-␣ and IL-1␤, increasing tory response. The C/EBP family of transcription factors exert
11␤-HSD1 expression, promoting local glucocorticoid avail- a major control over 11␤-HSD1 transcription (Williams et al.,
ability and hence local anti-inflammatory action. 2000) and family members play key roles in leukocyte differen-
tiation programmes (Chen et al., 1997; Sieweke and Graf, 1998;
4.3. 11β-HSD1−/− mice show a delay in the acquisition of Tavor et al., 2002) as well as the inflammatory response (Poli,
macrophage phagocytic capacity 1998). Whether they are as much a requirement for 11␤-HSD1
transcription in leukocytes as they are in metabolic tissues (e.g.
In vitro, corticosterone similarly promotes phagocytosis of liver and adipose tissue) remains to be determined. Our recent
apoptotic neutrophils by bone marrow derived macrophages, data has shown that in some tissues 11␤-HSD1 transcription ini-
irrespective of whether the macrophages originate from tiates from a novel upstream promoter (Bruley et al., submitted
11␤-HSD1−/− or control mice (Gilmour et al., 2005). 11- for publication); again, future work will establish whether this
Dehydrocorticosterone was equally effective in promoting is of relevance in immune cells. The benefits of glucocorti-
phagocytosis by control macrophages, but was completely coid therapy in arthritis and other inflammatory conditions is
K.E. Chapman et al. / Molecular and Cellular Endocrinology 248 (2006) 3–8 7

well accepted. A clearer understanding of the role and regu- drogenase type 1 permits regulation of glucocorticoid bioavailability by
lation of 11␤-HSD1 in macrophages and other leukocytes will human dendritic cells. Blood 106, 2042–2049.
Giles, K.M., Ross, K., Rossi, A.G., Hotchin, N.A., Haslett, C., Dransfield, I.,
have important therapeutic implications for rheumatoid arthritis,
2001. Glucocorticoid augmentation of macrophage capacity for phagocy-
atherogenesis and other inflammatory diseases. tosis of apoptotic cells is associated with reduced p130Cas expression,
loss of paxillin/pyk2 phosphorylation, and high levels of active Rac. J.
Acknowledgements Immunol. 167, 976–986.
Gilmour, J.S., 2003. Glucocorticoids, 11␤-hydroxysteroid dehydrogenases
and macrophage function. Ph.D. Thesis. University of Edinburgh.
Work in the authors’ laboratories is funded by The Wellcome Gilmour, J.S., Cailhier, J.-F., Man, T.Y., Coutinho, A.E., Clay, M., Thomas,
Trust, The National Kidney Research Fund and The Medical G., Harris, H.J., Mullins, J.J., Seckl, J.R., Savill, J.S., Chapman, K.E.,
Research Council. JSG was supported by a 4 year Ph.D. award 2005. Local amplification of glucocorticoids by 11␤-hydroxysteroid dehy-
from the Wellcome Trust. We thank members of the Centre for drogenase type 1 promotes macrophage phagocytosis of apoptotic leuko-
Inflammation Research and the Endocrinology Unit for many cytes, submitted for publication.
Gingras, M.C., Margolin, J.F., 2000. Differential expression of multiple unex-
stimulating discussions. pected genes during U937 cell and macrophage differentiation detected
by suppressive subtractive hybridization. Exp. Hematol. 28, 65–76.
References Haslett, C., 1999. Granulocyte apoptosis and its role in the resolution and
control of lung inflammation. Am. J. Respir. Crit. Care Med. 160, S5–S11.
Amelung, D., Hubener, H.J., Roka, L., Meyerheim, G., 1953. Conversion of Haslett, C., Lee, A., Savill, J.S., Meagher, L., Whyte, M.K., 1991. Apoptosis
cortisone to compound F. J. Clin. Endocrinol. Metab. 13, 1125–1126. (programmed cell death) and functional changes in aging neutrophils.
Ashwell, J.D., Lu, F.W., Vacchio, M.S., 2000. Glucocorticoids in T cell devel- Modulation by inflammatory mediators. Chest 99, 6S.
opment and function. Ann. Rev. Immunol. 18, 309–345. Heasman, S.J., Giles, K.M., Ward, C., Rossi, A.G., Haslett, C., Dransfield,
Barnes, P.J., 1998. Anti-inflammatory actions of glucocorticoids: molecular I., 2003. Glucocorticoid-mediated regulation of granulocyte apoptosis and
mechanisms. Clin. Sci. (Lond.) 94, 557–572. macrophage phagocytosis of apoptotic cells: implications for the resolu-
Berliner, M.L., Dougherty, T.F., 1964. Interconversion of cortisol and corti- tion of inflammation. J. Endocrinol. 178, 29–36.
sone by normal and leukemic murine lymphocytes. Acta Unio. Int. Contra. Hench, P.S., 1950. The reversibility of certain rheumatic and non-rheumatic
Cancrum. 20, 1133–1136. conditions by the use of cortisone or of the pituitary adrenocorticotrophic
Bertini, R., Bianchi, M., Ghezzi, P., 1988. Adrenalectomy sensitizes mice to hormone. In: Nobel Lectures, Physiology or Medicine. Elsevier, Amster-
the lethal effects of interleukin-1 and tumor necrosis factor. J. Exp. Med. dam, pp. 1942–1962.
167, 1708–1712. Hennebold, J.D., Mu, H.H., Poynter, M.E., Chen, X.P., Daynes, R.A., 1997.
Botto, M., Dell’Agnola, C., Bygrave, A.E., Thompson, E.M., Cook, H.T., Active catabolism of glucocorticoids by 11␤-hydroxysteroid dehydroge-
Petry, F., Loos, M., Pandolfi, P.P., Walport, M.J., 1998. Homozygous C1q nase in vivo is a necessary requirement for natural resistance to infection
deficiency causes glomerulonephritis associated with multiple apoptotic with Listeria monocytogenes. Int. Immunol. 9, 105–115.
bodies. Nat. Genet. 19, 56–59. Horigome, H., Horigome, A., Homma, M., Hirano, T., Oka, K.,
Bruley, C., Lyons, V., Worsley, A.G.F., Wilde, M.D., Darlington, G.D., 1999. Glycyrrhetinic acid-induced apoptosis in thymocytes: impact of
Morton, N.M., Seckl, J.R., Chapman, K.E. A novel promoter for the 11␤-hydroxysteroid dehydrogenase inhibition. Am. J. Physiol. 277,
11␤-hydroxysteroid dehydrogenase type 1 gene is active in lung and is E624–E630.
C/EBP␣-independent, submitted for publication. Hughes, J., Liu, Y., Van Damme, J., Savill, J., 1997. Human glomerular
Cai, T., Wong, B., Mundt, S.S., Thieringer, R., Wright, S.D., Hermanowski- mesangial cell phagocytosis of apoptotic neutrophils: mediation by a
Vosatka, A., 2001. Induction of 11␤-hydroxysteroid dehydrogenase type novel CD36-independent vitronectin receptor/thrombospondin recognition
1 but not 2 in human aortic smooth muscle cells by inflammatory stimuli. mechanism that is uncoupled from chemokine secretion. J. Immunol. 158,
J. Steroid. Biochem. Mol. Biol. 77, 117–122. 4389–4397.
Charriere, G., Cousin, B., Arnaud, E., Andre, M., Bacou, F., Penicaud, L., Justesen, J., Mosekilde, L., Holmes, M., Stenderup, K., Gasser, J., Mullins,
Casteilla, L., 2003. Preadipocyte conversion to macrophage. Evidence of J.J., Seckl, J.R., Kassem, M., 2004. Mice deficient in 11␤-hydroxysteroid
plasticity. J. Biol. Chem. 278, 9850–9855. dehydrogenase type 1 lack bone marrow adipocytes, but maintain normal
Chen, X.N., Liu, W.M., Ambrosino, C., Ruocco, M.R., Poli, V., Romani, bone formation. Endocrinology 145, 1916–1925.
L., Quinto, I., Barbieri, S., Holmes, K.L., Venuta, S., Scala, G., 1997. Klein, A., Bessler, H., Hoogervorst-Spalter, H., Kaufmann, H., Djaldetti, M.,
Impaired generation of bone marrow B lymphocytes in mice deficient in Joshua, H., 1980. A difference between human B and T lymphocytes
C/EBP-␤. Blood 90, 156–164. regarding their capacity to metabolize cortisol. J. Steroid. Biochem. 13,
Cooper, M.S., Bujalska, I., Rabbitt, E., Walker, E.A., Bland, R., Shep- 517–520.
pard, M.C., Hewison, M., Stewart, P.M., 2001. Modulation of 11␤- Kotelevtsev, Y., Holmes, M.C., Burchell, A., Houston, P.M., Schmoll, D.,
hydroxysteroid dehydrogenase isozymes by proinflammatory cytokines Jamieson, P., Best, R., Brown, R., Edwards, C.R.W., Seckl, J.R., Mullins,
in osteoblasts: an autocrine switch from glucocorticoid inactivation to J.J., 1997. 11␤-hydroxysteroid dehydrogenase type 1 knockout mice show
activation. J. Bone. Miner. Res. 16, 1037–1044. attenuated glucocorticoid inducible responses and resist hyperglycaemia
Dougherty, T.F., Schneebeli, G.L., 1955. The use of steroids as anti- on obesity or stress. Proc. Natl. Acad. Sci. U.S.A. 94, 14924–14929.
inflammatory agents. Ann. N.Y. Acad. Sci. 61, 328–348. Liu, Y.Q., Cousin, J.M., Hughes, J., VanDamme, J., Seckl, J.R., Haslett,
Escher, G., Galli, I., Vishwanath, B.S., Frey, B.M., Frey, F.J., 1997. Tumor C., Dransfield, I., Savill, J., Rossi, A.G., 1999. Glucocorticoids pro-
necrosis factor-␣ and interleukin-1␤ enhance the cortisone/cortisol shuttle. mote nonphlogistic phagocytosis of apoptotic leukocytes. J. Immunol.
J. Exp. Med. 186, 189–198. 162, 3639–3646.
Fadok, V.A., Bratton, D.L., Konowal, A., Freed, P.W., Westcott, J.Y., Henson, Mason, D., 1991. Genetic variation in the stress response: susceptibility
P.M., 1998. Macrophages that have ingested apoptotic cells in vitro inhibit to experimental allergic encephalomyelitis and implications for human
proinflammatory cytokine production through autocrine/paracrine mecha- inflammatory disease. Immunol. Today 12, 57–60.
nisms involving TGF-␣, PGE2, and PAF. J. Clin. Invest. 101, 890–898. McEwen, B.S., Biron, C.A., Brunson, K.W., Bulloch, K., Chambers, W.H.,
Finney, R.S.H., Somers, G.F., 1958. The anti-inflammatory activity of gly- Dhabhar, F.S., Goldfarb, R.H., Kitson, R.P., Miller, A.H., Spencer, R.L.,
cyrretinic acid and derivatives. J. Pharm. Pharmacol. 10, 613–620. Weiss, J.M., 1997. The role of adrenocorticoids as modulators of immune
Freeman, L., Hewison, M., Hughes, S.V., Evans, K.N., Hardie, D., Means, function in health and disease: neural, endocrine and immune interactions.
T.K., Chakraverty, R., 2005. Expression of 11␤-hydroxysteroid dehy- Brain Res. Rev. 23, 79–133.
8 K.E. Chapman et al. / Molecular and Cellular Endocrinology 248 (2006) 3–8

Meagher, L.C., Cousin, J.M., Seckl, J.R., Haslett, C., 1996. Opposing effects Sternberg, E.M., Chrousos, G.P., Wilder, R.L., Gold, P.W., 1992. The stress
of glucocorticoids on the rate of apoptosis in neutrophilic and eosinophilic response and the regulation of inflammatory disease. Ann. Intern. Med.
granulocytes. J. Immunol. 156, 4422–4428. 117, 854–866.
Meagher, L.C., Savill, J.S., Baker, A., Fuller, R.W., Haslett, C., 1992. Phago- Tavor, S., Vuong, P.T., Park, D.J., Gombart, A.F., Cohen, A.H., Koeffler,
cytosis of apoptotic neutrophils does not induce macrophage release of H.P., 2002. Macrophage functional maturation and cytokine production
thromboxane B2. J. Leukoc. Biol. 52, 269–273. are impaired in C/EBP epsilon-deficient mice. Blood 99, 1794–1801.
Morton, N.M., Holmes, M.C., Fiévet, C., Staels, B., Tailleux, A., Mullins, J.J., Taylor, P.R., Carugati, A., Fadok, V.A., Cook, H.T., Andrews, M., Carroll,
Seckl, J.R., 2001. Improved lipid and lipoprotein profile, hepatic insulin M.C., Savill, J.S., Henson, P.M., Botto, M., Walport, M.J., 2000. A hier-
sensitivity, and glucose tolerance in 11␤-hydroxysteroid dehydrogenase archical role for classical pathway complement proteins in the clearance
type 1 null mice. J. Biol. Chem. 276, 41293–41300. of apoptotic cells in vivo. J. Exp. Med. 192, 359–366.
Morton, N.M., Paterson, J.M., Masuzaki, H., Holmes, M.C., Staels, B., Tetsuka, M., Milne, M., Simpson, G.E., Hillier, S.G., 1999. Expression
Fievet, C., Walker, B.R., Flier, J.S., Mullins, J.J., Seckl, J.R., 2004. of 11␤-hydroxysteroid dehydrogenase, glucocorticoid receptor, and min-
Novel adipose tissue-mediated resistance to diet-induced visceral obesity eralocorticoid receptor genes in rat ovary. Biol. Reprod. 60, 330–
in 11␤-hydroxysteroid dehydrogenase type 1-deficient mice. Diabetes 53, 335.
931–938. Tetsuka, M., Thomas, F.J., Thomas, M.J., Anderson, R.A., Mason, J.I., Hillier,
Munck, A., Guyre, P.M., Holbrook, N.J., 1984. Physiological functions of S.G., 1997. Differential expression of messenger ribonucleic acids encod-
glucocorticoids in stress and their relationship to pharmacological actions. ing 11␤-hydroxysteroid dehydrogenase types 1 and 2 in human granulosa
Endocrinol. Rev. 5, 25–44. cells. J. Clin. Endocrinol. Metab. 82, 2006–2009.
Munck, A., Naray-Fejes-Toth, A., 1992. The ups and downs of glucocorti- Thieringer, R., Le Grand, C.B., Carbin, L., Cai, T.Q., Wong, B., Wright, S.D.,
coid physiology. Permissive and suppressive effects revisited. Mol. Cell Hermanowski-Vosatka, A., 2001. 11␤-Hydroxysteroid dehydrogenase type
Endocrinol. 90, C1–C4. 1 is induced in human monocytes upon differentiation to macrophages.
Newman, S.L., Henson, J.E., Henson, P.M., 1982. Phagocytosis of senescent J. Immunol. 167, 30–35.
neutrophils by human monocyte-derived macrophages and rabbit inflam- Tomlinson, J.W., Moore, J., Cooper, M.S., Bujalska, I., Shahmanesh, M.,
matory macrophages. J. Exp. Med. 156, 430–442. Burt, C., Strain, A., Hewison, M., Stewart, P.M., 2001. Regulation of
Poli, V., 1998. The role of C/EBP isoforms in the control of inflammatory expression of 11␤-hydroxysteroid dehydrogenase type 1 in adipose tissue:
and native immunity functions. J. Biol. Chem. 273, 29279–29282. tissue-specific induction by cytokines. Endocrinology 142, 1982–1989.
Pompei, R., Flore, O., Marccialis, M.A., Pani, A., Loddo, B., 1979. Gly- Tomlinson, J.W., Stewart, P.M., 2001. Cortisol metabolism and the role of
cyrrhizic acid inhibits virus growth and inactivates virus particles. Nature 11␤-hydroxysteroid dehydrogenase. Best Pract. Res. Clin. Endocrinol.
281, 689–690. Metab. 15, 61–78.
Rook, G., Baker, R., Walker, B., Honour, J., Jessop, D., Hernandez-Pando, R., Voll, R.E., Herrmann, M., Roth, E.A., Stach, C., Kalden, J.R., Girkontaite, I.,
Arriaga, K., Shaw, R., Zumla, A., Lightman, S., 2000. Local regulation 1997. Immunosuppressive effects of apoptotic cells. Nature 390, 350–351.
of glucocorticoid activity in sites of inflammation. Insights from the study Whyte, M.K., Meagher, L.C., MacDermot, J., Haslett, C., 1993. Impairment
of tuberculosis. Ann. N.Y. Acad. Sci. 917, 913–922. of function in aging neutrophils is associated with apoptosis. J. Immunol.
Sapolsky, R.M., Romero, L.M., Munck, A.U., 2000. How do glucocorticoids 150, 5124–5134.
influence stress responses? Integrating permissive, suppressive, stimula- Williams, L.J.S., Lyons, V., MacLeod, I., Rajan, V., Darlington, G.J., Poli, V.,
tory, and preparative actions. Endocrinol. Rev. 21, 55–89. Seckl, J.R., Chapman, K.E., 2000. C/EBP regulates hepatic transcription
Savill, J., Dransfield, I., Gregory, C., Haslett, C., 2002. A blast from the of 11␤-hydroxysteroid dehydrogenase type 1; a novel mechanism for
past: clearance of apoptotic cells regulates immune responses. Nat. Rev. cross talk between the C/EBP and glucocorticoid signalling pathways. J.
Immunol. 2, 965–975. Biol. Chem. 275, 30232–30239.
Savill, J.S., Wyllie, A.H., Henson, J.E., Walport, M.J., Henson, P.M., Haslett, Yau, J.L., Noble, J., Kenyon, C.J., Hibberd, C., Kotelevtsev, Y., Mullins,
C., 1989. Macrophage phagocytosis of aging neutrophils in inflamma- J.J., Seckl, J.R., 2001. Lack of tissue glucocorticoid reactivation in
tion. Programmed cell death in the neutrophil leads to its recognition by 11␤-hydroxysteroid dehydrogenase type 1 knockout mice ameliorates
macrophages. J. Clin. Invest. 83, 865–875. age-related learning impairments. Proc. Natl. Acad. Sci. U.S.A. 98,
Seckl, J.R., Walker, B.R., 2001. Minireview: 11␤-hydroxysteroid dehy- 4716–4721.
drogenase type 1—a tissue-specific amplifier of glucocorticoid action. Yeager, M.P., Guyre, P.M., Munck, A.U., 2004. Glucocorticoid regulation
Endocrinology 142, 1371–1376. of the inflammatory response to injury. Acta Anaesthesiol. Scand. 48,
Shanks, N., Windle, R.J., Perks, P.A., Harbuz, M.S., Jessop, D.S., Ingram, 799–813.
C.D., Lightman, S.L., 2000. Early-life exposure to endotoxin alters Yong, P.Y., Harlow, C., Thong, K.J., Hillier, S.G., 2002. Regulation of 11␤-
hypothalamic–pituitary–adrenal function and predisposition to inflamma- hydroxysteroid dehydrogenase type 1 gene expression in human ovarian
tion. Proc. Natl. Acad. Sci. U.S.A. 97, 5645–5650. surface epithelial cells by interleukin-1. Hum. Reprod. 17, 2300–2306.
Sieweke, M.H., Graf, T., 1998. A transcription factor party during blood cell Zhang, T.Y., Ding, X., Daynes, R.A., 2005. The expression of 11␤-
differentiation. Curr. Opin. Genet. Dev. 8, 545–551. hydroxysteroid dehydrogenase type I by lymphocytes provides a novel
Sternberg, E.M., 2001. Neuroendocrine regulation of autoimmune/ means for intracrine regulation of glucocorticoid activities. J. Immunol.
inflammatory disease. J. Endocrinol. 169, 429–435. 174, 879–889.

You might also like