You are on page 1of 5

Molecular and Cellular Endocrinology 246 (2006) 142–146

Non-genomic glucocorticoid effects to provide


the basis for new drug developments
In-Ho Song, Frank Buttgereit ∗
Department of Rheumatology and Clinical Immunology, Charité University Hospital, Schumannstrasse 20/21, 10117 Berlin, Germany

Abstract
Glucocorticoids act via genomic and non-genomic actions. The genomic glucocorticoid actions are well known and new details on processes of
transactivation and transrepression have been reported recently. Here we describe the current knowledge on non-genomic glucocorticoid actions
and discuss why these actions are considered to be of therapeutic relevance.
It is assumed that rapid non-genomic glucocorticoid effects are mediated by three different mechanisms: (1) physicochemical interactions with
cellular membranes (non-specific non-genomic effects); (2) membrane-bound glucocorticoid receptor (mGCR)-mediated non-genomic effects;
and (3) cytosolic glucocorticoid receptor (cGCR)-mediated non-genomic effects. With regard to the first mechanism, we discuss here lazaroids and
the novel development of drug targeting with liposomes as the carrier system for glucocorticoids. The clinical use of the latter two mechanisms is
still speculative, but intriguing ideas are being discussed in this regard.
© 2005 Elsevier Ireland Ltd. All rights reserved.

Keywords: Glucocorticoids; Methylprednisolone; Lazaroids

1. Introduction 1.1. Physicochemical interactions with cellular membranes


(non-specific non-genomic effects)
The mechanisms of glucocorticoid actions can be divided into
genomic and non-genomic effects. Genomic effects are known Rapid glucocorticoid actions (which occur within seconds)
to be mediated by transactivation and transrepression processes. are being considered as a result of physicochemical interactions
These mechanisms have been recently reviewed more in detail with cellular membranes. How can we explain these non-specific
(Schacke et al., 2004; Perretti et al., 2003; Song et al., 2005). non-genomic effects? Currently, the following hypothesis is
Here we discuss the therapeutic relevance of rapid non- favoured: Glucocorticoid molecules intercalate at high concen-
genomic glucocorticoid effects which are assumed to be medi- trations in cellular membranes (plasma and mitochondrial mem-
ated by three mechanisms: (1) physicochemical interactions branes) which alter cell functions by influencing cation transport
with cellular membranes (non-specific non-genomic effects) through the plasma membrane and by increasing the proton leak
(Buttgereit and Scheffold, 2002; Buttgereit et al., 2004); of the mitochondria. The impaired cation cycling across and
(2) cytosolic glucocorticoid receptor (cGCR)-mediated non- the compromised ATP production via oxidative phosphoryla-
genomic effects (Croxtall et al., 2000); and (3) membrane-bound tion are considered to result in immunosuppressive effects and
glucocorticoid receptor (mGCR)-mediated non-genomic effects to a reduced activity of inflammatory processes (Buttgereit and
(Bartholome et al., 2004). The therapeutic relevance of non- Scheffold, 2002; Buttgereit et al., 2004). In the following sec-
genomic glucocorticoid actions is an issue of ongoing discus- tion, we would like to explain more in detail the experimental
sion. For example, very recently, beneficial rapid non-genomic results that have led to this theory.
glucocorticoid effects have been described for the first time in The background to explain the rapid non-specific non-
vivo in the treatment of seasonal allergic rhinitis (Tillmann et genomic glucocorticoid effects is provided by considerations of
al., 2004). the cellular energy metabolism. Every organism and every cell
needs metabolic energy whereby ATP is the major source. Also
immune cells produce and consume certain amounts of ATP for
their housekeeping activities and for specific immune functions.
∗ Corresponding author. Tel.: +49 30 450 513125; fax: +49 30 450 513917. ATP-dependent immune functions include cytokinesis, migra-
E-mail address: frank.buttgereit@charite.de (F. Buttgereit). tion, phagocytosis, antigen processing and antigen presentation,

0303-7207/$ – see front matter © 2005 Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.mce.2005.11.012
I.-H. Song, F. Buttgereit / Molecular and Cellular Endocrinology 246 (2006) 142–146 143

signalling and effector functions such as the synthesis of anti- at concentrations that leave quiescent cells unaffec-
bodies, cytotoxicity and regulatory functions (Buttgereit et al., ted.
2000). The major sources for ATP production are glycolysis and 2. Methylprednisolone not only reverses but also prevents the
oxidative phosphorylation. The most important ATP-consuming Con A-effect on respiration in a dose dependent manner.
pathways are identified to be the transport of cations and syn- 3. Con A is known to produce a dramatic increase of cytoplas-
thesis of macromolecules (Buttgereit et al., 2000). In the case mic calcium concentration. This effect is clearly reduced in
of energy deficit, the functions of immune cells are known to the presence of therapeutically relevant drug concentrations
be impaired (Meldrum et al., 1994; Karlsson and Nassberger, or even abolished at suprapharmacological doses.
1992; Sanchez-Alcazar et al., 1995). 4. Methylprednisolone inhibits calcium and sodium cycling
Given this background, we have shown a clear hierarchy across the plasma membrane but has little effect on pro-
of energy-consuming pathways in case of reduced energy sup- tein synthesis. The inhibition of cation cycling in Con
ply: Pathways of macromolecule biosynthesis (protein synthesis A-stimulated thymocytes by the glucocorticoid is caused
and RNA/DNA synthesis) are most sensitive to energy restric- by direct effects and not by a reduction in ATP production
tion whereas cation transport ATPases are much less affected. even though methylprednisolone reduces ATP availability to
The mitochondrial proton leak is least sensitive to enery sup- some extent by inhibiting the reactions of substrate oxidation
ply. These results were obtained by quantitating main ATP- and by increasing mitochondrial proton leak.
consuming pathways under progressively restricted mitochon-
drial ATP production by using myxothiazol, a specific inhibitor The comparison of these methylprednisolone effects with
of the electron transport chain. Obviously, processes not essen- those of myxothiazol we have reported above leads to the follow-
tial for the immediate needs of the cell will be given up before ing hypothetical explanation: Methylprednisolone dissolves in
those that are more critical for ionic integrity (Buttgereit and membranes and affects physicochemical membrane properties
Brand, 1995; Buttgereit et al., 1999). and the activities of membrane-associated proteins (Buttgereit
Why are these experimental results important to under- and Scheffold, 2002; Buttgereit et al., 2004). The resulting inhi-
stand rapid glucocorticoid effects? If immune cells are treated bition of calcium and sodium entry across the plasma membrane
with high, but clinically relevant concentrations of methylpred- would explain (i) the decrease in ATP use (and therefore oxygen
nisolone (the glucocorticoid most commonly used for high- consumption, see Fig. 1) for plasma membrane ion cycling and
dose therapy), then the following effects on bioenergetics (ii) the drop in cytosolic free calcium. A direct effect on the mito-
are observed within seconds (Buttgereit and Scheffold, 2002; chondrial inner membrane would explain the observed increase
Buttgereit et al., 1997, 2004): in proton permeability and the consequent partial uncoupling
of oxidative phosphorylation. This is suggested to result in
1. The glucocorticoid instantly inhibits respiration of Con immunosuppression since immune cell function depends on
A-stimulated thymocytes and human immune cells (Fig. 1) proper functioning of these processes.

Fig. 1. Oxygen consumption of lymphocytes as stimulated by Con A and inhibited by methylprednisolone (original trace measured using a Clark electrode). This
figure shows an original trace of oxygen consumption in thymocytes measured amperometrically with a Clark electrode. The first part of the curve reflects the basal
rate of oxygen consumption, calculated by dividing the change in oxygen saturation (delta O2) by the time (delta t). Addition of concanavalin A (Con A) leads to
a significant reater rate of oxygen consumption within seconds as reflected by the changed slope of the line. Additon of methylprednisolone leads to a reduction of
oxygen consumption. This experiment was carried out by Robert Tripmacher from our laboratory.
144 I.-H. Song, F. Buttgereit / Molecular and Cellular Endocrinology 246 (2006) 142–146

Substances which act similarly to high-dose glucocorticoids effects” are suggested to be mediated by membrane-bound
are lazaroids (21-aminosteroids such as tirilazad). Lazaroids glucocorticoid receptors (mGCR). The existence of such
intercalate into biological membranes without binding to glu- membrane-bound glucocortioid receptors had already been
cocorticoid receptors. In this way, they cause a stabilisation shown in amphibian neuronal membranes (Orchinik et al., 1991)
of membranes and inhibition of destructive lipid peroxidation and in lymphoma cells (Gametchu et al., 1993; Chen et al., 1999;
which results in the prevention of oxidative cell damage. Tiri- Sackey et al., 1997). Recently, we were able to identify for the
lazad is used in neurotraumatology (Braughler and Pregenzer, first time mGCR in normal human peripheral blood mononuclear
1989; Dissemond et al., 2003; Bath et al., 2001). To our knowl- cells (Bartholome et al., 2004; Buttgereit et al., 2004; Song et al.,
edge, however, these drugs have not been used to investigate 2005). This observation was made using the novel technique of
their immunosuppressive effects in clinical trials. high-sensitivity immunofluorescent staining. The method uses
Coming back to glucocorticoids, a very interesting approach antibody-conjugated magnetofluorescent liposomes, which can
in the context of rapid non-specific non-genomic glucocorticoid increase fluorescence signal intensity up to 1000-fold compared
effects is drug targeting with liposomes as the carrier system with conventional methods and allows the detection of 50–100
for glucocorticoids. Newly developed glucocorticoid-containing target molecule per cell. Up to 9.2 and 12.3% of the monocytes
liposomes accumulate selectively at the site of inflammation and B lymphocytes were positive for mGCR, respectively. T
where there is an increased permeability of the local vascular lymphocytes never showed any significant mGCR expression
endothelium. By reaching very high concentrations (>10−5 M) (Bartholome et al., 2004). Further experiments demonstrated
at the site of inflammation for several hours, 100% utilisation of immunostimulation with lipopolysaccharide to increase signif-
the genomic actions and, in addition, significant therapeutically icantly the percentage of mGCR-positive monocytes. In the
beneficial non-genomic actions are achieved (Song et al., 2005). presence of brefeldin A, immunostimulation with lipopolysac-
Although the systemic glucocorticoid concentrations (plasma charide did not increase the number of mGCR-positive mono-
levels) remain relatively high, they give rise to fewer adverse cytes demonstrating the ability of brefeldin A to abrogate the
reactions because the steroid is encapsulated in the liposome. induced up-regulation of mGCR by inhibiting the secretory path-
Schmidt et al. (2003) recently used prednisolone liposomes to way. We concluded that immunostimulation induces cellular
demonstrate the success of this innovative approach in the treat- events whereby mGCR are actively upregulated and transported
ment of experimental autoimmune encephalomyelitis in Lewis through the cell.
rats (EAE). On the basis of this data, we hypothesized that there is a
Metselaar et al. (2003, 2004) have recently reported a com- clinically relevant correlation between the number of mGCR-
plete remission of experimental arthritis by joint targeting of positive cells and disease-related activity of the immune system.
glucocorticoids with long-circulating liposomes (Song et al., We indeed found a strong positive correlation between the fre-
2005). A single intravenous administration of 10 mg/kg lipo- quency of mGCR-positive monocytes and various parameters
somal prednisolone phosphate in rat experimental arthritis and of disease activity in patients suffering from rheumatoid arthri-
in murine collagen type II-induced arthritis showed a profound tis (Bartholome et al., 2004). One way to interpret these data is
antiinflammatory effect which lasted for more than 1 week. In that mGCR may play a role in the aetiopathogenesis of disease.
contrast, 10 mg/kg of unencapsulated liposomal prednisolone However, we suggest being more likely that immunostimula-
phosphate was less effective even after repeated daily injections. tion or high disease activity stimulate mGCR expression in
Moreover, these authors also found the process of cartilage ero- immune cells such as monocytes which in turn leads to a signif-
sion to be profoundly attenuated under this treatment. From the icantly higher percentage of cells undergoing mGCR-mediated
mechanistic point of view, the observation is interesting that the glucocorticoid-induced apoptosis (Sackey et al., 1997). This
liposomes accumulate in the inflamed joint, i.e. in the proxim- putative process would diminish the activity of the immune
ity of blood vessels mainly in the synovial lining. This strongly system and could be therefore considered as a mechanism of
suggested that liposomes selectively localise in cells with phago- negative feed-back regulation. Further experiments will have
cytotic capacity. to be made in order to prove this hypothesis. A new approach
These initial observations suggest targeted delivery using of optimising glucocorticoid therapy could be to develop drugs
long-circulating liposomal glucocorticoids to be an effective and selectively binding to the mGCR, but this is clearly specula-
novel therapeutic option in arthritis (Song et al., 2005). Hope- tive. First the function of mGCR needs to be investigated in
fully clinical trials in humans will yield similar intriguing results detail. Though there is no evidence for specific signalling path-
as in animal models. Future work will also have to optimise both ways associated with mGCR, this mechanism might explain the
the lipid shells and the incorporated glucocorticoids. rapid dexamethasone-induced adenylate cyclase/protein kinase
A-dependent inhibition of chloride ion secretion in bronchial
1.2. Membrane-bound glucocorticoid receptors as epithelium (Goulding, 2004; Urbach et al., 2002).
potential therapeutic targets
1.3. Rapid interaction with signalling processes via
Apart from the physicochemical interactions with cellu- cytosolic glucocorticoid receptors
lar membranes (non-specific non-genomic effects) mentioned
above, other mechanisms of rapid glucocorticoid actions are Another hypothesis to explain rapid glucocorticoid effects is
being discussed. So called “mGCR-mediated non-genomic that glucocorticoids bind to its cytosolic glucocorticoid recep-
I.-H. Song, F. Buttgereit / Molecular and Cellular Endocrinology 246 (2006) 142–146 145

tor where they not only cause classical genomic, but also rapid FlI1 fusion protein (Ewing sarcoma family of tumors), transloca-
non-genomic effects resulting in interactions with signalling tion fusion protein ETV6-NTRK3 (congenital fibrosarcoma or
processes. The underlying theory is the following: Arachi- cellular mesoblastic nephroma) and BCR-ABL kinase inhibitor
donic acid (AA) is an essential mediator which is important imatinib mesylate (chronic myeloid leukaemia). This is only a
for cell growth and several metabolic reactions (e.g. production selection to demonstrate that advances in molecular biology have
of inflammatory cytokines in the setting of the inflammatory already led to the identification of quite a lot new protein targets
cascade). AA release from cell membrane phospholipids is con- along with an increased understanding of the biologic role of
trolled by mediators which among others involve growth factors these proteins (Uren and Toretsky, 2005). This research is ulti-
(like insulin or PDGF), adaptor proteins such as SOS, Grb2, mately designed to improve therapeutic option as, e.g. demon-
the GTPase p21ras, erk2/ MAPK (mitogen activated protein strated with tyrosine kinase inhibitors such as gefitinib which is
kinases), cPLA2 (cytosolic phospholipaseA2) and especially currently tested in phase II trials in breast cancer. Monoclonal
lipocortin 1 (LC1) (Ahn et al., 1991; De Vries-Smits et al., 1992; antibodies targeting epidermal growth factor receptor (EGRF)
Lin et al., 1993; Croxtall et al., 1995, 1996, 1998). represent another approach to interfere with EGRF-driven signal
With regard to cGCR-mediated glucocorticoid effects, transduction. For example, the chimerized monoclonal anti-
Croxtall et al. (2000). have recently reported that epidermal body cetuximab is designed to treat advanced colorectal cancer
growth factor (EGF) stimulated cPLA2 (cytosolic PLA2) acti- (Caponigro et al., 2005). We have discussed these examples to
vation with subsequent arachidonic acid release can be inhibited illustrate that the cGCR-mediated non-genomic glucocorticoid
by dexamethasone. This effect is considered to be a glucocor- effects could provide the basis for targeted therapies in the future.
ticoid receptor-dependent (RU486-sensitive), but transcription-
independent (actinomycin-insensitive) mechanism. 1.4. Concluding remarks
What could be the explanation for this observed effect? To
answer this question, it should be noted that the unactivated Glucocorticoids are clinically very important drugs; how-
(unligated) cGCR is retained in the cytoplasm as a multi-protein ever, their use is limited by side effects. This drives interesting
complex consisting of heat shock proteins and several kinases of research in order to understand better the underlying mecha-
the MAPK signalling system (chaperones and co-chaperones), nisms of action. The non-genomic glucocorticoid effects we
including Src. Following glucocorticoid binding, the cGCR is describe here are currently in the focus of research. The ulti-
released from this complex, thus revealing domains of the recep- mate aim is to convert results of basic research in this regard
tor that are able to bind specifically to glucocorticoid respon- into novel therapeutic options to improve the benefit-risk-ratio
sive DNA elements. This mechanism is known to account for of glucocorticoid therapy.
genomic glucocorticoid effects. However, the role of the remain-
ing signalling components of the multi-protein complex is less References
clearly defined. In their study, Croxtall et al. (2000) showed
that there is a rapid release of not only glucocorticoid receptor Ahn, N.G., Seger, R., Bratlien, R.L., Diltz, C.D., Tonks, N.K., Krebs, E.G.,
protein, but also of Src from the multi-protein complex which 1991. Multiple components in an epidermal growth factor-stimulated pro-
tein kinase cascade. J. Biol. Chem. 266, 4220–4227.
follows the binding of dexamethasone to the cGCR. However, Bartholome, B., Spies, C.M., Gaber, T., Schuchmann, S., Berki, T., Kunkel,
the release of glucocorticoid receptor and Src appear to be dis- D., Bienert, M., Radbruch, A., Burmester, G.R., Lauster, R., Scheffold,
tinct events, since the nuclear translocation of glucocorticoid A., Buttgereit, F., 2004. Membrane glucocorticoid receptors (mGCR)
receptor is not required for subsequent activation of lipocortin are expressed in normal human peripheral blood mononuclear cells and
1 and rapid inhibition of arachidonic acid release. Rather, it up-regulated after in nitro stimulation and in patients with rheumatoid
arthritis. FASEB J. 18, 70–80.
appeared that it is the rapid effect of Scrd that fulfils this role. Bath, P.M., Iddenden, R., Bath, F.J., Orgogozo, J.M., 2001. Tirilazad for
In summary, it is suspected that the binding of glucocorti- acute ischaemic stroke. Cochrane Database Syst. Rev. 4:CD002087.
coids to the cGCR not only induces classical genomic effects, Braughler, J.M., Pregenzer, J.F., 1989. The 21-aminosteroid inhibitors of lipid
but also rapidly controls intracellular signalling through other peroxidation: reactions with lipid peroxyl and phenoxy radicals. Free
components of the multi-protein complex. It may be possible Radic. Biol. Med. 7, 125–130.
Buttgereit, F., Burmester, G.R., Brand, M.D., 2000. Bioenergetics of immune
in the future to make therapeutical use of these glucocorticoid functions: fundamental and therapeutic aspects. Immunol. Today 21,
effects. The clinical background for this assumption is that glu- 192–199.
cocorticoids have been shown recently to activate endothelial Buttgereit, F., Scheffold, A., 2002. Rapid glucocorticoid effects on immune
nitric oxide synthase in a non-genomic manner and mediated by cells. Steroids 67, 529–534.
PI3K and Akt phosphorylation. This ultimately leads to vasore- Buttgereit, F., Straub, H.S., Wehling, M., Burmester, G.R., 2004. Gluco-
corticoids in the treatment of rheumatic diseases. Arthritis Rheum. 50,
laxation, which might explain some of the cardioprotective 3408–3417.
effects of glucocorticoids (Lösel and Wehling, 2003; Hafezi- Buttgereit, F., Brand, D., 1995. A hierarchy of ATP-consuming processes in
Moghadam et al., 2002). There are other examples in the field mammalian cells. Biochem. J. 312, 163–167.
of oncology where blocking of small molecules represent very Buttgereit, F., Krauss, S., Brand, M.D., 1997. Methylprednisolone inhibits
promising therapeutic approaches. In this regard, we would like uptake of Ca2+ and Na+ ions into concanavalin A-stimulated thymocytes.
Biochem. J. 326, 329–332.
to mention the inhibition of IGFBP2 (insulin-like growth factor Buttgereit, F., Brand, M.D., Burmester, G.R., 1999. Equivalent doses and
binding protein) which enhances the invasion capacity of ovarian relative drug potencies for non-genomic glucocorticoid effects: a novel
cancer cells (Lee et al., 2005). Other examples include the EWS- glucocorticoid hierarchy. Biochem. Pharmacol. 58, 363–368.
146 I.-H. Song, F. Buttgereit / Molecular and Cellular Endocrinology 246 (2006) 142–146

Caponigro, F., Basile, M., Rosa, V.D., Normanno, N., 2005. New drugs in Meldrum, D.R., Ayala, A., Chaudry, I.H., 1994. Energetics of lympho-
cancer therapy, national tumor institute, Naples 17–18 June 2004. Anti- cyte “burnout” in late sepsis: adjuvant tratment with ATP-MgCl2
cancer Drugs 16, 211–221. improves energetics and decreases lethality. J. Surg. Res. 56, 537–
Chen, F., Watson, C.S., Gametchu, B., 1999. Association of the glucocorticoid 542.
receptor alternatively-spliced transcript 1A with the presence of the high Metselaar, J.M., Wauben, M.H., Wagenaar-Hilbers, J.P., Boermann, O.C.,
molecular weight membrane glucocorticoid receptor in mouse lymphoma Storm, G., 2003. Complete remission of experimental arthritis by joint
cells. J. Cell Biochem. 74, 430–446. targeting of glucocorticoids with long-circulating liposomes. Arthritis
Croxtall, J.D., Choudhury, Q., Flower, R.J., 2000. Glucocorticoids act within Rheum. 48, 1059–1066.
minutes to inhibit recruitment of signalling actors to activated EGF recep- Metselaar, J.M., van den Berg, W.B., Holthuysen, A.E., Wauben, M.H.,
tors through a receptor-dependent, transcription-independent mechanism. Storm, G., van Lent, P.L., 2004. Liposomal targeting of glucocorticoids
Br. J. Pharmacol. 130, 289–298. to synovial lining cells strongly increases therapeutic benefit in collagen
Croxtall, J.D., Choudhury, Q., Tokumoto, H., Flower, R.J., 1995. Lipocortin- type II arthritis. Ann. Rheum. Dis. 63, 348–353.
1 and the control of arachidonic acid release in cell signalling. Biochem. Orchinik, M., Murray, T.F., Moore, F.L., 1991. A corticosteroid receptor in
Pharmacol. 50, 465–474. neuronal membranes. Science 252, 1848–1851.
Croxtall, J.D., Choudhury, Q., Newman, S., Flower, R.J., 1996. Lipocortin 1 Perretti, M., Paul-Clark, M.J., Mancini, L., Flower, R.J., 2003. Generation of
and the control of cPLA2 activity in A549 cells. Biochem. Pharmacol. innovative anti-inflammatory and anti-arthritic glucocorticoid derivatives
52, 351–356. that release NO: the nitro-steroids. Digestive Liver Dis. 35 (Suppl 2),
Croxtall, J.D., Choudhury, Q., Flower, R.J., 1998. Inhibitory effect of peptides 41–48.
derived from the N-terminus of lipocortin 1 on arachidonic acid release Sackey, F.N., Watson, C.S., Gametchu, B., 1997. Cell cycle regulation of
and proliferation in the A549 cell line: identification of E-Q-E-Y-V as a membrane glucocorticoid receptor in CCRF-CEM human ALL cells: cor-
crucial element. Br. J. Pharmacol. 123, 975–983. relation to apoptosis. Am. J. Physiol. 273, E571–E583.
De Vries-Smits, A.M., Burgering, B.M., Leevers, S.J., Marshall, C.J., Bos, Sanchez-Alcazar, J.A., Hernandez, I., De la Torre, M.P., Garcia, I., Santiago,
J.L., 1992. Involvement of p21ras in activation of extracellular signal- E., Munoz-Yague, M.T., Solis-Herruzo, J.A., 1995. Down-regulation of
regulated kinase 2. Nature 357, 602–604. tumor necrosis factor receptors by blockade of mitochondrial respiration.
Dissemond, J., Schneider, L.A., Wlaschek, M., Brauns, T.C., Goos, M., J. Biol. Chem. 270, 23944–23950.
Scharfetter-Kochanek, K., 2003. The lazaroid tirilazad is a new inhibitor Schacke, H., Schottelius, A., Docke, W.D., Strehlke, P., Jaroch, S., Schmees,
of direct and indirect UVA-induced lipid peroxidation in human dermal N., Rehwinkel, H., Hennekes, H., Assadullah, K., 2004. Dissociation
fibroblasts. Arch. Dermatol. Res. 295, 287–292. of transactivation from transrepression activity by a selective glucocorti-
Gametchu, B., Watson, C.S., Wu, S., 1993. Use of receptor antibodies coid receptor agonist leads to separation of therapeutic effects from side
to demonstrate membrane glucocorticoid receptor in cells from human effects. Proc. Natl. Acad. Sci. U.S.A. 101, 227–232.
leukaemic patients. FASEB J. 7, 1283–1292. Schmidt, J., Metselaar, J.M., Wauben, M.H., Toyka, K.V., Storm, G., Gold, R.,
Goulding, N.J., 2004. The molecular complexity of glucocorticoid actions in 2003. Drug targeting by long circulating liposomal glucocorticosteroids
inflammation- a four-ring circus. Curr. Opin. Pharmacol. 4, 629–636. increases therapeutic efficacy in a model of multiple sclerosis. Brain 126,
Hafezi-Moghadam, A., et al., 2002. Acute cardiovascular protective effects of 1895–1904.
cortcosteroids are mediated by non-transscriptional activation of endothe- Song, I.H., Gold, R., Straub, R.H., Burmester, G.R., Buttgereit, F., 2005.
lial nitric oxide synthase. Nat. Med. 8, 473–479. New glucocorticoids on the horizon: repress, dont activate! J. Rheumatol.
Karlsson, H., Nassberger, L., 1992. In vitro metabolic inhibition of the 32, 1199–1207.
human lymphocyte: influence on the expression of interleukin-2 receptors. Tillmann, H.C., Stuck, B.A., Feuring, M., Rossol-Haseroth, K., Tran, B.M.,
Immunol. Cell Biol. 70, 309–313. Lösel, R., Schmidt, B.M., Hormann, K., Wehling, M., Schultz, A.,
Lee, E.J., Mircean, C., Shmulevich, I., Wang, H., Liu, J., Niemisto, A., 2004. Delayed genomic and acute nongenomic action of glucocorti-
Kavanagh, J.J., Lee, J.H., Zhang, W., 2005. Insulin-like growth factor costeroids in seasonal allergic rhinitis. Eur. J. Clin. Invest. 34, 67–
binding protein 2 promotes ovarian cancer cell invasion. Mol. Cancer 7 73.
(Epub ahead of print). Urbach, V., Walsh, D.E., Mainprice, B., Bousquet, J., Harvey, B.J., 2002.
Lin, L.L., Wartmann, M., Lin, A.Y., Knopf, J.L., Seth, A., Davis, R.J., 1993. Rapid non-genomic inhibition of ATP-induced Cl− secretion by dexam-
cPLA2 is phosphorylated and activated by MAP kinase. Cell 72, 269–278. ethasone in human bronchial epithelium. J. Physiol. 545, 869–878.
Lösel, F., Wehling, M., 2003. Nongenomic actions of steroid hormones. Nat. Uren, A., Toretsky, J.A., 2005. Pediatric malignancies provide unique cancer
Rev. Mol. Cell Biol. 4, 46–56. therapy targets. Curr. Opin. Pediatr. 1, 14–19.

You might also like