53
Crosstalk in Inflammation: The Interplay of Glucocorticoid Receptor-Based Mechanisms and Kinases and Phosphatases Ilse M. E. Beck, Wim Vanden Berghe, Linda Vermeulen, Keith R. Yamamoto, Guy Haegeman,* and Karolien De Bosscher* Laboratory of Eukaryotic Gene Expression and Signal Transduction (I.M.E.B., W.V.B., L.V., G.H., K.D.B.), Department of Physiology, Ghent University, B-9000 Ghent, Belgium; and Department of Cellular and Molecular Pharmacology (K.R.Y.), University of California, San Francisco, California 94143-2280 Glucocorticoids (GCs) are steroidal ligands for the GC receptor (GR), which can function as a ligand-activated transcription factor. These steroidal ligands and derivatives thereof are the first line of treatment in a vast array of inflammatory diseases. However, due to the general surge of side effects associated with long-term use of GCs and the potential problem of GC resistance in some patients, the scientific world continues to search for a better understanding of the GC-mediated antiinflammatory mechanisms. The reversible phosphomodification of various mediators in the inflammatory process plays a key role in modulating and fine-tuning the sensitivity, longevity, and intensity of the inflammatory response. As such, the antiinflammatory GCs can modulate the activity and/or expression of various kinases and phosphatases, thus affecting the signaling efficacy toward the propagation of proinflammatory gene expression and proinflammatory gene mRNA stability. Conversely, phosphorylation of GR can affect GR ligand- and DNA-binding affinity, mobility, and cofactor recruitment, culminating in altered transactiva- tion and transrepression capabilities of GR, and consequently leading to a modified antiinflammatory potential. Recently, new roles for kinases and phosphatases have been described in GR-based antiinflammatory mecha- nisms. Moreover, kinase inhibitors have become increasingly important as antiinflammatory tools, not only for research but also for therapeutic purposes. In light of these developments, we aim to illuminate the integrated interplay between GR signaling and its correlating kinases and phosphatases in the context of the clinically important combat of inflammation, giving attention to implications on GC-mediated side effects and therapy resistance. (Endocrine Reviews 30: 830 – 882, 2009) I. Introduction A. Inflammation at a molecular level B. Glucocorticoid receptor-mediated signaling II. Phosphoregulation of the Glucocorticoid Receptor A. GR phosphorylation B. GR dephosphorylation C. Other posttranslational modifications of GR III. Kinases Targeted by Glucocorticoid Receptor-Medi- ated Signaling A. Mitogen-activated protein kinases (MAPKs) B. MAPK-activated protein kinases (MKs) C. Cyclin-dependent kinases (Cdks) D. IB kinase (IKK) E. TANK-binding kinase 1 (TBK1) F. Other kinases IV. Phosphatases Targeted by Glucocorticoid Receptor- Mediated Signaling A. Dual specificity phosphatases (DUSPs) B. Other protein Y phosphatases C. Other phosphatases V. Kinase/Phosphatase Regulation in Glucocorticoid- Mediated Side Effects A. Skeleton and muscle effects B. Hyperglycemia and diabetes C. Other side effects VI. Kinase/Phosphatase Regulation in Glucocorticoid Resistance VII. Future Perspectives in the Combat of Inflammation A. New glucocorticoid receptor ligands B. Combination therapies C. MicroRNA-specific modulation of GR D. Epigenetic approaches VIII. Conclusions ISSN Print 0021-972X ISSN Online 1945-7197 Printed in U.S.A. Copyright © 2009 by The Endocrine Society doi: 10.1210/er.2009-0013 Received March 31, 2009. Accepted August 18, 2009. First Published Online November 4, 2009 * G.H. and K.D.B. share senior authorship. REVIEW 830 edrv.endojournals.org Endocrine Reviews, December 2009, 30(7):830 – 882 Downloaded from https://academic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

edrv0830.pdf - Oxford Academic

Embed Size (px)

Citation preview

Crosstalk in Inflammation: The Interplay ofGlucocorticoid Receptor-Based Mechanismsand Kinases and Phosphatases

Ilse M. E. Beck, Wim Vanden Berghe, Linda Vermeulen, Keith R. Yamamoto,Guy Haegeman,* and Karolien De Bosscher*

Laboratory of Eukaryotic Gene Expression and Signal Transduction (I.M.E.B., W.V.B., L.V., G.H., K.D.B.), Departmentof Physiology, Ghent University, B-9000 Ghent, Belgium; and Department of Cellular and Molecular Pharmacology(K.R.Y.), University of California, San Francisco, California 94143-2280

Glucocorticoids (GCs) are steroidal ligands for the GC receptor (GR), which can function as a ligand-activatedtranscription factor. These steroidal ligands and derivatives thereof are the first line of treatment in a vast arrayof inflammatory diseases. However, due to the general surge of side effects associated with long-term use ofGCs and the potential problem of GC resistance in some patients, the scientific world continues to search for abetter understanding of the GC-mediated antiinflammatory mechanisms.

The reversible phosphomodification of various mediators in the inflammatory process plays a keyrole in modulating and fine-tuning the sensitivity, longevity, and intensity of the inflammatory response.As such, the antiinflammatory GCs can modulate the activity and/or expression of various kinases andphosphatases, thus affecting the signaling efficacy toward the propagation of proinflammatory geneexpression and proinflammatory gene mRNA stability. Conversely, phosphorylation of GR can affect GRligand- and DNA-binding affinity, mobility, and cofactor recruitment, culminating in altered transactiva-tion and transrepression capabilities of GR, and consequently leading to a modified antiinflammatorypotential.

Recently, new roles for kinases and phosphatases have been described in GR-based antiinflammatory mecha-nisms. Moreover, kinase inhibitors have become increasingly important as antiinflammatory tools, not only forresearch but also for therapeutic purposes. In light of these developments, we aim to illuminate the integratedinterplay between GR signaling and its correlating kinases and phosphatases in the context of the clinicallyimportant combat of inflammation, giving attention to implications on GC-mediated side effects and therapyresistance. (Endocrine Reviews 30: 830–882, 2009)

I. IntroductionA. Inflammation at a molecular levelB. Glucocorticoid receptor-mediated signaling

II. Phosphoregulation of the Glucocorticoid ReceptorA. GR phosphorylationB. GR dephosphorylationC. Other posttranslational modifications of GR

III. Kinases Targeted by Glucocorticoid Receptor-Medi-ated SignalingA. Mitogen-activated protein kinases (MAPKs)B. MAPK-activated protein kinases (MKs)C. Cyclin-dependent kinases (Cdks)D. I�B kinase � (IKK�)

E. TANK-binding kinase 1 (TBK1)F. Other kinases

IV. Phosphatases Targeted by Glucocorticoid Receptor-Mediated SignalingA. Dual specificity phosphatases (DUSPs)B. Other protein Y phosphatasesC. Other phosphatases

V. Kinase/Phosphatase Regulation in Glucocorticoid-Mediated Side EffectsA. Skeleton and muscle effectsB. Hyperglycemia and diabetesC. Other side effects

VI. Kinase/Phosphatase Regulation in GlucocorticoidResistance

VII. Future Perspectives in the Combat of InflammationA. New glucocorticoid receptor ligandsB. Combination therapiesC. MicroRNA-specific modulation of GRD. Epigenetic approaches

VIII. Conclusions

ISSN Print 0021-972X ISSN Online 1945-7197Printed in U.S.A.Copyright © 2009 by The Endocrine Societydoi: 10.1210/er.2009-0013 Received March 31, 2009. Accepted August 18, 2009.First Published Online November 4, 2009* G.H. and K.D.B. share senior authorship.

R E V I E W

830 edrv.endojournals.org Endocrine Reviews, December 2009, 30(7):830–882

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

I. Introduction

According to the World Health Organization (WHO;2007 report), inflammation and inflammation-me-

diated illnesses are the biggest challenge in current medi-cine because 300 million people worldwide are estimatedto suffer from asthma and 210 million people live withmild or severe chronic obstructive pulmonary disease(COPD), the latter leading up to 5% of global deaths.Furthermore, many people live uncomfortably withchronic inflammatory disorders, such as rheumatoid ar-thritis and inflammatory bowel disease. Moreover, theonset of cancer and cardiovascular diseases has also beenlinked to inflammation, claiming 13 and 30% of globaldeaths, respectively (WHO). As the costs of treating thesedisorders mount up and life comfort and expectancy arethreatened, understanding and resolving inflammation iscurrently one of the main targets in science.

Today, glucocorticoid (GC)-based therapy is still themost commonly used treatment to combat chronic andacute inflammation. Since the discovery of the antiinflam-matory properties of human cortisone in rheumatoid ar-thritis (1) and the cloning of the GC receptor (GR) (2),tremendousprogresshasbeenmade inunderstandinghowGCs inhibit inflammation: the molecular antiinflamma-tory mechanism of GCs consists of GR-mediated transac-tivation and transrepression mechanisms, the latter ofwhich prominently features inhibition of nuclear fac-tor-�B (NF-�B) activation and activity.

GCs have multiple physiological actions. As a conse-quence, a chronic exposure to pharmacological hormonedoses becomes a problem in therapeutic settings, causingundesirable, yet on-target and thus GR-mediated, effects.The challenge is therefore not to develop more specificligands for GR, but to change the spectrum of GR-medi-ated events and try to skew it more toward antiinflamma-

tory pathways. This implies that selective (in terms of func-tionality) GR modulators could eliminate these adverseeffects. Besides the undesirable effects, GC resistance, inwhich the patients do not respond to GCs, may also occur.Therefore, the mainstay of antiinflammatory research ef-forts is focused on further characterizing the antiinflam-matory mechanisms of GCs in detail and developing newtherapeutic strategies to fight inflammation with a betterbenefit-to-risk-ratio.

Protein kinases (afterward referred to as kinases) areenzymes that can rapidly and reversibly phosphorylateS, T, or Y residues of cellular proteins and as such affecttheir structure, function, location or metabolism. Inturn, phosphatases function to revert the action of thesekinases by dephosphorylating specific target residues(3). The GR itself is on the one hand subject to intense phos-phoregulation, thus impacting its role in various antiinflam-matoryprocesses,andontheotherhandthisGRdeploysandaffects kinases and phosphatases as tools to implement itscellular antiinflammatory effects. In this review, we willfocus on the above events, providing a contemporary viewon the overall phosphomodulatory effects of and by theGR in the framework of inflammation. Additionally, therole of various phosphorylation events in the describedGC-mediated side effects and the reported phenomenonof GC resistance will be discussed. Ultimately, we willdiscuss future therapeutic implications of phosphoregu-lation in the context of GR-based antiinflammatorystrategies.

A. Inflammation at a molecular levelInflammation is an initially advantageous response to

intracellular damage or an extracellular challenger, pro-voking the activation of various proinflammatory medi-ators with the purpose to remove the damaging agent andto restore tissue structure and function. Physiologically,

Abbreviations: AF, Activation function; AP-1, activator protein-1; ARE, adenylate uridylate-rich element; ATF, activating transcription factor; C, carboxy; CBP, CREB-binding protein;cdc37, cell division cycle 37 protein; Cdk, cyclin-dependent kinase; C/EBP, CCAATenhancer-binding protein; COPD, chronic obstructive pulmonary disease; COX-2, cycloox-ygenase-2; CpdA, compound A; CREB, cAMP-responsive element-binding protein; CRM1,chromosome region maintenance (synonym, exportin1); CTD, C-terminal domain; DBD,DNA-binding domain; Dexras1, dexamethasone-induced Ras1; DUSP, dual specificityphosphatase; 4E-BP1, eukaryotic translation initiation factor 4E-binding protein 1; eIF-4E/F,eukaryotic translation initiation factor 4E/F; ELKS, protein rich in amino acids E, L, K and S;ENaC, epithelial Na� channel; FKBP, FK506-binding protein; GC, glucocorticoid; GILZ,GC-induced leucine zipper; GM-CSF, granulocyte monocyte-colony stimulating factor;G6Pase, glucose-6-phosphatase; GR, GC receptor; GRE, GC response element; GRIP1,GR-interacting protein 1; GSK, glycogen synthase kinase; H3, histone H3; HAT, histoneacetyl transferase; HDAC, histone deacetylase; hGR, human GR; HPA, hypothalamic-pi-tuitary-adrenocortical; Hsp, heat shock protein; ICAM, intercellular adhesion molecule; IE,immediate-early; Ifit1, IFN-induced with tetratricopeptide repeats 1; IFN, interferon; I�B,inhibitor of NF-�B; IKK, I�B kinase; IL1RI, IL-1 receptor I; IRAK, IL-1 receptor-associatedkinase; IRF, interferon regulatory factor; ISRE, IFN-stimulated response element; JAK, Januskinase; JDP, Jun dimerization protein; JNK, c-Jun N-terminal kinase; KO, knockout; LABA,long-acting �2 agonist; LBD, ligand-binding domain; Lck, lymphocyte kinase; LPS, lipo-polysaccharide; Mal, MyD88-adapter like protein; MED14, mediator complex subunit 14(synonym, DRIP150); MEKK, MKK kinase (synonyms, MKKK, MAPKKK, MAP3K); mGR,mouse/murine GR; miRNA, microRNA; MK, MAPK-activated kinase; MKK, MAPK kinase

(synonyms, MEK, MAPKK, MAP2K); MMP, matrix metalloproteinase; MMTV, mouse mam-mary tumor virus; MNK, MAPK-interacting kinase; MR, mineralocorticoid receptor; MSK,mitogen- and stress-activated protein kinase; MyD88, myeloid differentiation primary re-sponse gene 88; NCoR, nuclear corepressor; NF-�B, nuclear factor-�B; nGRE, negativeGRE; NHE, Na�/H� exchanger; NIK, NF-�B-inducing kinase; NLS, nuclear localizationsignal; p/CAF, p300/CBP-associated factor; PEPCK, phosphoenylpyruvate carboxykinase;PI3K, phosphatidylinositol-3-kinase; PKA, protein kinase A; PKB, protein kinase B (syn-onym, Akt); PKC, protein kinase C; PP, protein phosphatase; P-TEFb, positive transcriptionelongation factor b; PTP, protein tyrosine phosphatase; RANKL, receptor activator of NF-�Bligand; RANTES, regulated upon activation, normal T-cell expressed and secreted; rGR, ratGR; RIP, receptor-interacting protein; RNA pol II, RNA polymerase II; ROCK, Rho-dependentprotein kinase; RSK, ribosomal S6 kinase; SEGRA, selective GR agonist; SEGRM, selectiveGR modulator; SEK1, SAPK/ERK kinase 1; SGK, serum and GC-inducible kinase; S6K,70-kDa ribosomal protein S6 kinase; SLAP, Src-like adaptor protein; SOCS, suppressor ofcytokine signaling; Sp1, specificity protein 1; SP-A, surfactant protein-A; SRC, steroidreceptor coactivator; STAT, signal transducer and activator of transcription; SUMO, smallubiquitin-related modifier; TAD, transactivation domain; TAK1, TGF-activated kinase 1;TANK, TRAF family member-associated NF-�B activator; TBK1, TANK-binding kinase 1; TLR,Toll-like receptor; TNFR, TNF-� receptor; TPA, 12-O-tetradecanoylphorbol-13-acetate;TRADD, TNFR-associated death domain; TRAF, TNFR-associated factor; Tram, translocatingchain-associating membrane protein; TRE, TPA-response element; Trif, TIR domain-con-taining adapter-inducing IFN�; Trip6, thyroid receptor-interacting protein 6; TTP,tristetraprolin.

Endocrine Reviews, December 2009, 30(7):830–882 edrv.endojournals.org 831

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

inflammation is locally marked by swelling, redness, pain,heat, and loss of function. Although inflammation can bea beneficial reaction, return to homeostasis is of the utmostimportance to avoid the onset of an unfavorable chronicinflammation (4). The inflammatory reaction can be pro-voked by physical injury, tissue damage, or the invasion ofalien pathogens. Alternatively, inflammation can also beprovoked by an unwanted immune reaction of the body toits own proteins, i.e., an autoimmune reaction.

Biologically, inflammation progresses through differ-ent stages. First, local hyperemia is brought about by va-sodilators. Ensuing inflammation is characterized by anexudation or leakage of plasma from the blood vessels intothe inflamed tissue. This process is facilitated by an in-creased permeability of the endothelium and the aug-mented hydrostatic pressure in the blood capillaries. Next,cytokines and chemokines cause leukocytes (macro-phages, neutrophils, etc.) to infiltrate the inflamed tissueto accommodate the phagocytosis of cellular debris andpathogens. Finally, fibroblasts proliferate to reinstate tis-sue structure. All cells involved in the inflammatory pro-cess can sense the environment, responding to variousproinflammatory stimuli, and as a result initiate cytokineand chemokine cascades (5–7).

As such, the increase of inflammatory mediators, suchas cytokines [TNF� (afterward referred to as TNF), IL-1�,IL-6, granulocyte monocyte-colony stimulating factor(GM-CSF), etc.], chemokines [IL-8; regulated upon acti-vation, normal T-cell expressed and secreted (RANTES);melanoma growth-stimulating activity (Gro)-�; etc.],growth factors [fibroblast growth factor (FGF), epidermalgrowth factor (EGF), etc.], lipid-derived mediators (pro-stanoids, leukotrienes,), receptors [TNF receptor (TNFR),Toll-like receptor (TLR)], enzymes [inducible nitric oxidesynthase (iNOS), cyclooxygenase-2 (COX-2), matrix met-alloproteinases (MMPs), phospholipase A2, etc.), adhesionmolecules [intercellular adhesion molecule-1 (ICAM1), vas-cular cell adhesion molecule-1 (VCAM1), E-selectin, P-se-lectin], and peptides (bradykinin, tachykinin, endothelin), isdeemed pivotal for the propagation and progression of in-flammation. Conversely, to control the course of the inflam-matory process, modulatory and antiinflammatory cyto-kines such as IL-10, IL-4, and TGF� are released (8).

On a molecular level, inflammatory diseases are medi-ated by stimulation via proinflammatory signals, like thebacterial lipopolysaccharide (LPS), viral factors, or theself-produced IL-1� or TNF. Binding of these agents totheir respective receptors culminates predominantly inthe activation of activator protein-1 (AP-1) and NF-�B,but also in the activation of other transcription factors.In turn, both NF-�B and AP-1 stimulate the expression

of proinflammatory cytokines, chemokines, and adhe-sion molecules, thus propagating cellular inflammation(Fig. 1) (9 –12).

These activation pathways rely on a signaling cascadeof intermediary factors and especially kinases. For in-stance, soluble TNF binds to a membrane-imbeddedTNFR. Upon binding of TNF to TNFR1, the receptorhomotrimerizes and recruits the TNFR-associated deathdomain (TRADD) protein to the cytoplasmic death do-main of TNFR1 (13–16). Subsequently, the adhered pro-tein complex is supplemented by receptor-interacting pro-tein 1 (RIP1) and TNFR-associated factor 2 (TRAF2) (14,17–19). Ultimately, these adaptor proteins, specificallyTGF-activated kinase 1 (TAK1), NF-�B-inducing kinase(NIK), or MAPK kinase kinase (MEKK3), phosphorylatethe inhibitor of NF-�B (I�B) kinase (IKK) complex andtrigger its dissociation (12, 19–22). This process leads tothe activation of the IKK-NF-�B pathway, critical for in-ducing tissue inflammation. Additionally, TNFR ligandbinding results in the activation of ERK, p38, and c-JunN-terminal kinase (JNK) MAPK via a multilayered kinasecascade (23–25). The MAPK family of protein kinasescomprises ERK, JNK, and p38 MAPKs, of which the ac-tivation and function are regulated by upstream kinasesand stress-related inducers (25). The multilayered activa-tion cascade, enhancing the intracellular signal intensityand integrating various stimuli, is constructed bottom upby MAPKs, MAPK kinases (MAP2Ks or MKKs) andMAPK kinase kinases (MAP3Ks or MEKKs) (see Fig. 5).Preferentially, MAPKs target S/T protein residues, fol-lowed by a P (3, 25). These MAPKs diverge the web ofTNF-affected factors via multiple downstream kinase, co-factor, and transcription factor targets. Interestingly, theMAPK cascade is also involved in the posttranslationalcontrol of NF-�B and activation of AP-1 (26, 27).

Alternatively, IL-1� signals via activation of the type IIL-1 receptor (IL1RI) and its associate intermediates my-eloid differentiation primary response gene 88 (MyD88)and IL-1R-associated kinases (IRAK) 1, IRAK2, andIRAK4 and TAK1 toward activation of the MAPK path-way, AP-1, and NF-�B (11).

Lastly, various viral and bacterial factors can activatethe related Toll-like receptors (TLR1-10). For example,TLR3 is activated by double-stranded RNA and TLR4 bythe bacterial cell wall component LPS (28, 29). TLR3 andTLR4 signal via TIR domain-containing adapter-inducinginterferon (IFN) � (Trif) and MyD88, and via MyD88-adapter like protein (Mal) and translocating chain-asso-ciating membrane protein (Tram), respectively, to onceagain result in the activation and modulation of the MAPKpathway, AP-1 and NF-�B transcription factors (11, 12,

832 Beck et al. GR Crosstalk with Kinases and Phosphatases Endocrine Reviews, December 2009, 30(7):830–882

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

28, 29). Alternatively, TLR3 and TLR4 can signal viaTANK-binding kinase 1 (TBK1) and IKK� to activate IFNregulatory factor (IRF) 3 and to the subsequent transcrip-tion of type I IFN-inducible genes (30).

1. NF-�BBecause NF-�B is activated by a broad range of inflam-

matory and environmental stimuli, this transcription fac-tor, which plays critical roles in both innate and adaptiveimmunity, serves as a biological sensor. Moreover, NF-�Bis a pivotal regulator of inflammation because its activitylevel is raised in—and defective NF-�B signaling is asso-

ciated with—an ever increasing list of inflammatory andpathological conditions (31–36).

a. NF-�B structure. The transcription factor NF-�Bcomprises a family of five members: p65 (RelA), RelB,c-Rel, NF-�B1 (p50/p105), and NF-�B2 (p52/p100). Allmembers are characterized by an N-terminal Rel-homol-ogy domain, which is required for homo- or heterodimer-ization, sequence-specific DNA binding, and nucleartranslocation of NF-�B via its nuclear localization signal(NLS). Furthermore, RelA/p65, RelB, and c-Rel have C-terminal transactivation domains (TADs), whereas NF-

FIG. 1. Inflammation at a molecular level: a simplified scheme. TNFR activation by TNF, IL1RI by IL1�, TLR3 by double-stranded RNA (dsRNA), TLR4by bacterial LPS, and activation of other TLRs can signal via specific intermediary factors such as TRADD, TRAF2, RIP1, MEKK3, TAK1, TAB2/3, andNIK (for the TNFR), MyD88, IRAKs, TRAF6 and TAK1 (for IL1RI), Trif, RIP1, TRAF6 and TAK1 (for TLR3) and MyD88, Mal, Trif, Tram, RIP1, IRAKs, TRAF6and TAK1 (for TLR4) to the MAPK pathway and to the activation and regulation of NF-�B and AP-1. Additionally, triggering TLR3 or TLR4 signalingcascades can instigate IKK� and TBK1 activation and subsequent IRF3-regulated gene transcription (10–12, 29). TAB, TAK1-binding protein.

Endocrine Reviews, December 2009, 30(7):830–882 edrv.endojournals.org 833

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

�B1 and NF-�B2 are, respectively, p105 and p100 C-ter-minal ankyrin repeat-containing precursors (10), whichare proteasomally processed to yield N-terminal productsNF-�B p50 and p52, respectively. These mature species thenheterodimerize with a TAD-containing NF-�B family mem-ber or with the TAD-bearing I�B protein B cell lymphoma-3(Bcl-3) to form transcriptionally active complexes (37).

All NF-�B family members can form homo- or het-erodimers directing the dimer to a specific set of targetgenes. Recent evidence suggests that alternative splicing ofNF-�B components provides an additional way of con-trolling NF-�B signaling (38). NF-�B dimer function isspecific because the ablation of a certain NF-�B familymember cannot be compensated for (39). Because specificNF-�B dimer-promoter interaction is both context- andstimulus-dependent, the sequence of the NF-�B recogni-tion site is not the sole determinant of NF-�B dimer-pro-moter interfacing (40–43). The main research target ininflammation is the prevalent heterodimer NF-�B p65-p50, of which p50 can increase DNA binding (44) and p65confers transcriptional regulation (10).

b. NF-�B activation. With regard to the NF-�B activationmechanism, multiple pathways have been described. Themechanism by which NF-�B is activated by proinflamma-tory TNF is commonly referred to as the canonical acti-vation pathway (Fig. 1). In this canonical NF-�B activa-tion pathway, the transcription factor NF-�B dimerp65-p50 is, in its resting state, held in the cytoplasm by aninhibitory I�B molecule, most commonly I�B� (10, 45).I�B associates via its ankyrin repeats domain with NF-�B,which thus masks the NF-�B and I�B NLS motifs, conse-quently restricting inactive NF-�B to the cytoplasm. Whencells are challenged with a proinflammatory signal, such asTNF, the IKK complex becomes activated. The activatedIKK complex can phosphorylate I�B� on S32 and S36 orI�B� on S19 and S23, leading to polyubiquitination ofI�B� or I�B� and ultimately culminating in I�B degrada-tion by the 26S proteasome (10, 46, 47). Because NF-�Bis released from its cytoplasmic constraint, NF-�B trans-locates into the nucleus, guided by its NLS, where it canbind specific genomic target sequences.

The IKK complex comprises the catalytically activesubunits IKK� and IKK� together with the scaffold reg-ulatory subunit IKK� [NF-�B essential modulator(NEMO)] (48). Additional, but most likely transient, com-ponents of the IKK complex are the chaperoning heatshock protein 90 (Hsp90), cell division cycle 37 protein(cdc37), and protein rich in amino acids E, L, K, and S(ELKS) (49–51). Hsp90 is necessary to allow the relocal-ization and thus activation of the IKK complex to themembrane-associated and activated TNFR1 complex,

whereas cdc37 mediates the transient recruitment ofHsp90 to the IKK complex (49, 50, 52). Furthermore,ELKS is a necessary regulatory component of the IKKcomplex, serving as an auxiliary docking protein for I�B�

(51). The IKK complex-mediated phosphorylation of I�Brequires IKK� and IKK�, but does not necessitate IKK�,although IKK� can also phosphorylate I�B (47, 53).

Secondly, the noncanonical pathway, initiated via Bcell-activating factor (BAFF), lymphotoxin �, and otherinducers, entails cytokine, among which TNF, and virus-initiated NIK activation (54, 55). Activated NIK triggersIKK� homodimer activation, and subsequently S865,S869, and S871 of the p100 subunit in the p100-RelBcomplex becomes phosphorylated (56). This phosphory-lation induces proteasomal processing to the p52 NF-�Bsubunit (57–59). The p52-RelB complex, consequently,translocates to the nucleus, where it targets specific genepromoters, such as IL-2 (60, 61), for activation.

c. Posttranslational modifications of NF-�B. Subsequent tothe NF-�B activation process, NF-�B activity is substan-tially modulated by various posttranslational modifica-tions: acetylation (62–65), SUMOylation (66), and phos-phorylation (63, 67), of which the last is best characterizedso far. The intracellular control of NF-�B transactivation(duration and intensity) (64, 68, 69), subcellular localiza-tion (70), DNA-binding affinity (64, 65, 69, 71), andNF-�B interaction with cofactors and I�B� (72–74) viavarious posttranslational modifications forms an intricateweb of NF-�B management. Phosphorylation sites ofNF-�B p65 are spread out over the Rel-homology domainand TAD1 and TAD2 (Fig. 4C). Although some of thesephosphomodifications contribute to the transcriptionalactivity of NF-�B (73, 75–78), others do not (79).

In particular, the phosphorylation of NF-�B p65 S276by p38 and ERK MAPK-activated mitogen- and stress-activated protein kinase 1 (MSK1) or protein kinase A(PKA) is pivotal for proper initiation of specific inflam-matory gene expression (73, 74, 76, 80–84). Phosphor-ylation of NF-�B p65 S276 facilitates association of p65with the coactivators cAMP-responsive element-bindingprotein (CREB)-binding protein (CBP) and p300 (73, 74,83, 85) and the transcription elongation complex P-TEFb(positive transcription elongation factor b), consisting ofcyclin-dependent kinase, Cdk9 and cyclin T1 (81). Fur-thermore, NF-�B p65 S276 phosphorylation can enhancethe displacement of the inhibitory histone deacetylase 1(HDAC1)-NF-�B p50 complex to derepress proinflam-matory gene promoters (85). Therefore, NF-�B S276phosphorylation is a crucial step in NF-�B driven pro-moter activation of specific gene targets (73, 76, 80, 86).However, not all NF-�B-dependent genes require the

834 Beck et al. GR Crosstalk with Kinases and Phosphatases Endocrine Reviews, December 2009, 30(7):830–882

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

phosphorylation of NF-�B S276 for their transcription.Whereas the transcription of ICAM, VCAM, Gro-�, IL-8,and IL-6 depend on the NF-�B p65 S276 phosphorylation,the NF-�B-mediated transcription of major histocompat-ibility complex-I (MHC-I), mangano-superoxide dis-mutase (MnSOD), and I�B� do not (73, 81, 86). Of note,NF-�B-mediated gene transcription independent of NF-�BS276 phosphorylation shows constitutive binding of RNApolymerase II (RNA pol II) (81). This differentiation sug-gests that a NF-�B phosphorylation code controls NF-�B-mediated transactivation of specific target genes (87),possibly defined by the architecture and topology of thetarget promoter (88). Consequently, the selective phos-phorylation of S276 of NF-�B p65 may be responsible forsome of the differential sensitivity of certain �B sites torepression by GCs. Because the phosphorylation of NF-�Bp50 S337, which appears to be essential to NF-�B p50DNA binding, is mediated by PKAc and possibly MSK1,these kinases are attributed an important role in theregulation of NF-�B p65-p50-dependent gene tran-scription (89).

Various kinases can phosphorylate NF-�B S536: IKK�,IKK�, TBK1, IKK�, ribosomal S6 kinase 1 (RSK1), andglycogen synthase kinase (GSK) 3� (71, 90–96). Also, thephosphorylation of NF-�B p65 S536 can contribute to theactivity of NF-�B p65, most likely via facilitating the in-teraction between activated NF-�B and p300 (62, 93).Additionally, NF-�B p65 S536 phosphorylation is re-ported to weaken the binding of NF-�B to I�B, thus pro-longing the activity of NF-�B in the nucleus (71, 93). Ofnote, acetylation of NF-�B of multiple lysine residues,probably by the histone acetyl transferase (HAT) activityof CBP/p300, is preceded by and requires NF-�B S276 andS536 phosphorylation (62).

A third NF-�B phosphorylation that can positively af-fect NF-�B activity is the protein kinase C (PKC) �-medi-ated phosphorylation of NF-�B p65 S311. Similar toNF-�B p65 S276 and S536 phosphorylation, the phos-phorylation of NF-�B at S311 can enhance the interactionof NF-�B p65 with CBP. Moreover, NF-�B p65 S311phosphorylation augments NF-�B recruitment to �B sitesclose to the promoter of the proinflammatory cytokineIL-6 (75).

Phosphorylation of NF-�B p65 T254 by an unknownkinase can enhance the NF-�B p65 activity via inducingthe interaction of T254 and P255 of NF-�B p65 with thenuclear peptidyl-prolyl isomerase Pin1 [protein NIMA(never in mitosis gene a)-interacting]. This interaction re-sults in the isomerization of the P residue, entailing a con-formational change of NF-�B p65. Consequently, NF-�Bp65’s binding affinity for I�B� is decreased. Furthermore,

this modification stabilizes the NF-�B p65 protein andpromotes the nuclear translocation of NF-�B p65 (97).Conversely, ubiquitination of NF-�B p65, inducing itsproteolysis, is mediated by the E3-ubiquitin ligase sup-pressor of cytokine signaling (SOCS) 1. Because thebinding sites for Pin1 and SOCS1 lie in close proximity,competition for binding might be possible (98). The pro-teasomal degradation of DNA-bound NF-�B p65 pro-motes transcriptional termination (99).

For the phosphorylation of NF-�B p65 S529 by caseinkinase 2 (CK-2), it is still unclear whether this phosphor-ylation can affect NF-�B-mediated transcription (100,101). In contrast, phosphorylation of T505 via activationof checkpoint kinase 1 (Chk1) and ATM/Rad3-related(ATR) checkpoint kinase could decrease the activity ofNF-�B via enhancing its interaction with HDAC1 (102–104). Even so, the IKK�- or GSK3�-mediated phosphor-ylation of NF-�B p65 S468 negatively affects NF-�B ac-tivity (68, 79), whereas IKK�-mediated phosphorylationof this same residue was associated with NF-�B transac-tivation (105).

Conversely, endogenous protein phosphatase (PP) 2Acan associate with and dephosphorylate NF-�B p65 (106).Moreover, pharmacological blockage of PP2A leads toincreased phosphorylation of NF-�B p65 (68, 106). Ad-ditionally, association of PP4 has been linked to the acti-vation of NF-�B via the dephosphorylation of T505 (102,107, 108). Furthermore, it was suggested that a rapid de-phosphorylation of NF-�B S536 could contribute toswitching off NF-�B-dependent gene transcription (109).

d. NF-�B crosstalk. NF-�B-mediated transcription can fur-thermore be coregulated by crosstalk of NF-�B with othertranscription factors and association of NF-�B withvarious cofactors (110). Positive crosstalk of NF-�B-promoting proinflammatory gene transcription has beendescribed for aryl hydrocarbon receptor, specificity pro-tein 1 (Sp1), IRF, signal transducer and activator of tran-scription (STAT), activating transcription factor (ATF),CREB, and AP-1 (111–118). The binding of multiple dis-tinct transcription factor complexes occurs in a highly dy-namic manner (110, 119). An example of negativecrosstalk with NF-�B is shown by GR (120). Cofactorscan either coactivate or corepress NF-�B-mediated genetranscription. Functionally, these cofactors can stimulateor repress the transcriptional activity of the enhanceosome(i.e., the multiprotein complex mediating promoter acti-vation and gene transcription) (121) or alter the chromatinstructure. Many coactivators, including p300, CBP, p300/CBP- associated factor (p/CAF), and steroid receptor co-activator 1 (SRC1) have a HAT domain, capable of acety-lating histones but also other proteins (122–127).

Endocrine Reviews, December 2009, 30(7):830–882 edrv.endojournals.org 835

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

Interestingly, IKK� can phosphorylate CBP, increasingCBP activity and CBP binding to p65 (128). Conversely,corepressors often include HDAC activity (HDAC1,HDAC2, HDAC3) and can be directly or indirectly re-cruited to NF-�B-dependent gene promoters (85, 129–133). Other NF-�B-dependent gene-associated core-pressors are the NF-�B p65-p50-binding silencingmediator for retinoid and thyroid-hormone receptors(SMRT) and the NF-�B p50-binding nuclear corepres-sor (NCoR) (134, 135).

e. NF-�B-targeted genes. After NF-�B activation, nuclearNF-�B p65 can enhance gene expression of multiple proin-flammatory genes via the occupation of a NF-�B-specificpromoter recognition site (136). Because NF-�B can beconsidered a central regulator of proinflammatory genetranscription, NF-�B function is a highly dynamic signal-ing event, showing differential temporal expression pro-files for different genes (40, 137). Moreover, associationof NF-�B with its specific recognition sites is transient,with a half-life of seconds, suggesting a dynamic regula-tion of enhanceosome composition and gene transcriptionconstantly sensing the inflammatory status of its environ-ment (119).

The canonical or classical NF-�B activation pathwaycontrols among others leukocyte activation/chemotaxis,cellular metabolism, antigen processing, and negative reg-ulation of the TNF signaling pathway (138, 139). Theactivation of NF-�B results in gene transcription of cyto-kines (e.g., IL-6), chemokines (e.g., IL-8 and RANTES),adhesion molecules (e.g., E-selectin), enzymes (e.g.,iNOS), and other inflammatory mediators (8). Further-more, the cytokine TNF, but also IL-1�, can activate NF-�B, and these are thus mediators of a feedforward mech-anism perpetuating inflammation.

However, it is essential to prevent the uncontrolledpropagation of inflammation, which would cause sys-temic disorders. Therefore, NF-�B activation is also sub-ject to an autoregulatory negative feedback loop. First,gene transcription of I�B� is stimulated by NF-�B activa-tion. The newly synthesized I�B� replenishes the formerproteasomally degraded I�B� levels and can bind to activeNF-�B complexes in the nucleus, weakening NF-�B DNAbinding and subsequently transporting NF-�B back to thecytoplasm (10, 140, 141). Furthermore, upregulation ofthe anti-apoptotic protein A20, the A20-binding inhibitorof NF-�B activation, ABIN-1, the TNF�-converting en-zyme TACE, the tumor suppressor cylindromatosis(CYLD), the antiinflammatory cytokine IL-10, and themicroRNA miR-146 ultimately results in a negative reg-ulation of NF-�B activity (142–153).

2. AP-1AP-1 is a transcription factor of general importance for

many cellular processes in different organs, including in-flammation. Among the target genes of AP-1 are impor-tant regulators of cell proliferation, differentiation, andapoptosis. The DNA binding of the AP-1 complex to 12-O-tetradecanoylphorbol-13-acetate (TPA)-response ele-ment (TRE) sequences is rapidly induced by growth fac-tors, cytokines, and oncoproteins, which are implicated inthe proliferation, survival, differentiation, and transfor-mation of cells (154).

a. AP-1 structure. Similar to NF-�B, AP-1 is a homo- or het-erodimeric transcription factor complex that can be tar-geted to its regulatory sites in a sequence-specific manner(Fig. 1). The subunits of AP-1 are selected from the Jun(c-Jun, v-Jun, Jun B, and Jun D), Fos (c-Fos, Fos B, Fra-1,and Fra-2), activating transcription factor [ATF2, ATF3,B-ATF, Jun dimerization protein (JDP)-1, JDP-2], or MAF(MAFA, MAFB, c-MAF, NRL, MAFF, MAFG, andMAFK) families (154–156). The most predominantly oc-curring forms of AP-1 are Fos/Jun heterodimers, whichshow preferential binding to heptameric TREs when com-pared with ATF2/Jun homo- or heterodimers, which bindpreferentially to octameric TREs and are strongly inducedby the tumor promoter TPA (157). AP-1 proteins areknownasbasic leucine-zipperproteinsbecausetheydimerizethrough a leucine zipper motif and contain a basic domainfor interaction with the DNA backbone. While the Fos pro-teins do not form homodimers but can heterodimerize withmembers of the Jun family, the Jun proteins can both ho-modimerize and heterodimerize with other Jun or Fosmembers to form transcriptionally active complexes (155,158). In addition to Fos proteins, Jun proteins can alsoheterodimerize efficiently with other AP-1 family mem-bers, such as the ATF family (159), and other basiczipper-containing transcription factors (156, 160). Al-though members of the Jun and Fos families share a highdegree of structural homology, the individual AP-1dimers exert significant differences in their DNA-bind-ing affinity and their capability to activate or suppressgene expression (155).

b. AP-1 activation and activity. Regulation of net AP-1 ac-tivity can be achieved through changes in transcriptionof genes encoding AP-1 subunits, control of theirmRNA stability, posttranslational processing, turnover ofpreexisting or newly synthesized AP-1 subunits, and spe-cific interactions between AP-1 proteins and other tran-scription factors and cofactors.

Various stimuli, including physiological agents such asgrowth factors and cytokines, pharmacological com-

836 Beck et al. GR Crosstalk with Kinases and Phosphatases Endocrine Reviews, December 2009, 30(7):830–882

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

pounds, such as anisomycin, phorbol esters and okadaicacid, and stressors such as UV radiation, hyperosmoticand heavy metal stress, rapidly elicit transcription of“immediate early” (IE) genes, such as those of the Fos andJun families, by activation of MAPK cascades (161). Theseso-called “IE genes” are activated directly and require nonew transcription or translation for their induction. TheFos and Jun proteins then activate and repress other genes,thereby producing secondary transcriptional reprogram-ming appropriate to specific stimuli. Because these pro-teins are differentially expressed and regulated in a cell-and stimulus-specific manner, every cell type produces acomplex mixture of AP-1 dimers with subtly differentfunctions (154, 161). In most cases, enhanced expressionof c-jun gene, protein, and function is not a solitary eventbut can be accompanied by an induction of transcriptionfactors that are related to c-Jun (e.g., Jun B, Jun D), Fosfamily members (Fos, FosB, Fra-1/2), or ATF familymembers allowing the formation of functionally differ-ent heterodimers in a cell- and time-specific manner.Whereas some AP-1-regulated genes are preferentially in-duced by cJun-cFos dimers, others are mainly induced byJun D-Fra-1 dimers. Fra-1 and Fra-2 promoters are acti-vated by Jun-Fos dimers. The fos and jun genes are con-trolled by multiple upstream elements; for human c-fos,these include a cis-inducible element, a serum responseelement (SRE), a ternary complex factor site (TCF), anAP-1 site, an AP-1/CRE, a direct repeat, and a cAMP re-sponse element. The human c-jun promoter is controlledby the Jun2 AP-1 site, a footprint (FP), an NF-Jun site, twooverlapping Sp1 sites, a CCAAT box, the Jun1 AP-1 site,a related to serum response factor site (RSRF), and in the5�-untranslated region, two AP-2 sites and a weak AP-1site (158, 161, 162). These regulatory elements are highlyconserved between mouse and human.

AP-1 is induced by several external stimuli that increaseMAPK activity. Expression of c-Fos is induced by TCFs,which are activated through phosphorylation by the ERKMAPKs.

IE gene expression of c-Jun may be achieved throughATF or c-Jun. Alternatively, c fos and myocyte enhancerfactor 2 (MEF2), transcription factors can also induce c-jun expression in other contexts. The targeting of MAPKsto transcription factors controlling c-fos and c-jun geneexpression has been very clearly established, and phos-phorylation of these factors is further implicated in re-cruitment of coactivators such as p300/CBP and p/CAF, tothese promoters (163, 164).

c. Posttranslational regulation of AP-1. Posttranslational ac-tivation of AP-1 is produced by translocation of a stimu-lus-dependent kinase, such as MAPKs or their effector

kinases, which bind to and phosphorylate these factors,effecting transactivation and transcription of the associ-ated gene (154, 158). The mechanism of posttranslationalcontrol is most extensively documented in the case of mi-togen- and cellular stress-induced hyperphosphorylationand, in particular, activation of Jun through the JNK cas-cade (165, 166). Activated by a MAPK cascade, the JNKstranslocate to the nucleus, where they phosphorylate Junwithin its N-terminal TAD at S63 and S73 and therebyenhance its transactivation potential. The JNKs also phos-phorylate and potentiate the activity of JunD and ATF2.Alternatively, p38 MAPK-dependent phosphorylation ofc-Jun S63 and S73 has been demonstrated in response toa UV stimulus (167). Moreover, DNA-binding activity ofc-Fos, FosB, and JunB were also dependent on the p38protein kinase activity, whereas JunD, Fra-1, and Fra-2were not affected. A complex network of signaling path-ways that involves external signals for growth factors-Ras-Raf-MEK-ERK families also regulates AP-1 activity(168). Activated Ras or MEK1 primarily induces Fra-1and c-Jun after N-terminal phosphorylation by JNKs. Incontrast to c-Jun, JunB is not an efficient substrate forJNK. Furthermore, although JunD can be phosphorylatedby JNK, its phosphorylation requires interaction withpartners that provide a docking sequence (169). JunB andJunD are less potent collaborators of Ras in cell transfor-mation than c-Jun, which correlates with their lower tran-scriptional activity. ERKs are persistently activated bygrowth factors and oncogenic Ras in tumors and are pos-itive regulators of tumorigenesis (170). As such, they con-tribute substantially to the increased expression and acti-vation of AP-1 members in many tumor types. Potentialcandidates for kinases that regulate Fos activity are the Fos-regulating kinase (FRK), RSK2, and p38 and ERK MAPK(170). When the AP-1 complexes are present in larger quan-tities, glycogen synthase kinase 3� (GSK3�), RSK2, caseinkinase 2, cdc2, PKA, and PKC phosphorylate Fos and Junproteins, thereby regulating their protein stability, DNA-bindingactivity,andthetransactivatingpotentialof theAP-1family members (154, 158).

d. AP-1 dimers and crosstalk. The activities of AP-1 are par-tially modulated through the differential expression of itsindividual components, which determines their dimercomposition (154, 171), and partially through their spe-cific context—cell type, response element sequences andorganization, modification state, interaction with otherregulatory factors, promoter sequence and organization,etc. (172). Whereas Jun, Fos, and FosB are often associ-ated with a strong transactivation potential, JunB, JunD,Fra-1, and Fra-2 are usually found in a context in whichonly a weak transactivation potential is supported. Under

Endocrine Reviews, December 2009, 30(7):830–882 edrv.endojournals.org 837

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

specific circumstances, the latter might even act as repres-sors of AP-1 activity by competing for binding to AP-1sites or by forming “inactive” heterodimers with Jun, Fos,or FosB. The crucial and determining aspects of “context”are not yet fully understood. To illustrate the importance,it was described that the composition of AP-1 regulatorycomplexes and the biological activities of the bound fac-tors are dynamic and dependent on cell and response el-ement contexts. Col3A is the response element in the col-lagenase-3 gene that confers activation by phorbol estersand repression by GCs in human U2OS osteosarcomacells. The subunit composition and activity of AP-1, whichbinds ColA3, parallels the intracellular level of c-Fos,which is modulated by phorbol esters and GCs. A similarAP-1 site at the collagenase-1 gene, however, not induciblein U2OS cells, was not bound by AP-1, underscoring theimportance of a context-dependent gene regulation (172).

The decision as to whether AP-1 is oncogenic or anti-oncogenic might depend on the antagonistic activity ofdifferent Jun proteins, but it is probably also influenced bytumor type, tumor stage, and the genetic background oftumors (158, 168). For example, c-Jun-Fra-2, but not c-Jun-Fra-1 or c-Jun-c-Fos, inhibits the growth arrest of im-mortalized fibroblasts at confluence and under low-serumconditions. Using dimer-specific mutants of AP-1 pro-teins, in which manipulation of the leucine-zipper domainallows only specific dimers to form, it was demonstratedthat the c-Jun-induced transformation program can beseparated into two distinct pathways: c-Jun-ATF2 activitytriggers growth factor independence, and c-Jun-c-Fos ac-tivity causes anchorage-independent growth (158). Tofully elicit their oncogenic potential, most AP-1 compo-nents need the activity of “cooperating” oncoproteins,which often induce the expression of Jun and Fos proteinsbut also support AP-1-mediated cell transformation byposttranscriptional mechanisms (173, 174). The main co-operating partner of AP-1 is the Ras pathway because celltransformation by activated Ras or MEK1 (MAPK kinase)induces AP-1 protein expression (168). The oncogenic co-operation of c-Jun with Ras and other oncoproteins, func-tioning upstream of Ras, requires N-terminal phosphor-ylation of c-Jun by JNKs.

B. Glucocorticoid receptor-mediated signalingInflammation can be controlled by the stress-induced

release of GCs, mainly cortisol in humans. Besides beingefficient in combating inflammation, GCs display pleio-tropic effects in the regulation of protein, lipid, andcarbohydrate metabolism; innate and adaptive immunesystems; stress homeostatic regulation; reproductive pro-cesses; and growth and brain functions such as memoryand behavior (175).

1. GR domainsThe GR, which binds and mediates the signals of the

GCs, belongs to the superfamily of nuclear receptors. Thissuperfamily can be categorized according to ontogeny andfunction (176). In that respect, the GR is classified asNR3C1, most proximate to the mineralocorticoid recep-tor (MR). However, GRs and MRs nevertheless affect dis-tinct target genes (177). The GR consists of an N-terminaldomain, encompassing a first TAD [activation function 1(AF-1)] responsible for transcriptional activation and as-sociation with certain basal transcription factors (2, 178),a DNA-binding domain (DBD), in which the dimeriza-tion or D-loop within the two zinc fingers plays a rolein GR dimerization and DNA-binding functions (178 –183), and a C-terminal ligand-binding domain (LBD),containing a second TAD (AF-2) and also protein-bind-ing sites (178, 184 –189). Interestingly, in addition toGR ligands, different GR/DNA-binding sequences candifferentially affect GR conformation and regulatoryactivity; as such, DNA can be considered as a sequence-specific allosteric ligand of GR (190). Furthermore, theDBD can also account for GR-transcription factor as-sociation (191–193). In close proximity of the DBD andat the end of the LBD, two nuclear localization sites, theligand-independent NL1 and the ligand-dependentNL2, have been described that direct the activated GRtoward the nucleus (Fig. 2) (194, 195).

The GR can exist as multiple isoforms due to alternativesplicing (GR�, GR�, GR�, GR-A, GR-P) and differenttranslational start sites (GR�-A, GR�-B, GR�-C1, GR�-C2, GR�-C3, GR�-D1, GR�-D2, or GR�-D3). Both theN-terminal and C-terminal domain of GR can vary de-pending on the isotype, but the DBD most often remainsconstant. GR�, stretching to 777 amino acids in humans,is the most predominant, functional GR and currently themain research target. In contrast, GR� cannot bind GCsand is not ubiquitously expressed. However, GR� can actin a dominant-negative manner to suppress actions ofGR� and is implicated in GC resistance (196, 197). Al-though GR� is expressed throughout the body, the ex-pression pattern of the isoforms can be restricted tocertain cell types, possibly fine-tuning the GC-GR re-

FIG. 2. Structure and domain functions of the GR. NTD, N-Terminaldomain; HR, hinge region; TF, transcription factor.

838 Beck et al. GR Crosstalk with Kinases and Phosphatases Endocrine Reviews, December 2009, 30(7):830–882

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

sponse in various tissues (196, 198 –200). Additionally,various function-altering polymorphisms have been de-fined for the human GR (hGR) (199 –201). Membrane-associated and mitochondrial GR proteins have alsobeen described (202–206). Alternatively, GCs can actvia G protein-coupled receptors and its downstreamcascades (207). The precise role of GC signaling viathese receptors, however, still awaits further research. Itis expected that GR�, being the most predominant GRspecies, will remain a prime focus of therapeutic atten-tion for some time still.

In an uninduced state, GRs reside predominantly in thecell cytoplasm in association with a multimeric molecularchaperone complex, keeping the ligand-binding pocketreceptive to high-affinity hormone binding and inactivat-ing the NLS. This chaperone complex consists of severalHsps, such as Hsp90, Hsp70, and the Hsp90-bindingprotein p23, the hsp-organizing protein Hop, and tet-ratricopeptide repeat proteins that also bind Hsp90 suchas FK506-binding protein (FKBP) 51, FKBP52, cyclophilin40 (Cyp40), the C-terminus of Hsp70-interacting protein(CHIP), or the phosphatase PP5 (see Section II.B). However,in a single lysate, not all chaperone complexes are equallycomposed (186, 208).

2. Triggering GR-mediated signaling

Once their cellular target is reached, GCs can cross themembrane because they are small hydrophobic molecules.Alternatively, natural GCs can enter the cell via the steroidhormone recognition and effector complex (209). GCbinding to cytosolic GR instigates a conformationalchange in this receptor (185, 210). The active conforma-tional state of GR and its subsequent modifications allowGR to shed most of its chaperone complex, unmasking theNLS. These steroid-dependent changes allow GR to freelyand rapidly move along cytoskeletal tracts to ultimatelytranslocate into the nucleus (186). Subsequently, GR cangive rise to positive or negative transcriptional effects (Fig.3) (120, 211, 212) and rapid nontranscriptional effects(213). Together, these genomic and nongenomic path-ways controlled by GRs culminate in a multilayered andfine-tuned control mechanism for gene regulation. In ad-dition to the classic slow mode of GC action occurringbetween hours to days, increasing evidence is culminatingfor more rapid GC effects on cellular responses, takingplace within minutes. Because these GC effects are too fastto be regulated at the transcriptional level, they are termednongenomic, to distinguish them from the traditionalgenomic mode of GC action (214). Rapid GC effects may

FIG. 3. Activation and nuclear actions of the GR. The unactivated, cytoplasmic GR is complexed with chaperone proteins. Binding of GCs to theGR instigates the nuclear translocation of GR. The binding of dimeric, activated GR onto GREs, DNA binding of GR in a concerted manner withanother transcription factor (TF), or binding of GR onto a TF via a tethering mechanism can all result in GC-directed promoter activation. Thistransactivation results in the expression of metabolic gene products, associated with the occurrence of side effects, and to the expression of anumber of antiinflammatory proteins. The antiinflammatory effects of GCs are predominantly mediated via interference of monomeric GR with thetransactivation capacity of TFs, such as NF-�B, via a tethering mechanism. Otherwise, GC-activated GR can also negatively regulate genetranscription via competition for an overlapping binding site (competitive GRE) or via DNA-binding crosstalk with another TF (composite GRE), orelse via the sequestration of a DNA-bound TF. Although the nature of these sequences is not well-defined, gene repression via direct binding ofGR onto a so-called nGRE has also been reported.

Endocrine Reviews, December 2009, 30(7):830–882 edrv.endojournals.org 839

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

be transmitted by the GR or, more controversial, non-genomic GC activities might be mediated through non-specific physicochemical interactions with the plasmamembrane at high GC concentrations (215).

Although uninduced GR is mainly found in the cyto-plasm and GC-induced GR is largely nuclear, constitutiveshuttling between nucleus and cytoplasm has been re-ported for both nonactivated and activated forms of GR(216). It is, however, the import or export rate that deter-mines the location of the bulk of GR at any given time.Whereas importin�- or importin�-based nuclear import ofGR is mediated by the GR NLSs NL1 and NL2 (195, 217,218), calreticulin-based and chromosome region mainte-nance 1 (CRM1)/exportin1-based mechanisms have beendescribed to account for the nuclear export of liganded orunliganded GR (195, 219–221). Moreover, the locationof GR is codetermined by the recently discovered nuclearretention signal (NRS), which actively configures GR tothe nucleus (222). Additionally, the DNA-binding abilityof GR and ligand-specific conformational changes in GRhave been previously linked with GR’s nuclear mobilityand cellular location (223–225).

Once activated, several mechanisms are set in motion todiscontinue the GR response. Although GR is assumed tobe constitutively expressed under a vast array of physio-logical conditions, GR mRNA expression is subject to neg-ative regulation by GCs (226–228). This might be ex-plained by the presence of negative GC response element(nGRE), AP-1, NF-�B, and CREB regulatory motifs in thepromoter of GR, all of which are negatively regulated byGCs (196). Alternatively, GCs can destabilize GR mRNA(229). Moreover, ligand-activated GR protein is degradedupon prolonged exposure to GCs by the proteasome-ubiq-uitin degradation pathway (230, 231). However, muta-tion of the N-terminal GR PEST motif abrogates ligand-dependent downmodulation and consequently boostsGR-mediated transactivation (232). Lastly, it was shownthat protein degradation of GR is linked to its export.Although hormone dissociation results in a rapid releaseof GRs from chromatin, unliganded GR is delayed in itsexport. Accelerated nuclear export of a nuclear exportsequence-tagged GR chimera is associated with an in-creased rate of hormone-dependent down-regulation. Theprotracted rate of receptor nuclear export may be a way ofincreasing the efficiency of biological responses to second-ary hormone challenges, via a limitation on receptordown-regulation and hormone desensitization (233).

3. Nuclear activity of the GRa.GRtransactivationandtransrepressionmechanisms.Ligandbinding of GCs to GR results in the nuclear translocationof GR where this receptor can act to modulate transacti-

vation of typical GC response element (GRE)-containingor other promoters.

Ligand-activated, nuclear GR can stimulate the expres-sion of certain genes via DNA binding of a dimerized GR.These GR homodimers bind in the major groove of DNAvia their zinc finger DBD and target the imperfect palin-drome of the consensus GRE (5� GGT ACA nnn TGT TCT3�) (234–236). This GRE can differ among promoters insequence, copy number, and relative location in the pro-moter (in relation to the TATA box or other transcriptionfactor-binding sites), regulating the specificity and mag-nitude of its response. GR may thus be modified in anallosteric manner by its response elements to generate apattern of regulation that is appropriate to an individualgene (237, 238). In DNA-binding GR-mediated transac-tivation research, either a simple GRE or concatamer GREmouse mammary tumor virus (MMTV) reporter gene con-struct is often used, which GCs can transiently activate(239, 240). However, various studies revealed that manyknown GC-inducible genes do not contain consensus GREsites and do not require binding of dimerized GR. Some ofthese could be classified as promoters containing compos-ite elements, in which GR collaborates with another tran-scription factor to enhance transcription in a cooperativemanner (211, 241–245). Essentially all genes have GRbinding motifs reasonably close (at an average of 15 kb) tothe transcription start site, however few of those are func-tional. Many elements lie very far (�50kb) from thestart sites, and at present there are no simple ways toprove that a given element is controlling a given gene.Assignments are done essentially by conservation, dem-onstration of GR occupancy in vivo, and proximity(246). Lastly, tethering, i.e., direct binding of GR toDNA-bound transcription factors, has also been de-scribed to positively regulate DNA transcription uponGC administration (Fig. 3) (247–249).

Various transcription factors [e.g., Sp1, STAT1,STAT3, STAT5, CCAAT enhancer-binding protein (C/EBP), Ets, Egr-1, AP-2, AP-1, and NF-�B] can function inconcerted array with GR to regulate and fine-tune tran-scription in a positive or negative manner (250–252). Forexample, ligand-activated GR can inhibit most, but notall, NF-�B-driven gene expression (211). Indeed, GR actsselectively to inhibit NF-�B at some, but not all, NF-�Bsites. Notably, it inhibits NF-�B action at the IL-8 but notat the I�B� gene. NF�B at IL-8 is phosphorylated atSer276 and recruits P-TEFb to promote elongation. GRrepresses IL-8, where transcription elongation depends onP-TEFb-mediated phosphorylation of pol II C-terminaldomain (CTD) S2, by competing P-TEFb from p65 asso-ciation. In contrast, GR fails to repress I�B gene expres-sion, where CTD S2 phosphorylation proceeds without

840 Beck et al. GR Crosstalk with Kinases and Phosphatases Endocrine Reviews, December 2009, 30(7):830–882

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

p65 recruitment of P-TEFb, and GR binding to p65 there-fore poses no interference (253, 254). The findings presentedabove represent a good example of a situation in which thecontext dependency is mechanistically understood.

Likewise, activation of the transcription factor NF-�Bp65 can repress most, but not all, GR-transactivated genepromoters (255, 256). The NF-�B-mediated inhibition ofGR transactivation mechanisms is called “reciprocal re-pression” and is partially based on the mutual interactionof GR with NF-�B and partially on the cellular context.

Additionally, SRCs of the p160 family, such as SRC-1and SRC-2, interacting with GR via its LBD LxxLL motif,and various cofactors for chromatin remodeling and his-tone modification [e.g., SRC-1 and TIF-2 associated mod-ulatory protein (STAMP), CBP/p300, p/CAF, switching ofyeast mating type/sucrose nonfermenting (SWI/SNF), andcoactivator-associated arginine methyltransferase (CARM)1] can regulate GC-mediated promoter activation (240,257–261). The nature of the GR ligand impacts GR cofactorbinding. Although administration of the synthetic GC dexa-methasone leads toGRSRC-1binding, ligandbindingofGRto the GR antagonist RU486 stimulates binding of GR to thecorepressor NCoR (262). Furthermore, the requirement forcertain cofactors and different GR interfaces can depend onthe targeted gene promoter (244, 263). Concerning histonemodifications, aGC-inducedphosphorylationofhistoneH3(H3) S10 and acetylation of H3 K14 in MMTV promoterchromatin is associated with a transcriptionally active pro-moter (257).

The DNA binding of ligand-activated GR is, however,not a static phenomenon. Upon activation, GR rapidlyassembles onto known GREs. The chaperones Hsp90 andp23 can localize to GR-bound GREs in a GC-induciblemanner and promote disassembly of a functional GR-GRE transactivation complex (264). Furthermore, the GRtransactivation regulatory complex has proven to turnover in an extremely dynamic manner, via a ligand-dependent “hit and run” mechanism (223, 265, 266).Combined with the release of GCs from GRs (264), thesemechanisms continuously sense cellular stress hormonesand thus allow an appropriate cellular response to varyingGC levels.

b. GR transactivation and inflammation. GC-mediated up-regulation of I�B�, GC-induced leucine zipper (GILZ),dual specificity phosphatase (DUSP) 1 (see Section IV.A),lipocortin-1/annexin A1, secretory leukocyte protease in-hibitor SLPI, IL-10, the decoy IL-1 receptor type II, dexa-methasone-induced Ras1 (Dexras1), downstream of ty-rosine kinase 1 (Dok-1), Src-like adaptor protein (SLAP),p11/calpactin binding protein, thymosin �-4sulfoxide,Clara cell secretory 10-kDa protein (CC10), �-adrenergic

receptors, SOCS1, and tristetraprolin (TTP) have all beensuggested to be involved in the GC-mediated combat ofinflammation (211, 243, 267–270). For example, becauseDNaseI footprinting studies in T47D/A1–2 cells demon-strated that regulatory factors bind to the I�B-� promoterafter GC treatment, Deroo and Archer (271) proposedthat GCs may be required for transcription factor bindingand subsequent transactivation of the I�B-� promoter.Higher levels of I�B-� would then block the activation ofNF-�B. GILZ, a classical GRE-driven target gene, alsoseems to mimic some aspects of GC action and inhibitsinflammatory cytokine-induced COX-2 expression inbone marrow mesenchymal stem cells, via blockage of thenuclear translocation of NF-�B (272). Annexin A1, an-other GC-regulated target gene, is believed to exert itseffects through the FPR receptor family of G protein-cou-pled receptors, of which the implication in the regulationof many inflammatory processes is increasingly being rec-ognized (273). GCs transcriptionally stimulate the syn-thesis of TTP, a zinc finger protein capable of destabilizingseveral proinflammatory cytokine mRNAs by binding toadenylate uridylate-rich elements (AREs) within their 3�untranslated regions, subsequently targeting them fordegradation (see Section III.B.2) (274). The effect of(de)phosphorylation on the functionality of some of thegene products of the above-mentioned list or the effect ofthese proteins on other kinases/phosphatase signalingpathways is discussed further below.

The overall role and contribution of the different de-scribed GC-induced antiinflammatory proteins in the GC-mediated antiinflammatory mechanism remains some-what controversial (211, 275, 276). In some studies, GCscan repress proinflammatory gene expression of IL-6,ICAM1, and COX-2 without the need for de novo proteinsynthesis (277, 278), whereas in other studies urokinaseplasminogen activator, COX-2, and IL-8 mRNA tran-scriptional repression by GCs have been found to partiallyrely on de novo protein synthesis (279–284). Neverthe-less, although a body of evidence supports that the prin-cipal and initial antiinflammatory potential of GR mayreside in its direct repression of proinflammatory gene ex-pression (236, 277, 285, 286), it is apparent that GR-mediated transactivation also plays a role. Moreover, theprecise contributions of transrepression vs. transactiva-tion mechanisms in the antiinflammatory actions of GRseem highly context-dependent and make matters evenmore complex (120). Hence, the idea that dissociating li-gands would remain powerful antiinflammatories whilemodulating side effects is likely simplistic.

c. GR-mediated promoter inhibition. Besides GR-mediatedtransactivation, ligand-activated GR can also act as a

Endocrine Reviews, December 2009, 30(7):830–882 edrv.endojournals.org 841

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

DNA-binding factor to repress specific gene transcriptionvia composite response elements or competitive mecha-nisms (241, 251, 287–291) or can tether to another tran-scription factor to modulate transrepression of targetedgenes via crosstalk with kinases, cofactors, or other pro-moter-bound transcription factors (Fig. 3) (211, 212,251). In the latter mechanism, GR can bind and modulateDNA-bound transcription factors such as AP-1, NF-�B,STAT5, octamer-binding transcription factor 1 (Oct-1),CREB, Smad3, Smad6, Ets2, T-box expressed in T cells (T-bet), and GATA3 (211, 212, 251, 292). Typically, the teth-ering GR repression of transcription factor activity is re-flected inareciprocal repressionofGRtransactivationbythevery same transcription factors (192, 251, 255, 277).

The transcription factors NF-�B and AP-1 are key tothe propagation of inflammation and are also targets ofGC-dependent repression of proinflammatory gene tran-scription (31). Moreover, chromatin immunoprecipita-tion analysis has shown that GR binds proximal to theNF-�B or AP-1 binding site in various promoters (254,293–295). The nuclear interaction of GR with the C-ter-minal activation domains of NF-�B p65 is pivotal to theGC-mediated repressive effect on NF-�B-regulated geneexpression (192, 255, 277). Mechanistically, GR does notneed to bind the target genes’ promoter DNA to infer in-hibition of NF-�B and AP-1 function, yet mutation anal-ysis revealed that a functional GR DBD is necessary forrepression of AP-1- and NF-�B-regulated genes (192, 193,256, 296). This is explained by the finding that the GRDBD is involved in AP-1 tethering interactions (297). Evenso, the GR LBD has been implicated in repression of NF-�B-regulated genes (298). GR-interacting protein 1(GRIP1) was originally identified as a corepressor for GRduring tethering to AP-1, explaining the LBD requirementfor repression (297). Nonetheless, GR typically does notinhibit binding of AP-1 or NF-�B to its respective responseelement within the endogenous promoters (253, 262, 293,295, 299). In addition, the LIM-domain protein thyroidreceptor-interacting protein 6 (Trip6) is suggested tofunction as an essential intermediary interaction part-ner in the association of GR, and AP-1 or NF-�B, be-cause knockdown of Trip6 or abolishing the interactionof Trip6 with GR abrogates GR-mediated transrepres-sion (295, 300).

Additionally, GCs can directly affect various histonemodifications that combine into a so-called “histonecode,” thus influencing chromatin accessibility and theassociated gene transcription. Effects of GCs on histonephosphorylation will be discussed below. Furthermore,GCs can inhibit cellular TNF-induced histone H4 K8 andK12 acetylation via reducing the HAT activity of CBP.Moreover, GC administration can increase the expression

of HDAC2, target HDAC2 to NF-�B CBP complexes, andtarget HDAC1 to the SP-A gene promoter (130, 294, 301).These histone-deacetylating events are associated with ahalted transcription of NF-�B-driven genes (130, 294,301). At the single promoter level, recent studies revealeda GC-mediated decline of histone H3 and H4 acetylationon the SP-A and IL-8 gene promoters, respectively, whichwas associated with a decreased transcription of thesegenes (294, 302). Lastly, GCs instigate the dimethylationof H3 K9 at the SP-A promoter (294). This histone mod-ification constitutes a transcription-repressive chromatinmark (303).

Because besides DNA-bound transcription factors andthe basal transcription machinery, the activated NF-�B-driven gene promoters recruit various cofactors in a gene-and cell type-specific manner, specific research has aimedto unravel possible GC effects on the composition andmodulation of this enhanceosome. First, it was proposedthat GR competes with NF-�B or AP-1 for a limitedamount of cofactors, such as the HAT CBP/p300 or SRC-1(304). However, overexpression of these cofactors or mu-tation of the coactivator-interacting domains of GR orNF-�B did not lead to a reversal of the marked inhibition(295, 305–308). Furthermore, GC administration did notaffect NF-�B p65 association with CBP (307). Lastly, thehypothesis of involvement of GR-interacting cofactorswas challenged via a mutation experiment. Although themutation of E755A in the GR C terminus, abolishing theinteraction of GR with LxxLL-containing cofactors, sig-nificantly decreased GR-mediated transactivation of aGRE-regulated reporter gene construct, this mutation didnot alter GR-mediated transrepression of Gal4-p65 activ-ity (309). Nevertheless, gradual overexpression of SRC-1or SRC-2 combined with the comodulator STAMP resultsin a lower EC50 value and a higher fold repression forGR-mediated inhibition of AP-1-mediated reporter geneactivity, in which EC50 is defined as the GC concentrationrequired for a half maximal response (310). Involvementof SRC-2 in GR-mediated repression of AP-1- or NF-�B-dependent gene expression was also confirmed for theendogenous genes collagenase-3 and IL-8 (297). How-ever, because most data on cofactor function are derivedfrom overexpression experiments, physiological rele-vance of nuclear cofactor modulation of GR-induced tran-srepression is yet to be determined. Lastly, not only couldcofactor complex assembly be modulated, but like NF-�Band GR, cofactors such as SRC-1, SRC-2, SRC-3, PGC-1,CBP, NCoR, and SMRT are themselves also subject toextensive posttranslational modulation such as phosphor-ylation, methylation, SUMOylation, ubiquitination, andacetylation. These modifications can affect cofactor-nu-clear receptor binding, activity, localization, and half-life

842 Beck et al. GR Crosstalk with Kinases and Phosphatases Endocrine Reviews, December 2009, 30(7):830–882

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

(311–313). However, the impact of GCs on these modu-lations or vice versa, how these modulations impact GC-mediated mechanisms, is currently not very well known.

II. Phosphoregulation of the GlucocorticoidReceptor

Activational control of GR can be imposed via a combi-natorial mechanism involving ligand accessibility, GRconcentration, subcellular localization, and also post-translational modifications of GR. The activity of GR isaffected by various modifications, among which are phos-phorylation, acetylation, nitrosylation, redox regulation,ubiquitination, and SUMOylation (Fig. 4) (314, 315). Inthis section, we will discuss the impact of phosphomodu-lation of GR on various aspects of GR functionality andsignaling.

A. GR phosphorylationPhosphorylation is the reversible co-

valent association of a phospho groupon a protein. Phosphorylation is regu-lated by the balance between phos-phorylating kinases and dephosphory-lating phosphatases. This modificationmay affect GR hormone and DNAbinding and subcellular localization,alter GR interactions and protein half-life, ultimately affecting transactivatingand transrepressing capabilities of GRs.These phosphorylations of the GR aremediated by specifically targeted kinases.

In murine GR (mGR), researchershave identified eight phosphorylationsites: S122, S150, S212, S220, S234,S315, S412, and T159, most of whichreside in the N-terminal domain (Fig.4A) (316, 317). The rat GR (rGR) phos-phorylation sites correspond to those ofmGR (Fig. 4A) (318). Conversely, inhGR, five Ser residues were character-ized as phosphorylation targets: S113,S141, S203, S211, and S226, and re-cently S404 (319–321). These residuescould be sequence matched to the mGRphosphorylation targets S122, S150,S212, S220, S234, and S412, respec-tively (Fig. 4A). Additionally, recentmass spectrometry analysis of hGR con-firmed the phosphorylation of hGRS226, but also suggested a cell cycle-dependent potential phosphorylation ofT8, S45, S134, S234, and S267 (322), of

which only the latter four S residues present a conservedcounterpart in the mGR and rGR. However, additionalevidence to confirm in vivo phosphorylation of these sitesis yet to be reported. Of note, all phosphorylation sites arelocated in the AF-1-containing N-terminal domain of GR.

Phosphorylation of hGR S203 and S211 can both bemediated by Cdk2/cyclin A kinase complexes, whereasCdk2/cyclin E targets only hGR S203 (323). In support,murine embryonic fibroblast cells devoid of the Cdk in-hibitor p27Kip1, which affects Cdk2 activity, show en-hanced GR phosphorylation at the corresponding mGRS212 and S220 and an increase in GR transactivation po-tential (324). In general, phosphorylation of hGR S203and S211 or their murine counterparts is associated withan enhanced transactivation of GRE-regulated promoters(323, 325–327). However, the interaction of Cdk5 and itsactivator protein p35 and p25 with the GR LBD could also

FIG. 4. Structure and posttranslational modifications of the GR. A, The reportedphosphomodulated sites for the hGR, mGR, and rGR are depicted in relation to the knownfunctional domains of this receptor. B, The reported posttranslational modifications for thehGR, exempt from the phosphorylated sites, are depicted in relation to the knownfunctional domains of this receptor. C, The reported phosphomodulated sites for humanNF-�B-p65 are depicted in relation to the known functional domains of this transcriptionfactor. NTD, N-Terminal domain; HR, hinge region; P, phosphorylation site; h, human; m,murine; r, rat; aa, amino acids; SUMO, SUMOylation site; Ub, ubiquitination site; Ac,acetylation site; Rel-HD, Rel-homology domain; TA, transactivation.

Endocrine Reviews, December 2009, 30(7):830–882 edrv.endojournals.org 843

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

mediate phosphorylation of hGR S203 and hGR S211,remarkably resulting in a decreased GR transcriptionalactivation of the MMTV and serum- and GC-induciblekinase (SGK) promoter via attenuated GR-cofactor inter-actions (328).

The p38 MAPK could possibly also contribute to thephosphorylation of hGR S211 from 20 h of GC exposureonwards, in lymphoid cells. However, in these cells pre-treatment with p38 MAPK inhibitor SB203580 onlyslightly diminished hGR S211 phosphorylation. Further-more, in these cells, overexpression of the hGR S211Amutant severely impaired the GR transactivation potential(329). Conversely, p38 MAPK-mediated phosphorylationof hGR at an undefined residue was also associated witha decreased GR ligand-binding affinity and a slightly re-duced GC-dependent repression of GM-CSF production(330). Furthermore, activation of p38 MAPK via IL-1�

administration or overexpression resulted in a diminishedGR transactivation function and GR-GRE binding (331–333). Possibly, the p38 MAPK-mediated inhibition of GRactivity is not mediated via a direct GR phosphorylationbut is instigated via phosphorylation of a GR LBD-inter-acting factor (332).

Additionally, the direct interaction of GR via the JNKinteraction motif with JNK MAPK accommodates JNK-mediated phosphorylation of rGR S246, corresponding tohGR S226. This phosphorylation inhibits GR transcrip-tional activation (334, 335), but currently, the possibleeffect of this phosphorylation on GR transrepressionmechanisms has not been researched. In correspondencewith the above findings, overexpression of the JNK up-stream activator MKK7 inhibits GR transactivation to-ward an MMTV reporter gene construct (332). Mecha-nistically, activated JNK-instigated export of GR to thecytoplasm via a leptomycin B-sensitive, CRM1-dependentmechanism could contribute to the inhibition of GR ac-tivity. In support, UV-induced JNK MAPK or overex-pression of JNK expedited GR nuclear export and wasassociated with an inhibition of GR-mediated transcrip-tion, whereas expression of a GR S226A mutant showsno UV-induced export or associated diminished GR ac-tivity (334).

Furthermore, a GSK3-mediated phosphorylation ofrGR at T171 was shown in vitro. Overexpression of GSK3inhibited GR transcriptional activation but does not affectGR-mediated repression of an AP-1-driven reporter geneconstruct. However, this rGR T171 corresponds to a hGRA150, and GSK3 overexpression in human cells thus doesnot affect GR activity and shows a species-specific differ-ence in GR phosphorylation. Nevertheless, when hGRA150 is mutated to T, the GSK3-mediated inhibition ofGR transactivation can be restored (318). In contrast,

hGR S404 was recently identified as a target of GSK3�.The nuclear phosphorylation of this residue would lead tonuclear export of GR, an enhanced down-regulation ofGR and attenuated transactivation of GRE-containingpromoters, and a hampered transrepression of NF-�B-reg-ulated gene promoter activities (321).

Lastly, a ligand-independent association of GR withPKA has been reported, and overexpression of PKA canenhance basal and GC-induced MMTV reporter gene ac-tivity and GR-GRE binding (336–338). Although phos-phorylation of GR by PKAc was suggested in vitro, thiswas never confirmed in vivo (339).

Although GRs display a low basal phosphorylation,these receptors get hyperphosphorylated upon the addi-tion of agonist (316). In quiescent cells, hGR S211 phos-phorylation count is lower than that of hGR S203. TheGC-induced alterations in GR phosphorylation seem todepend on the preexisting phosphorylation status of GRbecause mutation of hGR S203 to A mildly impedes hGRS211 phosphorylation, while slightly enhancing hGRS226 phosphorylation, suggesting a possibly ordered, se-quential phosphorylation of GR and an intersite depen-dency (320, 340).

Although GCs strongly enhance both S203 and S211phosphorylations, hGR S211 phosphorylation is pro-posedasahallmark for the transactivationpotential ofGRbecause the antagonist RU486 still allows for GR S203 butnot S211 phosphorylation (320). It should be noted, how-ever, that RU486 can also behave as an agonist in a con-text-dependent manner (341). GR transactivation func-tion is found to be at its peak when the relativephosphorylation of hGR S211 surpasses that of S226(327). Although overexpression of hGR S211A only di-minishes GR transcriptional activity (329), mutation anal-ysis of mGR showed that S to A mutations for the S212 andS220 residues strongly decreased GR transactivation of aminimal GRE-regulated reporter gene construct (325).Not all phosphorylations lead to an increase in trans-activation. The recently identified interaction of ERK8with GR� via the LIM domain-containing Hic5 inter-mediate was suggested to function as a dampener of GRtransactivation, the mechanism of which remains un-known (342).

The localization of phosphorylated GR can also differ.Ligand-induced S211 phosphorylated GR appears to bemainly nuclear, whereas S203 phosphorylated GR pref-erentially resides in the cytoplasm (320, 326). In contrast,in the absence of ligand, basally phosphorylated GRs atS203 or S211 were both found in the cytoplasm (320,326). Conversely, a nuclear phosphorylation of hGR S404by GSK3� and rGR S246 by JNK seems to expedite nu-cleocytoplasmic transport of these GRs (321, 334). These

844 Beck et al. GR Crosstalk with Kinases and Phosphatases Endocrine Reviews, December 2009, 30(7):830–882

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

findings demonstrate the impact of GR phosphorylation sta-tus on its subcellular localization. Interestingly, pharmaco-logical inhibition of tyrosine phosphorylation—usinggenistein or tyrphostin AG126—is reported to stimulate nu-clear export of GR (343). Currently, it is not clear whetherthis relocalization effect should be attributed to the directmodulation of an unknown GR Y residue phosphoryla-tion. However, GR phosphorylation is not critical to thereceptors’ nuclear import function because mGR, with allphosphorylatable sites mutated to A, still undergoes li-gand-dependent nuclear translocation (325). In addition,RU486 can also elicit GR translocation (262).

A recent study has provided a link between cell com-partment-specific phosphorylation of the GR, induced byacute or chronic stress, and GR-dependent transcriptionalactivity in rat central nervous system tissue. Only acuteisolation stress resulted in an increase in serum cortico-sterone levels. Under the condition of chronic stress, de-spite unaltered levels of nuclear GR, a significant tran-scriptional activity was still observed. In fact, it turned outthat GR-dependent gene regulation patterns in the centralnervous system, for GR, corticotropin-releasing factor(CRF), and brain-derived neurotrophic factor (BDNF)were similar compared with the acute stress model. Theseresults may suggest that the transcriptional activity of GRis not solely regulated by the levels of hormone. Rather, thetranscriptional activity of GR under chronic isolation wasproposed to result from an increased Cdk5 activation andphosphorylation of the nuclear GR at S232 and a de-creased JNK activity, which was reflected in a decreasedphosphorylation of nuclear GR at S246 (344).

GR phosphorylation can impact its half-life becausehGR phosphorylated at S203 displays a more rapid decaythan GR phosphorylated at S211, and various mutationsablating the phospho-acceptor sites of mGR loweredligand-dependent down-regulation of mGR (320, 325). Inaddition, association of GR with the tumor suppressorgene TSG101 (tumor susceptibility gene 101), which canbind ubiquitin groups and negatively affect ubiquitin-de-pendent proteasomal degradation, occurs preferentiallywith a nonphosphorylated GR. This interaction ofTSG101 and hypophosphorylated GR then leads to unli-ganded GR protein stabilization (345, 346).

The GRE-regulated tyrosine aminotransferase (TAT)and GILZ gene promoters appear to preferentially bindGR phosphorylated at S211 or S226 (326). Mechanisti-cally, hGR phosphorylation of S211 would alter its con-formation and thus accommodate interactions of GR withvitamin D receptor-interacting protein 150/mediatorcomplex subunit 14 (DRIP150/MED14) (346). Indeed, inGR S211A-expressing cells, the expression of MED14-independent genes was not impaired, whereas the tran-

scription of MED14-dependent genes was attenuated,suggesting a link between GR phosphorylation andMED14 involvement in a gene promoter-specific manner(327). Promoter selectivity was also previously shown bymutation analyses of mGR showing differential effects onvarious GRE-containing reporter gene constructs (325)and mutation analysis of hGR showing differential effectson various GC-activated and GC-repressed genes (321). Inthat respect, it is widely accepted that the different mod-ification statuses of GR could lead to a variable cofactorinteraction profile.

Thus, ligand-dependent phosphomodulation of GRcould affect GR ligand binding, gene promoter-selectiveGR transactivation, GR DNA binding, cofactor recruit-ment, subcellular localization, and half-life.

B. GR dephosphorylationBecause phosphorylation is a reversible mechanism,

GR function is also regulated by various phosphatases. Insupport, administration of a pharmacological inhibitor ofPP1, PP2A, and PP5 function augments GR phosphory-lation and blocks nuclear import of ligand-activated GR.These agents, however, allow export of GR to the cyto-plasm but prevent its subsequent return to the nucleus,thus abrogating recycling of GR (340, 347, 348).

The tetratricopeptide repeat domain-containing PP5,via binding to Hsp90 in the GR chaperone complex, formsan indirect binding partner for GR (349–351). Knock-down of PP5 resulted in increased GR binding to DNA andGR transcriptional activity but did not affect the ability ofthe synthetic GC dexamethasone to bind to GR (352,353). In contrast, a similar experiment was recently re-ported to reduce GC-induced transcription of three en-dogenous genes (IRF8, IGF binding protein 1, Ladinin),while leaving GILZ expression unaffected. Concomi-tantly, this PP5 knockdown raised phosphorylation ofhGR at S203, S211, and even more pronounced at the GRactivity-inhibiting site S226 (340). This gene-specific con-trol of PP5 over GR activity may be regulated by promot-ing the ligand-binding affinity of GR (354). The questionarises whether these findings can be reconciled with oneanother on the basis of the existence of different contextsand/or a mix of primary and secondary effects. Alterna-tively, PP5 could mediate GC-instigated nuclear trans-location of GR via the interaction between PP5 and themotor protein dynein (355). In this respect, PP5 is be-lieved to dephosphorylate recycled GR proteins return-ing from the nucleus, thus resetting GR in a ligand-inducible state (340, 356).

Very recently, estrogen has been described to inhibitGC induction of DUSP1 and GSK genes in breast cancercells, providing a plausible explanation for why GC trialsin breast cancer are not overtly successful (357). The

Endocrine Reviews, December 2009, 30(7):830–882 edrv.endojournals.org 845

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

mechanism involved a reduced ligand-induced GR phos-phorylation at S211, which is associated with the activeform of GR. Estrogen increased the expression of PP5,which mediates the dephosphorylation of GR at S211.After PP5 knockdown, the estrogen-promoted cell prolif-eration was significantly suppressed by GCs, providingproof for a crosstalk between estrogen-induced PP5 andGR action (357).

In short, GR phosphomodulation is thus a flexiblemechanism, integrating cellular stimuli and modulatingmultiple receptor functionalities. Currently, no GR-tar-geting phosphatases have been linked to specific GR res-idues. Moreover, considering the plethora of kinases in-volved in GR phosphorylation, the small number of GR-targeting phosphatases, characterized until now, seemstoo constrained. Future research in this direction couldthus possibly unveil additional information on theseprocesses.

C. Other posttranslational modifications of GRIn this paragraph, we will discuss how other posttrans-

lational modifications of the GR can affect its phosphor-ylation status. SUMOylation involves an E1-activating en-zyme, an E2 conjugation enzyme, and an E3 ligase. Thecovalent attachment of a small ubiquitin-related modifier(SUMO) motif can affect protein stability, subcellular lo-calization, and transcriptional activity (358). The follow-ing SUMO-target sites have been identified in GR: K703in the C-terminal LBD and K277 and K293 in the N-terminal synergy control motif (which is defined to inhibitsynergistic transcription conferred by GREs containingmultiple GR binding sites). These latter SUMOylationscan thus act as inhibitory elements controlling GR activitytoward multiple, but not single, GREs (Fig. 4B) (359–361). Interestingly, the JNK-mediated phosphorylation ofhGR S226 appears to facilitate subsequent GR SUMOy-lation at the N-terminal SUMOylation sites, and thus GRSUMOylation could be considered a phosphorylation-di-rected posttranslational GR modification (359). Overex-pression of SUMO-1 can destabilize GR protein (362),whereas a mutation of the potential SUMOylation sites ofGR or inhibition of the SUMOylation enhances GR trans-activation in a gene-selective manner, with a preference formultiple GRE-containing gene promoters (359–361).Moreover, overexpression of the GR targeting SUMO-2protein decreased GR-mediated expression of endogenousGRE-regulated genes (359). Although SUMOylation ofGR can occur in a ligand-independent manner and doesnot require an intact GR LBD and DBD dimerization mo-tif, the integrity of the latter domains and especially bind-ing of GR to DNA appears necessary to implementSUMO-dependent transcriptional inhibition (360, 362).Although binding of death domain-associated protein

(DAXX) to SUMOylated GR has been proposed to me-diate the described, inhibitory effects (363), a recent reportfinds no effect of death DAXX overexpression or knock-down on SUMOylated GR activity (360). Currently, it isunclear how GR SUMOylation controls its transcriptionalactivity, but SUMO-interacting proteins are most likely tobe considered.

Additionally, it is interesting to note that the phosphor-ylatable hGR S404, which is phosphorylated by GSK3�,mGR S412, and rGR S424 are comprised in a PEST deg-radation motif (316, 321). Ligand-activated mGR couldbe ubiquitinated at K426, leading to degradation of theGR protein (232, 364). The residue K419 is the counter-part in hGR (Fig. 4B). Blocking mGR downmodulationvia proteasomal inhibitors or via mutation of K426 en-hances GR-mediated transactivation and retards GR mo-bility in the nucleus (232, 364). Moreover, because GRbecomes hyperphosphorylated upon ligand binding, it ap-pears that this phosphoregulation is key to the onset ofthe ubiquitination-mediated proteasomal degradationof GR, since a mutant GR with all possible phospho-sites mutated to A does not undergo ligand-dependentdownmodulation (325).

Because SUMOylation and ubiquitination appear to beaffected by a differential phosphorylation of GR (325,359), it would prove interesting to research the possibilityof other phospho-directed posttranslational modifica-tions of GR. In this perspective, interesting research couldfocus on the acetylation of hGR at K494 and K495, whichis a prerequisite for GR association with NF-�B p65 andassists the GC-instigated repression of GM-CSF gene ex-pression (365).

III. Kinases Targeted by GlucocorticoidReceptor-Mediated Signaling

Because kinases such as MAPKs, MSKs, and Cdks are veryimportant in controlling the expression of inflammatorycytokines, recent research has moved its focus to the mod-ulation of these kinases as alternative antiinflammatorytools beside GCs. However, various aspects in kinase sig-naling and expression are already positively or negativelyregulated by GCs. Therefore, this section will focus on theeffects of activated GR on kinase signaling in the frame-work of inflammation.

A. Mitogen-activated protein kinases (MAPKs)A dysregulated activation of MAPKs was traced in nu-

merous inflammatory diseases such as rheumatoid arthri-tis, psoriasis, systemic lupus erythematosus, asthma, andinflammatory bowel disease (3, 366). Interestingly, acti-vated GR forms a complex regulatory loop with the acti-

846 Beck et al. GR Crosstalk with Kinases and Phosphatases Endocrine Reviews, December 2009, 30(7):830–882

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

vated MAPK signaling pathway. In short, whereas GR candirectly and indirectly inactivate MAPKs, these MAPKscan affect GR via phosphorylating the receptor (see Sec-tion II.A). Depending on the cell type used, GCs have beenshown to suppress p38, JNK, and/or ERK MAPK phos-phorylation. The GC-induced dephosphorylations of p38,JNK, and ERK MAPK have all been suggested to occur viaactions of the GC-induced DUSP1 phosphatase (see Sec-tion IV.A).

1. JNK MAPKThe GR crosstalk with the JNK signaling pathway,

leading to repression of its downstream targets c-Jun,ATF2, and Elk-1, appears to have multiple mechanisticlayers (367, 368). Besides the GC-induced DUSP1-medi-ated dephosphorylation of JNK MAPK (369–371), GRcan also directly interact with JNK and thus interfere withJNK activity without the necessity of de novo gene ex-pression (367, 372). Furthermore, the JNK-binding se-quence of GR seemed to be required for GC-induced JNKinhibition and nuclear import of JNK (372). Notably, theinteraction of GR with JNK can indirectly decrease AP-1activity via binding of inactive JNK, together with GR, tothe AP-1-bound elements of the c-jun gene promoter(372–374). However, binding of GR with the antagonistRU486 does not support a GR JNK interaction or GR andJNK recruitment to the c-jun gene promoter. Also, JNKsignaling can be blocked by activated GR via interruptingthe association between the activating MKK7 and JNK(372). Additionally, activation of the upstream MEKK1can be hampered by GCs via its negative interference withthe MEKK1 Hsp90 association, culminating in a disrup-tion of the JNK activation (375). Recently, Kim et al. (376)demonstrated that the GC-inducible SGK1 kinase inhibitsthe activation of SAPK (stress-activated protein kinase)/ERK kinase 1 (SEK1) (the upstream JNK kinase, alsocalled MKK4), thereby negatively regulating the JNK sig-naling pathway. SGK1 was further found to physicallyassociate with SEK1. Because SGK1 has been implicatedin the promotion of cell survival and the protectionagainst cellular stress, the described mechanism ex-plains how SGK1 negatively regulates stress-activated sig-naling, namely through the inhibition of SEK1 function-ality (376). Finally, a GC-mediated inhibition of JNKMAPK activity has been shown to block the translation ofTNF in murine monocytes (377).

2. ERK MAPKInhibition of ERK MAPK activation can occur via

alternative GC-dependent mechanisms. The interactionof Raf-1, a MAP3K in the ERK signaling pathway, withits chaperone Hsp90 is inhibited by GCs. As a result,

Raf-1 can no longer associate with Ras, the activationof which remained unaffected by GCs. Consequently,the impaired Raf-1 activation leads to a decrease in ERKactivation (375, 378).

GCs can elevate not only DUSP1 expression but also theexpression of GRE-regulated GILZ in various cell lines(270, 280, 379–389). Interestingly, binding of GILZ toRaf-1 inhibits its phosphorylation and thus represses thephosphorylation of Raf-1’s downstream targets MKK1/2and ultimately ERK1/2 MAPK (390, 391). Of note, Raf-1can also associate with liganded GR, together with 14-3-3(392). Additionally, GILZ can directly bind to NF-�B andAP-1, and as such represses NF-�B- and AP-1-directedexpression of proinflammatory genes (270, 384, 386, 393,394). Other GC-induced genes, namely downstream oftyrosine kinase 1 (Dok-1), SLAP and Dexras1, have alsobeen associated with a GC-mediated inhibition of ERKMAPK activation and inflammatory signaling (395–398).

3. p38 MAPKAlthough in many cell lines GC treatment leads to a

decrease in p38 MAPK phosphorylation and activationvia a DUSP1-dependent regulation (275, 399, 400), pro-longed GC exposure of lymphoid cells is actually knownto increase p38 MAPK phosphorylation levels (329). Cell-specific effects should thus be considered.

B. MAPK-activated protein kinases (MKs)Although p38 and ERK MAPKs have their own specific

transcription factor and other protein targets, theseMAPKs can continue the MAPK cascade by activating yetanother level of kinases, the MKs (Fig. 5). Based on se-quence homology, these MKs comprise MSKs, 90-kDaRSKs, MAPK-interacting kinases (MNKs), MK2, MK3,and as a final group MK5 (25). Only ERK MAPKs canactivate RSKs and MNK2, and only p38 MAPKs can ac-

FIG. 5. The layered MAPK signaling cascade. A schematicrepresentation of the signaling cascades initiated by mitogens andstressors that lead to the activation of the MAP2Ks/MKKs, MAPKs, andultimately the various MKs.

Endocrine Reviews, December 2009, 30(7):830–882 edrv.endojournals.org 847

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

tivate MK2, MK3, and MK5, whereas both ERK and p38MAPKs can activate MSK1, MSK2, and MNK1 (25).

1. MSK1The two MSK proteins, MSK1 and MSK2, consist of

two kinase domains bound together by a linker region.The N-terminal kinase domain of MSK accommodatessubstrate and autophosphorylation, whereas the MSK(CTD) comprises a bipartite nuclear localization sequence(401), a MAPK-docking domain, and an autoinhibitorysequence (402, 403) and can only mediate autophosphor-ylation of MSK (25). At the cellular level, MSK1 andMSK2 are predominantly nuclear kinases, most likely dueto their bipartite NLS (401, 404). MSKs are activated viap38� and/or ERK1/2 MAPK phosphorylation in responseto various stimuli (84, 401, 405–408). Activation of theseupstream kinases leads to the phosphorylation of MSK1T581 or MSK2 T568, which is considered primordial tothe activation of the MSK1 C-terminal kinase domain andthe subsequent full activation of MSK1 (402, 403, 409,410). MSKs are involved in affecting chromatin structure,transcription factor accessibility, and ultimately genetranscription via the phosphorylation of CREB S133,ATF1 S63, and NF-�B S276, leading to their activation(73, 83, 84, 405, 407), and the phosphorylation of H3S10, contributing to chromatin relaxation and thus facil-itating transcription of select gene promoters (411–414).Interestingly, phosphorylation of H3 S10 and H3 S28 byMSKs creates confirmed binding sites for 14-3-3 regula-tory proteins at c-jun, c-fos, and HDAC1 gene promoters(415–417). Furthermore, it was demonstrated that theconcomitant acetylation of H3 K9 and K14 stabilizes theassociation between phospho-acetylated H3 and 14-3-3(416–418). Lastly, the phosphorylation of H3 S10 andattraction of 14-3-3 correlates with dissociation of thetranscription-repressive heterochromatin protein (HP) 1�

and recruitment of RNA pol II (417, 419).Recent investigation into the effect of GCs on MSK1

signaling revealed that, whereas GCs cannot profoundlyaffect MSK1 phosphorylation or activity, these steroidscan effectively abolish MSK1 recruitment and thus H3 S10phosphorylation at specific inflammatory gene promoters(420), which nuances an earlier report that GCs could notimpact the overall H3 S10 phosphorylation in these cells(421). Moreover, the GC-induced lack of MSK1 occu-pancy at these TNF-activated inflammatory gene promot-ers contributes to a decline in NF-�B transactivation(420). Remarkably, GCs not only impede recruitment ofMSK1 at inflammatory gene promoters, but actuallydrive certain MSK1 proteins to the cytoplasm in a GR-and CRM1-dependent manner and furthermore sup-port a physical interaction between GC-activated GRand activated MSK1 (420). However, in-depth mecha-

nistic studies of how GCs affect MSK1 localization arestill warranted. Because the GR modulator compoundA (CpdA), which can transrepress NF-�B-driven geneexpression without displaying any transactivatingproperties (293, 422), can export a fraction of the nu-clear MSK1 to the cytoplasm similar to classical GCs,the active GR-instigated redistribution of MSK1 to thecytoplasm fits in the framework of a general cellularmechanism of GR-mediated transrepression of NF-�B-mediated transcription (420).

MSK1 has also been implicated in the cytoplasmicphosphorylation of the eukaryotic translation initiationfactor 4E-binding protein 1 (4E-BP1) at S64 and possiblyalso T36 (423). In unstimulated cells, 4E-BP1 can bind tothe eukaryotic translation initiation factor-4E (eIF-4E)complex, thus inhibiting functioning of the eIF-4E holo-complex in translation initiation. Phosphorylation of 4E-BP1 relieves the translational block by dissociating 4E-BP1 from the eIF-4F member eIF-4E at the mRNA cap(423). Interestingly, the administration of GCs blockedphosphorylation of the translational repressor 4E-BP1,thus allowing reassociation of 4E-BP1 and eIF-4E (424–428) (see Section V.A). The latter interaction preventsmRNA cap-dependent initiation of translation. Currently,crosstalk of GR with MSK1 in this mechanism has notbeen fully researched. As such, the possible involvementof GC-translocated cytoplasmic MSK1 in these eventsand the overall implications of these inhibitory effectsfor the antiinflammatory potential of GCs are not fullyunderstood.

2. MK2The GC-mediated MAPK inhibition results in an inhi-

bition of proinflammatory gene transcription via repress-ing the phosphorylation of multiple factors, but it alsoaffects proinflammatory protein production becauseMAPKs and the MK2 are involved in mRNA stability.However, only the select mRNAs that contain AREs atthe 3�-untranslated end, which are often cytokine and che-mokine transcripts, are affected by MAPK regulation(429). Mechanistically, TTP can bind and destabilize theseARE-containing transcripts and trigger their degradationvia binding of exonucleases (430, 431). However, TTPfunction can be inhibited via phosphorylation by the p38MAPK-activated kinase MK2, thus stabilizing the ARE-containing transcripts such as TNF mRNA (432–438).Notably, TTP knockout (KO) mice display phenotypicallyinflammatory arthritis due to an increased stability ofTNF mRNA and an enhanced production of TNF protein(439). Because GCs enhance the expression of TTP in var-ious cell lines (243, 269, 274, 440) and concomitantlyinhibit p38 MAPK and thus most likely MK2 activity,these steroids promote the destabilization and degrada-

848 Beck et al. GR Crosstalk with Kinases and Phosphatases Endocrine Reviews, December 2009, 30(7):830–882

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

tion of, among others, TNF, COX-2, GM-CSF, vascularendothelial growth factor (VEGF), MMP1, MMP3, IL-6,and IL-8 mRNAs (243, 268, 279, 283, 441–443). In sup-port, GCs repress TNF mRNA in lung epithelial cells anddecrease luciferase expression of a TNF 3� untranslatedregion reporter plasmid in an orientation-dependent man-ner. Small interfering RNAs to TTP significantly preventthis effect, and a cell line stably expressing a short-hairpinRNA to TTP showed that TTP is critical for GC-inducedinhibition of TNF mRNA (269, 274). Comparing knock-down approaches with TTP�/� murine embryonic fibro-blasts, differential effects of the lack of TTP on the mRNAturnover of different target genes were observed. Despitethis, a certain degree of loss of GC-induced repression ofCCL2, CCL7, CXCL5, and IL-6, which demonstratedTTP binding, was found to be reproducible. The observeddifferences may be due to different levels of TTP repressionand to the diverse phenotype of cells with a chronic vs. anacute factor depletion (269, 274). Taken together, thesestudies reflect a novel inductive antiinflammatory signal-ing pathway for GCs that acts via posttranscriptionalmechanisms (269, 274). Not only p38 MAPK, but alsoJNK and ERK MAPK signaling has been linked to theposttranscriptional stabilization of TNF, IL-2, and IL-3mRNA (444–446). Combined, the GC-induced mRNA de-stabilization together with the inhibited gene transcriptioncauses a rapid clearance of existing inflammatory signalingmolecules. Of note, the GC-stimulated destabilization of GRmRNAismediatedthroughthissamemechanism(447),pro-viding an adequate feedback response.

Currently, no link has been demonstrated between GRsignaling and the RSKs, MNKs, MK3, or MK5.

C. Cyclin-dependent kinases (Cdks)The proinflammatory stimulus TNF promotes the re-

cruitment of RNA pol II at the IL-8 and ICAM1 promoterand the subsequent phosphorylation of the CTD of RNApol II at S2 and S5. Conversely, GCs can interfere with theS2 phosphorylation of the RNA pol II CTD, while leavingits recruitment to the promoter unaffected (254). This S2phosphorylation was mediated by Cdk9 of the Cdk9/cyclinT1 P-TEFb complex and is absolutely necessary for(NF-�B-mediated) transcription (448, 449). In support,knockdown or pharmacological blockage of P-TEFb se-verely impaired TNF stimulation of IL-8 gene transcrip-tion. Interestingly, at the IL-8 promoter, GR competeswith P-TEFb for binding to NF-�B p65, thus inhibitingIL-8 gene transcription and the phosphorylation of therecruited RNA pol II S2 (253, 254). Recently, it was alsodiscovered that the association of P-TEFb with NF-�Brequired NF-�B p65 phosphorylation at S276 (81), thusintegrating the GC-instigated blockage to recruit MSK1

(420) and P-TEFb (253, 254) at the inflammatory IL-8gene promoter.

Additionally, GCs can cell-dependently repress thegene expression of Cdk4 and Cdk 6 and their associatingcyclin D3 (450) and induce expression of the Cdk inhibitorp21Cip1 (451, 452). Furthermore, GCs can inhibit Cdk2and Cdk4 activity, an event that is associated with theantiproliferative effects of GCs and decreased GRE-mediated transcription (453–455). Experiments withseliciclib, a Cdk inhibitor targeting Cdk2, Cdk7, andCdk9, in a mouse model for the chronic inflammatorycondition systemic lupus erythematosus revealed that thisCdk inhibitor can lower kidney inflammation and prolongsurvival. Upon combining seliciclib with GCs, however,the therapy elicited a greater beneficial effect than eithertherapy could alone (456). Additional research into therole of Cdks in the GC-mediated antiinflammatory mech-anisms is still required.

D. I�B kinase � (IKK�)The multifactorial kinase complex IKK is essential in

relaying proinflammatory stimuli to NF-�B activation.When activated, IKK� and IKK� can phosphorylate I�B,thus targeting this inhibitory molecule for degradation viathe ubiquitin-proteasome degradation pathway and pro-gramming the subsequent release of NF-�B to translocateto the nucleus (10). Furthermore, IKK� can promoteNF-�B DNA binding on specific gene promoters (457).

GCs can, in particular cell lines, up-regulate the expres-sion of I�B�. The replenishment of the depleted I�B� poolthus redirects the activated NF-�B to the cytoplasm andultimately counteracts the former actions of the NF-�B-activating IKK complex. A critical discussion of the role ofGC-induced I�B� in the GR-mediated antiinflammatorymechanism is presented in Refs. 211 and 243.

In addition to MSK1 (411, 414), IKK� has also beendescribed to mediate H3 S10 phosphorylation (458, 459).Constitutive shuttling of IKK� between cytoplasm andnucleus has been reported (460), but upon TNF induction,IKK� translocates into the nucleus (458, 459). Ablation ofIKK� halts H3 S10 phosphorylation and decreases proin-flammatory gene expression, without affecting I�B� deg-radation or NF-�B DNA binding (458, 459, 461, 462).Chromatin immunoprecipitation analysis of the NF-�B-regulated surfactant protein-A (SP-A) gene promoter inhuman fetal lung type II cells recently revealed that GCscan diminish H3 S10 phosphorylation in the SP-A pro-moter and can block the recruitment of the H3 S10 kinaseIKK� (294). In this respect, it may also be interesting toinvestigate the recruitment dynamics of the H3 S10-phos-phorylating kinase MSK1 at the SP-A gene promoter.

Endocrine Reviews, December 2009, 30(7):830–882 edrv.endojournals.org 849

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

E. TANK-binding kinase 1 (TBK1)In a recent report, TRAF family member-associated

NF-�B activator (TANK)-binding kinase 1 (TBK1) wasidentified as a new target for GCs (463). This kinase isactivated downstream of both TLR3 and TLR4 signalingand functions together with the inducible I�B kinase IKK�

as an intermediate in the activation of the transcriptionfactor IRF3 (464, 465). Upon activation, IRF3 subse-quently dimerizes, associates with CBP, and can thentranslocate into the nucleus where it binds onto specificIFN-stimulated response elements (ISREs), e.g., in theRANTES gene promoter (466, 467). Currently, no effectsof GCs on IKK� have been described. However, GCs candiminish LPS (TLR4)- and poly I-C (TLR3)-stimulatedS127 phosphorylation of TBK1 and thus attenuate TBK1activity. Possible involvement of DUSP10 and the GC-inducible DUSP4 (275), but not DUSP1, was suggested viaanalysis of the effects of their overexpression on ISRE-containing reporter gene constructs. However, in this re-spect, (double) knockdown studies are still lacking. More-over, possible effects of GCs on the SH2-containingtyrosine phosphatase SHP-2, which is known to target thekinase domain of TBK1 (468), have not yet been re-searched. The described GC-mediated inhibition of TBK1would thus contribute to the steroid-initiated inhibition ofIRF3-regulated gene promoter activity of a reporter geneconstruct in U373 cells (463). Correspondingly, the LPS orpoly(I-C)-induced expression of RANTES, of which thepromoter contains an ISRE, can also be inhibited by ad-dition of GCs. Moreover, the poly(I-C)/TLR3-induced ex-pression of other IRF3-dependent genes such as IFN-�,IFN-inducible protein of 10 kDa (IP-10), ISG15, andISG56 and ISRE-luc reporter gene constructs is also sub-ject to a GC-mediated inhibition, which would be insti-gated via the disruption of IRF3 and GRIP1/nuclear co-activator-2) complexes in murine macrophages (469).

However, these events appear to be in contrast withfindings of Ogawa et al. (299), who showed in macro-phages that TLR4- or TLR9-mediated promoter activa-tion of the chemokines IFN-inducible protein of 10 kDa(IP-10) or IFN-induced with tetratricopeptide repeats 1(Ifit1) or of an IRF3-dependent reporter gene constructcan be inhibited by GCs, whereas the TLR3-mediated ac-tivation of the same gene promoters is refractory to GCs(299). Mechanistically, Ifit1 depends on activation via thetranscription factor IRF3, irrespective of the stimulus.However, the interaction of DNA-bound IRF3 withNF-�B p65 only occurs upon TLR4/TLR9-mediated Ifit1gene promoter activation. Under these circumstanceswhere p65 acts as a cofactor, the specific sensitivity of thispromoter to GC-mediated repression in a TLR4- or TLR9-stimulated environment is being guaranteed. The basis of

this repression was attributed to a binding competitionbetween IRF3 and GR for the same site in NF-�B p65, inwhich GR has a greater affinity for NF-�B p65 than IRF3(299). Interestingly, TBK1 is also a TNF-activated kinasefor NF-�B p65 S536, which plays a role in the full tran-scriptional activation of this transcription factor (470).Further research is needed to resolve this apparent para-dox between the results of McCoy et al. (463), Reily et al.(469), and Ogawa et al. (299).

F. Other kinasesThe STAT transcription factors are phosphoproteins,

activated by receptor-associated Janus kinases (JAKs), af-ter cytokine receptor stimulation. Administration of GCscan inhibit the IL-2-induced phosphorylation, nucleartranslocation, and DNA binding of STAT5 via a decreasedexpression of JAK3 and IL-2 receptor �. As a consequence,IL-2, IL-4, IL-7, and IL-15 signaling and ultimately T cellproliferation was impeded (471). Furthermore, GCs caninhibit the IL-12-induced phosphorylation of STAT4 onposition Y693 in T-lymphocytes, however, without af-fecting the IL-12-instigated JAK phosphorylation (472,473). The inhibition of IL-2-induced STAT5 phosphor-ylation by GCs in T cells was associated with a GC-mediated down-regulation of JAK3, but not JAK1, pro-tein levels (471).

PKC is a family of S/T kinases, which are ubiquitouslyexpressed under the control of diverse stimuli and havebeen implicated in the pathogenesis of asthma and COPD(474). As such, PKCs can function as an intermediate inNF-�B-dependent gene transcription (475, 476). An over-all GC-instigated decline in PKC activity was recentlyreported for mesenteric arteries from rats (477). In par-ticular, PKC�, which has been associated with the NF-�B-mediated activation of IL-8 gene promoter activity (476),is an interacting partner for the GC-induced Dexras1. Thisassociation results in a decline in PKC� phosphorylationand activity (478).

Protein kinase B (PKB), also known as Akt, is a phos-phatidylinositol-3-kinase (PI3K)-stimulated kinase, in-volved in cell cycle regulation. The GC-induced PKB ac-tivation can also result in the phosphorylation of itsdownstream target kinase, GSK3�, causing inhibition ofits activity (479, 480).

Upon GC administration, PKB can be rapidly activatedvia a PI3K-dependent pathway, leading to the release ofvasorelaxing nitric oxide (479, 481–484). In support, GRcan interact with the p85� subunit of PI3K (484). Theserapid nongenomic effects typically need high doses of GCsto occur.

Depending on the cell type, however, GCs may differ-entially target particular kinases. An important mecha-nism underlying long-term GC-induced bone loss is the

850 Beck et al. GR Crosstalk with Kinases and Phosphatases Endocrine Reviews, December 2009, 30(7):830–882

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

impairment of osteoblast function and bone formation.The Wnt signaling pathway has been hypothesized to playa critical role in the osteoblast differentiation-related cellcycle. A key negative regulator in the Wnt signaling path-way is the serine/threonine kinase GSK3�. Consequently,GC-mediated activation of GSK3� in osteoblasts resultedin an inhibition of the Wnt signaling pathway. In osteo-blasts, GCs do not stimulate but further inhibit phosphor-ylation of PKB/Akt on Ser473 and thus disrupt input fromthe PI3K/Akt pathway into the Wnt signaling pathway atthe level of GSK3� (485). GSK3�, a known phosphory-lator of NF-�B S468 (68), has recently also been shown tomodulate GR (321), adding on complexity (see also Sec-tion II.A). In mammary epithelial tumor cells, GCs inducethe phosphorylation of GSK3�, ultimately leading to itsdegradation via the ubiquitin 26S proteasome (480). Thismechanism is believed to be involved in the control of tightjunction formation in mammary epithelial tumor cells.Alternatively, GSK3� can also be activated via serum- andGC-inducible kinase 1 (SGK1). A high level of expressionof SGK in breast cancer cells suggests that this kinase mayfunction to protect tumor cells from apoptosis and thuscan act as an oncogene. As its name suggests, the expres-sion of SGK1 can be induced by GCs (486–488) and re-markably, this kinase can up-regulate also the transcrip-tional activity of NF-�B (489). In that respect, SGK1 wasrecently identified as the mediating kinase for PI3K-initi-ated phosphorylation of IKK� T23, which in turn facili-tates the phosphorylation of IKK� S180 and the subse-quent activation of NF-�B. Concomitantly, SGK1 wasfound to phosphorylate p300 S1834 and enhance NF-�B-mediated transcription (490). However, it is cur-rently unknown how GCs could affect these processesand what their role could be in an inflammatory re-sponse. It may seem paradoxical that GCs are able totranscriptionally stimulate a kinase that subsequentlyserves to activate NF-�B, but it is important to realizethat the functionality and role of SGK1 may be regu-lated not only transcriptionally but also posttransla-tionally by its upstream activating kinases, includingPI3K and MAPK (490).

Additionally, GCs can affect various members of theSrc family of tyrosine kinases. The c-Src kinase can—uponactivation—contribute to I�B� phosphorylation and sub-sequent NF-�B translocation (491), whereas p56lck [lym-phocyte kinase (Lck)] and p59fyn (Fyn) kinases and src-like spleen tyrosine kinase (Syk) have been reported to playa role in T cell receptor-mediated signal transduction(492). In contrast, GCs can rapidly inhibit the activity ofthese Lck and Fyn kinases via a membrane-associated GR,thus contributing to the GC-mediated inhibition of T-cellreceptor signaling (493–495). Moreover, the GC-induc-

ible increase in SLAP expression would inhibit Ag-inducedphosphorylation of the Syk in mast cells (396). In contrast,GCs can augment the phosphorylation of Lck and thedownstream activation of p59Fyn, Zap70, Rac1, and Vavin resting but not in activated T cells (496).

Furthermore, GCs can stimulate the protein and activ-ity levels of the Rho-dependent protein kinase (ROCK) 2but down-regulate the specific activity of ROCK1 in ratepithelial cells (497). The activation of ROCK2 is espe-cially implicated in lung inflammation and the associatedmyosin light chain phosphorylation (498). In support,GCs can impede myosin light chain phosphorylation inairway hyperresponsive rats (499).

The molecular mechanisms that underlie nongenomicGC-induced immunosuppression remain to be preciselydefined.

IV. Phosphatases Targeted by GlucocorticoidReceptor-Mediated Signaling

Phosphatases can be classified according to their substratespecificity. PPs targeting Ser/Thr and/or Tyr can be re-garded as three groups: the tyrosine-specific phosphatases[protein tyrosine phosphatases (PTPs)], the Ser/Thr spe-cific phosphatases, and the DUSPs, targeting phospho-Tyr/Ser/Thr (500, 501). Currently, the most intensivelyresearched GC-regulated phosphatase in the context ofinflammation is DUSP1 (400, 502). However, this phos-phatase is not the only phosphatase that is subjected toGC-mediated modulation.

A. Dual specificity phosphatases (DUSPs)The DUSP family comprises a subfamily of 10 enzy-

matically active DUSPs that target MAPKs, dephospho-rylating T and Y residues, and therefore these phospha-tases are also referred to as MAPK phosphatases. TheMAPK family of protein kinases includes extracellularsignal-related kinases ERK1 and ERK2; p38 MAPKs �, �,� and �; and the c-Jun N-terminal kinases JNK1, JNK2,and JNK3 (Fig. 5) (25, 503). Structurally, these DUSPs aremarked by a C-terminal catalytic domain, and DUSP1,DUSP2, DUSP4, and DUSP5 are considered inducible nu-clear members of this DUSP subfamily (504).

The promoter of DUSP1 can be driven by ligand-acti-vated GR, and overexpression of the dimerization-defec-tive GR mutant (193) is incapable of mediating activationof the murine DUSP1 promoter by GCs (193, 505). How-ever, GCs enhance DUSP1 mRNA expression in murinemacrophages expressing only this GRdim mutant (369).These findings per se are not necessarily in conflict becausethe group of Pearce (506) showed that GRdim can func-tion even better than wild-type GR at promoters with mul-

Endocrine Reviews, December 2009, 30(7):830–882 edrv.endojournals.org 851

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

tiple GR binding sites. Also, in the case of steroid recep-tors, the DBD dimer interface plays an important rolebecause specific receptor mutations or noncanonical spac-ing of half-sites that disfavor this dimer interface lead toenhanced synergy (507). However, GRE sequences werenever identified in DUSP1 promoters, and GCs failed toenhance human DUSP1 expression in cells solely express-ing a rGR LS7 (P493R/A494S) mutant, which is incapableof transactivating classical GRE-regulated promoters(192, 508, 509). Alternatively, GC-mediated DUSP1 pro-moter activation was attributed to an ERK- and JNK-dependent mechanism involving AP-1 and CREB bindingonto the DUSP1 promoter (510). Additionally, GR bind-ing onto the DUSP promoter, 1.5 kb upstream of the tran-scription start site, was suggested to occur via a tetheringmechanism onto C/EBP�, and mutation of this C/EBPbinding site attenuated GC-inducible DUSP1 reportergene expression (509). In this context, it is interesting tonote that GCs can enhance the function of C/EBP� byinducing its phosphorylation (511). DUSP expression inresponse to GC signaling seems cell type-dependent. Be-sides DUSP1, also DUSP2, DUSP9, the PEST domain-enriched tyrosine phosphatase, and especially DUSP4 areup-regulated in response to GCs in bone marrow-derivedmast cells (275), and GCs can also enhance the expressionof DUSP4 in adipocytes (512), whereas the expression ofDUSP2, DUSP4, and LC-PTP seems to be inhibited by GCsin various pre-B leukemia cell lines (513). Although, theabove-mentioned DUSPs can also modulate ERK, p38,and JNK MAPK phosphorylation, thus impacting the in-flammatory process (504), their role in the GR-mediatedantiinflammatory mechanism is currently unaddressed.To date, no reports have been published on GC modula-tion of the related family members DUSP5, DUSP7,DUSP8, or DUSP16.

Besides GCs, mitogens and stressors can also elevate theexpression of the inducible DUSPs, such as DUSP1, func-tioning as a negative feedback loop in MAPK regulation(504, 514). Mechanistically, evidence from KO cellsshowed an involvement of the p38 and ERK MAPK-activated MSK1 and ATF1, and of the JNK1 MAPK in theLPS-mediated stimulation of DUSP1 gene transcription(514, 515). However, stimulation of GR could furtherenhance the already LPS-induced DUSP1 promoter in asynergistic manner in macrophages (516). Very recently, aDUSP1/GRIP1-dependent combinatorial mechanism hasbeen identified for repression by GR of LPS-inducedCOX-2, CXCL-5, and IL-6. Activated GR could inhibitTLR4-dependent COX-2 gene induction in macrophagesvia a mechanism that involves both DUSP1-mediatedAP-1 inhibition and GR-GRIP1 recruitment to the p65NF-�B DNA complex (517).

Interestingly, SOCS1, which appears to be up-regu-lated by GCs in hematopoietic cells and immune cell can-cers (267, 388, 513, 518, 519), can hamper the transcrip-tion of DUSP1, probably via the association of GR andSOCS1 (520). In the inflammatory process, this SOCSis also involved in NF-�B p65 proteolysis (98) and cannegatively interfere with TLR2- and TLR4-mediatedsignaling (521).

Cell-specific GC-elevated levels of DUSP1 can then tar-get the activational phosphorylations of the MAPKs ERK,JNK, and p38, thus inactivating them (400). JNK and p38MAPK are especially subject to DUSP1-mediated dephos-phorylation because DUSP1 KO mice-derived immunecells display prolonged p38 and JNK activation, whereasthe GC-induced ERK dephosphorylation was not altered(275, 369). However, because the GC-induced ERK de-phosphorylation depends on de novo protein productionin RBL-2H3 mast cells, cell-specific effects may be at playhere or possibly other GC-inducible DUSP family mem-bers, such as DUSP2, DUSP4, or DUSP9, may intervene(275, 505).

Subsequent to the inactivation of the MAPKs, the on-going proinflammatory signaling pathway is halted, andgene expression and mRNA translation of proinflamma-tory cytokines is attenuated (reviewed in Refs. 243, 400,and 522) (see Sections III.A and III.B.2). Therefore,DUSP1 KO mice show an impaired GC-mediated re-pression of various proinflammatory cytokines. Never-theless, GCs can still inhibit the expression of certaincytokines. The difference in GC-induced repression be-tween DUSP1�/� and DUSP1�/� cells depends on theselected cytokine but also cell type. Furthermore, inhibi-tion of de novo protein synthesis in bone marrow-derivedmast cells of DUSP1�/� mice can still partly reverse theantiinflammatory potential of GCs, suggesting the in-volvement of other GC-induced antiinflammatory pro-teins and of direct GC-dependent antiinflammatory mech-anisms (275, 369). Although DUSP1 KO mice are viable,they appear more susceptible to inflammation than theirwild-type counterparts (275, 369, 523–526). However,the inflamed cells of these DUSP1 KO mice remain sensi-tive to the antiinflammatory mechanisms of GCs (275). Inconclusion, GC-instigated up-regulation of DUSP1 is con-sidered an additional layer in the overall antiinflammatorymechanism of GR.

In addition to GR and the MAPKs, DUSP1 itself is sub-ject to posttranslational modifications. Although an ex-treme C-terminal phosphorylation of DUSP1 (S359,S364) by ERK MAPK instigates DUSP stabilization (527),prolonged ERK MAPK activity results in other C-terminalphosphorylations of DUSP1 (S296, S323). However, thelatter phosphorylations promote DUSP degradation (528,

852 Beck et al. GR Crosstalk with Kinases and Phosphatases Endocrine Reviews, December 2009, 30(7):830–882

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

529). Interestingly, GCs can inhibit the proteasomal deg-radation of DUSP1 (505). Moreover, LPS-mediatedDUSP1 acetylation (K57) via p300 seems to stimulate thebinding affinity of DUSP1 for p38 MAPK (530, 531). It iscurrently unknown whether and how these processes areaffected by GCs and connected to inflammation.

B. Other protein Y phosphatasesIn the PTP family, PTP receptor type C (PTPRC, also

known as LCA or CD45) is also regulated by GCs. As anessential regulator of T and B cell antigen receptor signal-ing, this membrane-associated tyrosine kinase is a markerfor inflammation and is not surprisingly down-regulatedby GCs in synovial membranes of arthritic rats and leu-kocytes of rheumatoid arthritis patients (532, 533).

Alternatively, GCs have been reported to induce theexpression of the SH2 domain-containing protein Y phos-phatase PTP1C (also known as SHP1), but not PTP1Dand PTP1 in pancreatic rat cells (534). This PTP1C hasbeen implicated in the regulation of T cell antigen re-ceptor signaling (535), and a deficiency in PTP1C func-tion was recently linked to an increase in TLR-initiatedinflammation (536).

C. Other phosphatasesThe protein S/T phosphatases PP1 and PP2A family

(PPP) are most abundantly present in the cell. The target-specific activity of PP1 and PP2A is encoded by five cata-lytical subunits and a vast abundance of interacting reg-ulatory subunits (537). Although effects of PP1, PP2A,and PP5 on GR have been reported (see Section II.B),currently only a few effects of GCs on these PPs have beenreported. PP1 and/or PP2A may be involved in the GC-mediated inhibition of translation via a GR-dependent de-phosphorylation of p70 S6K (70-kDa ribosomal proteinS6 kinase) (427, 428) (see Section V.A). However, thisinvolvement is yet to be confirmed via knockdown studies.

The S/T PP2A, which is composed of a catalytical sub-unit PP2Ac, a regulatory subunit PP2A-A (PR65), and avariable regulatory subunit PP2A-B (500), was suggestedto target ERK, p38, JNK MAPK phosphorylations, andNF-�B phosphorylation in various cells (106, 538–544).Furthermore, PP2A can impose a genome-wide dephos-phorylation of H3 S10 in Drosophila (545). In light of thefact that GCs can also target H3 S10 phosphorylation(294, 420), it could be interesting to investigate possibleadditional effects of GCs on PP2A.

PP2B (also known as calcineurin or PP3) is a Ca2�-dependent phosphatase comprising a catalytic A-subunitand a regulatory calmodulin-binding B-subunit (500),which has been implicated in the negative regulation ofMyD88 and Trif and thus TLR-mediated signaling (546).

Functionally, PP2B inhibitors are often used together withGCs as an immunosuppressant and antiinflammatorytherapeutic (547, 548). However, both pathways do notsignal independently as GCs can also modulate PP2B ac-tivity (549, 550). Because GCs can trigger the release ofCa2�, GCs could also rapidly and transiently enhance theactivity of PP2B in T cells (549). Alternatively, GCs canstimulate PP2B activity in a Ca2�-independent mecha-nism, involving a GC-induced association of Hsp90 andPP2B in pancreatic �-cells (550, 551). However, GCs donot seem to affect the protein levels of PP2B in various cells(551, 552).

V. Kinase/Phosphatase Regulation inGlucocorticoid-Mediated Side Effects

Although GCs remain the mainstay in the treatment ofacute and chronic inflammatory afflictions, long-term GCtherapy of chronic inflammatory disorders could lead to adetrimental side-effect profile and possibly also GC resis-tance (see Section VI). Overall, prolonged GC treatmentand high doses of potent GCs are the main risk factors inthe onset of GC therapy-associated side effects. It is likelythat both “desired effects” and “adverse effects” reflectGR actions, not only on transcription/translation directlybut also on phosphorylation.

The most prevalent GC therapy-associated side effect isweight gain, followed by skin bruising/thinning and insom-nia. From the patients’ view point, these skin problems to-gether with a disturbed fat distribution are psychologicallydistressful. Clinically, however, hyperglycemia leading todiabetes mellitus, the enhanced susceptibility to infec-tions, and osteoporosis are most worthy of attention andpose the greatest challenge and management issues in thesepatients. For instance, osteoporotic fractures occur in upto 50% of patients on long-term GC therapy, and theGC-induced patients’ sensitivity to infections could dou-ble their hospitalizations (553). Note that we call thesesecondary effects “side effects,” but it should be kept inmind that all of these effects are actually exaggerations ofnormal physiological GC actions.

Mechanistically, the onset and maintenance of GC-associated side effects is complex. Some GC therapy-as-sociated side effects are mainly attributed to GR transac-tivation mechanisms (e.g., diabetes, glaucoma, myopathy,hypertension), whereas others originate from GR transre-pression mechanisms [e.g., suppression of hypothalamic-pituitary-adrenocortical (HPA) axis]. However, the mech-anisms of some side effects are not completely known orcould be attributed to both GR transactivation and tran-srepression mechanisms (e.g., osteoporosis) (554). Fur-ther clinical and molecular research in this field is required

Endocrine Reviews, December 2009, 30(7):830–882 edrv.endojournals.org 853

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

to deepen our knowledge about the prevalence and mech-anistic basis of GC therapy-associated side effects.

The GC-enhanced susceptibility to infections stemsfrom the plethora of antiinflammatory and immunosup-pressive actions of the activated GR, as discussed in thesections above. Below, we will highlight the involvementof phosphatase and kinase signaling in the mechanisms ofGC-instigated side effects, with particular attention to theclinically most important ones.

A. Skeleton and muscle effectsLong-term GC therapy-induced osteoporosis origi-

nates from a dual mechanism: a decrease of osteoblastproliferation and activity and an increase in osteoclastactivity (555, 556).

Key to the activation status of osteoclastogenesis is thereceptor activator of NF-�B ligand (RANKL)/osteoprote-gerin (OPG) ratio. GCs can enhance the expression ofRANKL while decreasing the expression of a RANKL-signaling inhibitor, namely osteoprotegerin (555, 556). Inthe GC-mediated stimulatory mechanism of bone catab-olism, abrogated expression of the IL-1R-associatedpseudokinase IRAK-M is coupled to an enhanced differ-entiation of hematopoietic precursor cells into osteoclastsand an increase in osteoclast activity (557). In that respect,GCs can impede the expression of this IRAK-M and thusstimulate bone resorption (558).

In osteoblasts, ERK MAPK is essential to accommodateosteoblast proliferation. However, the GC-inducedDUSP1 expression in these cells can dephosphorylate ERKMAPKs, an event that was thus linked to a tyrosine phos-phatase-mediated inhibition of osteoblast proliferation.GCs directly affect osteoblasts by decreasing prolifera-tion, and therefore this mechanism may contribute to thephenomenon of steroid-induced osteoporosis (559–562).Additionally, the GC-induced activation of GSK3� in os-teoblasts was recently shown to contribute to their apo-ptosis, whereas GC-activated p38 MAPKs rather inhib-ited the apoptosis of these osteoblasts (563). Furthermore,the cell-detachment-induced apoptosis of osteocytes,which are bone-embedded cells that can regulate osteo-blast activity via gap junctions, is regulated via the rapidactivation of protein tyrosine kinase 2� (PTK2beta) (alsoknown as RAFTK or Pyk2) and thus its downstream targetJNK MAPK (564–566). Lastly, a GC-mediated inhibitionof osteocalcin transcription, via a competitive nGRE (290,567) contributes to the inhibition of bone formation.

Long-term GC therapy in juvenile diseased patientsalso causes serious growth retardation (553). The GC-induced apoptosis of chondrocytes, linked to this pro-cess, has been attributed to GC-mediated caspase acti-vation as well as GC-mediated inhibition of PKBphosphorylation (568).

When a high-dose GC treatment is sustained, pa-tients may experience muscle atrophy, leading to my-opathy. This GC-induced loss of muscle tissue is mainlyattributed to enhanced protein degradation (cataboliceffects) and diminished protein synthesis (antianaboliceffects) (554, 569).

One of the factors in the antianabolic effects of GCs,hampering the cellular protein synthesis mechanism, is theGC-mediated inhibition of 4E-BP1 phosphorylation,which facilitates the translation repressing association of4E-BP1 with eIF-4E (424–428). However, okadaic acidcould perturb the GC-mediated inhibition of p70 S6K and4E-BP1 phosphorylation, thus pointing to a possible rolefor PP1 and/or PP2A as the PPs used (427, 428). Alterna-tively, the GC-mediated decrease in phosphorylation of4E-BP1 in myoblasts was suggested to occur via negativeinterference with upstream mammalian target of rapamy-cin (mTOR) signaling (570).

Additionally, the PI3K/PKB/GSK� signaling path-way can play an important role in the regulation ofmuscle atrophy. Not surprisingly, GCs can inhibit PKBactivity in myoblasts, thus allowing GSK3� activation,which is in turn associated with suppressed protein syn-thesis (569, 571).

B. Hyperglycemia and diabetesPatients who are subject to long-term GC therapy have

a tendency to develop hyperglycemia, with the risk of di-abetes because GCs not only decrease the stimulated in-sulin production but also lower their response to circulat-ing insulin (554). The former decrease in insulin levels wasattributed to a nGRE element in the insulin promoter aswell as GC-mediated PP2B-dependent apoptosis of insu-lin-secreting cells (550, 551, 572). The latter insulin re-sistance could in part occur via a GC-induced rapid andtransient inhibition of insulin receptor kinase activity andseveral downstream intermediates such as p70 S6K, PKB,3-phosphoinositide-dependent protein kinase (PDK),Fyn, and GSK3 (494, 512). Additionally, GCs could ac-tually enhance JNK MAPK phosphorylation in adipocytes,which was associated with a perturbed insulin receptor-de-pendent signaling resulting in insulin resistance (494). Inthese same cells, an increase in DUSP1 expression was sug-gested to associate with a decrease in cellular glucose uptake(512). However, additional evidence for a role of DUSP1 inGC-induced hyperglycemia is currently lacking.

Lastly, GCs can stimulate the expression of various glu-coneogenetic enzymes in the liver, among which TAT,pyruvate dehydrogenase kinase 4, glucose-6-phosphatase(G6Pase), and pyruvate carboxykinase (PEPCK), culmi-nating in elevated glucose levels (573–579).

854 Beck et al. GR Crosstalk with Kinases and Phosphatases Endocrine Reviews, December 2009, 30(7):830–882

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

C. Other side effectsGC-induced hypertension is in part caused by a dys-

regulation of Na� homeostasis. As an important factorin this event, GCs can enhance the GRE-regulated tran-scription of the epithelial Na� channel (�ENaC) geneand cell-specifically augment ENaC and Na(�)/H(�)exchanger 3 (NHE3) activity in a SGK1-dependentmanner (554, 580 –588). The GC-activated rise inNHE3 activity would involve a direct SGK1-mediatedNHE3 phosphorylation (588).

In the ENaC modulating mechanism, a SGK1-medi-ated inhibiting phosphorylation of ALL1-fused gene fromchromosome 9 (Af9) of the histone-methylating Dot1a(disruptor of telomeric silencing alternative splice varianta)-Af9 complex can derepress the ENaC promoter, thusfacilitating transcription (589). Furthermore, an aldoste-rone-induced SGK1 can phosphorylate the E3 ubiquitinligase Nedd4-2, which is stabilized by 14-3-3, and can thusdiminish the degradation rate of ENaC by reducing theinteraction affinity between Nedd4-2 and ENaC (590–593). Alternatively, a specific and direct phosphorylationof ENaC by overexpressed SGK1 can directly enhanceENaC activity (594). Moreover, a steroid-mediated rise inGILZ expression and the concomitant inhibition of ERKMAPK signaling was recently linked to an increase ofENaC expression and activity (379, 380, 390). In addi-tion, Na� retention could possibly be enhanced by GCs viaa SGK1-dependent increase in Na�/glucose cotransporterSGLT1 activity via a similar Nedd4-2- based mechanism(595). However, evidence to the specific GR-dependenceof the above mechanisms is currently still lacking.

Furthermore, a GC-induced increase in gastric acid se-cretion, thus contributing to an enhanced risk of gastro-intestinal bleeding and peptic ulcer development, occurs ina PI3K/SGK1-dependent manner of which the precisemechanism remains currently unclear (261, 596).

Lastly, long-term GC therapy can in some cases lead toeye problems, such as cataract. A contributing mechanismin the GC-induced development of cataract is an increasedrate of gluconeogenesis, thus implicating PEPCK andG6Pase regulation (554). Recently, it was discovered thatGCs can enhance GILZ and DUSP1 expression in lensepithelial cells, coinciding with a decrease of Raf, ERK,p38 MAPK, and PKB phosphorylation (597). However,currently a role for this mechanism in cataractogenesis hasnot been firmly established.

VI. Kinase/Phosphatase Regulation inGlucocorticoid Resistance

Patients suffering from GC resistance are refractory to anantiinflammatory GC treatment. Although GC resistance

can be innate, it can also be acquired due to a prolongedGC treatment (201). Here we will shortly discuss differentmechanisms that possibly lie at the basis of GC resistancein inflammation, with special attention for GR phosphor-ylation in this phenomenon.

Foremost, the cause of innate GC resistance lies in amutation of the GR itself, leading to abnormal GR con-centrations, ligand-binding affinity, GR stability, GC-induced nuclear translocation, or GR-cofactor interac-tions (201). Currently, the research field concerning theseveral known GR haplotypes with regard to its specificeffects on GR phosphorylation and GR effects on kinaseand phosphatase regulation remains largely unexplored.

Furthermore, innate and acquired GC resistance wasassociated with a decrease in GR� protein levels via ho-mologous down-regulation, an increase in the protein lev-els of the dominant-negatively acting GR�, a decreasedGR ligand-binding affinity, or GR DNA binding (598–601). Moreover, the sensitivity to GCs could be alignedwith the degree of GC-induced GR nuclear translocation(602). Alternatively, GC resistance has been linked to anelevated expression of FKBP51, an element of the GRchaperone complex (603). At the level of cofactor regu-lation, GC resistance was associated with a reducedbrahma-related gene (Brg1) 1 expression and a PI3K-reg-ulated decrease in HDAC2 activity and expression (365,604–606). However, in some cases GC resistance wasattributed to a failure of GCs to acetylate histone H4 K5and thus transactivate gene expression, rather than to adisturbed GR transrepression mechanism (602). How-ever, the existence of a multidrug resistance membranetransporter, extrudingGCsoutof the cell and thus limitingtheir activity, has also been reported in GC resistance andcan function as a time-restricting control mechanism ofactivated GR (607).

In addition, GC-induced alterations in the GR phos-phorylation status have been associated with acquired GCresistance. As such, GC resistance has also been linked tothe inflammatory status of the diseased tissue via an en-hanced kinase activity of JNK, ERK, and p38 MAPKs; anincreased synthesis and/or activity of the transcription fac-tors NF-�B and AP-1; and increased cytokine production(330, 331, 608–615). In this respect, p38 MAPK-, JNKMAPK-, and GSK3-mediated phosphorylations of GRhave been linked to a decrease in GR transcriptional ac-tivity (321, 330–335) (see Section II.A). Interestingly, ap38 MAPK- and GSK3-mediated phosphorylation of GRcoincided with an attenuated repression of NF-�B-medi-ated gene expression by GCs (321, 330). Conversely, GCresistance was associated with the inability of GCs to de-activate JNK MAPK, which was reflected in elevatedphosphorylated c-Jun and c-fos gene expression in GC

Endocrine Reviews, December 2009, 30(7):830–882 edrv.endojournals.org 855

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

resistance and coincided with an observed decrease of GR-AP-1 interaction intensity in steroid-resistant asthma pa-tients, when compared with samples from steroid-respon-sive patients (600, 609, 616–618). Moreover, the GCresponse of steroid-resistant patient samples could be re-stored via the addition of MAPK inhibitors (330, 612,613, 619), and thus the MAPK-mediated inhibition of GRfunction appears to be a central player in GC resistance. Ofinterest, GC-resistant T cells, in which the steroid respon-siveness was restored via the addition of IFN, featuredelevated DUSP1 expression levels and concomitantlyan inhibition of p38 MAPK phosphorylation (619). Inpatients with severe asthma, which showed decreasedresponsiveness to GCs, an increased p38 MAPKphosphorylation corresponds to reduced induction ofDUSP1 expression (614). Taken together, GC unre-sponsiveness features a central role for MAPK dysregu-lation and probably also impaired DUSP1 induction.

VII. Future Perspectives in the Combatof Inflammation

In this section, we will discuss new and upcoming GR-based therapeutics and therapeutic strategies, with spe-cial attention to the effect of these ligands on GR phos-phorylation and GR-based kinase and phosphatasemodulations.

A. New glucocorticoid receptor ligandsA long-standing hypothesis driving steroid develop-

ment pharmacology strategies for the past decade was thatthe side effects of GCs are mainly attributed to GR tran-scriptional activation, whereas the antiinflammatory ef-fects of GCs are predominantly mediated via GR transre-pression mechanisms. This viewpoint was supported bythe finding that GRdim, a GR variant with a mutationhampering GR homodimerization, thereby compromisingDNA-binding at some GRE-driven genes, and thus pre-venting the stimulation of a classical GRE, still allows forGC-mediated repression of AP-1- and NF-�B-mediatedproinflammatory gene expression (193, 285, 620). Morerecently, it was demonstrated that this model is too rigidand that GRdim still leads to transactivation of a numberof genes, exemplified by phenyl-N-methyl-transferase.Thus, on a subset of GR-responsive promoters, GRs canform concerted multimers in a manner that is independentof the DBD-dimer interface (506). Indeed, GRdim micestill suffer from some side effects in response to GCs (276),and clearly not all GR-mediated side effects are solely con-trolled by classical GR transactivation mechanisms (621).Of course, a number of side effects are also attributed totransrepression mechanisms, e.g., HPA axis suppression.

Although it is recognized that a few antiinflammatorygenes are under stimulatory control by GCs, the transcrip-tion of these genes sometimes depends on an atypical GRtransactivation mechanism, and their role in the GC-induced antiinflammatory mechanism is not completelyclear (243). Although the model in which GR transrepres-sion mechanisms are associated with antiinflammatory ef-fects and GR transactivation mechanisms are associatedwith undesired effects clearly has to be put into a morenuanced perspective, it is still believed that a selective mod-ulation of GR, resulting in distinct GR mechanisms, couldcontribute to yielding a more beneficial side-effect profile inantiinflammatory therapies (268,285,622). It isparamount,however, to obtain further insights into the “context-depen-dency” phenomenon of GR-mediated regulation. Com-pounds that could activate select GR mechanisms and thusalter GR-mediated gene expression profiles are beingdesignated as dissociated compounds, selective GR ago-nists (SEGRAs) or modulators (SEGRMs) (621–624). Theterminology “SEGRMs” can have the most broad inter-pretation and can thus also include molecules that are GRantagonists, or even molecules that could activate partic-ular functionalities of GR without actually being a ligand.Various compounds—steroidal and nonsteroidal—capa-ble of activating GR transrepression without inducing GRtransactivation mechanism are called dissociated com-pounds (625). However, when the dissociation is not ab-solute, it is semantically more correct to refer to them asSEGRAs or SEGRMs, depending on a reported binding ofthe compound in the ligand-binding pocket (623).

Multiple SEGRAs and SEGRMs have been reported:RU24858, RU24782, and RU40066 (286, 626, 627);A276575 (628); AL-438 (629, 630); compound 25 (631);CpdA (293, 422, 632); ZK216348 (633, 634); LGD-5552(635, 636); and various other compounds (622, 637–643).When one of the above-mentioned compounds stimulatesGR, cellular effects can mechanistically differ from clas-sical GCs in affinity for the GR LBD, GR GRE DNA bind-ing, dimerization, cofactor promoter occupancy, histonemodifications at targeted promoters, but also GR phos-phorylation—all resulting in a divergent gene expressionprofile (293, 422, 622, 623, 629, 636).

Currently, GR phosphorylation-related research hasonly been conducted with CpdA. As a dissociative GRmodulator, CpdA [i.e., 2-(4-acetoxyphenyl)-2-chloro-N-methylethylammonium chloride] can efficiently repressthe transcription of inflammatory genes via diminishingNF-�B p65 DNA binding and the overall NF-�B p65transactivation potential and promoting nuclear export ofthe transcription-facilitating kinase MSK1 (293, 420).Conversely, CpdA cannot stimulate GRE-mediated genepromoter activation (293, 422). Interestingly, unlike clas-

856 Beck et al. GR Crosstalk with Kinases and Phosphatases Endocrine Reviews, December 2009, 30(7):830–882

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

sical GCs, CpdA does not induce DUSP1 human or mousereporter genes, nor can it elevate DUSP1 protein levels invarious cell lines (I. M. E. Beck, unpublished results). Ofspecial interest, CpdA instigates a differential phosphor-ylation profile of GR in comparison to classical GCs be-cause it did not induce a hyperphosphorylation of hGRS211 (293). Although the N-terminal domain of GR is notnecessary for CpdA- or classical GC-mediated trans-repression of NF-�B-regulated gene expression, the lack ofhGR S211 phosphorylation in CpdA-stimulated cellscould possibly explain the deficiency in GR recruitment toand transactivation of GRE-regulated promoters (293).Taking into account that this phosphorylation site is con-sidered crucial in the GR transactivation process (320,327), this leads to a question about whether all dissociatedGR modulators share this feature. GR phosphorylationhas also been implicated in ligand-mediated GR down-regulation (320, 325, 345, 346). In this respect, it is in-teresting to note that CpdA does not evoke homologousGR down-regulation in fibroblast-like synovial cells,which is reflected in a diminished antiinflammatory ther-apy resistance in long-term treatment protocols (632).Currently, the mechanism lying at the basis of this CpdA-mediated GR preservation and thus the possible involve-ment of differential GR phosphorylation is unknown.Taken together, additional research into the phosphory-lation profiles of GRs liganded with SEGRMS, could openup new and interesting perspectives in SEGRM research.

With respect to side effects, we can note that SEGRMsthat cannot induce the GRE-regulated PEPCK andG6Pase, such as CpdA, typically do not induce hypergly-cemia and hyperinsulinemia (293, 422). Also, AL-438 andZK216348 cannot instigate hyperglycemia (629, 633).However, further investigations comparing the side-effectprofile of classical GCs vs. the SEGRMs in a more elab-orate manner and research into the in vivo antiinflamma-tory potential of these SEGRMs remain a future challengein which special attention for SEGRM-induced effects onkinases and phosphatases could shed a new light on themechanisms of these GR modulators.

B. Combination therapiesConsidering the detrimental side-effect profile evoked

by a long-term GC therapy, research efforts additionallyfocused on possible strategies to lower therapeutic GCconcentrations while maintaining a similar antiinflamma-tory potential. Mechanistically, the idea of combining twoor more different antiinflammatory compounds would al-low lower dosages of each, while maintaining the antiin-flammatory profile and attenuating possible side effects ofthese therapeutics (644).

The highly efficient antiinflammatory combinationof long-acting �2 agonists (LABAs) with GCs is already

clinically used for the treatment of asthma and COPD(645, 646). Interestingly, LABAs can stimulate phos-phorylation of hGR at, e.g., S211 and stimulate its nu-clear translocation (647– 649). Moreover, combiningLABAs with GCs leads to a more pronounced inhibitionof ERK MAPK, JNK MAPK, and I�B� phosphoryla-tions (650) and increased expression levels of the anti-inflammatory DUSP1, whereas the GC-induced expres-sion of GILZ or TTP was not increased by the additionof LABAs (648, 651).

Additionally, the antiinflammatory effects of combin-ing Cdk, MAPK, or MSK inhibitors with GCs has beenassessed, all of which resulted in an enhanced antiinflam-matory treatment profile (330, 331, 456, 652). Althoughthese results appear promising, additional immunologicalresearch remains necessary to investigate the immunolog-ical and clinical aspects of these combination therapies.

Although it may seem temptingly simple to combineGCs with inhibitors targeted at inflammatory process-in-volved kinases to maximize the inflammatory effect, thisreview shows that because of the widespread crosstalk ofkinases and phosphatases with all levels of GR signaling,caution is in order. Therefore, research into topical appli-cation or intermittent therapy strategies for GCs and ki-nase inhibitors, surpassing general systemic side effects;and more specific ligands for GR, such as the SEGRMs;and more specific kinase inhibitors and phosphatase mod-ulators could broaden the array of available therapeuticagents. From this review, it is apparent that further re-search into the effects of kinases and phosphatases onto orby GC-mediated signaling could conduce the developmentof various new drug targets in the combat against inflam-mation and the control of GC-mediated side effects andGC resistance. In the treatment of chronic diseases, a moreconstrained therapeutic window is required than in acuteafflictions, allowing for a short-term treatment. Prefer-ence should go to the targeting of kinases or phosphatasesdownstream or at the end of a signaling cascade, insteadof targeting kinases at the top of the cascade. However,because of their actions in multiple pathways and be-cause of a known difficulty in specific targeting, thedecision to target kinases/phosphatases has to be ap-proached with extreme caution, and the risk to off-tar-get effects should be investigated and carefully weighed.As a prerequisite, the in-depth knowledge of the stereo-dynamic structure of the active enzymatic pocket of thekinase and phosphatase and pharmaceutical design willhave to be broadened in the future.

C. MicroRNA-specific modulation of GRThe GC response varies among individuals, as well as

within tissues from the same individual, depending on pre-receptor stage, ligand metabolism, GR polymorphisms,

Endocrine Reviews, December 2009, 30(7):830–882 edrv.endojournals.org 857

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

selective expression of GR subtypes (197, 199, 653–659),kinase/phosphatase-dependent posttranslational modifi-cations of the GR (as discussed in the current manuscript),the cofactor environment (531, 660, 661), and cell type-specific hormone receptor crosstalk with aryl hydrocar-bon receptor, MR, and peroxisome proliferator-activatedreceptor (PPAR) (662–664).

MicroRNAs (miRNAs) are single-stranded small andnoncoding RNA molecules that can regulate gene expres-sion. miRNAs turn off target gene expression within cellsby binding complementary regions in mRNA transcripts,consequently affecting mRNA translation. The possibilityexists that various kinases/phosphatases may impact onmiRNA-dependent regulation of GR function, involved inthe control of developmental, tissue-specific, and individ-ual-specific GC responses in human health and disease.Interestingly, miR-18 and miR-124a reduced GR-medi-ated transactivation in addition to decreasing GR proteinlevels and GC responsiveness in a cell type-specific fashion(665). Another target of miR-124a includes the mRNA ofa small CTD phosphatase, an antineuronal factor ex-pressed in nonneuronal tissues, playing a role during em-bryonic development (666).

Because GR ligands themselves can also changemiRNA transcription profiles, miRNA control of GR andinflammatory stress-sensitive kinases and phosphataseswill become a hot area of further research in GR stressbiology and immune functionality (152, 667–670).

D. Epigenetic approachesEnvironmental exposure to low concentrations of hor-

mones, air pollution, or toxicants can have heritable epi-genetic DNA methylation effects in animals and humansand persistently change inflammatory stress responseslater in life, or even in next generations (671–677). Inter-estingly, life events occurring during the prenatal, neona-tal, and perinatal period have programming effects on theHPA axis, brain neurotransmitter systems, and cognitiveabilities of the offspring and long-term effects on the be-havioral and neuroendocrine response to stressors (678–680). Of special note, maternal stress during gestation,including maternal GCs, may predispose to immune-re-lated pathologies later in life or in offspring (681, 682). Inmale and female rats exposed to prenatal restraint stress,these effects include a long-lasting hyperactivation of theHPA response associated with an altered circadian rhythmof corticosterone secretion (682). More particularly, themethylation status of the hGR (NR3C1) gene promoter innewborns is sensitive to prenatal maternal mood and maystimulate a potential epigenetic process that links antena-tal maternal mood and altered HPA stress reactivity dur-ing infancy (683). DNA methylation of CpG dinucleotidesis generally associated with epigenetic silencing of tran-

scription and is heritable through cell division. MultipleCpG sequences are rare in mammalian genomes, but fre-quently occur near the transcriptional start site of activegenes, with clusters of CpGs (CpG islands) being hypo-methylated.TheGRCpGislandcontains sevenalternativefirst exons, and their promoters show highly variableDNA methylation patterns between individuals. As such,methylation may orchestrate alternative first exon usageand silencing and may control tissue-specific expression ofGR. The observed heterogeneity may reflect epigeneticmechanisms for fine-tuning GR activity, programmed byearly life environment and events (684). Of particular in-terest, various epigenetic (co)factors, including HDACs,HATs, polycomb proteins, methyl CpG binding proteins[methyl CpG binding protein 2 (MeCP2), methyl CpGbinding domain protein (MBD)], and even DNA methyl-transferases themselves are regulated by kinase/phospha-tase complexes during inflammatory stress (661,685–696). Reciprocally, lack of MeCP2 in a mouse modelof Rett syndrome increases various GC-regulated genes,including the SGK1 (697). It remains to be further inves-tigated whether kinase/phosphatase-dependent changesin DNA methylation at the GR gene promoter or GR tar-get genes are cyclical or persistent (698, 699). Conse-quently, combination therapy of GCs, kinase/phospha-tase inhibitors, and epigenetic drugs may hold promises tomodulate GR activity in a time-dependent, cell-specific, orgene-specific way.

VIII. Conclusions

GR activity is regulated by kinases and phosphatases at mul-tiple levels. Alternatively, both kinases and phosphatases aresubjected to regulation by the GR. The phosphomodulationof GR can affect its ligand affinity, DNA binding, cofactorrecruitment, cellular localization, half-life and recycling,overall posttranslational modification profile, and ulti-mately transactivational and transrepressional properties.Because these receptors exist in a dynamic regulation, cul-minating in different receptor functions at different times,GRs cannot be considered as a uniform population, andresearchers should be beware of this complication. In turn,GC-activated GRs can alter the expression level, activity,half-life, localization, and specific interactions of variouskinases and the expression and activity of various phos-phatases. In conclusion, the regulation and implications ofGR phosphorylation and the effects of GR on variousphosphorylation and dephosphorylation events in theframework of inflammatory models forms an intricateweb. Because the current use of kinase inhibitors in bothresearch and therapy has markedly increased in the field ofinflammatory disease control, this web of regulations

858 Beck et al. GR Crosstalk with Kinases and Phosphatases Endocrine Reviews, December 2009, 30(7):830–882

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

should be taken into account when interpreting results ofkinase inhibitors with regard to the antiinflammatorymechanism of GCs.

Current scientific knowledge may lead to the identifi-cation of new kinase or phosphatase drug targets and thedesign of novel combination therapies that target GR aswell as its associated kinases/phosphatases to promote theantiinflammatory efficacy, reduce side effects, tackle GCresistance, and ultimately culminate in a better therapeuticprofile. Recently, kinases/phosphatases have also enteredthe field of epigenetic and miRNA-dependent regulation ofGR function and are, as such, also involved in the control ofdevelopmental, tissue-specific, and individual-specific diver-sity in the GC pathway in human health and disease. How-ever, further research, both at the basic and clinical levels,is still required to understand and deepen knowledgeabout the specificity and intertwinement of GR-, kinase-,and phosphatase-mediated events.

In conclusion, as the plethora of GR-regulated effectson and by kinase and phosphatase activities unfolds, thein-depth understanding of the antiinflammatory mecha-nism of GCs opens up new perspectives in manipulatingthis process therapeutically in a focused manner.

Acknowledgments

Address all correspondence and requests for reprints to: Karolien DeBosscher, Laboratory of Eukaryotic Gene Expression and Signal Trans-duction, K.L. Ledeganckstraat 35, B-9000 Gent, Belgium. E-mail:[email protected].

This work was financially supported by grants from InteruniversityAttraction Poles 6/18 (to G.H.), a Concerted Research Actions GOAfrom Ghent University (to G.H.), and U.S. National Institutes of HealthGrant CA020535 (to K.R.Y.). I.M.E.B., W.V.B., L.V., and K.D.B. are post-doctoral fellows of the Research Foundation-Flanders (FWO–Vlaanderen).

Disclosure Summary: I.M.E.B., W.V.B., L.V., G.H., and K.D.B. havenothing to declare. K.R.Y. is a consultant for Merck and Co. andSangamo Biosciences.

References

1. Hench PS, Kendall EC, Slocumb CH 1949 The effect of ahormone of the adrenal cortex (17-hydroxy-11-dehydro-corticosterone; compound E) and of pituitary adrenocor-ticotropic hormone on rheumatoid arthritis. Mayo ClinProc 24:181–197

2. Hollenberg SM, Weinberger C, Ong ES, Cerelli G, Oro A,Lebo R, Thompson EB, Rosenfeld MG, Evans RM 1985Primary structure and expression of a functional humanglucocorticoid receptor cDNA. Nature 318:635–641

3. Sweeney SE, Firestein GS 2006 Mitogen activated proteinkinase inhibitors: where are we now and where are wegoing? Ann Rheum Dis 65(Suppl 3):iii83–iii88

4. Pascual G, Glass CK 2006 Nuclear receptors versus in-

flammation: mechanisms of transrepression. Trends En-docrinol Metab 17:321–327

5. Polito AJ, Proud D 1998 Epithelia cells as regulatorsof airway inflammation. J Allergy Clin Immunol 102:714 –718

6. Ritchlin C 2000 Fibroblast biology. Effector signals re-leased by the synovial fibroblast in arthritis. Arthritis Res2:356–360

7. Standiford TJ, Kunkel SL, Basha MA, Chensue SW, Lynch3rd JP, Toews GB, Westwick J, Strieter RM 1990 Inter-leukin-8 gene expression by a pulmonary epithelial cell line.A model for cytokine networks in the lung. J Clin Invest86:1945–1953

8. Barnes PJ, Chung KF, Page CP 1998 Inflammatory medi-ators of asthma: an update. Pharmacol Rev 50:515–596

9. GhoshS,KarinM2002Missingpieces in theNF-�Bpuzzle.Cell 109 Suppl:S81–S96

10. Hayden MS, Ghosh S 2008 Shared principles in NF-�Bsignaling. Cell 132:344–362

11. O’Neill LA 2008 The interleukin-1 receptor/Toll-like re-ceptor superfamily: 10 years of progress. Immunol Rev226:10–18

12. Verstrepen L, Bekaert T, Chau TL, Tavernier J, Chariot A,Beyaert R 2008 TLR-4, IL-1R and TNF-R signaling toNF-�B: variations on a common theme. Cell Mol Life Sci65:2964–2978

13. Vandevoorde V, Haegeman G, Fiers W 1997 Induced ex-pression of trimerized intracellular domains of the humantumor necrosis factor (TNF) p55 receptor elicits TNF ef-fects. J Cell Biol 137:1627–1638

14. Hsu H, Xiong J, Goeddel DV 1995 The TNF receptor1-associated protein TRADD signals cell death andNF-�B activation. Cell 81:495–504

15. Gaur U, Aggarwal BB 2003 Regulation of proliferation,survival and apoptosis by members of the TNF superfam-ily. Biochem Pharmacol 66:1403–1408

16. Chen G, Goeddel DV 2002 TNF-R1 signaling: a beautifulpathway. Science 296:1634–1635

17. Hsu H, Huang J, Shu HB, Baichwal V, Goeddel DV 1996TNF-dependent recruitment of the protein kinase RIPto the TNF receptor-1 signaling complex. Immunity 4:387–396

18. Hsu H, Shu HB, Pan MG, Goeddel DV 1996 TRADD-TRAF2 and TRADD-FADD interactions define two dis-tinct TNF receptor 1 signal transduction pathways. Cell84:299–308

19. Ting AT, Pimentel-Muinos FX, Seed B 1996 RIP mediatestumor necrosis factor receptor 1 activation of NF-�B but notFas/APO-1-initiated apoptosis. EMBO J 15:6189–6196

20. Devin A, Lin Y, Yamaoka S, Li Z, Karin M, Liu Zg 2001The � and � subunits of I�B kinase (IKK) mediate TRAF2-dependent IKK recruitment to tumor necrosis factor (TNF)receptor 1 in response to TNF. Mol Cell Biol 21:3986–3994

21. Malinin NL, Boldin MP, Kovalenko AV, Wallach D 1997MAP3K-related kinase involved in NF-�B induction byTNF, CD95 and IL-1. Nature 385:540–544

22. Yang J, Lin Y, Guo Z, Cheng J, Huang J, Deng L, Liao W,Chen Z, Liu Z, Su B 2001 The essential role of MEKK3 inTNF-induced NF-�B activation. Nat Immunol 2:620–624

23. Beyaert R, Van Loo G, Heyninck K, Vandenabeele P 2002

Endocrine Reviews, December 2009, 30(7):830–882 edrv.endojournals.org 859

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

Signaling to gene activation and cell death by tumor ne-crosis factor receptors and Fas. Int Rev Cytol 214:225–272

24. Boone E, Vandevoorde V, De Wilde G, Haegeman G 1998Activation of p42/p44 mitogen-activated protein kinases(MAPK) and p38 MAPK by tumor necrosis factor (TNF) ismediated through the death domain of the 55-kDa TNFreceptor. FEBS Lett 441:275–280

25. Roux PP, Blenis J 2004 ERK and p38 MAPK-activatedprotein kinases: a family of protein kinases with diversebiological functions. Microbiol Mol Biol Rev 68:320–344

26. Vanden Berghe W, Plaisance S, Boone E, De Bosscher K,Schmitz ML, Fiers W, Haegeman G 1998 p38 and extra-cellular signal-regulated kinase mitogen-activated proteinkinase pathways are required for nuclear factor-�B p65transactivation mediated by tumor necrosis factor. J BiolChem 273:3285–3290

27. Baud V, Karin M 2001 Signal transduction by tumor ne-crosis factor and its relatives. Trends Cell Biol 11:372–377

28. Kawai T, Akira S 2007 Signaling to NF-�B by Toll-likereceptors. Trends Mol Med 13:460–469

29. Kawai T, Akira S 2008 Toll-like receptor and RIG-I-likereceptor signaling. Ann NY Acad Sci 1143:1–20

30. Foster SL, Medzhitov R 2009 Gene-specific control of theTLR-induced inflammatory response. Clin Immunol 130:7–15

31. Barnes PJ 2006 Transcription factors in airway diseases.Lab Invest 86:867–872

32. Bonizzi G, Karin M 2004 The two NF-�B activation path-ways and their role in innate and adaptive immunity.Trends Immunol 25:280–288

33. D’Acquisto F, Ianaro A 2006 From willow bark to pep-tides: the ever widening spectrum of NF-�B inhibitors.Curr Opin Pharmacol 6:387–392

34. Kumar A, Takada Y, Boriek AM, Aggarwal BB 2004 Nu-clear factor-�B: its role in health and disease. J Mol Med82:434–448

35. Liou HC 2002 Regulation of the immune system by NF-�Band I�B. J Biochem Mol Biol 35:537–546

36. Smahi A, Courtois G, Rabia SH, Doffinger R, Bodemer C,Munnich A, Casanova JL, Israel A 2002 The NF-�B sig-nalling pathway in human diseases: from incontinentia pig-menti to ectodermal dysplasias and immune-deficiencysyndromes. Hum Mol Genet 11:2371–2375

37. Bours V, Franzoso G, Azarenko V, Park S, Kanno T,Brown K, Siebenlist U 1993 The oncoprotein Bcl-3 directlytransactivates through �B motifs via association withDNA-binding p50B homodimers. Cell 72:729–739

38. Leeman JR, Gilmore TD 2008 Alternative splicing in theNF-�B signaling pathway. Gene 423:97–107

39. Weih F, Carrasco D, Durham SK, Barton DS, Rizzo CA,Ryseck RP, Lira SA, Bravo R 1995 Multiorgan inflamma-tion and hematopoietic abnormalities in mice with a tar-geted disruption of RelB, a member of the NF-�B/Rel fam-ily. Cell 80:331–340

40. Hoffmann A, Leung TH, Baltimore D 2003 Genetic anal-ysis of NF-�B/Rel transcription factors defines functionalspecificities. EMBO J 22:5530–5539

41. Leung TH, Hoffmann A, Baltimore D 2004 One nucleo-tide in a �B site can determine cofactor specificity forNF-�B dimers. Cell 118:453–464

42. Chen-Park FE, Huang DB, Noro B, Thanos D, Ghosh G2002 The �B DNA sequence from the HIV long terminal

repeat functions as an allosteric regulator of HIV transcrip-tion. J Biol Chem 277:24701–24708

43. Natoli G, Saccani S, Bosisio D, Marazzi I 2005 Interactionsof NF-�B with chromatin: the art of being at the right placeat the right time. Nat Immunol 6:439–445

44. Phelps CB, Sengchanthalangsy LL, Malek S, Ghosh G2000 Mechanism of �B DNA binding by Rel/NF-�Bdimers. J Biol Chem 275:24392–24399

45. Hayden MS, Ghosh S 2004 Signaling to NF-�B. Genes Dev18:2195–2224

46. Ben-NeriahY2002Regulatory functionsofubiquitinationin the immune system. Nat Immunol 3:20–26

47. Brown K, Gerstberger S, Carlson L, Franzoso G, SiebenlistU 1995 Control of I�B-� proteolysis by site-specific, signal-induced phosphorylation. Science 267:1485–1488

48. Hacker H, Karin M 2006 Regulation and function of IKKand IKK-related kinases. Sci STKE 2006:re13

49. Chen G, Cao P, Goeddel DV 2002 TNF-induced recruit-ment and activation of the IKK complex require Cdc37 andHsp90. Mol Cell 9:401–410

50. Hinz M, Broemer M, Arslan SC, Otto A, Mueller EC, DettmerR, Scheidereit C 2007 Signal responsiveness of I�B kinasesis determined by Cdc37-assisted transient interaction withHsp90. J Biol Chem 282:32311–32319

51. Ducut Sigala JL, Bottero V, Young DB, Shevchenko A,Mercurio F, Verma IM 2004 Activation of transcriptionfactor NF-�B requires ELKS, an I�B kinase regulatory sub-unit. Science 304:1963–1967

52. Vanden Berghe T, Kalai M, van Loo G, Declercq W,Vandenabeele P 2003 Disruption of HSP90 function re-verts tumor necrosis factor-induced necrosis to apoptosis.J Biol Chem 278:5622–5629

53. Hu Y, Baud V, Delhase M, Zhang P, Deerinck T, EllismanM, Johnson R, Karin M 1999 Abnormal morphogenesisbut intact IKK activation in mice lacking the IKK� subunitof I�B kinase. Science 284:316–320

54. Xiao G, Rabson AB, Young W, Qing G, Qu Z 2006 Al-ternative pathways of NF-�B activation: a double-edgedsword in health and disease. Cytokine Growth Factor Rev17:281–293

55. Derudder E, Dejardin E, Pritchard LL, Green DR, KornerM, Baud V 2003 RelB/p50 dimers are differentially regu-lated by tumor necrosis factor-� and lymphotoxin-� re-ceptor activation: critical roles for p100. J Biol Chem 278:23278–23284

56. Solan NJ, Miyoshi H, Carmona EM, Bren GD, Paya CV2002 RelB cellular regulation and transcriptional activityare regulated by p100. J Biol Chem 277:1405–1418

57. Amir RE, Haecker H, Karin M, Ciechanover A 2004Mechanism of processing of the NF-�B2 p100 precursor:identification of the specific polyubiquitin chain-anchor-ing lysine residue and analysis of the role of NEDD8-mod-ification on the SCF(�-TrCP) ubiquitin ligase. Oncogene23:2540–2547

58. Fong A, Zhang M, Neely J, Sun SC 2002 S9, a 19 S pro-teasome subunit interacting with ubiquitinated NF-�B2/p100. J Biol Chem 277:40697–40702

59. Liang C, Zhang M, Sun SC 2006 �-TrCP binding and pro-cessing of NF-�B2/p100 involve its phosphorylation atserines 866 and 870. Cell Signal 18:1309–1317

60. Senftleben U, Cao Y, Xiao G, Greten FR, Krahn G, BonizziG, Chen Y, Hu Y, Fong A, Sun SC, Karin M 2001 Acti-

860 Beck et al. GR Crosstalk with Kinases and Phosphatases Endocrine Reviews, December 2009, 30(7):830–882

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

vation by IKK� of a second, evolutionary conserved,NF-�B signaling pathway. Science 293:1495–1499

61. Xiao G, Fong A, Sun SC 2004 Induction of p100 processingby NF-�B-inducing kinase involves docking I�B kinase �(IKK�) to p100 and IKK�-mediated phosphorylation.J Biol Chem 279:30099–30105

62. Chen LF, Williams SA, Mu Y, Nakano H, Duerr JM,Buckbinder L, Greene WC 2005 NF-�B RelA phosphory-lation regulates RelA acetylation. Mol Cell Biol 25:7966–7975

63. Perkins ND 2006 Post-translational modifications regu-lating the activity and function of the nuclear factor �Bpathway. Oncogene 25:6717–6730

64. Chen LF, Mu Y, Greene WC 2002 Acetylation of RelA atdiscrete sites regulates distinct nuclear functions of NF-�B.EMBO J 21:6539–6548

65. Kiernan R, Bres V, Ng RW, Coudart MP, El Messaoudi S,Sardet C, Jin DY, Emiliani S, Benkirane M 2003 Post-activation turn-off of NF-�B-dependent transcription isregulated by acetylation of p65. J Biol Chem 278:2758–2766

66. Mabb AM, Miyamoto S 2007 SUMO and NF-�B ties. CellMol Life Sci 64:1979–1996

67. Neumann M, Naumann M 2007 Beyond I�Bs: alternativeregulation of NF-�B activity. FASEB J 21:2642–2654

68. Buss H, Dorrie A, Schmitz ML, Frank R, Livingstone M,Resch K, Kracht M 2004 Phosphorylation of serine 468 byGSK-3� negatively regulates basal p65 NF-�B activity.J Biol Chem 279:49571–49574

69. Chen Lf, Fischle W, Verdin E, Greene WC 2001 Durationof nuclear NF-�B action regulated by reversible acetyla-tion. Science 293:1653–1657

70. Mattioli I, Sebald A, Bucher C, Charles RP, Nakano H, DoiT, Kracht M, Schmitz ML 2004 Transient and selectiveNF-�B p65 serine 536 phosphorylation induced by T cellcostimulation is mediated by I�B kinase � and controls thekinetics of p65 nuclear import. J Immunol 172:6336–6344

71. Bohuslav J, Chen LF, Kwon H, Mu Y, Greene WC 2004p53 induces NF-�B activation by an I�B kinase-indepen-dent mechanism involving phosphorylation of p65 by ri-bosomal S6 kinase 1. J Biol Chem 279:26115–26125

72. Chen ZJ 2005 Ubiquitin signalling in the NF-�B pathway.Nat Cell Biol 7:758–765

73. Vermeulen L, De Wilde G, Van Damme P, Vanden BergheW, Haegeman G 2003 Transcriptional activation of theNF-�B p65 subunit by mitogen- and stress-activated pro-tein kinase-1 (MSK1). EMBO J 22:1313–1324

74. Zhong H, Voll RE, Ghosh S 1998 Phosphorylation ofNF-�B p65 by PKA stimulates transcriptional activity bypromoting a novel bivalent interaction with the coactivatorCBP/p300. Mol Cell 1:661–671

75. Duran A, Diaz-Meco MT, Moscat J 2003 Essential role ofRelA Ser311 phosphorylation by �PKC in NF-�B tran-scriptional activation. EMBO J 22:3910–3918

76. Jamaluddin M, Wang S, Boldogh I, Tian B, Brasier AR2007 TNF-�-induced NF-�B/RelA Ser(276) phosphoryla-tion and enhanceosome formation is mediated by an ROS-dependent PKAc pathway. Cell Signal 19:1419–1433

77. Leitges M, Sanz L, Martin P, Duran A, Braun U, García JF,Camacho F, Diaz-Meco MT, Rennert PD, Moscat J 2001

Targeted disruption of the �PKC gene results in the im-pairment of the NF-�B pathway. Mol Cell 8:771–780

78. Wang D, Baldwin Jr AS 1998 Activation of nuclear factor-�B-dependent transcription by tumor necrosis factor-� ismediated through phosphorylation of RelA/p65 on serine529. J Biol Chem 273:29411–29416

79. Schwabe RF, Sakurai H 2005 IKK� phosphorylates p65 atS468 in transactivaton domain 2. FASEB J 19:1758–1760

80. Okazaki T, Sakon S, Sasazuki T, Sakurai H, Doi T, YagitaH, Okumura K, Nakano H 2003 Phosphorylation of serine276 is essential for p65 NF-�B subunit-dependent cellularresponses. Biochem Biophys Res Commun 300:807–812

81. Nowak DE, Tian B, Jamaluddin M, Boldogh I, VergaraLA, Choudhary S, Brasier AR 2008 RelA Ser276 phos-phorylation is required for activation of a subset of NF-�B-dependent genes by recruiting cyclin-dependent kinase9/cyclin T1 complexes. Mol Cell Biol 28:3623–3638

82. Zhong H, SuYang H, Erdjument-Bromage H, Tempst P,Ghosh S 1997 The transcriptional activity of NF-�B is reg-ulated by the I�B-associated PKAc subunit through a cyclicAMP-independent mechanism. Cell 89:413–424

83. Reber L, Vermeulen L, Haegeman G, Frossard N 2009Ser276 phosphorylation of NF-kB p65 by MSK1 controlsSCF expression in inflammation. PLoS ONE 4:e4393

84. Vermeulen L, Berghe WV, Beck IM, De Bosscher K,Haegeman G 2009 The versatile role of MSKs in transcrip-tional regulation. Trends Biochem Sci 34:311–318

85. Zhong H, May MJ, Jimi E, Ghosh S 2002 The phosphor-ylation status of nuclear NF-�B determines its associationwith CBP/p300 or HDAC-1. Mol Cell 9:625–636

86. Dong J, Jimi E, Zhong H, Hayden MS, Ghosh S 2008Repression of gene expression by unphosphorylatedNF-�B p65 through epigenetic mechanisms. Genes Dev 22:1159–1173

87. Anrather J, Racchumi G, Iadecola C 2005 Cis-acting,element-specific transcriptional activity of differentiallyphosphorylated nuclear factor-�B. J Biol Chem 280:244 –252

88. Foster SL, Hargreaves DC, Medzhitov R 2007 Gene-spe-cific control of inflammation by TLR-induced chromatinmodifications. Nature 447:972–978

89. Guan H, Hou S, Ricciardi RP 2005 DNA binding of re-pressor nuclear factor-�B p50/p50 depends on phosphor-ylation of Ser337 by the protein kinase A catalytic subunit.J Biol Chem 280:9957–9962

90. Sakurai H, Miyoshi H, Toriumi W, Sugita T 1999 Func-tional interactions of transforming growth factor �-acti-vated kinase 1 with I�B kinases to stimulate NF-�B acti-vation. J Biol Chem 274:10641–10648

91. Sakurai H, Chiba H, Miyoshi H, Sugita T, Toriumi W1999 I�B kinases phosphorylate NF-�B p65 subunit onserine 536 in the transactivation domain. J Biol Chem 274:30353–30356

92. Fujita F, Taniguchi Y, Kato T, Narita Y, Furuya A, OgawaT, Sakurai H, Joh T, Itoh M, Delhase M, Karin M,Nakanishi M 2003 Identification of NAP1, a regulatorysubunit of I�B kinase-related kinases that potentiatesNF-�B signaling. Mol Cell Biol 23:7780–7793

93. Sasaki CY, Barberi TJ, Ghosh P, Longo DL 2005 Phos-phorylation of RelA/p65 on serine 536 defines an I�B�-independent NF-�B pathway. J Biol Chem 280:34538–34547

Endocrine Reviews, December 2009, 30(7):830–882 edrv.endojournals.org 861

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

94. Jiang X, Takahashi N, Matsui N, Tetsuka T, Okamoto T2003 The NF-�B activation in lymphotoxin � receptor sig-naling depends on the phosphorylation of p65 at serine536. J Biol Chem 278:919–926

95. Buss H, Dorrie A, Schmitz ML, Hoffmann E, Resch K,Kracht M 2004 Constitutive and interleukin-1-induciblephosphorylation of p65 NF-�B at serine 536 is mediated bymultiple protein kinases including I�B kinase (IKK)-�,IKK�, IKK�, TRAF family member-associated (TANK)-binding kinase 1 (TBK1), and an unknown kinase and cou-ples p65 to TATA-binding protein-associated factor II31-mediated interleukin-8 transcription. J Biol Chem 279:55633–55643

96. Adli M, Baldwin AS 2006 IKK-i/IKK� controls constitu-tive, cancer cell-associated NF-�B activity via regulation ofSer-536 p65/RelA phosphorylation. J Biol Chem 281:26976–26984

97. Ryo A, Suizu F, Yoshida Y, Perrem K, Liou YC, Wulf G,Rottapel R, Yamaoka S, Lu KP 2003 Regulation of NF-�Bsignaling by Pin1-dependent prolyl isomerization andubiquitin-mediated proteolysis of p65/RelA. Mol Cell 12:1413–1426

98. Maine GN, Mao X, Komarck CM, Burstein E 2007COMMD1 promotes the ubiquitination of NF-�B sub-units through a cullin-containing ubiquitin ligase. EMBOJ 26:436–447

99. Saccani S, Marazzi I, Beg AA, Natoli G 2004 Degradationof promoter-bound p65/RelA is essential for the prompttermination of the nuclear factor �B response. J Exp Med200:107–113

100. Bird TA, Schooley K, Dower SK, Hagen H, Virca GD 1997Activation of nuclear transcription factor NF-�B by inter-leukin-1 is accompanied by casein kinase II-mediatedphosphorylation of the p65 subunit. J Biol Chem 272:32606–32612

101. Wang D, Westerheide SD, Hanson JL, Baldwin Jr AS 2000Tumor necrosis factor �-induced phosphorylation ofRelA/p65 on Ser529 is controlled by casein kinase II. J BiolChem 275:32592–32597

102. Yeh PY, Yeh KH, Chuang SE, Song YC, Cheng AL 2004Suppression of MEK/ERK signaling pathway enhances cis-platin-induced NF-�B activation by protein phosphatase4-mediated NF-�B p65 Thr dephosphorylation. J BiolChem 279:26143–26148

103. Rocha S, Garrett MD, Campbell KJ, Schumm K, PerkinsND 2005 Regulation of NF-�B and p53 through activationof ATR and Chk1 by the ARF tumour suppressor. EMBOJ 24:1157–1169

104. Campbell KJ, Witty JM, Rocha S, Perkins ND 2006 Cis-platin mimics ARF tumor suppressor regulation of RelA(p65) nuclear factor-�B transactivation. Cancer Res 66:929–935

105. Mattioli I, Geng H, Sebald A, Hodel M, Bucher C, KrachtM, Schmitz ML 2006 Inducible phosphorylation of NF-�Bp65 at serine 468 by T cell costimulation is mediated byIKK�. J Biol Chem 281:6175–6183

106. Yang J, Fan GH, Wadzinski BE, Sakurai H, Richmond A2001 Protein phosphatase 2A interacts with and directlydephosphorylates RelA. J Biol Chem 276:47828–47833

107. Zhou G, Mihindukulasuriya KA, MacCorkle-ChosnekRA, Van Hooser A, Hu MC, Brinkley BR, Tan TH 2002Protein phosphatase 4 is involved in tumor necrosis factor-

�-induced activation of c-Jun N-terminal kinase. J BiolChem 277:6391–6398

108. Hu MC, Tang-Oxley Q, Qiu WR, Wang YP,Mihindukulasuriya KA, Afshar R, Tan TH 1998 Proteinphosphatase X interacts with c-Rel and stimulates c-Rel/nuclear factor �B activity. J Biol Chem 273:33561–33565

109. Nelson DE, Ihekwaba AE, Elliott M, Johnson JR, GibneyCA, Foreman BE, Nelson G, See V, Horton CA, Spiller DG,Edwards SW, McDowell HP, Unitt JF, Sullivan E, GrimleyR, Benson N, Broomhead D, Kell DB, White MR 2004Oscillations in NF-�B signaling control the dynamics ofgene expression. Science 306:704–708

110. Hoffmann A, Natoli G, Ghosh G 2006 Transcriptionalregulation via the NF-�B signaling module. Oncogene 25:6706–6716

111. Vogel CF, Matsumura F 2009 A new cross-talk betweenthe aryl hydrocarbon receptor and RelB, a member of theNF-�B family. Biochem Pharmacol 77:734–745

112. Tian Y 2009 Ah receptor and NF-�B interplay on the stageof epigenome. Biochem Pharmacol 77:670–680

113. Basak S, Hoffmann A 2008 Crosstalk via the NF-�B sig-naling system. Cytokine Growth Factor Rev 19:187–197

114. Fujioka S, Niu J, Schmidt C, Sclabas GM, Peng B, UwagawaT, Li Z, Evans DB, Abbruzzese JL, Chiao PJ 2004 NF-�Band AP-1 connection: mechanism of NF-�B-dependentregulation of AP-1 activity. Mol Cell Biol 24:7806–7819

115. Kaisho T, Tanaka T 2008 Turning NF-�B and IRFs on andoff in DC. Trends Immunol 29:329–336

116. Kao YS, Fong JC 2008 Endothelin-1 induces glut1 tran-scription through enhanced interaction between Sp1 andNF-�B transcription factors. Cell Signal 20:771–778

117. Park JM, Greten FR, Wong A, Westrick RJ, Arthur JS,Otsu K, Hoffmann A, Montminy M, Karin M 2005 Sig-naling pathways and genes that inhibit pathogen-inducedmacrophage apoptosis—CREB and NF-�B as key regula-tors. Immunity 23:319–329

118. Yang J, Liao X, Agarwal MK, Barnes L, Auron PE, StarkGR 2007 Unphosphorylated STAT3 accumulates in re-sponse to IL-6 and activates transcription by binding toNF�B. Genes Dev 21:1396–1408

119. Bosisio D, Marazzi I, Agresti A, Shimizu N, Bianchi ME,Natoli G 2006 A hyper-dynamic equilibrium between pro-moter-bound and nucleoplasmic dimers controls NF-�B-dependent gene activity. EMBO J 25:798–810

120. De Bosscher K, Haegeman G 2009 Minireview: latest per-spectives on antiinflammatory actions of glucocorticoids.Mol Endocrinol 23:281–291

121. Thanos D, Maniatis T 1995 Virus induction of human IFN� gene expression requires the assembly of an enhanceo-some. Cell 83:1091–1100

122. Edelstein LC, Pan A, Collins T 2005 Chromatin modifi-cation and the endothelial-specific activation of the E-se-lectin gene. J Biol Chem 280:11192–11202

123. Gerritsen ME, Williams AJ, Neish AS, Moore S, Shi Y,Collins T 1997 CREB-binding protein/p300 are transcrip-tional coactivators of p65. Proc Natl Acad Sci USA 94:2927–2932

124. Perkins ND, Felzien LK, Betts JC, Leung K, Beach DH,Nabel GJ 1997 Regulation of NF-�B by cyclin-dependentkinases associated with the p300 coactivator. Science 275:523–527

125. Na SY, Lee SK, Han SJ, Choi HS, Im SY, Lee JW 1998

862 Beck et al. GR Crosstalk with Kinases and Phosphatases Endocrine Reviews, December 2009, 30(7):830–882

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

Steroid receptor coactivator-1 interacts with the p50 sub-unit and coactivates nuclear factor �B-mediated transac-tivations. J Biol Chem 273:10831–10834

126. Sheppard KA, Rose DW, Haque ZK, Kurokawa R, McInerneyE, Westin S, Thanos D, Rosenfeld MG, Glass CK, CollinsT 1999 Transcriptional activation by NF-�B requires mul-tiple coactivators. Mol Cell Biol 19:6367–6378

127. Ito K, Charron CE, Adcock IM 2007 Impact of proteinacetylation in inflammatory lung diseases. Pharmacol Ther116:249–265

128. Huang WC, Ju TK, Hung MC, Chen CC 2007 Phosphor-ylation of CBP by IKK� promotes cell growth by switchingthe binding preference of CBP from p53 to NF-�B. MolCell 26:75–87

129. Ashburner BP, Westerheide SD, Baldwin Jr AS 2001 Thep65 (RelA) subunit of NF-�B interacts with the histonedeacetylase (HDAC) corepressors HDAC1 and HDAC2to negatively regulate gene expression. Mol Cell Biol21:7065–7077

130. Ito K, Barnes PJ, Adcock IM 2000 Glucocorticoid receptorrecruitment of histone deacetylase 2 inhibits interleukin-1�-induced histone H4 acetylation on lysines 8 and 12.Mol Cell Biol 20:6891–6903

131. Brandl A, Heinzel T, Kramer OH 2009 Histone deacety-lases: salesmen and customers in the post-translationalmodification market. Biol Cell 101:193–205

132. Grabiec AM, Tak PP, Reedquist KA 2008 Targeting his-tone deacetylase activity in rheumatoid arthritis andasthma as prototypes of inflammatory disease: should wekeep our HATs on? Arthritis Res Ther 10:226

133. Calao M, Burny A, Quivy V, Dekoninck A, Van Lint C2008 A pervasive role of histone acetyltransferases anddeacetylases in an NF-�B-signaling code. Trends BiochemSci 33:339–349

134. Lee SK, Kim JH, Lee YC, Cheong J, Lee JW 2000 Silencingmediator of retinoic acid and thyroid hormone receptors,as a novel transcriptional corepressor molecule of activat-ing protein-1, nuclear factor-�B, and serum response fac-tor. J Biol Chem 275:12470–12474

135. Espinosa L, Ingles-Esteve J, Robert-Moreno A, Bigas A2003 I�B� and p65 regulate the cytoplasmic shuttling ofnuclear corepressors: cross-talk between Notch and NF�Bpathways. Mol Biol Cell 14:491–502

136. Ghosh S, May MJ, Kopp EB 1998 NF-� B and Rel proteins:evolutionarily conserved mediators of immune responses.Annu Rev Immunol 16:225–260

137. Saccani S, Pantano S, Natoli G 2001 Two waves of nuclearfactor �B recruitment to target promoters. J Exp Med 193:1351–1359

138. Tian B, Nowak DE, Brasier AR 2005 A TNF-induced geneexpression program under oscillatory NF-�B control.BMC Genomics 6:137

139. Tian B, Nowak DE, Jamaluddin M, Wang S, Brasier AR2005 Identification of direct genomic targets downstreamof the nuclear factor-�B transcription factor mediating tu-mor necrosis factor signaling. J Biol Chem 280:17435–17448

140. Tam WF, Wang W, Sen R 2001 Cell-specific associationand shuttling of I�B� provides a mechanism for nuclearNF-�B in B lymphocytes. Mol Cell Biol 21:4837–4846

141. Ghosh S, Hayden MS 2008 New regulators of NF-�B ininflammation. Nat Rev Immunol 8:837–848

142. Jaattela M, Mouritzen H, Elling F, Bastholm L 1996 A20zinc finger protein inhibits TNF and IL-1 signaling. J Im-munol 156:1166–1173

143. Wullaert A, Heyninck K, Janssens S, Beyaert R 2006 Ubiq-uitin: tool and target for intracellular NF-�B inhibitors.Trends Immunol 27:533–540

144. Mauro C, Pacifico F, Lavorgna A, Mellone S, Iannetti A,Acquaviva R, Formisano S, Vito P, Leonardi A 2006ABIN-1 binds to NEMO/IKK� and co-operates with A20in inhibiting NF-�B. J Biol Chem 281:18482–18488

145. Gallagher J, Howlin J, McCarthy C, Murphy EP, BresnihanB, FitzGerald O, Godson C, Brady HR, Martin F 2003Identification of Naf1/ABIN-1 among TNF-�-induced ex-pressed genes in human synoviocytes using oligonucleotidemicroarrays. FEBS Lett 551:8–12

146. Heyninck K, Kreike MM, Beyaert R 2003 Structure-func-tion analysis of the A20-binding inhibitor of NF-�B acti-vation, ABIN-1. FEBS Lett 536:135–140

147. Lantz M, Malik S, Slevin ML, Olsson I 1990 Infusion oftumor necrosis factor (TNF) causes an increase in cir-culating TNF-binding protein in humans. Cytokine 2:402– 406

148. Reddy P, Slack JL, Davis R, Cerretti DP, Kozlosky CJ,Blanton RA, Shows D, Peschon JJ, Black RA 2000 Func-tional analysis of the domain structure of tumor necrosisfactor-� converting enzyme. J Biol Chem 275:14608–14614

149. Kovalenko A, Chable-Bessia C, Cantarella G, Israel A,Wallach D, Courtois G 2003 The tumour suppressorCYLD negatively regulates NF-�B signalling by deubiqui-tination. Nature 424:801–805

150. Fukuda M, Fukuda F, Horiuchi Y, Oku Y, Suzuki S,Kusama K, Sakashita H 2006 Expression of CYLD, NF-�Band NF-�B-related factors in salivary gland tumors. InVivo 20:467–472

151. Platzer C, Meisel C, Vogt K, Platzer M, Volk HD 1995Up-regulation of monocytic IL-10 by tumor necrosis fac-tor-� and cAMP elevating drugs. Int Immunol 7:517–523

152. Taganov KD, Boldin MP, Chang KJ, Baltimore D 2006NF-�B-dependent induction of microRNA miR-146, aninhibitor targeted to signaling proteins of innate immuneresponses. Proc Natl Acad Sci USA 103:12481–12486

153. Werner SL, Kearns JD, Zadorozhnaya V, Lynch C, O’DeaE, Boldin MP, Ma A, Baltimore D, Hoffmann A 2008Encoding NF-�B temporal control in response to TNF:distinct roles for the negative regulators I�B� and A20.Genes Dev 22:2093–2101

154. Zenz R, Eferl R, Scheinecker C, Redlich K, Smolen J,Schonthaler HB, Kenner L, Tschachler E, Wagner EF 2008Activator protein 1 (Fos/Jun) functions in inflammatorybone and skin disease. Arthritis Res Ther 10:201

155. Hess J, Angel P, Schorpp-Kistner M 2004 AP-1 subunits:quarrel and harmony among siblings. J Cell Sci 117:5965–5973

156. Eychene A, Rocques N, Pouponnot C 2008 A new MAFiain cancer. Nat Rev Cancer 8:683–693

157. van Dam H, Castellazzi M 2001 Distinct roles of Jun:Fosand Jun:ATF dimers in oncogenesis. Oncogene 20:2453–2464

158. Eferl R, Wagner EF 2003 AP-1: a double-edged sword intumorigenesis. Nat Rev Cancer 3:859–868

159. Hai T, Hartman MG 2001 The molecular biology and

Endocrine Reviews, December 2009, 30(7):830–882 edrv.endojournals.org 863

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

nomenclature of the activating transcription factor/cAMPresponsive element binding family of transcription factors:activating transcription factor proteins and homeostasis.Gene 273:1–11

160. Xiao W, Hodge DR, Wang L, Yang X, Zhang X, FarrarWL 2004 NF-�B activates IL-6 expression through coop-eration with c-Jun and IL6-AP1 site, but is independent ofits IL6-NF�B regulatory site in autocrine human multiplemyeloma cells. Cancer Biol Ther 3:1007–1017

161. Hazzalin CA, Mahadevan LC 2002 MAPK-regulated tran-scription: a continuously variable gene switch? Nat RevMol Cell Biol 3:30–40

162. WagnerEF,EferlR2005Fos/AP-1proteins inboneand theimmune system. Immunol Rev 208:126–140

163. Bannister AJ, Kouzarides T 1995 CBP-induced stimula-tion of c-Fos activity is abrogated by E1A. EMBO J 14:4758–4762

164. Janknecht R, Nordheim A 1996 MAP kinase-dependenttranscriptional coactivation by Elk-1 and its cofactor CBP.Biochem Biophys Res Commun 228:831–837

165. Cahill MA, Janknecht R, Nordheim A 1996 Signallingpathways: jack of all cascades. Curr Biol 6:16–19

166. Janknecht R, Cahill MA, Nordheim A 1995 Signal inte-gration at the c-fos promoter. Carcinogenesis 16:443–450

167. Tanos T, Marinissen MJ, Leskow FC, Hochbaum D,Martinetto H, Gutkind JS, Coso OA 2005 Phosphoryla-tion of c-Fos by members of the p38 MAPK family. Role inthe AP-1 response to UV light. J Biol Chem 280:18842–18852

168. Espino PS, Li L, He S, Yu J, Davie JR 2006 Chromatinmodification of the trefoil factor 1 gene in human breastcancer cells by the Ras/mitogen-activated protein kinasepathway. Cancer Res 66:4610–4616

169. Kallunki T, Deng T, Hibi M, Karin M 1996 c-Jun canrecruit JNK to phosphorylate dimerization partners viaspecific docking interactions. Cell 87:929–939

170. Chalmers CJ, Gilley R, March HN, Balmanno K, CookSJ 2007 The duration of ERK1/2 activity determines theactivation of c-Fos and Fra-1 and the composition andquantitative transcriptional output of AP-1. Cell Signal19:695–704

171. Sutherland JA, Cook A, Bannister AJ, Kouzarides T 1992Conserved motifs in Fos and Jun define a new class ofactivation domain. Genes Dev 6:1810–1819

172. Rogatsky I, Zarember KA, Yamamoto KR 2001 Factorrecruitment and TIF2/GRIP1 corepressor activity at a col-lagenase-3 response element that mediates regulation byphorbol esters and hormones. EMBO J 20:6071–6083

173. Ozanne BW, Spence HJ, McGarry LC, Hennigan RF 2006Invasion is a genetic program regulated by transcriptionfactors. Curr Opin Genet Dev 16:65–70

174. Ozanne BW, Spence HJ, McGarry LC, Hennigan RF 2007Transcription factors control invasion: AP-1 the firstamong equals. Oncogene 26:1–10

175. Chrousos GP, Kino T 2007 Glucocorticoid action net-works and complex psychiatric and/or somatic disorders.Stress 10:213–219

176. Germain P, Staels B, Dacquet C, Spedding M, Laudet V2006 Overview of nomenclature of nuclear receptors.Pharmacol Rev 58:685–704

177. Datson NA, van der Perk J, de Kloet ER, Vreugdenhil E2001 Identification of corticosteroid-responsive genes in

rat hippocampus using serial analysis of gene expression.Eur J Neurosci 14:675–689

178. Kumar R, Thompson EB 1999 The structure of the nuclearhormone receptors. Steroids 64:310–319

179. Luisi BF, Xu WX, Otwinowski Z, Freedman LP,Yamamoto KR, Sigler PB 1991 Crystallographic analysisof the interaction of the glucocorticoid receptor with DNA.Nature 352:497–505

180. Dahlman-Wright K, Wright A, Gustafsson JA, Carlstedt-Duke J 1991 Interaction of the glucocorticoid receptorDNA-binding domain with DNA as a dimer is mediated bya short segment of five amino acids. J Biol Chem 266:3107–3112

181. Hard T, Kellenbach E, Boelens R, Maler BA, DahlmanK, Freedman LP, Carlstedt-Duke J, Yamamoto KR,Gustafsson JA, Kaptein R 1990 Solution structure of theglucocorticoid receptor DNA-binding domain. Science249:157–160

182. van Tilborg MA, Bonvin AM, Hård K, Davis AL, Maler B,Boelens R, Yamamoto KR, Kaptein R 1995 Structure re-finement of the glucocorticoid receptor-DNA binding do-main from NMR data by relaxation matrix calculations. JMol Biol 247:689–700

183. van Tilborg MA, Lefstin JA, Kruiskamp M, Teuben J,Boelens R, Yamamoto KR, Kaptein R 2000 Mutations inthe glucocorticoid receptor DNA-binding domain mimican allosteric effect of DNA. J Mol Biol 301:947–958

184. Robinson-Rechavi M, Escriva Garcia H, Laudet V 2003The nuclear receptor superfamily. J Cell Sci 116:585–586

185. Bledsoe RK, Stewart EL, Pearce KH 2004 Structure andfunction of the glucocorticoid receptor ligand binding do-main. Vitam Horm 68:49–91

186. Pratt WB, Galigniana MD, Morishima Y, Murphy PJ 2004Role of molecular chaperones in steroid receptor action.Essays Biochem 40:41–58

187. Darimont BD, Wagner RL, Apriletti JW, Stallcup MR,Kushner PJ, Baxter JD, Fletterick RJ, Yamamoto KR 1998Structure and specificity of nuclear receptor-coactivatorinteractions. Genes Dev 12:3343–3356

188. Hong H, Darimont BD, Ma H, Yang L, Yamamoto KR,Stallcup MR 1999 An additional region of coactivatorGRIP1 required for interaction with the hormone-bindingdomains of a subset of nuclear receptors. J Biol Chem 274:3496–3502

189. RicketsonD,HostickU,FangL,YamamotoKR,DarimontBD2007 A conformational switch in the ligand-binding do-main regulates the dependence of the glucocorticoid recep-tor on Hsp90. J Mol Biol 368:729–741

190. Meijsing SH, Pufall MA, So AY, Bates DL, Chen L,Yamamoto KR 2009 DNA binding site sequence directsglucocorticoid receptor structure and activity. Science 324:407–410

191. Bladh LG, Liden J, Dahlman-Wright K, Reimers M, Nilsson S,Okret S 2005 Identification of endogenous glucocorticoidrepressed genes differentially regulated by a glucocorticoidreceptor mutant able to separate between nuclear fac-tor-�B and activator protein-1 repression. Mol Pharmacol67:815–826

192. Liden J, Delaunay F, Rafter I, Gustafsson J, Okret S 1997A new function for the C-terminal zinc finger of the glu-cocorticoid receptor. Repression of RelA transactivation.J Biol Chem 272:21467–21472

864 Beck et al. GR Crosstalk with Kinases and Phosphatases Endocrine Reviews, December 2009, 30(7):830–882

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

193. Heck S, Kullmann M, Gast A, Ponta H, Rahmsdorf HJ,Herrlich P, Cato AC 1994 A distinct modulating domain inglucocorticoid receptor monomers in the repression of ac-tivity of the transcription factor AP-1. EMBO J 13:4087–4095

194. Picard D, Yamamoto KR 1987 Two signals mediate hor-mone-dependent nuclear localization of the glucocorticoidreceptor. EMBO J 6:3333–3340

195. Savory JG, Hsu B, Laquian IR, Giffin W, Reich T, HacheRJ, Lefebvre YA 1999 Discrimination between NL1- andNL2-mediated nuclear localization of the glucocorticoidreceptor. Mol Cell Biol 19:1025–1037

196. Duma D, Jewell CM, Cidlowski JA 2006 Multiple glu-cocorticoid receptor isoforms and mechanisms of post-translational modification. J Steroid Biochem Mol Biol102:11–21

197. Lu NZ, Cidlowski JA 2006 Glucocorticoid receptor iso-forms generate transcription specificity. Trends Cell Biol16:301–307

198. Lu NZ, Collins JB, Grissom SF, Cidlowski JA 2007 Selec-tive regulation of bone cell apoptosis by translational iso-forms of the glucocorticoid receptor. Mol Cell Biol 27:7143–7160

199. Gross KL, Cidlowski JA 2008 Tissue-specific glucocorti-coid action: a family affair. Trends Endocrinol Metab 19:331–339

200. Gross KL, Lu NZ, Cidlowski JA 2009 Molecular mecha-nisms regulating glucocorticoid sensitivity and resistance.Mol Cell Endocrinol 300:7–16

201. Charmandari E, Kino T, Ichijo T, Chrousos GP 2008 Gen-eralized glucocorticoid resistance: clinical aspects, molec-ular mechanisms, and implications of a rare genetic disor-der. J Clin Endocrinol Metab 93:1563–1572

202. Gametchu B, Chen F, Sackey F, Powell C, Watson CS 1999Plasma membrane-resident glucocorticoid receptors in ro-dent lymphoma and human leukemia models. Steroids 64:107–119

203. Guo Z, Chen YZ, Xu RB, Fu H 1995 Binding character-istics of glucocorticoid receptor in synaptic plasma mem-brane from rat brain. Funct Neurol 10:183–194

204. Sionov RV, Cohen O, Kfir S, Zilberman Y, Yefenof E 2006Role of mitochondrial glucocorticoid receptor in glucocor-ticoid-induced apoptosis. J Exp Med 203:189–201

205. Evans SJ, Murray TF, Moore FL 2000 Partial purificationand biochemical characterization of a membrane glucocor-ticoid receptor from an amphibian brain. J Steroid Bio-chem Mol Biol 72:209–221

206. Gametchu B, Watson CS, Wu S 1993 Use of receptor an-tibodies to demonstrate membrane glucocorticoid recep-tor in cells from human leukemic patients. FASEB J7:1283–1292

207. Tasker JG, Di S, Malcher-Lopes R 2006 Minireview: rapidglucocorticoid signaling via membrane-associated recep-tors. Endocrinology 147:5549–5556

208. Grad I, Picard D 2007 The glucocorticoid responses areshaped by molecular chaperones. Mol Cell Endocrinol275:2–12

209. Haller J, Mikics E, Makara GB 2008 The effects of non-genomic glucocorticoid mechanisms on bodily functionsand the central neural system. A critical evaluation of find-ings. Front Neuroendocrinol 29:273–291

210. Bledsoe RK, Montana VG, Stanley TB, Delves CJ, Apolito

CJ, McKee DD, Consler TG, Parks DJ, Stewart EL, WillsonTM,LambertMH,MooreJT,PearceKH,XuHE2002Crys-tal structure of the glucocorticoid receptor ligand binding do-main reveals a novel mode of receptor dimerization and co-activator recognition. Cell 110:93–105

211. De Bosscher K, Vanden Berghe W, Haegeman G 2003 Theinterplay between the glucocorticoid receptor and nuclearfactor-�B or activator protein-1: molecular mechanismsfor gene repression. Endocr Rev 24:488–522

212. De Bosscher K, Vanden Berghe W, Haegeman G 2006Cross-talk between nuclear receptors and nuclear factor�B. Oncogene 25:6868–6886

213. Song IH, Buttgereit F 2006 Non-genomic glucocorticoideffects to provide the basis for new drug developments.Mol Cell Endocrinol 246:142–146

214. Lowenberg M, Verhaar AP, van den Brink GR, HommesDW 2007 Glucocorticoid signaling: a non-genomic mech-anism for T-cell immunosuppression. Trends Mol Med 13:158–163

215. Falkenstein E, Wehling M 2000 Nongenomically initiatedsteroid actions. Eur J Clin Invest 30(Suppl 3):51–54

216. Hache RJ, Tse R, Reich T, Savory JG, Lefebvre YA 1999Nucleocytoplasmic trafficking of steroid-free glucocorti-coid receptor. J Biol Chem 274:1432–1439

217. Freedman ND, Yamamoto KR 2004 Importin 7 and im-portin �/importin � are nuclear import receptors for theglucocorticoid receptor. Mol Biol Cell 15:2276–2286

218. Tao T, Lan J, Lukacs GL, Hache RJ, Kaplan F 2006 Im-portin 13 regulates nuclear import of the glucocorticoidreceptor in airway epithelial cells. Am J Respir Cell MolBiol 35:668–680

219. Kumar S, Chaturvedi NK, Nishi M, Kawata M, Tyagi RK2004 Shuttling components of nuclear import machineryinvolved in nuclear translocation of steroid receptors exitnucleus via exportin-1/CRM-1 independent pathway. Bio-chim Biophys Acta 1691:73–77

220. Holaska JM, Black BE, Rastinejad F, Paschal BM 2002Ca2�-dependent nuclear export mediated by calreticulin.Mol Cell Biol 22:6286–6297

221. Walther RF, Lamprecht C, Ridsdale A, Groulx I, Lee S,Lefebvre YA, Hache RJ 2003 Nuclear export of the glu-cocorticoid receptor is accelerated by cell fusion-depen-dent release of calreticulin. J Biol Chem 278:37858–37864

222. Carrigan A, Walther RF, Salem HA, Wu D, Atlas E,Lefebvre YA, Hache RJ 2007 An active nuclear retentionsignal in the glucocorticoid receptor functions as a stronginducer of transcriptional activation. J Biol Chem 282:10963–10971

223. McNally JG, Muller WG, Walker D, Wolford R, Hager GL2000 The glucocorticoid receptor: rapid exchange withregulatory sites in living cells. Science 287:1262–1265

224. Sackey FN, Hache RJ, Reich T, Kwast-Welfeld J, LefebvreYA 1996 Determinants of subcellular distribution of theglucocorticoid receptor. Mol Endocrinol 10:1191–1205

225. Schaaf MJ, Lewis-Tuffin LJ, Cidlowski JA 2005 Ligand-selective targeting of the glucocorticoid receptor to nuclearsubdomains is associated with decreased receptor mobil-ity. Mol Endocrinol 19:1501–1515

226. Burnstein KL, Jewell CM, Sar M, Cidlowski JA 1994 In-tragenic sequences of the human glucocorticoid receptorcomplementary DNA mediate hormone-inducible recep-

Endocrine Reviews, December 2009, 30(7):830–882 edrv.endojournals.org 865

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

tor messenger RNA down-regulation through multiplemechanisms. Mol Endocrinol 8:1764–1773

227. Okret S, Poellinger L, Dong Y, Gustafsson JA 1986 Down-regulation of glucocorticoid receptor mRNA by glucocor-ticoid hormones and recognition by the receptor of a spe-cific binding sequence within a receptor cDNA clone. ProcNatl Acad Sci USA 83:5899–5903

228. Shimojo M, Hiroi N, Yakushiji F, Ueshiba H, YamaguchiN, Miyachi Y 1995 Differences in down-regulation of glu-cocorticoid receptor mRNA by cortisol, prednisolone anddexamethasone in HeLa cells. Endocr J 42:629–636

229. Vedeckis WV, Ali M, Allen HR 1989 Regulation of glu-cocorticoid receptor protein and mRNA levels. Cancer Res49:2295s–2302s

230. Alarid ET 2006 Lives and times of nuclear receptors. MolEndocrinol 20:1972–1981

231. Hoeck W, Rusconi S, Groner B 1989 Down-regulation andphosphorylation of glucocorticoid receptors in culturedcells. Investigations with a monospecific antiserum againsta bacterially expressed receptor fragment. J Biol Chem264:14396–14402

232. Wallace AD, Cidlowski JA 2001 Proteasome-mediatedglucocorticoid receptor degradation restricts transcrip-tional signaling by glucocorticoids. J Biol Chem 276:42714–42721

233. Liu J, DeFranco DB 2000 Protracted nuclear export ofglucocorticoid receptor limits its turnover and does notrequire the exportin 1/CRM1-directed nuclear exportpathway. Mol Endocrinol 14:40–51

234. La Baer J, Yamamoto KR 1994 Analysis of the DNA-bind-ing affinity, sequence specificity and context dependence ofthe glucocorticoid receptor zinc finger region. J Mol Biol239:664–688

235. Schena M, Freedman LP, Yamamoto KR 1989 Mutationsin the glucocorticoid receptor zinc finger region that dis-tinguish interdigitated DNA binding and transcriptionalenhancement activities. Genes Dev 3:1590–1601

236. Rhen T, Cidlowski JA 2005 Antiinflammatory action ofglucocorticoids—new mechanisms for old drugs. N EnglJ Med 353:1711–1723

237. Lefstin JA, Thomas JR, Yamamoto KR 1994 Influence ofa steroid receptor DNA-binding domain on transcriptionalregulatory functions. Genes Dev 8:2842–2856

238. Lefstin JA, Yamamoto KR 1998 Allosteric effects of DNAon transcriptional regulators. Nature 392:885–888

239. Ringold GM, Yamamoto KR, Tomkins GM, Bishop M,Varmus HE 1975 Dexamethasone-mediated induction ofmouse mammary tumor virus RNA: a system for studyingglucocorticoid action. Cell 6:299–305

240. Fryer CJ, Archer TK 1998 Chromatin remodelling by theglucocorticoid receptor requires the BRG1 complex. Na-ture 393:88–91

241. Diamond MI, Miner JN, Yoshinaga SK, Yamamoto KR1990 Transcription factor interactions: selectors of posi-tive or negative regulation from a single DNA element.Science 249:1266–1272

242. Miner JN, Yamamoto KR 1991 Regulatory crosstalk atcomposite response elements. Trends Biochem Sci 16:423– 426

243. Clark AR 2007 Anti-inflammatory functions of glucocor-ticoid-induced genes. Mol Cell Endocrinol 275:79–97

244. Rogatsky I, Wang JC, Derynck MK, Nonaka DF,

Khodabakhsh DB, Haqq CM, Darimont BD, GarabedianMJ, Yamamoto KR 2003 Target-specific utilization oftranscriptional regulatory surfaces by the glucocorticoidreceptor. Proc Natl Acad Sci USA 100:13845–13850

245. So AY, Chaivorapol C, Bolton EC, Li H, Yamamoto KR2007 Determinants of cell- and gene-specific transcrip-tional regulation by the glucocorticoid receptor. PLoSGenet 3:e94

246. So AY, Cooper SB, Feldman BJ, Manuchehri M,Yamamoto KR 2008 Conservation analysis predicts invivo occupancy of glucocorticoid receptor-binding se-quences at glucocorticoid-induced genes. Proc Natl AcadSci USA 105:5745–5749

247. De Martino MU, Bhattachryya N, Alesci S, Ichijo T,Chrousos GP, Kino T 2004 The glucocorticoid receptorand the orphan nuclear receptor chicken ovalbumin up-stream promoter-transcription factor II interact with andmutually affect each other’s transcriptional activities: im-plications for intermediary metabolism. Mol Endocrinol18:820–833

248. Doppler W, Windegger M, Soratroi C, Tomasi J, LechnerJ,RusconiS,CatoAC,AlmlofT,LidenJ,OkretS,GustafssonJA, Richard-Foy H, Starr DB, Klocker H, Edwards D,Geymayer S 2001 Expression level-dependent contribu-tion of glucocorticoid receptor domains for functional in-teraction with STAT5. Mol Cell Biol 21:3266–3279

249. Stocklin E, Wissler M, Gouilleux F, Groner B 1996 Func-tional interactions between Stat5 and the glucocorticoidreceptor. Nature 383:726–728

250. Pearce D, Matsui W, Miner JN, Yamamoto KR 1998 Glu-cocorticoid receptor transcriptional activity determined byspacing of receptor and nonreceptor DNA sites. J BiolChem 273:30081–30085

251. Kassel O, Herrlich P 2007 Crosstalk between the glucocor-ticoid receptor and other transcription factors: molecularaspects. Mol Cell Endocrinol 275:13–29

252. Rogatsky I, Ivashkiv LB 2006 Glucocorticoid modulationof cytokine signaling. Tissue Antigens 68:1–12

253. Luecke HF, Yamamoto KR 2005 The glucocorticoid re-ceptor blocks P-TEFb recruitment by NF�B to effectpromoter-specific transcriptional repression. Genes Dev19:1116 –1127

254. Nissen RM, Yamamoto KR 2000 The glucocorticoid re-ceptor inhibits NF�B by interfering with serine-2 phos-phorylation of the RNA polymerase II carboxy-terminaldomain. Genes Dev 14:2314–2329

255. Caldenhoven E, Liden J, Wissink S, Van de Stolpe A,Raaijmakers J, Koenderman L, Okret S, Gustafsson JA,Van der Saag PT 1995 Negative cross-talk between RelAand the glucocorticoid receptor: a possible mechanism forthe antiinflammatory action of glucocorticoids. Mol En-docrinol 9:401–412

256. McKay LI, Cidlowski JA 1998 Cross-talk between nuclearfactor-�B and the steroid hormone receptors: mechanismsof mutual antagonism. Mol Endocrinol 12:45–56

257. Li X, Wong J, Tsai SY, Tsai MJ, O’Malley BW 2003 Pro-gesterone and glucocorticoid receptors recruit distinct co-activator complexes and promote distinct patterns of localchromatin modification. Mol Cell Biol 23:3763–3773

258. Jenkins BD, Pullen CB, Darimont BD 2001 Novel glu-cocorticoid receptor coactivator effector mechanisms.Trends Endocrinol Metab 12:122–126

866 Beck et al. GR Crosstalk with Kinases and Phosphatases Endocrine Reviews, December 2009, 30(7):830–882

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

259. Perissi V, Rosenfeld MG 2005 Controlling nuclear recep-tors: the circular logic of cofactor cycles. Nat Rev Mol CellBiol 6:542–554

260. McKenna NJ, O’Malley BW 2002 Minireview: nuclearreceptor coactivators—an update. Endocrinology 143:2461–2465

261. Sandu C, Artunc F, Grahammer F, Rotte A, Boini KM,Friedrich B, Sandulache D, Metzger M, Just L, Mack A,Skutella T, Rexhepaj R, Risler T, Wulff P, Kuhl D, Lang F2007 Role of the serum and glucocorticoid inducible ki-nase SGK1 in glucocorticoid stimulation of gastric acidsecretion. Pflugers Arch 455:493–503

262. Garside H, Stevens A, Farrow S, Normand C, Houle B,Berry A, Maschera B, Ray D 2004 Glucocorticoid ligandsspecify different interactions with NF-�B by allosteric ef-fects on the glucocorticoid receptor DNA binding domain.J Biol Chem 279:50050–50059

263. Chen W, Rogatsky I, Garabedian MJ 2006 MED14 andMED1 differentially regulate target-specific gene activa-tion by the glucocorticoid receptor. Mol Endocrinol 20:560–572

264. Freeman BC, Yamamoto KR 2002 Disassembly of tran-scriptional regulatory complexes by molecular chaper-ones. Science 296:2232–2235

265. Stavreva DA, Muller WG, Hager GL, Smith CL, McNallyJG 2004 Rapid glucocorticoid receptor exchange at a pro-moter is coupled to transcription and regulated by chap-erones and proteasomes. Mol Cell Biol 24:2682–2697

266. Meijsing SH, Elbi C, Luecke HF, Hager GL, YamamotoKR 2007 The ligand binding domain controls glucocorti-coid receptor dynamics independent of ligand release. MolCell Biol 27:2442–2451

267. Chinenov Y, Rogatsky I 2007 Glucocorticoids and the in-nate immune system: crosstalk with the toll-like receptorsignaling network. Mol Cell Endocrinol 275:30–42

268. Newton R, Holden NS 2007 Separating transrepressionand transactivation: a distressing divorce for the glucocor-ticoid receptor? Mol Pharmacol 72:799–809

269. Ishmael FT, Fang X, Galdiero MR, Atasoy U, Rigby WF,Gorospe M, Cheadle C, Stellato C 2008 Role of the RNA-binding protein tristetraprolin in glucocorticoid-mediatedgene regulation. J Immunol 180:8342–8353

270. Eddleston J, Herschbach J, Wagelie-Steffen AL,Christiansen SC, Zuraw BL 2007 The anti-inflammatoryeffect of glucocorticoids is mediated by glucocorticoid-in-duced leucine zipper in epithelial cells. J Allergy Clin Im-munol 119:115–122

271. Deroo BJ, Archer TK 2001 Glucocorticoid receptor acti-vation of the I�B� promoter within chromatin. Mol BiolCell 12:3365–3374

272. Yang N, Zhang W, Shi XM 2008 Glucocorticoid-inducedleucine zipper (GILZ) mediates glucocorticoid action andinhibits inflammatory cytokine-induced COX-2 expres-sion. J Cell Biochem 103:1760–1771

273. D’Acquisto F, Perretti M, Flower RJ 2008 Annexin-A1: apivotal regulator of the innate and adaptive immune sys-tems. Br J Pharmacol 155:152–169

274. Smoak K, Cidlowski JA 2006 Glucocorticoids regulatetristetraprolin synthesis and posttranscriptionally regulatetumor necrosis factor � inflammatory signaling. Mol CellBiol 26:9126–9135

275. Maier JV, Brema S, Tuckermann J, Herzer U, Klein M,

Stassen M, Moorthy A, Cato AC 2007 Dual specificityphosphatase 1 knockout mice show enhanced susceptibil-ity to anaphylaxis but are sensitive to glucocorticoids. MolEndocrinol 21:2663–2671

276. Kleiman A, Tuckermann JP 2007 Glucocorticoid receptoraction in beneficial and side effects of steroid therapy: les-sons from conditional knockout mice. Mol Cell Endocri-nol 275:98–108

277. De Bosscher K, Schmitz ML, Vanden Berghe W, PlaisanceS, Fiers W, Haegeman G 1997 Glucocorticoid-mediatedrepression of nuclear factor-�B-dependent transcriptioninvolves direct interference with transactivation. Proc NatlAcad Sci USA 94:13504–13509

278. Wissink S, van Heerde EC, vand der Burg B, van der SaagPT 1998 A dual mechanism mediates repression of NF-�Bactivity by glucocorticoids. Mol Endocrinol 12:355–363

279. Chaudhary LR, Avioli LV 1996 Regulation of interleu-kin-8 gene expression by interleukin-1�, osteotropic hor-mones, and protein kinase inhibitors in normal humanbone marrow stromal cells. J Biol Chem 271:16591–16596

280. Chivers JE, Gong W, King EM, Seybold J, Mak JC, DonnellyLE, Holden NS, Newton R 2006 Analysis of the dissoci-ated steroid RU24858 does not exclude a role for induciblegenes in the anti-inflammatory actions of glucocorticoids.Mol Pharmacol 70:2084–2095

281. Henderson BR, Kefford RF 1993 Dexamethasone de-creases urokinase plasminogen activator mRNA stabilityin MAT 13762 rat mammary carcinoma cells. Br J Cancer67:99–101

282. Newton R, Seybold J, Kuitert LM, Bergmann M, Barnes PJ1998 Repression of cyclooxygenase-2 and prostaglandinE2 release by dexamethasone occurs by transcriptional andpost-transcriptional mechanisms involving loss of polyad-enylated mRNA. J Biol Chem 273:32312–32321

283. Tobler A, Meier R, Seitz M, Dewald B, Baggiolini M, FeyMF 1992 Glucocorticoids downregulate gene expressionof GM-CSF, NAP-1/IL-8, and IL-6, but not of M-CSF inhuman fibroblasts. Blood 79:45–51

284. Chang MM, Juarez M, Hyde DM, Wu R 2001 Mechanismof dexamethasone-mediated interleukin-8 gene suppres-sion in cultured airway epithelial cells. Am J Physiol LungCell Mol Physiol 280:L107–L115

285. Reichardt HM, Tuckermann JP, Gottlicher M, Vujic M,Weih F, Angel P, Herrlich P, Schutz G 2001 Repression ofinflammatory responses in the absence of DNA binding bythe glucocorticoid receptor. EMBO J 20:7168–7173

286. Belvisi MG, Wicks SL, Battram CH, Bottoms SE, RedfordJE, Woodman P, Brown TJ, Webber SE, Foster ML 2001Therapeutic benefit of a dissociated glucocorticoid and therelevance of in vitro separation of transrepression fromtransactivation activity. J Immunol 166:1975–1982

287. Dostert A, Heinzel T 2004 Negative glucocorticoid recep-tor response elements and their role in glucocorticoid ac-tion. Curr Pharm Des 10:2807–2816

288. Ki SH, Cho IJ, Choi DW, Kim SG 2005 Glucocorticoidreceptor (GR)-associated SMRT binding to C/EBP� TADand Nrf2 Neh4/5: role of SMRT recruited to GR in GSTA2gene repression. Mol Cell Biol 25:4150–4165

289. Malkoski SP, Dorin RI 1999 Composite glucocorticoidregulation at a functionally defined negative glucocorti-

Endocrine Reviews, December 2009, 30(7):830–882 edrv.endojournals.org 867

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

coid response element of the human corticotropin-releas-ing hormone gene. Mol Endocrinol 13:1629–1644

290. Stromstedt PE, Poellinger L, Gustafsson JA, Carlstedt-Duke J 1991 The glucocorticoid receptor binds to a se-quence overlapping the TATA box of the human osteocal-cin promoter: a potential mechanism for negativeregulation. Mol Cell Biol 11:3379–3383

291. Subramaniam N, Cairns W, Okret S 1998 Glucocorticoidsrepress transcription from a negative glucocorticoid re-sponse element recognized by two homeodomain-contain-ing proteins, Pbx and Oct-1. J Biol Chem 273:23567–23574

292. LibermanAC,Druker J,RefojoD,HolsboerF,ArztE2009Glucocorticoids inhibit GATA-3 phosphorylation and ac-tivity in T cells. FASEB J 23:1558–1571

293. De Bosscher K, Vanden Berghe W, Beck IM, Van Molle W,Hennuyer N, Hapgood J, Libert C, Staels B, Louw A,Haegeman G 2005 A fully dissociated compound of plantorigin for inflammatory gene repression. Proc Natl AcadSci USA 102:15827–15832

294. Islam KN, Mendelson CR 2008 Glucocorticoid/glucocor-ticoid receptor inhibition of surfactant protein-A (SP-A)gene expression in lung type II cells is mediated by repres-sive changes in histone modification at the SP-A promoter.Mol Endocrinol 22:585–596

295. Kassel O, Schneider S, Heilbock C, Litfin M, Gottlicher M,Herrlich P 2004 A nuclear isoform of the focal adhesionLIM-domain protein Trip6 integrates activating and re-pressing signals at AP-1- and NF-�B-regulated promoters.Genes Dev 18:2518–2528

296. Schule R, Rangarajan P, Kliewer S, Ransone LJ, Bolado J,Yang N, Verma IM, Evans RM 1990 Functional antago-nism between oncoprotein c-Jun and the glucocorticoidreceptor. Cell 62:1217–1226

297. Rogatsky I, Luecke HF, Leitman DC, Yamamoto KR 2002Alternate surfaces of transcriptional coregulator GRIP1function in different glucocorticoid receptor activation andrepression contexts. Proc Natl Acad Sci USA 99:16701–16706

298. Hollenberg SM, Evans RM 1988 Multiple and cooperativetrans-activation domains of the human glucocorticoid re-ceptor. Cell 55:899–906

299. Ogawa S, Lozach J, Benner C, Pascual G, Tangirala RK,Westin S, Hoffmann A, Subramaniam S, David M, Rosen-feld MG, Glass CK 2005 Molecular determinants ofcrosstalk between nuclear receptors and toll-like receptors.Cell 122:707–721

300. Diefenbacher M, Sekula S, Heilbock C, Maier JV, Litfin M,van Dam H, Castellazzi M, Herrlich P, Kassel O 2008Restriction to Fos family members of Trip6-dependentcoactivation and glucocorticoid receptor-dependenttrans-repression of activator protein-1. Mol Endocrinol22:1767–1780

301. Ito K, Lim S, Caramori G, Chung KF, Barnes PJ, AdcockIM 2001 Cigarette smoking reduces histone deacetylase 2expression, enhances cytokine expression, and inhibits glu-cocorticoid actions in alveolar macrophages. FASEB J 15:1110–1112

302. Tsaprouni LG, Ito K, Adcock IM, Punchard N 2007 Sup-pression of lipopolysaccharide- and tumour necrosis fac-tor-�-induced interleukin (IL)-8 expression by glucocorti-

coids involves changes in IL-8 promoter acetylation. ClinExp Immunol 150:151–157

303. Kouzarides T 2002 Histone methylation in transcriptionalcontrol. Curr Opin Genet Dev 12:198–209

304. Sheppard KA, Phelps KM, Williams AJ, Thanos D, GlassCK, Rosenfeld MG, Gerritsen ME, Collins T 1998 Nuclearintegration of glucocorticoid receptor and nuclear factor-�Bsignaling by CREB-binding protein and steroid receptorcoactivator-1. J Biol Chem 273:29291–29294

305. Burkhart BA, Hebbar PB, Trotter KW, Archer TK 2005Chromatin-dependent E1A activity modulates NF-�BRelA-mediated repression of glucocorticoid receptor-de-pendent transcription. J Biol Chem 280:6349–6358

306. De Bosscher K, Vanden Berghe W, Haegeman G 2001Glucocorticoid repression of AP-1 is not mediated bycompetition for nuclear coactivators. Mol Endocrinol15:219 –227

307. De Bosscher K, Vanden Berghe W, Vermeulen L, PlaisanceS, Boone E, Haegeman G 2000 Glucocorticoids repressNF-�B-driven genes by disturbing the interaction of p65with the basal transcription machinery, irrespective of co-activator levels in the cell. Proc Natl Acad Sci USA 97:3919–3924

308. McKay LI, Cidlowski JA 2000 CBP (CREB binding pro-tein) integrates NF-�B (nuclear factor-�B) and glucocorti-coid receptor physical interactions and antagonism. MolEndocrinol 14:1222–1234

309. Wu J, Li Y, Dietz J, Lala DS 2004 Repression of p65 tran-scriptional activation by the glucocorticoid receptor in theabsence of receptor-coactivator interactions. Mol Endo-crinol 18:53–62

310. Sun Y, Tao YG, Kagan BL, He Y, Jr SS 2008 Modulation oftranscription parameters in glucocorticoid receptor-mediatedrepression. Mol Cell Endocrinol 295:59–69

311. Jonas BA, Varlakhanova N, Hayakawa F, Goodson M,Privalsky ML 2007 Response of SMRT (silencing mediatorof retinoic acid and thyroid hormone receptor) and N-CoR(nuclear receptor corepressor) corepressors to mitogen-ac-tivated protein kinase kinase kinase cascades is determinedby alternative mRNA splicing. Mol Endocrinol 21:1924–1939

312. Wu RC, Feng Q, Lonard DM, O’Malley BW 2007 SRC-3coactivator functional lifetime is regulated by a phospho-dependent ubiquitin time clock. Cell 129:1125–1140

313. Wu RC, Smith CL, O’Malley BW 2005 Transcriptionalregulation by steroid receptor coactivator phosphoryla-tion. Endocr Rev 26:393–399

314. Faus H, Haendler B 2006 Post-translational modificationsof steroid receptors. Biomed Pharmacother 60:520–528

315. Rochette-Egly C 2003 Nuclear receptors: integration ofmultiple signalling pathways through phosphorylation.Cell Signal 15:355–366

316. Bodwell JE, Hu JM, Orti E, Munck A 1995 Hormone-induced hyperphosphorylation of specific phosphorylatedsites in the mouse glucocorticoid receptor. J Steroid Bio-chem Mol Biol 52:135–140

317. Bodwell JE, Ortí E, Coull JM, Pappin DJ, Smith LI, SwiftF 1991 Identification of phosphorylated sites in the mouseglucocorticoid receptor. J Biol Chem 266:7549–7555

318. Rogatsky I, Waase CL, Garabedian MJ 1998 Phosphoryla-tion and inhibition of rat glucocorticoid receptor transcrip-tional activation by glycogen synthase kinase-3 (GSK-3).

868 Beck et al. GR Crosstalk with Kinases and Phosphatases Endocrine Reviews, December 2009, 30(7):830–882

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

Species-specific differences between human and rat glu-cocorticoid receptor signaling as revealed through GSK-3phosphorylation. J Biol Chem 273:14315–14321

319. Ismaili N, Garabedian MJ 2004 Modulation of glucocor-ticoid receptor function via phosphorylation. Ann NYAcad Sci 1024:86–101

320. Wang Z, Frederick J, Garabedian MJ 2002 Decipheringthe phosphorylation “code” of the glucocorticoid receptorin vivo. J Biol Chem 277:26573–26580

321. Galliher-Beckley AJ, Williams JG, Collins JB, CidlowskiJA 2008 Glycogen synthase kinase 3�-mediated serinephosphorylation of the human glucocorticoid receptor re-directs gene expression profiles. Mol Cell Biol 28:7309–7322

322. Dephoure N, Zhou C, Villen J, Beausoleil SA, BakalarskiCE, Elledge SJ, Gygi SP 2008 A quantitative atlas of mitoticphosphorylation.ProcNatlAcadSciUSA105:10762–10767

323. Krstic MD, Rogatsky I, Yamamoto KR, Garabedian MJ1997 Mitogen-activated and cyclin-dependent protein ki-nases selectively and differentially modulate transcrip-tional enhancement by the glucocorticoid receptor. MolCell Biol 17:3947–3954

324. Wang Z, Garabedian MJ 2003 Modulation of glucocor-ticoid receptor transcriptional activation, phosphoryla-tion, and growth inhibition by p27Kip1. J Biol Chem 278:50897–50901

325. Webster JC, Jewell CM, Bodwell JE, Munck A, Sar M,Cidlowski JA 1997 Mouse glucocorticoid receptor phos-phorylation status influences multiple functions of the re-ceptor protein. J Biol Chem 272:9287–9293

326. Blind RD, Garabedian MJ 2008 Differential recruitmentof glucocorticoid receptor phospho-isoforms to glucocor-ticoid-induced genes. J Steroid Biochem Mol Biol 109:150–157

327. Chen W, Dang T, Blind RD, Wang Z, Cavasotto CN,Hittelman AB, Rogatsky I, Logan SK, Garabedian MJ2008 Glucocorticoid receptor phosphorylation differen-tially affects target gene expression. Mol Endocrinol 22:1754–1766

328. Kino T, Ichijo T, Amin ND, Kesavapany S, Wang Y, KimN, Rao S, Player A, Zheng YL, Garabedian MJ, KawasakiE, Pant HC, Chrousos GP 2007 Cyclin-dependent kinase5 differentially regulates the transcriptional activity of theglucocorticoid receptor through phosphorylation: clinicalimplications for the nervous system response to glucocor-ticoids and stress. Mol Endocrinol 21:1552–1568

329. Miller AL, Webb MS, Copik AJ, Wang Y, Johnson BH,Kumar R, Thompson EB 2005 p38 Mitogen-activatedprotein kinase (MAPK) is a key mediator in glucocorticoid-induced apoptosis of lymphoid cells: correlation betweenp38 MAPK activation and site-specific phosphorylation ofthe human glucocorticoid receptor at serine 211. Mol En-docrinol 19:1569–1583

330. Irusen E, Matthews JG, Takahashi A, Barnes PJ, ChungKF, Adcock IM 2002 p38 Mitogen-activated protein ki-nase-induced glucocorticoid receptor phosphorylation re-duces its activity: role in steroid-insensitive asthma. J Al-lergy Clin Immunol 109:649–657

331. Bantel H, Schmitz ML, Raible A, Gregor M, Schulze-Osthoff K 2002 Critical role of NF-�B and stress-activatedprotein kinases in steroid unresponsiveness. FASEB J 16:1832–1834

332. Szatmary Z, Garabedian MJ, Vilcek J 2004 Inhibition ofglucocorticoid receptor-mediated transcriptional activa-tion by p38 mitogen-activated protein (MAP) kinase. J BiolChem 279:43708–43715

333. Wang X, Wu H, Miller AH 2004 Interleukin 1� (IL-1�)induced activation of p38 mitogen-activated protein ki-nase inhibits glucocorticoid receptor function. Mol Psy-chiatry 9:65–75

334. ItohM,AdachiM,YasuiH,TakekawaM,TanakaH, ImaiK 2002 Nuclear export of glucocorticoid receptor is en-hanced by c-Jun N-terminal kinase-mediated phosphory-lation. Mol Endocrinol 16:2382–2392

335. Rogatsky I, Logan SK, Garabedian MJ 1998 Antago-nism of glucocorticoid receptor transcriptional activa-tion by the c-Jun N-terminal kinase. Proc Natl Acad SciUSA 95:2050 –2055

336. Espinas ML, Roux J, Pictet R, Grange T 1995 Glucocor-ticoids and protein kinase A coordinately modulate tran-scription factor recruitment at a glucocorticoid-responsiveunit. Mol Cell Biol 15:5346–5354

337. Doucas V, Shi Y, Miyamoto S, West A, Verma I, Evans RM2000 Cytoplasmic catalytic subunit of protein kinase Amediates cross-repression by NF-�B and the glucocorticoidreceptor. Proc Natl Acad Sci USA 97:11893–11898

338. Rangarajan PN, Umesono K, Evans RM 1992 Modulationof glucocorticoid receptor function by protein kinase A.Mol Endocrinol 6:1451–1457

339. Haske T, Nakao M, Moudgil VK 1994 Phosphorylation ofimmunopurified rat liver glucocorticoid receptor by thecatalytic subunit of cAMP-dependent protein kinase. MolCell Biochem 132:163–171

340. Wang Z, Chen W, Kono E, Dang T, Garabedian MJ 2007Modulation of glucocorticoid receptor phosphorylationand transcriptional activity by a C-terminal-associatedprotein phosphatase. Mol Endocrinol 21:625–634

341. Zhang S, Jonklaas J, Danielsen M 2007 The glucocorticoidagonist activities of mifepristone (RU486) and progester-one are dependent on glucocorticoid receptor levels but noton EC50 values. Steroids 72:600–608

342. Saelzler MP, Spackman CC, Liu Y, Martinez LC, Harris JP,Abe MK 2006 ERK8 down-regulates transactivation of theglucocorticoid receptor through Hic-5. J Biol Chem 281:16821–16832

343. Yang J, Liu J, DeFranco DB 1997 Subnuclear trafficking ofglucocorticoid receptors in vitro: chromatin recycling andnuclear export. J Cell Biol 137:523–538

344. Adzic M, Djordjevic J, Djordjevic A, Niciforovic A,Demonacos C, Radojcic M, Krstic-Demonacos M 2009Acute or chronic stress induce cell compartment-specificphosphorylation of glucocorticoid receptor and alter itstranscriptional activity in Wistar rat brain. J Endocrinol202:87–97

345. Ismaili N, Blind R, Garabedian MJ 2005 Stabilization ofthe unliganded glucocorticoid receptor by TSG101. J BiolChem 280:11120–11126

346. Hittelman AB, Burakov D, Iniguez-Lluhí JA, FreedmanLP, Garabedian MJ 1999 Differential regulation of glu-cocorticoid receptor transcriptional activation via AF-1-associated proteins. EMBO J 18:5380–5388

347. DeFranco DB, Qi M, Borror KC, Garabedian MJ, BrautiganDL 1991 Protein phosphatase types 1 and/or 2A regulate

Endocrine Reviews, December 2009, 30(7):830–882 edrv.endojournals.org 869

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

nucleocytoplasmic shuttling of glucocorticoid receptors.Mol Endocrinol 5:1215–1228

348. Galigniana MD, Housley PR, DeFranco DB, Pratt WB1999 Inhibition of glucocorticoid receptor nucleocytoplas-mic shuttling by okadaic acid requires intact cytoskeleton.J Biol Chem 274:16222–16227

349. Banerjee A, Periyasamy S, Wolf IM, Hinds Jr TD, Yong W,Shou W, Sanchez ER 2008 Control of glucocorticoid andprogesterone receptor subcellular localization by the li-gand-binding domain is mediated by distinct interactionswith tetratricopeptide repeat proteins. Biochemistry 47:10471–10480

350. Silverstein AM, Galigniana MD, Chen MS, Owens-GrilloJK, Chinkers M, Pratt WB 1997 Protein phosphatase 5 isa major component of glucocorticoid receptor.hsp90 com-plexes with properties of an FK506-binding immunophi-lin. J Biol Chem 272:16224–16230

351. Chen MS, Silverstein AM, Pratt WB, Chinkers M 1996 Thetetratricopeptide repeat domain of protein phosphatase 5mediates binding to glucocorticoid receptor heterocom-plexes and acts as a dominant negative mutant. J Biol Chem271:32315–32320

352. Somers JP, DeFranco DB 1992 Effects of okadaic acid, aprotein phosphatase inhibitor, on glucocorticoid receptor-mediated enhancement. Mol Endocrinol 6:26–34

353. Zuo Z, Urban G, Scammell JG, Dean NM, McLean TK,Aragon I, Honkanen RE 1999 Ser/Thr protein phospha-tase type 5 (PP5) is a negative regulator of glucocorticoidreceptor-mediated growth arrest. Biochemistry 38:8849–8857

354. Davies TH, Ning YM, Sanchez ER 2005 Differential con-trol of glucocorticoid receptor hormone-binding functionby tetratricopeptide repeat (TPR) proteins and the immu-nosuppressive ligand FK506. Biochemistry 44:2030–2038

355. Galigniana MD, Radanyi C, Renoir JM, Housley PR, PrattWB 2001 Evidence that the peptidylprolyl isomerase do-main of the hsp90-binding immunophilin FKBP52 is in-volved in both dynein interaction and glucocorticoid re-ceptor movement to the nucleus. J Biol Chem 276:14884–14889

356. Hinds Jr TD, Sanchez ER 2008 Protein phosphatase 5. IntJ Biochem Cell Biol 40:2358–2362

357. Zhang Y, Leung DY, Nordeen SK, Goleva E 2009 Estrogeninhibits glucocorticoid action via protein phosphatase 5(PP5) mediated glucocorticoid receptor dephosphoryla-tion. J Biol Chem 284:24542–24552

358. Hay RT 2006 Role of ubiquitin-like proteins in transcrip-tional regulation. Ernst Schering Res Found Workshop 57:173–192

359. Davies L, Karthikeyan N, Lynch JT, Sial EA, Gkourtsa A,Demonacos C, Krstic-Demonacos M 2008 Cross talk ofsignaling pathways in the regulation of the glucocorticoidreceptor function. Mol Endocrinol 22:1331–1344

360. Holmstrom SR, Chupreta S, So AY, Iniguez-Lluhí JA 2008SUMO-mediated inhibition of glucocorticoid receptorsynergistic activity depends on stable assembly at the pro-moter but not on DAXX. Mol Endocrinol 22:2061–2075

361. Tian S, Poukka H, Palvimo JJ, Janne OA 2002 Small ubiq-uitin-related modifier-1 (SUMO-1) modification of theglucocorticoid receptor. Biochem J 367:907–911

362. Le Drean Y, Mincheneau N, Le Goff P, Michel D 2002

Potentiation of glucocorticoid receptor transcriptional ac-tivity by sumoylation. Endocrinology 143:3482–3489

363. Lin DY, Huang YS, Jeng JC, Kuo HY, Chang CC, ChaoTT, Ho CC, Chen YC, Lin TP, Fang HI, Hung CC, SuenCS, Hwang MJ, Chang KS, Maul GG, Shih HM 2006 Roleof SUMO-interacting motif in Daxx SUMO modification,subnuclear localization, and repression of sumoylatedtranscription factors. Mol Cell 24:341–354

364. Deroo BJ, Rentsch C, Sampath S, Young J, DeFranco DB,Archer TK 2002 Proteasomal inhibition enhances glu-cocorticoid receptor transactivation and alters its sub-nuclear trafficking. Mol Cell Biol 22:4113–4123

365. Ito K, Yamamura S, Essilfie-Quaye S, Cosio B, Ito M, BarnesPJ, Adcock IM 2006 Histone deacetylase 2-mediateddeacetylation of the glucocorticoid receptor enables NF-�Bsuppression. J Exp Med 203:7–13

366. Thalhamer T, McGrath MA, Harnett MM 2008 MAPKsand their relevance to arthritis and inflammation. Rheu-matology (Oxford) 47:409–414

367. Caelles C, Gonzalez-Sancho JM, Munoz A 1997 Nuclearhormone receptor antagonism with AP-1 by inhibition ofthe JNK pathway. Genes Dev 11:3351–3364

368. Ventura JJ, Roncero C, Fabregat I, Benito M 1999 Glu-cocorticoid receptor down-regulates c-Jun amino terminalkinases induced by tumor necrosis factor � in fetal rat he-patocyte primary cultures. Hepatology 29:849–857

369. Abraham SM, Lawrence T, Kleiman A, Warden P,Medghalchi M, Tuckermann J, Saklatvala J, Clark AR2006 Antiinflammatory effects of dexamethasone arepartly dependent on induction of dual specificity phospha-tase 1. J Exp Med 203:1883–1889

370. Chen P, Li J, Barnes J, Kokkonen GC, Lee JC, Liu Y 2002Restraint of proinflammatory cytokine biosynthesis bymitogen-activated protein kinase phosphatase-1 inlipopolysaccharide-stimulated macrophages. J Immu-nol 169:6408 – 6416

371. Issa R, Xie S, Khorasani N, Sukkar M, Adcock IM, Lee KY,Chung KF 2007 Corticosteroid inhibition of growth-re-lated oncogene protein-� via mitogen-activated kinasephosphatase-1 in airway smooth muscle cells. J Immunol178:7366–7375

372. Bruna A, Nicolas M, Munoz A, Kyriakis JM, Caelles C2003 Glucocorticoid receptor-JNK interaction mediatesinhibition of the JNK pathway by glucocorticoids. EMBOJ 22:6035–6044

373. Gonzalez MV, Jimenez B, Berciano MT, Gonzalez-SanchoJM, Caelles C, Lafarga M, Munoz A 2000 Glucocorticoidsantagonize AP-1 by inhibiting the activation/phosphory-lation of JNK without affecting its subcellular distribution.J Cell Biol 150:1199–1208

374. Hirasawa N, Izumi S, Linwong W, Ohuchi K 2003 Inhi-bition by dexamethasone of interleukin 13 production viaglucocorticoid receptor-mediated inhibition of c-Jun phos-phorylation. FEBS Lett 554:489–493

375. Cissel DS, Beaven MA 2000 Disruption of Raf-1/heatshock protein 90 complex and Raf signaling by dexameth-asone in mast cells. J Biol Chem 275:7066–7070

376. Kim MJ, Chae JS, Kim KJ, Hwang SG, Yoon KW, Kim EK,Yun HJ, Cho JH, Kim J, Kim BW, Kim HC, Kang SS, LangF, Cho SG, Choi EJ 2007 Negative regulation of SEK1signaling by serum- and glucocorticoid-inducible proteinkinase 1. EMBO J 26:3075–3085

870 Beck et al. GR Crosstalk with Kinases and Phosphatases Endocrine Reviews, December 2009, 30(7):830–882

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

377. Swantek JL, Cobb MH, Geppert TD 1997 Jun N-terminalkinase/stress-activated protein kinase (JNK/SAPK) is re-quired for lipopolysaccharide stimulation of tumor necro-sis factor � (TNF-�) translation: glucocorticoids inhibitTNF-� translation by blocking JNK/SAPK. Mol Cell Biol17:6274–6282

378. Rider LG, Hirasawa N, Santini F, Beaven MA 1996 Acti-vation of the mitogen-activated protein kinase cascade issuppressed by low concentrations of dexamethasone inmast cells. J Immunol 157:2374–2380

379. Bhalla V, Soundararajan R, Pao AC, Li H, Pearce D 2006Disinhibitory pathways for control of sodium transport:regulation of ENaC by SGK1 and GILZ. Am J PhysiolRenal Physiol 291:F714–F721

380. Soundararajan R, Zhang TT, Wang J, Vandewalle A,Pearce D 2005 A novel role for glucocorticoid-inducedleucine zipper protein in epithelial sodium channel-medi-ated sodium transport. J Biol Chem 280:39970–39981

381. Wang JC, Derynck MK, Nonaka DF, Khodabakhsh DB,Haqq C, Yamamoto KR 2004 Chromatin immunoprecipi-tation (ChIP) scanning identifies primary glucocorticoidreceptor target genes. Proc Natl Acad Sci USA 101:15603–15608

382. Cohen N, Mouly E, Hamdi H, Maillot MC, Pallardy M,Godot V, Capel F, Balian A, Naveau S, Galanaud P, LemoineFM, Emilie D 2006 GILZ expression in human dendriticcells redirects their maturation and prevents antigen-spe-cific T lymphocyte response. Blood 107:2037–2044

383. Ehrchen J, Steinmuller L, Barczyk K, Tenbrock K, NackenW, Eisenacher M, Nordhues U, Sorg C, Sunderkotter C,Roth J 2007 Glucocorticoids induce differentiation of aspecifically activated, anti-inflammatory subtype of hu-man monocytes. Blood 109:1265–1274

384. Berrebi D, Bruscoli S, Cohen N, Foussat A, Migliorati G,Bouchet-Delbos L, Maillot MC, Portier A, Couderc J,Galanaud P, Peuchmaur M, Riccardi C, Emilie D 2003Synthesis of glucocorticoid-induced leucine zipper (GILZ)by macrophages: an anti-inflammatory and immunosup-pressive mechanism shared by glucocorticoids and IL-10.Blood 101:729–738

385. D’Adamio F, Zollo O, Moraca R, Ayroldi E, Bruscoli S,Bartoli A, Cannarile L, Migliorati G, Riccardi C 1997 Anew dexamethasone-induced gene of the leucine zipperfamily protects T lymphocytes from TCR/CD3-activatedcell death. Immunity 7:803–812

386. Mittelstadt PR, Ashwell JD 2001 Inhibition of AP-1 by theglucocorticoid-inducible protein GILZ. J Biol Chem 276:29603–29610

387. Galon J, Franchimont D, Hiroi N, Frey G, Boettner A,Ehrhart-Bornstein M, O’Shea JJ, Chrousos GP, BornsteinSR 2002 Gene profiling reveals unknown enhancing andsuppressive actions of glucocorticoids on immune cells.FASEB J 16:61–71

388. Tonko M, Ausserlechner MJ, Bernhard D, Helmberg A,Kofler R 2001 Gene expression profiles of proliferatingvs. G1/G0 arrested human leukemia cells suggest a mech-anism for glucocorticoid-induced apoptosis. FASEB J 15:693–699

389. Godot V, Garcia G, Capel F, Arock M, Durand-Gasselin I,Asselin-Labat ML, Emilie D, Humbert M 2006 Dexameth-asone and IL-10 stimulate glucocorticoid-induced leucinezipper synthesis by human mast cells. Allergy 61:886–890

390. Ayroldi E, Zollo O, Macchiarulo A, Di Marco B, Marchetti C,Riccardi C 2002 Glucocorticoid-induced leucine zipper in-hibits the Raf-extracellular signal-regulated kinase path-way by binding to Raf-1. Mol Cell Biol 22:7929–7941

391. Ayroldi E, Zollo O, Bastianelli A, Marchetti C, AgostiniM, Di Virgilio R, Riccardi C 2007 GILZ mediates the an-tiproliferative activity of glucocorticoids by negative reg-ulation of Ras signaling. J Clin Invest 117:1605–1615

392. Widen C, Zilliacus J, Gustafsson JA, Wikstrom AC 2000Glucocorticoid receptor interaction with 14-3-3 andRaf-1, a proposed mechanism for cross-talk of two signaltransduction pathways. J Biol Chem 275:39296–39301

393. Di Marco B, Massetti M, Bruscoli S, Macchiarulo A, DiVirgilio R, Velardi E, Donato V, Migliorati G, Riccardi C2007 Glucocorticoid-induced leucine zipper (GILZ)/NF-�B interaction: role of GILZ homo-dimerization andC-terminal domain. Nucleic Acids Res 35:517–528

394. Ayroldi E, Migliorati G, Bruscoli S, Marchetti C, Zollo O,Cannarile L, D’Adamio F, Riccardi C 2001 Modulation ofT-cell activation by the glucocorticoid-induced leucine zip-per factor via inhibition of nuclear factor �B. Blood 98:743–753

395. Hiragun T, Peng Z, Beaven MA 2005 Dexamethasone up-regulates the inhibitory adaptor protein Dok-1 and sup-presses downstream activation of the mitogen-activatedprotein kinase pathway in antigen-stimulated RBL-2H3mast cells. Mol Pharmacol 67:598–603

396. Hiragun T, Peng Z, Beaven MA 2006 Cutting edge: dexa-methasone negatively regulates Syk in mast cells by up-regulating SRC-like adaptor protein. J Immunol 177:2047–2050

397. Graham TE, Prossnitz ER, Dorin RI 2002 Dexras1/AGS-1inhibits signal transduction from the Gi-coupled formylpeptide receptor to Erk-1/2 MAP kinases. J Biol Chem 277:10876–10882

398. Kemppainen RJ, Behrend EN 1998 Dexamethasone rap-idly induces a novel ras superfamily member-related genein AtT-20 cells. J Biol Chem 273:3129–3131

399. Bhattacharyya S, Brown DE, Brewer JA, Vogt SK, MugliaLJ 2007 Macrophage glucocorticoid receptors regulateToll-like receptor 4-mediated inflammatory responses byselective inhibition of p38 MAP kinase. Blood 109:4313–4319

400. Clark AR, Martins JR, Tchen CR 2008 Role of dual spec-ificity phosphatases in biological responses to glucocorti-coids. J Biol Chem 283:25765–25769

401. Deak M, Clifton AD, Lucocq LM, Alessi DR 1998 Mitogen-and stress-activated protein kinase-1 (MSK1) is directly ac-tivated by MAPK and SAPK2/p38, and may mediate activa-tion of CREB. EMBO J 17:4426–4441

402. McCoy CE, Macdonald A, Morrice NA, Campbell DG,Deak M, Toth R, McIlrath J, Arthur JS 2007 Identificationof novel phosphorylation sites in MSK1 by precursor ionscanning MS. Biochem J 402:491–501

403. Tomas-Zuber M, Mary JL, Lamour F, Bur D, Lesslauer W2001 C-terminal elements control location, activationthreshold, and p38 docking of ribosomal S6 kinase B(RSKB). J Biol Chem 276:5892–5899

404. Dunn KL, Espino PS, Drobic B, He S, Davie JR 2005 TheRas-MAPK signal transduction pathway, cancer and chro-matin remodeling. Biochem Cell Biol 83:1–14

Endocrine Reviews, December 2009, 30(7):830–882 edrv.endojournals.org 871

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

405. Arthur JS 2008 MSK activation and physiological roles.Front Biosci 13:5866–5879

406. Darragh J, Soloaga A, Beardmore VA, Wingate AD, WigginGR, Peggie M, Arthur JS 2005 MSKs are required for thetranscription of the nuclear orphan receptors Nur77,Nurr1 and Nor1 downstream of MAPK signalling. Bio-chem J 390:749–759

407. Wiggin GR, Soloaga A, Foster JM, Murray-Tait V, CohenP, Arthur JS 2002 MSK1 and MSK2 are required for themitogen- and stress-induced phosphorylation of CREBand ATF1 in fibroblasts. Mol Cell Biol 22:2871–2881

408. Beardmore VA, Hinton HJ, Eftychi C, Apostolaki M,Armaka M, Darragh J, McIlrath J, Carr JM, Armit LJ,Clacher C, Malone L, Kollias G, Arthur JS 2005 Genera-tion and characterization of p38� (MAPK11) gene-tar-geted mice. Mol Cell Biol 25:10454–10464

409. McCoy CE, Campbell DG, Deak M, Bloomberg GB,Arthur JS 2005 MSK1 activity is controlled by multiplephosphorylation sites. Biochem J 387:507–517

410. Tomas-Zuber M, Mary JL, Lesslauer W 2000 Control sitesof ribosomal S6 kinase B and persistent activation throughtumor necrosis factor. J Biol Chem 275:23549–23558

411. Davie JR 2003 MSK1 and MSK2 mediate mitogen- andstress-induced phosphorylation of histone H3: a contro-versy resolved. Sci STKE 2003:PE33

412. Dunn KL, Davie JR 2005 Stimulation of the Ras-MAPKpathway leads to independent phosphorylation of histoneH3 on serine 10 and 28. Oncogene 24:3492–3502

413. Dyson MH, Thomson S, Inagaki M, Goto H, Arthur SJ,Nightingale K, Iborra FJ, Mahadevan LC 2005 MAP ki-nase-mediated phosphorylation of distinct pools of histoneH3 at S10 or S28 via mitogen- and stress-activated kinase1/2. J Cell Sci 118:2247–2259

414. Soloaga A, Thomson S, Wiggin GR, Rampersaud N,Dyson MH, Hazzalin CA, Mahadevan LC, Arthur JS 2003MSK2 and MSK1 mediate the mitogen- and stress-inducedphosphorylation of histone H3 and HMG-14. EMBO J22:2788–2797

415. Macdonald N, Welburn JP, Noble ME, Nguyen A, YaffeMB, Clynes D, Moggs JG, Orphanides G, Thomson S,Edmunds JW, Clayton AL, Endicott JA, Mahadevan LC2005 Molecular basis for the recognition of phosphory-lated and phosphoacetylated histone h3 by 14-3-3. MolCell 20:199–211

416. Winter S, Fischle W, Seiser C 2008 Modulation of 14-3-3interaction with phosphorylated histone H3 by combina-torial modification patterns. Cell Cycle 7:1336–1342

417. Winter S, Simboeck E, Fischle W, Zupkovitz G, Dohnal I,Mechtler K, Ammerer G, Seiser C 2008 14-3-3 proteinsrecognize a histone code at histone H3 and are required fortranscriptional activation. EMBO J 27:88–99

418. Walter W, Clynes D, Tang Y, Marmorstein R, Mellor J,Berger SL 2008 14-3-3 interaction with histone H3 in-volves a dual modification pattern of phosphoacetylation.Mol Cell Biol 28:2840–2849

419. Vicent GP, Ballare C, Nacht AS, Clausell J, Subtil-Rodríguez A, Quiles I, Jordan A, Beato M 2006 Inductionof progesterone target genes requires activation of Erk andMsk kinases and phosphorylation of histone H3. Mol Cell24:367–381

420. Beck IM, Vanden Berghe W, Vermeulen L, Bougarne N,Vander Cruyssen B, Haegeman G, De Bosscher K 2008

Altered subcellular distribution of MSK1 induced by glu-cocorticoids contributes to NF-�B inhibition. EMBO J 27:1682–1693

421. Hasegawa Y, Tomita K, Watanabe M, Yamasaki A, SanoH, Hitsuda Y, Shimizu E 2005 Dexamethasone inhibitsphosphorylation of histone H3 at serine 10. Biochem Bio-phys Res Commun 336:1049–1055

422. Dewint P, Gossye V, De Bosscher K, Vanden Berghe W,Van Beneden K, Deforce D, Van Calenbergh S, Muller-Ladner U, Vander Cruyssen B, Verbruggen G, HaegemanG, Elewaut D 2008 A plant-derived ligand favoring mo-nomeric glucocorticoid receptor conformation with im-paired transactivation potential attenuates collagen-in-duced arthritis. J Immunol 180:2608–2615

423. Liu G, Zhang Y, Bode AM, Ma WY, Dong Z 2002 Phos-phorylation of 4E-BP1 is mediated by the p38/MSK1 path-way in response to UVB irradiation. J Biol Chem 277:8810–8816

424. Otulakowski G, Duan W, Gandhi S, O’brodovich H 2007Steroid and oxygen effects on eIF4F complex, mTOR, andENaC translation in fetal lung epithelia. Am J Respir CellMol Biol 37:457–466

425. Liu Z, Jahn LA, Long W, Fryburg DA, Wei L, Barrett EJ2001 Branched chain amino acids activate messenger ri-bonucleic acid translation regulatory proteins in humanskeletal muscle, and glucocorticoids blunt this action.J Clin Endocrinol Metab 86:2136–2143

426. Liu Z, Li G, Kimball SR, Jahn LA, Barrett EJ 2004 Glu-cocorticoids modulate amino acid-induced translation ini-tiation in human skeletal muscle. Am J Physiol EndocrinolMetab 287:E275–E281

427. Shah OJ, Iniguez-Lluhi JA, Romanelli A, Kimball SR,Jefferson LS 2002 The activated glucocorticoid receptormodulates presumptive autoregulation of ribosomal pro-tein S6 protein kinase, p70 S6K. J Biol Chem 277:2525–2533

428. Shah OJ, Kimball SR, Jefferson LS 2000 Glucocorticoidsabate p70(S6k) and eIF4E function in L6 skeletal myo-blasts. Am J Physiol Endocrinol Metab 279:E74–E82

429. Dong C, Davis RJ, Flavell RA 2002 MAP kinases in theimmune response. Annu Rev Immunol 20:55–72

430. Carballo E, Lai WS, Blackshear PJ 1998 Feedback inhibi-tion of macrophage tumor necrosis factor-� production bytristetraprolin. Science 281:1001–1005

431. Carrick DM, Lai WS, Blackshear PJ 2004 The tandemCCCH zinc finger protein tristetraprolin and its relevanceto cytokine mRNA turnover and arthritis. Arthritis ResTher 6:248–264

432. Carballo E, Cao H, Lai WS, Kennington EA, Campbell D,Blackshear PJ 2001 Decreased sensitivity of tristetrapro-lin-deficient cells to p38 inhibitors suggests the involve-ment of tristetraprolin in the p38 signaling pathway. J BiolChem 276:42580–42587

433. Chrestensen CA, Schroeder MJ, Shabanowitz J, Hunt DF,Pelo JW, Worthington MT, Sturgill TW 2004 MAPKAPkinase 2 phosphorylates tristetraprolin on in vivo sites in-cluding Ser178, a site required for 14-3-3 binding. J BiolChem 279:10176–10184

434. Hitti E, Iakovleva T, Brook M, Deppenmeier S, GruberAD, Radzioch D, Clark AR, Blackshear PJ, Kotlyarov A,Gaestel M 2006 Mitogen-activated protein kinase-acti-vated protein kinase 2 regulates tumor necrosis factor

872 Beck et al. GR Crosstalk with Kinases and Phosphatases Endocrine Reviews, December 2009, 30(7):830–882

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

mRNA stability and translation mainly by altering tristet-raprolin expression, stability, and binding to adenine/uri-dine-rich element. Mol Cell Biol 26:2399–2407

435. Brook M, Tchen CR, Santalucia T, McIlrath J, Arthur JS,Saklatvala J, Clark AR 2006 Posttranslational regulationof tristetraprolin subcellular localization and protein sta-bility by p38 mitogen-activated protein kinase and extra-cellular signal-regulated kinase pathways. Mol Cell Biol26:2408–2418

436. Neininger A, Kontoyiannis D, Kotlyarov A, Winzen R,Eckert R, Volk HD, Holtmann H, Kollias G, Gaestel M2002 MK2 targets AU-rich elements and regulates biosyn-thesis of tumor necrosis factor and interleukin-6 indepen-dently at different post-transcriptional levels. J Biol Chem277:3065–3068

437. Stoecklin G, Stubbs T, Kedersha N, Wax S, Rigby WF,Blackwell TK, Anderson P 2004 MK2-induced tristetra-prolin:14-3-3 complexes prevent stress granule associationand ARE-mRNA decay. EMBO J 23:1313–1324

438. Dean JL, Sarsfield SJ, Tsounakou E, Saklatvala J 2003 p38Mitogen-activated protein kinase stabilizes mRNAs thatcontain cyclooxygenase-2 and tumor necrosis factor AU-rich elements by inhibiting deadenylation. J Biol Chem278:39470–39476

439. Taylor GA, Carballo E, Lee DM, Lai WS, Thompson MJ,Patel DD, Schenkman DI, Gilkeson GS, Broxmeyer HE,Haynes BF, Blackshear PJ 1996 A pathogenetic role forTNF � in the syndrome of cachexia, arthritis, and auto-immunity resulting from tristetraprolin (TTP) deficiency.Immunity 4:445–454

440. Stojadinovic O, Lee B, Vouthounis C, Vukelic S, Pastar I,Blumenberg M, Brem H, Tomic-Canic M 2007 Novelgenomic effects of glucocorticoids in epidermal keratino-cytes: inhibition of apoptosis, interferon-� pathway, andwound healing along with promotion of terminal differ-entiation. J Biol Chem 282:4021–4034

441. Lasa M, Abraham SM, Boucheron C, Saklatvala J, ClarkAR 2002 Dexamethasone causes sustained expression ofmitogen-activated protein kinase (MAPK) phosphatase 1and phosphatase-mediated inhibition of MAPK p38. MolCell Biol 22:7802–7811

442. Lasa M, Brook M, Saklatvala J, Clark AR 2001 Dexa-methasone destabilizes cyclooxygenase 2 mRNA by inhib-iting mitogen-activated protein kinase p38. Mol Cell Biol21:771–780

443. Stellato C 2004 Post-transcriptional and non-genomic ef-fects of glucocorticoids. Proc Am Thorac Soc 1:255–263

444. Chen CY, Del Gatto-Konczak F, Wu Z, Karin M 1998Stabilization of interleukin-2 mRNA by the c-Jun NH2-terminal kinase pathway. Science 280:1945–1949

445. Dumitru CD, Ceci JD, Tsatsanis C, Kontoyiannis D,Stamatakis K, Lin JH, Patriotis C, Jenkins NA, CopelandNG, Kollias G, Tsichlis PN 2000 TNF-� induction by LPSis regulated posttranscriptionally via a Tpl2/ERK-depen-dent pathway. Cell 103:1071–1083

446. Ming XF, Kaiser M, Moroni C 1998 c-jun N-terminal ki-nase is involved in AUUUA-mediated interleukin-3 mRNAturnover in mast cells. EMBO J 17:6039–6048

447. Schaaf MJ, Cidlowski JA 2003 Molecular determinants ofglucocorticoid receptor mobility in living cells: the impor-tance of ligand affinity. Mol Cell Biol 23:1922–1934

448. Price DH 2000 P-TEFb, a cyclin-dependent kinase con-

trolling elongation by RNA polymerase II. Mol Cell Biol20:2629–2634

449. Barboric M, Nissen RM, Kanazawa S, Jabrane-Ferrat N,Peterlin BM 2001 NF-�B binds P-TEFb to stimulate tran-scriptional elongation by RNA polymerase II. Mol Cell8:327–337

450. Rogatsky I, Trowbridge JM, Garabedian MJ 1997 Glu-cocorticoid receptor-mediated cell cycle arrest is achievedthrough distinct cell-specific transcriptional regulatorymechanisms. Mol Cell Biol 17:3181–3193

451. Cha HH, Cram EJ, Wang EC, Huang AJ, Kasler HG,Firestone GL 1998 Glucocorticoids stimulate p21 gene ex-pression by targeting multiple transcriptional elementswithina steroid responsive regionof thep21waf1/cip1pro-moter in rat hepatoma cells. J Biol Chem 273:1998–2007

452. Cram EJ, Ramos RA, Wang EC, Cha HH, Nishio Y,Firestone GL 1998 Role of the CCAAT/enhancer bindingprotein-� transcription factor in the glucocorticoid stim-ulation of p21waf1/cip1 gene promoter activity in growth-arrested rat hepatoma cells. J Biol Chem 273:2008–2014

453. Greenberg AK, Hu J, Basu S, Hay J, Reibman J, Yie TA,Tchou-Wong KM, Rom WN, Lee TC 2002 Glucocorti-coids inhibit lung cancer cell growth through both the ex-tracellular signal-related kinase pathway and cell cycle reg-ulators. Am J Respir Cell Mol Biol 27:320–328

454. Bhattacharjee RN, Banks GC, Trotter KW, Lee HL, ArcherTK 2001 Histone H1 phosphorylation by Cdk2 selectivelymodulates mouse mammary tumor virus transcriptionthrough chromatin remodeling. Mol Cell Biol 21:5417–5425

455. Banks GC, Deterding LJ, Tomer KB, Archer TK 2001Hormone-mediated dephosphorylation of specific histoneH1 isoforms. J Biol Chem 276:36467–36473

456. Zoja C, Casiraghi F, Conti S, Corna D, Rottoli D, CavinatoRA, Remuzzi G, Benigni A 2007 Cyclin-dependent kinaseinhibition limits glomerulonephritis and extends lifespanof mice with systemic lupus. Arthritis Rheum 56:1629–1637

457. Gloire G, Horion J, El Mjiyad N, Bex F, Chariot A,Dejardin E, Piette J 2007 Promoter-dependent effect ofIKK� on NF-�B/p65 DNA binding. J Biol Chem282:21308–21318

458. Anest V, Hanson JL, Cogswell PC, Steinbrecher KA, StrahlBD, Baldwin AS 2003 A nucleosomal function for I�Bkinase-� in NF-�B-dependent gene expression. Nature423:659–663

459. Yamamoto Y, Verma UN, Prajapati S, Kwak YT, GaynorRB 2003 Histone H3 phosphorylation by IKK-� is crit-ical for cytokine-induced gene expression. Nature 423:655– 659

460. Park GY, Wang X, Hu N, Pedchenko TV, Blackwell TS,Christman JW 2006 NIK is involved in nucleosomal reg-ulation by enhancing histone H3 phosphorylation byIKK�. J Biol Chem 281:18684–18690

461. Li Q, Lu Q, Hwang JY, Buscher D, Lee KF, Izpisua-Belmonte JC, Verma IM 1999 IKK1-deficient mice exhibitabnormal development of skin and skeleton. Genes Dev13:1322–1328

462. Sizemore N, Lerner N, Dombrowski N, Sakurai H, StarkGR 2002 Distinct roles of the I� B kinase � and � subunitsin liberating nuclear factor � B (NF-� B) from I� B and in

Endocrine Reviews, December 2009, 30(7):830–882 edrv.endojournals.org 873

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

phosphorylating the p65 subunit of NF-�B. J Biol Chem277:3863–3869

463. McCoy CE, Carpenter S, Pålsson-McDermott EM, GearingLJ, O’Neill LA 2008 Glucocorticoids inhibit IRF3 phos-phorylation in response to Toll-like receptor-3 and -4 bytargeting TBK1 activation. J Biol Chem 283:14277–14285

464. McWhirter SM, Fitzgerald KA, Rosains J, Rowe DC,Golenbock DT, Maniatis T 2004 IFN-regulatory factor3-dependent gene expression is defective in Tbk1-deficientmouse embryonic fibroblasts. Proc Natl Acad Sci USA 101:233–238

465. Hemmi H, Takeuchi O, Sato S, Yamamoto M, Kaisho T,Sanjo H, Kawai T, Hoshino K, Takeda K, Akira S 2004The roles of two I�B kinase-related kinases in lipopolysac-charide and double stranded RNA signaling and viral in-fection. J Exp Med 199:1641–1650

466. Honda K, Taniguchi T 2006 IRFs: master regulators of sig-nalling by Toll-like receptors and cytosolic pattern-recogni-tion receptors. Nat Rev Immunol 6:644–658

467. Servant MJ, Grandvaux N, tenOever BR, Duguay D, LinR, Hiscott J 2003 Identification of the minimal phosphoac-ceptor site required for in vivo activation of interferon reg-ulatory factor 3 in response to virus and double-strandedRNA. J Biol Chem 278:9441–9447

468. An H, Zhao W, Hou J, Zhang Y, Xie Y, Zheng Y, Xu H,Qian C, Zhou J, Yu Y, Liu S, Feng G, Cao X 2006 SHP-2phosphatase negatively regulates the TRIF adaptor pro-tein-dependent type I interferon and proinflammatory cy-tokine production. Immunity 25:919–928

469. Reily MM, Pantoja C, Hu X, Chinenov Y, Rogatsky I2006 The GRIP1:IRF3 interaction asa target forglucocor-ticoid receptor-mediated immunosuppression. EMBO J 25:108–117

470. Buss H, Dorrie A, Schmitz ML, Hoffmann E, Resch K,Kracht M 2004 Constitutive and interleukin-1-induciblephosphorylation of p65 NF-�B at serine 536 is mediated bymultiple protein kinases including I�B kinase (IKK)-�,IKK�, IKK�, TRAF family member-associated (TANK)-binding kinase 1 (TBK1), and an unknown kinase and cou-ples p65 to TATA-binding protein-associated factor II31-mediated interleukin-8 transcription. J Biol Chem 279:55633–55643

471. Bianchi M, Meng C, Ivashkiv LB 2000 Inhibition of IL-2-induced Jak-STAT signaling by glucocorticoids. Proc NatlAcad Sci USA 97:9573–9578

472. Franchimont D, Galon J, Gadina M, Visconti R, Zhou Y,Aringer M, Frucht DM, Chrousos GP, O’Shea JJ 2000Inhibition of Th1 immune response by glucocorticoids:dexamethasone selectively inhibits IL-12-induced Stat4phosphorylation in T lymphocytes. J Immunol 164:1768–1774

473. Fahey AJ, Robins RA, Kindle KB, Heery DM, ConstantinescuCS 2006 Effects of glucocorticoids on STAT4 activation inhuman T cells are stimulus-dependent. J Leukoc Biol 80:133–144

474. Dempsey EC, Cool CD, Littler CM 2007 Lung disease andPKCs. Pharmacol Res 55:545–559

475. Catley MC, Cambridge LM, Nasuhara Y, Ito K, ChiversJE, Beaton A, Holden NS, Bergmann MW, Barnes PJ,Newton R 2004 Inhibitors of protein kinase C (PKC) pre-vent activated transcription: role of events downstream ofNF-�B DNA binding. J Biol Chem 279:18457–18466

476. Page K, Li J, Zhou L, Iasvovskaia S, Corbit KC, Soh JW,Weinstein IB, Brasier AR, Lin A, Hershenson MB,Iasvoyskaia S 2003 Regulation of airway epithelial cellNF-�B-dependent gene expression by protein kinase C �.J Immunol 170:5681–5689

477. Aras-Lopez R, Xavier FE, Ferrer M, Balfagon G 2009Dexamethasone decreases neuronal nitric oxide release inmesenteric arteries from hypertensive rats through de-creased protein kinase C activation. Clin Sci (Lond) 117:305–312

478. Nguyen CH, Watts VJ 2006 Dexamethasone-induced Rasprotein 1 negatively regulates protein kinase C�: implica-tions for adenylyl cyclase 2 signaling. Mol Pharmacol 69:1763–1771

479. Matthews L, Berry A, Ohanian V, Ohanian J, Garside H,Ray D 2008 Caveolin mediates rapid glucocorticoid effectsand couples glucocorticoid action to the antiproliferativeprogram. Mol Endocrinol 22:1320–1330

480. Failor KL, Desyatnikov Y, Finger LA, Firestone GL 2007Glucocorticoid-induced degradation of glycogen synthasekinase-3 protein is triggered by serum- and glucocorticoid-induced protein kinase and Akt signaling and controls�-catenin dynamics and tight junction formation in mam-mary epithelial tumor cells. Mol Endocrinol 21:2403–2415

481. Hafezi-Moghadam A, Simoncini T, Yang Z, Limbourg FP,Plumier JC, Rebsamen MC, Hsieh CM, Chui DS, ThomasKL, Prorock AJ, Laubach VE, Moskowitz MA, French BA,Ley K, Liao JK 2002 Acute cardiovascular protective ef-fects of corticosteroids are mediated by non-transcrip-tional activation of endothelial nitric oxide synthase. NatMed 8:473–479

482. Limbourg FP, Huang Z, Plumier JC, Simoncini T, FujiokaM, Tuckermann J, Schutz G, Moskowitz MA, Liao JK2002 Rapid nontranscriptional activation of endothelialnitric oxide synthase mediates increased cerebral bloodflow and stroke protection by corticosteroids. J Clin Invest110:1729–1738

483. Langdown ML, Holness MJ, Sugden MC 2001 Earlygrowth retardation induced by excessive exposure to glu-cocorticoids in utero selectively increases cardiac GLUT1protein expression and Akt/protein kinase B activity inadulthood. J Endocrinol 169:11–22

484. Limbourg FP, Liao JK 2003 Nontranscriptional actions ofthe glucocorticoid receptor. J Mol Med 81:168–174

485. Smith E, Frenkel B 2005 Glucocorticoids inhibit the tran-scriptional activity of LEF/TCF in differentiating osteoblastsin a glycogen synthase kinase-3�-dependent and -indepen-dent manner. J Biol Chem 280:2388–2394

486. Wu W, Zou M, Brickley DR, Pew T, Conzen SD 2006Glucocorticoid receptor activation signals through fork-head transcription factor 3a in breast cancer cells. MolEndocrinol 20:2304–2314

487. Itani OA, Liu KZ, Cornish KL, Campbell JR, Thomas CP2002 Glucocorticoids stimulate human sgk1 gene expres-sion by activation of a GRE in its 5�-flanking region. Am JPhysiol Endocrinol Metab 283:E971–E979

488. Mikosz CA, Brickley DR, Sharkey MS, Moran TW,Conzen SD 2001 Glucocorticoid receptor-mediated pro-tection from apoptosis is associated with induction of theserine/threonine survival kinase gene, sgk-1. J Biol Chem276:16649–16654

489. Zhang L, Cui R, Cheng X, Du J 2005 Antiapoptotic effect

874 Beck et al. GR Crosstalk with Kinases and Phosphatases Endocrine Reviews, December 2009, 30(7):830–882

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

of serum and glucocorticoid-inducible protein kinase ismediated by novel mechanism activating I�B kinase. Can-cer Res 65:457–464

490. Tai DJ, Su CC, Ma YL, Lee EH 2009 SGK1 phosphory-lation of I�B kinase � and p300 up-regulates NF-�B ac-tivity and increases N-methyl-D-aspartate receptor NR2Aand NR2B expression. J Biol Chem 284:4073–4089

491. Abu-Amer Y, Ross FP, McHugh KP, Livolsi A, Peyron JF,Teitelbaum SL 1998 Tumor necrosis factor-� activation ofnuclear transcription factor-�B in marrow macrophages ismediated by c-Src tyrosine phosphorylation of I� B�. J BiolChem 273:29417–29423

492. Wong WS 2005 Inhibitors of the tyrosine kinase signalingcascade for asthma. Curr Opin Pharmacol 5:264–271

493. LowenbergM,TuynmanJ,BilderbeekJ,GaberT,ButtgereitF,van Deventer S, Peppelenbosch M, Hommes D 2005 Rapidimmunosuppressive effects of glucocorticoids mediatedthrough Lck and Fyn. Blood 106:1703–1710

494. Lowenberg M, Tuynman J, Scheffer M, Verhaar A,Vermeulen L, van Deventer S, Hommes D, PeppelenboschM 2006 Kinome analysis reveals non-genomic glucocor-ticoid receptor-dependent inhibition of insulin signaling.Endocrinology 147:3555–3562

495. Lowenberg M, Verhaar AP, Bilderbeek J, Marle J, Buttgereit F,Peppelenbosch MP, van Deventer SJ, Hommes DW 2006Glucocorticoids cause rapid dissociation of a T-cell-recep-tor-associated protein complex containing LCK and FYN.EMBO Rep 7:1023–1029

496. Ghosh MC, Baatar D, Collins G, Carter A, Indig F, BiragynA, Taub DD 2009 Dexamethasone augments CXCR4-me-diated signaling in resting human T cells via the activationof the Src kinase Lck. Blood 113:575–584

497. Rubenstein NM, Callahan JA, Lo DH, Firestone GL 2007Selective glucocorticoid control of Rho kinase isoformsregulate cell-cell interactions. Biochem Biophys Res Com-mun 354:603–607

498. Mong PY, Wang Q 2009 Activation of Rho kinase iso-forms in lung endothelial cells during inflammation. J Im-munol 182:2385–2394

499. Goto K, Chiba Y, Sakai H, Misawa M 2008 Glucocorti-coids inhibited airway hyperresponsiveness throughdownregulation of CPI-17 in bronchial smooth muscle.Eur J Pharmacol 591:231–236

500. Barford D 1996 Molecular mechanisms of the proteinserine/threonine phosphatases. Trends Biochem Sci 21:407–412

501. Camps M, Nichols A, Arkinstall S 2000 Dual specificityphosphatases: a gene family for control of MAP kinasefunction. FASEB J 14:6–16

502. Abraham SM, Clark AR 2006 Dual-specificity phospha-tase 1: a critical regulator of innate immune responses.Biochem Soc Trans 34:1018–1023

503. Turjanski AG, Vaque JP, Gutkind JS 2007 MAP kinasesand the control of nuclear events. Oncogene 26:3240-3253

504. Dickinson RJ, Keyse SM 2006 Diverse physiological func-tions for dual-specificity MAP kinase phosphatases. J CellSci 119:4607–4615

505. Kassel O, Sancono A, Kratzschmar J, Kreft B, Stassen M,Cato AC 2001 Glucocorticoids inhibit MAP kinase viaincreased expression and decreased degradation ofMKP-1. EMBO J 20:7108–7116

506. Adams M, Meijer OC, Wang J, Bhargava A, Pearce D 2003Homodimerization of the glucocorticoid receptor is notessential for response element binding: activation of thephenylethanolamine N-methyltransferase gene by dimer-ization-defective mutants. Mol Endocrinol 17:2583–2592

507. Iniguez-Lluhí JA, Pearce D 2000 A common motif withinthe negative regulatory regions of multiple factors inhibitstheir transcriptional synergy. Mol Cell Biol 20:6040-6050

508. Helmberg A, Auphan N, Caelles C, Karin M 1995 Glu-cocorticoid-induced apoptosis of human leukemic cells iscaused by the repressive function of the glucocorticoid re-ceptor. EMBO J 14:452–460

509. Johansson-Haque K, Palanichamy E, Okret S 2008 Stim-ulation of MAPK-phosphatase 1 gene expression by glu-cocorticoids occurs through a tethering mechanism involv-ing C/EBP. J Mol Endocrinol 41:239–249

510. Furst R, Zahler S, Vollmar AM 2008 Dexamethasone-in-duced expression of endothelial mitogen-activated proteinkinase phosphatase-1 involves activation of the transcrip-tion factors activator protein-1 and 3�,5�-cyclic adenosine5�-monophosphate response element-binding protein andthe generation of reactive oxygen species. Endocrinology149:3635–3642

511. Berg T, Didon L, Barton J, Andersson O, Nord M 2005Glucocorticoids increase C/EBP� activity in the lung epi-thelium via phosphorylation. Biochem Biophys Res Com-mun 334:638–645

512. Bazuine M, Carlotti F, Tafrechi RS, Hoeben RC, MaassenJA 2004 Mitogen-activated protein kinase (MAPK) phos-phatase-1 and -4 attenuate p38 MAPK during dexameth-asone-induced insulin resistance in 3T3–L1 adipocytes.Mol Endocrinol 18:1697–1707

513. Yoshida NL, Miyashita T, U M, Yamada M, Reed JC,Sugita Y, Oshida T 2002 Analysis of gene expression pat-terns during glucocorticoid-induced apoptosis using oli-gonucleotide arrays. Biochem Biophys Res Commun 293:1254–1261

514. Sanchez-Tillo E, Comalada M, Xaus J, Farrera C, ValledorAF, Caelles C, Lloberas J, Celada A 2007 JNK1 is requiredfor the induction of Mkp1 expression in macrophages dur-ing proliferation and lipopolysaccharide-dependent acti-vation. J Biol Chem 282:12566–12573

515. Ananieva O, Darragh J, Johansen C, Carr JM, McIlrath J,Park JM,WingateA,MonkCE,TothR,SantosSG, IversenL, Arthur JS 2008 The kinases MSK1 and MSK2 act asnegative regulators of Toll-like receptor signaling. Nat Im-munol 9:1028–1036

516. Hammer M, Mages J, Dietrich H, Schmitz F, Striebel F,Murray PJ, Wagner H, Lang R 2005 Control of dual-spec-ificity phosphatase-1 expression in activated macrophagesby IL-10. Eur J Immunol 35:2991–3001

517. Cho IJ, Kim SG 2009 A novel mitogen-activated proteinkinase phosphatase-1 and glucocorticoid receptor (GR) in-teracting protein-1-dependent combinatorial mechanismof gene transrepression by GR. Mol Endocrinol 23:86–99

518. Chauhan S, Leach CH, Kunz S, Bloom JW, Miesfeld RL2003 Glucocorticoid regulation of human eosinophil geneexpression. J Steroid Biochem Mol Biol 84:441–452

519. Schmidt S, Rainer J, Riml S, Ploner C, Jesacher S, AchmullerC, Presul E, Skvortsov S, Crazzolara R, Fiegl M, Raivio T,Janne OA, Geley S, Meister B, Kofler R 2006 Identification

Endocrine Reviews, December 2009, 30(7):830–882 edrv.endojournals.org 875

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

of glucocorticoid-response genes in children with acutelymphoblastic leukemia. Blood 107:2061–2069

520. Haffner MC, Jurgeit A, Berlato C, Geley S, Parajuli N,Yoshimura A, Doppler W 2008 Interaction and functionalinterference of glucocorticoid receptor and SOCS1. J BiolChem 283:22089–22096

521. Mansell A, Smith R, Doyle SL, Gray P, Fenner JE, Crack PJ,Nicholson SE, Hilton DJ, O’Neill LA, Hertzog PJ 2006Suppressor of cytokine signaling 1 negatively regulatesToll-like receptor signaling by mediating Mal degradation.Nat Immunol 7:148–155

522. Wang X, Liu Y 2007 Regulation of innate immune re-sponse by MAP kinase phosphatase-1. Cell Signal19:1372–1382

523. Chi H, Barry SP, Roth RJ, Wu JJ, Jones EA, Bennett AM,Flavell RA 2006 Dynamic regulation of pro- and anti-in-flammatory cytokines by MAPK phosphatase 1 (MKP-1)in innate immune responses. Proc Natl Acad Sci USA 103:2274–2279

524. Hammer M, Mages J, Dietrich H, Servatius A, Howells N,Cato AC, Lang R 2006 Dual specificity phosphatase 1(DUSP1) regulates a subset of LPS-induced genes and pro-tects mice from lethal endotoxin shock. J Exp Med 203:15–20

525. Salojin KV, Owusu IB, Millerchip KA, Potter M, Platt KA,Oravecz T 2006 Essential role of MAPK phosphatase-1 inthe negative control of innate immune responses. J Immu-nol 176:1899–1907

526. Zhao Q, Wang X, Nelin LD, Yao Y, Matta R, Manson ME,Baliga RS, Meng X, Smith CV, Bauer JA, Chang CH, LiuY 2006 MAP kinase phosphatase 1 controls innate im-mune responses and suppresses endotoxic shock. J ExpMed 203:131–140

527. Brondello JM, Pouyssegur J, McKenzie FR 1999 ReducedMAP kinase phosphatase-1 degradation after p42/p44MAPK-dependent phosphorylation. Science 286:2514–2517

528. Lin YW, Chuang SM, Yang JL 2003 ERK1/2 achieves sus-tained activation by stimulating MAPK phosphatase-1degradation via the ubiquitin-proteasome pathway. J BiolChem 278:21534–21541

529. Lin YW, Yang JL 2006 Cooperation of ERK and SCFSkp2for MKP-1 destruction provides a positive feedback regu-lation of proliferating signaling. J Biol Chem 281:915–926

530. Cao W, Bao C, Padalko E, Lowenstein CJ 2008 Acetyla-tion of mitogen-activated protein kinase phosphatase-1 in-hibits Toll-like receptor signaling. J Exp Med 205:1491–1503

531. Chi H, Flavell RA 2008 Acetylation of MKP-1 and thecontrol of inflammation. Sci Signal 1:pe44

532. Pelegrí C, Franch A, Castellote C, Castell M 1995 Immu-nohistochemical changes in synovial tissue during thecourse of adjuvant arthritis. J Rheumatol 22:124–132

533. Nielsen H, Petersen AA, Skjødt H, Hørslev-Petersen K,Bendtzen K 1999 Blood levels of CD11b� memory T lym-phocytes are selectively upregulated in patients with activerheumatoid arthritis. APMIS 107:1124–1130

534. Cambillau C, Rauly I, Sarfati P, Saint-Laurent N, Esteve JP,Fanjul M, Svoboda M, Prats H, Hollande E, Vaysse N1995 Regulation of the src homology 2 domain-containingprotein tyrosine phosphatase PTP1C by glucocorticoids in

rat pancreatic AR42J cells. Endocrinology 136:5476–5484

535. Pani G, Fischer KD, Mlinaric-Rascan I, Siminovitch KA1996 Signaling capacity of the T cell antigen receptor isnegatively regulated by the PTP1C tyrosine phosphatase. JExp Med 184:839–852

536. Croker BA, Lawson BR, Rutschmann S, Berger M,EidenschenkC,BlasiusAL,MorescoEM,SovathS,CengiaL, Shultz LD, Theofilopoulos AN, Pettersson S, Beutler BA2008 Inflammation and autoimmunity caused by a SHP1mutation depend on IL-1, MyD88, and a microbial trigger.Proc Natl Acad Sci USA 105:15028–15033

537. Virshup DM, Shenolikar S 2009 From promiscuity to pre-cision: protein phosphatases get a makeover. Mol Cell 33:537–545

538. Boudreau RT, Hoskin DW, Lin TJ 2004 Phosphatase in-hibition potentiates IL-6 production by mast cells in re-sponse to Fc�RI-mediated activation: involvement of p38MAPK. J Leukoc Biol 76:1075–1081

539. Kins S, Kurosinski P, Nitsch RM, Gotz J 2003 Activationof the ERK and JNK signaling pathways caused by neuron-specific inhibition of PP2A in transgenic mice. Am J Pathol163:833–843

540. Millward TA, Zolnierowicz S, Hemmings BA 1999 Reg-ulation of protein kinase cascades by protein phosphatase2A. Trends Biochem Sci 24:186–191

541. Silverstein AM, Barrow CA, Davis AJ, Mumby MC 2002Actions of PP2A on the MAP kinase pathway and apopto-sis are mediated by distinct regulatory subunits. Proc NatlAcad Sci USA 99:4221–4226

542. SonodaY,KasaharaT,YamaguchiY,KunoK,MatsushimaK,Mukaida N 1997 Stimulation of interleukin-8 productionby okadaic acid and vanadate in a human promyelocytecell line, an HL-60 subline. Possible role of mitogen-acti-vated protein kinase on the okadaic acid-induced NF-�Bactivation. J Biol Chem 272:15366–15372

543. Shanley TP, Vasi N, Denenberg A, Wong HR 2001 Theserine/threonine phosphatase, PP2A: endogenous regu-lator of inflammatory cell signaling. J Immunol 166:966 –972

544. Amaral MC, Casillas AM, Nel AE 1993 Contrasting ef-fects of two tumour promoters, phorbol myristate acetateand okadaic acid, on T-cell responses and activation of p42MAP-kinase/ERK-2. Immunology 79:24–31

545. Nowak SJ, Pai CY, Corces VG 2003 Protein phosphatase2A activity affects histone H3 phosphorylation and tran-scription in Drosophila melanogaster. Mol Cell Biol 23:6129–6138

546. Kang YJ, Kusler B, Otsuka M, Hughes M, Suzuki N, SuzukiS, Yeh WC, Akira S, Han J, Jones PP 2007 Calcineurinnegatively regulates TLR-mediated activation pathways.J Immunol 179:4598–4607

547. Braun WE 2003 Renal transplantation: basic concepts andevolution of therapy. J Clin Apher 18:141–152

548. Kitahara K, Kawai S 2007 Cyclosporine and tacrolimus forthe treatment of rheumatoid arthritis. Curr Opin Rheu-matol 19:238–245

549. Tong DC, Buck SM, Roberts BR, Klein JD, Tumlin JA2004 Calcineurin phosphatase activity: activation by glu-cocorticoids and role of intracellular calcium. Transplan-tation 77:259–267

550. Ranta F, Avram D, Berchtold S, Dufer M, Drews G, Lang

876 Beck et al. GR Crosstalk with Kinases and Phosphatases Endocrine Reviews, December 2009, 30(7):830–882

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

F, Ullrich S 2006 Dexamethasone induces cell death ininsulin-secreting cells, an effect reversed by exendin-4. Di-abetes 55:1380–1390

551. Ranta F, Dufer M, Stork B, Wesselborg S, Drews G, HaringHU, Lang F, Ullrich S 2008 Regulation of calcineurinactivity in insulin-secreting cells: stimulation by Hsp90during glucocorticoid-induced apoptosis. Cell Signal20:1780 –1786

552. Lumbers ER, Boyce AC, Joulianos G, Kumarasamy V,Barner E, Segar JL, Burrell JH 2005 Effects of cortisol oncardiac myocytes and on expression of cardiac genes infetal sheep. Am J Physiol Regul Integr Comp Physiol 288:R567–R574

553. McDonough AK, Curtis JR, Saag KG 2008 The epidemi-ology of glucocorticoid-associated adverse events. CurrOpin Rheumatol 20:131–137

554. Schacke H, Docke WD, Asadullah K 2002 Mechanismsinvolved in the side effects of glucocorticoids. PharmacolTher 96:23–43

555. Vega D, Maalouf NM, Sakhaee K 2007 Clinical Review:the role of receptor activator of nuclear factor-�B (RANK)/RANK ligand/osteoprotegerin: clinical implications. J ClinEndocrinol Metab 92:4514–4521

556. Cooper MS 2004 Sensitivity of bone to glucocorticoids.Clin Sci (Lond) 107:111–123

557. Li H, Cuartas E, Cui W, Choi Y, Crawford TD, Ke HZ,Kobayashi KS, Flavell RA, Vignery A 2005 IL-1 receptor-associated kinase M is a central regulator of osteoclastdifferentiation and activation. J Exp Med 201:1169–1177

558. Soares-Schanoski A, Gomez-Pina V, del Fresno C,Rodríguez-Rojas A, García F, Glaría A, Sanchez M, Vallejo-Cremades MT, Baos R, Fuentes-Prior P, Arnalich F,Lopez-Collazo E 2007 6-Methylprednisolone down-regu-lates IRAK-M in human and murine osteoclasts and boostsbone-resorbing activity: a putative mechanism for corti-coid-induced osteoporosis. J Leukoc Biol 82:700–709

559. Horsch K, de Wet H, Schuurmans MM, Allie-Reid F, CatoAC, Cunningham J, Burrin JM, Hough FS, Hulley PA 2007Mitogen-activated protein kinase phosphatase 1/dualspecificity phosphatase 1 mediates glucocorticoid inhibi-tion of osteoblast proliferation. Mol Endocrinol 21:2929–2940

560. Engelbrecht Y, de Wet H, Horsch K, Langeveldt CR,Hough FS, Hulley PA 2003 Glucocorticoids induce rapidup-regulation of mitogen-activated protein kinase phos-phatase-1 and dephosphorylation of extracellular signal-regulated kinase and impair proliferation in human andmouse osteoblast cell lines. Endocrinology 144:412–422

561. Conradie MM, de Wet H, Kotze DD, Burrin JM, HoughFS, Hulley PA 2007 Vanadate prevents glucocorticoid-in-duced apoptosis of osteoblasts in vitro and osteocytes invivo. J Endocrinol 195:229–240

562. HulleyPA,ConradieMM,LangeveldtCR,HoughFS2002Glucocorticoid-induced osteoporosis in the rat is pre-vented by the tyrosine phosphatase inhibitor, sodium or-thovanadate. Bone 31:220–229

563. Yun SI, Yoon HY, Jeong SY, Chung YS 2009 Glucocor-ticoid induces apoptosis of osteoblast cells through the ac-tivation of glycogen synthase kinase 3�. J Bone MinerMetab 27:140–148

564. Plotkin LI, Manolagas SC, Bellido T 2007 Glucocorticoidsinduce osteocyte apoptosis by blocking focal adhesion ki-

nase-mediated survival. Evidence for inside-out signalingleading to anoikis. J Biol Chem 282:24120–24130

565. Chauhan D, Hideshima T, Pandey P, Treon S, Teoh G,RajeN,RosenS,KrettN,HussonH,AvrahamS,KharbandaS,Anderson KC 1999 RAFTK/PYK2-dependent and -inde-pendent apoptosis in multiple myeloma cells. Oncogene18:6733–6740

566. Tokiwa G, Dikic I, Lev S, Schlessinger J 1996 Activation ofPyk2 by stress signals and coupling with JNK signalingpathway. Science 273:792–794

567. Meyer T, Carlstedt-Duke J, Starr DB 1997 A weak TATAbox is a prerequisite for glucocorticoid-dependent repres-sion of the osteocalcin gene. J Biol Chem 272:30709–30714

568. Chrysis D, Zaman F, Chagin AS, Takigawa M, SavendahlL 2005 Dexamethasone induces apoptosis in proliferativechondrocytes through activation of caspases and suppres-sion of the Akt-phosphatidylinositol 3�-kinase signalingpathway. Endocrinology 146:1391–1397

569. Schakman O, Gilson H, Thissen JP 2008 Mechanisms ofglucocorticoid-induced myopathy. J Endocrinol 197:1–10

570. Wang H, Kubica N, Ellisen LW, Jefferson LS, Kimball SR2006 Dexamethasone represses signaling through themammalian target of rapamycin in muscle cells by enhanc-ing expression of REDD1. J Biol Chem 281:39128–39134

571. Schakman O, Kalista S, Bertrand L, Lause P, Verniers J,Ketelslegers JM, Thissen JP 2008 Role of Akt/GSK-3�/�-catenin transduction pathway in the muscle anti-atrophyaction of insulin-like growth factor-I in glucocorticoid-treated rats. Endocrinology 149:3900–3908

572. Goodman PA, Medina-Martinez O, Fernandez-Mejia C1996 Identification of the human insulin negative regula-tory element as a negative glucocorticoid response element.Mol Cell Endocrinol 120:139–146

573. Yoshiuchi I, Shingu R, Nakajima H, Hamaguchi T,Horikawa Y, Yamasaki T, Oue T, Ono A, Miyagawa JI,Namba M, Hanafusa T, Matsuzawa Y 1998 Mutation/polymorphism scanning of glucose-6-phosphatase genepromoter in noninsulin-dependent diabetes mellitus pa-tients. J Clin Endocrinol Metab 83:1016–1019

574. Kwon HS, Huang B, Unterman TG, Harris RA 2004Protein kinase B-� inhibits human pyruvate dehydroge-nase kinase-4 gene induction by dexamethasone throughinactivation of FOXO transcription factors. Diabetes53:899 –910

575. Nakae J, Kitamura T, Silver DL, Accili D 2001 The fork-head transcription factor Foxo1 (Fkhr) confers insulin sen-sitivity onto glucose-6-phosphatase expression. J Clin In-vest 108:1359–1367

576. Rigaud G, Roux J, Pictet R, Grange T 1991 In vivo foot-printing of rat TAT gene: dynamic interplay between theglucocorticoid receptor and a liver-specific factor. Cell 67:977–986

577. Arizmendi C, Liu S, Croniger C, Poli V, Friedman JE 1999The transcription factor CCAAT/enhancer-binding pro-tein � regulates gluconeogenesis and phosphoenolpyru-vate carboxykinase (GTP) gene transcription during dia-betes. J Biol Chem 274:13033–13040

578. Crosson SM, Roesler WJ 2000 Hormonal regulation of thephosphoenolpyruvate carboxykinase gene. Role of specificCCAAT/enhancer-binding protein isoforms. J Biol Chem275:5804–5809

Endocrine Reviews, December 2009, 30(7):830–882 edrv.endojournals.org 877

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

579. Yamada K, Duong DT, Scott DK, Wang JC, Granner DK1999 CCAAT/enhancer-binding protein � is an accessoryfactor for the glucocorticoid response from the cAMP re-sponse element in the rat phosphoenolpyruvate carboxyki-nase gene promoter. J Biol Chem 274:5880–5887

580. Itani OA, Auerbach SD, Husted RF, Volk KA, Ageloff S,Knepper MA, Stokes JB, Thomas CP 2002 Glucocorti-coid-stimulated lung epithelial Na(�) transport is associ-ated with regulated ENaC and sgk1 expression. Am JPhysiol Lung Cell Mol Physiol 282:L631–L641

581. Sayegh R, Auerbach SD, Li X, Loftus RW, Husted RF,Stokes JB, Thomas CP 1999 Glucocorticoid induction ofepithelial sodium channel expression in lung and renal ep-ithelia occurs via trans-activation of a hormone responseelement in the 5�-flanking region of the human epithelialsodium channel � subunit gene. J Biol Chem 274:12431–12437

582. Brennan FE, Fuller PJ 2000 Rapid upregulation of serumand glucocorticoid-regulated kinase (sgk) gene expres-sion by corticosteroids in vivo. Mol Cell Endocrinol 166:129 –136

583. Chen SY, Bhargava A, Mastroberardino L, Meijer OC,Wang J, Buse P, Firestone GL, Verrey F, Pearce D 1999Epithelial sodium channel regulated by aldosterone-in-duced protein sgk. Proc Natl Acad Sci USA 96:2514–2519

584. Boyd C, Naray-Fejes-Toth A 2007 Steroid-mediated reg-ulation of the epithelial sodium channel subunits in mam-mary epithelial cells. Endocrinology 148:3958–3967

585. Vallon V, Lang F 2005 New insights into the role of serum-and glucocorticoid-inducible kinase SGK1 in the regula-tion of renal function and blood pressure. Curr OpinNephrol Hypertens 14:59–66

586. Choi JY, Kim SY, Son EJ, Kim JL, Shin JH, Song MH,Moon UY, Yoon JH 2006 Dexamethasone increases fluidabsorption via Na�/H� exchanger (NHE) 3 activation innormal human middle ear epithelial cells. Eur J Pharmacol536:12–18

587. Wang D, Zhang H, Lang F, Yun CC 2007 Acute activationof NHE3 by dexamethasone correlates with activation ofSGK1 and requires a functional glucocorticoid receptor.Am J Physiol Cell Physiol 292:C396–C404

588. Wang D, Sun H, Lang F, Yun CC 2005 Activation ofNHE3 by dexamethasone requires phosphorylation ofNHE3 at Ser663 by SGK1. Am J Physiol Cell Physiol 289:C802–C810

589. Zhang W, Xia X, Reisenauer MR, Rieg T, Lang F, Kuhl D,Vallon V, Kone BC 2007 Aldosterone-induced Sgk1 re-lieves Dot1a-Af9-mediated transcriptional repression ofepithelial Na� channel �. J Clin Invest 117:773–783

590. Debonneville C, Flores SY, Kamynina E, Plant PJ, Tauxe C,Thomas MA, Munster C, Chraïbi A, Pratt JH, Horisberger JD,Pearce D, Loffing J, Staub O 2001 Phosphorylation ofNedd4-2 by Sgk1 regulates epithelial Na(�) channel cellsurface expression. EMBO J 20:7052–7059

591. Snyder PM, Olson DR, Thomas BC 2002 Serum and glu-cocorticoid-regulated kinase modulates Nedd4-2-medi-ated inhibition of the epithelial Na� channel. J Biol Chem277:5–8

592. Ichimura T, Yamamura H, Sasamoto K, Tominaga Y,Taoka M, Kakiuchi K, Shinkawa T, Takahashi N, ShimadaS, Isobe T 2005 14-3-3 proteins modulate the expression ofepithelial Na� channels by phosphorylation-dependent

interaction with Nedd4-2 ubiquitin ligase. J Biol Chem280:13187–13194

593. Nagaki K, Yamamura H, Shimada S, Saito T, Hisanaga S,Taoka M, Isobe T, Ichimura T 2006 14-3-3 Mediatesphosphorylation-dependent inhibition of the interactionbetween the ubiquitin E3 ligase Nedd4-2 and epithelialNa� channels. Biochemistry 45:6733–6740

594. Diakov A, Korbmacher C 2004 A novel pathway of epi-thelial sodium channel activation involves a serum- andglucocorticoid-inducible kinase consensus motif in the Cterminus of the channel’s �-subunit. J Biol Chem279:38134–38142

595. Dieter M, Palmada M, Rajamanickam J, Aydin A, BusjahnA, Boehmer C, Luft FC, Lang F 2004 Regulation of glucosetransporter SGLT1 by ubiquitin ligase Nedd4-2 and ki-nases SGK1, SGK3, and PKB. Obes Res 12:862–870

596. Lang PA, Schniepp R, Kirchhoff P, Socrates T, Sidani SM,Geibel JP 2007 PI3 kinase dependent stimulation of gastricacid secretion by dexamethasone. Cell Physiol Biochem20:527–534

597. Gupta V, Awasthi N, Wagner BJ 2007 Specific activationof the glucocorticoid receptor and modulation of signaltransduction pathways in human lens epithelial cells. In-vest Ophthalmol Vis Sci 48:1724–1734

598. Webster JC, Oakley RH, Jewell CM, Cidlowski JA 2001Proinflammatory cytokines regulate human glucocorti-coid receptor gene expression and lead to the accumulationof the dominant negative � isoform: a mechanism for thegeneration of glucocorticoid resistance. Proc Natl Acad SciUSA 98:6865–6870

599. Sousa AR, Lane SJ, Cidlowski JA, Staynov DZ, Lee TH2000 Glucocorticoid resistance in asthma is associatedwith elevated in vivo expression of the glucocorticoid re-ceptor �-isoform. J Allergy Clin Immunol 105:943–950

600. Adcock IM, Lane SJ, Brown CR, Lee TH, Barnes PJ 1995Abnormal glucocorticoid receptor-activator protein 1 in-teraction in steroid-resistant asthma. J Exp Med 182:1951–1958

601. Adcock IM, Lane SJ, Brown CR, Peters MJ, Lee TH, BarnesPJ 1995 Differences in binding of glucocorticoid receptorto DNA in steroid-resistant asthma. J Immunol 154:3500–3505

602. Matthews JG, Ito K, Barnes PJ, Adcock IM 2004 Defectiveglucocorticoid receptor nuclear translocation and alteredhistone acetylation patterns in glucocorticoid-resistant pa-tients. J Allergy Clin Immunol 113:1100–1108

603. Woodruff PG, Boushey HA, Dolganov GM, Barker CS,Yang YH, Donnelly S, Ellwanger A, Sidhu SS, Dao-PickTP, Pantoja C, Erle DJ, Yamamoto KR, Fahy JV 2007Genome-wide profiling identifies epithelial cell genes as-sociated with asthma and with treatment response to cor-ticosteroids. Proc Natl Acad Sci USA 104:15858–15863

604. Bilodeau S, Vallette-Kasic S, Gauthier Y, Figarella-BrangerD, Brue T, Berthelet F, Lacroix A, Batista D, Stratakis C,Hanson J, Meij B, Drouin J 2006 Role of Brg1 and HDAC2in GR trans-repression of the pituitary POMC gene andmisexpression in Cushing disease. Genes Dev 20:2871–2886

605. Ito K, Herbert C, Siegle JS, Vuppusetty C, Hansbro N,Thomas PS, Foster PS, Barnes PJ, Kumar RK 2008 Steroid-resistant neutrophilic inflammation in a mouse model of an

878 Beck et al. GR Crosstalk with Kinases and Phosphatases Endocrine Reviews, December 2009, 30(7):830–882

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

acute exacerbation of asthma. Am J Respir Cell Mol Biol39:543–550

606. Marwick JA, Caramori G, Stevenson CS, Casolari P,Jazrawi E, Barnes PJ, Ito K, Adcock IM, Kirkham PA, PapiA 2009 Inhibition of PI3K� restores glucocorticoid func-tion in smoking-induced airway inflammation in mice.Am J Respir Crit Care Med 179:542–548

607. Pariante CM 2008 The role of multi-drug resistance p-glycoprotein in glucocorticoid function: studies in animalsand relevance in humans. Eur J Pharmacol 583:263–271

608. Pace TW, Hu F, Miller AH 2007 Cytokine-effects on glu-cocorticoid receptor function: relevance to glucocorticoidresistance and the pathophysiology and treatment of majordepression. Brain Behav Immun 21:9–19

609. Sousa AR, Lane SJ, Soh C, Lee TH 1999 In vivo resistanceto corticosteroids in bronchial asthma is associated withenhanced phosphorylation of JUN N-terminal kinase andfailure of prednisolone to inhibit JUN N-terminal kinasephosphorylation. J Allergy Clin Immunol 104:565–574

610. Adcock IM, Lane SJ 2003 Corticosteroid-insensitive asth-ma: molecular mechanisms. J Endocrinol 178:347–355

611. Leung DY, Bloom JW 2003 Update on glucocorticoidaction and resistance. J Allergy Clin Immunol 111:3–22;quiz 23

612. Tsitoura DC, Rothman PB 2004 Enhancement of MEK/ERK signaling promotes glucocorticoid resistance inCD4� T cells. J Clin Invest 113:619–627

613. Li LB, Goleva E, Hall CF, Ou LS, Leung DY 2004 Supe-rantigen-induced corticosteroid resistance of human Tcells occurs through activation of the mitogen-activatedprotein kinase kinase/extracellular signal-regulated kinase(MEK-ERK) pathway. J Allergy Clin Immunol 114:1059–1069

614. Bhavsar P, Hew M, Khorasani N, Torrego A, Barnes PJ,Adcock I, Chung KF 2008 Relative corticosteroid insen-sitivity of alveolar macrophages in severe asthma com-pared with non-severe asthma. Thorax 63:784–790

615. Goleva E, Hauk PJ, Hall CF, Liu AH, Riches DW, MartinRJ, Leung DY 2008 Corticosteroid-resistant asthma is as-sociated with classical antimicrobial activation of airwaymacrophages. J Allergy Clin Immunol 122:550–559.e3

616. Loke TK, Mallett KH, Ratoff J, O’Connor BJ, Ying S,Meng Q, Soh C, Lee TH, Corrigan CJ 2006 Systemic glu-cocorticoid reduces bronchial mucosal activation of acti-vator protein 1 components in glucocorticoid-sensitive butnot glucocorticoid-resistant asthmatic patients. J AllergyClin Immunol 118:368–375

617. Lane SJ, Adcock IM, Richards D, Hawrylowicz C, BarnesPJ, Lee TH 1998 Corticosteroid-resistant bronchialasthma is associated with increased c-fos expression inmonocytes and T lymphocytes. J Clin Invest 102:2156–2164

618. Takahashi E, Onda K, Hirano T, Oka K, Maruoka N,Tsuyuguchi M, Matsumura Y, Niitsuma T, Hayashi T2002 Expression of c-fos, rather than c-jun or glucocorti-coid-receptor mRNA, correlates with decreased glucocor-ticoid response of peripheral blood mononuclear cells inasthma. Int Immunopharmacol 2:1419–1427

619. Goleva E, Li LB, Leung DY 2009 IFN-� reverses IL-2- andIL-4-mediated T-cell steroid resistance. Am J Respir CellMol Biol 40:223–230

620. Reichardt HM, Kaestner KH, Tuckermann J, Kretz O,

Wessely O, Bock R, Gass P, Schmid W, Herrlich P, AngelP, Schutz G 1998 DNA binding of the glucocorticoid re-ceptor is not essential for survival. Cell 93:531–541

621. Schacke H, Hennekes H, Schottelius A, Jaroch S, LehmannM, Schmees N, Rehwinkel H, Asadullah K 2002 SEGRAs:a novel class of anti-inflammatory compounds. ErnstSchering Res Found Workshop 40:357–371

622. Schacke H, Berger M, Rehwinkel H, Asadullah K 2007Selective glucocorticoid receptor agonists (SEGRAs):novel ligands with an improved therapeutic index. MolCell Endocrinol 275:109–117

623. Rosen J, Miner JN 2005 The search for safer glucocorti-coid receptor ligands. Endocr Rev 26:452–464

624. Song IH, Gold R, Straub RH, Burmester GR, Buttgereit F2005 New glucocorticoids on the horizon: repress, don’tactivate! J Rheumatol 32:1199–1207

625. Hudson AR, Roach SL, Higuchi RI 2008 Recent develop-ments in the discovery of selective glucocorticoid receptormodulators (SGRMs). Curr Top Med Chem 8:750–765

626. Vanden Berghe W, Francesconi E, De Bosscher K, Resche-Rigon M, Haegeman G 1999 Dissociated glucocorticoidswith anti-inflammatory potential repress interleukin-6gene expression by a nuclear factor-�B-dependent mech-anism. Mol Pharmacol 56:797–806

627. Vayssiere BM, Dupont S, Choquart A, Petit F, Garcia T,Marchandeau C, Gronemeyer H, Resche-Rigon M 1997Synthetic glucocorticoids that dissociate transactivationand AP-1 transrepression exhibit antiinflammatory activ-ity in vivo. Mol Endocrinol 11:1245–1255

628. Lin CW, Nakane M, Stashko M, Falls D, Kuk J, Miller L,Huang R, Tyree C, Miner JN, Rosen J, Kym PR, CoghlanMJ, Carter G, Lane BC 2002 Trans-activation and repres-sion properties of the novel nonsteroid glucocorticoidreceptor ligand 2,5-dihydro-9-hydroxy-10-methoxy-2,2,4-trimethyl-5-(1-methylcyclohexen-3-y 1)-1H-[1]benzopyrano[3,4-f]quinoline (A276575) and its four stereoisomers. MolPharmacol 62:297–303

629. Coghlan MJ, Jacobson PB, Lane B, Nakane M, Lin CW,Elmore SW, Kym PR, Luly JR, Carter GW, Turner R, TyreeCM, Hu J, Elgort M, Rosen J, Miner JN 2003 A novelantiinflammatory maintains glucocorticoid efficacy withreduced side effects. Mol Endocrinol 17:860–869

630. Owen HC, Miner JN, Ahmed SF, Farquharson C 2007 Thegrowth plate sparing effects of the selective glucocorticoidreceptor modulator, AL-438. Mol Cell Endocrinol 264:164–170

631. Ali A, Thompson CF, Balkovec JM, Graham DW,Hammond ML, Quraishi N, Tata JR, Einstein M, Ge L,Harris G, Kelly TM, Mazur P, Pandit S, Santoro J, SitlaniA, Wang C, Williamson J, Miller DK, Thompson CM,Zaller DM, Forrest MJ, Carballo-Jane E, Luell S 2004Novel N-arylpyrazolo[3,2-c]-based ligands for the glu-cocorticoid receptor: receptor binding and in vivo activity.J Med Chem 47:2441–2452

632. Gossye V, Elewaut D, Van Beneden K, Dewint P, HaegemanG, De Bosscher K 9 February 2009 A plant-derived glu-cocorticoid receptor modulator attenuates inflammationwithout provoking ligand-induced resistance. Ann RheumDis doi:10.1136/ard.2008.102871

633. Schacke H, Schottelius A, Docke WD, Strehlke P, Jaroch S,Schmees N, Rehwinkel H, Hennekes H, Asadullah K 2004Dissociation of transactivation from transrepression by a

Endocrine Reviews, December 2009, 30(7):830–882 edrv.endojournals.org 879

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

selective glucocorticoid receptor agonist leads to separa-tion of therapeutic effects from side effects. Proc Natl AcadSci USA 101:227–232

634. Humphrey EL, Williams JH, Davie MW, Marshall MJ2006 Effects of dissociated glucocorticoids on OPG andRANKL in osteoblastic cells. Bone 38:652–661

635. Lopez FJ, Ardecky RJ, Bebo B, Benbatoul K, De GrandpreL, Liu S, Leibowitz MD, Marschke K, Rosen J, Rungta D,Viveros HO, Yen WC, Zhi L, Negro-Vilar A, Miner JN2008 LGD-5552, an antiinflammatory glucocorticoid re-ceptor ligand with reduced side effects, in vivo. Endocri-nology 149:2080–2089

636. Miner JN, Ardecky B, Benbatoul K, Griffiths K, Larson CJ,Mais DE, Marschke K, Rosen J, Vajda E, Zhi L, Negro-Vilar A 2007 Antiinflammatory glucocorticoid receptorligand with reduced side effects exhibits an altered protein-protein interaction profile. Proc Natl Acad Sci USA 104:19244–19249

637. BarkerM,ClackersM,CopleyR,DemaineDA,HumphreysD,Inglis GG, Johnston MJ, Jones HT, Haase MV, House D,Loiseau R, Nisbet L, Pacquet F, Skone PA, Shanahan SE,Tape D, Vinader VM, Washington M, Uings I, Upton R,McLay IM, Macdonald SJ 2006 Dissociated nonsteroidalglucocorticoid receptor modulators; discovery of the ago-nist trigger in a tetrahydronaphthalene-benzoxazine series.J Med Chem 49:4216–4231

638. Shah N, Scanlan TS 2004 Design and evaluation of novelnonsteroidal dissociating glucocorticoid receptor ligands.Bioorg Med Chem Lett 14:5199–5203

639. Thompson CF, Quraishi N, Ali A, Mosley RT, Tata JR,Hammond ML, Balkovec JM, Einstein M, Ge L, Harris G,Kelly TM, Mazur P, Pandit S, Santoro J, Sitlani A, Wang C,Williamson J, Miller DK, Yamin TT, Thompson CM,O’Neill EA, Zaller D, Forrest MJ, Carballo-Jane E, Luell S2007 Novel glucocorticoids containing a 6,5-bicyclic corefused to a pyrazole ring: synthesis, in vitro profile, molec-ular modeling studies, and in vivo experiments. BioorgMed Chem Lett 17:3354–3361

640. Thompson CF, Quraishi N, Ali A, Tata JR, HammondML, Balkovec JM, Einstein M, Ge L, Harris G, Kelly TM,Mazur P, Pandit S, Santoro J, Sitlani A, Wang C, WilliamsonJ, Miller DK, Yamin TT, Thompson CM, O’Neill EA,Zaller D, Forrest MJ, Carballo-Jane E, Luell S 2005 Novelheterocyclic glucocorticoids: in vitro profile and in vivoefficacy. Bioorg Med Chem Lett 15:2163–2167

641. Wang JC, Shah N, Pantoja C, Meijsing SH, Ho JD, ScanlanTS, Yamamoto KR 2006 Novel arylpyrazole compoundsselectively modulate glucocorticoid receptor regulatory ac-tivity. Genes Dev 20:689–699

642. Biggadike K, Boudjelal M, Clackers M, Coe DM, DemaineDA, Hardy GW, Humphreys D, Inglis GG, Johnston MJ,Jones HT, House D, Loiseau R, Needham D, Skone PA,Uings I, Veitch G, Weingarten GG, McLay IM, MacdonaldSJ 2007 Nonsteroidal glucocorticoid agonists: tetrahy-dronaphthalenes with alternative steroidal A-ring mimet-ics possessing dissociated (transrepression/transactiva-tion) efficacy selectivity. J Med Chem 50:6519–6534

643. Robinson RP, Buckbinder L, Haugeto AI, McNiff PA,MillhamML,ReeseMR,Schaefer JF,AbramovYA,BordnerJ, Chantigny YA, Kleinman EF, Laird ER, Morgan BP,Murray JC, Salter ED, Wessel MD, Yocum SA 2009Octahydrophenanthrene-2,7-diol analogues as dissoci-

ated glucocorticoid receptor agonists: discovery and leadexploration. J Med Chem 52:1731–1743

644. Chou TC 2006 Theoretical basis, experimental design, andcomputerized simulation of synergism and antagonism indrug combination studies. Pharmacol Rev 58:621–681

645. Barnes PJ 2006 Corticosteroids: the drugs to beat. EurJ Pharmacol 533:2–14

646. Calverley PM, Anderson JA, Celli B, Ferguson GT, JenkinsC, Jones PW, Yates JC, Vestbo J 2007 Salmeterol and flu-ticasone propionate and survival in chronic obstructivepulmonary disease. N Engl J Med 356:775–789

647. Eickelberg O, Roth M, Lorx R, Bruce V, Rudiger J, JohnsonM, Block LH 1999 Ligand-independent activation of the glu-cocorticoid receptor by �2-adrenergic receptor agonists inprimary human lung fibroblasts and vascular smooth musclecells. J Biol Chem 274:1005–1010

648. Usmani OS, Ito K, Maneechotesuwan K, Ito M, JohnsonM, Barnes PJ, Adcock IM 2005 Glucocorticoid receptornuclear translocation in airway cells after inhaled combi-nation therapy. Am J Respir Crit Care Med 172:704–712

649. Adcock IM, Maneechotesuwan K, Usmani O 2002 Mo-lecular interactions between glucocorticoids and long-act-ing �2-agonists. J Allergy Clin Immunol 110:S261–268

650. Fragaki K, Kileztky C, Trentesaux C, Zahm JM, Bajolet O,Johnson M, Puchelle E 2006 Downregulation by a long-acting �2-adrenergic receptor agonist and corticosteroid ofStaphylococcus aureus-induced airway epithelial inflam-matory mediator production. Am J Physiol Lung Cell MolPhysiol 291:L11–L18

651. Kaur M, Chivers JE, Giembycz MA, Newton R 2008 Long-acting �2-adrenoceptor agonists synergistically enhanceglucocorticoid-dependent transcription in human airwayepithelial and smooth muscle cells. Mol Pharmacol 73:203–214

652. Beck IM, Berghe WV, Gerlo S, Bougarne N, Vermeulen L,De Bosscher K, Haegeman G 2009 Glucocorticoids andmitogen- and stress-activated protein kinase 1 inhibitors:possible partners in the combat against inflammation. Bio-chem Pharmacol 77:1194–1205

653. Lim-Tio SS, Fuller PJ 1998 Intracellular signaling path-ways confer specificity of transactivation by mineralocor-ticoid and glucocorticoid receptors. Endocrinology 139:1653–1661

654. Derijk RH, de Kloet ER2008 Corticosteroid receptor poly-morphisms: determinants of vulnerability and resilience.Eur J Pharmacol 583:303–311

655. Aubry EM, Odermatt A 2009 Retinoic acid reduces glu-cocorticoid sensitivity in C2C12 myotubes by decreasing11�-hydroxysteroid dehydrogenase type 1 and glucocor-ticoid receptor activities. Endocrinology 150:2700–2708

656. Odermatt A, Gumy C 2008 Glucocorticoid and mineralo-corticoid action: why should we consider influences byenvironmental chemicals? Biochem Pharmacol 76:1184–1193

657. Atanasov AG, Odermatt A 2007 Readjusting the glucocor-ticoid balance: an opportunity for modulators of 11�-hy-droxysteroid dehydrogenase type 1 activity? Endocr MetabImmune Disord Drug Targets 7:125–140

658. Kolber BJ, Wieczorek L, Muglia LJ 2008 Hypothalamic-pituitary-adrenal axis dysregulation and behavioral anal-ysis of mouse mutants with altered glucocorticoid or min-eralocorticoid receptor function. Stress 11:321–338

880 Beck et al. GR Crosstalk with Kinases and Phosphatases Endocrine Reviews, December 2009, 30(7):830–882

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

659. Gruber G, Carlet M, Turtscher E, Meister B, Irving JA,Ploner C, Kofler R 2009 Levels of glucocorticoid receptorand its ligand determine sensitivity and kinetics of glucocor-ticoid-induced leukemia apoptosis. Leukemia 23:820–823

660. Rosenfeld MG, Lunyak VV, Glass CK 2006 Sensors andsignals: a coactivator/corepressor/epigenetic code for inte-grating signal-dependent programs of transcriptional re-sponse. Genes Dev 20:1405–1428

661. Brush MH, Guardiola A, Connor JH, Yao TP, ShenolikarS 2004 Deactylase inhibitors disrupt cellular complexescontaining protein phosphatases and deacetylases. J BiolChem 279:7685–7691

662. Pascussi JM, Gerbal-Chaloin S, Duret C, Daujat-Chavanieu M, Vilarem MJ, Maurel P 2008 The tangle ofnuclear receptors that controls xenobiotic metabolism andtransport: crosstalk and consequences. Annu Rev Pharma-col Toxicol 48:1–32

663. Bougarne N, Paumelle R, Caron S, Hennuyer N, MansouriR, Gervois P, Staels B, Haegeman G, De Bosscher K 2009PPAR� blocks glucocorticoid receptor �-mediated trans-activation but cooperates with the activated glucocorticoidreceptor � for transrepression on NF-�B. Proc Natl AcadSci USA 106:7397–7402

664. Nishi M, Kawata M 2007 Dynamics of glucocorticoid re-ceptorandmineralocorticoid receptor: implications fromlivecell imaging studies. Neuroendocrinology 85:186–192

665. Vreugdenhil E, Verissimo CS, Mariman R, Kamphorst JT,Barbosa JS, Zweers T, Champagne DL, Schouten T, MeijerOC, de Kloet ER, Fitzsimons CP 2009 MicroRNA 18and 124a down-regulate the glucocorticoid receptor:implications for glucocorticoid responsiveness in thebrain. Endocrinology 150:2220 –2228

666. Visvanathan J, Lee S, Lee B, Lee JW, Lee SK 2007 ThemicroRNA miR-124 antagonizes the anti-neural REST/SCP1 pathway during embryonic CNS development.Genes Dev 21:744–749

667. Rainer J, Ploner C, Jesacher S, Ploner A, Eduardoff M,Mansha M, Wasim M, Panzer-Grumayer R, Trajanoski Z,Niederegger H, Kofler R 2009 Glucocorticoid-regulatedmicroRNAs and mirtrons in acute lymphoblastic leuke-mia. Leukemia 23:746–752

668. Bethke A, Fielenbach N, Wang Z, Mangelsdorf DJ, AntebiA 2009 Nuclear hormone receptor regulation of microRNAscontrols developmental progression. Science 324:95–98

669. Dai R, Phillips RA, Zhang Y, Khan D, Crasta O, Ahmed SA2008 Suppression of LPS-induced interferon-� and nitricoxide in splenic lymphocytes by select estrogen-regulatedmicroRNAs: a novel mechanism of immune modulation.Blood 112:4591–4597

670. Asirvatham AJ, Gregorie CJ, Hu Z, Magner WJ, TomasiTB 2008 MicroRNA targets in immune genes and theDicer/Argonaute and ARE machinery components. MolImmunol 45:1995–2006

671. Burkhart BA, Ivey ML, Archer TK 2009 Long-term lowlevel glucocorticoid exposure induces persistent repres-sion in chromatin. Mol Cell Endocrinol 298:66 –75

672. Adcock IM, Tsaprouni L, Bhavsar P, Ito K 2007 Epigeneticregulation of airway inflammation. Curr Opin Immunol19:694–700

673. Nawrot TS, Adcock I 2009 The detrimental health effectsof traffic-related air pollution: a role for DNA methyl-ation? Am J Respir Crit Care Med 179:523–524

674. Kagoshima M, Wilcke T, Ito K, Tsaprouni L, Barnes PJ,Punchard N, Adcock IM 2001 Glucocorticoid-mediatedtransrepression is regulated by histone acetylation andDNA methylation. Eur J Pharmacol 429:327–334

675. Baccarelli A, Wright RO, Bollati V, Tarantini L, LitonjuaAA, Suh HH, Zanobetti A, Sparrow D, Vokonas PS,Schwartz J 2009 Rapid DNA methylation changes afterexposure to traffic particles. Am J Respir Crit Care Med179:572–578

676. Backdahl L, Bushell A, Beck S 2009 Inflammatory signal-ling as mediator of epigenetic modulation in tissue-specificchronic inflammation. Int J Biochem Cell Biol 41:176–184

677. Vanden Berghe W, Ndlovu MN, Hoya-Arias R, DijsselbloemN, Gerlo S, Haegeman G 2006 Keeping up NF-�B appear-ances: epigenetic control of immunity or inflammation-triggered epigenetics. Biochem Pharmacol 72:1114–1131

678. van Bel F, Heijnen CJ 2009 Perinatal programming andreprogramming by glucocorticoid therapy and perinatalstress. Semin Fetal Neonatal Med 14:127–129

679. Champagne DL, de Kloet ER, Joels M 2009 Fundamentalaspects of the impact of glucocorticoids on the (immature)brain. Semin Fetal Neonatal Med 14:136–142

680. Weaver IC 2009 Epigenetic effects of glucocorticoids. SeminFetal Neonatal Med 14:143–150

681. Merlot E, Couret D, Otten W 2008 Prenatal stress, fetalimprinting and immunity. Brain Behav Immun 22:42–51

682. Darnaudery M, Maccari S 2008 Epigenetic programmingof the stress response in male and female rats by prenatalrestraint stress. Brain Res Rev 57:571–585

683. Oberlander TF, Weinberg J, Papsdorf M, Grunau R, MisriS, Devlin AM 2008 Prenatal exposure to maternal depres-sion, neonatal methylation of human glucocorticoid re-ceptor gene (NR3C1) and infant cortisol stress responses.Epigenetics 3:97–106

684. Turner JD, Pelascini LP, Macedo JA, Muller CP 2008Highly individual methylation patterns of alternativeglucocorticoid receptor promoters suggest individual-ized epigenetic regulatory mechanisms. Nucleic AcidsRes 36:7207–7218

685. Schwermann J, Rathinam C, Schubert M, Schumacher S,Noyan F, Koseki H, Kotlyarov A, Klein C, Gaestel M 2009MAPKAP kinase MK2 maintains self-renewal capacity ofhaematopoietic stem cells. EMBO J 28:1392–1406

686. Voncken JW, Niessen H, Neufeld B, Rennefahrt U, DahlmansV, Kubben N, Holzer B, Ludwig S, Rapp UR 2005MAPKAP kinase 3pK phosphorylates and regulateschromatin association of the polycomb group proteinBmi1. J Biol Chem 280:5178 –5187

687. Fischle W 2008 Talk is cheap—cross-talk in establish-ment, maintenance, and readout of chromatin modifica-tions. Genes Dev 22:3375–3382

688. Adhvaryu KK, Selker EU 2008 Protein phosphatase PP1 isrequired for normal DNA methylation in Neurospora.Genes Dev 22:3391–3396

689. Hodge DR, Cho E, Copeland TD, Guszczynski T, Yang E,Seth AK, Farrar WL 2007 IL-6 enhances the nuclear trans-location of DNA cytosine-5-methyltransferase 1 (DNMT1) viaphosphorylation of the nuclear localization sequence bythe AKT kinase. Cancer Genomics Proteomics 4:387–398

690. Gazin C, Wajapeyee N, Gobeil S, Virbasius CM, GreenMR 2007 An elaborate pathway required for Ras-me-diated epigenetic silencing. Nature 449:1073–1077

Endocrine Reviews, December 2009, 30(7):830–882 edrv.endojournals.org 881

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022

691. Merienne K, Pannetier S, Harel-Bellan A, Sassone-Corsi P2001 Mitogen-regulated RSK2-CBP interaction controls theirkinase and acetylase activities. Mol Cell Biol 21:7089–7096

692. Thambirajah AA, Eubanks JH, Ausio J 2009 MeCP2 post-translational regulation through PEST domains: two novelhypotheses: potential relevance and implications for Rettsyndrome. Bioessays 31:561–569

693. Chao HT, Zoghbi HY 2009 The yin and yang of MeCP2phosphorylation. Proc Natl Acad Sci USA 106:4577–4578

694. Tao J, Hu K, Chang Q, Wu H, Sherman NE, MartinowichK, Klose RJ, Schanen C, Jaenisch R, Wang W, Sun YE 2009Phosphorylation of MeCP2 at serine 80 regulates its chro-matin association and neurological function. Proc NatlAcad Sci USA 106:4882–4887

695. KameshitaI,SekiguchiM,HamasakiD,SugiyamaY,HatanoN, Suetake I, Tajima S, Sueyoshi N 2008 Cyclin-dependentkinase-like 5 binds and phosphorylates DNA methyltrans-ferase 1. Biochem Biophys Res Commun 377:1162–1167

696. Miyake K, Nagai K 2007 Phosphorylation of methyl-CpGbinding protein 2 (MeCP2) regulates the intracellular lo-calization during neuronal cell differentiation. NeurochemInt 50:264–270

697. Nuber UA, Kriaucionis S, Roloff TC, Guy J, Selfridge J,Steinhoff C, Schulz R, Lipkowitz B, Ropers HH, HolmesMC, Bird A 2005 Up-regulation of glucocorticoid-regu-lated genes in a mouse model of Rett syndrome. Hum MolGenet 14:2247–2256

698. Kangaspeska S, Stride B, Metivier R, Polycarpou-SchwarzM, Ibberson D, Carmouche RP, Benes V, Gannon F, ReidG 2008 Transient cyclical methylation of promoter DNA.Nature 452:112–115

699. Metivier R, Gallais R, Tiffoche C, Le Peron C, JurkowskaRZ, Carmouche RP, Ibberson D, Barath P, Demay F, ReidG, Benes V, Jeltsch A, Gannon F, Salbert G 2008 CyclicalDNA methylation of a transcriptionally active promoter.Nature 452:45–50

882 Beck et al. GR Crosstalk with Kinases and Phosphatases Endocrine Reviews, December 2009, 30(7):830–882

Dow

nloaded from https://academ

ic.oup.com/edrv/article/30/7/830/2355077 by guest on 11 August 2022