Journal of Pediatrics & Child Care

Download PDF
Review Article

Functional Links Between Glucocorticoids and Cytokines In DBA

Yoan Konto-Ghiorghi*

  • Department of Pediatrics, Stanford University School of Medicine, Stanford University, Stanford, California, USA

*Address for Correspondence: Yoan Konto-Ghiorghi, Department of Pediatrics, Stanford University School of Medicine, CCSR Building, Room 1210, 269 Campus Drive, Stanford University, Stanford, California, 94305-5162 USA, E-mail: yoan.konto@gmail.com
 
Citation: Konto-Ghiorghi Y. Functional Links Between Glucocorticoids and Cytokines In DBA. J Pediatrics Child Care. 2013;1(1): 5.
 
Copyright © 2013 Yoan Konto-Ghiorghi. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
 
Journal of Pediatrics & Child Care | ISSN: 2380-0534 | Volume: 1, Issue: 1
 
Submission: 02 April 2013 | Accepted: 29 April 2013 | Published: 30 April 2013

Abstract

Diamond Blackfan anaemia (DBA) is a red blood cell aplasia characterized by erythropoietic defects as well as congenital anomalies. Forty percent of patients with DBA are treated with glucocorticoid steroids, which remain the mainstay of treatment in DBA. Many advances in the understanding of the physiological role of the glucocorticoid receptor have been made since the first introduction of glucocorticoids to the clinic, but their mechanism of action in the treatment of DBA is still under investigation. This review is intended to summarize the mechanism of glucocorticoid action specifically as related to erythropoiesis, focusing on the functional links between glucocorticoids and cytokines.

Introduction

Diamond Blackfan anemia (DBA) was first described as a disorder of impaired red blood cell production in children [1,2]. While most of the DBA cases are diagnosed in early infancy, a recent case report reveals that DBA can occur during fetal development. This suggests that severely affected DBA fetus likely die to hydrops fetalis and result in undiagnosed miscarriages [3]. This disorder results from a cellular defect in which erythroid progenitors are highly sensitive to death by apoptosis, leading to erythropoietic failure [4]. The etiology of DBA has been the subject of continuous discussions, and while the early success of treatment with glucocorticoids (GCs) in 1951 [5] led to the idea that the pathogenesis of DBA could have an immunologic basis [6], it is now accepted that DBA is in fact a member of a rare group of genetic disorders.

Approximately 50% of DBA patients have a single mutation in a gene encoding a ribosomal protein, which indicates that DBA is associated with a disorder of ribosome biogenesis and/or function [4]. The pronounced erythroid defect suggests that erythroid progenitors may express specific mRNAs that are hypersensitive to the decreased translation capacity [7]. Twenty-five percent of patients have a mutation in the ribosomal protein S19 (RPS19) gene [8], and two independent studies have demonstrated that over expression of the RPS19 transgene increases the number of erythroid colonies in RPS19 deficient hematopoietic progenitor cells in vitro [9,10]. In a mouse model of DBA, a high expression of RPS19 can rescue the erythroid development, and the corrected DBA cells show a survival advantage in vivo [11]. As zebrafish hematopoietic regulation is conserved with mammals, zebrafish models have also been reported to be useful in studying DBA [12-14], recapitulating many aspects of the DBA phenotype, including hematopoietic specific defects and p53 activation [14]. The list of genes that are mutated in DBA has been updated in 2013, to include ten ribosomal genes and one transcriptional regulator: RPS19, RPS24, RPS17, RPL35A, RPL5, RPL11, RPS7, RPS10, RPS26, RPL26, and GATA1 [15].

More than 50 years after their introduction to the clinic, GCs are still the most effective drugs used in DBA. Nevertheless, the reported side effects of GCs include decreased growth velocity in infants, neuromotor dysfunction in toddlers, and significant immune suppression, rendering GCs an unsuitable therapeutic choice for children under the age of one [4]. Today, the only curative treatment for DBA is hematopoietic stem cell transplantation, a treatment that relies on the availability of an HLA-matched related donor [4]. To reduce the side effects of GCs and increase their efficacy, the mechanism of action of GCs has been – and still is – the subject of intensive research.

This short review will therefore focus on (a) the molecular mechanisms of GCs reported in erythropoiesis, and (b) the relationship between GCs and cytokines, where cytokines will be discussed as new potential drug targets in DBA treatment.

Molecular mechanism of Glucocorticoids

Glucocorticoids (GCs) have pleiotropic effects on hematopoietic cells. They have been shown to induce apoptosis in lymphoid cells [16], prevent apoptosis in granulocytes [17], and drive the proliferation of erythroid progenitors [18,19].

Endogenous GCs

GCs play a pivotal role in several critical biologic processes including growth, reproduction, intermediary metabolism, immune and inflammatory reactions and they also have specific functions in the central nervous and cardiovascular systems [20,21]. The principal endogenous GCs are the hormones cortisol and corticosterone. Cortisol and corticosterone are both synthesized from cholesterol in cells of the zona fasciculate of the adrenal cortex (cortisol is the predominant GC in human, whereas rodents produce mainlycorticosterone). The release of both GCs into the blood is pulsatile and varies according to a distinct circadian pattern. Additional secretions can occur independent of circadian tone in response to physical and/or emotional trauma [22].

In vitro, GCs enhance the formation of murine erythroid colonies [23] and increase proliferation of erythroid cells in the presence of limiting amounts of erythropoietin (Epo) [24]. Interestingly, GCs also stimulate erythropoiesis indirectly by increasing Epo production in the kidney [25].

Other studies have shown a direct correlation between GCs concentration and erythropoiesis by studying individuals that have pathological changes in the production of GCs. For example, Addison’s disease, a disorder caused by insufficient corticosteroid production, is associated with anemia, whereas increased red blood cell count, hemoglobin, and hematocrit values are observed in Cushing’s syndrome, a disease characterized by elevated GCs levels [26].

The human glucocorticoid receptors

The glucocorticoid receptor (GR) is an intracellular receptor. It is the first member of the subfamily of steroid hormone receptors, is widely distributed in the body, and has a variety of functions that are essential for survival. Genetically modified mice lacking the GR die at birth and reveal impaired development of several organs [27]. Cell responsiveness to GCs depends not only on the presence of the GR but also on its concentration – which is known to fluctuate during development and the cell cycle, and following disturbances in endocrine status [28]. The human GR (hGR) gene consists of nine exons and is located on chromosome 5. Alternative splicing of the hGR gene in exon 9 generates two highly homologous isoforms of the receptor, termed α and β [29]. The GRα binds and is activated by corticosteroids, whereas the unique structure of the GRβ impairs the ligand-binding domain and induces nuclear retention [30]. The GRβ has been implicated in steroid resistance in asthma [31], and observed to exert dominant-negative effects on the GRα [32,33]. The belief that the GRβ is a general negative modulator of GRα function was challenged by cotransfection studies in COS-7 cells, where the GRβ was not observed to inhibit the effects of dexamethasone-activated GRα on a glucocorticoid-responsive reporter gene [34]. Most actions of GCs are mediated by the GR, and several polymorphisms in the hGR gene are associated with altered GC sensitivity [35]. As there is currently no reliable way to predict steroid responsiveness in DBA patients [4], these polymorphisms very likely affect the response of patients to GC in the treatment of DBA and represent another reason for the development of non GC-based DBA drugs.

Activation and targets of glucocorticoid receptor

Activation of the glucocorticoid receptor (GR) leads to subsequent intracellular signaling. Heat Shock Protein 90 (HSP90) regulates ligand binding and cytoplasmic retention of the GRα by exposing the ligand-binding site and masking the two nuclear localization sequences [36]. Upon ligand-induced activation, the GR undergoes a conformational change, resulting in its dissociation from a multiprotein complex that includes HSP90 and its translocation into the nucleus.

Within the nucleus, the GR binds as a homodimer to Glucocorticoid Response Elements (GREs) in the promoter region of target genes, regulating their expression positively or negatively depending on the GRE sequence and promoter context [37]. Alternatively, the ligand-activated GRα can interact as a monomer with other transcription factors, modulating their transcriptional activity through direct protein-protein interaction, independent of direct DNA binding [38].

DNA-independent actions of GR: transrepression

The suppression of transactivation of other transcription factors through protein-protein interactions is particularly important in the suppression of immune function and inflammation by GCs [39]. Most of the effects of GCs on the immune system may be mediated by the interaction between GR and NF-κB, AP-1, and signal transducers and activators of transcription (STATs) [40-43].

The example of transrepression of the proinflammatory transcription factor AP-1 is particularly interesting since DNAbinding inactive mutants of the GR are fully capable of AP-1 transrepression [44]. In erythroid progenitors, several members of NF-κB family are also expressed and it has been suggested that NF-κB factors could function to regulate specific genes involved in erythropoiesis [45]. However, since induction of proliferation of erythroid progenitors by GCs requires DNA binding by the GR [18,36], the transrepression action has appeared not to be essential to the function of the GR in erythroid progenitors.

Transcriptional activation by GR: transactivation

DNA-dependent transactivation by the GR requires dimerization of the receptor [21,46]. Mice carrying a point mutation in the GR, which prevents receptor dimerization (GRdim/dim), are viable [46] and GRdim/dim mice display normal erythrocyte numbers in peripheral blood [47]. GRdim/dim mice were then challenged either by drug-induced hemolytic anemia or by hypoxia to address the question of whether or not the GR would be important for stress erythropoiesis. In both cases, wild-type mice responded with strongly increased numbers of colony-forming units–erythroid (CFU-Es) in their spleens, whereas GRdim/dim mice showed no response. GRdependent expansion upon stress was restricted to a specific erythroid compartment [47]. In vitro, erythroid cells from fetal livers GRdim/ dim mice fail to undergo sustained proliferation in contrast to wildtype cells [47].

Genes differentially regulated by GCs

Upon hormone binding, the GR associates with high affinity to genomic GR binding sequence, typically an imperfect palindromic hexameric half site, separated by 3-base pair spacers [48,49]. Genomewide in silico identification of GR binding sequences could thus be a powerful method to reveal the transcriptional regulatory network of GCs. However, focusing on the role of the GC dexamethasone (Dex) in erythropoiesis, the Dex-induced genes are not enriched for GR binding sites in their promoter regions but instead for hypoxiainducible factor 1α (HIF1α), suggesting that HIF1α activation would enhance the biologic function of GR activation [50]. Another oligonucleotide microarray study to survey gene expression in RPS19-deficient CD34+ cells showed that Dex decreases the expression of 3 cytokine genes: interleukin-1 beta (IL-1B), lymphotoxin-beta (LTB), and macrophage inflammatory protein-1-alpha (MIP-1α, CC chemokine ligand 3 [CCL3]) [51]. All of these cytokines are active in myeloid lineage cells, and Mip-1α is a cytokine that blocks proliferation of hematopoietic stem cells [52].

Targets of the GR include genes involved in proliferation of erythroblasts such as the SCF receptor Kit and the transcription factor Myb [18,53], and cytokines have been shown to modulate GCs function. In absence of the cytokines Epo and SCF, activation of the GR in erythroid progenitors causes cell cycle arrest [18,54]. Dex treatment leads to up-regulation of the antiproliferative genes Btg1 and GilZ [55]. In 3T3 fibroblasts, Btg1 expression is maximal in the G0/G1 phases of the cell cycle and is down-regulated when cells progress throughout G1, suggesting the existence of a functional link between Btg1 and cell cycle progression [56]. GilZ is known to interfere with signaling-controlled activity of NF-κB, AP-1 and the tyrosine kinase JAK2 inihibitor CIS [57,58]. In erythroid progenitors, GilZ is up-regulated by Dex alone, down-regulated by Epo and Epo/SCF, while coregulation by Epo, SCF, and Dex abolishes Epo dependent down-regulation [55]. These conclusions show that Dex is able to attenuate the effect of Epo and/or SCF. Furthermore, CXC chemokine receptor 4 (CXCR4) is up-regulated in the presence of Dex plus Epo/SCF, and CXCR4 is down regulated in GRdim/dim when compared with wt erythroid progenitors, thus confirming an activation of CXCR4 expression by GCs [56].

Suppressor of cytokine signaling 1 (SOCS1) up-regulation by Dex in erythroid progenitors has been confirmed by three different groups [50,55,59]. Mice lacking SOCS1 have been shown to have a subnormal hematocrit and an accumulation of immature red blood cells [60]. Furthermore, SOCS1 is associated with the phosphorylated form of JAK2 (pJAK2) [61] and dysregulation of pJAK2 degradation is involved in polycythemia [62]. Polycythemia is characterized by a net increase in the total number of red blood cells, and can arise when an intrinsic defect in erythroid progenitors render them hypersensitive or independent of erythropoietin stimulation [63]. These reports suggest that SOCS1 might be one of the GCs targets in DBA.

Other mechanisms of action of GCs

The effects of GCs on inflammation occur in a time-frame of 30 minutes to 18 hours [64]. However, the previously described mechanisms cannot explain the rapid effects observed within minutes or seconds following administration. As reported in several other cellular systems [65], the GR can activate a rapid membrane associated signaling in erythroid cells. In human erythroblasts, growth factors stimulation induces association of the GR on the membrane, where the GR forms a complex with the EPO receptor to antagonize its ability to phosphorylate STAT5 [66]. This reveals that GCs are able to inhibit erythroid maturation through a rapid membrane associated pathway that interferes with EPO receptor signaling.

Another mechanism is based on a direct interaction of the GR with mRNA. Whereas some RNA-binding proteins (RBPs) such as ubiquitous HuR mainly act as a positive regulator of mRNA stability, others such as the GCs-induced zinc finger RBP tristetraprolin (TTP) limit the inflammatory response by accelerating the mRNA decay of its targets [67]. Interacting directly with the chemokines (C-C motif) ligand 2 and 7 (CCL2 and CCL7) mRNAs in human airway epithelial cell line BEAS-2B, the GR accelerates the decay of their respective mRNA [68]. The identification of GR-mediated acceleration of chemokine mRNA decay and of this additional function of GR in the cytoplasmic compartment opens a new paradigm in the GCs mechanism of action. Human CD34+ cells and hematopoietic precursors secrete numerous regulatory molecules that form the basis of intercellular cross-talk networks and regulate in an autocrine and/or a paracrine manner the various stages of normal human hematopoiesis [69]. Thus, this new role of GCs could be particularly relevant if specific chemokines / cytokines were found to be differentially expressed and affecting proliferation or differentiation of erythroid progenitors in DBA.

Immunomodulatory GCs-inspired drugs tested in DBA

The broad anti-inflammatory profile of GCs probably accounts for their marked clinical effectiveness in many types of disease [70]. Since the pathophysiology of DBA was first attributed to immune dysregulation and putative suppressor T cells [6], different immunomodulatory agents were tested, and showed different efficacy profiles: 6-mercaptopurine, cyclophosphamide, vincristine, intravenous immunoglobulin, and anti-thymocyte globulin were tried in patients and found to be largely ineffective [4]. On the other hand, in 10 out of 20 cases treated with a combination of cyclosporine and corticosteroids, transient responses were observed, whereas cyclosporine alone has been reported to cause a sustained response in 2 out of 10 patients [71-78]. Interestingly, the mechanisms of action of the two groups of drugs are different: the drugs of the first group (6-mercaptopurine, cyclophosphamide, vincristine, intravenous immunoglobulin, and anti-thymocyte globulin) have been shown to directly target the T cells, inhibiting their proliferation [79-83], whereas cyclosporine is known to have a milder effect, and modulate the expression of cytokines in T-Lymphocytes and in other cell types [84,85].

Conclusion

Today, 40% of patients with DBA receive steroid treatment, and the prolonged duration of steroid therapy is associated with the risk of side effects [4]; thus, the identification of new compounds that can be used to treat DBA is a priority.

Taken together, the evidence seems to indicate that the therapeutic effect of GCs in DBA could also be linked to the inhibition of cytokine function. In this hypothesis, some cytokines are specifically produced and interfere with normal erythropoiesis in DBA patient erythroid progenitors. This cytokine-mediated hypothesis of DBA is supported by in vitro experiments showing that normal erythroid progenitors secrete cytokines/chemokines, thus regulating hematopoiesis in an autocrine/paracrine manner [69].

SOCS proteins are physiological suppressors of cytokine signaling [86]. Proliferation induction and differentiation arrest caused by the GR mainly depends on mechanisms involving transactivation [18], and by inducing SOCS1 up-regulation in erythroid progenitors [50,55,59], GCs could also act in favor of an inhibition of cytokine pathways. At the same time, this proposition does not exclude the possibility that transrepression (dependent on activations between GR and AP1 or NF-κB) also occurs when erythroid progenitors are subjected to GCs treatment. Since several members of NF-κB family are also expressed [45] in erythroid progenitors, GCs could then inhibit NF-κB-induced cytokines expression, as they do in many other types of cells [41]. Finally, the acceleration of chemokine mRNAs decay by direct interaction between the GR and chemokine mRNAs in human airway epithelial cell line BEAS-2B helps to better understand the rapid action of GCs [68]. This specific degradation of chemokine mRNAs by GCs might partially explain the beneficial effect of GCs treatment in DBA.

These general conclusions on the functional links between cytokines and glucocorticoids should thus be confirmed in the specific field of DBA, in order to determine if drugs targeting cytokines could indeed pave the way for new treatments in DBA.

Acknowledgements

The author is eternally grateful to Dr. Kathleen Sakamoto for her support, invaluable discussions, and her help in preparing this manuscript, Nicole Clarke, Hee-Don Chae, and Grace Aldana-Masangkay for helpful comments on the manuscript, Elena Bibikova, and Michael Strohman for fruitful discussions.

References

  1. Josephs HW (1936) Anaemia of Infancy and Early Childhood. Medicine 15: 307-451.
  2. Diamond LK, Blackfan K (1938) Hypoplastic anemia. American Journal of Diseases of Children 56: 464–467.
  3. Lydie DC, Geneviève CP, Maud S, Martine F, Raymonde B, et al. (2013) First de novo mutation in RPS19 gene as the cause of hydrops fetalis in Diamond-Blackfan anemia. Am J Hematol 88: 160.
  4. Vlachos A, Ball S, Dahl N, Alter BP, Sheth S, et al. (2008) Diagnosing and treating Diamond Blackfan anaemia: results of an international clinical consensus conference. Br J Haematol 142: 859-876.
  5. Gasser C (1951) Aplastische Anamie (chronische Erythroblastophthise) und Cortison. Schweizerische Medizinische Wochenschrift 81: 1241–1242.
  6. Hoffman R, Zanjani ED, Vila J, Zalusky R, Lutton JD, et al. (1976) Diamond-Blackfan syndrome: lymphocyte-mediated suppression of erythropoiesis. Science 193: 899-900.
  7. Horos R. Von Lindern M (2012) Molecular mechanisms of pathology and treatment in Diamond Blackfan Anaemia. Br J Haematol 159: 514-527.
  8. Draptchinskaia N, Gustavsson P, Andersson B, Pettersson M, Willig TN, et al. (1999) The gene encoding ribosomal protein S19 is mutated in Diamond-Blackfan anaemia. Nat Genet 21: 169-175.
  9. Hamaguchi I, Ooka A, Brun A, Richter J, Dahl N, et al, (2002) Gene transfer improves erythroid development in ribosomal protein S19-deficient Diamond-Blackfan anemia. Blood 100: 2724-2731.
  10. Flygare J, Kiefer T, Miyake K, Utsugisawa T, Hamaguchi I, et al, (2005) Deficiency of ribosomal protein S19 in CD34+ cells generated by siRNA blocks erythroid development and mimics defects seen in Diamond-Blackfan anemia. Blood 105: 4627-34.
  11. Flygare J, Olsson K, Richter J, Karlsson S (2008) Gene therapy of Diamond Blackfan anemia CD34(+) cells leads to improved erythroid development and engraftment following transplantation. Exp Hematol 36: 1428-1435.
  12. Taylor AM, Humphries JM, White RM, Murphey RD, Burns CE, et al. (2012) Hematopoietic defects in rps29 mutant zebrafish depend upon p53 activation. Exp Hematol 40: 228-237 e5.
  13. Torihara H, Uechi T, Chakraborty A, Shinya M, Sakai N, et al. (2011) Erythropoiesis failure due to RPS19 deficiency is independent of an activated Tp53 response in a zebrafish model of Diamond-Blackfan anaemia. Br J Haematol 152: 648-654.
  14. Danilova N, Sakamoto KM, Lin S (2011) Ribosomal protein L11 mutation in zebrafish leads to haematopoietic and metabolic defects. Br J Haematol 152: 217-28.
  15. Vlachos A, Dahl N, Dianzani I, Lipton JM (2013) Clinical utility gene card for: Diamond - Blackfan anemia - update. Eur J Hum Genet.
  16. Schmidt S, Rainer J, Ploner C, Presul E, Riml S,et al. (2004) Glucocorticoidinduced apoptosis and glucocorticoid resistance: molecular mechanisms and clinical relevance. Cell Death Differ Suppl 1: S45-55.
  17. Liles WC, Dale DC, Klebanoff SJ (1995) Glucocorticoids inhibit apoptosis of human neutrophils. Blood 86: 3181-3188.
  18. Wessely O, Deiner EM, Beug H, Von Lindern M (1997) The glucocorticoid receptor is a key regulator of the decision between self-renewal and differentiation in erythroid progenitors. EMBO J 16: 267-80.
  19. Migliaccio G, Di Pietro R, di Giacomo V, Di Baldassarre A, Migliaccio AR, et al. (2002) In vitro mass production of human erythroid cells from the blood of normal donors and of thalassemic patients. Blood Cells Mol Dis 28: 169-180.
  20. Chrousos GP, Charmandari E, Kino T (2004) Glucocorticoid action networks-an introduction to systems biology. J Clin Endocrinol Metab,. 89: 563-564.
  21. Nicolaides NC, Galata Z, Kino T, Chrousos GP, Charmandari E (2010) The human glucocorticoid receptor: molecular basis of biologic function. Steroids 75: 1-12.
  22. Buckingham JC (2006) Glucocorticoids: exemplars of multi-tasking. Br J Pharmacol 147 Suppl 1: S258-268.
  23. Golde DW, Bersch N, Cline MJ (1976) Potentiation of erythropoiesis in vitro by dexamethasone. J Clin Invest 57: 57-62.
  24. Udupa KB, Crabtree HM, Lipschitz DA (1986) In vitro culture of proerythroblasts: characterization of proliferative response to erythropoietin and steroids. Br J Haematol 62: 705-714.
  25. Fisher JW (1998) A quest for erythropoietin over nine decades. Annu Rev Pharmacol Toxicol 38: 1-20.
  26. Miller WL, Tyrrell JB (1995) The Adrenal Cortex. Endocrinology and metabolism. (ed. P. Fehlig, J.D. Baxter, and L.A. Frohman) 555-571.
  27. Cole TJ, Blendy JA, Monaghan AP, Krieglstein K, Schmid W, et al. (1995) Targeted disruption of the glucocorticoid receptor gene blocks adrenergic chromaffin cell development and severely retards lung maturation. Genes Dev 9: 1608-21.
  28. De Kloet ER, Vreugdenhil E, Oitzl MS, Joëls M (1998) Brain corticosteroid receptor balance in health and disease. Endocr Rev 19: 269-301.
  29. Yudt MR, Cidlowski JA (2002) The glucocorticoid receptor: coding a diversity of proteins and responses through a single gene. Mol Endocrinol 16: 1719-1726.
  30. Yudt MR, Jewell CM, Bienstock RJ, Cidlowski JA (2003) Molecular origins for the dominant negative function of human glucocorticoid receptor beta. Mol Cell Biol 23: 4319-30.
  31. Leung DY, Hamid Q, Vottero A, Szefler SJ, Surs W et al. (1997) Association of glucocorticoid insensitivity with increased expression of glucocorticoid receptor beta. J Exp Med 186: 1567-74.
  32. Oakley, RH, Sar M, Cidlowski JA (1996) The human glucocorticoid receptor beta isoform. Expression, biochemical properties, and putative function. J Biol Chem 271: 9550-9559.
  33. Bamberger CM, Bamberger AM, de Castro M, Chrousos GP (1995) Glucocorticoid receptor beta, a potential endogenous inhibitor of glucocorticoid action in humans. J Clin Invest 95: 2435-2441.
  34. Hecht K, Carlstedt DJ, Stierna P, Gustafsson J, Bronnegard M, et al. (1997) Evidence that the beta-isoform of the human glucocorticoid receptor does not act as a physiologically significant repressor. J Biol Chem 272: 26659-26664.
  35. Van Rossum EF, Lamberts SW (2004) Polymorphisms in the glucocorticoid receptor gene and their associations with metabolic parameters and body composition. Recent Prog Horm Res 59: 333-357.
  36. Pratt WB (1993) The role of heat shock proteins in regulating the function, folding, and trafficking of the glucocorticoid receptor. J Biol Chem 268: 21455-21458.
  37. Bamberger CM, Schulte HM, Chrousos GP (1996) Molecular determinants of glucocorticoid receptor function and tissue sensitivity to glucocorticoids. Endocr Rev 17: 245-261.
  38. Gottlicher M, Heck S, Herrlich P (1998) Transcriptional cross-talk, the second mode of steroid hormone receptor action. J Mol Med (Berl) 76: 480-489.
  39. Reichardt HM, Tuckermann JP, Gottlicher M, Vujic M, Weih F, et al. (2001) Repression of inflammatory responses in the absence of DNA binding by the glucocorticoid receptor. EMBO J 20: 7168-7173.
  40. Karin M, Chang L (2001) AP-1--glucocorticoid receptor crosstalk taken to a higher level. J Endocrinol 169: 447-451.
  41. Barnes PJ, Karin M (1997) Nuclear factor-kappaB: a pivotal transcription factor in chronic inflammatory diseases. N Engl J Med 336: 1066-1071.
  42. Didonato JA, Saatcioglu F, Karin M (1996) Molecular mechanisms of immunosuppression and anti-inflammatory activities by glucocorticoids. Am J Respir Crit Care Med 154: S11-15.
  43. Beato, M., P. Herrlich, and Schutz G (1995) Steroid hormone receptors: many actors in search of a plot. Cell 83: 851-857.
  44. Heck S, Kullmann M, Gast A, Ponta H, Rahmsdorf HJ, et al. (1994) A distinct modulating domain in glucocorticoid receptor monomers in the repression of activity of the transcription factor AP-1. EMBO J 13: 4087-4095.
  45. Zhang MY, Sun SC, Bell L, Miller BA (1998) NF-kappaB transcription factors are involved in normal erythropoiesis. Blood 91: 4136-4144.
  46. Reichardt HM, Kaestner KH, Tuckermann J, Kretz O, Wessely O, et al. (1998) DNA binding of the glucocorticoid receptor is not essential for survival. Cell 93: 531-541.
  47. Bauer A, Tronche F, Wessely O, Kellendonk C, Reichardt HM, et al. (1999) The glucocorticoid receptor is required for stress erythropoiesis. Genes Dev 13: 2996-3002.
  48. Strahle U, Klock G, Schutz G (1987) A DNA sequence of 15 base pairs is sufficient to mediate both glucocorticoid and progesterone induction of gene expression. Proc Natl Acad Sci USA 84: 7871-7875.
  49. Meijsing SH, Pufall MA, So AY, Bates DL, Chen L, et al. (2009) DNA binding site sequence directs glucocorticoid receptor structure and activity. Science 324: 407-410.
  50. Flygare J, Rayon Estrada V, Shin C, Gupta S, Lodish HF (2011) HIF1alpha synergizes with glucocorticoids to promote BFU-E progenitor self-renewal. Blood 117: 3435-44.
  51. Ebert BL, Lee MM, Pretz JL, Subramanian A, Mak R,et al. (2005) An RNA interference model of RPS19 deficiency in Diamond-Blackfan anemia recapitulates defective hematopoiesis and rescue by dexamethasone: identification of dexamethasone-responsive genes by microarray. Blood 105: 4620-4626.
  52. Graham GJ, Wright EG, Hewick R, Wolpe SD, Wilkie NM, et al. (1990) Identification and characterization of an inhibitor of haemopoietic stem cell proliferation. Nature 344: 442-444.
  53. Von Lindern M, Zauner W, Mellitzer G, Steinlein P, Fritsch G, et al. (1999) The glucocorticoid receptor cooperates with the erythropoietin receptor and c-Kit to enhance and sustain proliferation of erythroid progenitors in vitro. Blood 94: 550-559.
  54. Dolznig H, Habermann B, Stangl K, Deiner EM, Moriggl R, et al. (2002) Apoptosis protection by the Epo target Bcl-X(L) allows factor-independent differentiation of primary erythroblasts. Curr Biol 12: 1076-1085.
  55. Kolbus A, Blázquez DM, Carotta S, Bakker W, Luedemann S, et al. (2003) Cooperative signaling between cytokine receptors and the glucocorticoid receptor in the expansion of erythroid progenitors: molecular analysis by expression profiling. Blood 102: 3136-3146.
  56. Rouault JP, Rimokh R, Tessa C, Paranhos G, Ffrench M, et al. (1992) BTG1, a member of a new family of antiproliferative genes. EMBO J 11: 1663-1670.
  57. Riccardi C, Cifone MG, Migliorati G (1999) Glucocorticoid hormone-induced modulation of gene expression and regulation of T-cell death: role of GITR and GILZ, two dexamethasone-induced genes. Cell Death Differ 6: 1182-1189.
  58. Mittelstadt PR, Ashwell JD (2001) Inhibition of AP-1 by the glucocorticoidinducible protein GILZ. J Biol Chem 276: 29603-29610.
  59. Narla A, Dutt S, McAuley JR, Al-Shahrour F, Hurst S, et al. (2011) Dexamethasone and lenalidomide have distinct functional effects on erythropoiesis. Blood 118: 2296-304.
  60. Metcalf D, Alexander WS, Elefanty AG, Nicola NA, Hilton DJ, et al. (1999) Aberrant hematopoiesis in mice with inactivation of the gene encoding SOCS-1. Leukemia 13: 926-934.
  61. Ingley E (2012) Integrating novel signaling pathways involved in erythropoiesis. IUBMB Life 64: 402-410.
  62. Russell RC, Sufan RI, Zhou B, Heir P, Bunda S, et al. (2011) Loss of JAK2 regulation via a heterodimeric VHL-SOCS1 E3 ubiquitin ligase underlies Chuvash polycythemia. Nat Med 17: 845-853.
  63. Messinezy M, Pearson TC (1999) The classification and diagnostic criteria of the erythrocytoses (polycythaemias). Clin Lab Haematol 21: 309-316.
  64. Goulding NJ (2004) The molecular complexity of glucocorticoid actions in inflammation - a four-ring circus. Curr Opin Pharmacol 4: 629-636.
  65. Tasker JG, Di S, Malcher LR (2006) Minireview: rapid glucocorticoid signaling via membrane-associated receptors. Endocrinology147: 5549-56.
  66. Stellacci E, Di Noia A, Di Baldassarre A, Migliaccio G, Battistini A, et al.(2009) Interaction between the glucocorticoid and erythropoietin receptors in human erythroid cells. Exp Hematol 37: 559-572.
  67. Anderson P (2008) Post-transcriptional control of cytokine production. Nat Immunol 9: 353-359.
  68. Ishmael FT, Fang X, Houser KR, Pearce K, Abdelmohsen K, et al. (2011) The human glucocorticoid receptor as an RNA-binding protein: global analysis of glucocorticoid receptor-associated transcripts and identification of a target RNA motif. J Immunol 186: 1189-98.
  69. Majka M, Janowska WA, Ratajczak J, Ehrenman K, Pietrzkowski Z, et al. (2001) Numerous growth factors, cytokines, and chemokines are secreted by human CD34(+) cells, myeloblasts, erythroblasts, and megakaryoblasts and regulate normal hematopoiesis in an autocrine/paracrine manner. Blood 97: 3075-3085.
  70. Barnes PJ (2001) Molecular mechanisms of corticosteroids in allergic diseases. Allergy 56: 928-936.
  71. Alessandri AJ, Rogers PC, Wadsworth LD, Davis JH (2000) Diamondblackfan anemia and cyclosporine therapy revisited. J Pediatr Hematol Oncol 22: 176-179.
  72. Bobey NA, Carcao M, Dror Y, Freedman MH, Dahl N, et al. (2003) Sustained cyclosporine-induced erythropoietic response in identical male twins with diamond-blackfan anemia. J Pediatr Hematol Oncol 25: 914-918.
  73. Leonard EM, Raefsky E, Griffith P, Kimball J, Nienhuis AW, et al. (1989) Cyclosporine therapy of aplastic anaemia, congenital and acquired red cell aplasia. Br J Haematol 72: 278-84.
  74. Garcia Vela JA, Monteserin MC, Oña F, Barea LM, Lastra A, et al. (1993) Cyclosporine A used as a single drug in the treatment of pure red cell aplasia associated with thymoma. Am J Hematol 42: 238-239.
  75. Seip M, Zanussi GF (1988) Cyclosporine in steroid-resistant Diamond- Blackfan anaemia. Acta Paediatr Scand 77: 464-466.
  76. Splain J, Berman BW (1992) Cyclosporin A treatment for Diamond-Blackfan anemia. Am J Hematol 39: 208-211.
  77. Tötterman TH, Nisell J, Killander A, Gahrton G, Lönnqvist B (1984) Successful treatment of pure red-cell aplasia with cyclosporin. Lancet 2: 693.
  78. Williams DL, Mageed AS, Findley H, Ragab AH (1987) Cyclosporine in the treatment of red cell aplasia. Am J Pediatr Hematol Oncol 9: 314-316.
  79. Atreya I, Neurath MF (2009) Understanding the delayed onset of action of azathioprine in IBD: are we there yet? Gut 58: 325-326.
  80. Qiu R, Kalhorn TF, Slattery JT (2004) ABCC2-mediated biliary transport of 4-glutathionylcyclophosphamide and its contribution to elimination of 4-hydroxycyclophosphamide in rat. J Pharmacol Exp Ther 308: 1204-1212.
  81. Thomadaki H, Floros KV, Scorilas (2009) A Molecular response of HL-60 cells to mitotic inhibitors vincristine and taxol visualized with apoptosis-related gene expressions, including the new member BCL2L12. Ann N Y Acad Sci 1171: 276-283.
  82. Sewell WA, Jolles S (2002) Immunomodulatory action of intravenous immunoglobulin. Immunology 107: 387-393.
  83. Duftner C, Dejaco C, Hengster P, Bijuklic K, Joannidis M, et al. (2012) Apoptotic effects of antilymphocyte globulins on human pro-inflammatory CD4+CD28- T-cells. PLoS One 7: e33939.
  84. Khanna A, Li B, Stenzel KH, Suthanthiran M (1994) Regulation of new DNA synthesis in mammalian cells by cyclosporine. Demonstration of a transforming growth factor beta-dependent mechanism of inhibition of cell growth. Transplantation 57: 577-582.
  85. Matsuda S, Koyasu S (2000) Mechanisms of action of cyclosporine.Immunopharmacology 47: 119-125.
  86. Krebs DL, Hilton DJ (2000) SOCS: physiological suppressors of cytokine signaling. J Cell Sci 113: 2813-2819.