Trends in Cell Biology
Volume 15, Issue 3, March 2005, Pages 146-155
Journal home page for Trends in Cell Biology

Turning cells red: signal transduction mediated by erythropoietin

https://doi.org/10.1016/j.tcb.2005.01.007Get rights and content

Erythropoietin (EPO) is the crucial cytokine regulator of red blood-cell production. Since the discovery of EPO in 1985 and the isolation of its cognate receptor four years later, there has been significant interest in understanding the unique ability of this ligand–receptor pair to promote erythroid mitogenesis, survival and differentiation. The development of knockout mice has elucidated the precise role of the ligand, receptor and downstream players in murine erythroid development. In this review, we summarize EPO-mediated signaling pathways and examine their significance in vivo.

Introduction

The identification of erythropoietin (EPO) dramatically improved the quality of life of patients suffering from chronic kidney disease and EPO is the world's leading biopharmaceutical [1]. EPO is the crucial regulator of red blood-cell production and delivers essential growth, differentiation and survival signals to erythroid progenitors in the foetal liver, bone marrow and spleen (Figure 1; Box 1). The biosynthesis of EPO is regulated by oxygen tension and many details of this cascade have been elucidated recently (reviewed in [2]).

The EPO receptor (EPO-R), which was described initially in 1989 [3], is a member of the cytokine receptor superfamily. The EPO-R is expressed in bone marrow-derived erythroid progenitors and several non-hematopoietic tissues including myocytes, cortical neurons, and prostatic, breast and ovarian epithelia. The EPO-R is believed to exist in an unliganded, dimeric state. On binding EPO, it undergoes a conformational change that activates the pre-bound, cytoplasmic, tyrosine kinase, Janus kinase 2 (JAK2) [4]. In turn, JAK2 (and, potentially, other tyrosine kinases) phosphorylate several cytoplasmic tyrosine residues in the cytoplasmic tail of the EPO-R that act as docking sites for proteins that contain Src-homology 2 (SH2) domains. EPO activates tyrosine phosphorylation of the SH2 domain-containing transcription factors, signal transducer and activator of transcription 1 (STAT1), STAT3 and STAT5a/b. The number of citations that involve the EPO-R is >1000 and we know much about the plethora of signaling proteins that are recruited to and/or activated downstream of the EPO-R.

The significance of these proteins in mediating erythropoiesis is known because of the availability of knockout mice. In this review, we discuss EPO-mediated signaling pathways with an emphasis on recent developments that exploit knockout mice to delineate crucial steps in erythroid maturation. It updates our knowledge about the relevant players in mouse erythroid development in vivo and is relevant for those interested in hematopoiesis, cytokine biology and signal transduction. Earlier work is discussed in several excellent reviews 5, 6, 7.

Section snippets

Mechanism of activation of the EPO-R

In this section, we discuss the domain architecture and regulation of the murine EPO-R, which is a membrane protein with 507 amino-acids and a single transmembrane domain (Figure 2). The extracellular domain of the EPO-R has two fibronectin type II domains. Crystallographic studies demonstrate that the EPO-R extracellular domain exists as a dimer in the absence of EPO. A high-affinity receptor is generated when one EPO molecule binds to a preformed EPO-R dimer. Binding of EPO (or other

Gene targeting reveals non-redundant roles for EPO, EPO-R and JAK2 in erythropoiesis

The physiological role of EPO [13] and EPO-R 13, 14, 15 in erythropoiesis has been determined, in part, using knockout mice. Mice with null mutations in genes encoding either EPO or EPO-R die during embryogenesis at embryonic day E12.5–E13.5, a stage of massive erythroid expansion. However, intact colony forming unit-erythroid (CFU-E) cells are obtained, indicating that EPO signaling is dispensable at earlier stages of hematopoiesis including the burst forming unit-erythroid (BFU-E) (Figure 1).

EPO-dependent tyrosine kinase activation: JAK2 and friends

Higher mammals express four JAKs. EPO stimulation leads to robust tyrosine phosphorylation of JAK2, which is time and dose dependent [4]. All JAK kinases have seven unique domains, termed JAK-homology (JH) domains (Figure 3). A catalytically active tyrosine kinase domain (JH1) and adjacent pseudokinase domain (JH2) are located at the C terminus. Although the pseudokinase domain has the architecture of a tyrosine kinase domain, key catalytic residues are not conserved and it might have a

Lyn

Lyn and other Src family tyrosine kinases are substrates of EPO signaling. Evidence of their involvement has emerged following the isolation of a human erythroid cell line, J2E-NR, which is Lyn-deficient and has impaired erythroid differentiation [30]. The J2E-NR cell line has decreased concentrations of the erythroid transcription factors GATA-1 and erythroid kruppel-like factor (EKLF), and STAT5a [31]. Similarly, erythroblasts from Lyn-deficient mice do not express GATA-1 and EKLF, and

Btk

Several reports have examined the activation of Tec family tyrosine kinases, including Btk, following EPO-receptor activation. Although Btk-knockout mice have no overt defect in erythropoiesis, evidence indicates that Btk is crucial for EPO-mediated signaling. Erythroblasts isolated from Btk-deficient mice have enhanced erythroid differentiation, even when cultured in media that supports self-renewal rather than differentiation [32]. Tyrosine phosphorylation of STAT5 is delayed following EPO

Activation of EPO and STAT

Seven members of the STAT family are expressed in mammals. These proteins are SH2-domain-containing transcription factors that play an integral role in cytokine signaling. Studies utilizing cells in culture show that EPO activates STAT1, STAT3 and STAT5a/b (Figure 4) 5, 6, 7. Analysis of erythropoiesis is complete in STAT1-deficient and STAT5a/b-knockout mice.

Studies of cell lines illustrate that EPO-dependent activation of STAT1 is dependent on JAK2 but does not require cytoplasmic tyrosines

EPO activates Erk1/2, SAP kinase/Jun kinase and p38

EPO activates the catalytic activity of several mitogen-activated protein kinases, including extracellular-regulated kinases 1/2 (Erk1/2), SAP kinase (SAPK)/Jun kinase (Jnk) and p38. Erk1/2 is thought to participate in mitogenesis, whereas the roles of SAPK/Jnk and p38 are more complicated.

Many growth factors and cytokines activate Ras by recruiting Grb2-Sos to their receptors. There is significant redundancy in how the Grb2-Sos exchange complex is recruited to the EPO-R. For example, Grb2 can

Cell survival mediated through the Bcl family

Proteins of the B cell lymphoma 2 (Bcl2) family are key effectors of survival signals mediated by growth factor receptors. Several studies have shown that Bcl2 and B cell lymphoma-XL (Bcl-XL) protect erythroid cell lines and primary erythroid cells from apoptosis. Bcl2-knockout mice have no overt erythroid abnormalities, probably because Bcl2 is not expressed in erythroid progenitors. However, Bcl-XL-knockout mice display embryonic lethality at E12.5 because of brain defects and severe anaemia

EPO and Ca2+

Unlike antigen receptors, cytokine receptors do not have a rapid, phospholipase Cγ-mediated Ca2+ flux on receptor engagement. However, Ca2+ does play a role in erythroid development: Ca2+ influx is required for murine erythroleukemia cell differentiation and intracellular Ca2+ increases in human BFU-E culture (reviewed in [7]).

Utilizing a series of deletion and tyrosine mutants of the EPO-R, Miller and colleagues have demonstrated that Y460 of the EPO-R is necessary and sufficient for

Turning signals off

Although the signals that regulate mitogenesis are well understood, less is known about the mechanisms of negative regulation. This discussion is limited to SOCS, tyrosine phosphatases and inositol phosphatases because these are the best-studied negative regulators (Figure 6).

SOCS

The SOCS family of proteins, which was discovered in 1997, plays a role in negative regulation of cytokine signaling. The eight family members have a unique N-terminal region, a central SH2 domain and a C-terminal SOCS box.

SOCS proteins are transcriptionally regulated immediate early genes that feedback on EPO-signaling pathways. Induction of cytokine-inducible SH2 domain (Cis), SOCS-1 and SOCS-3 mRNA transcripts is observed in cell lines and primary cells that have been stimulated with EPO [63]

Tyrosine phosphatases

Four tyrosine phosphatases are involved in regulating erythropoiesis. The SH2-domain containing phosphatases, Shp1 and Shp2, cluster designation 45 (CD45) and protein tyrosine phosphatase 1B (PTP-1B) all modulate activity of JAK2 kinase (Figure 6).

The motheaten (me) mouse has germline mutations in Shp1 (reviewed in [73]). Erythroid progenitors from mev/mev mice are either hypersensitive to EPO or show EPO independent-growth. Importantly, Shp1 associates with Y429 and Y431 of the EPO-R [74].

The

Inositol phosphatases

Another manner of desensitization involves the regulation of metabolic intermediates of inositol. Phosphatidylinositol (3,4,5)-trisphosphate [PtdIns(3,4,5)P3] activates many crucial signaling events. Several enzymes dephosphorylate PtdIns(3,4,5)P3, including phosphatase and tensin homolog (which dephosphorylate the 3′ phosphate) and SH2-inositol phosphatase, SHIP-1 and SHIP-2 (which dephosphorylate the 5′ phosphate). SHIP-1 is recruited to Y401 of the EPO-R [41] and SHIP-1 null mice show

Concluding remarks

Many studies provide details of the intricacies of signaling events that are activated by EPO. Analysis of erythropoiesis in gene-targeted mice has allowed determination of the signaling events that are required for normal erythropoiesis. Although the importance of EPO, EPO-R and JAK2 is undeniable in foetal erythropoiesis, other signaling proteins might have important roles in signal integration in adults. These underlying effects might only be observed under conditions of erythroid stress.

Acknowledgements

Research in the author's laboratory is supported by Canadian Institutes of Health Research, National Cancer Institute of Canada and Leukemia and Lymphoma Society. DLB is a National Cancer Institute of Canada Research Scientist. We thank Sam Benchimol, Martin Carroll and Madeleine Bonnard for helpful comments on the manuscript and apologize to many colleagues whose contributions we were unable to list because of reference citation limitations.

References (81)

  • H. Wu

    Generation of committed erythroid BFU-E and CFU-E progenitors does not require erythropoietin or the erythropoietin receptor

    Cell

    (1995)
  • E. Parganas

    Jak2 is essential for signaling through a variety of cytokine receptors

    Cell

    (1998)
  • H. Neubauer

    Jak2 deficiency defines an essential developmental checkpoint in definitive hematopoiesis

    Cell

    (1998)
  • K. Li

    Attenuated signaling by a phosphotyrosine-null Epo receptor form in primary erythroid progenitor cells

    Blood

    (2003)
  • N. Suzuki

    Erythroid-specific expression of the erythropoietin receptor rescued its null mutant mice from lethality

    Blood

    (2002)
  • A.C. Heinrich

    A mouse model for visualization and conditional mutations in the erythroid lineage

    Blood

    (2004)
  • M. von Lindern

    The glucocorticoid receptor cooperates with the erythropoietin receptor and c-Kit to enhance and sustain proliferation of erythroid progenitors in vitro

    Blood

    (1999)
  • S. Carotta

    Directed differentiation and mass cultivation of pure erythroid progenitors from mouse embryonic stem cells

    Blood

    (2004)
  • A. Halupa

    A novel role for STAT1 in regulating murine erythropoiesis: deletion of STAT1 results in overall reduction of erythroid progenitors and alters their distribution

    Blood

    (2005)
  • K. Kirito

    Identification of the human erythropoietin receptor region required for Stat1 and Stat3 activation

    Blood

    (2002)
  • C.K. Lee

    STAT3 is a negative regulator of granulopoiesis but is not required for G-CSF-dependent differentiation

    Immunity

    (2002)
  • S. Teglund

    Stat5a and Stat5b proteins have essential and nonessential, or redundant, roles in cytokine responses

    Cell

    (1998)
  • M. Socolovsky

    Fetal anemia and apoptosis of red cell progenitors in Stat5a−/−5b−/− mice: a direct role for Stat5 in Bcl-X(L) induction

    Cell

    (1999)
  • H.L. Bradley

    Cell intrinsic defects in cytokine responsiveness of STAT5-deficient hematopoietic stem cells

    Blood

    (2002)
  • D.L. Barber

    Erythropoietin activates Raf1 by an Shc-independent pathway in CTLL-EPO-R cells

    Blood

    (1997)
  • J.M. Mason

    The SH2 inositol 5-phosphatase Ship1 is recruited in an SH2-dependent manner to the erythropoietin receptor

    J. Biol. Chem.

    (2000)
  • T. Tauchi

    Tyrosine 425 within the activated erythropoietin receptor binds Syp, reduces the erythropoietin required for Syp tyrosine phosphorylation, and promotes mitogenesis

    Blood

    (1996)
  • R. Haq

    Regulation of erythropoietin-induced STAT serine phosphorylation by distinct mitogen-activated protein kinases

    J. Biol. Chem.

    (2002)
  • S.M. Jacobs-Helber

    JNK and p38 are activated by erythropoietin (EPO) but are not induced in apoptosis following EPO withdrawal in EPO-dependent HCD57 cells

    Blood

    (2000)
  • K. Tamura

    Requirement for p38alpha in erythropoietin expression: a role for stress kinases in erythropoiesis

    Cell

    (2000)
  • H. Bao

    Protein kinase B (c-Akt), phosphatidylinositol 3-kinase, and STAT5 are activated by erythropoietin (EPO) in HCD57 erythroid cells but are constitutively active in an EPO-independent, apoptosis-resistant subclone (HCD57-SREI cells)

    Blood

    (1999)
  • H. Huddleston

    Functional p85alpha gene is required for normal murine fetal erythropoiesis

    Blood

    (2003)
  • D. Bouscary

    Critical role for PI 3-kinase in the control of erythropoietin-induced erythroid progenitor proliferation

    Blood

    (2003)
  • Y. Kashii

    A member of Forkhead family transcription factor, FKHRL1, is one of the downstream molecules of phosphatidylinositol 3-kinase-Akt activation pathway in erythropoietin signal transduction

    Blood

    (2000)
  • H. Dolznig

    Apoptosis protection by the Epo target Bcl-X(L) allows factor-independent differentiation of primary erythroblasts

    Curr. Biol.

    (2002)
  • J.N. Ihle

    The Stat family in cytokine signaling

    Curr. Opin. Cell Biol.

    (2001)
  • M. Socolovsky

    Ineffective erythropoiesis in Stat5a(−/−)5b(−/−) mice due to decreased survival of early erythroblasts

    Blood

    (2001)
  • B.A. Miller

    Identification of the erythropoietin receptor domain required for calcium channel activation

    J. Biol. Chem.

    (1999)
  • X. Chu

    Erythropoietin modulates calcium influx through TRPC2

    J. Biol. Chem.

    (2002)
  • X. Chu

    Interaction of TRPC2 and TRPC6 in erythropoietin modulation of calcium influx

    J. Biol. Chem.

    (2004)
  • Cited by (291)

    • Erythropoietin and skeletal cells crosstalks in physiology and disease

      2023, Current Opinion in Endocrine and Metabolic Research
    • Iron-loaded transferrin potentiates erythropoietin effects on erythroblast proliferation and survival: a novel role through transferrin receptors

      2021, Experimental Hematology
      Citation Excerpt :

      Thus, it has been used during the treatment of anemia from several causes [18]. Production of red blood cells is dependent on EpoR-mediated pathways, including STAT5, MAPK/ERK, and PI3K/AKT [19]. In addition to transferrin, TfR1 is a receptor for pIgA1.

    View all citing articles on Scopus
    View full text