As essential mediators of red cell production, erythropoietin (EPO) and its cell surface receptor (EPO receptor [EPOR]) have been intensely studied. Early investigations defined basic mechanisms for hypoxia-inducible factor induction of EPO expression, and within erythroid progenitors EPOR engagement of canonical Janus kinase 2/signal transducer and activator of transcription 5 (JAK2/STAT5), rat sarcoma/mitogen-activated protein kinase/extracellular signal-regulated kinase (RAS/MEK/ERK), and phosphatidylinositol 3-kinase (PI3K) pathways. Contemporary genetic, bioinformatic, and proteomic approaches continue to uncover new clinically relevant modulators of EPO and EPOR expression, and EPO’s biological effects. This Spotlight review highlights such factors and their emerging roles during erythropoiesis and anemia.

Early parabiotic experiments with anemic and nephrectomized rats predicted the existence of erythropoietin (EPO) as a blood-borne kidney-derived activator of erythropoiesis.1  Evidence that EPO occurs as a unique glycoprotein hormone was further advanced via arduous fractionations and bioassays of urinary proteins from anemia patients.2  The purification, partial sequencing, and cloning of erythropoietin3  have led to the generation of recombinant human EPO (rhEPO) (and derivatives) for the treatment of anemia associated with chronic kidney disease, chemotherapy, and low-risk myelodysplastic syndrome.4  The subsequent discovery of the EPO receptor (EPOR) as a plasma membrane single-pass homodimer5,6  elevated the EPO/EPOR system as a paradigm for hematopoietic cytokine receptor action. The EPOR, for example, was among the first discovered to associate with a Janus kinase (JAK),7  to transduce signals via transmembrane conformational mechanisms,8  and to be causally associated with polycythemia.9  EPO’s clinical and scientific successes have prompted in-depth investigations into EPO/EPOR biology. This review focuses on intriguing advances in understanding the regulation of EPO and EPOR expression, and the nature of novel EPO/EPOR signals that regulate erythroid progenitor cell (EPC) development. EPO also has been reported to exert survival, proliferative, and/or developmental effects in a wide range of nonhematopoietic tissues.10-16  In such cell types, however, EPOR protein expression (including cell surface levels) can be nominal,17  thereby complicating interpretations for direct vs indirect effects. Nonetheless, incisive EPOR loss-of-function approaches have revealed interesting EPO effects in cardiomyocyte mitochondrial biogenesis,18  retinal cell cytoprotection,19  melanoma cell survival,20  and adipogenesis.21  This broad area of investigation, however, lies beyond the scope of the present report.

The nature of rare Epo-producing cells is first becoming more clearly defined. During primitive erythropoiesis, studies using an Epo gene green fluorescent protein knock-in mouse indicate predominant Epo expression by neural crest and neuroepithelial cells.22  Tracking studies of myelin protein-zero23  marked peripheral neural cells demonstrate that Epopos embryonic neural crest fibroblasts migrate to the kidney,24  and perinatally reside within peritubular interstitia.25  Renal fibrosis due to ureteral obstruction can promote transdifferentiation of Epohigh fibroblasts to Epolow myofibroblasts.24 Epo levels in myofibroblasts can be increased, however, via neurotropin or dexamethasone dosing. During stress erythropoiesis, Epo expression can also be induced in the liver,26  as well as bone marrow (BM) osteoblasts as demonstrated upon von Hippel–Lindau factor (VHL) inactivation.27 

New insight has also been gained into EPO gene regulation. Early investigations of hypoxia-induced EPO expression established important roles for a downstream EPO enhancer (E-3′) as a binding site for hypoxia-inducible factor (HIF) and hepatocyte nuclear factor 4 transcriptional regulators.28  In vivo studies in mice with a green fluorescent protein-marked Epo allele demonstrate that E-3′ deletion results in embryonic and neonatal anemia.25,26  In juvenile and adult kidney, however, Epo production is unexpectedly regained in the absence of E-3′,26  whereas hepatic Epo production continues to depend upon E-3′ effects.26  For renal EPO production, this raises new questions concerning activation mechanisms.

Among HIF1α, -2α, and -3α, HIF2α has been defined as a prime component of an EPO gene activating complex.29  New insight into HIF2α regulation (beyond requisite heterodimerization with HIFβ/aryl hydrocarbon receptor nuclear translocator)30  has also been gained. Hif2α’s translation first has been shown to be suppressed via iron response element binding protein 1 (Irp1) (in a knockout [KO] mouse model), thus connecting iron levels to Hif2α/HIFβ-regulated Epo gene expression.31  HIF2α’s activity is also modulated by lysine acetylation and deacetylation via CREB-binding protein (CBP) and sirtuin, respectively.32  For acetylation, acetyl-CoA levels during stress erythropoiesis can become physiologically limiting. Specifically, Hif2α’s acetylation, CBP association, and enhanced activity have been shown in an Acss2-KO model to depend upon acetyl-CoA synthetase-2.33  Moreover, acetate supplementation in vivo elevates Epo levels as well as hematocrits in hemolytic, partial nephrectomy chronic kidney disease, and Kit mutant models. The turnover of HIFs is promoted via hydroxylation by prolyl 4-hydroxylases (PHDs), and ubiquitination by VHL.30  Notably, several PHD inhibitors have been developed as HIF stabilizers to enhance EPO and erythrocyte production (eg, roxadustat [FibroGen/Astellas], AKB-6548 [Akebia Therapeutics], and GSK1278863 [GlaxoSmithKline]).34 

The expression of EPO’s receptor is stringently regulated and is at a low level (∼1100 EPORs per primary human EPC and ∼300 per late-stage erythroblast) as determined via 125I-EPO binding studies.35  At the EpoR locus, Gata1,36  Sp1,37  and Scl/Tal138  stimulate transcription, but additional regulators are not well defined. For EPOR trafficking, certain new insights have been gained. Over-expression studies in murine myeloid 32D cells suggest ligand-independent EpoR turnover, with replenishment from a predicted large intracellular pool.39  For the endogenous EPOR, however, studies in human UT7epo and/or primary EPCs demonstrate substantial up-modulation of cell surface EPORs when EPO is limited, marked down-modulation upon EPO exposure, and only modest intracellular EPOR pools.40  During EPOR endocytosis, coordinated roles for p85-α (PI3 kinase regulatory subunit), ubiquitinated casitas B-lineage lymphoma and Epsin1 have been described.41,42  In UT7epo cells, β-transducin repeat containing E3 ubiquitin ligase (β-TRCP) subsequently promotes EPOR degradation.43  Dynamic down-modulation of low-level EPOR cell surface expression emphasizes a need for cautious interpretation of apparent EPOR levels, and the use of high-specificity reagents.17,40 

Several new EPOR interacting factors have been described. Proteomic analyses of biotin-EPO/EPOR complexes have identified transferrin receptor 2 (TFR2) as an EPOR partner.44  In UT7epo cells, TFR2 facilitates EPOR processing and transport to the cell surface.44  In primary human EPCs, TFR2 knockdown decreased hemoglobinized cell formation, and increased numbers of early stage EPCs.44  EPCs from Tfr2−/− mice exhibit decreased EPO sensitivity and erythroid colony-forming unit formation.44  During iron deficiency, Tfr2 also acts to balance erythrocyte production with available iron.45  Beyond its established roles in hepatocyte iron transport,46  Tfr2 also therefore modulates EPO-dependent erythropoiesis. In addition, phospho-proteomic analyses have identified the integral plasma membrane protein regulator of human erythroid cell expansion (RHEX) as a new EPOR-associated factor. In UT7epo cells, RHEX co-IP’s with EPOR/JAK2 complexes, and its tyrosine phosphorylation is strongly induced by EPO. In primary EPCs, RHEX exhibits stage-specific expression,47  and its knockdown attenuates extracellular signal-regulated kinase 1/2 (ERK1/2) activation as well as late-stage human erythroblast development. Interestingly, RHEX is not represented among rat, mouse, or lower vertebrates.

Transferrin receptor 1 (TFR1) can also modulate EPOR signaling. Specifically, Tfr1 ligation by polymeric-IgA1 (p-IgA1) in murine erythroblasts increases EPO/EPOR-dependent mitogen-activated protein kinase and phosphatidylinositol 3-kinase signaling.48  This occurs in the absence of transferrin binding, but depends upon a Tfr1 endocytic motif. In a knock-in model, human p-IgA1 enhances recovery from anemia due to 5-fluorouracil, hypoxia, and hemolytic anemia. p-IgA1 also binds CD89, an Fc-α receptor and suppressor of inflammatory cytokine production.49  By speculation, p-IgA1 might also aid stress erythropoiesis by lessening inflammation, a mechanism implicated for an activin receptor-IIA ligand trap as a new anti-anemia agent.50 

As demonstrated in primary human EPCs, balanced activation of RAS pathways is required for effective EPO-dependent erythroblast formation.51  In mouse models, the deletion of K-Ras (but not H-Ras or N-Ras) generates severe anemia,52  and compromises EPO-dependent fetal liver EPC development. Activated K-RasG12D likewise induces severe anemia due to ineffective fetal erythropoiesis,53  and persistently stimulates Erk1/2, Akt, and signal transducer and activator of transcription 5 (Stat5).54  Of clinical interest, the inhibition of RAS farnesylation by tipifarnib decreases polycythemia vera erythroid burst-forming unit hyperproliferation.55  During erythropoiesis, RAS is also regulated by newly emerging guanosine triphosphate (GTP)/guanosine diphosphate exchange factors. One is the Ras-GTPase activating protein Rasa3, which when mutated in Scat mice leads to anemia and thrombocytopenia.56  A second is neurofibromin (Nf1), for which mutations have been associated with juvenile myelomonocytic leukemia, including anemia due to limited EPC differentiation.57 

Regulation of RAS-modulated targets is also important for EPO-dependent erythropoiesis. C-Raf deletion results in embryonic anemia.58  And in β-thalassemia proerythroblasts, phospho-C-Raf levels correlate with increased ERK activation.59  Mek2 is dispensable for mouse development,60  implicating prime roles for Mek1 in Erk1/2 signaling. In mice expressing a truncated EpoR-HM allele, pharmacologic inhibition of Mek reverses stage-specific EPC differentiation defects,61  whereas in mice with somatic inactivation of Nf1, Mek1/2 inhibition decreases splenomegaly and enhances erythropoiesis.57  For ERKs, Erk1−/− mice exhibit heightened splenic erythropoiesis and hematocrits.62 

RAS-like GTPases can also regulate EPO-dependent EPC formation. As a new EPO/EPOR target gene and Roco family GTPase, malignant fibrous histiocytoma-amplified sequences with leucine-rich tandem repeats 1 (MASL1) supports C-RAF/MEK/ERK activation and primary human erythroblast development.63  As Ras homolog family GTPases, RACs also regulate EPC development and erythroblast enucleation.64  In 32D-EpoR and UT7epo cells, EPO rapidly activates RAC1, implicating possible EPO/EPOR regulation of RACs.65  In oncology contexts, as new inhibitors of RAS and RAS-like factors are developed,66  their potential negative effects on erythropoiesis should therefore be evaluated.

EPO’s best-known effects are cytoprotective.67  Koulnis et al have described EPO’s slow yet persistent down-modulation of proapoptotic Bcl2-like 11 (Bim) in murine splenic EPCs.68  Prior studies in HCD57 cells and primary murine EPCs also demonstrated EPO-induced Bim phosphorylation and proteasomal degradation.69  The inhibition of Bim therefore represents one EPO-induced EPC survival mechanism. Post-EPO dosing, Bcl-xL levels in splenic EPCs transiently increase,68  and in 32D-EPOR cells Bcl-xL is an EPO/EPOR/STAT5 target gene.70  These latter EPO effects, however, are not observed in erythroid colony-forming unit-like murine BM EPCs,71  and EPO can efficiently cytoprotect Bclx-KO EPCs.72  Important questions therefore arise concerning possible additional mediators of EPO/EPOR cytoprotection.

Via gene profiling of murine BM EPCs, an intracellular Spi2A serpin has been identified as a new EPO/EPOR/JAK2/Stat5 target, and cytoprotective factor.71  Spi2A inhibits B- and L-cathepsins, which when leached from damaged lysosomes can trigger apoptosis.73  Spi2A-KO mice exhibit compromised EPO-induced EPC formation, and worsened anemia due to hemolysis or irradiation. Spi2A further cytoprotects erythroblasts against ROS, an effect that is phenocopied by a cathepsin-B inhibitor. Pharmacologically, selective cathepsin inhibitors therefore might act to limit cell loss due to oxidant damage in thalassemia and/or sickle cell EPCs.74,75  During stress erythropoiesis, Fas ligand (FasL)/Fas levels can also substantially modulate murine splenic EPC survival.76  In human EPCs, tumor necrosis factor (TNF)-related apoptosis inducing ligand or TRAIL may be a more potent pro-apoptotic TNF, and FASL may support caspase-dependent late differentiation events.77  As further illustrated by recent gene profiling studies of human and murine EPCs,78  complexities can exist in erythroid regulator utilization among species (and erythroid tissues) that require reconciliation.

In the context of erythroid neoplasia, JAK2 hyperactivation due to a V617F mutation (within an inhibitory pseudokinase domain) is a frequently contributing factor.79  Polycythemia vera V617F EPCs can develop in the absence of EPO, although at lower efficiencies.80  In a Ba/F3 cell model, JAK2V617F’s transforming potential is also promoted by EPOR (or thrombopoietin receptor [MPL]) expression.81  For JAK2 R867Q or S755R/R938Q mutations (as associated primarily with thrombocytosis), however, transformation is supported by MPL and not the EPOR. These findings implicate selective EPOR (and MPL) interactions with mutated JAK2 alleles in a context of myeloproliferative disease. JAK2’s degradation interestingly has been demonstrated to involve VHL-mediated ubiquitination. This is illustrated in Chuvash polycythemia in which the VHL mutation R200W alters properties of VHL-SOCS1 E3 ligase complexes, and limits activated phospho-JAK2 turnover.82 

Although EPO/EPOR effects at large require JAK2, supporting roles for Src family kinases are also emerging. Murine EPCs deficient in Lyn exhibit diminished EPO-dependent erythroblast formation.83  And Src, but not Jak2, may mediate posttranslational modification of Cbl, a ubiquitin ligase which promotes EPOR down-modulation (as studied in F-36P cells).84  Mouse KO models additionally have revealed nonredundant Stat5-independent roles for phospholipase-cγ1 (Plc-γ1) in promoting EPOR/Jak2 signals for EPC development.85  In oncology contexts, promising inhibitors of SRC are being developed that additionally can affect Plc-γ1 and/or RAS circuits.86,87  For these agents, potential compromising effects on erythropoiesis should also be considered.

Via EPOR/Jak2/Stat5 signaling, EPO can also induce cytokine expression by EPCs,88  with the C1q/TNF cytokine family member erythroferrone (ERFE/Ctrp-15) as a new example.89  Specifically, ERFE has been discovered to act on hepatocytes to suppress hepcidin production, thereby lessening hepcidin’s inhibitory effects on iron efflux from enterocytes, hepatocytes, and macrophages.90  Following phlebotomy, ERFE expression is heightened, with ERFE-KO mice exhibiting delayed recovery from blood-loss–induced anemia. By comparison, the disruption of ERFE in β-thalassemia mice diminishes iron overload.89  ERFE modulation therefore has therapeutic potential for balancing systemic iron levels. Unexpectedly, EPO has also been shown to exert effects on pluripotent hematopoietic progenitor cells. Specifically, EPO at elevated levels can alter the transcriptomes of multi- and bi-potent progenitors.91  This generates lineage bias, and increases erythroid output while decreasing myelopoiesis.91  EPO therefore may guide EPC differentiation, as previously implicated in studies of EPO/EPOR-stimulated Akt phosphorylation of Gata1.92  Such EPO actions might contribute to rhEPO’s enhancement of erythroid recovery following allogeneic transplant.93 

Within EPO and EPOR circuits, important new components are being revealed. Several regulate endogenous EPO expression (Figure 1A). Apo-IRP1 inhibits Hif2α transcript translation,31  whereas iron reverses this effect, heightening HIF2α and EPO levels. Pharmacologically, PHD inhibitors that stabilize HIF2α can likewise increase EPO expression,34  as does acetate supplementation via enhancement of Hif2α acetylation during stress erythropoiesis.33 

Figure 1

Emerging EPO and EPOR regulators, and action circuits. (A) Regulators of EPO expression in renal peritubular interstitial fibroblasts: during embryogenesis, neural tissue derived EPOpos protein-zeropos fibroblasts occupy interstitial peritubular sites within the neonatal kidney. Renal damage and fibrosis can convert these cells to EPOlow myofibroblasts.24  Within EPOpos interstitial fibroblasts, the EPO production is modulated in part by HIF2α, which itself is regulated at multiple levels. Iron reverses apo-IRP inhibition of HIF2α translation.31  HIF2α turnover is promoted by VHL and PHDs,30  and pharmacologic inhibitors of PHDs stabilize HIF2α.34  During stress erythropoiesis, acetate supplementation can further enhance HIF2α complex acetylation, activity, and EPO production via an Acss2-CBP circuit.33  (B) Modulation of EPOR signaling by interacting plasma membrane proteins: TFR2 associates with the EPOR and can modulate its trafficking.44  Upon p-IgA1 ligation, Tfr1 can also enhance EPOR signaling.48  RHEX also associates with the hEPOR, and promotes EPO-dependent human erythroblast formation.47  (C) Recently defined EPO/EPOR signal transduction circuits: newly discovered EPO/EPOR response genes include ERFE, Spi2A, and MASL1. As a secreted TNF-related cytokine, ERFE completes a circuit between EPO action, and regulation of systemic iron levels.89  By inhibiting leached lysosomal cathepsins, Spi2A cytoprotects erythroblasts against consequences of oxidative damage.71  MASL1 acts within a central RAS/MEK/ERK circuit,63  together with RHEX, to reinforce ERK1/2 activation.47  Further dynamic balancing of essential RAS/MEK/ERK signals (and of EPC formation) occurs via RAS down-modulation by Rasa356  and Nf1.57  Pro-erythropoietic effects also are being established for Akt, Plc-γ1, Lyn, and Src kinases. Akt can affect erythroid development via serine phosphorylation of Gata1,92  whereas Lyn and Src can act to enhance EPO/EPOR activated growth/development signals,83,84  and to modulate Cbl’s E3 ligase effects on EPOR turnover.74,75  For each of these EPO/EPOR signal transducers, their engagement and actions appear to become especially important during anemia and/or stress erythropoiesis.

Figure 1

Emerging EPO and EPOR regulators, and action circuits. (A) Regulators of EPO expression in renal peritubular interstitial fibroblasts: during embryogenesis, neural tissue derived EPOpos protein-zeropos fibroblasts occupy interstitial peritubular sites within the neonatal kidney. Renal damage and fibrosis can convert these cells to EPOlow myofibroblasts.24  Within EPOpos interstitial fibroblasts, the EPO production is modulated in part by HIF2α, which itself is regulated at multiple levels. Iron reverses apo-IRP inhibition of HIF2α translation.31  HIF2α turnover is promoted by VHL and PHDs,30  and pharmacologic inhibitors of PHDs stabilize HIF2α.34  During stress erythropoiesis, acetate supplementation can further enhance HIF2α complex acetylation, activity, and EPO production via an Acss2-CBP circuit.33  (B) Modulation of EPOR signaling by interacting plasma membrane proteins: TFR2 associates with the EPOR and can modulate its trafficking.44  Upon p-IgA1 ligation, Tfr1 can also enhance EPOR signaling.48  RHEX also associates with the hEPOR, and promotes EPO-dependent human erythroblast formation.47  (C) Recently defined EPO/EPOR signal transduction circuits: newly discovered EPO/EPOR response genes include ERFE, Spi2A, and MASL1. As a secreted TNF-related cytokine, ERFE completes a circuit between EPO action, and regulation of systemic iron levels.89  By inhibiting leached lysosomal cathepsins, Spi2A cytoprotects erythroblasts against consequences of oxidative damage.71  MASL1 acts within a central RAS/MEK/ERK circuit,63  together with RHEX, to reinforce ERK1/2 activation.47  Further dynamic balancing of essential RAS/MEK/ERK signals (and of EPC formation) occurs via RAS down-modulation by Rasa356  and Nf1.57  Pro-erythropoietic effects also are being established for Akt, Plc-γ1, Lyn, and Src kinases. Akt can affect erythroid development via serine phosphorylation of Gata1,92  whereas Lyn and Src can act to enhance EPO/EPOR activated growth/development signals,83,84  and to modulate Cbl’s E3 ligase effects on EPOR turnover.74,75  For each of these EPO/EPOR signal transducers, their engagement and actions appear to become especially important during anemia and/or stress erythropoiesis.

Close modal

Within EPCs, EPOR activity can be unexpectedly augmented by interactions with several plasma membrane proteins (Figure 1B). TRF2 acts via association with EPOR complexes, whereas Tfr1 is engaged upon p-IgA1 ligation, with each bolstering EPC formation.44,48  This ties two iron importers to EPOR’s effects. In addition, the novel hEPC protein RHEX associates with the hEPOR, enhances ERK1/2 activation, and supports erythroblast development.47 

Important downstream EPOR signal transducers are also being discovered (Figure 1C). Within a central RAS/MEK module, these include MASL1,63  Rasa3,56  and neurofibrin,57  that act to balance ERK1/2 signaling and EPC production. For EPC cytoprotection, an EPO-induced Spi2A serpin and small molecule inhibitors of leached lysosomal cathepsins have emerged that lessen ROS-associated damage. Pro-erythropoietic actions of Akt, Plc-γ1, and Src family kinases are also being more clearly defined. Finally, EPO is proving to exert guiding effects on early hematopoietic progenitors,91  pointing to new EPO target populations (and indicating an ability of EPO to affect cells harboring few EPORs). High merit therefore persists for continued investigations of novel EPO/EPOR action mechanisms.

This study was supported by grants from the National Institutes of Health National Heart, Lung, and Blood Institute grant R01 HL044491 and National Insitute of Diabetes and Digestive and Kidney Diseases grant R01 DK089439 (D.M.W.) and National Heart, Lung, and Blood Institute grant F32 HL120596 (D.K.).

Contribution: D.K. and D.M.W. contributed in substantial ways to review concepts and construction.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Don M. Wojchowski, Center of Excellence in Stem and Progenitor Cell Biology and Regenerative Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME 04074; e-mail: wojchd@mmc.org.

1
Reissmann
 
KR
Studies on the mechanism of erythropoietic stimulation in parabiotic rats during hypoxia.
Blood
1950
, vol. 
5
 
4
(pg. 
372
-
380
)
2
Miyake
 
T
Kung
 
CK
Goldwasser
 
E
Purification of human erythropoietin.
J Biol Chem
1977
, vol. 
252
 
15
(pg. 
5558
-
5564
)
3
Jacobs
 
K
Shoemaker
 
C
Rudersdorf
 
R
, et al. 
Isolation and characterization of genomic and cDNA clones of human erythropoietin.
Nature
1985
, vol. 
313
 
6005
(pg. 
806
-
810
)
4
Rizzo
 
JD
Brouwers
 
M
Hurley
 
P
, et al. 
American Society of Hematology and the American Society of Clinical Oncology Practice Guideline Update Committee
American Society of Hematology/American Society of Clinical Oncology clinical practice guideline update on the use of epoetin and darbepoetin in adult patients with cancer.
Blood
2010
, vol. 
116
 
20
(pg. 
4045
-
4059
)
5
D’Andrea
 
AD
Lodish
 
HF
Wong
 
GG
Expression cloning of the murine erythropoietin receptor.
Cell
1989
, vol. 
57
 
2
(pg. 
277
-
285
)
6
Constantinescu
 
SN
Wu
 
H
Liu
 
X
Beyer
 
W
Fallon
 
A
Lodish
 
HF
The anemic Friend virus gp55 envelope protein induces erythroid differentiation in fetal liver colony-forming units-erythroid.
Blood
1998
, vol. 
91
 
4
(pg. 
1163
-
1172
)
7
Witthuhn
 
BA
Quelle
 
FW
Silvennoinen
 
O
, et al. 
JAK2 associates with the erythropoietin receptor and is tyrosine phosphorylated and activated following stimulation with erythropoietin.
Cell
1993
, vol. 
74
 
2
(pg. 
227
-
236
)
8
Livnah
 
O
Johnson
 
DL
Stura
 
EA
, et al. 
An antagonist peptide-EPO receptor complex suggests that receptor dimerization is not sufficient for activation.
Nat Struct Biol
1998
, vol. 
5
 
11
(pg. 
993
-
1004
)
9
Sokol
 
L
Luhovy
 
M
Guan
 
Y
Prchal
 
JF
Semenza
 
GL
Prchal
 
JT
Primary familial polycythemia: a frameshift mutation in the erythropoietin receptor gene and increased sensitivity of erythroid progenitors to erythropoietin.
Blood
1995
, vol. 
86
 
1
(pg. 
15
-
22
)
10
Bartnicki
 
P
Kowalczyk
 
M
Rysz
 
J
The influence of the pleiotropic action of erythropoietin and its derivatives on nephroprotection.
Med Sci Monit
2013
, vol. 
19
 (pg. 
599
-
605
)
11
Debeljak
 
N
Solár
 
P
Sytkowski
 
AJ
Erythropoietin and cancer: the unintended consequences of anemia correction.
Front Immunol
2014
, vol. 
5
 pg. 
563
 
12
Mastromarino
 
V
Musumeci
 
MB
Conti
 
E
Tocci
 
G
Volpe
 
M
Erythropoietin in cardiac disease: effective or harmful?
J Cardiovasc Med (Hagerstown)
2013
, vol. 
14
 
12
(pg. 
870
-
878
)
13
Nguyen
 
AQ
Cherry
 
BH
Scott
 
GF
Ryou
 
MG
Mallet
 
RT
Erythropoietin: powerful protection of ischemic and post-ischemic brain.
Exp Biol Med (Maywood)
2014
, vol. 
239
 
11
(pg. 
1461
-
1475
)
14
Sanchis-Gomar
 
F
Garcia-Gimenez
 
JL
Pareja-Galeano
 
H
Romagnoli
 
M
Perez-Quilis
 
C
Lippi
 
G
Erythropoietin and the heart: physiological effects and the therapeutic perspective.
Int J Cardiol
2014
, vol. 
171
 
2
(pg. 
116
-
125
)
15
Wang
 
L
Di
 
L
Noguchi
 
CT
Erythropoietin, a novel versatile player regulating energy metabolism beyond the erythroid system.
Int J Biol Sci
2014
, vol. 
10
 
8
(pg. 
921
-
939
)
16
Zhang
 
Y
Wang
 
L
Dey
 
S
, et al. 
Erythropoietin action in stress response, tissue maintenance and metabolism.
Int J Mol Sci
2014
, vol. 
15
 
6
(pg. 
10296
-
10333
)
17
Elliott
 
S
Sinclair
 
A
Collins
 
H
Rice
 
L
Jelkmann
 
W
Progress in detecting cell-surface protein receptors: the erythropoietin receptor example.
Ann Hematol
2014
, vol. 
93
 
2
(pg. 
181
-
192
)
18
Carraway
 
MS
Suliman
 
HB
Jones
 
WS
Chen
 
CW
Babiker
 
A
Piantadosi
 
CA
Erythropoietin activates mitochondrial biogenesis and couples red cell mass to mitochondrial mass in the heart.
Circ Res
2010
, vol. 
106
 
11
(pg. 
1722
-
1730
)
19
Mowat
 
FM
Gonzalez
 
F
Luhmann
 
UF
, et al. 
Endogenous erythropoietin protects neuroretinal function in ischemic retinopathy.
Am J Pathol
2012
, vol. 
180
 
4
(pg. 
1726
-
1739
)
20
Kumar
 
SM
Zhang
 
G
Bastian
 
BC
, et al. 
Erythropoietin receptor contributes to melanoma cell survival in vivo.
Oncogene
2012
, vol. 
31
 
13
(pg. 
1649
-
1660
)
21
Teng
 
R
Gavrilova
 
O
Suzuki
 
N
, et al. 
Disrupted erythropoietin signalling promotes obesity and alters hypothalamus proopiomelanocortin production.
Nat Commun
2011
, vol. 
2
 pg. 
520
 
22
Suzuki
 
N
Hirano
 
I
Pan
 
X
Minegishi
 
N
Yamamoto
 
M
Erythropoietin production in neuroepithelial and neural crest cells during primitive erythropoiesis.
Nat Commun
2013
, vol. 
4
 pg. 
2902
 
23
Yamaguchi
 
Y
Hayashi
 
A
Campagnoni
 
CW
Kimura
 
A
Inuzuka
 
T
Baba
 
H
L-MPZ, a novel isoform of myelin P0, is produced by stop codon readthrough.
J Biol Chem
2012
, vol. 
287
 
21
(pg. 
17765
-
17776
)
24
Asada
 
N
Takase
 
M
Nakamura
 
J
, et al. 
Dysfunction of fibroblasts of extrarenal origin underlies renal fibrosis and renal anemia in mice.
J Clin Invest
2011
, vol. 
121
 
10
(pg. 
3981
-
3990
)
25
Pan
 
X
Suzuki
 
N
Hirano
 
I
Yamazaki
 
S
Minegishi
 
N
Yamamoto
 
M
Isolation and characterization of renal erythropoietin-producing cells from genetically produced anemia mice.
PLoS ONE
2011
, vol. 
6
 
10
pg. 
e25839
 
26
Suzuki
 
N
Obara
 
N
Pan
 
X
, et al. 
Specific contribution of the erythropoietin gene 3′ enhancer to hepatic erythropoiesis after late embryonic stages.
Mol Cell Biol
2011
, vol. 
31
 
18
(pg. 
3896
-
3905
)
27
Rankin
 
EB
Wu
 
C
Khatri
 
R
, et al. 
The HIF signaling pathway in osteoblasts directly modulates erythropoiesis through the production of EPO.
Cell
2012
, vol. 
149
 
1
(pg. 
63
-
74
)
28
Bunn
 
HF
Erythropoietin.
Cold Spring Harb Perspect Med
2013
, vol. 
3
 
3
pg. 
a011619
 
29
Scortegagna
 
M
Ding
 
K
Zhang
 
Q
, et al. 
HIF-2alpha regulates murine hematopoietic development in an erythropoietin-dependent manner.
Blood
2005
, vol. 
105
 
8
(pg. 
3133
-
3140
)
30
Haase
 
VH
Regulation of erythropoiesis by hypoxia-inducible factors.
Blood Rev
2013
, vol. 
27
 
1
(pg. 
41
-
53
)
31
Anderson
 
SA
Nizzi
 
CP
Chang
 
YI
, et al. 
The IRP1-HIF-2α axis coordinates iron and oxygen sensing with erythropoiesis and iron absorption.
Cell Metab
2013
, vol. 
17
 
2
(pg. 
282
-
290
)
32
Chen
 
R
Xu
 
M
Hogg
 
RT
, et al. 
The acetylase/deacetylase couple CREB-binding protein/Sirtuin 1 controls hypoxia-inducible factor 2 signaling.
J Biol Chem
2012
, vol. 
287
 
36
(pg. 
30800
-
30811
)
33
Xu
 
M
Nagati
 
JS
Xie
 
J
, et al. 
An acetate switch regulates stress erythropoiesis.
Nat Med
2014
, vol. 
20
 
9
(pg. 
1018
-
1026
)
34
Krishnan
 
M
Nissenson
 
AR
New alternatives in anemia treatment: biosimilars and HIF stabilizers.
Nephrol News Issues
2014
, vol. 
28
 
6
(pg. 
24
-
28
)
35
Broudy
 
VC
Lin
 
N
Brice
 
M
Nakamoto
 
B
Papayannopoulou
 
T
Erythropoietin receptor characteristics on primary human erythroid cells.
Blood
1991
, vol. 
77
 
12
(pg. 
2583
-
2590
)
36
Zon
 
LI
Youssoufian
 
H
Mather
 
C
Lodish
 
HF
Orkin
 
SH
Activation of the erythropoietin receptor promoter by transcription factor GATA-1.
Proc Natl Acad Sci USA
1991
, vol. 
88
 
23
(pg. 
10638
-
10641
)
37
Feng
 
D
Kan
 
YW
The binding of the ubiquitous transcription factor Sp1 at the locus control region represses the expression of beta-like globin genes.
Proc Natl Acad Sci USA
2005
, vol. 
102
 
28
(pg. 
9896
-
9900
)
38
Lacombe
 
J
Krosl
 
G
Tremblay
 
M
, et al. 
Genetic interaction between Kit and Scl.
Blood
2013
, vol. 
122
 
7
(pg. 
1150
-
1161
)
39
Becker
 
V
Schilling
 
M
Bachmann
 
J
, et al. 
Covering a broad dynamic range: information processing at the erythropoietin receptor.
Science
2010
, vol. 
328
 
5984
(pg. 
1404
-
1408
)
40
Singh
 
S
Verma
 
R
Pradeep
 
A
, et al. 
Dynamic ligand modulation of EPO receptor pools, and dysregulation by polycythemia-associated EPOR alleles.
PLoS ONE
2012
, vol. 
7
 
1
pg. 
e29064
 
41
Sulahian
 
R
Cleaver
 
O
Huang
 
LJ
Ligand-induced EpoR internalization is mediated by JAK2 and p85 and is impaired by mutations responsible for primary familial and congenital polycythemia.
Blood
2009
, vol. 
113
 
21
(pg. 
5287
-
5297
)
42
Bulut
 
GB
Sulahian
 
R
Yao
 
H
Huang
 
LJ
Cbl ubiquitination of p85 is essential for Epo-induced EpoR endocytosis.
Blood
2013
, vol. 
122
 
24
(pg. 
3964
-
3972
)
43
Meyer
 
L
Deau
 
B
Forejtníková
 
H
, et al. 
beta-Trcp mediates ubiquitination and degradation of the erythropoietin receptor and controls cell proliferation.
Blood
2007
, vol. 
109
 
12
(pg. 
5215
-
5222
)
44
Forejtnikovà
 
H
Vieillevoye
 
M
Zermati
 
Y
, et al. 
Transferrin receptor 2 is a component of the erythropoietin receptor complex and is required for efficient erythropoiesis.
Blood
2010
, vol. 
116
 
24
(pg. 
5357
-
5367
)
45
Nai
 
A
Lidonnici
 
MR
Rausa
 
M
, et al. 
The second transferrin receptor regulates red blood cell production in mice.
Blood
2015
, vol. 
125
 
7
(pg. 
1170
-
1179
)
46
Chen
 
J
Chloupková
 
M
Abnormal iron uptake and liver cancer.
Cancer Biol Ther
2009
, vol. 
8
 
18
(pg. 
1699
-
1708
)
47
Verma
 
R
Su
 
S
McCrann
 
DJ
, et al. 
RHEX, a novel regulator of human erythroid progenitor cell expansion and erythroblast development.
J Exp Med
2014
, vol. 
211
 
9
(pg. 
1715
-
1722
)
48
Coulon
 
S
Dussiot
 
M
Grapton
 
D
, et al. 
Polymeric IgA1 controls erythroblast proliferation and accelerates erythropoiesis recovery in anemia.
Nat Med
2011
, vol. 
17
 
11
(pg. 
1456
-
1465
)
49
Wu
 
J
Ji
 
C
Xie
 
F
, et al. 
FcalphaRI (CD89) alleles determine the proinflammatory potential of serum IgA.
J Immunol
2007
, vol. 
178
 
6
(pg. 
3973
-
3982
)
50
Dussiot
 
M
Maciel
 
TT
Fricot
 
A
, et al. 
An activin receptor IIA ligand trap corrects ineffective erythropoiesis in β-thalassemia.
Nat Med
2014
, vol. 
20
 
4
(pg. 
398
-
407
)
51
Arcasoy
 
MO
Jiang
 
X
Co-operative signalling mechanisms required for erythroid precursor expansion in response to erythropoietin and stem cell factor.
Br J Haematol
2005
, vol. 
130
 
1
(pg. 
121
-
129
)
52
Khalaf
 
WF
White
 
H
Wenning
 
MJ
Orazi
 
A
Kapur
 
R
Ingram
 
DA
K-Ras is essential for normal fetal liver erythropoiesis.
Blood
2005
, vol. 
105
 
9
(pg. 
3538
-
3541
)
53
Braun
 
BS
Archard
 
JA
Van Ziffle
 
JA
Tuveson
 
DA
Jacks
 
TE
Shannon
 
K
Somatic activation of a conditional KrasG12D allele causes ineffective erythropoiesis in vivo.
Blood
2006
, vol. 
108
 
6
(pg. 
2041
-
2044
)
54
Zhang
 
J
Liu
 
Y
Beard
 
C
, et al. 
Expression of oncogenic K-ras from its endogenous promoter leads to a partial block of erythroid differentiation and hyperactivation of cytokine-dependent signaling pathways.
Blood
2007
, vol. 
109
 
12
(pg. 
5238
-
5241
)
55
Larghero
 
J
Gervais
 
N
Cassinat
 
B
, et al. 
Farnesyltransferase inhibitor tipifarnib (R115777) preferentially inhibits in vitro autonomous erythropoiesis of polycythemia vera patient cells.
Blood
2005
, vol. 
105
 
9
(pg. 
3743
-
3745
)
56
Blanc
 
L
Ciciotte
 
SL
Gwynn
 
B
, et al. 
Critical function for the Ras-GTPase activating protein RASA3 in vertebrate erythropoiesis and megakaryopoiesis.
Proc Natl Acad Sci USA
2012
, vol. 
109
 
30
(pg. 
12099
-
12104
)
57
Chang
 
T
Krisman
 
K
Theobald
 
EH
, et al. 
Sustained MEK inhibition abrogates myeloproliferative disease in Nf1 mutant mice.
J Clin Invest
2013
, vol. 
123
 
1
(pg. 
335
-
339
)
58
Rubiolo
 
C
Piazzolla
 
D
Meissl
 
K
, et al. 
A balance between Raf-1 and Fas expression sets the pace of erythroid differentiation.
Blood
2006
, vol. 
108
 
1
(pg. 
152
-
159
)
59
Wannatung
 
T
Lithanatudom
 
P
Leecharoenkiat
 
A
Svasti
 
S
Fucharoen
 
S
Smith
 
DR
Increased erythropoiesis of beta-thalassaemia/Hb E proerythroblasts is mediated by high basal levels of ERK1/2 activation.
Br J Haematol
2009
, vol. 
146
 
5
(pg. 
557
-
568
)
60
Bélanger
 
LF
Roy
 
S
Tremblay
 
M
, et al. 
Mek2 is dispensable for mouse growth and development.
Mol Cell Biol
2003
, vol. 
23
 
14
(pg. 
4778
-
4787
)
61
Menon
 
MP
Fang
 
J
Wojchowski
 
DM
Core erythropoietin receptor signals for late erythroblast development.
Blood
2006
, vol. 
107
 
7
(pg. 
2662
-
2672
)
62
Guihard
 
S
Clay
 
D
Cocault
 
L
, et al. 
The MAPK ERK1 is a negative regulator of the adult steady-state splenic erythropoiesis.
Blood
2010
, vol. 
115
 
18
(pg. 
3686
-
3694
)
63
Kumkhaek
 
C
Aerbajinai
 
W
Liu
 
W
, et al. 
MASL1 induces erythroid differentiation in human erythropoietin-dependent CD34+ cells through the Raf/MEK/ERK pathway.
Blood
2013
, vol. 
121
 
16
(pg. 
3216
-
3227
)
64
Kalfa
 
TA
Zheng
 
Y
Rho GTPases in erythroid maturation.
Curr Opin Hematol
2014
, vol. 
21
 
3
(pg. 
165
-
171
)
65
Arai
 
A
Kanda
 
E
Miura
 
O
Rac is activated by erythropoietin or interleukin-3 and is involved in activation of the Erk signaling pathway.
Oncogene
2002
, vol. 
21
 
17
(pg. 
2641
-
2651
)
66
Cox
 
AD
Fesik
 
SW
Kimmelman
 
AC
Luo
 
J
Der
 
CJ
Drugging the undruggable RAS: mission possible?
Nat Rev Drug Discov
2014
, vol. 
13
 
11
(pg. 
828
-
851
)
67
Koury
 
MJ
Bondurant
 
MC
Erythropoietin retards DNA breakdown and prevents programmed death in erythroid progenitor cells.
Science
1990
, vol. 
248
 
4953
(pg. 
378
-
381
)
68
Koulnis
 
M
Porpiglia
 
E
Porpiglia
 
PA
, et al. 
Contrasting dynamic responses in vivo of the Bcl-xL and Bim erythropoietic survival pathways.
Blood
2012
, vol. 
119
 
5
(pg. 
1228
-
1239
)
69
Abutin
 
RM
Chen
 
J
Lung
 
TK
Lloyd
 
JA
Sawyer
 
ST
Harada
 
H
Erythropoietin-induced phosphorylation/degradation of BIM contributes to survival of erythroid cells.
Exp Hematol
2009
, vol. 
37
 
2
(pg. 
151
-
158
)
70
Socolovsky
 
M
Fallon
 
AE
Wang
 
S
Brugnara
 
C
Lodish
 
HF
Fetal anemia and apoptosis of red cell progenitors in Stat5a-/-5b-/- mice: a direct role for Stat5 in Bcl-X(L) induction.
Cell
1999
, vol. 
98
 
2
(pg. 
181
-
191
)
71
Dev
 
A
Byrne
 
SM
Verma
 
R
Ashton-Rickardt
 
PG
Wojchowski
 
DM
Erythropoietin-directed erythropoiesis depends on serpin inhibition of erythroblast lysosomal cathepsins.
J Exp Med
2013
, vol. 
210
 
2
(pg. 
225
-
232
)
72
Rhodes
 
MM
Kopsombut
 
P
Bondurant
 
MC
Price
 
JO
Koury
 
MJ
Bcl-x(L) prevents apoptosis of late-stage erythroblasts but does not mediate the antiapoptotic effect of erythropoietin.
Blood
2005
, vol. 
106
 
5
(pg. 
1857
-
1863
)
73
Repnik
 
U
Hafner Cesen
 
M
Turk
 
B
Lysosomal membrane permeabilization in cell death: concepts and challenges.
Mitochondrion
2014
, vol. 
19
 
pt A
(pg. 
49
-
57
)
74
De Franceschi
 
L
Bertoldi
 
M
Matte
 
A
, et al. 
Oxidative stress and β-thalassemic erythroid cells behind the molecular defect.
Oxid Med Cell Longev
2013
, vol. 
2013
 pg. 
985210
 
75
Sangokoya
 
C
Telen
 
MJ
hi
 
JT
microRNA miR-144 modulates oxidative stress tolerance and associates with anemia severity in sickle cell disease.
Blood
2010
, vol. 
116
 
20
(pg. 
4338
-
4348
)
76
Koulnis
 
M
Liu
 
Y
Hallstrom
 
K
Socolovsky
 
M
Negative autoregulation by Fas stabilizes adult erythropoiesis and accelerates its stress response.
PLoS ONE
2011
, vol. 
6
 
7
pg. 
e21192
 
77
Carlile
 
GW
Smith
 
DH
Wiedmann
 
M
A non-apoptotic role for Fas/FasL in erythropoiesis.
FEBS Lett
2009
, vol. 
583
 
4
(pg. 
848
-
854
)
78
An
 
X
Schulz
 
VP
Li
 
J
, et al. 
Global transcriptome analyses of human and murine terminal erythroid differentiation.
Blood
2014
, vol. 
123
 
22
(pg. 
3466
-
3477
)
79
Kralovics
 
R
Passamonti
 
F
Buser
 
AS
, et al. 
A gain-of-function mutation of JAK2 in myeloproliferative disorders.
N Engl J Med
2005
, vol. 
352
 
17
(pg. 
1779
-
1790
)
80
Jedidi
 
A
Marty
 
C
Oligo
 
C
, et al. 
Selective reduction of JAK2V617F-dependent cell growth by siRNA/shRNA and its reversal by cytokines.
Blood
2009
, vol. 
114
 
9
(pg. 
1842
-
1851
)
81
Marty
 
C
Saint-Martin
 
C
Pecquet
 
C
, et al. 
Germ-line JAK2 mutations in the kinase domain are responsible for hereditary thrombocytosis and are resistant to JAK2 and HSP90 inhibitors.
Blood
2014
, vol. 
123
 
9
(pg. 
1372
-
1383
)
82
Russell
 
RC
Sufan
 
RI
Zhou
 
B
, et al. 
Loss of JAK2 regulation via a heterodimeric VHL-SOCS1 E3 ubiquitin ligase underlies Chuvash polycythemia.
Nat Med
2011
, vol. 
17
 
7
(pg. 
845
-
853
)
83
Karur
 
VG
Lowell
 
CA
Besmer
 
P
Agosti
 
V
Wojchowski
 
DM
Lyn kinase promotes erythroblast expansion and late-stage development.
Blood
2006
, vol. 
108
 
5
(pg. 
1524
-
1532
)
84
Shintani
 
T
Ohara-Waki
 
F
Kitanaka
 
A
Tanaka
 
T
Kubota
 
Y
Cbl negatively regulates erythropoietin-induced growth and survival signaling through the proteasomal degradation of Src kinase.
Blood Cells Mol Dis
2014
, vol. 
53
 
4
(pg. 
211
-
218
)
85
Schnöder
 
TM
Arreba-Tutusaus
 
P
Griehl
 
I
, et al. 
Epo-induced erythroid maturation is dependent on Plcγ1 signaling [published online ahead of print November 14, 1014].
Cell Death Differ
86
Girotti
 
MR
Lopes
 
F
Preece
 
N
, et al. 
Paradox-breaking RAF inhibitors that also target SRC are effective in drug-resistant BRAF mutant melanoma.
Cancer Cell
2015
, vol. 
27
 
1
(pg. 
85
-
96
)
87
de Lavallade
 
H
Khoder
 
A
Hart
 
M
, et al. 
Tyrosine kinase inhibitors impair B-cell immune responses in CML through off-target inhibition of kinases important for cell signaling.
Blood
2013
, vol. 
122
 
2
(pg. 
227
-
238
)
88
Menon
 
MP
Karur
 
V
Bogacheva
 
O
Bogachev
 
O
Cuetara
 
B
Wojchowski
 
DM
Signals for stress erythropoiesis are integrated via an erythropoietin receptor-phosphotyrosine-343-Stat5 axis.
J Clin Invest
2006
, vol. 
116
 
3
(pg. 
683
-
694
)
89
Kautz
 
L
Jung
 
G
Valore
 
EV
Rivella
 
S
Nemeth
 
E
Ganz
 
T
Identification of erythroferrone as an erythroid regulator of iron metabolism.
Nat Genet
2014
, vol. 
46
 
7
(pg. 
678
-
684
)
90
Rochette
 
L
Gudjoncik
 
A
Guenancia
 
C
Zeller
 
M
Cottin
 
Y
Vergely
 
C
The iron-regulatory hormone hepcidin: a possible therapeutic target?
Pharmacol Ther
2015
, vol. 
146
 (pg. 
35
-
52
)
91
Grover
 
A
Mancini
 
E
Moore
 
S
, et al. 
Erythropoietin guides multipotent hematopoietic progenitor cells toward an erythroid fate.
J Exp Med
2014
, vol. 
211
 
2
(pg. 
181
-
188
)
92
Zhao
 
W
Kitidis
 
C
Fleming
 
MD
Lodish
 
HF
Ghaffari
 
S
Erythropoietin stimulates phosphorylation and activation of GATA-1 via the PI3-kinase/AKT signaling pathway.
Blood
2006
, vol. 
107
 
3
(pg. 
907
-
915
)
93
Hellström-Lindberg
 
E
van de Loosdrecht
 
A
Erythropoiesis stimulating agents and other growth factors in low-risk MDS.
Best Pract Res Clin Haematol
2013
, vol. 
26
 
4
(pg. 
401
-
410
)
Sign in via your Institution