Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Hematology

ISSN: (Print) (Online) Journal homepage: www.tandfonline.com/journals/yhem20

Regulation of erythropoiesis: emerging concepts


and therapeutic implications

Pu Tang & Huaquan Wang

To cite this article: Pu Tang & Huaquan Wang (2023) Regulation of erythropoiesis:
emerging concepts and therapeutic implications, Hematology, 28:1, 2250645, DOI:
10.1080/16078454.2023.2250645

To link to this article: https://doi.org/10.1080/16078454.2023.2250645

© 2023 The Author(s). Published by Informa


UK Limited, trading as Taylor & Francis
Group

Published online: 28 Aug 2023.

Submit your article to this journal

Article views: 4817

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=yhem20
HEMATOLOGY
2023, VOL. 28, NO. 1, 2250645
https://doi.org/10.1080/16078454.2023.2250645

REVIEW ARTICLE

Regulation of erythropoiesis: emerging concepts and therapeutic implications


Pu Tang and Huaquan Wang
Department of Hematology, General Hospital, Tianjin Medical University, Tianjin, People’s Republic of China

ABSTRACT ARTICLE HISTORY


The process of erythropoiesis is complex and involves the transfer of cells from the yolk sac to Received 19 April 2023
the fetal hepar and, ultimately, to the bone marrow during embryonic development. Within the Accepted 17 August 2023
bone marrow, erythroid progenitor cells undergo several stages to generate reticulocytes that
KEYWORDS
enter the bloodstream. Erythropoiesis is regulated by various factors, with erythropoietin (EPO) Erythropoiesis; embryonic
synthesized by the kidney being the promoting factor and hepcidin synthesized by the hepar erythropoiesis; human
inhibiting iron mobilization. Transcription factors, such as GATA and KLF, also play a crucial role erythropoietin (EPO);
in erythropoiesis. Disruption of any of these factors can lead to abnormal erythropoiesis, erythroferrone (ERFE);
resulting in red cell excess, red cell deficiency, or abnormal morphological function. This ferritin; transferrin; anemia;
review provides a general description of erythropoiesis, as well as its regulation, highlighting polycythemia
the significance of understanding the process for the diagnosis and treatment of various
hematological disorders.

Introduction absorbed during development [3]. Blood islands,


which contain primitive erythroid progenitor cells,
Erythropoiesis is a process by which red blood cells
form in the mesoderm layer of the yolk sac. These pro-
(erythrocytes) are produced in the marrow. It involves
genitor cells differentiate into erythroblasts, which
the differentiation of erythroid progenitor cells into
produce embryonic hemoglobin (α2ε2) [4]. α2ε2 is
mature RBCs, which transport oxygen from the lungs
composed of a pair of alpha and a pair of epsilon
to the body’s tissues. erythropoiesis is a complex
globin chains and has a stronger affinity for oxygen
process that is strictly regulated by varieties of
than adult hemoglobin.
factors, including hormones, cytokines, and growth
Around 6–8 weeks of gestation, erythropoiesis shifts
factors.
to the hepar and spleen [5], as these organs become
In recent years, there have been vital advances in
capable of producing erythropoietin (EPO), the
our awareness of the molecular mechanisms that
hormone that stimulates erythropoiesis. Studies show
govern erythropoiesis. These advances have led to
that EMP produced in the mouse yolk sac migrates
the positive result of new therapeutic strategies for
to the hepar and colonizes [6], and hematopoietic
the treatment of erythropoietic disorders. In the
stem cells also migrate to the hepar [7]. The hepar
article, we review the issues of embryonic erythropoi-
becomes the primary site of erythropoiesis from
esis, the cytokines involved in erythropoiesis, signaling
around weeks 10–28 of gestation, while the spleen
cascades, and possible problems in erythropoiesis.
mainly produces RBCs in the second trimester.
By the end of the second trimester, erythropoiesis
The normal physiological course of begins to shift to the marrow, which becomes the
erythropoiesis initial site of erythropoiesis by the time of birth
(Figure 1). At this stage, fetal hemoglobin (HbF) is the
Erythropoiesis in embryos
primary type of hemoglobin produced, which pos-
In embryos, erythropoiesis occurs in a different sesses a stronger affinity for oxygen than adult hemo-
location than in adults. During embryonic develop- globin (HbA) and helps to facilitate oxygen transport
ment, erythropoiesis initially takes place in the yolk across the placenta. After birth, the output of HbF
sac, and later shifts to the hepar and spleen, before gradually decreases and is replaced by the output of
ultimately transitioning to the marrow [1]. HbA.
The Yolk sac is the original site of erythropoiesis Fetal hematopoiesis is regulated by multiple factors.
during the first few weeks of embryonic development For example, ferritin IRP2 regulates iron infusion from
[2], and it’s a short-lived membrane which is gradually the maternal placenta [8], and EpoR can lengthen the

CONTACT Huaquan Wang wanghuaquan@tmu.edu.cn


© 2023 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/), which permits unrest-
ricted use, distribution, and reproduction in any medium, provided the original work is properly cited. The terms on which this article has been published allow the posting of the
Accepted Manuscript in a repository by the author(s) or with their consent.
2 P. TANG AND H. WANG

Figure 1. An overview of erythropoiesis in the embryo. Erythropoiesis in the embryo can be divided into three stages. The first
stage is the third to sixth week of embryogenesis, during which the yolk sac, formed by mesodermal cells and angioplastic cells,
produces platelets, PEs, and macrophages, with the erythrocyte-containing PEs entering the circulation. The second stage is the
third to sixth month of embryonic development, during which EMPs produced by the yolk sac migrate to the liver and proliferate
and differentiate into red cells in the liver. The third stage is after six months of embryonic development, during which the hema-
topoietic center is located in the bone marrow, and hematopoietic stem cells differentiate into red cells and enter the circulation.

early stages of the erythrocyte cycle by increasing red In the bone marrow, erythroid progenitor cells
cell cycle length [9]. Furthermore, growth factors (SCF) differentiate into erythroblasts, which undergo
are critical for erythropoiesis and are critical for main- several rounds of cell division and synthesis of hemo-
taining normal adult erythrocyte numbers [10]. globin, the protein that brings oxygen in RBCs. The
Erythropoiesis first occurs in the yolk sac, then metas- cells gradually reduce their size and nucleus, and
tasizes to the fetal liver, and finally resides in the bone increase their cytoplasmic volume, ultimately becom-
marrow after birth. Switching between these sites is ing reticulocytes.
associated with changes in globin expression and The output of erythroblasts is regulated by a
oxygen-binding affinity of erythrocytes. When erythro- number of cytokines and growth factors, including ery-
cyte proliferation metastasizes from the yolk sac to the thropoietin (EPO), stem cell factor (SCF), and interleu-
fetal liver, a primordial to final erythroblastic transform- kin-3 (IL-3), among others. EPO, which is produced
ation occurs. Fetal to adult globin conversion occurs primarily by the kidneys in response to low oxygen
when erythropoiesis is transferred from the fetal liver levels in the body, excites the differentiation and pro-
to the bone marrow. Fetal globin has a higher oxygen liferation of erythroid progenitor cells.
affinity than adult globin and can better extract The process of erythropoiesis in the bone marrow is
oxygen from maternal blood. These spatial and tem- also affected by the microenvironment, or niche, in
poral switches during erythropoiesis allow the develop- which the cells remain. This niche consists of various
ing embryo/fetus to adapt to the changing oxygen cell types, containing osteoblasts, endothelial cells,
requirements during development [2]. stromal cells, as well as extracellular matrix com-
ponents [12].
Osteoblasts, which are bone-forming cells, have
Erythropoiesis in bone marrow
been shown to regulate erythropoiesis through the
Erythropoiesis in bone marrow is the process by which output of cytokines and growth factors, as well as
progenitor cells become mature red blood cells (RBCs), direct cell-to-cell contacts with erythroid progenitor
about 20 billion erythrocytes emerge in the marrow cells. Endothelial cells also play a role in regulating ery-
every day [11]. This course is regulated and controlled thropoiesis by producing cytokines and growth
by a heterogeneous network of signaling pathways factors, and by providing a source of oxygen and
and transcription factors. other nutrients. Stromal cells, which are mesenchymal
HEMATOLOGY 3

cells that support hematopoiesis, also contribute to impaired erythropoiesis and anemia. Stress erythropoi-
the regulation of erythropoiesis by producing growth esis is highly dependent on signals such as EPO/EPOR/
factors and providing a supportive niche for erythroid JAK2/STAT5, as well as activation signals in erythroid
progenitor cells (Figure 2). islets such as BMP4/SMAD5 and integrin signals [15].
Terminal enucleation is the process by which the
nucleus is extruded from late erythrocytes before
Stages of RBC maturation and differentiation
they mature into reticular cells. This is a unique
feature of mammalian erythrocyte production [13]. Erythropoiesis is a multi-stage process that typically
Some congenital dysplasia of erythropoietic anemia lasts for approximately 14 days. It includes the
(CDAs) and myelodysplastic syndrome (MDS) may differentiation of hematopoietic stem cells into ery-
have a pathological phenomenon of erythrocyte enu- throid progenitor cells and megakaryocytes. Ery-
cleation disorder. In MDS, abnormal erythrocyte throid progenitor cells consist of erythroid bursting
islands and poor erythrocyte segregation may indi- forming units (BFU-E) and erythroid colony forming
cate impaired peripheral enucleation. In CDA type I, units (CFU-E), which are a continuous stage of ery-
caused by CDAN1 or C15orf41 mutations, the throid progenitor cells [16]. These cells then differ-
encoded proteins are thought to have roles in chro- entiate into erythroid precursors of varying
matin remodeling during nuclear extrusion. In CDA morphologies.
type II caused by SEC23B mutations, SEC23B may The earliest recognizable erythroid precursor cells
be involved in mesomeric formation during cell div- are pro-erythroblasts (pro-E), which mark the begin-
ision, and its dysfunction can lead to impaired enu- ning of the second stage of red blood cell (RBC) matu-
cleation [14]. In CDA type III, caused by a KIF23 ration. Pro-E cells develop into basophilic (Bas-E),
mutation, the mutant protein disrupts cytokinesis polychromatic (Poly-E), and orthochromatic erythro-
through effects on the mitotic spindle apparatus. In cytes (Ortho-E) in a sequential manner, ultimately
these cases, the molecular mechanisms of impaired leading to the formation of reticulocytes that lack a
enucleation are not fully understood, but may nucleus [17]. During this process, the size of the cells
involve protein dysfunction involved in the regu- and nuclei gradually decreases while the accumulation
lation of chromatin remodeling, cell division, and of hemoglobin increases [1].
erythrocyte island formation. Late-stage erythropoiesis is characterized by the
loss of the nucleus, loss of surface biomarkers, and
the formation of a strong and extendable cytoskeleton.
Erythroblast island
These changes allow for the final maturation and
Erythroblast island refers to a unique microenviron- differentiation of erythroid precursors to fully mature
ment in the marrow where erythroblasts, the precur- erythrocytes [18].
sors of erythrocytes, interact with macrophages. The
erythroblast island is symbolized by a central macro-
Biomarkers of erythropoiesis
phage surrounded by a circle of developing
erythroblasts. Studies utilizing single-cell genomics have demon-
Macrophages within the erythroblast island play an strated that the output of erythrocytes (RBCs) is an
important role in supporting erythropoiesis, the course ongoing process characterized by varying cellular
by which progenitor cells differentiate into mature states and the expression of different genes [16].
erythrocytes. Macrophages provide various factors The progenitor cells, BFU-E and CFU-E, are highly
such as transferrin, iron, cytokines, and growth enriched in the umbilical cord and bone marrow
factors to support erythropoiesis [12]. as well as peripheral blood, as indicated by the
The erythroblasts in the erythroblast island depend expression of CD34, CD105, CD36, CD71, and
on the macrophages for survival and differentiation. CD45RA markers. By using CD34/CD105 and GPA/
Macrophages provide adhesion molecules and phago- CD105 spectra, all four progenitor stages and five
cytose the extruded nuclei and organelles from eryth- terminal erythroid differentiation stages can be
roblasts. Additionally, macrophages produce growth identified [19].
factors and cytokines like erythropoietin, stem cell Further differentiation of human erythroid precur-
factor, and insulin-like growth factor-1, which sors can be classified by surface expression of the
promote erythropoiesis and the proliferation of transferrin receptor (CD71) and glycoprotein A
erythroblasts. (CD235a). Additionally, the expression of Integrin
The close association between macrophages and α4 (CD49D) and band three protein has been
erythroblasts within the erythroblast island creates a found to promote the resolution of multifarious
supportive microenvironment that stimulates the stages of erythroid precursor differentiation [1]. A
efficient output of functional erythrocytes. Dysfunction combination of GYPA (CD235A) and CD105
or disruption of this microenvironment can lead to expression or the gain of band3/SLC4A1 (CD233)
4 P. TANG AND H. WANG

Figure 2. An overview of erythropoiesis in the bone marrow. The formation of reticulocytes from hematopoietic stem cells (HSPC)
in the bone marrow takes about seven steps. During the first stage of erythropoiesis, hematopoietic stem cells produce mega-
karyocytes in addition to erythroid bursting-forming units (BFU-E)and erythroid colony-forming units(CFU-E). The second stage
of erythropoiesis is marked by the production of proerythroblasts (Pro-E), the first erythroid cells to be morphologically recogniz-
able. The pro-E then produces basophils (Bas-E), which cease to produce ribosomes during basophilization. Bas-E subsequently
produces polychromic phage (Poly-E), Poly-E subsequently produces positively stained erythrocytes (Ortho-E), and Ortho-E even-
tually produces reticulocytes. During erythropoiesis, the size of the cells decreases continuously, and the intracellular hemoglobin
content gradually increases.

expression and loss of CD49d can be used to track maturation [20]. In mice, Ter119, CD71, and HE4
erythroid maturation [16]. are markers of progenitor cells and erythrocytes,
Biomarkers of erythropoiesis in human and mouse and Ter119 is a marker of mature red blood cells.
is totally different. In humans, miR-451 and miR-144 In addition to EPO, SCF and IL-3 are key cytokines
are highly expressed during erythropoiesis and play that regulate early erythrocyte production in mice.
an important role in promoting erythrocyte differen- Similar to humans, TGF-β signaling also negatively
tiation. They are located in the same gene cluster regulates late erythropoiesis in mice. Transcription
and are regulated by the red transcription factor factors such as GATA-1 and KLF1 are key mouse ery-
GATA1. miR-486 is also upregulated during erythropoi- throid regulators [21].
esis and collaborates with GATA1 as a regulator of
erythrocyte differentiation. miR-221 and miR-222 are
down-regulated during the differentiation of human Regulatory mechanisms of erythropoiesis
hematopoietic progenitor cells into red blood cells.
Exogenous regulation and control of
miR-320 is associated with the expression of CD71
erythropoiesis
protein, a marker of reticular cell maturation in
human sickle cell anemia. CD71 and CD36 in early pro- Varieties of external elements can activate essential
genitor cells, glycoprotein A in late progenitor cells/ downstream signaling pathways that regulate the
precursors, and CD235a in mature red blood cells are differentiation, proliferation, and survival of erythro-
key markers of human erythropoiesis. cytes. One such factor is IL-3, which can stimulate the
In mice, as in humans, miR-451 and miR-144 are proliferation of early progenitors, containing BFU-E.
key red blood cell biomarkers in mice that promote Additionally, KIT ligands can bind to KIT (CD117), pro-
erythropoiesis when overexpressed. miR-150 is moting the proliferation of BFU-E, CFU-E, and erythro-
down-regulated during erythrocyte terminal differen- blasts [16].
tiation in mice. miR-221, miR-222 and miR-223 were Wnt signaling also plays a part in the regulation of
down-regulated during erythropoiesis in mice. let-7 erythropoiesis, particularly in the terminal differen-
miRNA is upregulated during mouse erythrocyte tiation of erythrocytes in myeloid sinusoidal
HEMATOLOGY 5

endothelial cells, as well as in the maturity of reticulo- 1000-fold in response to tissue hypoxia. In mice,
cytes and the overall function of red blood cells and HIF2a specifically activates EPO, which is required for
hematopoietic cells in mesenchymal stromal cell popu- EPO expression in the kidney and hepar. Destruction
lations [22]. of HIF2a leads to anemia and hematopoietic defects.
EPO binds to EPOR in the hepar and inhibits the
expression of hepcidin, which can also be inhibited
Regulation of erythropoiesis by EPO by EPO-derived factors [27]. Osteoblasts can also gen-
erate EPO to promote erythropoiesis under stable con-
EPO is a glycoprotein that plays a crucial role in adjust- ditions of constitutive HIF. Exogenous EPO and the
ing erythrocyte quality and hematopoiesis in transloca- formation of bone have a connection with raised
tion and formation of erythrocyte transcriptional bone marrow vascular density and angiogenesis in
programs [11]. During stress erythropoiesis, EPO many repair models. EPO directly stimulates osteogen-
quickly improves the appearance of the anti-apoptotic esis by targeting mesenchymal stromal cells and
regulator BCL-XL in early erythrocytes by STAT5 and hematopoietic stem cells, and indirectly stimulates
inhibits the pro-apoptotic proteins BIMsponse to alter- osteoblast differentiation by irritating HSCs to secrete
nations in tissue oxygenation [23]. EPO is produced by bone morphogenetic proteins. EPO also directly stimu-
renal stromal cells in adults and mainly by the hepar in lates osteoblast differentiation toward mature osteo-
embryos. EPO contributes to hematopoiesis of hema- clasts via Jak2 and PI3K pathways, resulting in an
topoietic stem and progenitor cells (HSPC) by promot- according decline in EPOR transcript levels [11]
ing erythroid proliferation and survival from CFU-E, (Figure 3).
which rapidly decreases with the terminal maturation
of the cell [16].
Regulation of erythropoiesis and iron
EPO induces erythropoiesis by binding to EPO
metabolism by the ERFE-ferritin-transferritin
receptor (EPOR) on precursors of erythroid cells in
axis
the marrow, promoting their survival, proliferation,
and differentiation. EPOR is expressed primarily on The regulation of iron metabolism and erythropoiesis
CFU-E, erythrocytes, and early basophils. Binding to is crucial, and it also involves the ERFE-ferritin-trans-
EPO prevents apoptosis of these erythroid progenitors ferrin axis [16]. Approximately two-thirds of the
[24]. body’s iron is stored in RBCs [28]. RBCs require 20–
The EPO/EPOR pathway stimulates erythroid devel- 25 mg of iron daily, which is supplied by iron trans-
opment by activating the JAK2/STAT3/STAT5 pathway, port proteins that contain only 3 milligrams of iron at
which aids in Nucl, NOXA, Fas, and FasL [25]. EPO also any given moment and are replaced every several
stimulates erythroid precursor cells in the marrow and hours [29]. Since the human body cannot excrete
spleen to quickly produce erythroferrone (ERFE) by iron, the balance between iron intake, transport, util-
STAT5 [26]. ization, and storage must be tightly regulated [30].
Activation of STAT3 by EPO/EPO receptors leads to Iron required by RBCs is obtained largely from
increased expression of genes involved in red blood macrophages phagocytosing old RBCs, and is sup-
cell progenitor cell survival (Bcl-xL), proliferation, and plied daily by 20–25 mg of iron, which is absorbed
differentiation. A key target of STAT3 is the transcrip- in the intestine by 1–2 mg (0.05% of total iron
tion factor GATA-1, which regulates the expression of content in the body) to make up the iron loss due
many red blood-cell specific genes, including those to intestinal epithelial cell loss and a small amount
involved in hemoglobin production. STAT3 is rigidly of blood loss in the skin [31].
and briefly regulated during erythropoiesis through Non-heme iron can be absorbed from the lumen by
EPO-mediated JAK2 activation, followed by rapid feed- the divalent metal transport protein 1 (DMT1) located
back inhibition through SOCS protein, phosphatase, at the apical surface of epithelial cells in the duodenum
and proteasome degradation to prevent overactivity. by the duodenal cytochrome B reductase (DCYTB),
This allows STAT3 to induce erythroid gene expression which reduces iron to ferrous metal, and heme iron
in a precise manner to match the physiological require- is absorbed more efficiently in the gut than non-
ments of red blood cell production [11]. heme iron [32]. Absorbed iron can be utilized by intes-
JAK2 inactivation leads to anemia, whereas JAK2 tinal cells, kept in ferritin, or transported to the circula-
mutations lead to raised erythrocyte quantity and ery- tion by iron a basolateral membrane of the intestinal
throcytosis. EPO and EPOR play critical roles in erythro- cells [33]. Iron absorption and tissue distribution are
cyte quality control, and their dysregulation can result regulated mainly by the interaction of the hepar hor-
in various hematological disorders. mones hepcidin and ferritin. Ferritin exerts its function
During hypoxia, the highly conserved hypoxia-indu- through three common forms: ferritin coordinated by
cible factor (HIF) signaling pathway is activated, and the side chains of proteins, ferritin in the heme loop,
the first target of HIF is EPO. EPO levels increase by and ferritin in the iron-sul cluster. The iron homeostasis
6 P. TANG AND H. WANG

Figure 3. An overview of the mechanism by which EPO regulates erythropoiesis. Hypoxia-inducible factor 2 (HIF-2) induces EPO
production, which in combination with EPOR activates MAPK, JAK2, and PI3K signaling and directly inhibits the expression of hep-
cidin. JAK2 subsequently activated both STAT3 and STAT5 signaling, in which STAT5 activated erythroid receptor (ERFE) and anti-
apoptotic regulator BCL-XL, promoting the development of nuclear translocation NRD. Stat5 also inhibited BIM, NOXA, Fas, and
FasL.

system is mainly affected by erythrocytes (erythrocytes similar to general ligand-induced endocytosis in vivo.
and their precursors in erythropoiesis-producing This process is triggered by transporter conformational
organs), two storage locations (hepatocytes in the changes induced by ferritin, leading to ubiquitin of the
hepar, macrophages in the spleen and hepar), lysine-rich cytoplasmic fraction that links the two six-
plasma transporting iron between tissues and organs, helix regions of ferritin. Ferritin is then targeted for
and absorptive intestinal cells in the duodenum [34]. lysosomal and proteasomal degradation through ubi-
Erythroferrone (ERFE) is a protein synthesized and quitination, which is facilitated by Rnf217, an impor-
secreted by red blood cells in the marrow and tant E3 ubiquitin ligase [26,34].
extramedullary regions, and is induced by EPO Hepcidin is regulated by various factors including
output. ERFE belongs to the C1Q tumor necrosis plasma iron concentration (majorly relying on the
factor-related protein family, and acts directly in contact of ferritin with the transferrin receptors TFR1
the hepar to decrease hepcidin expression. Unlike and TFR2), hepar iron reserve, systemic inflammation
EPO, which can stimulate erythropoiesis in both signals transmitted majorly by IL-6 to hepatocytes,
stress and homeostatic conditions, ERFE only and erythroid activity signals transmitted mainly
responds to stress erythropoiesis. ERFE inhibits hep- through ERFE concentration [34]. Excessive iron can
cidin expression, increases the supply of iron, and be toxic and cause oxidative DNA damage [35], and
helps maintain iron balance during periods of high in patients with iron overload, hepcidin levels are elev-
erythropoietic demand [26]. ated. The combination of hepcidin and ferritin results
Ferritin is synthesized in the hepar and upon in the rapid degradation of hepcidin. When systemic
binding to iron, it undergoes ubiquitination and gets iron levels are low, this results in decreased expression
degraded in the lysosomes. This process reduces the of hepcidin and raised iron mobilization. Hypoxia inhi-
transfer of ferritin to the cell membrane, keeps iron bits hepcidin expression, and the hypoxia-induced
inside the cell, and lowers the amount of iron in the hepcidin inhibition, unrelated to EPO and erythrocyte
blood. The regulation of ferritin by endocytosis is output, is mediated by hypoxia-mediated degradation
HEMATOLOGY 7

of C/EBPA, a key transcription factor required for hep- addition to hepcidin, inflammatory stimulation in the
cidin basal expression. Hepar hypoxia is a strong form of Toll-like receptor ligands straightly inhibits cel-
inhibitor of hepcidin expression during hepar inflam- lular ferritin mRNA quantity and reduces cellular
mation and IL-6 output. HIF has a hand in the suppres- plasma iron output [34].
sion of hepcidin, and animal studies have shown that The transferrin (Tf) complex binds one iron mol-
HIF activation in the mouse hepar decreases hepcidin ecule and has two receptors, TFR1 and TFR2. TFR1
expression and increases ferritin in the intestine and mediates the internalization of Tf in the iron-transfer
macrophages [27]. Stressful and ineffective erythropoi- pathway, and it can bind partially saturated transferri-
esis can also inhibit ferritin. GDF15 and TWSG1, tin, as well as iron in both the light and heavy chains of
members of the transforming growth factor B super- ferritin. The affinity of TFR1 for c-valve and n-valve
family, are considered to be pathological inhibitors of saturated transferritin is only 5 and 6 times that of
hepcidin in ineffective erythropoiesis., GDF15 and Apo-Tf, respectively. At physiological pH levels, Tf is
TWSG1 inhibit hepcidin expression [26] (Figure 4). discharged from TFR1 to the serum, where it can
HIF2α is activated in the intestine when there are combine with iron again [37]. The IRE/IRP system
low levels of iron in the epithelium, leading to an increases TFR1 expression. The IRE/IRP system func-
increase in hepcidin-mediated degradation of ferritin. tions to regulate cellular iron homeostasis, ensuring
During iron deficiency, PHD activity is reduced, which that cells can increase TfR1 expression when the iron
further enhances the expression of HIF2α. Although supply is insufficient, thereby enhancing iron absorp-
intestinal iron levels do not change during erythropoi- tion and utilization efficiency. Once the cellular iron
esis, the decreased oxygen levels in the intestine result level reaches an appropriate level, IRP dissociates
in HIF2α activation. Hepcidin exhibits bacteriostatic from the IRE, leading to a decrease in TfR1 expression
effects on several bacteria and fungi [27]. to prevent excessive iron uptake. When the cellular
The transferrin (Tf) complex consists of two lobes, iron level is low, IRP binds to the IRE (iron-responsive
each of which can bind one iron atom. The degree of element) located in the 3′ UTR (untranslated region)
Tf saturation reports the body’s iron capacity, which of TfR1 mRNA, stabilizing its structure and promoting
is typically between 20% and 45% in healthy human the stability and translation of TfR1 mRNA. As a
beings [36]. Transferrin is the only iron transport result, the expression level of TfR1 increases, allowing
protein known so far. Stable transferrin binding to cells to more effectively uptake iron [38]. TFR1 is over-
the cell membrane stimulates iron absorption by the expressed in differentiated erythroid cells. Nuclear
duodenum, enhances iron recovery by macrophages endosomes including Holo-Tf-TFR1 approach the mito-
from old red blood cells and other cells, and allows chondria and transfer iron to the organelle by rapid
the transfer of iron contained in the hepar [26]. Most and transient contact with the mitochondria, restrain-
transferrin molecules are bound with hepcidin most ing the cytoplasmic iron pool without triggering the
of the time, and this mechanism is quickly adjustable adjustment of the IR/IRP system [36].
when the concentration of hepcidin is reduced. In

Figure 4. Mechanism of the action of hepcidin. Various systemic factors regulate the expression of hepcidin. Plasma iron levels
and liver iron reserve levels are factors promoting ferritin expression. In contrast, systemic hypoxia, low iron levels, high IL-6 levels
in the inflammatory state, and HIF and ERFE were inhibitory factors of hepcidin. Ferritin can cooperate with the E3 ubiquitin ligase
Rnf217 for ubiquitylation and degradation of ferritin.
8 P. TANG AND H. WANG

Tfr2 is a transmembrane glycoprotein that acts as a (NURD) complexes, promote the binding of GATA1 to
receptor for circulating iron. It’s similar to the classical regulatory sites [16].
transferrin receptor Tfr1. Tfr2 plays an important part in During erythroid cell development, GATA factor
regulating iron homeostasis by controlling hepcidin switching occurs, whereby GATA2 protein levels are
levels. Loss-of-function mutations in Tfr2 result in low decreased and GATA1 protein levels are raised.
levels of hepcidin, excess circulating iron, and hemo- GATA1 targets several genes that are regulated by
chromatosis [39], while mutations in hepcidin cause GATA2 in hematopoietic stem and progenitor cells
severe hereditary iron overload [40]. [43]. In erythroid cells, GATA1 occupies both the pro-
Unlike TFR1, TFR2 is not adjusted by the IR/IRP moter and enhancer, but the enhancer activity
system. TFR2 expression is instead regulated post- decreases during differentiation, with GATA1 predomi-
translationally by its ligand, Holo-Tf [36]. TFR2 protects nantly binding to the promoter. Enhancers play a more
ferrous plasma Tf from lysosomal degradation and is a significant role in the early stages of cell differentiation,
part of the erythroid progenitor EpoR complex, critical while promoters are more critical in the terminal
for valid transfer of EpoR to the cell surface and final stages.
differentiation. In mouse erythroid progenitors, the GATA2 tran-
Hepatic TfR2 stimulates iron signaling to ferritin scription factor increases the production of the stem
to repress further iron influx into the blood when cell cytokine receptor KIT, while GATA1 is responsible
iron is present. In contrast, erythroid TfR2 limits for reducing KIT expression to promote terminal differ-
Epo sensitivity to prevent overdose erythropoiesis. entiation. However, in human erythroid progenitors,
Under iron-deficient conditions, TfR2 becomes high levels of KIT expression require GATA1 binding
unstable, leading to hepatic TfR2 downregulation, on erythroid-specific SE.
which inhibits iron signaling to ferritin, stimulates GATA1 is regulated by various factors at the post-
cellular iron excretion, and increases iron supply to transcriptional level. HSP27 and HSP70 members of
RBC. Reduction of erythroid TfR2 promotes Epo’s the heat shock protein family, along with ribosomal
sensitivity to erythropoiesis and contributes to the proteins, promote the ubiquitination and proteaso-
suppression of hepcidin by ERFE [41]. mal degradation of GATA1. HSP70 also plays a role
in preventing the caspase-3-mediated intranuclear
division of GATA1, while ribosomal proteins regulate
Transcriptional control of erythropoiesis
the normal translation of GATA1 [45]. Additionally,
RBC output is regulated by intrinsic transcription there is an interaction between GATA1 and p53,
factors that regulate the differentiation of erythroid which inhibits RNA Pol I transcription by restricting
cells. Among these transcription factors, GATA1 plays the RNA Pol I complex from assembling on the
an important part in directing the differentiation of rDNA facilitator. In cancer, the loss of function of
erythroid cells towards RBC lineage. Another key tran- p53 and mass output of ribosomes may lead to
scription factor involved in RBC output is KLF1. the inhibition of GATA1 by p53 [17]. On the other
Mutations in GATA1 and KLF1 are associated with hand, downregulation of p19INK4d reduces GATA1
various types of anemia in humans [1]. These transcrip- protein levels and impairs human terminal erythroid
tion factors activate and suppress regulatory networks differentiation, which is adjusted by the p-ERK-
involved in RBC output and have different functions in HSP70-GATA1 pathway, where PEBP1 serves as a
this process [16]. connection between p19INK4d and the p-ERK-
HSP70-GATA1 pathway [45].
Transcriptional regulation of GATA in
erythropoiesis Transcriptional regulation of KLF1 in
erythropoiesis
The GATA transcription factor, which contains zinc-
finger DNA binding domains, is critical for various bio- KLF1 is a protein that is abundant in erythroid cells and
logical processes, including hematopoiesis [42]. GATA contains three C2H2 zinc fingers at its C-terminus. It
factors interact with specific regulatory elements to connects its DNA consensus sequence (5′ ccmcrcccn)
mediate transcriptional changes [43]. GATA-1 is essen- mainly in the distal regulatory region, and is expressed
tial for the existence and differentiation of progenitors in both primitive and nucleated erythrocytes within
of red blood cells, while GATA-2 adjusts the survival the hematopoietic compartment [46]. Its primary role
and proliferation of hematopoietic stem and progeni- in erythropoiesis is the activation of gene transcription,
tor cells [12]. GATA1 has three functional domains, and it inhibits the differentiation of megakaryocytes
including an N-terminal activation domain and two while promoting early differentiation of RBCs [47].
homologous Zn finger domains located in the C-term- KLF1 plays an important part in the final stages of
inal region [44]. Various cofactors, such as FOG-1/ the cell cycle and chromatin condensation, which
ZFPM1, nuclear remodeling bodies, and deacetylase stimulate RBC output. In the absence of KLF1, RBC
HEMATOLOGY 9

denuclearization is impaired, CDKN2C and CDKN1A Reduced erythropoiesis


levels are reduced, and RBC proliferation is raised.
Anemia due to hematopoietic stem cell
Although KLF1 is predominantly a transcriptional acti-
abnormalities
vator, it can also act as a transcriptional inhibitor [16].
Coactivator-coinhibitory interactions under KLF1 MDS refers to a cluster of clonal diseases characterized
acetylation dynamically regulate downstream tran- by morphological dysplasia of hematopoietic stem
scriptional effects [47]. cells, hematopoietic failure, and peripheral cell loss,
leading to decreased erythropoiesis. MDS cells,
usually derived from pluripotent hematopoietic stem
Transcriptional regulation of erythropoiesis by cells, are malignant tumors that typically cannot be
other factors cured except through allogeneic stem cell transplan-
tation (SCT) [55]. About 50% of MDS patients possess
TGF-beta is a crucial factor in cell differentiation and
somatic mutations in the spliceosome gene, with
has significant effects on hematopoiesis [48]. TGFβ1
SF3B1 being the most frequent one [56], and SF3B1
is involved in cell survival, proliferation, and differen-
mutations primarily linked to abnormal 3′ mRNA spli-
tiation. Megakaryocytes are the primary source of
cing. In vitro and in vivo, SF3B1K700E cells showed
TGFβ1 in the bone marrow, but they can also be syn-
raised sensitivity to the spliceosome regulator E7107,
thesized by immune cells, macrophages, and marrow
and the usage of spliceosome regulators may prove
stromal cells. TGFβ1 inhibits early progenitor cell pro-
effective in treating SF3B1 mutational hematologic
liferation and stimulates erythroid terminal differen-
malignancies [57]. Moreover, loss or mutations in MDS
tiation through Smad2/3-Smad4 or tiff-1g signaling.
and TET2(4q24) have been linked, and TET2 mutations
Dysregulation of TGFβ1 may lead to myelofibrosis,
can cause various disorders such as myeloproliferative
while blocking it may result in the proliferation of ery-
neoplasms, chronic myelomonocytic leukemia, acute
throid progenitors [49]. Additionally, members of the
myeloid leukemia, and decreased erythropoiesis [58].
TGFβ family promote the degradation of HIF1α by inhi-
Aplastic anemia (AA) is characterized by marrow
biting VHL, an E3 ubiquitin ligase, which enhances
failure (BMF) status, which results in cytopenia and
hypoxia responses [50].
ineffective hematopoiesis. The disease is classified as
hereditary or acquired, with the latter usually associ-
ated with established germline mutations, and occurs
Unusual and common erythropoietic
in two peaks, one in patients aged 15–25 years and
disorders
another in those older than 60 years. The prognosis
Anemia is a condition characterized by a hemoglobin of severe or very severe acquired AA with supportive
level below 13 g/dL in adult males and below 12 g/ therapy alone is poor, with mortality rates exceeding
dL in non-pregnant females [51], and can result from 80% after two years [59]. The major factors that lead
decreased erythropoiesis, excessive red blood cell to AA are direct bone marrow injury, genetic mutations
destruction, or blood loss [52]. Reduced erythropoiesis that reduce the DNA repair capacity of hematopoietic
can be sorted into several categories, including anemia stem cells, immune factors, and possibly environ-
caused by abnormalities in hematopoietic stem cells mental contamination and viral infections [60]. Hema-
(e.g. aplastic anemia, PRCA, myelodysplastic syndrome, topoietic stem cell transplantation is the curative
leukemia), abnormalities in hematopoietic regulation treatment for AA, while immunosuppressive treatment
(e.g. myeloid necrosis, myelofibrosis, tumor disease, (IST) with horse anti thymocyte globulin and cyclos-
myeloid metastasis, bone marrow stromal-cell- porine A is the first-line treatment for elderly patients
involved diseases), hyperlymphocyte dysfunction (e.g. and all patients lacking matched sibling donors [61].
AA and immune-associated pancytopenia), abnormal-
ities in hematopoietic regulatory factors (e.g. chronic
disease-associated anemia), hyperapoptosis (e.g. par-
Anemia due to abnormal hematopoietic
oxysmal nocturnal hemoglobinuria), or deficiency of
regulation
hematopoietic materials (e.g. iron-deficiency anemia,
deficiency of iron, leucine, and vitamin B12, and mega- Chronic disease anemia (ACD), also acknowledged as
loblastic anemia) [53]. Additionally, low levels of sel- inflammatory anemia (AI), is the second most
enium, which is involved in redox reactions by common type of anemia worldwide. This type of
binding to the selenium family of cysteine-containing anemia is primarily induced by hepcidin, an inflamma-
proteins and has an antioxidant effect, can lead to tory cytokine that regulates iron homeostasis by block-
reduced erythroid cell number and differentiation ing intestinal iron absorption, trapping iron in
under stress, resulting in an ineffective response. It is reticuloendothelial cells, and reducing iron availability
important not to overlook selenium deficiency in for erythropoiesis [62]. The transferring of iron from
cases of reduced erythropoiesis [54]. macrophages to developing erythroid cells also
10 P. TANG AND H. WANG

requires transferrin FPN. In the absence of FPN, iron In beta-thalassemia, mutations in the HBB gene lead
remains trapped in macrophages, leading to impaired to insufficient production of beta-globin. This leads to
iron transferring and erythroid maturation arrest in excessive amounts of unstable alpha-globin chains
developing erythroid cells. Inflammatory cytokines that aggregate in red blood cell precursors, leading
also decrease EPO synthesis, impair erythroid progeni- to ineffective production of red blood cells and prema-
tor differentiation, and shorten the lifespan of mature ture destruction of red blood cells [70].
erythroid cells [63]. These signals also increase periph- There are two alpha-globin genes on chromosome
eral red cell depletion [50]. 16 – α1 and α2, and alpha-thalassemia can be caused
Oncogenic anemia is a type of chronic disease by a deletion or point mutation of one or both alpha-
anemia associated with the activation of cytokines globin genes, resulting in a defect in the alpha-globin
like interferon-gamma, interleukin-1, and tumor necro- chain. When alpha-globin production is impaired, excess
sis factor (TNF), which can suppress the output of β-globin chains form tetramers (HbH) with high oxygen
endogenous EPO, damage iron utilization, and affinity and poor stability, leading to hemolysis [71].
decrease erythroid precursor proliferation [64]. Apop- The severity of thalassemia is determined by the
tosis is a significant contributor to the pathogenesis degree of imbalance in the alpha – / non-alpha
of anemia in chronic kidney disease, and approxi- globin ratio. Patients with transfusion-dependent tha-
mately half of the patients with chronic heart failure lassemia (TDT) have severe imbalances and require life-
anemia also have evidence of endogenous apoptosis long transfusions. Patients with non-transfusion-
and/or functional iron deficiency [65]. dependent thalassemia (NTDT) have less imbalance
A new treatment for anemia in chronic kidney and anemia.
disease is the hypoxia-inducible factor proline The body tries to compensate for anemia by
hydroxylase inhibitor, which blocks the degradation increasing red blood cell production in the bone
of the transcription factor hypoxia-inducible factor, sti- marrow. However, this enlarged but ineffective eryth-
mulating erythropoiesis to physiological levels [66]. rocyte production leads to further abnormalities. For
example, it increases the intestinal absorption of iron
and inhibits the iron-regulating hormone hepcidin.
Anemia with abnormal erythrocyte morphology
This can lead to an iron overload in the liver and
Erythrocyte abnormalities can manifest as large, small, heart. Ineffective erythropoiesis also causes bone
or misshapen cells. Normal erythrocytes have a diam- marrow dilatation and extramedullary hematopoiesis,
eter of 7–8 μm and a mean cell volume (MCV) of 80– leading to bone abnormalities and splenomegaly [72].
95 fl, with larger erythrocytes above this range and
smaller erythrocytes below. The different subgroups
Erythrocytosis
of erythrocyte abnormalities include globin chain
defects (such as hemoglobinemia or thalassemia), Polycythemia is a condition where the erythrocyte
heme synthesis defects, iron availability defects, or pre- count exceeds the normal sex range, and it can be
cursor iron acquisition defects [67]. Macrocytic anemia, categorized into relative polycythemia induced by a
which is characterized by enlarged erythrocytes, can decrease in plasma volume and absolute polycythemia
be further categorized into megaloblastic anemia induced by an increase in the quality of red blood cells.
(caused by defects in RNA and DNA synthesis) and Primary polycythemia occurs due to the spontaneous
non-megaloblastic anemia [68]. output of erythrocytes, usually from myeloproliferative
Plasmodium infection not only reduces erythropoi- neoplasms (also known as pseudo polycythemia or
esis but also inhibits the bone marrow’s response to PV). On the other hand, secondary polycythemia
EPO and leads to abnormalities in nuclear ultrastruc- occurs when the body responds appropriately to elev-
ture, including polynuclei, nuclear fragments, internuc- ated serum erythropoietin levels [73], such as in post-
lear bridges, and irregular nuclear shapes. renal transplant polycythemia where erythropoiesis is
Furthermore, parasites have immune escape mechan- driven by both allografts and native kidneys [74]. Poly-
isms that modify host immune responses by increasing cythemia includes various types such as thrombosis,
cytokine output, including interleukin-6 (IL-6), IL-12, IL- idiopathic polycythemia, congenital polycythemia,
1, IL-10, tumor necrosis factor-alpha (TNFalpha), and hypoxic lung disease, and post-transplant polycythe-
interferon-gamma (IFN-gamma). These inflammatory mia [75]. The majority of polycythemia vera cases are
cytokines can strongly impact erythropoiesis [69]. linked to acquired JAK2 variants. Different types of
familial polycythemia are associated with various
Thalassemia defects, such as erythropoietin hypersensitivity,
Mutations in the hemoglobin synthesis gene lead to defects in the oxygen sensitivity pathway, or an
reduced or absent production of α or β-globin chains increase in hemoglobin affinity for oxygen [76]. Poly-
and an imbalance in the ratio of α / non-α-globin cythemia vera may also result from mutations in the
chains, resulting in thalassemia. congenital hypoxia-inducible factor pathway [77]. It is
HEMATOLOGY 11

Figure 5. Classification of Common Abnormal Erythropoietic Diseases. Abnormalities of erythropoiesis may be classified as
increased erythropoiesis. Reduced erythropoiesis and abnormal erythrocyte morphology. Reduced erythropoiesis can be
divided into hematopoietic stem cell abnormalities, dysregulation of hematopoiesis, and obstruction of hematopoietic material
utilization. Hypererythropoiesis may be classified as primary or secondary, or as relative or absolute, depending on the
amount of red blood cell mass.

crucial to distinguish PV from polycythemia due to erythroid cells by removing aging and impaired red
other causes because early detection and treatment blood cells, and by interacting with developing proto-
of PV can prevent numerous vasomotor and thrombo- red blood cells [80]. Moreover, macrophages interact
tic complications. Patients with PV often have an with erythroblasts through direct adhesion and indirect
enlarged plasma volume that can mask the polycythe- cells, such as erythrocyte-release positive regulators
mia, making diagnosis difficult if this basic fact is that inhibit the interaction of specific protein 6 (Gas6),
ignored [78]. Polycythemia may not cause any symp- vascular endothelial growth factor A (VEGF-A), and pla-
toms, or it may present with thrombosis, vasomotor cental growth factor (PlGF) to regulate stress erythropoi-
symptoms, or splenomegaly [73]. esis [81] (Figure 5).
When erythropoiesis is reduced, the body compen-
sates by inducing extramedullary erythropoiesis, also
known as stress erythropoiesis. Unlike the continuous
Therapeutic significance
output of erythrocytes in steady-state erythropoiesis, Anemia has a complex multi-factorial etiology. The main
stress-induced erythropoiesis produces a large number causes include nutritional deficiencies (iron, vitamin A,
of new erythrocytes through synchronous differentiation B12, folate), infectious diseases (malaria, HIV, hookworm),
of progenitor cells. The bone marrow temporarily inherited hemoglobin disorders (sickle cell, thalassemia),
migrates hematopoietic cells to the spleen to re-establish and inflammation. Fetal hemoglobin (HbF) induction
this process [50]. Early progenitor cells, which are usually remains a promising approach for the treatment of
in the quiescent phase, are favored by the HSPC com- sickle cell disease and beta-thalassemia. Strategies
ponent in extramedullary tissue, but proliferation is inhib- include targeting transcription factors that regulate
ited [79]. Additionally, anemia stress increases fetal to adult hemoglobin conversions, such as BCL11A,
erythrocyte phagocytosis, and splenic macrophages KLF1, and MYB, or using epigenetic regulators such as
promote CCL2 output, which recruits circulating mono- HDAC and LSD1 inhibitors. Gene therapy using lentiviral
cytes and forms new hematopoietic islands in the vectors to express the β-globin gene or shRNA targeting
spleen. Macrophages regulate the circulation of splenic BCL11A in hematopoietic stem cells has shown the
12 P. TANG AND H. WANG

potential to permanently correct hemoglobinopathy. ORCID


Gene editing using CRISPR/Cas9 to correct potential
Huaquan Wang http://orcid.org/0000-0003-2402-2717
mutations is also being explored. Erythropoietic drugs
such as PHD inhibitors and activin receptor traps (such
as Sotercept) are emerging to treat inflammatory
anemia, chronic kidney disease, cancer, and more. Man- References
agement of iron homeostasis with hepcidin modulators, [1] Nandakumar SK, Ulirsch JC, Sankaran VG. Advances in
small hepcidin, or TMPRSS6 inhibitors may help treat iron understanding erythropoiesis: evolving perspectives.
overload in patients with transfusion-dependent thalas- Br J Haematol. 2016;173(2):206–218. DOI:10.1111/bjh.
13938
semia. Anti-inflammatory therapies such as selectin
[2] Dzierzak E, Philipsen S. Erythropoiesis: development
inhibitors, anti-CXCR2, or iNKT cell blocking may alleviate and differentiation. Cold Spring Harb Perspect Med.
vaso-blocking crises and organ damage in sickle cell 2013;3(4):011–601.
disease. Enzyme or molecular replacement gene [3] Yumine A, Fraser ST, Sugiyama D. Regulation of the
therapy appears to hold promise for treating rare inher- embryonic erythropoietic niche: a future perspective.
ited anemia such as pyruvate kinase deficiency and con- Blood Res. 2017;52(1):10–17. DOI:10.5045/br.2017.52.1.10
[4] Yamane T. Cellular basis of embryonic hematopoiesis
genital erythropoietic porphyria [53].
and its implications in prenatal erythropoiesis. Int J
For polycythemia vera, maintaining hematocrit Mol Sci. 2020;21(24):9346. DOI:10.3390/ijms21249346
targets of <45% in men and <42% in women by phle- [5] Popescu DM, Botting RA, Stephenson E, et al. Decoding
botomy is the basis of treatment to prevent thrombo- human fetal liver haematopoiesis. Nature. 2019;574
sis, which is a major immediate health threat. Targeted (7778):365–371. DOI:10.1038/s41586-019-1652-y
[6] Palis J. Interaction of the macrophage and primitive ery-
therapies such as ruxolitinib and pegylated interferon
throid lineages in the mammalian embryo. Front
can be used for symptom control and to reduce sple- Immunol. 2016;7:669.
nomegaly, but their role in preventing thrombosis is [7] Baron MH, Vacaru A, Nieves J. Erythroid development in
unclear. Hydroxyurea is effective in reducing cells, the mammalian embryo. Blood Cells, Molecules, &
but does not affect disease progression, with risks Diseases. 2013;51(4):213–219. DOI:10.1016/j.bcmd.
such as myelofibrosis and leukemia transformation. 2013.07.006
[8] Chakraborty S, Andrieux G, Kastl P, et al. Erythropoietin-
Therefore, its use should be limited. Treatment for
driven dynamic proteome adaptations during erythro-
pregnant patients includes strict blood pressure poiesis prevent iron overload in the developing
control and phlebotomy, but aspirin and anticoagu- embryo. Cell Rep. 2022;40(12):111360. DOI:10.1016/j.
lants are avoided unless at high risk. Pegylated inter- celrep.2022.111360
feron is safe when necessary. For advanced [9] Hidalgo D, Bejder J, Pop R, et al. Epor stimulates rapid
cycling and larger red cells during mouse and human
myelofibrosis, ruxolitinib, thalidomide-prednisone,
erythropoiesis. Nat Commun. 2021;12(1):7334. DOI:10.
and azacytidine may be selected, but hematopoietic 1038/s41467-021-27562-4
stem cell transplantation is curable when feasible [78]. [10] Barminko J, Reinholt B, Baron MH. Development and
In conclusion, erythropoiesis is a complex and differentiation of the erythroid lineage in mammals.
dynamic process that occurs primarily in the marrow, Dev Comp Immunol. 2016;58:18–29. DOI:10.1016/j.dci.
but can also occur extramedullary under stress con- 2015.12.012
[11] Eggold JT, Rankin EB. Erythropoiesis, EPO, macro-
ditions. The process is influenced by various factors phages, and bone. Bone. 2019;119:36–41. DOI:10.
including bone marrow microenvironment, macro- 1016/j.bone.2018.03.014
phages, exogenous and endogenous factors, and [12] Zivot A, Lipton JM, Narla A, et al. Erythropoiesis: insights
intrinsic transcriptional regulators such as GATA and into pathophysiology and treatments in 2017. Mol Med.
KLF. Abnormalities in erythropoiesis can lead to 2018;24(1):11–11. DOI:10.1186/s10020-018-0011-z
[13] Iolascon A, Andolfo I, Russo R. Congenital dyserythro-
various hematologic disorders such as anemia, erythro-
poietic anemias. Blood. 2020;136(11):1274–1283.
cytosis, and polycythemia vera. Studies on erythropoi- DOI:10.1182/blood.2019000948
esis have provided valuable clinical insights, leading to [14] Valent P, Büsche G, Theurl I, et al. Normal and patho-
the development of new treatments for hematologic logical erythropoiesis in adults: from gene regulation
malignancies and anemia, and early recognition and to targeted treatment concepts. Haematologica.
prevention of thrombosis in polycythemia vera. 2018;103(10):1593–1603. DOI:10.3324/haematol.2018.
192518
[15] Ramos P, Casu C, Gardenghi S, et al. Macrophages
Disclosure statement support pathological erythropoiesis in polycythemia
No potential conflict of interest was reported by the author(s). vera and beta-thalassemia. Nat Med. 2013;19(4):437–
445. DOI:10.1038/nm.3126
[16] Caulier AL, Sankaran VG. Molecular and cellular mech-
Funding anisms that regulate human erythropoiesis. Blood.
2022;139(16):2450–2459. DOI:10.1182/blood.
The work is funded by National Natural Science Foundation 2021011044
of China (81170472), the Key Technology Research and [17] Le Goff S, Boussaid I, Floquet C, et al. P53 activation
Development Program of Tianjin China (18ZXDBSY00140). during ribosome biogenesis regulates normal erythroid
HEMATOLOGY 13

differentiation. Blood. 2021;137(1):89–102. DOI:10. [36] Kohgo Y, Niitsu Y, Kondo H, et al. Serum transferrin
1182/blood.2019003439 receptor as a new index of erythropoiesis. Blood.
[18] Gibson JS, Rees DC. Lipid metabolism in terminal ery- 1987;70(6):1955–1958. DOI:10.1182/blood.V70.6.1955.
thropoiesis. Blood. 2018;131(26):2872–2874. DOI:10. 1955
1182/blood-2018-05-850255 [37] Blanchette NL, Manz DH, Torti FM, et al. Modulation of
[19] Yan H, Ali A, Blanc L, et al. Comprehensive phenotyping hepcidin to treat iron deregulation: potential clinical
of erythropoiesis in human bone marrow: evaluation of applications. Expert Rev Hematol. 2016;9(2):169–186.
normal and ineffective erythropoiesis. Am J Hematol. DOI:10.1586/17474086.2016.1124757
2021;96(9):1064–1076. DOI:10.1002/ajh.26247 [38] Zhou ZD, Tan EK. Iron regulatory protein (IRP)-iron
[20] Sun L, Yu Y, Niu B, et al. Red blood cells as potential responsive element (IRE) signaling pathway in human
repositories of MicroRNAs in the circulatory system. neurodegenerative diseases. Mol Neurodegener.
Front Genet. 2020;11:442. DOI:10.3389/fgene.2020. 2017;12(1):75. DOI:10.1186/s13024-017-0218-4
00442 [39] Ginzburg YZ, Fleming RE. Tfr2 suppression benefits B-
[21] Parisi S, Finelli C, Fazio A, et al. Clinical and molecular thalassemic erythropoiesis. Blood. 2018;132(21):2215–
insights in erythropoiesis regulation of signal transduc- 2216. DOI:10.1182/blood-2018-10-876110
tion pathways in myelodysplastic syndromes and β-tha- [40] Kim A, Nemeth E. New insights into iron regulation and
lassemia. Int J Mol Sci. 2021;22(2):827. DOI:10.3390/ erythropoiesis. Curr Opin Hematol. 2015;22(3):199–205.
ijms22020827 DOI:10.1097/MOH.0000000000000132
[22] Heil J, Olsavszky V, Busch K, et al. Bone marrow sinusoi- [41] Pantopoulos K. Tfr2 links iron metabolism and erythro-
dal endothelium controls terminal erythroid differen- poiesis. Blood. 2015;125(7):1055–1056. DOI:10.1182/
tiation and reticulocyte maturation. Nat Commun. blood-2014-12-617571
2021;12(1):6963. DOI:10.1038/s41467-021-27161-3 [42] Fujiwara T. GATA transcription factors: basic principles
[23] Tsiftsoglou AS. Erythropoietin (EPO) as a key regulator and related human disorders. Tohoku J Exp Med.
of erythropoiesis, bone remodeling and endothelial 2017;242(2):83–91. DOI:10.1620/tjem.242.83
transdifferentiation of multipotent mesenchymal stem [43] Romano O, Petiti L, Felix T, et al. GATA factor-mediated
cells (MSCs): implications in regenerative medicine. gene regulation in human erythropoiesis.
Cells. 2021;10(8):2140. DOI:10.3390/cells10082140 iScience. 2020;23(4):101018. DOI:10.1016/j.isci.2020.
[24] Shih H, Wu C, Lin S. Physiology and pathophysiology of 101018
renal erythropoietin-producing cells. J Formos Med [44] Barbarani G, Fugazza C, Strouboulis J, et al. The pleiotro-
Assoc. 2018;117(11):955–963. DOI:10.1016/j.jfma.2018. pic effects of GATA1 and KLF1 in physiological erythro-
03.017 poiesis and in dyserythropoietic disorders. Front
[25] Oikonomidou PR, Rivella S. What can we learn from Physiol. 2019;10:91. DOI:10.3389/fphys.2019.00091
ineffective erythropoiesis in thalassemia? Blood Rev. [45] Han X, Zhang J, Peng Y, et al. Unexpected role for
2018;32(2):130–143. DOI:10.1016/j.blre.2017.10.001 p19INK4d in posttranscriptional regulation of GATA1
[26] Kautz L, Nemeth E. Molecular liaisons between erythro- and modulation of human terminal erythropoiesis.
poiesis and iron metabolism. Blood. 2014;124(4):479– Blood. 2017;129(2):226–237. DOI:10.1182/blood-2016-
482. DOI:10.1182/blood-2014-05-516252 09-739268
[27] Shah YM, Xie L. Hypoxia-inducible factors link iron [46] Gnanapragasam MN, Bieker JJ. Orchestration of late
homeostasis and erythropoiesis. Gastroenterology. events in erythropoiesis by KLF1/EKLF. Curr Opin
2014;146(3):630–642. DOI:10.1053/j.gastro.2013.12.031 Hematol. 2017;24(3):183–190. DOI:10.1097/MOH.
[28] Zhang A, Enns CA. Grab and go: transferrin uptake in 0000000000000327
erythropoiesis. Blood. 2022;140(10):1061–1063. DOI:10. [47] Kim MY, Yan B, Huang S, et al. Regulating the regula-
1182/blood.2022017638 tors: the role of histone deacetylase 1 (HDAC1) in ery-
[29] Ginzburg YZ. Chapter two – hepcidin-ferroportin axis in thropoiesis. Int J Mol Sci. 2020;21(22):8460. DOI:10.
health and disease. In: G Litwack, editor. Vitamins and 3390/ijms21228460
hormones. Academic Press; 2019. p. 17–45. [48] Nakamura K, Smyth MJ. Aberrant erythropoiesis fuels
[30] Sinha S, Pereira-Reis J, Guerra A, et al. The role of iron in tumor growth. Cell Res. 2018;28(6):611–612. DOI:10.
benign and malignant hematopoiesis. Antioxid Redox 1038/s41422-018-0047-1
Signal. 2021;35(6):415–432. DOI:10.1089/ars.2020.8155 [49] Villeval J, Vainchenker W. Megakaryocytes tame ery-
[31] Chifman J, Laubenbacher R, Torti SV. A systems biology thropoiesis with TGFβ1. Blood. 2020;136(9):1016–
approach to iron metabolism. In: J Chifman, R 1017. DOI:10.1182/blood.2020006906
Laubenbacher, SV Torti, editors. Advances in [50] Paulson RF, Ruan B, Hao S, et al. Stress erythropoiesis is
Experimental Medicine and Biology. New York, NY: a key inflammatory response. Cells. 2020;9(3). DOI:10.
Springer New York; 2014. p. 201–225. 3390/cells9030634
[32] Camaschella C, Nai A, Silvestri L. Iron metabolism and [51] Tomasz G, Ewa W, Jolanta M. Biomarkers of iron metab-
iron disorders revisited in the hepcidin era. olism in chronic kidney disease. Int Urol Nephrol.
Haematologica. 2020;105(2):260–272. DOI:10.3324/ 2021;53(5):935–944. DOI:10.1007/s11255-020-02663-z
haematol.2019.232124 [52] Chaparro CM, Suchdev PS. Anemia epidemiology,
[33] Wojtaszek E, Glogowski T, Malyszko J. Iron and chronic pathophysiology, and etiology in low- and middle-
kidney disease: still a challenge. Front Med (Lausanne). income countries. Ann N Y Acad Sci. 2019;1450
2020;7:565135. DOI:10.3389/fmed.2020.565135 (1):15–31.
[34] Nemeth E, Ganz T. Hepcidin-ferroportin interaction con- [53] Sankaran VG, Weiss MJ. Anemia: progress in molecular
trols systemic iron homeostasis. Int J Mol Sci. 2021;22 mechanisms and therapies. Nat Med. 2015;21(3):221–
(12):6493. DOI:10.3390/ijms22126493 230. DOI:10.1038/nm.3814
[35] Morales M, Xue X. Targeting iron metabolism in cancer [54] Dulmovits BM, Blanc L. Stress erythropoiesis: selenium
therapy. Theranostics. 2021;11(11):8412–8429. DOI:10. to the rescue!. Blood. 2018;131(23):2512–2513. DOI:10.
7150/thno.59092 1182/blood-2018-04-844720
14 P. TANG AND H. WANG

[55] Hellstrom-Lindberg E, Tobiasson M, Greenberg P. [69] Dumarchey A, Lavazec C, Verdier F. Erythropoiesis and
Myelodysplastic syndromes: moving towards personal- malaria, a multifaceted interplay. Int J Mol Sci. 2022;23
ized management. Haematologica. 2020;105(7):1765– (21):12762. DOI:10.3390/ijms232112762
1779. DOI:10.3324/haematol.2020.248955 [70] Musallam KM, Bou-Fakhredin R, Cappellini MD, et al.
[56] Malcovati L, Stevenson K, Papaemmanuil E, et al. SF3B1- 2021 update on clinical trials in β-thalassemia. Am J
mutant MDS as a distinct disease subtype: a proposal Hematol. 2021;96(11):1518–1531. DOI:10.1002/ajh.
from the international working group for the prognosis 26316
of MDS. Blood. 2020;136(2):157–170. DOI:10.1182/ [71] Munkongdee T, Chen P, Winichagoon P, et al. Update in
blood.2020004850 laboratory diagnosis of thalassemia. Front Mol Biosci.
[57] Obeng EA, Chappell R, Seiler M, et al. Physiologic 2020;7:74–74. DOI:10.3389/fmolb.2020.00074
expression of SF3B1K700E causes impaired erythropoi- [72] Motta I, Bou-Fakhredin R, Taher AT, et al. Beta thalasse-
esis, aberrant splicing, and sensitivity to pharmacologic mia: new therapeutic options beyond transfusion and
spliceosome modulation. Cancer Cell. 2016;30(3):404– iron chelation. Drugs (New York, NY). 2020;80
417. DOI:10.1016/j.ccell.2016.08.006 (11):1053–1063.
[58] Wojchowski DM. Ineffective erythropoiesis of TET2 [73] Mithoowani S, Laureano M, Crowther MA, et al.
deficiency. Blood. 2018;132(22):2320–2321. DOI:10. Investigation and management of erythrocytosis.
1182/blood-2018-10-878645 CMAJ. 2020;192(32):913–918. DOI:10.1503/cmaj.
[59] Schoettler ML, Nathan DG. The pathophysiology of 191587
acquired aplastic anemia: current concepts revisited. [74] Alzoubi B, Kharel A, Machhi R, et al. Post-transplant ery-
Hematol Oncol Clin North Am. 2018;32(4):581–594. throcytosis after kidney transplantation: a review. World
DOI:10.1016/j.hoc.2018.03.001 J Transplant. 2021;11(6):220–230. DOI:10.5500/wjt.v11.
[60] NS Y. Aplastic anemia. N Engl J Med. 2018;17 i6.220
(379):1643–1656. [75] McMullin MFF, Mead AJ, Ali S, et al. A guideline for the
[61] Bacigalupo A, Passweg J. Diagnosis and treatment of management of specific situations in polycythaemia
acquired aplastic anemia. Hematol Oncol Clin North vera and secondary erythrocytosis: a British society for
Am. 2009;23(2):159–170. DOI:10.1016/j.hoc.2009.01.005 haematology guideline. Br J Haematol. 2019;184
[62] Weiss G, Ganz T, Goodnough LT. Anemia of inflam- (2):161–175. DOI:10.1111/bjh.15647
mation. Blood. 2019 ;133(1):40–50. DOI:10.1182/blood- [76] Gaspersic J, Kristan A, Kunej T, et al. Erythrocytosis:
2018-06-856500 genes and pathways involved in disease development.
[63] Fraenkel PG. Anemia of inflammation: a review. Med Blood Transfus. 2021;19(6):518–532.
Clin N Am. 2017;101(2):285–296. DOI:10.1016/j.mcna. [77] Cazzola M, Cazzola M. Introduction to a review series on
2016.09.005 normal and pathologic erythropoiesis. Blood. 2022;139
[64] Abdel-Razeq H, Hashem H. Recent update in the patho- (16):2413–2414. DOI:10.1182/blood.2022015497
genesis and treatment of chemotherapy and cancer [78] Spivak JL. How I treat polycythemia vera. Blood. 2019;4
induced anemia. Crit Rev Oncol Hematol. (134):345–352.
2020;145:102837. DOI:10.1016/j.critrevonc.2019.102837 [79] Mende N, Bastos HP, Santoro A, et al. Unique molecular
[65] Cazzola M. Introduction to a How I treat series on and functional features of extramedullary hematopoie-
anemia. Blood. 2020. tic stem and progenitor cell reservoirs in humans.
[66] Hanna RM, Streja E, Kalantar-Zadeh K. Burden of anemia Blood. 2022;139(23):3387–3401. DOI:10.1182/blood.
in chronic kidney disease: beyond erythropoietin. Adv 2021013450
Ther. 2021;38(1):52–75. DOI:10.1007/s12325-020- [80] Haldar M. Stressed about erythropoiesis: EBI macro-
01524-6 phages. Blood. 2018;132(24):2530–2532. DOI:10.1182/
[67] Cappellini MD, Russo R, Andolfo I, et al. Inherited micro- blood-2018-10-882183
cytic anemias. Hematology. 2020;2020(1):465–470. [81] Ulyanova T, Phelps SR, Papayannopoulou T. The macro-
DOI:10.1182/hematology.2020000158 phage contribution to stress erythropoiesis: when less
[68] Newhall DA, Oliver R, Lugthart S. Anaemia: a disease or is enough. Blood. 2016;128(13):1756–1765. DOI:10.
symptom. Neth J Med. 2020;3(78):104–110. 1182/blood-2016-05-714527

You might also like