You are on page 1of 26

1104 SECTION XVIII — Developmental Hematopoiesis

FLSEC: Fetal liver sinusoidal endothelial cell 13. Notta F, Zandi S, Takayama N, et al. Distinct routes of lineage develop-
ment reshape the human blood hierarchy across ontogeny. Science.
GA: Gestational age 2016;351(6269):aab2116.
GGA: Golgi-localized γ-adaptin ear homology ARF binding protein 22. Popescu DM, Botting RA, Stephenson E, et al. Decoding human fetal liver hae-
GMP: Granulocyte monocyte progenitor matopoiesis. Nature. 2019;574(7778):365–371.
GPCR: G protein-coupled receptors 23. Höfer T, Rodewald HR. Differentiation-based model of hematopoietic stem cell
functions and lineage pathways. Blood. 2018;132(11):1106–1113.
HSC: Hematopoietic stem cell 51. Bjerregaard MD, Jurlander J, Klausen P, Borregaard N, Cowland JB. The in vivo
HSPA1A: Heat shock protein 1 profile of transcription factors during neutrophil differentiation in human
KC: Keratinocyte chemoattractant bone marrow. Blood. 2003;101(11):4322–4332.
LAD: Leukocyte adhesive deficiency 52. Cowland JB, Borregaard N. Granulopoiesis and granules of human neutrophils.
Immunol Rev. 2016;273(1):11–28.
LMPP: Lymphoid-primed multipotent progenitor 65. Hoogendijk AJ, Pourfarzad F, Aarts CEM, et al. Dynamic transcriptome-pro-
LPS: Lipopolysaccharide teome correlation networks reveal human myeloid differentiation and neutro-
LXA4: Lipoxin A4 phil-specific programming. Cell Rep. 2019;29(8):2505–2519.
MEP: Megakaryocyte erythroid progenitor 71. Grassi L, Pourfarzad F, Ullrich S, et al. Dynamics of transcription regulation in
human bone marrow myeloid differentiation to mature blood neutrophils. Cell
MIP: Macrophage inflammation protein Rep. 2018;24(10):2784–2794.
MPO: Myeloperoxidase 75. Mistry JJ, Marlein CR, Moore JA, et al. ROS-mediated PI3K activation drives
MPP: Multipotent progenitor mitochondrial transfer from stromal cells to hematopoietic stem cells in
Myd88: Myeloid differentiation factor 88 response to infection. Proc Natl Acad Sci USA. 2019:201913278.
139. Rørvig S, Østergaard O, Heegaard NH, Borregaard N. Proteome profiling of
NADPH: Nicotinamide adenine dinucleotide phosphate human neutrophil granule subsets, secretory vesicles, and cell membrane: cor-
NET: Neutrophil extracellular trap relation with transcriptome profiling of neutrophil precursors. J Leuko Biol.
NICU: Neonatal intensive care unit 2013;94(4):711–721.
NOD: Nod-like receptors 141. Lawrence SM, Corriden R, Nizet V. The ontogeny of a neutrophil: mechanisms
of granulopoiesis and homeostasis. Microbiol Mol Biol Rev. 2018;82(1):
NSP4: Neutrophil serine protease 4 e00057-00017.
OLFM-4: Olfactomedin 4 143. Le Cabec V, Cowland JB, Calafat J, Borregaard N. Targeting of proteins to gran-
PAMP: Pathogen-associated molecular pattern ule subsets is determined by timing and not by sorting: the specific granule
PGC: Primordial germ cells protein NGAL is localized to azurophil granules when expressed in HL-60 cells.
Proc Natl Acad Sci USA. 1996;93(13):6454–6457.
PIK3: Phosphoinositide 3-kinase 157. Morioka S, Maueröder C, Ravichandran KS. Living on the edge: efferocytosis at
PRM: Pattern recognition molecules the interface of homeostasis and pathology. Immunity. 2019;50(5):1149–1162.
PRR: Pattern recognition receptors 171. Deshmukh HS, Liu Y, Menkiti OR, et al. The microbiota regulates neutrophil
PTEN: Phosphate and tensin homolog homeostasis and host resistance to Escherichia coli K1 sepsis in neonatal mice.
Nat Med. 2014;20(5):524–530.
ROS: Reactive oxygen species 173. Cotten CM, Taylor S, Stoll B, et al. Prolonged duration of initial empirical antibi-
SCN: Severe congenital neutropenia otic treatment is associated with increased rates of necrotizing enterocolitis and
SGA: Small for gestational age death for extremely low birth weight infants. Pediatrics. 2009;123(1):58–66.
SGD: Neutrophil-Specific Granule Deficiency 174. Greenwood C, Morrow AL, Lagomarcino AJ, et al. Early empiric antibiotic use in
preterm infants is associated with lower bacterial diversity and higher relative
SLPI: Secretory leukocyte protease inhibitor abundance of Enterobacter. J Pediatr. 2014;165(1):23–29.
SNAP-23: 23-kDA synaptosome-associated protein 176. Christensen RD,Yoder BA, Baer VL, Snow GL, Butler A. Early-onset neutropenia
SNARE: Soluble N-ethylmaleimide-sensitive factor attachment in small-for-gestational-age infants. Pediatrics. 2015;136(5):e1259–1267.
protein receptor 177. Koenig JM, Christensen RD. Incidence, neutrophil kinetics, and natural history
of neonatal neutropenia associated with maternal hypertension. N Engl J Med.
TLR:Toll-like receptor 1989;321(9):557–562.
TNF:Tumor necrosis factor 178. Christensen RD, Henry E, Wiedmeier SE, Stoddard RA, Lambert DK. Low blood
UPR: Unfolded-protein response neutrophil concentrations among extremely low birth weight neonates: data
  
from a multihospital health-care system. J Perinatol. 2006;26(11):682–687.
185. Schmutz N, Henry E, Jopling J, Christensen RD. Expected ranges for blood neu-
A complete reference list is available at www.ExpertConsult.com. trophil concentrations of neonates: the Manroe and Mouzinho charts revisited.
J Perinatol. 2008;28(4):275–281.
187. Adrover JM, Del Fresno C, Crainiciuc G, et al. A neutrophil timer coordinates
SELECT REFERENCES immune defense and vascular protection. Immunity. 2019;50(2):390–402.
188. Winter C, Silvestre-Roig C, Ortega-Gomez A, et al. Chrono-pharmacological
6. Easterbrook J, Rybtsov S, Gordon-Keylock S, et al. Analysis of the spatiotem-
targeting of the CCL2-CCR2 axis ameliorates atherosclerosis. Cell Metab.
poral development of hematopoietic stem and progenitor cells in the early
2018;28(1):175–182.
human embryo. Stem Cell Reports. 2019;12(5):1056–1068.
189. Zhang D, Chen G, Manwani D, et al. Neutrophil ageing is regulated by the
8. Ivanovs A, Rybtsov S, Anderson RA, Turner ML, Medvinsky A. Identification of
microbiome. Nature. 2015;525(7570):528–532.
the niche and phenotype of the first human hematopoietic stem cells. Stem
190. Lawrence SM, Corriden R, Nizet V. Age-appropriate functions and dysfunc-
Cell Reports. 2014;2(4):449–456.
tions of the neonatal neutrophil. Front Pediatr. 2017;5(23):1–15.

109 Developmental Erythropoiesis


Timothy M. Bahr | Robin K. Ohls

present in the mesenchymal and endodermal layers of the yolk


ERYTHROCYTE KINETICS sac.1 These red blood cells, or hematocytoblasts, are the product
of primitive megaloblastic erythropoiesis,2 and they differ
SITES AND STAGES OF FETAL RED BLOOD CELL from erythrocytes formed later in gestation when definitive
PRODUCTION normoblastic erythropoiesis occurs. Primitive erythroblasts
EXTRAEMBRYONIC ERYTHROPOIESIS are nucleated and macrocytic (20 to 25 μm in diameter). They
Extraembryonic erythropoiesis begins in the fetal yolk sac have a mean corpuscular volume (MCV) of more than 180
by 14 days’ gestation. Small nests of nucleated blood cells are fL and have a characteristic fine nuclear chromatin pattern

Descargado para Mirko Flores Gonzalez (mfloresg8@upao.edu.pe) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en octubre 31,
2023. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
CHAPTER 109 — Developmental Erythropoiesis 1105

and a polychromatophilic cytoplasm containing abundant during fetal development.Yolk sac cells are capable of producing
hemoglobin.2,3 Red blood cells enter the embryonic circulation granulocytic, megakaryocytic, and erythroid colonies when
at 3 to 4 weeks’ gestation, coincident with joining of the vitelline transplanted into adult irradiated recipients and in conditioned
and umbilical circulations.4 newborn recipients,9,10 a finding that supports the view that
fetal stem cells possess the capacity to be pluripotent and their
INTRAEMBRYONIC ERYTHROPOIESIS differentiation is controlled by microenvironmental factors.
Definitive normoblastic erythropoiesis of the fetus begins
in the liver during the early first trimester.5 By 6 to 8 weeks’ MARROW, LIVER, AND BLOOD DIFFERENTIALS
gestation, the liver replaces the yolk sac as the primary site of Table 109.1 describes the differential counts in fetal liver, marrow,
red blood cell production, and by 10 to 12 weeks’ gestation, and circulating blood according to gestational age.11–14 The liver
extraembryonic erythropoiesis has essentially ceased. Red blood of the second-trimester human fetus is the primary organ for
cell production occurs in the liver throughout the remainder erythropoiesis,with a myeloid-to-erythroid ratio ranging from 0.07
of gestation, although production begins to diminish during at 14 to 17 weeks’ gestation to 0.2 at 21 to 24 weeks’ gestation.11
the second trimester as bone marrow erythropoiesis increases By 24 weeks the composition of the fetal bone marrow begins
(Fig. 109.1). Erythroblasts are first noted in the marrow at 8 to to resemble that of adult bone marrow, differing by the presence
9 weeks’ gestation.5 By the end of the third trimester, almost all of a large number of stromal elements, the absence of plasma
erythropoiesis is occurring in the bone marrow, although resi­ cells and lymph follicles, and an overall increased cellularity in
dual erythropoiesis may continue in the liver and may be found the fetal bone marrow. Unlike adult bone marrow, large fat cells
in other sites. In rodents, the spleen is also a site of erythropoiesis are not present in fetal bone marrow. Between 18 and 22 weeks’
before the onset of marrow red blood cell production. As with gestation, the mitotic index of the fetal bone marrow becomes
the liver, this site ceases production shortly after birth.6 It is virtually identical to that of adult bone marrow. The fetal bone
unclear whether the spleen contributes to erythropoiesis in the marrow myeloid-to-erythroid ratio starts to exceed the normal
human fetus. Although erythroid precursors have been identi­ adult bone marrow ratio (1.5 ± 0.4) early in midgestation and
fied as early as 6 to 7 weeks’ gestation in human splenic tissue,7 remains elevated even at the time of birth.
it is not clear whether such cells are actually developing within
splenic tissue or whether they are simply part of the circulation.8 PROGENITOR CELL CONCENTRATIONS
Studies of bone marrow cells in tissue culture have identified
ONTOGENY OF STEM CELLS specific committed red blood cell precursors, termed erythroid
Red blood cell precursors in the yolk sac are extremely primitive progenitors,15–18 on the basis of their characteristic growth
cells, and they may either disappear or seed other areas where in vitro. When bone marrow cells are placed in semisolid media
erythropoiesis later becomes prominent. The unicentric theory culture systems for 5 to 7 days, an erythropoietin (EPO)-sensitive
proposes that all hematopoietic stem cells originate from the erythroid progenitor cell, termed colony-forming unit–erythroid
yolk sac, then migrate from one hematopoietic site to another. (CFU-E), clonally matures into a single cluster containing 30 to
Moore and Metcalf9 demonstrated circulating pluripotent stem 100 normoblasts (Fig. 109.2). An erythroid-specific progenitor
cells in the peripheral blood just before the development of that is less differentiated than a CFU-E (and therefore a more
liver erythropoiesis. They suggested that the development of primitive cell) is termed a burst-forming unit–erythroid (BFU-
intraembryonic hematopoiesis requires an intact yolk sac, and E). Twelve to 14 days after cultures of bone marrow cells are
migration of stem cells from the yolk sac to hematopoietic tissue is initiated, a BFU-E develops into a large, multicentered colony
necessary for the development of intraembryonic hematopoiesis. of normoblasts, in which each center contains 200 to 10,000
The development of other organ systems via migration of cells normoblasts. Finally, the most primitive erythroid progenitor cell
(such as neural crest tissue) gives credence to this theory. identifiable through in vitro culture is termed a colony-forming
The multicentric theory suggests that a new clonal formation of unit–granulocyte, erythrocyte, macrophage, megakaryocyte
hematopoietic stem cells occurs at different sites of hematopoiesis (CFU-GEMM, or CFU-MIX). Twelve to 14 days after marrow cells
are placed in culture, this multipotent progenitor develops into
a mixed colony of both normoblast clusters and granulocyte-
Primitive Definitive
erythropoiesis erythropoiesis
macrophage clusters.
The ability of an organ to produce red blood cells is based on
Yolk sac the number of progenitor cells it contains, as well as the growth
100 Liver Marrow
factors stimulating those cells to proliferate. Determination of
erythroid progenitor numbers obtained from cell suspensions
80 of liver, marrow, spleen, and blood of second-trimester human
% Production

fetuses (Table 109.2) showed twice the number of multipotent


60 progenitors and erythroid progenitors in the fetal liver compared
with marrow.8,11,12 Erythroid progenitors from fetal liver also
40 appeared to be more sensitive to EPO (the primary erythroid
growth factor) than were progenitors from fetal marrow.
20 Progenitor cell concentrations in the spleen were nearly
identical to concentrations in the circulation.8–12 Whether the
progenitors isolated from liver and marrow represent different
0
subpopulations of progenitors (e.g., populations that express
0 4 8 12
16 20 24 28 32 36 40 different erythroid growth factor receptors, different receptor
Gestational age (wk) Birth numbers, different receptor affinity, or different cycling rates) is
Fig. 109.1 Sites and stages of fetal erythropoiesis. Primitive erythro- not known.
poiesis begins in the yolk sac at 2 to 3 weeks after conception. By the
end of the first trimester the liver has become the primary erythroid REGULATION OF ERYTHROPOIESIS
organ. The liver is the primary source of red blood cells during the Erythropoietic regulation in the human fetus differs markedly
second trimester, whereas the bone marrow is the primary source of from that in the adult. In the adult, erythropoietic regulation
red blood cells during the last trimester. primarily involves maintaining the red blood cell mass. In contrast,

Descargado para Mirko Flores Gonzalez (mfloresg8@upao.edu.pe) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en octubre 31,
2023. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
1106 SECTION XVIII — Developmental Hematopoiesis

Table 109.1 Differential Counts in Liver, Marrow, and Blood During Gestation.

Differential Counts (Percentage ± SD) of Liver Cell Suspensions From Fetuses at a Gestational Age of 14–24 wk
Gestational Age (wk)
Cells 14–17 (n = 5) 18–20 (n = 7) 21–24 (n = 8)
Normoblast
Pronormoblast 3.1 ± 1.4 3.4 ± 1.0 2.9 ± 1.8
Basophilic 18.4 ± 7.6 13.7 ± 1.6 13.7 ± 4.7
Polychromatophilic 57.5 ± 12.6 55.3 ± 7.5 51.1 ± 5.5
Orthochromic 13.9 ± 5.2 15.9 ± 3.7 14.2 ± 5.0
Total erythroid cells 9.3 ± 6.3 87.8 ± 4.8 81.9 ± 5.0
Neutrophil
Promyelocyte 0±0 0.2 ± 0.2a 1.2 ± 0.4a,b
Myelocyte 0±0 0±0 0.2 ± 0.2a,b
Metamyelocyte 0±0 0±0 0±0
Band 0±0 0±0 0±0
Segmented 0±0 0±0 0±0
Total neutrophils 0±0 0.2 ± 0.2 1.4 ± 0.5a,b
Undifferentiated blast 0.5 ± 0.6 3.1 ± 1.8a 2.2 ± 0.7a
Macrophage 0.5 ± 0.6 1.2 ± 4.0a 1.3 ± 5.0
Lymphocyte 5.4 ± 2.6 3.9 ± 3.6 11.3 ± 4.2a,b
Eosinophil 0±0 0±0 0±0
Otherc 0.8 ± 0.8 3.4 ± 2.4 1.9 ± 2.4
Differential Counts (Percentage ± SD) of Marrow Cell Suspensions From Fetuses at a Gestational Age of
14–24 wk
Gestational Age (wk)
Cells 14–17 (n = 6) 18–20 (n = 6) 21–24 (n = 8)
Normoblast
Pronormoblast 0.3 ± 0.2d 1.1 ± 0.4d 0.5 ± 0.2d
Basophilic 1.4 ± 0.6d 3.1 ± 0.8d 1.7 ± 1.0d
Polychromatophilic 9.2 ± 5.0d 12.9 ± 6.2d 12.3 ± 5.2d
Orthochromic 12.1 ± 11.1 20.7 ± 12.2 8.1 ± 4.4
Total erythroid cells 23.0 ± 13.3d 37.8 ± 16.8d 22.6 ± 9.0d
Neutrophil
Promyelocyte 5.9 ± 2.9d 5.8 ± 2.6d 5.0 ± 1.7d
Myelocyte 2.7 ± 1.4d 3.4 ± 1.6d 2.1 ± 1.0d
Metamyelocyte 2.1 ± 1.0d 2.5 ± 1.8d 1.7 ± 1.0d
Band 3.1 ± 1.8d 3.2 ± 2.7d 1.6 ± 1.2d
Segmented 1.2 ± 1.3d 1.1 ± 1.3d 0.4 ± 0.2d
Total neutrophils 13.8 ± 6.1d 16.1 ± 8.0d 10.7 ± 5.0d
Undifferentiated blast 8.9 ± 12.4d 11.4 ± 5.4d 11.6 ± 3.6d
Macrophage 5.4 ± 3.8d 4.2 ± 2.4d 3.1 ± 1.6d
Lymphocyte 35.3 ± 15.8d 28.8 ± 8.0d 50.3 ± 6.8d
Eosinophil 0.5 ± 0.6 1.1 ± 1.2 1.2 ± 0.8d
Othere 0.8 ± 0.4 0.5 ± 0.4 0.4 ± 0.2
Differential Counts (Percentage ± SD) of Umbilical Cord Blood Samples From Fetuses at a Gestational Age of 18–29 wk
Gestational Age (wk)
Cells 18–21 (n = 186) 22–25 (n = 230) 26–29 (n = 144)
Normoblast (%WBCs) 45.0 ± 86.0 21.0 ± 23.0 21.0 ± 67.0
Basophilic 0.5 ± 1.0 0.5 ± 1.0 0.5 ± 1.0
Neutrophil 6.0 ± 4.0 6.5 ± 3.5 8.5 ± 4.0
Lymphocyte 88.0 ± 7.0 87.0 ± 6.0 85.0 ± 6.0
Eosinophil 2.0 ± 3.0 3.0 ± 3.0 4.0 ± 3.0
Monocyte 3.5 ± 2.0 3.0 ± 2.5 3.0 ± 2.5

aP < .05 versus 14 to 17 weeks.


bP < .05 versus 18 to 20 weeks.
cHepatocyte, megakaryocyte, or cell of undetermined origin.
dP < .05 versus fetal liver.
eMegakaryocyte or cell of undetermined origin.

WBCs, White blood cells.

Descargado para Mirko Flores Gonzalez (mfloresg8@upao.edu.pe) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en octubre 31,
2023. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
CHAPTER 109 — Developmental Erythropoiesis 1107

Pluripotent stem CFU-GEMM BFU-E CFU-E Normoblast Reticulocyte Mature


cell erythrocyte
Stem cell factor

IL-6

IL-9

IL-3

GM-CSF

EPO

Fig. 109.2 Erythropoietic progenitors and the growth factors influencing erythropoiesis. BFU-E, Burst-forming unit–erythroid; CFU-E, colony-
forming unit–erythroid; CFU-GEMM, colony-forming unit–granulocyte, erythrocyte, macrophage, and megakaryocyte; EPO, erythropoietin; GM-
CSF, granulocyte-macrophage colony-stimulating factor; IL-3, interleukin-3; IL-6, interleukin-6; IL-9, interleukin-9.

interleukin (IL)-3, IL-6, IL-9, and IL-12.23–26 Thrombopoietin, a


Table 109.2 Progenitor Cell Concentrations in growth factor involved in platelet production whose receptor is
Fetal Liver, Marrow, Spleen, and Blood. similar in structure to the EPO receptor, also stimulates erythroid
colony formation.27 IL-3 and GM-CSF were the first growth
CFU-MIX BFU-E factors noted to have erythroid-stimulating properties, initially
Liver 12.7 ± 2.1 20.7 ± 3.1 described as “burst-promoting activity.” In combination with
Marrow 6.7 ± 1.4a 9.3 ± 2.7a EPO, these factors synergistically stimulate differentiation and
Spleen 4.2 ± 2.7a 5.9 ± 3.7a proliferation of BFUs-E and CFUs-GEMM.
Blood – 8.0 ± 5.4a
Stem cell factor is a multipotent growth factor that, in
aP
combination with other factors, supports clonal maturation of
< .05 versus liver.
Numbers represent cell concentrations per 5 × 103 cells plated.
hematopoietic progenitors. Murine studies indicate that stem cell
BFU-E, Burst-forming unit–erythroid; CFU-MIX, colony-forming unit– factor may be expressed during embryogenesis, thereby affecting
granulocyte, erythrocyte, macrophage, and megakaryocyte. embryonic and fetal erythropoiesis. In vitro studies show that
stem cell factor alone stimulates clonal maturation of fetal
(but not adult) multipotent progenitors. Erythroid progenitors
isolated from term umbilical cord blood are more responsive to
constant and dramatic changes characterize erythropoiesis in the stem cell factor, alone and in synergism with GM-CSF and IL-3,
embryo and fetus. The incredible rate of somatic growth and the than are adult marrow progenitors.22
resultant need to constantly increase the fetal red blood cell mass Term circulating erythroid progenitors are also more sensitive to
necessitate an extraordinary erythropoietic effort. Moreover, IL-6 and IL-9 than are adult progenitors.24 IL-6 is a multifunctional, 22-
the relatively low oxygen tensions but high metabolic rates to 26-kDa glycoprotein cytokine involved in B-cell stimulation and
of fetal tissues require a system of oxygen delivery that differs immunoglobulin production, acute-phase reactions, and induction
significantly from the adult system. of hematopoietic progenitors from a noncycling (G0) phase into an
active cycling (S) phase. IL-6 alone supports clonogenic maturation of
ERYTHROID GROWTH FACTORS newborn umbilical cord blood BFUs-E and CFUs-GEMM and induces
The production of erythrocytes, from pluripotent stem cell to progenitor cell cycling.24 IL-9 is also a multipotent cytokine and is
mature red blood cell, is governed by various growth factors. similar to IL-6 in its ability to stimulate primitive erythroid progenitors
These erythropoietic growth factors are produced by accessory and multipotent progenitors isolated from umbilical cord blood.25
cells such as liver macrophages and marrow stromal cells, and Insulin-like growth factor 1 has been shown to cause clonal expansion
they stimulate maturation, growth, and differentiation at various of erythroid progenitors isolated from adult marrow28,29 and on
stages of red blood cell production. The progenitors involved in erythroid and mesenchymal-like cells in the fetal spleen. A recently
red blood cell production and the factors that stimulate their identified growth factor termed regulator of human erythroid cell
cellular maturation are depicted in Fig. 109.2. Although these expansion is an erythroid growth factor that promotes formation
growth factors all facilitate production of red blood cells, none of hemoglobinizing erythroblasts.30 It comprises a new EPO/EPO
plays a more important regulatory role than EPO. EPO is a 30- receptor target and regulator of human erythroid cell expansion that
to 39-kDa glycoprotein that binds to specific receptors on the additionally acts to support late-stage erythroblast development.
surface of erythroid precursors and stimulates their differentiation
and clonal maturation into mature erythrocytes.19,20 The EPO UNIQUE FEATURES OF FETAL PROGENITORS
gene (EPO) is located on chromosome band 7q21-22.21 During Fetal erythroid progenitors respond in a slightly different fashion
fetal erythropoiesis, EPO is produced principally by cells of than do adult erythroid progenitors. In addition to the features
monocyte/macrophage origin residing in the liver. Postnatally, EPO noted earlier, fetal progenitors appear more sensitive to EPO than
is produced almost exclusively by peritubular cells of the kidney. adult erythroid progenitors.31 Specifically, BFUs-E of fetal origin
Other growth factors besides EPO play a role in the develop more rapidly into erythroid colonies, and the colonies
differentiation and clonal expansion of erythroid progenitors. generally contain significantly more normoblasts. In addition,
These include granulocyte-macrophage colony-stimulating factor BFUs-E from adult bone marrow require a combination of EPO
(GM-CSF),15 stem cell factor (also known as c-kit ligand),22 and plus another factor, such as IL-3 or GM-CSF, to mature clonally;

Descargado para Mirko Flores Gonzalez (mfloresg8@upao.edu.pe) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en octubre 31,
2023. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
1108 SECTION XVIII — Developmental Hematopoiesis

Table 109.3 Blood Cell Indices During Gestation.

Gestational Age White Blood Total White Blood Platelets Red Blood Cells Hemoglobin Hematocrit Mean Corpuscular
(wk) Cellsa (×109/L) Cells (×109/L) (×109/L) (×1012/L) (g/dL) (%) Volume (fL)
18–21 (n = 760) 4.68 ± 2.96 2.57 ± 0.42 234 ± 57 2.85 ± 0.36 11.69 ± 1.27 37.3 ± 4.3 131.1 ± 11.0
22–25 (n = 1200) 4.72 ± 2.82 3.73 ± 2.17 247 ± 59 3.09 ± 0.34 12.2 ± 1.6 38.6 ± 3.9 125.1 ± 7.8
26–29 (n = 460) 5.16 ± 2.53 4.08 ± 0.84 242 ± 69 3.46 ± 0.41 12.91 ± 1.38 40.9 ± 4.4 118.5 ± 8.0
>30 (n = 440) 7.71 ± 4.99 6.40 ± 2.99 232 ± 87 3.82 ± 0.64 13.64 ± 2.21 43.6 ± 7.2 114.4 ± 9.3

aIncluding
normoblasts.
From Forestier F, Daffos F, Catherine N. Developmental hematopoiesis in normal human fetal blood. Blood. 1991;77:2360.

however, many fetal BFUs-E mature in the presence of EPO of EPO mRNA,59 and quantitative mRNA studies in second-
alone.32 Studies have shown that fetal clones produce GM-CSF trimester human fetuses reveal that the fetal kidney produces
and IL-3, which may explain their unique capability for growth approximately 5% of the amount of EPO mRNA that the fetal
factor independence and auto-stimulation.33 liver produces during the second trimester.60 Thus it appears that
regulation of EPO gene transcription differs between the liver
CONTROL OF ERYTHROPOIETIN PRODUCTION and the kidney in utero.
Erythropoiesis in utero is controlled by erythroid growth factors One mechanism by which gene expression can be
produced by the fetus, not the mother. EPO is the primary differentially regulated is through methylation of promoter and
regulator of erythropoiesis in adults and appears to be the enhancer regions of a gene, and another is through deacetylation
controlling factor for fetal erythropoiesis, especially during of histones.61–65 Increased methylation generally results in
late gestation. EPO does not cross the placenta in humans,34,35 decreased gene expression, whereas histone deacetylation causes
monkeys, or sheep,36 although it has been reported to do so in relaxation of DNA structures and allows gene expression to
mice.37 In the mouse, suppression of maternal erythropoiesis occur.These two mechanisms may be involved in developmental
by hypertransfusion does not suppress fetal erythropoiesis.38 In EPO gene expression. For example, methylation of the enhancer
humans, stimulation of maternal EPO production does not result region (leading to decreased expression) of the EPO gene in
in stimulation of fetal red blood cell production.39 developing human kidney during the second trimester was
The expression of EPO is thought to be controlled by an oxygen- much greater than in the liver.66
sensing mechanism in the liver and kidney.40 Both hypoxia and EPO levels have been measured in umbilical cord blood
anemia stimulate erythropoiesis by stimulating messenger RNA during the third trimester, and these levels gradually increase
(mRNA) transcription and EPO production.41 Two factors, hepatic throughout later development. 67–69 From EPO measurements
nuclear factor 4 and hypoxia-inducible factor 1 (HIF-1), serve as made in umbilical cord blood from infants of laboring and
transcription factors for EPO as well as other hypoxia-inducible genes. nonlaboring mothers70 and from infants undergoing labor
Hepatic nuclear factor 4 binds to the EPO promoter and enhancer stress,71–75 it seems likely that individual umbilical cord
regions of the gene. HIF-1 is a basic helix-loop-helix transcription EPO levels primarily reflect hypoxic stress during labor
factor composed of HIF-1α and HIF-1β subunits that bind to cis- and delivery. Serum EPO concentrations at birth normally
acting hypoxia-response elements and induce EPO transcription. range from 5 to 100 mU/mL. For comparison, serum EPO
HIF-1 is expressed in many cells and is involved in up-regulating a concentrations in anemic, nonuremic adults may be as high
variety of oxygen-regulated proteins, including vascular endothelial as 300 to 400 mU/mL.76,77
growth factor. HIF-1α appears to be constitutively expressed and
rapidly degraded under normoxic conditions. RNA stability depends RED BLOOD CELL INDICES IN THE FETUS AND
on the ubiquitin proteasome degradation system; inhibition of NEONATE
this system leads to increased levels of HIF-1 and EPO, even under RED BLOOD CELL CONCENTRATIONS AND
normoxic conditions. Promoter and enhancer elements within the HEMATOCRIT
EPO gene are responsive to hypoxia, as well as to cobalt exposure Red blood cell indices change during gestation and continue to
in vitro.42 In animal models, the liver-sensing mechanism has a change through the first year of life. Circulating red blood cell
decreased sensitivity to hypoxia, producing one tenth the amount concentrations gradually increase during the second trimester,
of EPO in response to comparable stimuli in the kidney.43,44 The from (2.85 ± 0.36) × 106/μL at 18 to 21 weeks to (3.82 ±
liver also appears to require more prolonged hypoxia to achieve 0.64) × 106/μL at 30 weeks (Table 109.3).12 At term gestation,
an EPO response.45,46 Other substances, including testosterone,47 circulating red blood cell concentrations range from 5.0 × 106
estrogen,48 thyroid hormone,49 prostaglandins,50 vitamin E,51 and to 5.5 × 106/μL.78 In parallel with increasing red blood cell
lipoproteins,52 have been shown to enhance EPO production or its concentrations, hematocrits increase from 30% to 40% during the
effects, both in vivo and in vitro.53,54 second trimester and continue to increase to term values over
It is not known what factors regulate the switch of EPO the latter part of the third trimester.78 Changes in hematocrit
production from the liver to the kidney. Renal production of EPO from 22 to 42 weeks’ gestation can be seen in Fig. 109.3.79 Term
is not necessary for normal fetal erythropoiesis.55 The lack of a hematocrits range from 50% to 63%, with some variability noted
renal contribution to EPO production is illustrated by the normal because of delayed clamping of the umbilical cord.80 Values
serum EPO concentrations and normal hematocrits of anephric are also dependent on the sampling site. Capillary hemoglobin
fetuses.56 In the sheep, EPO production in both the liver and the concentrations are as much as 3.5 g/dL higher than venous
kidney is highest at 60 days’ gestation (term being 140 days).57 hemoglobin concentrations.81
EPO production in the fetal liver decreases by 90 days’ gestation;
however, increased renal production of EPO continues until 130 HEMOGLOBIN
days’ gestation, when production falls to levels seen in adult sheep.58 The hemoglobin concentration gradually rises during gestation.82
Studies in human fetal and neonatal kidney obtained from At 10 weeks gestation the average hemoglobin concentration
postmortem specimens also report measurable quantities is approximately 9 g/dL.83 By 22 to 24 weeks’ gestation, fetal

Descargado para Mirko Flores Gonzalez (mfloresg8@upao.edu.pe) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en octubre 31,
2023. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
CHAPTER 109 — Developmental Erythropoiesis 1109

70 (3) plasma dilution and an increase in blood volume related to


growth. Multiple groups have also hypothesized that a process
60 termed neocytolysis (the selective lysis of young erythrocytes)
contributes to excessive hemolysis after birth, and thus a rapid
50 decrease in hematocrit as the neonate adapts to its new, relatively
hyperoxic environment.88–90
HCT (%)

40 The nadir of hemoglobin concentration in term infants


occurs at approximately 8 weeks, with an average hemoglobin
30 concentration of 11.2 g/dL.78,83 Hemoglobin values gradually
rise such that, by 6 months, the average term infant has a
20
hemoglobin concentration of 12.1 g/dL. Altitude may have
a modest effect on the postnatal changes in hemoglobin
10
concentration; infants living at 1600 m had higher hemoglobin
0 concentrations (by 0.4 g/dL) by 6 months of age than infants
22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42
living at sea level.91,92
The average decline in the hemoglobin concentration of
Gestational age (wk)
preterm very low-birth-weight infants (<1500 g) is remarkably
different from that of term infants, in part due to phlebotomy
losses that invariably occur in preterm infants, as well as the
20 effects of transfusions on endogenous erythropoiesis. Very low-
birth-weight infants reach a nadir of hemoglobin concentration
of 8 g/dL at 4 to 8 weeks of age.93 Fig. 109.4 and Table 109.4
15
HGB (g/dL)

demonstrate relationships among birth weight, chronologic age,


and red blood cell indices in term and preterm infants.78,79,83,92–96
10 Red blood cell indices may differ from normal ranges in
infants born small for their gestational age, in whom placental
insufficiency and secondary polycythemia are common.91–98
5 Infants of diabetic mothers, infants of mothers who smoke,
and infants born at higher altitudes also tend to have higher
hemoglobin concentrations at birth.91–96,98–100 In growth-
0
restricted infants born to hypertensive mothers, the blood
22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42
supply to the placenta and the capacity to deliver oxygen to
Gestational age (wk) the fetus are diminished. Accelerated erythropoiesis is likely
Fig. 109.3 Reference ranges for hematocrit and hemoglobin from 22 part of a compensating mechanism that raises oxygen-carrying
to 42 weeks’ gestation. Reference ranges are shown for hematocrit capacity, maintaining an adequate supply to the fetus. In infants
(HCT; upper panel) and hemoglobin concentration (HGB; lower panel) of diabetic mothers, increased metabolic demands of the fetus
at 22 to 42 weeks’ gestation. Tests were done during a 6.5-year pe- (resulting from increased glucose availability) may account
riod on more than 20,000 neonates. Values were excluded when the for higher fetal oxygen needs and compensatory increase in
diagnosis included abruption, placenta previa, or known cases of fetal hemoglobin concentration by the fetus. The increased red
anemia or when a blood transfusion was given before the first hema- blood cell mass in infants of diabetic mothers does not result
tocrit was measured. The solid line represents the mean value, and from higher maternal levels of hemoglobin A1C (a high-affinity
the dashed lines represent the 5% and 95% reference range. (From hemoglobin capable of decreasing the oxygen transferred to
Christensen RD, Jopling J, Henry E, et al. The erythrocyte indices of the fetus). In mothers who smoke, the increase in fetal carbon
neonates, defined using data from over 12,000 patients in a multihos- monoxide and the subsequent decrease in available oxygen are
pital system. J Perinatal. 2008;28:24–28.) the likely causes of the compensatory increase in hemoglobin
levels in the fetus.

hemoglobin values reach 11 to 12 g/dL, and by 30 weeks the MEAN CORPUSCULAR VOLUME
hemoglobin concentrations are 13 to 14 g/dL.12 Premature The size of the red blood cell gradually decreases during
male infants reach term umbilical cord hemoglobin values development. The MCV is more than 180 fL in the embryo, falls
earlier than premature female infants,84 possibly because to 130 fL by midgestation, and decreases to 115 fL by the end
of the erythropoietic effects of testosterone.47 Hemoglobin of pregnancy (Fig. 109.5). By 1 year of age, the MCV reaches
concentrations are relatively constant during the last 6 to an average of 82 fL.95 Similar to cells of other organs in infants
8 weeks of gestation (see Fig. 109.3); at term the average born prematurely, the red blood cell MCV declines quickly
hemoglobin concentration is approximately 16 to 17 g/dL.85–87 after birth, and the postpartum changes in MCV appear to be
An increase in hemoglobin concentration by 2 hours of related to chronologic age rather than postmenstrual age.94 The
postnatal life occurs in most infants, resulting from a decrease mean corpuscular hemoglobin concentrations remain relatively
in plasma volume. By 8 to 12 hours after birth, the hemoglobin constant, and the mean corpuscular hemoglobin decreases
concentration achieves a relatively constant level. Red blood cell slightly.100
production decreases significantly at birth, so that hemoglobin
concentrations gradually decline by the end of the first postnatal BLOOD VOLUME
week.80 The decrease in red blood cell production after birth The placenta and umbilical cord contain 75 to 125 mL of blood
is predominantly the result of increased availability of oxygen at term, or approximately one fourth to one third of the fetal
in the extrauterine environment, which greatly reduces EPO blood volume.78 Umbilical arteries constrict shortly after birth,
production and endogenous erythropoiesis.The continued fall in but the umbilical vein remains dilated, and blood flows in the
hemoglobin concentration over the next several weeks results direction of gravity. Infants held at or below the level of the
from (1) decreased red blood cell production, (2) a shortened placenta can receive half of the placental blood volume (30 to
red blood cell life span of the fetal/neonatal erythrocyte, and 50 mL) in 1 minute. Blood can also travel from the neonate into

Descargado para Mirko Flores Gonzalez (mfloresg8@upao.edu.pe) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en octubre 31,
2023. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
1110 SECTION XVIII — Developmental Hematopoiesis

65 25

60

55
20
50

45
40 15

HGB (g/dL)
HCT (%)

35
30

25 10

20

15
5
10

5
0 0
0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28
A Day of life B Day of life

65 25
60
55
20
50

45
40 15
35 HGB (g/dL)
HCT (%)

30

25 10

20

15
5
10

5
0 0
0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29
C Day of life D Day of life
Fig. 109.4 Hematocrit and hemoglobin reference ranges for term and preterm infants in the first month of life. (A) Hematocrit (HCT) over the
first 28 days of life for neonates born at 35 to 42 weeks’ gestation. (B) Blood hemoglobin (HGB) concentration over the first 28 days of life for
neonates born at 35 to 42 weeks’ gestation. (C) HCT over the first 28 days of life for neonates born at 29 to 34 weeks’ gestation. (D) Blood HGB
concentration over the first 28 days of life for neonates born at 29 to 34 weeks’ gestation. (Reprinted with permission from Henry E, Christensen
RD. Reference intervals in neonatal hematology. Clin Perinatol. 2015;42:483–497.)

Table 109.4 Mean Hemoglobin Values (± 1 SD; g/dL) in Low-Birth-Weight Infants.

Age (wk)
Birth Weight (g) 2 4 6 8 10
800–1000 16.0 (14.8–17.2) 10.0 (6.8–13.2) 8.7 (7.0–10.2) 8.0 (7.1–9.8) 8.0 (6.9–10.2)
1001–1200 16.4 (14.1–18.7) 12.8 (7.8–15.3) 10.5 (7.2–12.3) 9.1 (7.8–10.4) 8.5 (7.0–10.0)
1201–1400 16.2 (13.6–18.8) 13.4 (8.8–16.2) 10.9 (8.5–13.3) 9.9 (8.0–11.8) 9.8 (8.4–11.3)
1401–1500 15.6 (13.4–17.8) 11.7 (9.7–13.7) 10.5 (9.1–11.9) 9.8 (8.4–12.0) 9.9 (8.4–11.4)
1501–2000 15.6 (13.5–17.7) 11.0 (9.6–14.0) 9.6 (8.8–11.5) 9.8 (8.4–12.1) 10.1 (8.6–11.8)

Data from Williams ML, Shoot RJ, O’Neal PL, et al. Role of dietary iron and fat in vitamin E deficiency anemia of infancy. N Engl J Med. 1975;292:887.

the placenta if there is a significant gradient, resulting in 20 to 30 1 month of age, blood volumes in term infants average 73 to
mL of blood loss per minute.101 Studies evaluating the benefits of 77 mL/kg.
delayed cord clamping or cord milking showed improvements
in blood volume, hematocrit, decreased transfusions, decreased NUCLEATED RED BLOOD CELLS
intraventricular hemorrhage (IVH), decreased late onset sepsis, Though nucleated red blood cells (NRBCs) are rarely observed
and improved iron stores.102 in healthy adults and children, they are often observed in term
The blood volume of infants with early umbilical cord and preterm neonates.103–105 Multiple studies have hypothesized
clamping averages 72 mL/kg, while the blood volume of that high concentrations of NRBCs at birth (or within the first
infants with delayed umbilical cord clamping averages 93 hours to days of life) are a marker of in utero hypoxia. Indeed,
mL/kg. Preterm infants have slightly larger blood volumes a high concentration of NRBCs at birth has been associated
(89 to 105 mL/kg), owing to an increased plasma volume. By with an increased risk of developing IVH and/or retinopathy of

Descargado para Mirko Flores Gonzalez (mfloresg8@upao.edu.pe) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en octubre 31,
2023. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
CHAPTER 109 — Developmental Erythropoiesis 1111

140 120
MCV (fL)
120 100

100

NRBC/100 WBC
80
80
60
60
MCH (pg)

40
40
20
20

A 0
0
22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42
10,000
Gestational age (wk)
Fig. 109.5 Mean corpuscular volume (MCV) and mean corpuscular
hemoglobin reference ranges from 22 to 42 weeks. Reference ranges 8,000
are shown for the (MCV; upper set of lines, measured in femtoliters)
and the mean corpuscular hemoglobin (MCH) concentration (lower
6,000

NRBC/µL
set of lines, measured in picograms) at 22 to 42 weeks’ gestation.
Tests were done during a 6.5-year period on more than 20,000 neo-
nates. 4,000

prematurity.106–108 Placental abruption, intraamniotic infection,


and intrauterine growth restriction have also been associated 2,000
with significantly higher NRBCs at birth.108 NRBCs are measured
as either NRBC/μL or NRBC per 100 white blood cells (WBC;
NRBC/100 WBC). The normative patterns of NRBCs are similar, 0
regardless of units used. The number of NRBCs on the day of 23 31 33 35 37 39 41
25 27 29
birth decreases from a median of ∼25 NBRC/100 WBC for B Gestation (wk)
a neonate born at 23 to 28 weeks gestation to a median of 7 Fig. 109.6 Reference ranges for blood concentrations of nucleated
NRBC/100 WBC for a term neonate (Fig. 109.6).108 red blood cell (NRBC) on the day of birth by gestational age. The
lower and upper lines represent the 5th and 95th percentile limits.
RETICULOCYTE INDICES The middle line represents the mean value. (A) Data expressed as
Erythrocytes are released from the marrow into the blood NRBC/100 white blood cell (WBC). (B) Data expressed as NRBC/μL.
as reticulocytes. The cytoplasmic organelles (e.g., ribosomes, (From Christensen RD, Henry E, Andres RL, et al. Reference ranges
mitochondria, Golgi apparatus) persist for 1 to 2 days after for blood concentrations of nucleated red blood cells in neonates.
release before involution. Special staining detects elements of the Neonatology. 2011;99[4]:289–294.)
residual organelles and allows for a manual reticulocyte count.
Automated hematology counters that utilize flow cytometry have the first 3 days of life, and 14% have hematocrits over 70%.112
made more accurate reticulocyte enumeration possible because Widness and colleagues reported elevated EPO concentrations
much larger samples of cells are evaluated than in a manual count. in umbilical cord blood samples of neonates with trisomy 21,
These analyzers also make it possible to identify subpopulations speculating that hypoxemia in utero resulted in elevated EPO
of reticulocytes that appear to be clinically helpful. and consequent polycythemia. In addition, neonates with trisomy
The reticulocyte count reflects the rate of effective 21 have an elevated MCV in the first 3 days of life.112
erythropoiesis. Because EPO production and endogenous Less data is available on the hematologic abnormalities in
erythropoiesis falls dramatically after birth, the number of neonates with trisomy 18 and trisomy 13. Like neonates with
reticulocytes in circulation also falls. In an otherwise well trisomy 21, platelet and leukocyte abnormalities are more
neonate, the low number of reticulocytes in circulation persists pronounced. Wiedmeier and colleagues observed anemia in 40%
through the first 90 days of life until the physiologic nadir of and polycythemia in 17% of neonates with trisomy 18. Only 1
hemoglobin concentration is reached.109 of 22 neonates in their cohort with trisomy 13 had abnormal
The immature reticulocyte fraction (IRF) quantifies the erythrocyte findings.113
proportion of reticulocytes that are particularly high in RNA as a
ratio to total reticulocytes, and is an early and sensitive index of FETOMATERNAL RED BLOOD CELL TRANSFER
marrow erythropoietic activity. Because flow cytometric gating Maternal and fetal circulating cells may, at different times,
on automated hematology analyzers is not standardized, the IRF cross the placental barrier. Fetal contamination of the maternal
may vary among analyzer manufacturers for any given sample. circulation can occur before delivery, as evidenced by studies
of maternal blood group immunization. About 50% to 75% of
RED BLOOD CELL INDICES IN NEONATES WITH pregnancies are associated with some degree of fetomaternal
ANEUPLOIDY transfer of blood. This event is uncommon in the first trimester
Neonates with aneuploidy have consistent differences in their (3%). The volumes of fetal transplacental transfer are relatively
RBC indices. This phenomenon has been best described in small, usually on the order of 0.01 to 0.1 mL, but on occasion
neonates with trisomy 21.110,111 Polycythemia at birth is common they may be much greater. About one pregnancy in 400 is
among this population. Henry and colleagues estimated that 33% associated with fetal transplacental bleeding of 30 mL or greater,
of neonates with trisomy 21 have hematocrits over 65% during and about one pregnancy in 2000 is associated with a potential

Descargado para Mirko Flores Gonzalez (mfloresg8@upao.edu.pe) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en octubre 31,
2023. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
1112 SECTION XVIII — Developmental Hematopoiesis

fetal transplacental hemorrhage of 100 mL or more.114 The 5' 3'


overall risk of Rh immunization occurring in an Rh-incompatible Chromosome 11
pregnancy is 16% if the fetus is Rh positive and ABO compatible
with its mother. This risk decreases to 1.5% if the fetus is Rh LCR ε Gγ Aγ ψβ1 δ β
positive and ABO incompatible. Fetal transfer of cells to the
mother occurs during abortions as well (about a 2% incidence ψα2 α2 θ1
of such a transfer with spontaneous abortion and a 4% to 5% Chromosome 16
rate if abortion is induced).115 Because fetal hemoglobin is
resistant to acid elution, cells containing fetal hemoglobin ζ2 ψζ1 ψα1 α1
can be distinguished from cells containing hemoglobin A. The
Kleihauer-Betke stain of peripheral maternal blood uses this
characteristic of fetal hemoglobin to detect fetal cells in the
maternal circulation,116 although results from mothers with 0 10 20 30 40 50 60 70
increased fetal hemoglobin synthesis (i.e., sickle cell disease, kb ladder
thalassemia, and hereditary persistence of fetal hemoglobin) are
Embryonic: Hemoglobin Gower 1 ζ2ε2
not reliable. Diagnosis of fetomaternal hemorrhage may also be Hemoglobin Gower 2 α2ε2
missed when the mother and infant are ABO incompatible. In Hemoglobin Portland I ζ2γ2
these cases, the fetal cells are rapidly cleared from the maternal
circulation by maternal anti-A or anti-B antibodies. Fetal: Hemoglobin F α2γ2

Adult: Hemoglobin A α2β2


ERYTHROCYTE BIOCHEMISTRY Hemoglobin A2 α2δ2

During fetal erythropoiesis, an orderly evolution of the Fig. 109.7 Organization of the globin genes. Transcription of mes-
production of different hemoglobins occurs. Eight globin senger RNA occurs from the 5′ to the 3′ end, and for both chromo-
genes direct the synthesis of six different polypeptide chains, somes the genes are arranged in order of their developmental ac-
designated α, β, γ, δ, ε, and ζ. These globin chains combine in tivation. The upper segment represents the β-like globin genes on
the developing erythroblast to form seven different hemoglobin the short arm of chromosome 11, and the lower segment represents
tetramers: hemoglobin Gower 1 (ζ2-ε2), hemoglobin Gower 2 (α2- the α-like genes on the distal part of the short arm of chromosome
ε2), hemoglobin Portland I (ζ2-γ2), hemoglobin Portland II (ζ2-β2), 16. Regions of the gene that code for primary globin proteins are
fetal hemoglobin (α2-γ2), hemoglobin A (α2-β2), and hemoglobin shown as shaded ovals, and regions that code for pseudogenes (ψ-
A2 (α2-δ2). nonexpressed remnants, which have a number of inactivating muta-
tions that prevent transcription and translation into functional globin
GLOBIN GENES protein) are shown as open ovals. θ1 is a globin-like gene without
The globin genes are organized into two clusters (Fig. 109.7).The inactivating mutations. The locus control region (LCR) is shown as
α-like genes are located along a 20-kb distal segment of the short a hatched segment and contains five DNase I hypersensitivity sites,
arm of chromosome 16. The cluster contains three functional represented by rectangles. A downstream hypersensitivity site known
genes (α1, α2, and ζ2), three pseudogenes (evolutionary remnants as 3′ hypersensitivity site 1 is shown at the 3′ end. The composition of
of genes that are not expressed because of inactivating mutations embryonic, fetal, and adult hemoglobins is listed.
that prevent production of a functional globin protein), and
one gene of undetermined function (a globin-like gene without Like the α1 and α2 genes, the Gγ and Aγ genes appear to be
inactivating mutations). The β-like gene cluster is located along a virtually identical over a span of 1.5 kb, suggesting a mechanism
60-kb segment of the short arm of chromosome 11 and contains for gene matching during evolution. Amino acid sequencing of
five functional genes (β, δ, Gγ, Aγ, and ε) and one pseudogene. normal umbilical cord blood has shown that either glycine (Gγ)
Within each complex, the genes are all in the same 5′ to 3′ or alanine (Aγ) is present at the 136 position in the γ chain.120
orientation and are arranged in the order in which they are Short-chain organic acids, such as butyrate and acetate, increase
expressed during development.117 γ-gene promoter activity. This activity can be enhanced even
The α genes are duplicated in humans. Each α gene further by incorporation of sections of the β-gene locus control
is approximately 4 kb long, interrupted by two small region.121,122
nonhomologous regions. The exons and first introns of the two The β-globin gene cluster occupies a region of approximately
α-globin genes have identical sequences, but the second intron 17 kb on the short arm of chromosome 11.117 Each of its
of α1 is nine bases longer and differs by three bases from that constituent genes, their flanking regions, and large stretches of
of the α2 gene. Despite the high degree of homology between the regions between them have been sequenced. Studies suggest
these two genes, the sequences diverge in the 3′ untranslated that the transcription factor NF-E1 regulates increased β-globin
regions, 13 bases beyond the TAA stop codon. Because of these gene expression during erythroid maturation. Deletion of the
sequence differences, the relative output of the two genes can be NF-E1 binding site in the upstream segment of the β-globin
measured: α2 mRNA is produced 2 to 3 times more abundantly promoter blocks induction of transcription in murine leukemia
than α1 mRNA in fetal liver and marrow and in fetal and adult cells.123
erythrocytes.117 δ-Globin gene expression occurs early at the erythroid
The ζ gene appears critical to normal fetal development. progenitor stage. By the reticulocyte stage, no δ-globin mRNA
In a murine model, both α-gene and ζ-gene expression can be synthesis can be detected.The δ-globin gene promoter functions
measured from the onset of erythropoiesis in the yolk sac.118 In at a much lower level than the β-globin gene promoter, resulting
addition, expression of both genes occurs concomitantly within in significantly lower levels of δ-globin mRNA production. In
the same cells. Expression of the ζ gene occurs predominantly addition, decreased expression occurs because of a globin
through the first 6 to 8 weeks of gestation, although minute promoter–specific silencer element located upstream of the
quantities of ζ-globin can be measured in fetal and neonatal red δ-globin gene.124 Moreover, δ-globin mRNA is less stable than
blood cells.119 β-globin mRNA, which likely accounts for the early cessation of

Descargado para Mirko Flores Gonzalez (mfloresg8@upao.edu.pe) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en octubre 31,
2023. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
CHAPTER 109 — Developmental Erythropoiesis 1113

60
Globin chain production (% of total) 100
HGB A: α2β2
50 α HGB F: α2γ2 97%

40 80
Gγ β 75%

% HGB
30 60
HGB Gower 1: ζ2ε2
20 40 HGB Portland I: ζ2γ2
ζ Aγ
HGB Gower 2: α2ε2 25% HGB A2: α2δ2
ε
10 20
δ 2%
1%
10 20 30 Birth 10 20 30 40 10 20 30 Birth 10 20 30 40

A Gestational age (wk) Weeks after birth B Gestational age (wk) Weeks after birth
Fig. 109.8 Changes in globin subunits (A) and hemoglobin tetramers (B) during human development from the embryo to early infancy. HGB,
Hemoglobin. (Data collated from Bunn HF, Forget BG. Hemoglobin: Molecular, Genetic and Clinical Aspects. Philadelphia: WB Saunders;
1986:68.)

δ-globin synthesis and significantly lower levels of hemoglobin A2 in normal adults. The relative rates of Gγ-chain and Aγ-chain
compared with hemoglobin A. production are also constant throughout fetal life at a Gγ/Aγ ratio
The ε gene is located 5′ of the other β-like genes, and it is of approximately 3:1.120 It is not known whether these pairs of
expressed during embryonic development. The ε gene is similar genes are initially activated at this ratio; however, this production
in sequence to the γ genes. A silencing element of the promoter ratio is reached early in development.
region may be responsible for inactivating gene expression after Somewhere between the 32nd week and the 36th week of
the embryonic stage.125 The only known mutation that results in gestation, the relative rate of β-chain synthesis increases and that
persistent expression of this globin gene occurs in trisomy 13, of γ-chain production declines such that at birth β-chain synthesis
in which there is a delay in the switch from ε-globin to γ-globin makes up 30% to 50% of non–α-chain synthesis. It is generally
production.126 held that the transition from fetal to adult erythropoiesis starts
at 30 to 36 weeks after conception, but the rate of the transition
GLOBIN CHAIN SYNTHESIS has been controversial. A gradual transition from fetal to adult
It has been possible to analyze the patterns of globin chain hemoglobin synthesis occurs, starting in the first trimester.128,129
production at early ages of embryonic development during Considerable variation among infants occurs, however, because
the transition from yolk sac (primitive) to hepatic (definitive) many infants show prolonged dependence on fetal hemoglobin.
erythropoiesis. It is unknown why primitive erythroid After birth the level of γ-chain production steadily declines and
progenitors programmed to produce one type of hemoglobin, that of β-chain production increases, so by the end of the first
such as hemoglobin Gower 1, give way to definitive progenitors year, γ-chain synthesis reaches the low level characteristic of adult
programmed to produce a different type of hemoglobin, such life. The normal range of postnatal fetal hemoglobin production
as fetal hemoglobin. Quantification of globin gene synthesis can be seen in Fig. 109.8. During the first few months of life, the
reflects production by numerous red blood cells, and production Gγ/Aγ ratio changes from 3:1 to 2:3, although this ratio is variable

of a specific hemoglobin is usually reported as a percentage of in adults.130,131


the total hemoglobin measured. Studies evaluating hemoglobin Production of the δ chain has been observed as early as 32
production by erythroid colonies in culture, however, show that weeks. Activation of the δ gene lags β-gene activation, so the
individual cells in a colony produce predominantly one type of adult β/δ synthesis ratio is not reached until 4 to 6 months after
hemoglobin.127 birth.
During the fourth to fifth week, ζ, δ, and ε chains are the
main globin chains synthesized (Fig. 109.8). During the sixth HEMOGLOBIN PRODUCTION
to seventh week, α, δ, ε, Gγ, and Aγ chains are produced in the Developmental changes in the production of the various
remaining primitive erythroblasts, and α, ε, Gγ, and Aγ chains are hemoglobins are noted in Fig. 109.8. Before the onset of formation
produced in definitive erythrocytes. By the seventh to eighth of other chains, unpaired globin chains may form tetramers,
week, ε- and ζ-chain synthesis is no longer detectable, and the resulting in the presence of ε4.132 Almost immediately thereafter,
main globin chains produced are α, Gγ, and Aγ. Production of α-chain and ζ-chain production begins, and hemoglobins Gower
the β chain is just barely detectable at this time and gradually 1 (ζ2-ε2), Gower 2 (α2-ε2), and Portland I (ζ2-γ2) are formed.133
increases, such that by 10 weeks, it makes up 10% of total By 5 to 6 weeks’ gestation, hemoglobins Gower 1 and Gower
non–α-chain production. As soon as β-chain production occurs, 2 constitute 42% and 24% of the total hemoglobin, respectively,
genetic disorders associated with β-chain synthetic or structural with fetal hemoglobin (α2-γ2) making up the remainder. By 14 to
abnormalities may be detected in utero, a finding suggesting an 16 weeks, hemoglobin F constitutes 50% of the total hemoglobin,
asynchronous transition from ζ-chain to α-chain production as and by 20 weeks, it forms more than 90% of the hemoglobin.131,134
compared with ε-chain to γ-chain production, with the ζ-α switch Small quantities of hemoglobin A (α2-β2) are found, beginning
occurring slightly earlier.128 at 6 to 8 weeks’ gestation. The increase in β-chain production
From the 10th week to about the 33rd week of gestation, the occurring between 12 and 20 weeks’ gestation accounts for the
main globin chains synthesized are α, Gγ, Aγ, and β. Assessment of sudden rise in the amount of hemoglobin A found at the end
the output of the two linked α-globin genes by mRNA analysis of the first trimester of pregnancy. Tetramers of γ chains (γ4, or
suggests that they are expressed in a ratio ranging from 1.5:1 hemoglobin Barts) and β-chains (β4, or hemoglobin H) can be
to 3.0:1 (α2/α1) throughout fetal life. This does not appear to found in conditions in which α-chain synthesis is impaired or
change during development and is the same as that observed absent, such as the α-thalassemia syndromes.

Descargado para Mirko Flores Gonzalez (mfloresg8@upao.edu.pe) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en octubre 31,
2023. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
1114 SECTION XVIII — Developmental Hematopoiesis

Fetal hemoglobin is easily distinguished immunologically


and biochemically from adult hemoglobin. The most significant
physiologic characteristic of fetal hemoglobin is the decreased 140
interaction with 2,3-diphosphoglycerate (2,3-DPG). 2,3-DPG
binds to deoxyhemoglobin in a cavity between the β chains and 120
stabilizes the deoxy form of hemoglobin, resulting in a reduced
hemoglobin-oxygen affinity. 2,3-DPG binds less effectively 100
to the γ-globin chains, because of the differing amino acid
sequence in the non-α chain. Consequently, 2,3-DPG does not

HbF (g/L)
80
reduce the oxygen affinity of hemoglobin F as much as that of
hemoglobin A. 60
Other differences in physical properties exist between fetal
and adult hemoglobin. Hemoglobin F is more soluble in strong
phosphate buffers than hemoglobin A.116 Hemoglobin F is 40
oxidized to methemoglobin more easily than is hemoglobin A,
and it has a considerably greater affinity for oxygen than does 20
adult hemoglobin as a result of differences in binding to 2,3-
DPG. Fetal hemoglobin is resistant to acid elution, which allows 0
differentiation of cells containing fetal hemoglobin from cells
containing hemoglobin A.116 35 40 45 50 55 60 65 70
The total γ chains in the blood of the fetus and newborn Postconceptional age (wk)
comprise 70% to 80% of Gγ chains. This fraction falls to about Fig. 109.9 Correlation between postconceptional age and absolute
40% by 5 months of age. This unique difference in Gγ chain levels of fetal hemoglobin. Postnatal fetal hemoglobin concentrations
production found in the fetus helps to distinguish fetal (HbF) are shown for 280 infants, examined at postnatal ages of 6
hematopoiesis from that found in later life. Under stress, the weeks (circles), 12 weeks (triangles), and 6 months (diamonds). (From
older infant and adult revert to this intrauterine form of fetal Berglund SK, Lindberg J, Westrup B, et al. Effects of iron supplements
hemoglobin structure. This often occurs in leukemic states and perinatal factors on fetal hemoglobin disappearance in LBW in-
in children and adults, and in other conditions as well.135,136 fants. Pediatr Res. 2014;76:477–482.)
The delay in the switch of hemoglobin F to hemoglobin A has
been noted in conditions of maternal hypoxia,137 in infants
small for their gestational age,138 and in infants of diabetic the appearance of predominantly adult hemoglobin–containing
mothers.139,140 Elevated levels of fetal hemoglobin may have cells during the second and third months of life. These findings,
protective effects in some disease states, and much research together with the results of analyses of the intercellular
has gone into identifying fetal to adult hemoglobin transition, distribution of fetal and adult hemoglobin by sensitive imm­
in order to “switch on” γ-globin gene expression and increase unologic methods, suggest, although they do not prove, that the
fetal hemoglobin production.141 The regulators implicated in transition from fetal to adult hemoglobin production occurs
hemoglobin F production include B-cell lymphoma/leukemia in the same erythrocyte population. This conclusion is also
11A, myeloblastosis protooncogene protein, and Krüppel-like consistent with the patterns of fetal and β-chain production in
factor 1. In addition, microRNAs 15a and 16-1 play a role in red blood cell colonies grown from neonatal blood.143
gene regulation.142 Studies show that the type of globin chains produced at
The postpartum decline of fetal hemoglobin production and different stages of development are not closely related to the site
of the intercellular distribution of fetal and adult hemoglobins of erythropoiesis. It appears that ζ and ε chains are synthesized in
has been extensively examined during the first few months of both primitive and definitive cell lines. Moreover, the switch from
life. Immediately after birth, there is a brief rise in hemoglobin F γ-chain to β-chain production occurs synchronously throughout
concentration, followed by a steady decline (Fig. 109.9). Studies of the liver and bone marrow during the later stages of fetal
the intercellular distribution of hemoglobin F, using the relatively development. The transition from γ-chain to β-chain synthesis is
insensitive acid-elution technique, have shown that during the most closely related to postconceptional age and not chronologic
first few months of life the distribution of hemoglobin F is quite age.135 Thus premature infants continue to synthesize relatively
heterogeneous. At 3 months the distribution of hemoglobin F large quantities of γ chains (and fetal hemoglobin) until 40
becomes bimodal, with populations of cells that contain acid- weeks’ gestation.
resistant hemoglobin F and populations of adult “ghost” cells.
These observations have suggested that fetal hemoglobin-
containing cells are replaced by a population of cells containing ERYTHROCYTE METABOLISM
adult hemoglobin.
Profound changes occur in the rates of red blood cell The mature red blood cell differs from other cells in the body in
production immediately before birth and during the first few that it has no nucleus; it consequently lacks the ability to engage
months after birth. On a body-weight basis, red blood cell in de novo protein synthesis. It has no mitochondria or ribosomes,
production during the latter months of gestation is significantly no nucleic acid or deoxyribonucleic acid synthesis, no Krebs
greater compared with that in adult life. Immediately after cycle of intermediary metabolism, and no electron transport
birth, erythropoiesis is considerably reduced, presumably as an system for oxidative phosphorylation. Thus cellular metabolism
adaptation to the extrauterine environment, and red blood cell is dependent on a limited supply of preexisting enzymes. These
production occurs at a low level for the first few weeks of life. enzymes, coenzymes, and the substrates of glucose metabolism
It is clear from globin-chain synthetic studies that there is a interact with hemoglobin and the red blood cell membrane to
steady and linear decline in γ-chain synthesis during the period perform all the primary functions of the red blood cell, the most
of reduced neonatal erythropoiesis. Newly synthesized red important being oxygen transport.
blood cells appearing in the circulation when erythropoiesis Other important functions of red blood cell metabolism
resumes contain predominantly adult hemoglobin. These include maintaining adequate amounts of energy in the form of
observations may explain the short plateau in the proportion adenosine triphosphate (ATP), producing reducing substances to
of fetal hemoglobin (but not absolute levels) after birth and act as antioxidants, and maintaining appropriate amounts of red

Descargado para Mirko Flores Gonzalez (mfloresg8@upao.edu.pe) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en octubre 31,
2023. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
CHAPTER 109 — Developmental Erythropoiesis 1115

Box 109.1 Metabolic Characteristics of Neonatal Erythrocytes

Carbohydrate Metabolism Accelerated decline of 2,3-diphosphoglycerate into red blood


Glucose consumption increased cell incubationa
Galactose more completely utilized as a substrate both under Increased ATP levels
normal circumstances and for methemoglobin reductiona Accelerated decline of ATP levels during brief incubation
Decreased activity of sorbitol pathwaya Storage Characteristics
Decreased triokinase activitya
Increased potassium efflux and greater degrees of hemolysis
Glycolytic Enzymes during short periods of storage
Increased activity of hexokinase, phosphoglucose isomerase,a More rapid assumption of altered morphologic forms on stor-
aldolase, glyceraldehyde 3-phosphate dehydrogenase,a phos- age or incubationa
phoglycerate kinase,a phosphoglycerate mutase, enolase,a Membrane
pyruvate kinase, lactate dehydrogenase, glucose 6-phosphate
dehydrogenase, 6-phosphogluconic dehydrogenase, galacto- Decreased ouabain-sensitive ATPasea
kinase, and galactose 1-phosphate uridyltransferase Decreased potassium influxa
Decreased activity of phosphofructokinasea Decreased permeability to glycerol and thioureaa
Distribution of hexokinase isoenzymes differs from that of adultsa Decreased membrane filterabilitya
Increased sphingomyelin content, decreased lecithin content
Nonglycolytic Enzymes of stromal phospholipids
Increased activity of glutamic oxaloacetic transaminase and Decreased content of linoleic acida
glutathione reductase Increase in content of lipid phosphorus and cholesterol per cell
Decreased activity of NADP-dependent methemoglobin Greater affinity for glucosea
reductase,a catalase,a glutathione peroxidase, carbonic Other
anhydrase,a adenylate kinase,a and glutathione synthetasea
Presence of α-glycerol-3-phosphate dehydrogenasea Increased methemoglobin contenta
Increased affinity of hemoglobin for oxygena
Adenosine Triphosphate and Phosphate Metabolism Glutathione instabilitya
Decreased phosphate uptake,a slower incorporation into ATP Increased tendency for Heinz body formation in the presence
and 2,3-diphosphoglyceratea of oxidant compoundsa

aAppears to be a unique characteristic of the newborn’s erythrocytes and not merely a function of the presence of young red blood cells.
From Oski FA, Naiman JL. Hematologic Problems in the Newborn. 3rd ed. Philadelphia: WB Saunders; 1982:107.
ATP, Adenosine triphosphate; NADP, nicotinamide adenine dinucleotide phosphate.

blood cell 2,3-DPG to assist in the modulation of hemoglobin’s less than 1% of glucose is metabolized to glycogen within the red
oxygen affinity. Energy metabolism is largely a function of the blood cell.145
Embden-Meyerhof pathway. This pathway also regulates the
quantity of red blood cell 2,3-DPG within the cell. The pentose EMBDEN-MEYERHOF PATHWAY
phosphate pathway, among other functions, has a vital role in the At least 90% of glucose is metabolized via the Embden-Meyerhof
production of reducing substances such as reduced nicotinamide pathway.146,147 Two moles of ATP are produced for every mole
adenine dinucleotide phosphate (NADPH). Box 109.1 lists the of glucose catabolized, yielding two moles of lactic acid. This
metabolic characteristics of neonatal erythrocytes. A few of the potential for the production of ATP is not fully achieved because
variations seen in comparison with adult red blood cells are approximately 20% of metabolized glucose traverses the 2,3-DPG
clinically significant in that they affect the life span of neonatal cycle, thus bypassing one of the kinase steps, which is a site of
red blood cells. ATP generation.148
The Embden-Meyerhof pathway has several unique
GLYCOLYSIS characteristics with respect to fetal and newborn cells. These
Red blood cells need a constant supply of carbohydrate to cells consume greater quantities of glucose than do the red
maintain adequate levels of ATP. Although glucose is the pre­ blood cells of adults.149 Galactose metabolism in the newborn
fer­red carbohydrate, the red blood cell metabolizes fructose or red blood cell also differs.150 Galactokinase activity is three
mannose almost as readily. Galactose is metabolized more slowly. times greater in the erythrocytes of newborns, and these cells
Intracellular glucose concentrations equilibrate immediately consume galactose more rapidly than do those of the adult.The
with changes in plasma glucose concentrations. Glucose enters glycolytic enzymes phosphoglycerate kinase and enolase are
the human erythrocyte by facilitated transfer, and it is either much more active in the cells of the fetus and newborn infant
converted to glucose 6-phosphate or reduced to its polyol than would be anticipated from their young cell age.151,152 In
derivative, sorbitol, which is then converted to fructose.144 Once contrast, the activity of phosphofructokinase (a rate-controlling
formed, glucose 6-phosphate is metabolized by one of three enzyme in glycolysis) is lower than normal in the erythrocytes
pathways. The Embden-Meyerhof pathway converts glucose from newborn infants.151,153,154 Developmental changes in
6-phosphate to lactate or pyruvate and in the process generates the activities of these three enzymes towards normal adult
ATP. Metabolism by way of the pentose phosphate pathway values during the first year of life appear to be independent
produces reduced intermediates and a phosphorylated pentose of red blood cell age and reflect a transition from fetal to adult
sugar (ribulose 5-phosphate).This sugar ultimately returns to the erythropoiesis. The decreased phosphofructokinase activity of
Embden-Meyerhof pathway. Finally, glucose 6-phosphate may be fetal cells may be a consequence of accelerated decay of an
converted to glucose 1-phosphate and then to glycogen, although unstable enzyme.The relative deficiency of this enzyme appears

Descargado para Mirko Flores Gonzalez (mfloresg8@upao.edu.pe) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en octubre 31,
2023. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
1116 SECTION XVIII — Developmental Hematopoiesis

to result in alterations in glucose metabolism, and it could be methemoglobin reduction, the reduction of glutathione, and the
functionally significant. Several of the other enzymes of the stabilization of certain enzymes.160 NADPH serves as a hydrogen
Embden-Meyerhof pathway have shown differences in the donor in the presence of the enzyme glutathione reductase,
staining intensity of certain isoenzyme zones as compared with resulting in reduction of glutathione. Reduced glutathione
adult controls, although the significance of this observation is ultimately serves as a substrate for the enzyme glutathione
not known. peroxidase, which is responsible for the detoxification of
The ATP generated via the Embden-Meyerhof pathway hydrogen peroxide. Hydrogen peroxide is a byproduct of the
is necessary for the maintenance of the normal biconcave conversion of oxyhemoglobin to methemoglobin, which is
shape of the erythrocyte.155 It is also necessary for pyrimidine a naturally occurring reaction inside the red blood cell in the
nucleotide synthesis, completion of purine nucleotide synthesis presence of oxidative stress. The absence of NADPH (or anyth­
and glutathione synthesis,156 incorporation of fatty acids into ing that interferes with the production of reduced glutathione),
membrane phospholipids,157 active cation transport,154 and the the synthesis of glutathione, or the inability to detoxify hydrogen
initial step in the phosphorylation of glucose by the enzyme peroxide severely impairs the viability of the red blood cell.
hexokinase. Not all these synthesizing functions are important This is the case in G6PD deficiency. Patients with this condition
in the mature red blood cell, which lacks a nucleus. However, can experience hemolytic crises when they experience oxidative
loss of red blood cell ATP produces a marked decrease in red stress from illness, certain foods, and certain drugs.
blood cell deformability. As the cell ages, ATP levels fall, and The pentose phosphate pathway in the newborn red blood
older cells have lower glucose utilization, greater osmotic cell differs from that in the adult red blood cell. Two enzymes
fragility on incubation, lower membrane lipid content, lower of the pentose pathway, G6PD and 6-phosphogluconic acid
potassium concentration, and greater sodium concentration. dehydrogenase, are active at levels higher than those seen in adult
They are less deformable and have a shorter life span. The red red blood cells.161 Carbon dioxide production by erythrocytes of
blood cells of term and preterm infants, when studied in the first term and preterm infants is equal to or greater than that seen in
several days of life, contain higher levels of ATP than do cells red blood cells of adults.
from adults.158,159 The small premature infant has even higher Although there are suggestions that the pentose phosphate
ATP levels.158 pathway activity in the newborn is normal, there is also
Newborn red blood cells seem to demonstrate a transient evidence that newborn infants are more susceptible to oxidant-
immaturity in their metabolism. This results in a slower uptake induced injury, leading to glutathione instability, Heinz body
of phosphorus, a delayed incorporation into 2,3-DPG, and a formation, and the development of methemoglobinemia.162,163
marked decline in 2,3-DPG levels, as well as ATP levels, during This oxidant vulnerability may be caused by factors unrelated
short periods of incubation in vitro. The precise reason for to the pentose phosphate pathway. For example, the red blood
this relatively transient immaturity is not known, but it may cell membrane in the fetus and newborn may have a decreased
explain why the newborn red blood cell loses potassium at an number of membrane sulfhydryl groups, making these cells
accelerated rate and undergoes marked morphologic alteration more susceptible than mature red blood cells to Heinz body
during short incubations in vitro. formation.163 Additionally, NADPH-methemoglobin reductase
Pyruvate kinase deficiency is a well-known enzymopathy that activity is decreased in neonatal red blood cells, and the plasma
affects the normal function of the Embden-Meyerhof pathway of newborn infants appears to have a diminished antioxidant
and results in clinically significant erythrocyte pathology. capacity.164 The precise mechanisms surrounding the newborn
Deleterious mutations in the PKLR (pyruvate kinase L/R) gene red blood cell vulnerability to oxidant injury are not known.
result in low levels of pyruvate kinase. This results in abnormal Fetal and newborn red blood cells have diminished levels
erythrocyte morphology (echinocytes, anisocytes, poikilocytes) of glutathione peroxidase, which may render the cells more
and a lifelong hemolytic anemia. The exact mechanisms of vulnerable to hydrogen peroxide–induced oxidant injury. In
hemolysis are not known. Though other Embden-Meyerhof addition, the newborn red blood cell may have a diminished
pathway enzymopathies have been described in the literature, capacity for handling other activated oxygen radicals, such
they are even more rare than pyruvate kinase deficiency. as singlet oxygen and the superoxide radical.165 The latter
is converted to hydrogen peroxide in the presence of the
PENTOSE PHOSPHATE PATHWAY enzyme superoxide dismutase. Superoxide dismutase levels
Glucose 6-phosphate undergoes oxidative decarboxylation differ widely among infants. Diminished activity of superoxide
through the pentose phosphate pathway, consuming oxygen dismutase could result in accumulation of superoxide radicals.
and producing carbon dioxide. The pentose pathway requires Free radicals are generally detoxified by antioxidants such as
NADPH as a cofactor. In the first step, oxidation of glucose α-tocopherol (vitamin E). However, if superoxide dismutase
6-phosphate to 6-phosphogluconolactone is catabolized levels are increased (as has been described in some infants), the
by glucose 6-phosphate dehydrogenase (G6PD), generating hydrogen peroxide presented to reduced glutathione may not
NADPH. This step is followed by enzymatic hydrolysis of be adequately detoxified.Therefore a delicate balance appears to
6-phosphogluconolactone to 6-phosphogluconate, which is exist between enzymes involved in production and detoxification
then oxidized (in the presence of 6-phosphogluconic dehy­ of free radicals and oxidative intermediates. The use of inhaled
drogenase) to ribulose 5-phosphate, with the production nitric oxide as treatment for pulmonary hypertension in sick
of carbon dioxide. Approximately 3% to 10% of all glucose neonates may affect this balance, and further studies are required
metabolized by the cell is cycled through the pentose phosphate to determine its impact on oxidant injury beyond an increase in
pathway. Hypoxia and acidosis increase the proportion of methemoglobin formation.
glucose metabolism shunted through this pathway.
The pentose phosphate shunt results in the production of 2,3-DIPHOSPHOGLYCERATE METABOLISM
two important products, ribose 5-phosphate and NADPH. Ribose The affinity of a hemoglobin solution for oxygen can be
5-phosphate is a vital constituent of the pyridine nucleotides decreased by interaction of hemoglobin with certain organic
nicotinamide adenine dinucleotide phosphate and NADPH phosphates.166 Among the organic phosphates tested, 2,3-DPG
and the purine nucleotides adenosine diphosphate and ATP. and ATP are the most effective in lowering oxygen affinity. The
There is no pathway for de novo purine formation inside the highly charged anion 2,3-DPG binds to deoxyhemoglobin but
red blood cell; however, the mature red blood cell does retain not to oxyhemoglobin. Various conditions increase the amount
the ability for pyridine nucleotide formation. NADPH is critical of 2,3-DPG present within the red blood cell, as regulated in
for preservation of cell integrity because it is necessary for the Embden-Meyerhof pathway. Once formed, each molecule

Descargado para Mirko Flores Gonzalez (mfloresg8@upao.edu.pe) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en octubre 31,
2023. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
CHAPTER 109 — Developmental Erythropoiesis 1117

of 2,3-DPG binds reversibly to one deoxyhemoglobin tetramer in much greater numbers in the peripheral blood of newborn
under physiologic conditions of solute concentration and pH. infants than in that of adults.175 Target cells, acanthocytes,
Fetal deoxyhemoglobin does not possess as great an affinity puckered immature erythrocytes, and other irregular projections
for 2,3-DPG as does adult deoxyhemoglobin, and therefore it may normally be found. For example, a greater percentage of
cannot bind 2,3-DPG to the same degree as adult hemoglobin. neonatal red blood cells have membrane surface pits, which are
Thus the fetal leftward-shifted hemoglobin-oxygen dissociation most likely the sites of formation of endocytic vacuoles. In normal
curve is not easily modulated by the presence of 2,3-DPG. The adults, 2.6% of erythrocytes appear to have surface pits or craters
half-saturation pressure (P50) of fetal blood is 19 to 21 mm ranging in size from 0.2 to 0.5 μm in diameter, demonstrated
Hg, some 6 to 8 mm Hg lower than that of adult blood. As the by interference-contrast microscopy.176 In contrast, pits can be
fetal hemoglobin concentration declines (see Fig. 109.9), the found in almost half of the erythrocytes of preterm infants and in
increasing percentage of adult hemoglobin and increasing red one fourth of the erythrocytes of term infants.
blood cell 2,3-DPG content produce a marked rightward shift in Red blood cell deformability is principally governed by three
the hemoglobin-oxygen equilibrium curve. Infants with a greater factors: the surface area to volume relationship of the red blood
proportion of adult hemoglobin but less 2,3-DPG may have the cell, the viscosity of the cytoplasm of the cell, and intrinsic red
same P50 as those with increased quantities of fetal hemoglobin blood cell membrane rigidity.174 The deformability of erythrocytes
but a high red blood cell 2,3-DPG content. is important for several reasons. First, red blood cell deformability
appears to be an important determinant of red blood cell life span
in vivo. The removal of a red blood cell from the circulation is
ERYTHROCYTE PHYSIOLOGY thought to be a consequence of declining deformability, making
the red blood cell susceptible to sequestration in the spleen and
PHYSICAL PROPERTIES OF NEONATAL other organs, where it must negotiate extraordinarily narrow
ERYTHROCYTES passages. Second, red blood cell deformability directly influences
The fetal and neonatal red blood cell differs from the mature blood flow in the peripheral circulation. Third, red blood cell
red blood cell of the older infant, child, and adult in various deformability affects whole blood viscosity, which, in turn, affects
ways. Specific characteristics of fetal red blood cells include a peripheral vascular resistance and cardiac workload.177
shortened life span, macrocytosis, high fetal hemoglobin content Neonatal red blood cells with the greatest density (representing
with a Gγ/Aγ ratio of 3:1, the presence of i antigen,167 and low the oldest cells in the circulation) lose more volume than adult
carbonic anhydrase enzyme activity.49 red blood cells, have a higher mean corpuscular hemoglobin
concentration than adult red blood cells, and are less deformable
LIFE SPAN than the oldest red blood cells seen in adults. This suggests an
The differences in physical properties of red blood cells accelerated decrease in deformability of aging red blood cells
derived from term and preterm infants may in part account for related to a more pronounced increase in the mean corpuscular
the decreased life span of neonatal red blood cells within the hemoglobin concentration, the principal determinant of the
circulation. The average life span for a neonatal red blood cell is internal viscosity of the red blood cell. Neonatal red blood cell
60 to 90 days,168 approximately half to two thirds that of an adult membranes deform more readily to a given shear force than do
red blood cell. When neonatal red blood cells are transfused into adult red blood cell membranes, resulting in greater susceptibility
adults, they exhibit a shortened life span, owing to alterations of neonatal cell membranes to yield and fragment.177 These
intrinsic to the neonatal red blood cell.169 In contrast, cells mechanical properties may lead to accelerated membrane loss
transfused from adult donors appear to survive normally in and a decreased life span. Recent studies evaluating the red blood
newborns.169 With increasing degrees of prematurity, remarkably cell deformability of preterm infants receiving recombinant EPO
shorter red blood cell life spans (35 to 50 days) are found. Fetal reported that erythrocyte deformability was significantly related
studies using [14C] cyanate-labeled red blood cells in sheep to the reticulocyte count, indicating that the improvement of
revealed an average red blood cell life span of 63.6 ± 5.8 days.170 erythrocyte deformability with recombinant EPO therapy was
The mean red blood cell life span increased linearly from 35 to due to the formation of well-deformable young erythrocytes.178
107 days as the fetal age increased from 97 days (midgestation)
to 136 days (term). SURFACE CHARGE
The shortened red blood cell life span of the preterm and The surface charge of newborn red blood cells is more negative
term neonate may be explained by some of the characteristics than that of adult red blood cells. The negative charge at the red
specific to newborn cells—namely, a rapid decline in intracellular blood cell surface is largely responsible for the electrophoretic
enzyme activity and ATP levels,171 loss of membrane surface mobility of the cell, and it appears to reflect the sialic acid
area by internalization of membrane lipids, decreased levels of content of the red blood cell membrane.179,180 Proteases expose
intracellular carnitine,172 increased susceptibility of membrane more negative sites on neonatal red blood cells than on adult
lipids and protein to peroxidation,173 and increased mechanical red blood cells and increase the electrophoretic mobility to a
fragility resulting from increased membrane deformability.174 greater degree.181 The sialic acid content of the newborn infant’s
erythrocyte membrane shows a gradual but significant decrease
SIZE in the first several weeks of life.182 Most studies, however, have
Red blood cell dimensions change markedly during fetal and shown that the electrophoretic mobility of the neonatal cell is
neonatal development. Early in embryogenesis, cell diameters similar to that of the adult cell.180 The more negative surface
range from 20 to 25 μm, and the MCV averages 150 to 180 fL.3 charge of the neonatal red blood cell membrane is one of the
During fetal development red blood cells gradually decrease in many characteristics that results in a decreased sedimentation
size and volume (see Fig. 109.5); at term the average cell is 8 to 10 rate in newborns.183
μm in diameter, with an MCV between 108 and 118 fL. During the
first year of life red blood cells continue to diminish in size, and OSMOTIC FRAGILITY
at 1 year of life they resemble adult red blood cells (Table 109.5). The osmotic fragility of red blood cells is a composite index of
their shape, hydration, and, within certain limitations, proneness
SHAPE AND DEFORMABILITY to in vivo destruction.174,184 Preterm and term infants have an
Just as there is variation in the size of newborn red blood cells, increased osmotic resistance.185The osmotic fragility of neonatal
there is variation in shape. Irregularly shaped cells are present cells begins to revert towards adult values shortly after birth, and

Descargado para Mirko Flores Gonzalez (mfloresg8@upao.edu.pe) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en octubre 31,
2023. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
1118 SECTION XVIII — Developmental Hematopoiesis

Table 109.5 Postnatal Changes in Red Blood Cell Indices in Term Infants.

Days Weeks Months


Birth
(Umbilical
RBC Indices Cord Blood) 1 3 1 2 4 2 3 4 6 9 12
Hemoglobin 16.5 18.5 18.6 17.5 16.6 13.9 11.2 (9.4) 11.5 12.2 12.6 12.7 12.7
(g/dL) (13.0) (14.5) (16.5) (13.5) (13.4) (10.7) (9.5) (10.3) (11.1) (11.4) (11.3)
Hematocrit 51 (42) 56 (45) 55 (42) 54 (41) 53 (33) 44 (28) 35 (29) 35 (32) 38 (31) 36 (32) 36 (33) 37
(%)
Red blood 4.7 (3.9) 5.3 (4.0) 5.6 (3.9) 5.1 (3.9) 4.9 (3.3) 4.3 (3.1) 3.7 (3.1) 3.8 (3.5) 4.3 (3.9) 4.7 (4.0) 4.7 (4.1) 4.7
cells
(×1012/L)
Mean 108 (98) 108 (95) 110 107 (88) 105 (88) 101 (91) 95 (84) 91 (74) 87 (76) 76 (68) 78 (70) 78 (71)
corpuscular (104)
volume (fL)
MCH (pg) 34 (31) 34 (31) 36.7 34 (28) 33.6 32.5 (29) 30.4 (27) 30 (25) 28.6 (25) 26.8 (24) 27.3 (25) 26.8 (24)
(30.0)
MCHC (g/dL) 33 (30) 33 (29) 33.1 33 (28) 31.4 31.8 31.8 33 (30) 32.7 35 (32.7) 34.9 34.3
(28.1) (28.1) (28.3) (28.8) (32.4) (32.1)

The values are the means (values in parentheses are −2 standard deviations).
MCH, Mean corpuscular hemoglobin; MCHC, mean corpuscular hemoglobin concentration; RBC, red blood cell.
Data from Saarinen UM, Siimes MA. Developmental changes in red blood cell counts and indices of infants after exclusion of iron deficiency by laboratory criteria and
continuous iron supplementation. J Pediatr. 1978;92(3):412–416; and Dallman PR. In: Rudolph A, ed. Pediatrics. 16th ed. New York: Appleton-Century-Crofts;
1977:1111.

osmotic resistance reaches adult values by 4 to 6 weeks of age. by a common control mechanism; therefore the presence of i
There is no known advantage to the increased osmotic antigen can serve as a marker of fetal hematopoiesis.
resistance seen in the neonatal red blood cell. However, studies Red blood cell antigens in the ABO, MN, Rh, Kell, Duffy, and
of osmotic fragility in neonates have practical implications. It Vel systems are well developed in early intrauterine life.194 They
has been suggested that the diagnostic criteria for hereditary are easily demonstrated in the fifth to seventh gestational weeks
spherocytosis used in adults and older children are unreliable and remain constant through the remainder of intrauterine
in newborn infants. For example, spherocytes are not regularly development. Other antigens, such as the Lutheran and Xg
seen in all infants with hereditary spherocytosis. Indeed, systems, develop more slowly but are present at birth. Lewis
spherocytes may be frequently found in ABO incompatibility. antigens are lacking in the newborn. By approximately 2 years of
When one is performing an osmotic fragility test, a neonatal age, the child’s red blood cell and plasma antigens have developed
osmotic fragility curve must be used rather than an adult a pattern that is seen throughout the remainder of life.78
curve.186 A relatively new diagnostic test for hereditary Although A and B antigens are present early in utero, A and B
spherocytosis, eosin-5-maleimide (EMA)-flow cytometry, isoagglutinin production occurs much later.195 By 30 to 34 weeks’
quantifies the erythrocyte membrane band 3 complex using gestation, however, about 50% of infants have some measurable
an eosin-5-maleimide dye flow cytometric analysis.187,188 A anti-A or anti-B antibodies.The fetal production of such antibodies
reduction in band 3 complex is consistent with hereditary is not related to maternal ABO blood type. Intrauterine exposure
spherocytosis. This test appears to perform very well in to gram-negative organisms, whose antigens are chemically related
neonates even in the first days of life.189,190 to those of blood groups A and B, is a potent stimulus for the
development of these antibodies. Isohemagglutinin antibodies
SURFACE RECEPTORS AND ANTIGENS ultimately are demonstrable in normal infants by 6 months of age
The neonatal red blood cell differs from the adult red blood cell and approach adult values at 2 years of age. Low to absent titers
in its ability to bind various substances. For example, the neonatal after this time are suggestive of immune deficiency.196
red blood cell binds more insulin than the adult red blood cell
because of the presence of greater numbers of insulin receptors OXYGEN TRANSPORT
per cell.191,192 Newborn red blood cells have approximately 2.5 At no other time of life are the mechanisms controlling oxygen
times the number of digoxin receptors in comparison with adult transport more complicated than in utero and during the immediate
red blood cells,193 and as a consequence they have erythrocyte- postpartum period. During prenatal life the fetal arterial oxygen
to-plasma digoxin ratios three times those of adults. This may tension (Po2) is approximately 30 mm Hg, and the venous Po2 is
explain the greater tolerance of newborns who are receiving approximately 15 mm Hg (Fig. 109.10). This low Po2 contributes
maintenance digoxin therapy. to the development of relative polycythemia in the fetus. After
Another unique characteristic of the fetal red blood cell is the birth, numerous factors affect oxygenation, including the inspired
manifestation of the i antigen on the cell surface (adult red blood gas mixture, pulmonary function, the arterial oxygen dissociation
cells express i antigen). Membrane i antigen is a carbohydrate curve, and the ability to extract oxygen at the tissue level.197,198
moiety located on protein membrane band 3, which, during It has been speculated that the actual amount of oxygen released
development, is converted from a linear polylactosamine to a to tissues may be greater in utero, given the characteristics of the
branched carbohydrate chain of N -acetyllactosamine units.167 hemoglobin-oxygen dissociation curve (Fig. 109.11).
Red blood cells bearing the i antigen are usually not detectable If pulmonary function is normal, there will be a rise in the Po2
by the first year of age. It has been suggested that the switch from of pulmonary blood in adults and neonates, from the 40 mm Hg of
fetal to adult hemoglobin and the transformation of i antigen pulmonary arterial blood to the 100 mm Hg of pulmonary venous
expression that occur during the first year of life are governed blood. Because of the shape of the hemoglobin-oxygen dissociation

Descargado para Mirko Flores Gonzalez (mfloresg8@upao.edu.pe) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en octubre 31,
2023. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
CHAPTER 109 — Developmental Erythropoiesis 1119

curve, these Po2 values permit 95% saturation of hemoglobin by shifted to the left, the affinity of hemoglobin for oxygen is increased.
oxygen. Further increases in Po2 produce little additional rise in Thus at any given Po2, less oxygen is released to the tissues.
saturation. In the normal adult (see Fig. 109.10), approximately Certain factors are known to alter hemoglobin’s affinity for
50% of hemoglobin will be saturated with oxygen when the Po2 oxygen (Box 109.2). The most important of these are the fetal
has fallen to 27 mm Hg (P50 = 27). In situations in which the hemoglobin concentration and the red blood cell 2,3-DPG
hemoglobin-oxygen dissociation curve has shifted to the right, the content. The level of red blood cell 2,3-DPG gradually increases
affinity of hemoglobin for oxygen is reduced. Thus at any given with gestation. By the end of the first postnatal week, the 2,3-DPG
Po2, more oxygen is released to tissues. Conversely, if the curve is levels are considerably higher than they are at birth (Table 109.6).
After the first week, red blood cell 2,3-DPG levels remain relatively
NORMAL TERM INFANTS
unchanged for the next 6 months. In term infants the hemoglobin-
oxygen dissociation curve gradually shifts to the right, and by 4
90 to 6 months of age, the P50 values approximate those of the adult.
1 2 3 4 5 6 7
The situation is somewhat different in preterm infants. Fetal
hemoglobin synthesis is still quite active; therefore, increases
80 in P50 seen in term infants as a result of the switch from fetal to
adult hemoglobin do not occur. The red blood cell 2,3-DPG
concentrations are slightly lower in preterm infants as well.96 These
70
concentrations can be increased with the use of EPO, thereby
shifting the hemoglobin-oxygen dissociation curve to the right.199
HbO2 saturation (%)

60 A precise relationship between the decrease in oxygen affinity of


a neonate’s blood and the progressive decline in the concentration
(1) Day 1 of fetal hemoglobin does not exist.198 Rather, changes in P50 reflect
50 (2) Day 5 the interplay between the levels of red blood cell 2,3-DPG, the
(3) 3 wk decline in fetal hemoglobin levels, and the subsequent increase in
(4) 6–9 wk hemoglobin A levels. A unifying concept has emerged consisting
40
(5) 3–4 mo of a “functioning 2,3-DPG fraction,”96 which determines how well
hemoglobin is able to release oxygen to tissues. The functioning
(6) 6 mo
30 2,3-DPG fraction (expressed as nanomoles per milliliter of red
(7) 8–11 mo blood cells) is calculated by multiplying the red blood cell 2,3-DPG
content by the percentage of adult hemoglobin93:
20
[2,3‐DPG ]functional = [2,3‐DPG ]observed × [100− % hemoglobin F]
10 
[109.1]
If P50 cannot be measured directly, it may be calculated from
the following equation86 after determining the functioning 2,3-
10 20 30 40 50 60 DPG fraction:
PO2 (mm Hg, pH 7.4) P50 = 18.4 + 0.0016[2,3‐DPG]functional [109.2]
Fig. 109.10 Oxygen equilibrium curve of blood from term infants
at different postnatal ages. The oxygen half-saturation pressure of Table 109.6 illustrates the expected changes in hemoglobin
hemoglobin (P50) on day 1 is 19.4 ± 1.8 mm Hg and has shifted to concentration, 2,3-DPG concentration, functional 2,3-DPG
30.3 ± 0.7 at age 11 months (normal adult 27.0 ± 1.1 mm Hg). (From fraction, and P50 during the first 3 months of life in low-birth-
Oski FA, Naiman JL. Hematologic Problems in the Newborn. 3rd ed. weight infants.96 In low-birth-weight infants, the amount of
Philadelphia: WB Saunders; 1982:250.) oxygen released by hemoglobin can be determined (on the basis

mm Hg
20 40 60 80 100
100 10.0
31 mmol/L

16 mmol/L

0.80 Fetal 8.0


45 mmol/L

0.60 Adult 6.0


mmol/L
ctO2
SO2

0.40 4.0
40 mmol/L

0.20 Adult 2.0


Adult
venous arterial
Fetal Fetal
venous arterial
0 0
4 8 12
PO2/kPa
Fig. 109.11 Differences in the hemoglobin-oxygen dissociation curve between the newborn and the adult. This figure demonstrates the effects
of these differences on oxygen tension (ctO2) and oxygen saturation (SO2). (From Stockman JA. Fetal Hematology. In: Eden RD, Boehm J, eds.
Assessment and Care of the Fetus. Norwalk, CT: Appleton & Lange; 1989:124.)

Descargado para Mirko Flores Gonzalez (mfloresg8@upao.edu.pe) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en octubre 31,
2023. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
1120 SECTION XVIII — Developmental Hematopoiesis

Fig. 109.13 illustrates the arterial Po2 below which shifts to the
Box 109.2 Factors Affecting right in the neonatal hemoglobin-oxygen dissociation curve are
Hemoglobin-Oxygen Affinity no longer advantageous. The gas tension at which this occurs is
known as the crossover Po2.201 The crossover Po2 is dependent
Increased Amount of Red Blood Cell 2,3-Diphospho- on how low the venous Po2 falls before oxygen delivery ceases.
glycerate, Increased P50 Wimberley202 calculated that if the arterial Po2 fell to less than 32
mm Hg, and if the venous Po2 fell to 10 mm Hg (a value found
Adaptation to high altitude
in the cerebral venous blood of some sick newborns), the infant
Hypoxemia associated with chronic pulmonary disease
would achieve better oxygen delivery with a fetal hemoglobin-
Hypoxemia associated with cyanotic heart disease
oxygen dissociation curve than with an adult curve. Conversely,
Anemia
if arterial Po2 can be maintained at a higher value, better oxygen
Secondary to iron deficiency
delivery would exist if the hemoglobin-oxygen dissociation
Secondary to chronic renal disease
curve were shifted to the right.
Caused by sickle cell anemia
In neonates with hypoxemia (e.g., infants with cyanotic
Decreased red blood cell mass
congenital heart disease), a shift to the right (a higher P50) in
Chronic liver disease
the hemoglobin-oxygen dissociation curve tends to lower the
Hyperthyroidism
arterial and venous oxygen saturation at any given Po2. It has
Red blood cell pyruvate kinase deficiency
been documented that if arterial hypoxemia results from right-
Decreased Amount of Red Blood Cell to-left shunting, shifts to the right in the hemoglobin-oxygen
2,3-Diphosphoglycerate, Decreased P50 dissociation curve always tend to increase the arterial-venous
oxygen difference and improve oxygen delivery.201
Septic shock
After intrauterine transfusion, infants have oxygen-unloading
Severe acidosis
properties characteristic of those of adult blood. Despite the
Following massive transfusions of stored blood
decrease in oxygen affinity that accompanies intrauterine
Neonatal respiratory distress syndrome
transfusion, no deleterious effects of this procedure with respect
Increased P50, No Consistent Alteration in Amount to oxygen uptake by the fetus have been documented.203 The
of Red Blood Cell 2,3-Diphosphoglycerate physiologic significance of manipulating the hemoglobin-oxygen
affinity of fetuses and extremely preterm infants remains to be
Abnormal hemoglobins (Kansas, Seattle, Hammersmith,
fully studied.
Tacoma, E)
Vigorous exercise

Decreased P50, No Consistent Alteration in Amount PHYSIOLOGIC ANEMIA IN NEONATES


of Red Blood Cell 2,3-Diphosphoglycerate AND THE ANEMIA OF PREMATURITY
Abnormal hemoglobins (Kempsey, Chesapeake, J Cape
A gradual decrease in hemoglobin concentration occurs during
Town, Yakima, Rainier)
the first 2 to 3 months of life in term and preterm infants. The
hemoglobin concentration remains stable during the next several
weeks and then slowly rises. In term infants the fall in red blood
cell production after birth is the result of improved oxygenation
of unloading from a normal arterial Po2 adjusted for age to an and occurs as a natural adaptation to extrauterine life. Because
arbitrary central venous Po2 of 40 mm Hg).93 Although oxygen- of this, the decrease in hemoglobin concentration has been
carrying capacity (hemoglobin concentration × percentage of termed a physiologic nadir rather than true anemia. However,
oxygen saturation × 1.36 mL oxygen per gram of hemoglobin) in preterm infants, adaptive mechanisms may not be complete.
decreases during the first few months of life as a consequence Preterm infants generally decrease hemoglobin concentration
of a decline in hemoglobin concentration, the amount of to values lower than those seen in term infants, and the nadir
oxygen capable of being delivered to tissues actually increases. varies with the degree of prematurity. Fig. 109.4 illustrates the
For example, a newborn weighing 1000 g with a hemoglobin decline in hemoglobin concentration in the first month of life on
concentration of 15 g/dL, a P50 of 19, and a central venous Po2 of the basis of birth weight and gestational age. Hemoglobin values
40 mm Hg will unload 1 mL of oxygen to tissues for every 100 as low as 7 g/dL are common in preterm infants who have not
mL of blood that passes through the capillary bed. At 10 weeks undergone phlebotomies.94,204
of age, the P50 has shifted to the right and is now 24 mm Hg.This Assessment of the factors that characterize oxygen supply and
same infant will now deliver 2.1 mL of oxygen per 100 mL of oxygen demand has provided insight into the adaptive changes
blood, even though the hemoglobin concentration has declined occurring in preterm infants in response to low hemoglobin
to 8 g/dL (see Fig. 109.11). concentrations. In preterm infants, a low central venous Po2
These calculations emphasize the importance of understanding correlates with a mild increase in EPO production (Fig. 109.14).205
an infant’s ability to deliver oxygen to tissues when one is The decline in central venous Po2 may indicate the presence of
determining whether to administer an erythrocyte transfusion. anemia, representing the integration of variables that determine
The decision to transfuse erythrocytes should not be based oxygen supply and demand: hemoglobin concentration, red
on hemoglobin concentration alone. Transfusions significantly blood cell and oxygen affinity, intravascular volume, oxygen
affect an infant’s endogenous erythropoiesis. For infants who consumption, heart rate, cardiac stroke volume, and arterial
undergo exchange transfusion or multiple transfusions, both EPO oxygenation.The hemoglobin concentration is only one of many
concentrations and reticulocyte counts are lower at any given important variables ensuring adequate oxygen delivery in both
hemoglobin concentration (Fig. 109.12).93,200 term and preterm infants.186
It is often assumed that oxygen delivery is decreased in
newborns because of the presence of high-affinity hemoglobin. ANEMIA OF PREMATURITY
In fact, a leftward shift in the hemoglobin-oxygen dissociation CHARACTERISTICS
curve resulting from high levels of fetal hemoglobin may better Despite the slight increase in EPO production that occurs
maintain oxygen delivery during episodes of severe hypoxemia. in response to declining central venous Po2 values, EPO

Descargado para Mirko Flores Gonzalez (mfloresg8@upao.edu.pe) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en octubre 31,
2023. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
CHAPTER 109 — Developmental Erythropoiesis 1121

Table 109.6 Changes in Hemoglobin Concentration, Hematocrit, and Other Markers of Oxygen Delivery During
the First 3 Months of Life in Low-Birth-Weight Infants.

Total Fetal
Hemoglobin O2 Capacity P50 at pH 7.40 2,3-Diphosphoglycerate Hemoglobin FFDPG
Age Blood (g/dL) Hematocrit (%) MCHC (%) Blood (mL/dL) (mm Hg) (nmol/mL) (% of Total) (nmol/mL)
Group I (<1000 g)a
2 wk 17.2 47.0 36.6 23.9 18.0 6255 83.0 1002
4 wk 8.5 26.0 32.7 11.8 15.0 3923 81.0 761
9 wk 7.2 22.0 32.7 10.0 15.0 4636 87.1 974
11 wk 7.7 22.5 34.2 10.7 17.0 5867 78.0 1290
Group II (1001–1500 g)b
1–2 days 15.1 ± 1.3 45.7 ± 3.7 33.0 ± 0.7 21.0 ± 1.8 18.0 ± 1.7 4124 ± 1562 86.6 ± 3.1 580 ± 287
5–8 days 13.4 ± 1.1 41.4 ± 3.2 33.5 ± 2.9 18.7 ± 1.5 18.9 ± 3.0 4501 ± 1919 84.4 ± 3.8 903 ± 689
2–3 wk 12.6 ± 3.1 33.6 ± 6.0 34.2 ± 1.1 15.9 ± 3.1 21.2 ± 1.9 5721 ± 1375 83.3 ± 5.1 1119 ± 557
4–5 wk 8.8 ± 0.9 25.3 ± 1.8 34.9 ± 1.7 12.3 ± 1.3 20.5 ± 1.7 6095 ± 2081 85.2 ± 2.3 931 ± 456
6–9 wk 9.1 ± 1.7 24.5 ± 5.8 35.1 ± 2.2 11.8 ± 2.4 23.4 ± 1.1 8734 ± 1834 77.2 ± 1.9 1995 ± 480
9–10 wkc 8.2 24.0 34.0 11.1 24.0 9000 77.0 2070
Group III (1501–2000 g)b
1–2 days 16.1 ± 0.9 47.8 ± 1.9 33.7 ± 1.9 22.4 ± 1.2 19.3 ± 0.9 4475 ± 1174 87.2 ± 3.6 703 ± 331
5–8 days 16.8 ± 3.3 48.5 ± 10.0 34.7 ± 0.5 25.3 ± 4.7 19.8 ± 1.3 5489 ± 1428 79.4 ± 5.0 1056 ± 590
2–3 wk 13.6 ± 3.0 40.4 ± 9.8 34.4 ± 1.5 18.8 ± 4.0 21.3 ± 1.8 6002 ± 998 80.6 ± 5.8 1184 ± 329
4–5 wk 11.2 ± 2.8 31.9 ± 9.9 35.5 ± 2.2 15.5 ± 3.8 20.8 ± 1.6 5841 ± 839 75.8 ± 7.8 1569 ± 577
6–9 wk 8.0 ± 0.7 22.1 ± 1.7 35.9 ± 0.7 11.1 ± 1.0 24.0 ± 0.9 7290 ± 634 67.5 ± 6.2 2457 ± 575
Group IV (2001–2500 g)b
1–2 days 15.9 ± 0.9 46.2 ± 5.8 35.8 ± 1.9 21.9 ± 1.5 20.2 ± 1.6 5306 ± 1075 76.8 ± 5.43 1258 ± 392
5–8 days 15.6 ± 1.7 47.0 ± 5.0 34.2 ± 1.1 21.5 ± 2.4 21.3 ± 3.3 6417 ± 1527 77.7 ± 6.3 1457 ± 603
2–3 wk 12.3 ± 1.1 35.1 ± 3.2 34.9 ± 0.5 17.1 ± 1.5 22.0 ± 1.3 7145 ± 1737 76.9 ± 4.7 1666 ± 472
6–9 wkc 14.0 44.0 34.0 19.5 25.5 7100 43.0 3212

aOnly one patient.


bValues are given as the mean ± the standard deviation.
cFewer than five infants.
FFDPG, Functioning fraction of 2,3-diphosphoglycerate; MCHC, mean corpuscular hemoglobin concentration; RBC, red blood cell.
From Delivoria-Papadopoulos M, Roncevic N, Oski FA. Postnatal changes in oxygen transport of term, preterm and sick infants: the role of red cell 2,3
diphosphoglycerate in adult hemoglobin. Pediatr Res. 1971;5:235.

concentrations in preterm infants are still significantly low, given are highly sensitive to EPO,214,215 and the concentrations of other
the degree of anemia (Fig. 109.15).76,111,205,206 This anemia, termed erythropoietic growth factors, including IL-3 and GM-CSF, appear
the anemia of prematurity, affects infants born at less than to be normal.216
approximately 32 weeks gestation, and it is the most common Infants born prematurely lack a normal response to anemia
anemia encountered in the neonatal period. It is a normocytic, and fail to increase EPO production despite an apparent need
normochromic anemia, generally associated with hemoglobin for improved tissue oxygenation.217 This occurs regardless of the
concentrations less than 10 g/dL and low reticulocyte counts. cause of anemia, whether it is the early anemia of phlebotomy
Some infants may be asymptomatic, whereas others demonstrate loss or the later anemia of prematurity. Possible molecular and
signs of anemia that are alleviated by transfusion. These signs cellular mechanisms responsible for this lack of responsiveness
traditionally include tachycardia, increased episodes of apnea and include defects in transferring the hypoxic signal to the nucleus,
bradycardia, poor weight gain, an increased oxygen requirement, diminished or defective binding of transcription factors to the
and elevated serum lactate concentrations that decrease after promoter or enhancer regions of the gene, decreased production
transfusion.207–210 or stability of the transcriptional factors, decreased production
Anemia of prematurity was first described by Schulman,211 or stability of EPO mRNA, diminished or unstable protein
who divided the anemia into three phases. Early anemia was production, or increased production of counterregulatory
marked by an initial fall in hemoglobin concentration. The second, proteins such as IL-1 and tumor necrosis factor. The inability of
or intermediate, phase was characterized by maintenance of the kidney or liver in the premature infant to produce EPO mRNA
low hemoglobin concentrations. The third phase, late anemia of with magnitude equal to that seen in the fetus may also involve
prematurity, resulted in hemoglobin concentrations that continued a developmental delay in expression of transcriptional factors
to fall, despite symptoms of anemia. The anemia of prematurity responsible for increasing EPO production, or in persistence of
usually resolved spontaneously by 3 to 6 months of life. fetal EPO gene methylation patterns. Because EPO produced by
Although much information has accumulated since the 1970s, the liver is indistinguishable from that produced by the kidney,
detailed mechanisms responsible for the anemia of prematurity it is unknown whether preterm infants rely on EPO produced
remain to be determined. Shortened erythrocyte survival,212 by the liver, the kidney, or a combination of the two. However,
hemodilution associated with a rapidly increasing body mass,213 macrophages from preterm infants generate EPO mRNA and
and the transition from fetal to adult hemoglobin96 have all been protein as effectively as do those from term infants and adults.218
implicated. Despite diminished levels of oxygen available to The anemia of prematurity likely involves some sort of delay
tissues93 and the appearance of signs of anemia,206 serum EPO in activation of EPO gene expression, from prenatal (liver) to
concentrations remain low.206 However, erythroid progenitors postnatal (kidney) sites of production.

Descargado para Mirko Flores Gonzalez (mfloresg8@upao.edu.pe) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en octubre 31,
2023. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
1122 SECTION XVIII — Developmental Hematopoiesis

PREMATURE BABIES (1000–1500 g)

Theoretical arteriovenous oxygen saturation difference (%)


100
20 P50 = 22 torr
1.0 P50 = 27 torr
1–2 days P50 = 32 torr
80
Oxygen content (m/100 mL blood)

1.2

15
1.6 60
5–8 days

6–9 wk 1.8
40
10 2–3 wk 2.1

20

5
0
0 20 40 60 80 100
9–10 wk Arterial PO2
Fig. 109.14 The effect of arterial partial pressure of oxygen (Po2)
on theoretical arteriovenous oxygen content difference when venous
10 20 30 40 50 60 70 150 partial pressure of oxygen is 40 mm Hg (bottom set of curves), 20
Oxygen tension (PO2 mm Hg) mm Hg (middle set of curves), or 10 mm Hg (top set of curves) at
various oxygen half-saturation pressures of hemoglobin (P50). (From
Fig. 109.12 Oxygen equilibrium curves of blood from premature in- Woodson RD. Physiological significance of oxygen dissociation curve
fants (1000 to 1500 g) at different postnatal ages. Double-headed ar- shifts. Crit Care Med. 1979;7:368.)
rows represent the oxygen unloading capacity between a given arterial
and venous Po2. Points corresponding to 150 mm Hg on the abscissa
are the oxygen capacities. Each curve represents the mean value of CLINICAL TRIALS
the infants studied in each group. (From Delivoria-Papadopoulos M, Multiple clinical studies have been performed that have
Roncevic N, Oski FA. Postnatal changes in oxygen transport of term, evaluate EPO administered to preterm infants.219 Randomized
preterm and sick infants: the role of red cell 2,3 diphosphoglycerate in controlled studies using EPO dosages greater than 500 U/kg/
adult hemoglobin. Pediatr Res. 1971;5:235.) week for at least 2 weeks220–233 have been reviewed elsewhere.
A randomized placebo-controlled study of darbepoetin and
EPO evaluated infants with a birth weight of 500 to 1250 g and
48 hours of age or younger who were randomized to receive
35
darbepoetin (10 μg/kg, once per week subcutaneously), EPO
(400 U/kg, three times per week subcutaneously), or placebo
30 (sham dosing) through 35 weeks’ gestation.234 Infants in the
darbepoetin and EPO groups received significantly fewer
25 transfusions (P = 0.015) and, importantly, were exposed to fewer
donors (P = 0.044) than the placebo group (darbepoetin, 1.2 ±
HbF >60%
2.4 transfusions and 0.7 ± 1.2 donors per infant; EPO, 1.2 ± 1.6
EP (mU/mL)

20 transfusions and 0.8 ± 1.0 donors per infant; placebo, 2.4 ± 2.9
transfusions and 1.2 ± 1.3 donors per infant). Morbidities were
15 similar among groups, including the incidence of retinopathy of
∆2.8 prematurity. In this population of preterm infants, darbepoetin
and EPO successfully serve as adjuncts to transfusions in
10 ∆3.0
maintaining red blood cell mass.
Recently published trials evaluating higher doses of EPO
HbF <30% ∆3.4
5 administered to extremely preterm infants evaluated potential
neuroprotective effects, with varied outcomes.235,236 It is clear
from the most recent trials that EPO administration to preterm
0
infants significantly decreases transfusions and improves
0 4 6 8 10 12 14 16 18
hematocrit during the initial neonatal intensive care unit
Hemoglobin (g/dL) (NICU) hospitalization. Further evaluation regarding potential
Fig. 109.13 Relationship between erythropoietin concentrations and neuroprotection is ongoing.
postnatal hemoglobin. In infants born prematurely who have under-
gone exchange transfusion or multiple transfusion (fetal hemoglobin PHARMACOKINETICS
[HbF] <30%), the hemoglobin concentration may fall several grams Pharmacokinetic studies in newborn monkeys and sheep
per deciliter before resumption of erythropoietin production (EP). indicate that neonates have a larger volume of distribution and
(From Stockman JA, Garcia JF, Oski FA. The anemia of prematuri- a faster elimination of EPO,237,238 necessitating the use of doses
ty: factors governing the erythropoietin response. New Engl J Med. higher than those required for adults.239 These pharmacokinetic
1977;296:647.) differences apply to preterm infants as well.240,241 Although

Descargado para Mirko Flores Gonzalez (mfloresg8@upao.edu.pe) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en octubre 31,
2023. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
CHAPTER 109 — Developmental Erythropoiesis 1123

42 neutropenia, was noted in early studies but has not been reported
in the last decade.245–247 The neutropenia was not associated
with depletion of either neutrophil reserves or colony-forming
unit–granulocyte-macrophage concentrations, but it appeared
to involve reduced production of neutrophils from granulocytic
progenitors248,249; it resolves after discontinuation of EPO
20
administration. In newborn mice, neutropenia occurs after
administration of very high doses of recombinant EPO as a result
of decreased production of neutrophils from progenitors.248
Overall, the side effects of EPO administration in preterm
infants have been minimal compared with those reported in
adults (hypertension, bone pain, rash, and, rarely, seizures). None
15 of these effects were reported in preterm infants involved in
clinical trials. Moreover, no studies have reported differences
EP mU/mL

between placebo and EPO recipients in the incidence of neonatal


morbidities such as chronic lung disease, IVH, necrotizing
enterocolitis, or late-onset sepsis. Meta-analyses first published in
2006 reported that, compared with late EPO administration, early
10 (first week of life) EPO administration increased the incidence
of retinopathy of prematurity (ROP) greater than stage 2.250,251
Revised meta-analyses corrected a previous misclassification of a
single-center EPO study into the late EPO administration group.
The association between retinopathy of prematurity and EPO
is no longer statistically significant.252,253 No differences were
5 seen in recently completed multicenter trials evaluating EPO
administered to over 2000 extremely preterm infants.235,236,254 In
fact, when those infants are included in updated meta-analyses,
the trend in greater than stage 2 ROP is greater in those infants
randomized to placebo or control groups. Close ophthalmologic
evaluation for retinopathy of prematurity continues to be a
0 priority in clinical trials in preterm infants. Although there
25 30 35 40 have been few long-term follow-up studies, it appears that the
administration of EPO has no adverse effect on developmental
PVO2 (torr)
outcome or growth, measured at 18 to 22 months.255
Fig. 109.15 Changes in plasma erythropoietin concentrations in re-
sponse to declines in central venous oxygen tension (PVO2) in pre- NUTRITIONAL SUPPLEMENTATION
term neonates. The arrow represents the position of 38 mm Hg, which Functional iron deficiency has frequently been reported in
for the purpose of this figure is the lower limit of normal for venous pediatric and adult patients receiving EPO, and it likely limited
oxygen tension. The horizontal dashed line is the upper limit of normal the success of some of the early EPO trials in preterm infants.
for erythropoietin when venous oxygen tension of 38 Torr or greater is Studies evaluating iron stores in EPO recipients220,221 noted
taken as normal. EP, Erythropoietin production. (From Stockman JA, increased iron requirements in infants receiving EPO, as
Graeber JE, Clark DA, et al. Anemia of prematurity: determinants of evidenced by diminished ferritin concentrations and elevated
the erythropoietin response. Pediatr. 1984;105:786.) numbers of hypochromic red blood cells, despite iron doses of 6
mg/kg/day. Infants receiving EPO are likely at greater risk for iron
deficiency than for iron overload and increased oxidant stress.
various EPO doses and dosing schedules have been evaluated However, further evaluation is still required to determine the
in preterm infants, the erythrokinetics of EPO (the interaction optimal dose and most effective route of administration of iron
between erythroid progenitors and their growth factors) in in preterm infants receiving EPO.
preterm infants are still being explored. Vitamin E is an antioxidant, inhibiting peroxidation of
The route of administration also influences the effectiveness polyunsaturated fatty acids in the lipid bilayers of all cell
of EPO. Rapid intravenous administration of EPO generates membranes.Vitamin E requirements may be increased secondary
peak serum concentrations that far exceed physiologic to increased iron supplementation, because iron promotes
concentrations.242 This may result in wasted drug via increased oxidation of polyunsaturated fatty acids. Pathak and colleagues228
renal excretion. Dosing strategies that achieve lower peak evaluated high-dose vitamin E in preterm infants receiving EPO
serum concentrations over a more prolonged period may be and found it showed no benefit compared with doses of 50
more effective. Dosages in the range of 500 to 1400 U/kg/week, IU/kg/day. Further study is needed to determine the optimal dose
with administration subcutaneously every day or every other of vitamin E in preterm infants receiving EPO.
day, result in an adequate erythropoietic response. Newer, long- Folate and vitamin B12 have recently been evaluated in
acting erythropoiesis-stimulating agents such as darbepoetin combination with EPO and iron, and have shown significant
are currently being studied.243,244 Data from preterm infants promise.232,256 In an evaluation of preterm infants weighing
(considering half-life, volume of distribution, and clearance) 800 g or less, Haiden and colleagues232 achieved significantly
suggest that higher doses will be required, but dosing intervals greater success (38% of infants did not receive transfusion) when
may be as great as once every 2 to 3 weeks. vitamin B12 at a dosage of 21 mg/kg/week subcutaneously was
added to a regimen of EPO, iron, vitamin E, and folate.239 When
SIDE EFFECTS OF ERYTHROPOIESIS-STIMULATING combined with limited phlebotomy losses, this therapy shows
AGENTS great promise in extremely low-birth-weight infants.
Side effects in neonates receiving erythropoiesis-stimulating Regardless of the treatment strategy, a critical understanding
agents have been rare. A side effect unique to neonates, transient of the developmental, physiologic, and pathologic influences

Descargado para Mirko Flores Gonzalez (mfloresg8@upao.edu.pe) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en octubre 31,
2023. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
1124 SECTION XVIII — Developmental Hematopoiesis

affecting oxygen delivery in term and preterm infants is important 96. Delivoria-Papadopoulos M, Roncevic NP, Oski FA. Postnatal changes in oxygen
transport of term, premature, and sick infants: the role of red cell 2,3-diphos-
in determining the optimal strategy in maintaining or altering phoglycerate and adult hemoglobin. Pediatr Res. 1971;5(6):235–245. https://
the hematocrit through the administration of an erythrocyte doi.org/10.1203/00006450-197106000-00001.
transfusion or the administration of an erythropoiesis-stimulating 101. Oh W, Lind J. Venous and capillary hematocrit in newborn infants and
agent. placental transfusion. Acta Paediatr. 1966;55(1):38–48. https://doi.
org/10.1111/j.1651-2227.1966.tb15207.x.
108. Christensen RD, Henry E, Andres RL, Bennett ST. Reference ranges for blood
concentrations of nucleated red blood cells in neonates. Neonatology.
CONCLUSION 2011;99(4):289–294. https://doi.org/10.1159/000320148.
111. Christensen RD, ed. Hematologic Problems of the Neonate. Philadelphia:
Saunders; 2000.
The fetal and neonatal periods mark a time when erythrocyte 120. Masala B, Manca L, Formato M, Pilo G. A study of the switch of fetal hemoglo-
indices and characteristics differ remarkably from indices of bin in newborn erythrocytes fractionated by density gradient. Hemoglobin.
children and adults. The stages of erythropoiesis move from 1983;7(6):567–572. https://doi.org/10.3109/03630268309027937.
yolk sack to liver to marrow, whereas production of erythrocyte 128. Peschle C, Mavilio F, Carè A, et al. Haemoglobin switching in human embryos:
asynchrony of ζ → α and ε → γ-globin switches in primitive and defini-
growth factors migrates from liver to marrow and kidney. Infants tive erythropoietic lineage. Nature. 1985;313(5999):235–238. https://doi.
born prematurely have altered production of erythrocytes; the org/10.1038/313235a0.
ability to deliver oxygen to tissues based on fetal and adult 135. Bard H, Lachance C, Widness JA, Gagnon C. The reactivation of fetal hemoglo-
hemoglobin concentrations varies greatly during the early bin synthesis during anemia of prematurity. Pediatr Res. 1994;36(2):253–256.
https://doi.org/10.1203/00006450-199408000-00018.
months of life in extremely low-birth-weight infants. A detailed 143. Peschle C, Condorelli M. Regulation of fetal and adult erythropoiesis. In: Porter
understanding of erythrocyte physiology and the nutritional and R, Fitzsimons DW, eds. Ciba Foundation Symposium 37 ‐ Congenital Disor-
growth factor needs of preterm and term infants will aid the ders of Erythropoiesis. Chichester, UK: John Wiley & Sons, Ltd.; 2008:25–47.
clinician in caring for neonates in the first weeks of life. https://doi.org/10.1002/9780470720196.ch3.
177. Linderkamp O, Stadler AA, Zilow EP. Blood viscosity and optimal hemato-
ACKNOWLEDGMENTS crit in preterm and full-term neonates in 50– to 500–μm tubes. Pediatr Res.
1992;32(1):97–102. https://doi.org/10.1203/00006450-199207000-00019.
Parts of this chapter were adapted from sections of Chapter 134 203. Novy MJ, Frigoletto FD, Easterday CL, Umansky I, Nelson NM. Changes in
by authors James A. Stockman III and Pedro A. DeAlarcon in the umbilical-cord blood oxygen affinity after intrauterine transfusions for eryth-
roblastosis. N Engl J Med. 1971;285(11):589–595. https://doi.org/10.1056/
first edition of this book. We thank Erin Adair for assistance with NEJM197109092851101.
illustrations. 204. Williams ML, Shoot RJ, O’Neal PL, Oski FA. Role of dietary iron and fat on
vitamin E deficiency anemia of infancy. N Engl J Med. 1975;292(17):887–890.
A complete reference list is available at www.ExpertConsult.com. https://doi.org/10.1056/NEJM197504242921704.
205. Stockman JA, Graeber JE, Clark DA, McClellan K, Garcia JF, Kavey RE. Ane-
mia of prematurity: determinants of the erythropoietin response. J Pediatr.
SELECT REFERENCES 1984;105(5):786–792. https://doi.org/10.1016/s0022-3476(84)80308-x.
206. Brown MS, Garcia JF, Phibbs RH, Dallman PR. Decreased response of plasma immu-
9. Moore MAS, Metcalf D. Ontogeny of the haemopoietic system: yolk sac origin of noreactive erythropoietin to “available oxygen” in anemia of prematurity. J Pedi-
in vivo and in vitro colony forming cells in the developing mouse embryo. Br atr. 1984;105(5):793–798. https://doi.org/10.1016/s0022-3476(84)80309-1.
J Haematol. 1970;18(3):279–296. https://doi.org/10.1111/j.1365-2141.1970. 208. Bifano EM, Smith F, Borer J. Relationship between determinants of oxygen deliv-
tb01443.x. ery and respiratory abnormalities in preterm infants with anemia. J Pediatr.
10. Yoder MC, Hiatt K. Engraftment of embryonic hematopoietic cells in con- 1992;120(2 Pt 1):292–296. https://doi.org/10.1016/s0022-3476(05)80447-0.
ditioned newborn recipients. Blood. 1997;89(6):2176–2183. https://doi. 209. Keyes WG, Donohue PK, Spivak JL, Jones MD, Oski FA. Assessing the need for
org/10.1182/blood.V89.6.2176. transfusion of premature infants and role of hematocrit, clinical signs, and
12. Forestier F, Daffos F, Catherine N, Renard M, Andreux JP. Developmental erythropoietin level. Pediatrics. 1989;84(3):412–417.
hematopoiesis in normal human fetal blood. Blood. 1991;77(11):2360–2363. 215. Rhondeau SM, Christensen RD, Ross MP, Rothstein G, Simmons MA. Responsive-
19. Spivak JL. The mechanism of action of erythropoietin. Int J Cell Cloning. ness to recombinant human erythropoietin of marrow erythroid progenitors
1986;4(3):139–166. https://doi.org/10.1002/stem.5530040302. from infants with the “anemia of prematurity. J Pediatr. 1988;112(6):935–940.
21. Jacobs K, Shoemaker C, Rudersdorf R, et al. Isolation and characteriza- https://doi.org/10.1016/s0022-3476(88)80223-3.
tion of genomic and cDNA clones of human erythropoietin. Nature. 216. Ohls RK, Liechty KW, Turner MC, Kimura R, Christensen RD. Erythroid “burst
1985;313(6005):806–810. https://doi.org/10.1038/313806a0. promoting” activity in serum of patients with the anemia of prematurity. J Pedi-
34. Zanjani ED, Pixley JS, Slotnick N, MacKintosh R, Ekhterae D, Clemons G. Eryth- atr. 1990;116(5):786–789. https://doi.org/10.1016/s0022-3476(05)82672-1.
ropoietin does not cross the placenta into the fetus. Pathobiology. 1993;61(3– 220. Maier RF, Obladen M, Scigalla P, et al. The effect of epoetin beta (recombinant
4):211–215. https://doi.org/10.1159/000163796. human erythropoietin) on the need for transfusion in very-low-birth-weight
40. Goldberg MA, Dunning SP, Bunn HF. Regulation of the erythropoietin gene: evi- infants. European Multicentre Erythropoietin Study Group. N Engl J Med.
dence that the oxygen sensor is a heme protein. Science. 1988;242(4884):1412– 1994;330(17):1173–1178. https://doi.org/10.1056/NEJM199404283301701.
1415. https://doi.org/10.1126/science.2849206. 222. Shannon KM, Keith JF, Mentzer WC, et al. Recombinant human erythropoietin
41. Peschle C, Marone G, Genovese A, Cillo C, Magli C, Condorelli M. Erythropoi- stimulates erythropoiesis and reduces erythrocyte transfusions in very low
etin production by the liver in fetal-neonatal life. Life Sci. 1975;17(8):1325– birth weight preterm infants. Pediatrics. 1995;95(1):1–8.
1330. https://doi.org/10.1016/0024-3205(75)90146-0. 223. Ohls RK, Osborne KA, Christensen RD. Efficacy and cost analysis of treating
44. Erslev AJ, Caro J, Kansu E, Silver R. Renal and extrarenal erythropoietin pro- very low birth weight infants with erythropoietin during their first two weeks
duction in anaemic rats. Br J Haematol. 1980;45(1):65–72. https://doi. of life: a randomized, placebo-controlled trial. J Pediatr. 1995;126(3):421–426.
org/10.1111/j.1365-2141.1980.tb03811.x. https://doi.org/10.1016/s0022-3476(95)70462-0.
55. Zanjani ED, Poster J, Burlington H, Mann LI,Wasserman LR. Liver as the primary site 225. Donato H, Vain N, Rendo P, et al. Effect of early versus late administration of
of erythropoietin formation in the fetus. J Lab Clin Med. 1977;89(3):640–644. human recombinant erythropoietin on transfusion requirements in premature
60. Ohls RK. Erythropoietin and hypoxia inducible factor-1 expression in the infants: results of a randomized, placebo-controlled, multicenter trial. Pediat-
midtrimester human fetus. Acta Paediatr. 2002;91(9):27–30. https://doi. rics. 2000;105(5):1066–1072. https://doi.org/10.1542/peds.105.5.1066.
org/10.1080/080352502320764166. 226. Ohls RK, Ehrenkranz RA, Wright LL, et al. Effects of early erythropoietin
76. Erslev AJ, Wilson J, Caro J. Erythropoietin titers in anemic, nonuremic patients. therapy on the transfusion requirements of preterm infants below 1250
J Lab Clin Med. 1987;109(4):429–433. grams birth weight: a multicenter, randomized, controlled trial. Pediatrics.
79. Christensen RD, Jopling J, Henry E, Wiedmeier SE. The erythrocyte indices 2001;108(4):934–942. https://doi.org/10.1542/peds.108.4.934.
of neonates, defined using data from over 12 000 patients in a multihospital 227. Maier RF, Obladen M, Müller-Hansen I, et al. Early treatment with erythropoi-
health care system. J Perinatol. 2008;28(1):24–28. https://doi.org/10.1038/ etin beta ameliorates anemia and reduces transfusion requirements in infants
sj.jp.7211852. with birth weights below 1000 g. J Pediatr. 2002;141(1):8–15. https://doi.
88. Song J, Sundar K, Gangaraju R, Prchal JT. Regulation of erythropoiesis after org/10.1067/mpd.2002.124309.
normoxic return from chronic sustained and intermittent hypoxia. J Appl 235. Juul SE, Comstock BA, Wadhawan R, et al. A randomized trial of erythropoietin
Physiol Bethesda Md. 2017;123(6):1671–1675. https://doi.org/10.1152/jap- for neuroprotection in preterm infants. N Engl J Med. 2020;382(3):233–243.
plphysiol.00119.2017. 1985. https://doi.org/10.1056/NEJMoa1907423.
93. Stockman JA, Garcia JF, Oski FA.The anemia of prematurity. Factors governing 239. Eschbach JW, Egrie JC, Downing MR, Browne JK, Adamson JW. Correction
the erythropoietin response. N Engl J Med. 1977;296(12):647–650. https://doi. of the anemia of end-stage renal disease with recombinant human eryth-
org/10.1056/NEJM197703242961202. ropoietin. Results of a combined phase I and II clinical trial. N Engl J Med.
1987;316(2):73–78. https://doi.org/10.1056/NEJM198701083160203.

Descargado para Mirko Flores Gonzalez (mfloresg8@upao.edu.pe) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en octubre 31,
2023. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
REFERENCES 29. Miharada K-I, Hiroyama T, Sudo K, Nagasawa T, Nakamura Y. Refinement of cyto-
kine use in the in vitro expansion of erythroid cells. Hum Cell. 2006;19(1):30–
1. Takashina T. Haemopoiesis in the human yolk sac. J Anat. 1987;151:125–135. 37. https://doi.org/10.1111/j.1749-0774.2005.00005.x.
2. Palis J. Primitive and definitive erythropoiesis in mammals. Front Physiol. 30. Verma R, Su S, McCrann DJ, et al. RHEX, a novel regulator of human ery-
2014;5:3. https://doi.org/10.3389/fphys.2014.00003. throid progenitor cell expansion and erythroblast development. J Exp Med.
3. Kelemen E, Calvo W, Fliedner TM. Atlas of Human Hemopoietic Develop- 2014;211(9):1715–1722. https://doi.org/10.1084/jem.20130624.
ment. Berlin: Springer-Verlag; 1979. 31. Emerson SG, Thomas S, Ferrara JL, Greenstein JL. Developmental regulation
4. Moore KL, Persaud TVN, Torchia MG. The Developing Human: Clinically Ori- of erythropoiesis by hematopoietic growth factors: analysis on popula-
ented Embryology. 10th ed. Philadelphia: Elsevier; 2016. tions of BFU-E from bone marrow, peripheral blood, and fetal liver. Blood.
5. Baron MH, Isern J, Fraser ST. The embryonic origins of erythropoiesis in mam- 1989;74(1):49–55.
mals. Blood. 2012;119(21):4828–4837. https://doi.org/10.1182/blood-2012- 32. Valtieri M, Gabbianelli M, Pelosi E, et al. Erythropoietin alone induces ery-
01-153486. throid burst formation by human embryonic but not adult BFU-E in unicellular
6. Wood WG. Haemoglobin synthesis during human fetal development. Br Med serum-free culture. Blood. 1989;74(1):460–470.
Bull. 1976;32(3):282–287. https://doi.org/10.1093/oxfordjournals.bmb. 33. Schibler KR, Li Y, Ohls RK, et al. Possible mechanisms accounting for the
a071376. growth factor independence of hematopoietic progenitors from umbilical
7. Djaldetti M. Hemopoietic events in human embryonic spleens at early gestational cord blood. Blood. 1994;84(11):3679–3684.
stages. Biol Neonate. 1979;36(3–4):133–144. https://doi.org/10.1159/000241218. 34. Zanjani ED, Pixley JS, Slotnick N, MacKintosh R, Ekhterae D, Clemons G. Eryth-
8. Calhoun DA, Li Y, Braylan RC, Christensen RD. Assessment of the contribu- ropoietin does not cross the placenta into the fetus. Pathobiology. 1993;61(3–
tion of the spleen to granulocytopoiesis and erythropoiesis of the mid- 4):211–215. https://doi.org/10.1159/000163796.
gestation human fetus. Early Hum Dev. 1996;46(3):217–227. https://doi. 35. Malek A, Sager R, Eckardt K-U, Bauer C, Schneider H. Lack of transport of eryth-
org/10.1016/0378-3782(96)01765-3. ropoietin across the human placenta as studied by an in vitro perfusion system.
9. Moore MAS, Metcalf D. Ontogeny of the haemopoietic system: yolk sac origin of Pflugers Arch Eur J Physiol. 1994;427(1–2):157–161. https://doi.org/10.1007/
in vivo and in vitro colony forming cells in the developing mouse embryo. Br BF00585955.
J Haematol. 1970;18(3):279–296. https://doi.org/10.1111/j.1365-2141.1970. 36. Widness JA, Sawyer ST, Schmidt RL, Chestnut DH. Lack of maternal to fetal trans-
tb01443.x. fer of 125I-labelled erythropoietin in sheep. J Dev Physiol. 1991;15(3):139–143.
10. Yoder MC, Hiatt K. Engraftment of embryonic hematopoietic cells in con- 37. Koury MJ, Bondurant MC, Graber SE, Sawyer ST. Erythropoietin messenger
ditioned newborn recipients. Blood. 1997;89(6):2176–2183. https://doi. RNA levels in developing mice and transfer of 125I-erythropoietin by the pla-
org/10.1182/blood.V89.6.2176. centa. J Clin Invest. 1988;82(1):154–159. https://doi.org/10.1172/JCI113564.
11. Ohls RK, Li Y, Abdel-Mageed A, Buchanan G, Mandell L, Christensen RD. Neu- 38. Jacobson LO, Marks EK, Gaston EO. Studies on erythropoiesis. XII. The effect
trophil pool sizes and granulocyte colony-stimulating factor production in of transfusion-induced polycythemia in the mother on the fetus. Blood.
human mid-trimester fetuses. Pediatr Res. 1995;37(6):806–811. https://doi. 1959;14(6):644–653.
org/10.1203/00006450-199506000-00022. 39. Matoth Y, Zaizov R. Regulation of erythropoiesis in the fetal rat. Isr J Med Sci.
12. Forestier F, Daffos F, Catherine N, Renard M, Andreux JP. Developmental 1971;7(7):839–845.
hematopoiesis in normal human fetal blood. Blood. 1991;77(11):2360–2363. 40. Goldberg MA, Dunning SP, Bunn HF. Regulation of the erythropoietin gene: evi-
13. Rosse C, Kraemer MJ, Dillon TL, McFarland R, Smith NJ. Bone marrow cell pop- dence that the oxygen sensor is a heme protein. Science. 1988;242(4884):1412–
ulations of normal infants; the predominance of lymphocytes. J Lab Clin Med. 1415. https://doi.org/10.1126/science.2849206.
1977;89(6):1225–1240. 41. Peschle C, Marone G, Genovese A, Cillo C, Magli C, Condorelli M. Erythropoi-
14. Carbonell F, Calvo W, Fliedner TM. Cellular composition of human fetal etin production by the liver in fetal-neonatal life. Life Sci. 1975;17(8):1325–
bone marrow. Cells Tissues Organs. 1982;113(4):371–375. https://doi. 1330. https://doi.org/10.1016/0024-3205(75)90146-0.
org/10.1159/000145570. 42. Imagawa S, Goldberg M, Doweiko J, Bunn H. Regulatory elements of the eryth-
15. Sieff CA, Emerson SG, Mufson A, Gesner TG, Nathan DG. Dependence of highly ropoietin gene. Blood. 1991;77(2):278–285. https://doi.org/10.1182/blood.
enriched human bone marrow progenitors on hemopoietic growth factors and V77.2.278.278.
their response to recombinant erythropoietin. J Clin Invest. 1986;77(1):74–81. 43. Fried W. The Liver as a source of extrarenal erythropoietin production. Blood.
https://doi.org/10.1172/JCI112305. 1972;40(5):671–677. https://doi.org/10.1182/blood.V40.5.671.671.
16. Lipton JM, Kudisch M, Nathan DG. Response of three classes of human ery- 44. Erslev AJ, Caro J, Kansu E, Silver R. Renal and extrarenal erythropoietin pro-
throid progenitors to the absence of erythropoietin in vitro as a measure of duction in anaemic rats. Br J Haematol. 1980;45(1):65–72. https://doi.
progenitor maturity. Exp Hematol. 1981;9(10):1035–1041. org/10.1111/j.1365-2141.1980.tb03811.x.
17. Golde DW,Takaku F, eds. Hematopoietic Stem Cells. New York: Dekker; 1985. 45. Naughton BA, Dornfest BS, Preti RA, Johnson R, Gordon AS. Hepatic and renal
18. Heath DS, Axelrad AA, McLeod DL, Shreeve MM. Separation of the erythro- erythropoietin production during various intervals of short-term hypoxia. J
poietin-responsive progenitors BFU-E and CFU-E in mouse bone marrow by Med. 1984;15(1):45–58.
unit gravity sedimentation. Blood. 1976;47(5):777–792. 46. Beru N, McDonald J, Lacombe C, Goldwasser E. Expression of the erythro-
19. Spivak JL. The mechanism of action of erythropoietin. Int J Cell Cloning. poietin gene. Mol Cell Biol. 1986;6(7):2571–2575. https://doi.org/10.1128/
1986;4(3):139–166. https://doi.org/10.1002/stem.5530040302. mcb.6.7.2571.
20. Dessypris E, Graber SE, Krantz SB, Stone WJ. Effects of recombinant erythropoi- 47. Zanjani ED, Banisadre M. Hormonal stimulation of erythropoietin production
etin on the concentration and cycling status of human marrow hematopoietic and erythropoiesis in anephric sheep fetuses. J Clin Invest. 1979;64(5):1181–
progenitor cells in vivo. Blood. 1988;72(6):2060–2062. 1187. https://doi.org/10.1172/JCI109571.
21. Jacobs K, Shoemaker C, Rudersdorf R, et al. Isolation and characteriza- 48. Udupa KB, Crabtree HM, Lipschitz DA. In vitro culture of proerythroblasts: charac-
tion of genomic and cDNA clones of human erythropoietin. Nature. terization of proliferative response to erythropoietin and steroids. Br J Haematol.
1985;313(6005):806–810. https://doi.org/10.1038/313806a0. 1986;62(4):705–714. https://doi.org/10.1111/j.1365-2141.1986.tb04094.x.
22. Schibler KR, Ohls RK, Le T, Liechty KW, Christensen RD. Effect of recombi- 49. Stamatoyannopoulos G, Nienhuis AW, eds. Cellular and Molecular Regulation
nant stem cell factor on clonogenic maturation and cycle status of human of Hemoglobin Switching. Grune & Stratton; 1979.
fetal hematopoietic progenitors. Pediatr Res. 1994;35(3):303–306. https://doi. 50. Datta MC. Prostaglandin E2 mediated effects on the synthesis of fetal and adult
org/10.1203/00006450-199403000-00004. hemoglobin in blood erythroid bursts. Prostaglandins. 1985;29(4):561–577.
23. Umemura T, Papayannopoulou T, Stamatoyannopoulos G. The mechanism of https://doi.org/10.1016/0090-6980(85)90080-2.
expansion of late erythroid progenitors during erythroid regeneration: target 51. Drake JR, Fitch CD. Status of vitamin E as an erythropoietic factor. Am J Clin
cells and effects of erythropoietin and interleukin-3. Blood. 1989;73(7):1993– Nutr. 1980;33(11):2386–2393. https://doi.org/10.1093/ajcn/33.11.2386.
1998. 52. Konwalinka G, Breier C, Peschel C, Geissler D, Lisch HJ, Braunsteiner H. Effect
24. Gardner JD, Liechty KW, Christensen RD. Effects of interleukin-6 on fetal of human plasmalipoproteins on erythropoietic progenitor cells in serum-free
hematopoietic progenitors. Blood. 1990;75(11):2150–2155. cultures. Blut. 1986;52(3):191–195. https://doi.org/10.1007/BF00320536.
25. Holbrook ST, Ohls RK, Schibler KR,Yang YC, Christensen RD. Effect of interleu- 53. Arnaud S, Blanchet JP. Mouse serum enables CFU-E to grow under physiologic
kin-9 on clonogenic maturation and cell-cycle status of fetal and adult hemato- concentration of erythropoietin in vitro. Exp Hematol. 1986;14(2):143–146.
poietic progenitors. Blood. 1991;77(10):2129–2134. 54. de Witte T, Holdrinet R, Raymakers R, Wessels J, Haanen C. Influence of
26. Gluzman-Poltorak Z, Vainstein V, Basile LA. Recombinant interleukin-12, but peripheral blood admixture on the number of hematopoietic progenitor
not granulocyte-colony stimulating factor, improves survival in lethally irradi- cells (CFU-GM and BFU-E) in human bone marrow aspirates. Acta Haematol.
ated nonhuman primates in the absence of supportive care: evidence for the 1985;74(2):70–74. https://doi.org/10.1159/000206172.
development of a frontline radiation medical countermeasure. Am J Hematol. 55. Zanjani ED, Poster J, Burlington H, Mann LI,Wasserman LR. Liver as the primary
2014;89(9):868–873. https://doi.org/10.1002/ajh.23770. site of erythropoietin formation in the fetus. J Lab Clin Med. 1977;89(3):640–
27. Kaushansky K, Broudy VC, Sitnicka E, Lofton-Day C, Grossman A, Sprugel K. Do 644.
the preclinical effects of thrombopoietin correlate with its in vitro properties? 56. Widness JA, Philipps AF, Clemons GK. Erythropoietin levels and erythropoiesis
Stem Cell. 1996;14(S1):108–111. https://doi.org/10.1002/stem.5530140713. at birth in infants with Potter syndrome. J Pediatr. 1990;117(1 Pt 1):155–158.
28. Miyagawa S, Kobayashi M, Konishi N, Sato T, Ueda K. Insulin and insulin-like https://doi.org/10.1016/s0022-3476(05)82459-x.
growth factor I support the proliferation of erythroid progenitor cells in bone 57. Lim G, Jeyaseelan K, Wintour E. Ontogeny of erythropoietin gene expression
marrow through the sharing of receptors. Br J Haematol. 2000;109(3):555– in the sheep fetus: effect of dexamethasone at 60 days of gestation. Blood.
562. https://doi.org/10.1046/j.1365-2141.2000.02047.x. 1994;84(2):460–466. https://doi.org/10.1182/blood.V84.2.460.460.

1124.e1
Descargado para Mirko Flores Gonzalez (mfloresg8@upao.edu.pe) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en octubre 31,
2023. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
1124.e2SECTION XVIII — Developmental Hematopoiesis

58. Nagai T,Yasuoka Y, Izumi Y, et al. Reevaluation of erythropoietin production by 86. Guest GM. Erythrocytes and hemoglobin of the blood in infancy and childhood:
the nephron. Biochem Biophys Res Commun. 2014;449(2):222–228. https:// III. Factors in variability, statistical studies. AMA J Dis Child. 1957;93(5):486.
doi.org/10.1016/j.bbrc.2014.05.014. https://doi.org/10.1001/archpedi.1957.02060040488002.
59. Dame C, Fahnenstich H, Freitag P, et al. Erythropoietin mRNA expression in 87. Mollison PL, Veall N, Cutbush M. Red cell and plasma volume in newborn
human fetal and neonatal tissue. Blood. 1998;92(9):3218–3225. infants. Arch Dis Child. 1950;25(123):242–253. https://doi.org/10.1136/
60. Ohls RK. Erythropoietin and hypoxia inducible factor-1 expression in the adc.25.123.242.
midtrimester human fetus. Acta Paediatr. 2002;91(9):27–30. https://doi. 88. Song J, Sundar K, Gangaraju R, Prchal JT. Regulation of erythropoiesis after
org/10.1080/080352502320764166. normoxic return from chronic sustained and intermittent hypoxia. J Appl
61. Naveh-Many T, Cedar H. Active gene sequences are undermethylated. Pro­c Physiol Bethesda Md 1985. 2017;123(6):1671–1675. https://doi.org/10.1152/
Natl Acad Sci. 1981;78(7):4246–4250. https://doi.org/10.1073/pnas.78.7.4246. japplphysiol.00119.2017.
62. Singal R, Ginder GD. DNA methylation. Blood. 1999;93(12):4059–4070. 89. Song J, Yoon D, Christensen RD, Horvathova M, Thiagarajan P, Prchal JT. HIF-
63. Yin H, Blanchard KL. DNA methylation represses the expression of the human mediated increased ROS from reduced mitophagy and decreased catalase
erythropoietin gene by two different mechanisms. Blood. 2000;95(1):111– causes neocytolysis. J Mol Med Berl Ger. 2015;93(8):857–866. https://doi.
119. org/10.1007/s00109-015-1294-y.
64. Wenger RH, Kvietikova I, Rolfs A, Camenisch G, Gassmann M. Oxygen-regu- 90. Rice L, Alfrey CP. The negative regulation of red cell mass by neocytolysis:
lated erythropoietin gene expression is dependent on a CpG methylation-free physiologic and pathophysiologic manifestations. Cell Physiol Biochem Int
hypoxia-inducible factor-1 DNA-binding site. Eur J Biochem. 1998;253(3):771– J Exp Cell Physiol Biochem Pharmacol. 2005;15(6):245–250. https://doi.
777. https://doi.org/10.1046/j.1432-1327.1998.2530771.x. org/10.1159/000087234.
65. Keohane AM, Lavender JS, O’Neill LP, Turner BM. Histone acetylation and 91. Moore LG, Newberry MA, Freeby GM, Crnic LS. Increased incidence of neonatal
X inactivation. Dev Genet. 1998;22(1):65–73. https://doi.org/10.1002/ hyperbilirubinemia at 3,100 m in Colorado. Am J Dis Child. 1984;138(2):157–
(SICI)1520-6408(1998)22:1<65::AID-DVG7>3.0.CO;2-5. 161. https://doi.org/10.1001/archpedi.1984.02140400039009.
66. Padilla MT, Dai A, Palmisano W, Ohls RK. Developmental regulation of erythro- 92. Stockman JA, Pochedly C, eds. Developmental and Neonatal Hematology.
poietin gene expression through methylation. J Investig Med. 2006;54(1):S160. New York: Raven Press; 1988.
4-S160. https://doi.org/10.2310/6650.2005.X0004.470. 93. Stockman JA, Garcia JF, Oski FA.The anemia of prematurity. Factors governing
67. Halvorsen S, Finne PH. Erythropoietin production in the human fetus and the erythropoietin response. N Engl J Med. 1977;296(12):647–650. https://doi.
newborn. Ann N Y Acad Sci. 1968;149(erythropoietin 1):576–577. https://doi. org/10.1056/NEJM197703242961202.
org/10.1111/j.1749-6632.1968.tb15199.x. 94. Stockman JA. RBC Values in low-birth-weight infants during the first seven
68. Meberg A. Haemoglobin Concentrations and erythropoietin levels in appropri- weeks of life. Arch Pediatr Adolesc Med. 1980;134(10):945. https://doi.
ate and small for gestational age infants. Scand J Haematol. 2009;24(2):162– org/10.1001/archpedi.1980.02130220023007.
168. https://doi.org/10.1111/j.1600-0609.1980.tb02362.x. 95. Grauel L, Müller S, Gross J, Syllm-Rapoport I. Separation and characterization
69. Thomas RM, Canning CE, Cotes PM, et al. Erythropoietin and cord blood hae- of red blood cells from newborns and infants during the first trimenon of
moglobin in the regulation of human fetal erythropoiesis. BJOG An Int J Obstet life using a dextran density gradient: mean cellular volume and mean cellular
Gynaecol. 1983;90(9):795–800. https://doi.org/10.1111/j.1471-0528.1983. haemoglobin concentration. Acta Haematol. 1982;67(2):102–108. https://doi.
tb09318.x. org/10.1159/000207034.
70. Widness JA, Clemons GK, Garcia JF, Oh W, Schwartz R. Increased immuno- 96. Delivoria-Papadopoulos M, Roncevic NP, Oski FA. Postnatal changes in oxygen
reactive erythropoietin in cord serum after labor. Am J Obstet Gynecol. transport of term, premature, and sick infants: the role of red cell 2,3-diphos-
1984;148(2):194–197. https://doi.org/10.1016/s0002-9378(84)80175-1. phoglycerate and adult hemoglobin. Pediatr Res. 1971;5(6):235–245. https://
71. Stevenson DK, Bucalo LR, Cohen RS, Vreman HJ, Ferguson JE, Schwartz doi.org/10.1203/00006450-197106000-00001.
HC. Increased immunoreactive erythropoietin in cord plasma and neona- 97. Humbert JR, Abelson H, Hathaway WE, Battaglia FC. Polycythemia in small
tal bilirubin production in normal term infants after labor. Obstet Gynecol. for gestational age infants. J Pediatr. 1969;75(5):812–819. https://doi.
1986;67(1):69–73. org/10.1016/S0022-3476(69)80304-5.
72. Hashimoto K, Clapp JF. The effect of nuchal cord on amniotic fluid and cord 98. Hakanson DO, Oh W. Hyperviscosity in the small-for-gestational age infant. Biol
blood erythropoietin at delivery. J Soc Gynecol Investig. 2003;10(7):406–411. Neonate. 1980;37(1–2):109–112. https://doi.org/10.1159/000241263.
https://doi.org/10.1016/s1071-5576(03)00139-4. 99. Bureau MA, Shapcott D, Berthiaume Y, et al. Maternal cigarette smoking and fetal
73. Teramo KA, Hiilesmaa VK, Schwartz R, Clemons GK, Widness JA. Amniotic oxygen transport: a study of P50, 2,3-diphosphoglycerate, total hemoglobin,
fluid and cord plasma erythropoietin levels in pregnancies complicated by hematocrit, and type F hemoglobin in fetal blood. Pediatrics. 1983;72(1):22–26.
preeclampsia, pregnancy-induced hypertension and chronic hypertension. J 100. Matoth Y, Zaizov R, Varsano I. Postnatal changes in some red cell
Perinat Med. 2004;32(3):240–247. https://doi.org/10.1515/JPM.2004.045. parameters. Acta Paediatr Scand. 1971;60(3):317–323. https://doi.
74. Finne PH. Erythropoietin levels in cord blood as an indicator of intra- org/10.1111/j.1651-2227.1971.tb06663.x.
uterine hypoxia. Acta Paediatr Scand. 1966;55(5):478–489. https://doi. 101. Oh W, Lind J. Venous and capillary hematocrit in newborn infants and placental
org/10.1111/j.1651-2227.1966.tb15239.x. ­transfusion. Acta Paediatr. 1966;55(1):38–48. https://doi.org/10.1111/
75. Widness JA, Teramo KA, Clemons GK, et al. Correlation of the interpretation j.1651-2227.1966.tb15207.x.
of fetal heart rate records with cord plasma erythropoietin levels. Br J Obstet 102. DuPont TL, Ohls RK. Placental transfusion: current practices and future direc-
Gynaecol. 1985;92(4):326–332. https://doi.org/10.1111/j.1471-0528.1985. tions. NeoReviews. 2018;19(1):e1–e10. https://doi.org/10.1542/neo.19-1-e1.
tb01104.x. 103. Perrone S, Vezzosi P, Longini M, et al. Nucleated red blood cell count in term
76. Erslev AJ, Wilson J, Caro J. Erythropoietin titers in anemic, nonuremic patients. and preterm newborns: reference values at birth. Arch Dis Child Fetal Neona-
J Lab Clin Med. 1987;109(4):429–433. tal Ed. 2005;90(2):F174–F175. https://doi.org/10.1136/adc.2004.051326.
77. Fisher JW. Erythropoietin: physiology and pharmacology update. Exp Biol 104. Baschat AA, Gungor S, Kush ML, Berg C, Gembruch U, Harman CR. Nucleated
Med. 2003;228(1):1–14. https://doi.org/10.1177/153537020322800101. red blood cell counts in the first week of life: a critical appraisal of relationships
78. Orkin SH, Nathan DG, Ginsburg D, Look AT, Fisher DE, Lux S. Nathan and with perinatal outcome in preterm growth-restricted neonates. Am J Obstet
Oski’s Hematology of Infancy and Childhood. Philadelphia: Saunders; Gynecol. 2007;197(3):286–e1-8. https://doi.org/10.1016/j.ajog.2007.06.020.
2008. 105. Rolfo A, Maconi M, Cardaropoli S, Biolcati M, Danise P, Todros T. Nucleated red
79. Christensen RD, Jopling J, Henry E, Wiedmeier SE. The erythrocyte indices blood cells in term fetuses: reference values using an automated analyzer. Neo-
of neonates, defined using data from over 12 000 patients in a multihospital natology. 2007;92(3):205–208. https://doi.org/10.1159/000102096.
health care system. J Perinatol. 2008;28(1):24–28. https://doi.org/10.1038/ 106. Green DW, Hendon B, Mimouni FB. Nucleated erythrocytes and intraventricu-
sj.jp.7211852. lar hemorrhage in preterm neonates. Pediatrics. 1995;96(3 Pt 1):475–478.
80. Usher R, Shephard M, Lind J. The blood volume of the newborn infant and 107. Buonocore G, Perrone S, Gioia D, et al. Nucleated red blood cell count at birth as
placental transfusion. Acta Paediatr. 1963;52(5):497–512. https://doi. an index of perinatal brain damage. Am J Obstet Gynecol. 1999;181(6):1500–
org/10.1111/j.1651-2227.1963.tb03809.x. 1505. https://doi.org/10.1016/s0002-9378(99)70396-0.
81. Oettinger L, Mills WB. Simultaneous capillary and venous hemoglobin deter- 108. Christensen RD, Henry E, Andres RL, Bennett ST. Reference ranges for blood
minations in the newborn infant. J Pediatr. 1949;35(3):362–365. https://doi. concentrations of nucleated red blood cells in neonates. Neonatology.
org/10.1016/s0022-3476(49)80008-4. 2011;99(4):289–294. https://doi.org/10.1159/000320148.
82. Bratteby LE. Studies on erythro-kinetics in infancy. X. Red cell volume 109. Christensen RD, Henry E, Bennett ST, Yaish HM. Reference intervals for reticu-
of newborn infants in relation to gestational age. Acta Paediatr Scand. locyte parameters of infants during their first 90 days after birth. J Perinatol.
1968;57(2):132–136. https://doi.org/10.1111/j.1651-2227.1968.tb04665.x. 2016;36(1):61–66. https://doi.org/10.1038/jp.2015.140.
83. Walker J, Turnbull EP. Haemoglobin and red cells in the human foetus and 110. Miller M, Cosgriff JM. Hematological abnormalities in newborn infants
their relation to the oxygen content of the blood in the vessels of the umbili- with Down syndrome. Am J Med Genet. 1983;16(2):173–177. https://doi.
cal cord. Lancet. 1953;265(6781):312–318. https://doi.org/10.1016/s0140- org/10.1002/ajmg.1320160207.
6736(53)91158-1. 111. Christensen RD, ed. Hematologic Problems of the Neonate. Philadelphia:
84. Burman D, Morris AF. Cord haemoglobin in low birthweight infants. Arch Dis Saunders; 2000.
Child. 1974;49(5):382–385. https://doi.org/10.1136/adc.49.5.382. 112. Henry E, Walker D, Wiedmeier SE, Christensen RD. Hematological abnormali-
85. Marks J, Gairdner D, Roscoe JD. Blood formation in infancy: part III. Cord ties during the first week of life among neonates with Down syndrome: data
blood. Arch Dis Child. 1955;30(150):117–120. https://doi.org/10.1136/ from a multihospital healthcare system. Am J Med Genet. 2007;143A(1):42–50.
adc.30.150.117. https://doi.org/10.1002/ajmg.a.31442.

Descargado para Mirko Flores Gonzalez (mfloresg8@upao.edu.pe) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en octubre 31,
2023. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
CHAPTER 109 — Developmental Erythropoiesis1124.e3

113. Wiedmeier SE, Henry E, Christensen RD. Hematological abnormalities during 139. Bard H, Prosmanne J. Relative rates of fetal hemoglobin and adult hemoglobin
the first week of life among neonates with trisomy 18 and trisomy 13: data synthesis in cord blood of infants of insulin-dependent diabetic mothers. Pedi-
from a multi-hospital healthcare system. Am J Med Genet. 2008;146A(3):312– atrics. 1985;75(6):1143–1147.
320. https://doi.org/10.1002/ajmg.a.32107. 140. Perrine SP, Greene MF, Faller DV. Delay in the fetal globin switch in infants
114. Scott JR,Warenski JC.Tests to detect and quantitate fetomaternal bleeding. Clin of diabetic mothers. N Engl J Med. 1985;312(6):334–338. https://doi.
Obstet Gynecol. 1982;25(2):277–282. https://doi.org/10.1097/00003081- org/10.1056/NEJM198502073120602.
198206000-00007. 141. Sankaran VG, Orkin SH. The switch from fetal to adult hemoglobin. Cold
115. Eden RD, Boehm FH, Haire M, eds. Assessment and Care of the Fetus: Spring Harb Perspect Med. 2013;3(1):a011643. https://doi.org/10.1101/csh-
Physiological, Clinical, and Medicolegal Principles. Norwalk, CN: Appleton perspect.a011643.
& Lange; 1990. 142. Ginder GD. Epigenetic regulation of fetal globin gene expression in adult
116. Kleihauer E, Braun H, Betke K. [Demonstration of fetal hemoglobin in erythro- erythroid cells. Transl Res J Lab Clin Med. 2015;165(1):115–125. https://doi.
cytes of a blood smear]. Klin Wochenschr. 1957;35(12):637–638. https://doi. org/10.1016/j.trsl.2014.05.002.
org/10.1007/BF01481043. 143. Peschle C, Condorelli M. Regulation of fetal and adult erythropoiesis. In: Porter
117. Wood WG, Weatherall DJ. Developmental genetics of the human haemoglo- R, Fitzsimons DW, eds. Ciba Foundation Symposium 37 ‐ Congenital Disor-
bins. Biochem J. 1983;215(1):1–10. https://doi.org/10.1042/bj2150001. ders of Erythropoiesis. Chichester, UK: John Wiley & Sons, Ltd.; 2008:25–47.
118. Leder A, Kuo A, Shen MM, Leder P. In situ hybridization reveals co-expression https://doi.org/10.1002/9780470720196.ch3.
of embryonic and adult alpha globin genes in the earliest murine erythrocyte 144. Travis SF, Morrison AD, Clements RS, Winegrad AI, Oski FA. Metabolic altera-
progenitors. Dev Camb Engl. 1992;116(4):1041–1049. tions in the human erythrocyte produced by increases in glucose concen-
119. Chui DH, Mentzer WC, Patterson M, et al. Human embryonic zeta-globin chains tration. J Clin Invest. 1971;50(10):2104–2112. https://doi.org/10.1172/
in fetal and newborn blood. Blood. 1989;74(4):1409–1414. JCI106704.
120. Masala B, Manca L, Formato M, Pilo G. A study of the switch of fetal hemoglo- 145. Moses SW, Chayoth R, Levin S, Lazarovitz E, Rubinstein D. Glucose and glycogen
bin in newborn erythrocytes fractionated by density gradient. Hemoglobin. metabolism in erythrocytes from normal and glycogen storage disease type
1983;7(6):567–572. https://doi.org/10.3109/03630268309027937. III subjects. J Clin Invest. 1968;47(6):1343–1348. https://doi.org/10.1172/
121. Safaya S, Ibrahim A, Rieder R. Augmentation of gamma-globin gene promoter JCI105826.
activity by carboxylic acids and components of the human beta-globin locus 146. Murphy JR. Erythrocyte metabolism. II. Glucose metabolism and pathways. J
control region. Blood. 1994;84(11):3929–3935. https://doi.org/10.1182/ Lab Clin Med. 1960;55:286–302.
blood.V84.11.3929.bloodjournal84113929. 147. Oski FA. Red cell metabolism in the premature infant. II. The pentose phos-
122. Stamatoyannopoulos G, Blau CA, Nakamoto B, et al. Fetal hemoglobin induction phate pathway. Pediatrics. 1967;39(5):689–695.
by acetate, a product of butyrate catabolism. Blood. 1994;84(9):3198–3204. 148. Brewer GJ, ed. Red Cell Metabolism and Function: Proceedings of the First
123. deBoer E, Antoniou M, Mignotte V, Wall L, Grosveld F. The human beta- International Conference on Red Cell Metabolism and Function. Vol. 6. Ann
globin promoter; nuclear protein factors and erythroid specific induc- Arbor: Held at the University of Michigan; 1969:1–3. October, Springer US;
tion of transcription. EMBO J. 1988;7(13):4203–4212. https://doi.org/10. 1970. https://doi.org/10.1007/978-1-4684-3195-7.
1002/j.1460-2075.1988.tb03317.x. 149. Oski FA, Naiman JL. Red cell metabolism in the premature infant. I. Adenosine
124. Vitale M, Dimarzo R, Calzolari R, et al. Evidence for a globin promoter-spe- triphosphate levels, adenosine triphosphate stability, and glucose consump-
cific silencer element located upstream of the human δ-globin gene. Bio- tion. Pediatrics. 1965;36:104–112.
chem Biophys Res Commun. 1994;204(1):413–418. https://doi.org/10.1006/ 150. Ng WG, Donnell GN, Bergren WR. Galactokinase activity in human erythro-
bbrc.1994.2474. cytes of individuals at different ages. J Lab Clin Med. 1965;66(1):115–121.
125. Cao SX, Gutman PD, Dave HP, Schechter AN. Identification of a transcriptional 151. Travis SF, Kumar SP, Paez PC, Delivoria-Papadopoulos M. Red cell metabolic
silencer in the 5’-flanking region of the human epsilon-globin gene. Proc Natl alterations in postnatal life in term infants. Glycolytic enzymes and glucose-
Acad Sci. 1989;86(14):5306–5309. https://doi.org/10.1073/pnas.86.14.5306. 6-phosphate dehydrogenase. Pediatr Res. 1980;14(12):1349–1352. https://doi.
126. Huehns ER, Hecht F, Keil JV, Motulsky AG. Developmental hemoglobin anoma- org/10.1203/00006450-198012000-00016.
lies in a chromosomal triplication: D1 trisomy syndrome. Proc Natl Acad Sci. 152. Konrad PN, Valentine WN, Paglia DE. Enzymatic activities and glutathione
1964;51(1):89–97. https://doi.org/10.1073/pnas.51.1.89. content of erythrocytes in the newborn: comparison with red cells of older
127. Papayannopoulou T, Kurachi S, Brice M, Nakamoto B, Stamatoyannopoulos normal subjects and those with comparable reticulocytosis. Acta Haematol.
G. Asynchronous synthesis of HbF and HbA during erythroblast maturation. 1972;48(4):193–201. https://doi.org/10.1159/000208458.
II. Studies of G gamma, A gamma, and beta chain synthesis in individual ery- 153. Gross RT, Schroeder EAR. The relationship of triphosphopyridine nucleotide
throid clones from neonatal and adult BFU-E cultures. Blood. 1981;57(3): content to abnormalities in the erythrocytes of premature infants. J Pediatr.
531–536. 1963;63(4):823–825. https://doi.org/10.1016/S0022-3476(63)80274-7.
128. Peschle C, Mavilio F, Carè A, et al. Haemoglobin switching in human embryos: 154. Whittam R. Transport and Diffusion in Red Blood Cells. Vol. 13. London:
asynchrony of ζ → α and ε → γ-globin switches in primitive and defini- Edward Arnold; 1964.
tive erythropoietic lineage. Nature. 1985;313(5999):235–238. https://doi. 155. Nakao M, Nakao T, Yamazoe S, Yoshikawa H. Adenosine triphosphate and
org/10.1038/313235a0. shape of erythrocytes. J Biochem (Tokyo). 1961;49(6):487–492. https://doi.
129. Phillips HM, Holland BM, Jones JG, Abdel-Moiz AL, Turner TL, Wardrop CAJ. org/10.1093/oxfordjournals.jbchem.a127333.
Definitive estimate of rate of hemoglobin switching: measurement of percent 156. Koj A. Biosynthesis of glutathione in human blood cells. Acta Biochim Pol.
hemoglobin F in neonatal reticulocytes. Pediatr Res. 1988;23(6):595–597. 1962;9:11–25.
https://doi.org/10.1203/00006450-198806000-00013. 157. Oliveira MM, Vaughan M. Incorporation of fatty acids into phospholipids of
130. Schroeder WA, Huisman THJ, Brown AK, et al. Postnatal changes in the chemi- erythrocyte membranes. J Lipid Res. 1964;5(2):156–162.
cal heterogeneity of human fetal hemoglobin. Pediatr Res. 1971;5(9):493. 158. Stave U, Cara J. Adenosinphosphate im Blut Frühgeborener. Neonatology.
https://doi.org/10.1203/00006450-197109000-00008. 1961;3(2–3):160–173. https://doi.org/10.1159/000239813.
131. Huisman THJ, Schroeder WA, Brown AK, Hyman CB, Ortega JA, Sukumaran PK. 159. De Luca C, Stevenson JH, Kaplan E. Simultaneous multiple-column chromatog-
Further studies of the postnatal change in chemical heterogeneity of human raphy: its application to the separation of the adenine nucleotides of human
fetal hemoglobin in several abnormal conditions. Pediatr Res. 1975;9(1):1–6. erythrocytes. Anal Biochem. 1962;4(1):39–45. https://doi.org/10.1016/0003-
https://doi.org/10.1203/00006450-197501000-00001. 2697(62)90017-9.
132. Szelengi JG, Hollán SR. Studies on the structure of human embryonic hemoglo- 160. Huennekens FM, Caffrey RW, Gabrio BW. The electron transport sequence of
bin. Acta Biochim Biophys. 1969;4(1):47–55. methemoglobin reductase. Ann N Y Acad Sci. 2006;75(1):167–174. https://doi.
133. Hecht F, Motulsky AG, Lemire RJ, Shepard TE. Predominance of hemoglobin org/10.1111/j.1749-6632.1958.tb36862.x.
Gower 1 in early human embryonic development. Science. 1966;152(3718):91– 161. Gross RT, Hurwitz RE. The pentose phosphate pathway in human erythro-
92. https://doi.org/10.1126/science.152.3718.91. cytes; relationship between the age of the subject and enzyme activity. Pedi-
134. Huehns ER, Dance N, Beaven GH, Hecht F, Motulsky AG. Human embryonic atrics. 1958;22(3):453–460.
hemoglobins. Cold Spring Harb Symp Quant Biol. 1964;29:327–331. https:// 162. Schröter W, Tillmann W. Heinz body susceptibility of red cells and
doi.org/10.1101/sqb.1964.029.01.035. exchange transfusion. Acta Haematol. 1973;49(2):74–79. https://doi.
135. Bard H, Lachance C, Widness JA, Gagnon C. The reactivation of fetal hemoglo- org/10.1159/000208387.
bin synthesis during anemia of prematurity. Pediatr Res. 1994;36(2):253–256. 163. Tillmann W, Menke J, Schroter W. The formation of Heinz bodies in ghosts
https://doi.org/10.1203/00006450-199408000-00018. of human erythrocytes of adults and newborn infants. Klin Wochenschr.
136. Bard H, Fouron JC, Gagnon C, Gagnon J. Hypoxemia and increased fetal hemo- 1973;51(4):201–203. https://doi.org/10.1007/BF01468345.
globin synthesis. J Pediatr. 1994;124(6):941–943. https://doi.org/10.1016/ 164. Stockman JA, Clark DA, Mcclellan K. Diminished antioxidant activity (aoa) of
s0022-3476(05)83188-9. newborn plasma.Pediatr Res.1981;15:684. https://doi.org/10.1203/00006450-
137. Bromberg YM, Abrahamov A, Salzberger M. The effect of maternal anoxaemia 198104001-01473.
on the foetal haemoglobin of the newborn. BJOG An Int J Obstet Gynaecol. 165. Carrell RW, Winterbourn CC, Rachmilewitz EA. Activated oxygen
1956;63(6):875–877. https://doi.org/10.1111/j.1471-0528.1956.tb05577.x. and haemolysis. Br J Haematol. 1975;30(3):259–264. https://doi.
138. Bard H.The effect of placental insufficiency on fetal and adult hemoglobin syn- org/10.1111/j.1365-2141.1975.tb00540.x.
thesis. Am J Obstet Gynecol. 1974;120(1):67–72. https://doi.org/10.1016/0002- 166. Benesch R, Benesch RE, Yu CI. Reciprocal binding of oxygen and diphospho-
9378(74)90181-1. glycerate by human hemoglobin. Proc Natl Acad Sci. 1968;59(2):526–532.
https://doi.org/10.1073/pnas.59.2.526.

Descargado para Mirko Flores Gonzalez (mfloresg8@upao.edu.pe) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en octubre 31,
2023. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
1124.e4SECTION XVIII — Developmental Hematopoiesis

167. Childs RA, Feizi T, Fukuda M, Hakomori SI. Blood-group-I activity associated 193. Kearin M, Kelly JG, O’Malley K. Digoxin “receptors” in neonates: an expla-
with band 3, the major intrinsic membrane protein of human erythrocytes. nation of less sensitivity to digoxin than in adults. Clin Pharmacol Ther.
Biochem J. 1978;173(1):333–336. https://doi.org/10.1042/bj1730333. 1980;28(3):346–349. https://doi.org/10.1038/clpt.1980.172.
168. Wranne L. Studies on erythro-kinetics in infancy.: VII. Quantitative esti- 194. Toivanen P, Hirvonen T. Iso- and heteroagglutinins in human fetal
mation of the haemoglobin catabolism by carbon monoxide tech- and neonatal sera. Scand J Haematol. 2009;6(1):42–48. https://doi.
nique in young infants. Acta Paediatr. 1967;56(4):381–390. https://doi. org/10.1111/j.1600-0609.1969.tb01802.x.
org/10.1111/j.1651-2227.1967.tb15396.x. 195. Thomaidis T, Agathopoulos A, Matsaniotis N. Natural isohemagglutinin pro-
169. Mollison PL. The survival of transfused erythrocytes in haemolytic disease of duction by the fetus. J Pediatr. 1969;74(1):39–48. https://doi.org/10.1016/
the newborn. Arch Dis Child. 1943;18(96):161–172. https://doi.org/10.1136/ s0022-3476(69)80006-5.
adc.18.96.161. 196. Gartner OT, Gilbert R, McDermott M, Benovitz S, Wolf AM. Anti-A and anti-B
170. Brace RA, Langendörfer C, Song TB, Mock DM. Red blood cell life span in the antibodies in children. J Am Med Assoc. 1967;201(3):206–207. https://doi.
ovine fetus. Am J Physiol Regul Integr Comp Physiol. 2000;279(4):R1196– org/10.1001/jama.1967.03130030076024.
1204. https://doi.org/10.1152/ajpregu.2000.279.4.R1196. 197. Stockman JA. Anemia of prematurity. Clin Perinatol. 1977;4(2):239–257.
171. Komazawa M, Oski FA. Biochemical characteristics of “young” and “old” eryth- https://doi.org/10.1016/S0095-5108(18)31207-7.
rocytes of the newborn infant. J Pediatr. 1975;87(1):102–106. https://doi. 198. Stockman JA. Anemia of prematurity. Current concepts in the issue of when
org/10.1016/S0022-3476(75)80082-5. to transfuse. Pediatr Clin North Am. 1986;33(1):111–128. https://doi.
172. Schmidt-Sommerfeld E, Penn D. Carnitine and total parenteral nutri- org/10.1016/s0031-3955(16)34972-0.
tion of the neonate. Biol Neonate. 1990;58(suppl 1):81–88. https://doi. 199. Soubasi V, Kremenopoulos G,Tsantali C, Savopoulou P, Mussafiris C, Dimitriou
org/10.1159/000243302. M. Use of erythropoietin and its effects on blood lactate and 2,3-diphospho-
173. Bracci R, Martini G, Buonocore G, et al. Changes in erythrocyte properties glycerate in premature neonates. Neonatology. 2000;78(4):281–287. https://
during the first hours of life: electron spin resonance of reacting sulfydryl doi.org/10.1159/000014280.
groups. Pediatr Res. 1988;24(3):391–395. https://doi.org/10.1203/00006450- 200. Oski FA, Stockman JA. Anaemia in early infancy. Br J Haematol. 1974;
198809000-00022. 27(2):195–200. https://doi.org/10.1111/j.1365-2141.1974.tb06786.x.
174. Böhler T, Leo A, Stadler A, Linderkamp O. Mechanical fragility of erythrocyte 201. Rossoff L, Zeldin R, Hew E, Aberman A. Changes in blood P50. Chest.
membrane in neonates and adults. Pediatr Res. 1992;32(1):92–96. https://doi. 1980;77(2):142–146. https://doi.org/10.1378/chest.77.2.142.
org/10.1203/00006450-199207000-00018. 202. Wimberley PD. Fetal hemoglobin, 2,3-diphosphoglycerate and oxygen trans-
175. Zipursky A, Brown E, Palko J, Brown EJ. The erythrocyte differential count in port in the newborn premature infant. Scand J Clin Lab Investig Suppl.
newborn infants. Am J Pediatr Hematol Oncol. 1983;5(1):45–51. 1982;160:1–149.
176. Holroyde CP, Oski FA, Gardner FH.The “pocked” erythrocyte. Red-cell surface 203. Novy MJ, Frigoletto FD, Easterday CL, Umansky I, Nelson NM. Changes in
alterations in reticuloendothelial immaturity of the neonate. N Engl J Med. umbilical-cord blood oxygen affinity after intrauterine transfusions for eryth-
1969;281(10):516–520. https://doi.org/10.1056/NEJM196909042811002. roblastosis. N Engl J Med. 1971;285(11):589–595. https://doi.org/10.1056/
177. Linderkamp O, Stadler AA, Zilow EP. Blood viscosity and optimal hemato- NEJM197109092851101.
crit in preterm and full-term neonates in 50– to 500–μm tubes. Pediatr Res. 204. Williams ML, Shoot RJ, O’Neal PL, Oski FA. Role of dietary iron and fat on
1992;32(1):97–102. https://doi.org/10.1203/00006450-199207000-00019. vitamin E deficiency anemia of infancy. N Engl J Med. 1975;292(17):887–890.
178. Klipp M, Holzwarth A-U, Poeschl JM, Nelle M, Linderkamp O. Effects of https://doi.org/10.1056/NEJM197504242921704.
erythropoietin on erythrocyte deformability in non-transfused preterm 205. Stockman JA, Graeber JE, Clark DA, McClellan K, Garcia JF, Kavey RE. Ane-
infants. Acta Paediatr. 2007;96(2):253–256. https://doi.org/10.1111/j.1651- mia of prematurity: determinants of the erythropoietin response. J Pediatr.
2227.2007.00101.x. 1984;105(5):786–792. https://doi.org/10.1016/s0022-3476(84)80308-x.
179. Cook GM, Heard DH, Seaman GV. Sialic acids and the electrokinetic 206. Brown MS, Garcia JF, Phibbs RH, Dallman PR. Decreased response of plasma
charge of the human erythrocyte. Nature. 1961;191:44–47. https://doi. immunoreactive erythropoietin to “available oxygen” in anemia of pre-
org/10.1038/191044a0. maturity. J Pediatr. 1984;105(5):793–798. https://doi.org/10.1016/s0022-
180. Eylar EH, Madoff MA, Brody OV, Oncley JL. The contribution of sialic 3476(84)80309-1.
acid to the surface charge of the erythrocyte. J Biol Chem. 1962;237: 207. Ross MP, Christensen RD, Rothstein G, et al. A randomized trial to develop
1992–2000. criteria for administering erythrocyte transfusions to anemic preterm infants 1
181. Kosztolányi GY, Jobst K. Electrokinetic analysis of the fetal erythrocyte mem- to 3 months of age. J Perinatol. 1989;9(3):246–253.
brane after trypsin digestion. Pediatr Res. 1980;14(2):138–141. https://doi. 208. Bifano EM, Smith F, Borer J. Relationship between determinants of oxygen
org/10.1203/00006450-198002000-00014. delivery and respiratory abnormalities in preterm infants with anemia. J Pedi-
182. Calatroni A, Cordaro V, Salpietro C, Barberi I. Erythrocyte membrane sialic atr. 1992;120(2 Pt 1):292–296. https://doi.org/10.1016/s0022-3476(05)80447-
acid in new-born infants. Acta Haematol. 1984;71(3):198–203. https://doi. 0.
org/10.1159/000206584. 209. Keyes WG, Donohue PK, Spivak JL, Jones MD, Oski FA. Assessing the need for
183. Adler SM, Denton RL. The erythrocyte sedimentation rate in the newborn transfusion of premature infants and role of hematocrit, clinical signs, and
period. J Pediatr. 1975;86(6):942–948. https://doi.org/10.1016/s0022- erythropoietin level. Pediatrics. 1989;84(3):412–417.
3476(75)80233-2. 210. Izraeli S, Ben-Sira L, Harell D, Naor N, Ballin A, Davidson S. Lactic acid as a pre-
184. Linderkamp O, Meiselman HJ. Geometric, osmotic, and membrane mechanical dictor for erythrocyte transfusion in healthy preterm infants with anemia of
properties of density-separated human red cells. Blood. 1982;59(6):1121–1127. prematurity. J Pediatr. 1993;122(4):629–631. https://doi.org/10.1016/s0022-
185. Goldbloom A. Icterus neonatorum. Arch Pediatr Adolesc Med. 1929;38(1):57. 3476(05)83551-6.
https://doi.org/10.1001/archpedi.1929.01930070069009. 211. Schulman I.The anemia of prematurity. J Pediatr. 1959;54(5):663–672. https://
186. Schröter W, Kashnitz E. Diagnosis of hereditary spherocytosis in newborn doi.org/10.1016/s0022-3476(59)80130-x.
infants. J Pediatr. 1983;103(3):460–463. https://doi.org/10.1016/S0022- 212. Pearson HA. Life-span of the fetal red blood cell. J Pediatr. 1967;70(2):166–171.
3476(83)80428-4. https://doi.org/10.1016/s0022-3476(67)80410-4.
187. King M-J, Smythe JS, Mushens R. Eosin-5-maleimide binding to band 3 and 213. Bratteby LE. Studies on erythro-kinetics in infancy. IX. Prediction of red
Rh-related proteins forms the basis of a screening test for hereditary sphero- cell volume from venous haematocrit in early infancy. Acta Paediatr
cytosis. Br J Haematol. 2004;124(1):106–113. https://doi.org/10.1046/j.1365- Scand. 1968;57(2):125–131. https://doi.org/10.1111/j.1651-2227.1968.
2141.2003.04730.x. tb04664.x.
188. Bianchi P, Fermo E, Vercellati C, et al. Diagnostic power of laboratory tests for 214. Shannon KM, Naylor GS, Torkildson JC, et al. Circulating erythroid progenitors
hereditary spherocytosis: a comparison study in 150 patients grouped accord- in the anemia of prematurity. N Engl J Med. 1987;317(12):728–733. https://
ing to molecular and clinical characteristics. Haematologica. 2012;97(4):516– doi.org/10.1056/NEJM198709173171203.
523. https://doi.org/10.3324/haematol.2011.052845. 215. Rhondeau SM, Christensen RD, Ross MP, Rothstein G, Simmons MA. Responsive-
189. Andres O, Eber S, Speer CP. Early postnatal diagnosis of hereditary spherocy- ness to recombinant human erythropoietin of marrow erythroid progenitors
tosis by combining light microscopy, acidified glycerol lysis test and eosin- from infants with the “anemia of prematurity. J Pediatr. 1988;112(6):935–940.
5’-maleimide binding assay. Ann Hematol. 2015;94(12):1959–1964. https:// https://doi.org/10.1016/s0022-3476(88)80223-3.
doi.org/10.1007/s00277-015-2491-z. 216. Ohls RK, Liechty KW, Turner MC, Kimura R, Christensen RD. Erythroid “burst
190. Christensen RD, Agarwal AM, Nussenzveig RH, Heikal N, Liew MA, Yaish HM. promoting” activity in serum of patients with the anemia of prematurity. J Pedi-
Evaluating eosin-5-maleimide binding as a diagnostic test for hereditary sphe- atr. 1990;116(5):786–789. https://doi.org/10.1016/s0022-3476(05)82672-1.
rocytosis in newborn infants. J Perinatol. 2015;35(5):357–361. https://doi. 217. Ohls RK, Harcum J, Li Y, Davila G, Christensen RD. Serum erythropoietin con-
org/10.1038/jp.2014.202. centrations fail to increase after significant phlebotomy losses in ill preterm
191. Hendricks SA, Lippe BM, Kaplan SA, Landaw EM, Hertz D, Scott M. Insu- infants. J Perinatol. 1997;17(6):465–467.
lin binding to erythrocytes of normal infants, children, and adults: variation 218. Ohls RK, Li Y, Trautman MS, Christensen RD. Erythropoietin production by
with age and sex. J Clin Endocrinol Metab. 1981;52(5):969–974. https://doi. macrophages from preterm infants: implications regarding the cause of
org/10.1210/jcem-52-5-969. the anemia of prematurity. Pediatr Res. 1994;35(2):169–170. https://doi.
192. Herzberg VL, Boughter MJ, Carlisle SK, Ahmad F, Hill DE. Insulin recep- org/10.1203/00006450-199402000-00008.
tor binding to cord blood erythrocytes of varying gestational age and 219. Ohls RK. Human recombinant erythropoietin in the prevention and treat-
comparison with adult values. Pediatr Res. 1980;14(1):4–7. https://doi. ment of anemia of prematurity. Pediatr Drugs. 2002;4(2):111–121. https://doi.
org/10.1203/00006450-198001000-00002. org/10.2165/00128072-200204020-00004.

Descargado para Mirko Flores Gonzalez (mfloresg8@upao.edu.pe) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en octubre 31,
2023. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
CHAPTER 109 — Developmental Erythropoiesis1124.e5

220. Maier RF, Obladen M, Scigalla P, et al. The effect of epoetin beta (recombinant Pediatr Res. 1990;28(3):284. https://doi.org/10.1203/00006450-199009000-
human erythropoietin) on the need for transfusion in very-low-birth-weight 00066.
infants. European Multicentre Erythropoietin Study Group. N Engl J Med. 239. Eschbach JW, Egrie JC, Downing MR, Browne JK, Adamson JW. Correction
1994;330(17):1173–1178. https://doi.org/10.1056/NEJM199404283301701. of the anemia of end-stage renal disease with recombinant human eryth-
221. Meyer MP, Meyer JH, Commerford A, et al. Recombinant human erythropoi- ropoietin. Results of a combined phase I and II clinical trial. N Engl J Med.
etin in the treatment of the anemia of prematurity: results of a double-blind, 1987;316(2):73–78. https://doi.org/10.1056/NEJM198701083160203.
placebo-controlled study. Pediatrics. 1994;93(6 Pt 1):918–923. 240. Brown MS, Jones MA, Ohls RK, Christensen RD. Single-dose pharmacokinet-
222. Shannon KM, Keith JF, Mentzer WC, et al. Recombinant human erythropoietin ics of recombinant human erythropoietin in preterm infants after intravenous
stimulates erythropoiesis and reduces erythrocyte transfusions in very low and subcutaneous administration. J Pediatr. 1993;122(4):655–657. https://doi.
birth weight preterm infants. Pediatrics. 1995;95(1):1–8. org/10.1016/s0022-3476(05)83559-0.
223. Ohls RK, Osborne KA, Christensen RD. Efficacy and cost analysis of treating 241. Ohls RK, Veerman MW, Christensen RD. Pharmacokinetics and effectiveness
very low birth weight infants with erythropoietin during their first two weeks of recombinant erythropoietin administered to preterm infants by continuous
of life: a randomized, placebo-controlled trial. J Pediatr. 1995;126(3):421–426. infusion in total parenteral nutrition solution. J Pediatr. 1996;128(4):518–523.
https://doi.org/10.1016/s0022-3476(95)70462-0. https://doi.org/10.1016/s0022-3476(96)70363-3.
224. Maier RF, Obladen M, Kattner E, et al. High-versus low-dose erythropoietin in 242. Cogar AA, Hartenberger CH, Ohls RK. Endothelin concentrations in pre-
extremely low birth weight infants. The European Multicenter rhEPO Study term infants treated with human recombinant erythropoietin. Neonatology.
Group. J Pediatr. 1998;132(5):866–870. https://doi.org/10.1016/s0022- 2000;77(2):105–108. https://doi.org/10.1159/000014203.
3476(98)70320-8. 243. Warwood TL, Ohls RK,Wiedmeier SE, et al. Single-dose darbepoetin administra-
225. Donato H, Vain N, Rendo P, et al. Effect of early versus late administration of tion to anemic preterm neonates. J Perinatol. 2005;25(11):725–730. https://
human recombinant erythropoietin on transfusion requirements in premature doi.org/10.1038/sj.jp.7211387.
infants: results of a randomized, placebo-controlled, multicenter trial. Pediat- 244. Warwood TL, Ohls RK, Lambert DK, et al. Intravenous administration of dar-
rics. 2000;105(5):1066–1072. https://doi.org/10.1542/peds.105.5.1066. bepoetin to NICU patients. J Perinatol. 2006;26(5):296–300. https://doi.
226. Ohls RK, Ehrenkranz RA, Wright LL, et al. Effects of early erythropoietin org/10.1038/sj.jp.7211498.
therapy on the transfusion requirements of preterm infants below 1250 245. Halpérin DS, Wacker P, Lacourt G, et al. Effects of recombinant human eryth-
grams birth weight: a multicenter, randomized, controlled trial. Pediatrics. ropoietin in infants with the anemia of prematurity: a pilot study. J Pediatr.
2001;108(4):934–942. https://doi.org/10.1542/peds.108.4.934. 1990;116(5):779–786. https://doi.org/10.1016/s0022-3476(05)82671-x.
227. Maier RF, Obladen M, Müller-Hansen I, et al. Early treatment with erythropoi- 246. Ohls RK, Christensen RD. Recombinant erythropoietin compared with
etin beta ameliorates anemia and reduces transfusion requirements in infants erythrocyte transfusion in the treatment of anemia of prematurity. J Pediatr.
with birth weights below 1000 g. J Pediatr. 2002;141(1):8–15. https://doi. 1991;119(5):781–788. https://doi.org/10.1016/s0022-3476(05)80303-8.
org/10.1067/mpd.2002.124309. 247. Beck D, Masserey E, Meyer M, Calame A. Weekly intravenous administration of
228. Pathak A, Roth P, Piscitelli J, Johnson L. Effects of vitamin E supplementation recombinant human erythropoietin in infants with the anaemia of prematurity.
during erythropoietin treatment of the anaemia of prematurity. Arch Dis Eur J Pediatr. 1991;150(11):767–772. https://doi.org/10.1007/BF02026707.
Child Fetal Neonatal Ed. 2003;88(4):F324–F328. https://doi.org/10.1136/ 248. Christensen RD, Liechty KW, Koenig JM, Schibler KR, Ohls RK. Administration
fn.88.4.f324. of erythropoietin to newborn rats results in diminished neutrophil produc-
229. Meyer MP, Sharma E, Carsons M. Recombinant erythropoietin and blood tion. Blood. 1991;78(5):1241–1246.
transfusion in selected preterm infants. Arch Dis Child Fetal Neonatal Ed. 249. Christensen RD, Rothstein G. Erythropoietin affects the maturation pattern
2003;88(1):F41–F45. https://doi.org/10.1136/fn.88.1.f41. of fetal G-CSF-responsive progenitors. Am J Hematol. 1992;39(2):108–112.
230. Türker G, Sarper N, Gökalp AS, Usluer H. The effect of early recombinant https://doi.org/10.1002/ajh.2830390207.
erythropoietin and enteral iron supplementation on blood transfusion in pre- 250. Aher SM, Ohlsson A. Early versus late erythropoietin for preventing red
term infants. Am J Perinatol. 2005;22(08):449–455. https://doi.org/10.105 blood cell transfusion in preterm and/or low birth weight infants. Cochrane
5/s-2005-918888. Database Syst Rev. 2006;3:CD004865. https://doi.org/10.1002/14651858.
231. Pollak A, Hayde M, Hayn M, et al. Effect of intravenous iron supplementation CD004865.pub2.
on erythropoiesis in erythropoietin-treated premature infants. Pediatrics. 251. Ohlsson A, Aher SM. Early erythropoietin for preventing red blood cell
2001;107(1):78–85. https://doi.org/10.1542/peds.107.1.78. transfusion in preterm and/or low birth weight infants. Cochrane Data-
232. Haiden N, Schwindt J, Cardona F, et al. Effects of a combined therapy of base Syst Rev. 2006;3:CD004863. https://doi.org/10.1002/14651858.
erythropoietin, iron, folate, and vitamin b12 on the transfusion requirements CD004863.pub2.
of extremely low birth weight infants. Pediatrics. 2006;118(5):2004–2013. 252. Aher SM, Ohlsson A. Late erythropoietin for preventing red blood cell transfu-
https://doi.org/10.1542/peds.2006-1113. sion in preterm and/or low birth weight infants. Cochrane Database Syst Rev.
233. Ohls RK, Harcum J, Schibler KR, Christensen RD. The effect of erythropoie- 2014;4:CD004868. https://doi.org/10.1002/14651858.CD004868.pub4.
tin on the transfusion requirements of preterm infants weighing 750 grams 253. Ohlsson A, Aher SM. Early erythropoietin for preventing red blood cell
or less: a randomized, double-blind, placebo-controlled study. J Pediatr. transfusion in preterm and/or low birth weight infants. Cochrane Data-
1997;131(5):661–665. https://doi.org/10.1016/s0022-3476(97)70089-1. base Syst Rev. 2014;4:CD004863. https://doi.org/10.1002/14651858.
234. Ohls RK, Christensen RD, Kamath-Rayne BD, et al. A randomized, masked, CD004863.pub4.
placebo-controlled study of darbepoetin alfa in preterm infants. Pediatrics. 254. Natalucci G, Latal B, Koller B, et al. Effect of early prophylactic high-dose
2013;132(1):e119–e127. https://doi.org/10.1542/peds.2013-0143. recombinant human erythropoietin in very preterm infants on neurodevel-
235. Juul SE, Comstock BA, Wadhawan R, et al. A randomized trial of erythropoietin opmental outcome at 2 years: a randomized clinical trial. J Am Med Assoc.
for neuroprotection in preterm infants. N Engl J Med. 2020;382(3):233–243. 2016;315(19):2079. https://doi.org/10.1001/jama.2016.5504.
https://doi.org/10.1056/NEJMoa1907423. 255. Ohls RK, Ehrenkranz RA, Das A, et al. Neurodevelopmental outcome and
236. Song J, Sun H, Xu F, et al. Recombinant human erythropoietin improves neu- growth at 18 to 22 months’ corrected age in extremely low birth weight infants
rological outcomes in very preterm infants. Ann Neurol. 2016;80(1):24–34. treated with early erythropoietin and iron. Pediatrics. 2004;114(5):1287–1291.
https://doi.org/10.1002/ana.24677. https://doi.org/10.1542/peds.2003-1129-L.
237. George JW, Bracco CA, Shannon KM, et al. Age-related differences in erythro- 256. Haiden N, Klebermass K, Cardona F, et al. A randomized, controlled trial of
poietic response to recombinant human erythropoietin: comparison in adult the effects of adding vitamin b12 and folate to erythropoietin for the treat-
and infant rhesus monkeys. Pediatr Res. 1990;28(6):567–571. https://doi. ment of anemia of prematurity. Pediatrics. 2006;118(1):180–188. https://doi.
org/10.1203/00006450-199012000-00003. org/10.1542/peds.2005-2475.
238. Widness JA,Veng-Pedersen P, Modi NB, Schmidt RL, Chestnut DH. Developmen-
tal changes in erythropoietin (Ep) pharmacokinetics in fetal and neonatal sheep.

Descargado para Mirko Flores Gonzalez (mfloresg8@upao.edu.pe) en Antenor Orrego Private University de ClinicalKey.es por Elsevier en octubre 31,
2023. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.

You might also like