Anda di halaman 1dari 12

Core Concepts: The Biology of Hemoglobin

Robin K. Ohls
NeoReviews 2011;12;e29-e38
DOI: 10.1542/neo.12-1-e29

The online version of this article, along with updated information and services, is
located on the World Wide Web at:
http://neoreviews.aappublications.org/cgi/content/full/neoreviews;12/1/e29

NeoReviews is the official journal of the American Academy of Pediatrics. A monthly publication,
it has been published continuously since 2000. NeoReviews is owned, published, and trademarked
by the American Academy of Pediatrics, 141 Northwest Point Boulevard, Elk Grove Village,
Illinois, 60007. Copyright © 2011 by the American Academy of Pediatrics. All rights reserved.
Online ISSN: 1526-9906.

Downloaded from http://neoreviews.aappublications.org by Sreeram Subramanian on February 28, 2011


Article core concepts

Core Concepts: The Biology of Hemoglobin


Robin K. Ohls, MD*
Abstract
A consistent and organized transition from embryonic to fetal to adult hemoglobin
Author Disclosure (Hgb) occurs during human fetal development. Hgb concentrations gradually in-
Dr Ohls has disclosed crease, averaging 18 g/dL (180 g/L) by 40 weeks’ gestation. The ability to deliver
no financial oxygen to tissues in the fetus and neonate is primarily determined by the percentage of
fetal versus adult Hgb and the concentration of 2,3 diphosphoglycerate (2,3-DPG).
relationships relevant
Studies continue to evaluate the relationship between Hgb concentrations and oxygen
to this article. This
delivery in neonates to determine what Hgb concentrations best meet the needs of a
commentary does not wide variety of clinical situations from the critically ill extremely low-birthweight
contain a discussion infant to the stable growing preterm infant. Biochemical interactions between nitric
of an unapproved/ oxide (NO) and Hgb beyond the production of methemoglobin do occur and may be
investigative use of a a source of deliverable NO to the microcirculation under hypoxic conditions.
commercial
product/device.
Objectives After completing the article, readers should be able to:

1. Describe the development of globin gene synthesis and Hgb formation.


2. Explain fetal to neonatal transition of Hgb F to Hgb A.
3. Review the development and function of 2,3-DPG.
4. Describe the relationship between NO and Hgb.

Hemoglobin Concentration During Development


Red blood cell indices such as Hgb, hematocrit, mean cell volume, and red cell distribution
width change during gestation and continue to change through the first postnatal year. (1)
Hgb concentrations gradually rise during gestation. At 10 weeks’ gestation, the average
concentration is approximately 9 g/dL (90 g/L); (1)(2) by the start of the third trimester,
values in the developing fetus reach 11 to 12 g/dL (110 to 120 g/L); and by 30 weeks,
Hgb concentrations are 13 to 14 g/dL (130 to 140 g/L). (1) Jopling and associates (3)
have identified reference ranges based on approximately 25,000 preterm and term infants.
From 22 to 40 weeks’ gestation, there is a consistent increase in Hgb of 0.21 g/dL
(2.1 g/L) per week (Fig. 1). (3) In this large cross-sectional study, no sex differences were
noted in Hgb concentrations, but some studies have reported a slight difference in Hgb
concentrations between white and African American preterm infants. (4) At delivery, a 1-
to 2-g/dL (10- to 20-g/L) rise in Hgb may result from transfusion of placental blood into
the infant. In term and late preterm infants, Hgb concentrations increase by approximately
4% at 4 hours of postnatal age, resulting from a decrease in plasma volume. (5) By 8 to
12 hours of age, Hgb concentrations achieve a relatively constant level. In contrast, in an
evaluation of more than 20,000 preterm infants (29 to 34 weeks’ gestation), a decrease of
approximately 6% was measured at 4 hours of age. (3) This decrease might be due to a lack
of placental transfusion because the umbilical cord in preterm infants is rapidly clamped to
expedite resuscitation.
Red blood cell production decreases significantly after birth, primarily as a result of the
increased availability of oxygen, which greatly reduces erythropoietin (Epo) production
and endogenous erythropoiesis. By the end of the first postnatal week, Hgb concentrations
begin to decline (3) and continue to decrease over the next several weeks as a result of
decreased erythrocyte production, a shortened erythrocyte life span, and an increase in

*Professor of Pediatrics, University of New Mexico, Albuquerque, NM.

NeoReviews Vol.12 No.1 January 2011 e29


Downloaded from http://neoreviews.aappublications.org by Sreeram Subramanian on February 28, 2011
core concepts hemoglobin

count for the higher oxygen needs


and the compensatory increase in
erythropoiesis and Hgb concentra-
tion. (15)

Hemoglobin Synthesis
During fetal erythropoiesis, an or-
derly evolution of the production
of various Hgbs occurs. Eight glo-
bin genes direct the synthesis of six
different polypeptide chains, desig-
nated alpha (␣), beta (␤), gamma
(␥), delta (␦), epsilon (␧), and zeta
(␨). These globin chains combine
in the developing erythroblast to
form seven different Hgb tetramers:
Gower 1 (␨2-␧2), Gower 2 (␣2-␧2),
Portland (␨2-␥2), fetal hemoglobin
Figure 1. Reference ranges for blood hemoglobin concentrations at birth in 24,416 (Hgb F: ␣2-␥2), and two types of
patients at 22 to 42 weeks’ gestation. The solid line represents the mean value and the adult hemoglobin: ␣2-␤2, known as
dashed lines represent the 5% to 95% reference range. Reprinted with permission from Hgb A, and ␣2-␦2, known as Hgb
Jopling. (3) A2 (Table 1).

blood volume related to growth (Fig. 2). Term infants Globin Genes
reach their Hgb nadir at approximately 8 weeks, with The globin genes are organized into two clusters
an average Hgb concentration of 11.2 g/dL (112 g/L). (Fig. 3). The ␣-like genes are located along a 20-kb distal
(6) Hgb concentrations subsequently rise so that by segment of the short arm of chromosome 16. The cluster
6 months, the concentration averages 12.1 g/dL contains three functional genes (␣1, ␣2, and ␨2), three
(121 g/L). (7) pseudogenes (evolutionary remnants of genes that are
The decline in Hgb in very low-birthweight infants is not expressed because of inactivating mutations that
greater than that in term infants, in part because of prevent production of a functional globin protein), and
phlebotomy losses and in part because of the suppressive one gene of undetermined function (a globin-like gene
impact of transfusions on endogenous erythropoiesis. without inactivating mutations). The ␤-like gene cluster
Such infants reach the Hgb nadir of 8 g/dL (80 g/L) is located along a 60-kb segment of the short arm of
at 4 to 8 weeks of age. (8) Figure 2 demonstrates chromosome 11, and it contains five functional genes
relationships among birthweight, chronologic age, and (␤, ␦, A␥, G␥, and ␧) and one pseudogene. Within each
Hgb in term and preterm infants. (3) complex, the genes are all in the same 5⬘ to 3⬘ orienta-
Maternal conditions can influence fetal Hgb concen- tion, and they are arranged in the order in which they are
trations. Infants born small for gestational age can have expressed during development. (16)
higher Hgb concentrations due to placental insufficiency
and secondary polycythemia. (9)(10) Infants of dia- Timing of Globin Chain Synthesis
betic mothers, infants of smoking mothers, and infants Globin chain production has been determined at each
born at higher altitudes also tend to have higher Hgb stage of development, from initial yolk sac (primitive) to
concentrations at birth (11)(12)(13)(14) In all of these hepatic (definitive) and marrow erythropoiesis. It is not
conditions, accelerated erythropoiesis is believed to be clear why or how primitive erythroid progenitors pro-
part of a compensating mechanism designed to raise grammed to produce embryonic Hgb transition to de-
oxygen-carrying capacity to maintain an adequate oxy- finitive progenitors programmed to produce Hgb F.
gen supply to the fetus. In the case of the fetus of a Because quantification of globin gene expression using
mother who has diabetes, increased metabolic demands real time polymerase chain reaction methods reflects
of the fetus (as evidenced by a positive correlation be- production by a heterogeneous source of erythrocytes,
tween maternal Hgb A1c and neonatal Hgb) may ac- production of a specific Hgb is usually reported as a

e30 NeoReviews Vol.12 No.1 January 2011


Downloaded from http://neoreviews.aappublications.org by Sreeram Subramanian on February 28, 2011
core concepts hemoglobin

During the sixth to seventh week of


gestation, primitive erythroblasts
continue to produce ␣, ␨, and ␧
chains, while definitive erythrocytes
produce ␣, ␧, G␥, and A␥ chains. By
the seventh to eighth week, ␧- and
␨-chain synthesis is no longer de-
tectable, and the primary globin
chains produced are ␣, G␥, and A␥.
␤-chain production is just barely
detectable at this time and gradu-
ally increases, comprising up to 10%
of total non–␣-chain production by
10 weeks of gestation. (18) Genetic
disorders associated with ␤-chain
synthetic or structural abnormali-
ties may be detected in utero as
soon as ␤-chain production occurs
but are often not clinically apparent
until after birth.
From 10 to 33 weeks of gesta-
tion, the primary globin chains syn-
thesized are ␣, G␥, A␥, and ␤. As-
sessment of the output of the two
linked ␣-globin genes by mRNA
analysis suggests that they are ex-
pressed in the ratio (␣2:␣1) rang-
ing from 1.5 to 3.0:1 throughout
fetal life, and this ratio continues
through normal adulthood. The
relative rates of G␥-chain and A␥-
chain production are also constant
throughout fetal life at a G␥:A␥ ratio
of approximately 3:1. (19) During
the transition from 32 to 36 weeks
of gestation, the relative rate of
␤-chain synthesis increases and that
of ␥-chain production declines, so
at birth, ␤-chain synthesis makes
Figure 2. Reference ranges for blood hemoglobin concentrations in 39,559 patients
during the 28 days after birth in late preterm and term infants 35 to 42 weeks’ gestation up approximately 50% of non–␣-
(panel A) and in preterm infants 29 to 34 weeks’ gestation (panel B). The solid line chain synthesis. There is consider-
represents the mean value and the dashed lines represent the 5% to 95% reference range. able variation among infants, how-
Reprinted with permission from Jopling. (3) ever, with many infants showing
prolonged dependence on Hgb F.
After birth, the level of ␤-chain pro-
percentage of the total Hgb measured. Studies measur- duction increases, while the level of ␥-chain production
ing Hgb production by erythroid colonies in culture steadily declines, so by the end of the first year, ␥-chain
show that individual cells in a colony produce predomi- synthesis reaches the low concentration that is character-
nantly one type of Hgb. (17) istic of adult life (⬍2%). Over the first few months after
During the fourth to fifth week of gestation, ␣, ␨, and birth, the G␥:A␥ ratio changes from 3:1 to 2:3, although
␧ chains are the primary globin chains produced (Fig. 4). this ratio varies in adults. (20)(21)(22)

NeoReviews Vol.12 No.1 January 2011 e31


Downloaded from http://neoreviews.aappublications.org by Sreeram Subramanian on February 28, 2011
core concepts hemoglobin

Globin Chain Composition


Table 1. ing in the presence of ␧4. (23) Soon thereafter, ␣- and
␨-chain production begins, and Hgb Gower 1 (␨2-␧2),
of Common Hemoglobin Gower 2 (␣2-␧2), and Portland (␨2-␥2) are formed. (24)
By 5 to 6 weeks’ gestation, Hgb Gower 1 and Gower 2
Type of Hemoglobin Composition
constitute 42% and 24% of the total Hgb, respectively,
Embryonic Hemoglobin: with Hgb F (␣2-␥2) making up the remainder. By 14 to
Gower 1 ␨2-␧2 16 weeks, Hgb F constitutes 50% of the total Hgb, and
Gower 2 ␣2-␧2
Portland ␨2-␥2 by 20 weeks, it forms more than 90% of the Hgb. (25)
Fetal Hemoglobin: Small quantities of Hgb A (␣2-␤2) are found beginning
F ␣2-␥2 at 6 to 8 weeks’ gestation. The increase in ␤-chain
Adult Hemoglobin: production occurring between 12 and 20 weeks’ gesta-
A ␣2-␤2 tion accounts for the sudden rise in Hgb A found at
A2 ␣2-␦2
the end of the first trimester of pregnancy. Tetramers of
␥-chains (␥4 or Hgb Barts) and ␤-chains (␤4 or Hgb H)
Delta-chain production has been observed as early as can be found in conditions in which ␣-chain synthesis is
32 weeks’ gestation. Delta-gene production lags behind impaired or absent, such as ␣-thalassemia syndromes.
␤-gene production, so the adult ␤/␦ synthesis ratio is not Hgb F is easily distinguished immunologically and bio-
reached until 4 to 6 months after birth. chemically from Hgb A. The primary differentiating physi-
ologic characteristic of Hgb F is its decreased interaction
with 2,3-DPG (also known as 2, 3 bisphosphoglycerate).
Hemoglobin Production During Development 2,3-DPG binds to deoxyhemoglobin in a cavity between
Developmental changes in the production of Hgb can be the ␤-chains and stabilizes the deoxy form of Hgb, resulting
seen in Figure 5. Before the onset of other chain forma- in reduced Hgb-oxygen affinity. 2,3-DPG binds less effec-
tion, unpaired globin chains may form tetramers, result- tively to ␥-globin chains because of the differing amino acid
sequence in the non–␣-chain. Con-
sequently, 2,3-DPG does not reduce
the oxygen affinity of Hgb F as much
as that of Hgb A.
Fetal and adult Hgb also differ in
solubility. Hgb F is more soluble in
strong phosphate buffers than Hgb A
and is oxidized to methemoglobin
more easily than Hgb A. Hgb F has a
considerably greater affinity for oxy-
gen as a result of differences in bind-
ing to 2,3-DPG mentioned previ-
ously. Hgb F is resistant to acid
elution, which allows differentiation
of cells containing Hgb F from cells
containing Hgb A. (26) This prop-
erty forms the basis of differentiating
fetal from maternal red cells using the
Kleihauer Betke stain.
Figure 3. Organization of the globin genes. Transcription takes place from the 5ⴕ to the 3ⴕ G
␥-chains represent 70% to 80%
end; for both chromosomes, the genes are arranged in order of their developmental activation.
of the total ␥-chains in the blood of
The upper part of the figure represents the beta-like globin genes on the short arm of
the fetus and newborn. The percent
chromosome 11, and the lower part of the figure represents the alpha-like genes on the distal
short arm of chromosome 16. Regions of the gene that code for primary globin proteins are of ␥-chains made up of ␥ falls to
G

shown as shaded ovals. Regions that code for pseudogenes (y-nonexpressed remnants that about 40% by 5 months of age. This
have a number of inactivating mutations that prevent transcription and translation into unique difference in ␥-chain pro-
G

functional globin protein) are shown as open ovals. ␪-1 is a globin-like gene without duction found in the fetus helps to
inactivating mutations. The locus control region (LCR) is shown as a hatched segment. distinguish fetal hematopoiesis from

e32 NeoReviews Vol.12 No.1 January 2011


Downloaded from http://neoreviews.aappublications.org by Sreeram Subramanian on February 28, 2011
core concepts hemoglobin

of diabetic mothers. (30)(31) Ele-


vated concentrations of Hgb F may
have protective effects in some dis-
ease states. For example, a high con-
centration of fetal Hgb F in patients
who have sickle cell disease may be a
predictor of increased adult life ex-
pectancy. (32)(33)
On a body weight basis, red blood
cell production during the latter
months of gestation is significantly
greater compared with that in adult
life. Immediately after birth, erythro-
poiesis is considerably reduced, asso-
ciated with a steady and linear decline
in ␥-chain synthesis during the pe-
riod of reduced neonatal erythropoi-
esis. Newly synthesized red blood cells
appearing in the circulation when
erythropoiesis resumes contain pre-
Figure 4. Production of globin chains during the fetal and neonatal period. dominantly Hgb A. The postpartum
decline of Hgb F production and of
that found in later life. Under stress, the older infant and the intercellular distribution of fetal and adult Hgb over
adult increase production of Hgb F. Increased Hgb F the first postnatal months has been extensively examined.
production often occurs in leukemic states and in other Immediately after birth, there is a brief rise in Hgb F,
conditions. (27)(28) The delay in the switch of Hgb F to followed by a steady decline. Studies of the intercellular
Hgb A has been noted in conditions of maternal hypoxia, in distribution of Hgb F (using an acid-elution technique)
infants who are small for gestational age, (29) and in infants have shown that the distribution of Hgb F is heterogeneous
over the first few months after birth.
At 3 months, the distribution of
Hgb F becomes bimodal, with pop-
ulations of cells that contain acid-
resistant Hgb F and populations of
cells containing Hgb A. These obser-
vations have suggested that Hgb
F-containing cells are replaced by a
population of cells containing Hgb A
during the early postnatal period.
Studies show that the type of
globin chains produced at different
stages of development are not closely
related to the site of erythropoiesis.
It appears that ␨- and ␧-chains are
synthesized in both primitive and
definitive cell lines. Moreover, dur-
ing the later stages of fetal develop-
ment, the switch from ␥- to ␤-chain
production occurs synchronously
throughout the liver and bone
marrow. The transition from ␥- to
Figure 5. Hemoglobin production during the fetal and neonatal period. ␤-chain synthesis is most closely re-

NeoReviews Vol.12 No.1 January 2011 e33


Downloaded from http://neoreviews.aappublications.org by Sreeram Subramanian on February 28, 2011
core concepts hemoglobin

lated to postconceptional age and not chronologic age. Certain factors are known to alter the affinity of Hgb for
(27) Thus, infants born preterm continue to synthesize oxygen (Table 2). The most important of these are the Hgb
significant amounts of ␥-globin (and fetal Hgb) until F concentration and the red cell 2,3-DPG content. The
40 weeks’ gestation. concentration of red blood cell 2,3-DPG gradually in-
creases with gestation. At term, the concentration is similar
2,3-DPG Metabolism to that of adults. By the end of the first postnatal week, the
The affinity of Hgb for oxygen can be decreased by 2,3-DPG concentrations are considerably higher than they
interaction with certain organic phosphates, such as are at birth. After the first week, red blood cell 2,3-DPG
2,3-DPG and adenosine triphosphate. (34) The highly concentrations remain relatively unchanged for the next
charged anion 2,3-DPG binds to deoxyhemoglobin but 6 months. In term infants, the Hgb-oxygen dissociation
not to oxyhemoglobin. Deoxyhemoglobin F does not curve gradually shifts to the right, and by 4 to 6 months of
possess as great an affinity for 2,3-DPG as does deoxy- age, the P50 values approximate those of the adult.
hemoglobin A and, therefore, cannot bind 2,3-DPG to The situation is somewhat different in preterm in-
the same degree as Hgb A. Thus, the fetal leftward- fants. Because Hgb F synthesis is still active, increases in
shifted Hgb oxygen dissociation curve (Fig. 6) is not P50 seen in term infants as a result of the switch from
easily modulated in the presence of 2,3-DPG. Hgb F to Hgb A do not occur as rapidly. The red blood
The P50 (partial pressure of oxygen at which half of cell 2,3-DPG concentrations also are slightly lower in
Hgb is saturated) of fetal blood is 19 to 21 mm Hg, some preterm infants. (35) These concentrations are increased
6 to 8 mm Hg lower than that of adult blood. As Hgb F with the use of human recombinant Epo, which shifts the
concentration declines after birth, however, there is a oxygen dissociation curve to the right. (36)(37)
marked rightward shift in the postnatal Hgb oxygen
equilibrium curve. The percentage of Hgb A and the red Nitric Oxide-hemoglobin Interactions
cell 2,3-DPG content play the greatest roles in altering NO plays a significant role in vasoactive regulation.
the position of the Hgb oxygen dissociation curve. As a Under baseline conditions, NO is produced by endo-
result, preterm infants who have a greater proportion of thelial NO synthase and diffuses into surrounding smooth
Hgb A but less 2,3-DPG (which occurs following packed muscle cells, activating soluble guanylyl cyclase to produce
red blood cell transfusion) may have a similar P50 as those cyclic guanosine 5⬘-monophosphate, and regulates vascular
who have increased quantities of Hgb F. tone. NO reacts with oxyhemoglobin to form methemo-
globin, which is reversed by erythro-
cytic methemoglobin reductase. A
second reaction also can occur, in
which NO reacts with deoxyhemo-
globin to form nitrosyl hemoglobin
(NO-Hgb). There is some evidence
that erythrocytes containing NO-
Hgb may be able to release NO into
the circulation, thus causing vasodi-
latation in the microvasculature. A
third reaction has been studied that
involves the binding of NO to the
␤-chain cysteine amino acid to form
S-nitrosyl-hemoglobin (SNO-Hgb).
It has been postulated that nitrite
ions within erythrocytes can be re-
duced to NO by deoxyhemoglobin,
so NO is generated as erythrocytes
enter hypoxic regions. All of these
potential mechanisms result in NO
Figure 6. Hemoglobin-oxygen dissociation curve. The curve representing fetal hemoglo- controlling blood flow via hypoxic
bin is on the left, and the curve representing adult hemoglobin is on the right. The P50 is vasodilatation. These mechanisms
shown as a hatched line for each curve. are especially important in preterm

e34 NeoReviews Vol.12 No.1 January 2011


Downloaded from http://neoreviews.aappublications.org by Sreeram Subramanian on February 28, 2011
core concepts hemoglobin

tissues may be greater in utero because of the character-


Factors Affecting
Table 2. istics of the Hgb-oxygen dissociation curve.
If pulmonary function is normal, the PO2 of pulmo-
Hemoglobin-Oxygen Affinity nary blood rises from the 40 mm Hg of pulmonary
Increased P50, increased red blood cell 2,3-DPG: arterial blood to the 100 mm Hg of pulmonary venous
blood. Because of the shape of the Hgb-oxygen dissoci-
● Adaptation to high altitude
● Hypoxemia associated with chronic pulmonary disease ation curve, these PO2 values permit 95% saturation of
● Hypoxemia associated with cyanotic heart disease Hgb by oxygen. Further increases in PO2 produce little
● Anemia additional increase in saturation. In the healthy adult,
● Decreased red blood cell mass approximately 50% of Hgb is saturated with oxygen
● Hyperthyroidism
when the PO2 falls to 27 mm Hg (P50⫽27 mm Hg). In
● Red cell pyruvate kinase deficiency
situations in which the Hgb-oxygen dissociation curve
Increased P50, no consistent alteration in red blood cell has shifted to the right, the affinity of Hgb for oxygen is
DPG:
reduced. Thus, at any given PO2, more oxygen is released
● Abnormal hemoglobins (Kansas, Seattle, to tissues. Conversely, if the curve is shifted to the left,
Hammersmith, Tacoma, E) the affinity of Hgb for oxygen is increased. Therefore, at
● Vigorous exercise
any given PO2, less oxygen is released to the tissues.
Decreased P50, decreased red blood cell 2,3-DPG: A precise relationship does not exist between the decline
● Septic shock in Hgb F and the decrease in oxygen affinity of a neonate’s
● Severe acidosis blood. (38) Rather, changes in P50 reflect the interaction
● Following massive transfusions of stored blood between red blood cell 2,3-DPG, the increase in Hgb A
● Neonatal respiratory distress syndrome
that occurs after birth, and the decline in Hgb F. Although
Decreased P50, no consistent alteration in red blood cell oxygen-carrying capacity (Hgb concentration ⫻ oxygen
DPG: saturation ⫻ 1.36 mL oxygen/g of Hgb) decreases over
● Abnormal hemoglobins (Kempsey, Chesapeake, the first few postnatal months as Hgb concentration de-
Capetown, Yakima, Rainier) clines, the amount of oxygen delivery can remain similar or
2,3-DPG⫽2,3 diphosphoglycerate, P50⫽partial pressure of oxygen at even increase. (35) For example, a preterm infant born with
which half of hemoglobin is saturated
a Hgb concentration of 15 g/dL (150 g/L) delivers 1 mL
of oxygen to the tissues for every 100 mL of circulating
infants because vulnerable vascular beds such as the splanch- blood (based on a P50 of 19 and a central venous PO2 of
nic system are at risk for hypoxic injury via shunting of 40 mm Hg). As the percent of Hgb A increases over time,
blood to the brain and heart. Blood that is collected for the P50 shifts to the right. The infant can now deliver
transfusion loses its SNO-Hgb within hours, so transfused 2.1 mL of oxygen per 100 mL of blood, despite a decrease
packed red blood cells (PRBCs) lack NO. Transfusing in total Hgb to 8 g/dL (80 g/L) (based on a P50 of 24 mm
PRBCs that cannot deliver NO to the microvasculature Hg and a central venous PO2 of 40 mm Hg). (35)
may, in fact, reduce local oxygen delivery to tissues through After intrauterine transfusion, infants have oxygen-
vasoconstriction. Studies are in progress to evaluate the unloading properties characteristic of adult blood. Despite
renitrosylization of Hgb before transfusion. the decrease in oxygen affinity that accompanies intrauter-
ine transfusion, no deleterious effects of this procedure with
Oxygen Transport respect to oxygen uptake by the fetus have been docu-
The mechanisms controlling oxygen transport in utero mented. (39) The physiologic significance of manipulating
and immediately postpartum are complex. During pre- the Hgb-oxygen affinity of extremely preterm infants via
natal life, the fetal arterial oxygen tension (PO2) is ap- PRBC transfusions continues to be studied. It is important
proximately 30 mm Hg, and the venous PO2 is approxi- to understand an infant’s ability to deliver oxygen to tissues
mately 15 mm Hg (Fig. 6). These low PO2s contribute to when determining whether to administer an erythrocyte
the development of relative polycythemia in the fetus. transfusion. The decision to transfuse should not be based
After birth, numerous factors affect oxygenation, includ- on Hgb concentration alone. Transfusions significantly af-
ing the inspired gas mixture, pulmonary function, the fect an infant’s endogenous erythropoiesis: for infants who
arterial oxygen dissociation curve, and the ability to undergo exchange transfusion or multiple transfusions,
extract oxygen at the tissue level. (38) It has been spec- both Epo concentrations and reticulocyte counts are lower
ulated that the actual amount of oxygen released to at any given Hgb concentration. (8)(40) The search con-

NeoReviews Vol.12 No.1 January 2011 e35


Downloaded from http://neoreviews.aappublications.org by Sreeram Subramanian on February 28, 2011
core concepts hemoglobin

tinues for a specific marker that reflects the need for im- DG, eds. Hematology of Infancy and Childhood. Philadelphia, PA:
proved oxygen delivery to tissues (via red blood cell trans- WB Saunders; 1993:18 – 43
7. Kling PJ, Schmidt RL, Roberts RA, Widness JA. Serum eryth-
fusion). Currently, no ideal marker that is simple, requires
ropoietin levels during infancy: associations with erythropoiesis.
little or no blood, is reproducible, and can be applied to J Pediatr. 1996;128:791
preterm infants exists for clinical use in neonates. 8. Stockman JA 3rd, Garcia JF, Oski FA. The anemia of prematu-
rity: factors governing the erythropoietin response. N Engl J Med.
Summary 1977;296:647
The organized transition from embryonic to fetal to adult 9. Humbert JR, Abelson H, Hathaway WE, Battaglia FC. Poly-
Hgb has been extensively studied, providing significant cythemia in small for gestational age infants. J Pediatr. 1969;75:812
10. Hakanson DO, Oh W. Hyperviscosity in the small for gesta-
understanding of the molecular basis of Hgb development. tional age infant. Pediatr Res. 1977;11:472A
Studies continue to evaluate the relationship between Hgb 11. Moore LG, Newberry MA, Freeby GM, Crnic LS. Increased
concentrations and oxygen delivery in neonates to best incidence of neonatal hyperbilirubinemia at 3100 m in Colorado.
determine what Hgb concentrations best meet the needs of Am J Dis Child. 1984;138:158
a wide variety of clinical situations from the critically ill 12. Bureau MA, Shapcott D, Berthiaumey, et al. Maternal ciga-
rette smoking and fetal oxygen transport: a study of P50, 2,3-
extremely low-birthweight infant to the stable growing diphosphoglycerate, total hemoglobin, hematocrit, and type F he-
preterm infant. Studies are underway to explore the mech- moglobin in fetal blood. Pediatrics. 1983;2:22
anisms linking Hgb metabolism and the transfer of NO by 13. Matoth Y, Zaizove R, Varsano I. Postnatal changes in some red
erythrocytes, and these studies have the potential to add cell parameters. Acta Paediatr Scand. 1971;60:317
greatly to the body of evidence regarding transfusion guide- 14. Gonzales GF, Steenland K, Tapia V. Maternal hemoglobin
level and fetal outcome at low and high altitudes. Am J Physiol Regul
lines in various neonatal populations.
Integr Comp Physiol. 2009;297:R1477–R1485
15. Nelson SM, Freeman DJ, Sattar N, Lindsay RS. Erythrocytosis
in offspring of mothers with type 1 diabetes—are factors other than
insulin critical determinants? Diabet Med. 2009;26:887– 892
American Board of Pediatrics Neonatal-Perinatal 16. Weatherall DJ, Wood WG, Jones RW, Clegg JB. The develop-
Medicine Content Specifications mental genetics of human hemoglobin. In: Stamatoyannopoulos G,
• Know the biochemical characteristics of Nienhuis AW, eds. Experimental Approaches for the Study of Hemo-
fetal hemoglobin. globin Switching. New York, NY: Alan R Liss; 1985:3–25
• Know the developmental biology of 17. Papayannopoulou T, Kurachi S, Brice M, Nakamoto B, Stama-
hemoglobin types. toyannopoulos G. Asynchronous synthesis of HbF and HbA during
• Know normal erythropoiesis in the fetus erythroblast maturation. II. Studies of G gamma, A gamma, and
and neonate. beta chain synthesis in individual erythroid clones from neonatal
and adult BFU-E cultures. Blood. 1981;57:531
18. Kleihauer E. The hemoglobins. In: Stave U, ed. Physiology of the
Perinatal Period. Vol 1. New York, NY: Appleton-Century-Crofts;
ACKNOWLEDGMENTS. I wish to thank Rebecca Mo- 1970:255
ran, MD, and Andrea Duncan, MD, for their thoughtful 19. Masala B, Manca L, Formato M, Pilo G. A study of the switch
review and comments and Ann Chavez for her assistance of fetal hemoglobin and newborn erythrocytes fractionated by
in completing the manuscript. density gradient. Hemoglobin. 1983;7:567
20. Schröeder WA, Shelton JR, Shelton JB, Apell G, Huisman TH,
Bouver NG. Worldwide occurrence of nonallelic genes for the gamma-
References chain of human foetal haemoglobin in newborns. Nature. 1972;240:273
1. Forestier F, Daffos F, Catherine N, Renard M, Andreux JP. 21. Schröeder WA, Huisman THJ, Brown AK, et al. Postnatal
Developmental hematopoiesis in normal human fetal blood. Blood. changes in chemical heterogeneity of human fetal hemoglobin.
1991;77:2360 Pediatr Res. 1971;5:473
2. Bratteby L. Studies on the erythro-kinetics in infancy: red cell 22. Schröeder WA, Huisman THJ. Human gamma-chains: struc-
volume of newborn infants in relation to gestational age. Acta tural features. In: Stamatoyannopoulos G, Nienhuis AW, eds. Cel-
Paediatr Scand. 1968;57:132 lular and Molecular Regulation of Hemoglobin Switching. New
3. Jopling J, Henry E, Wiedmeier SE, Christensen RD. Reference York, NY: Grune & Stratton; 1979:29 – 45
ranges for hematocrit and blood hemoglobin concentration during 23. Szelengi JG, Holland SR. Studies on the structure of human
the neonatal period: data from a multihospital health care system. embryonic hemoglobin. Acta Biochim Biophys Acad Sci Hung. 1969;4:47
Pediatrics. 2009;123:e333– e337 24. Hecht F, Motulsky AG, Lemire RJ, Shepard TE. Predomi-
4. Alur P, Devapatla SS, Super DM, et al. Impact of race and nance of hemoglobin Gower 1 in early human embryonic develop-
gestational age on red blood cell indices in very low birth weight ment. Science. 1966;152:91
infants. Pediatrics. 2000;106:306 –310 25. Heizman THJ, Schroeder WA, Brown AK, Hyman CB, Or-
5. Usher R, Shephard M, Lind J. The blood volume of a newborn tega JA, Sukumaran PK. Further studies of the postnatal change in
infant and placental transfusion. Acta Paediatr Scand. 1963;52:497 chemical heterogeneity of human fetal hemoglobin in several ab-
6. Oski FA. The erythrocyte and its disorders. In: Oski FA, Nathan normal conditions. Pediatr Res. 1975;9:1

e36 NeoReviews Vol.12 No.1 January 2011


Downloaded from http://neoreviews.aappublications.org by Sreeram Subramanian on February 28, 2011
core concepts hemoglobin

26. Kleihauer E, Braun H, Betke K. Demonstration of fetal hemoglo- and diphosphoglycerate by human hemoglobin. Proc Natl Acad Sci
bin in the erythrocytes of a blood smear. Klin Wochenschr. 1957;35:637 U S A. 1968;59:526
27. Bard H, Lachance C, Widness JA, Gagnon C. The reactivation 35. Delivoria-Papadopoulos M, Roncevic NP, Oski FA. Postnatal
of fetal hemoglobin synthesis during anemia of prematurity. Pediar changes in oxygen transport of term, preterm and sick infants: the role
Res. 1994;36:253 of red cell 2,3 diphosphoglycerate in adult hemoglobin. Pediatr Res.
28. Bard H, Fouron JC, Gagnon C, Gagnon J. Hypoxemia and 1971;5:235
increased fetal hemoglobin synthesis. J Pediatr. 1994;124:941 36. Soubasi V, Kremenopoulos G, Tsantal C, Savopoulou P, Mus-
29. Bard H. The effect of placental insufficiency on fetal hemoglobin safiris C, Dimitriou M. Use of erythropoietin and its effects on blood
and adult hemoglobin synthesis. Am J Obstet Gynecol. 1974;120:67 lactate and 2, 3-diphosphoglycerate in premature neonates. Biol Neo-
30. Bard H, Prosmanne J. Relative rates of fetal hemoglobin and nate. 2000;78:281
adult hemoglobin synthesis in the cord blood of infants of insulin- 37. Debska-Slizień A, Owczarzak A, Lysiak-Szydlowska W, Rutkowski
dependent diabetic mothers. Pediatrics. 1985;75:1143 B. Erythrocyte metabolism during renal anemia treatment with recombi-
31. Perrine SP, Greene MF, Faller DV. Delay in fetal hemoglobin nant human erythropoietin. Int J Artif Organs. 2004;27:935–942
switch in infants of diabetic mothers. N Engl J Med. 1985;312:334 38. Stockman JA. Anemia of prematurity: current concepts in the
32. Platt OS, Brambilla DJ, Rosse WF, et al. Mortality in sickle cell issue of when to transfuse. Pediatr Clin North Am. 1986;33:111
disease. Life expectancy and risk factors for early death. N Engl 39. Novy MJ, Frigoletto FD, Easterday CL, Umansky I, Nelson NM.
J Med. 1994;330:1639 Changes in umbilical-cord blood oxygen affinity after intrauterine
33. Trompeter S, Roberts I. Haemoglobin F modulation in child- transfusions for erythroblastosis. N Engl J Med. 1971;285:589
hood sickle cell disease. Br J Haematol. 2009;144:308 –316 40. Oski FA, Stockman JA. Anaemia in early infancy. Br J Haema-
34. Benesch R, Benesch RE, Yu CL. Reciprocal binding of oxygen tol. 1974;27:195

NeoReviews Vol.12 No.1 January 2011 e37


Downloaded from http://neoreviews.aappublications.org by Sreeram Subramanian on February 28, 2011
core concepts hemoglobin

NeoReviews Quiz
9. Red blood cell production decreases after birth, primarily as a result of increased availability of oxygen,
which greatly reduces erythropoietin production and endogenous erythropoiesis. A decrease in hemoglobin
concentration follows this reduced red blood cell production. Of the following, the hemoglobin nadir in
healthy term infants is reached at a postnatal age closest to:
A. 8 weeks.
B. 12 weeks.
C. 16 weeks.
D. 20 weeks.
E. 24 weeks.

10. Hemoglobin consists of heme, an iron-containing protoporphyrin, and globin, a polypeptide. Eight globin
genes direct the synthesis of six different polypeptide chains, designated as alpha (␣), beta (␤), gamma
(␥), delta (␦), epsilon (␧), and zeta (␨). These globin chains combine in the developing erythroblast to
form seven different hemoglobin tetramers: Gower 1 (␨2-␧2), Gower 2 (␣2-␧2), Portland (␨2-␥2), fetal
hemoglobin (Hgb F: ␣2-␥2), adult hemoglobin (Hgb A: ␣2-␤2), and adult hemoglobin A2 (Hgb A2: ␣2-␦2).
Of the following, the most prevalent hemoglobin tetramer in the fetus at 18 weeks of gestational age is:
A. Gower 1.
B. Hgb A.
C. Hgb A2.
D. Hgb F.
E. Portland.

11. A term infant is born with severe anemia. A Kleihauer Betke test is performed on maternal blood to
determine whether fetomaternal hemorrhage is the cause. Of the following, the property of fetal
hemoglobin that best differentiates fetal from maternal red blood cells using the Kleihauer Betke test is
that the fetal hemoglobin, relative to adult hemoglobin, is/has:
A. Decreased interaction with 2,3-diphosphoglycerate.
B. Greater affinity for oxygen.
C. Greater solubility in strong phosphate buffer.
D. Readily oxidized to methemoglobin.
E. Resistant to acid elution.

e38 NeoReviews Vol.12 No.1 January 2011


Downloaded from http://neoreviews.aappublications.org by Sreeram Subramanian on February 28, 2011
Core Concepts: The Biology of Hemoglobin
Robin K. Ohls
NeoReviews 2011;12;e29-e38
DOI: 10.1542/neo.12-1-e29

Updated Information including high-resolution figures, can be found at:


& Services http://neoreviews.aappublications.org/cgi/content/full/neoreview
s;12/1/e29
Permissions & Licensing Information about reproducing this article in parts (figures,
tables) or in its entirety can be found online at:
http://neoreviews.aappublications.org/misc/Permissions.shtml
Reprints Information about ordering reprints can be found online:
http://neoreviews.aappublications.org/misc/reprints.shtml

Downloaded from http://neoreviews.aappublications.org by Sreeram Subramanian on February 28, 2011

Anda mungkin juga menyukai