Sie sind auf Seite 1von 12

Biochemical Basis of Hypoxic-Ischemic Encephalopathy

Maria Delivoria-Papadopoulos and Peter J. Marro


NeoReviews 2010;11;e184-e193
DOI: 10.1542/neo.11-4-e184

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;11/4/e184

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 © 2010 by the American Academy of Pediatrics. All rights reserved.
Online ISSN: 1526-9906.

Downloaded from http://neoreviews.aappublications.org. Provided by USUHS LRC on July 18, 2010


Article neurology

Biochemical Basis of Hypoxic-Ischemic


Encephalopathy
Maria Delivoria-
Papadopoulos, MD,*
Objectives After completing this article, readers should be able to:
Peter J. Marro, MD† 1. Describe the steps of posthypoxic brain injury.
2. Explain the role of N-methyl-D-aspartate (NMDA) receptors in hypoxia.
3. Delineate the contribution of free radicals to neuronal injury during hypoxia.
Author Disclosure
4. Explain how hypoxia affects calcium influx and modification of apoptotic proteins.
Drs Delivoria-
Papadopoulos and
Marro have disclosed Abstract
no financial Despite improved methods of intrapartum monitoring and advances in neonatal care
and treatment, neonatal hypoxic-ischemic injury continues to produce significant
relationships relevant
morbidity and mortality, often leading to long-term neurologic consequences. Hyp-
to this article. This
oxia creates an imbalance in metabolic demand and cellular energy supply, resulting
commentary does not in the disruption of critical cellular functions and the activation of excitatory neuro-
contain a discussion transmitters. In addition, the structure, function, and modification of cellular pro-
of an unapproved/ cesses, such as the N-methyl-D-aspartate (NMDA) receptor and intracellular calcium
investigative use of a regulation, are affected. Nuclear calcium signals control critical nuclear functions,
including regulation of transcription factors and cell cycle, gene transcription, DNA
commercial
replication, and nuclear envelope breakdown. Nitric oxide synthase and the genera-
product/device.
tion of nitric oxide during hypoxia may contribute significantly to altered cell
function, disruption in calcium homeostasis, and the activation of caspases, leading to
programmed cell death. The biochemical mechanisms involved in hypoxic-ischemic
neuronal injury and death are exceedingly complex and interdependent. This discus-
sion focuses primarily on some of the major cellular and molecular mechanisms of
hypoxic neuronal injury in the newborn brain.

Introduction
Perinatal hypoxia-ischemia is the most common cause of
neurologic disease during the neonatal period. Hypoxic-
Abbreviations ischemic encephalopathy (HIE) is associated with high mor-
tality and morbidity rates, including cerebral palsy, intellec-
Apaf-1: apoptotic protease activation factor
tual disability, and seizures. The incidence of perinatal
ATP: adenosine triphosphate
asphyxia is about 1.0% to 1.5% in most centers and usually is
CaM kinase: calcium/calmodulin kinase
related to gestational age and birthweight. It occurs in 9.0%
CNS: central nervous system
of infants younger than 36 weeks’ gestation and in 0.5% of
CREB: cAMP response element binding
infants older than 36 weeks’ gestation. (1) Perinatal HIE
cyt c: cytochrome C
may develop during the antepartum (20%), intrapartum
HIE: hypoxic-ischemic encephalopathy
(30%), antepartum and intrapartum (35%), or postpartum
ICAD: inhibitor of caspase-activated DNase
(10%) period. (2) HIE develops in the setting of perinatal
IP3: inositol triphosphate
asphyxia, which is a multiorgan system disease and includes
NMDA: N-methyl-D-aspartate
circumstances affecting the cerebral blood flow in the fetus
NNLA: Nw-nitro-l-arginine
and newborn that compromise the supply of oxygen to the
nNOS: neuronal nitric oxide synthase
brain. Assessment and management of these complications is
NO: nitric oxide
an integral part of the treatment of perinatal asphyxia/HIE.
NOS: nitric oxide synthase
(3)
PARP: poly-ADP-ribose-polymerase
A large amount of information has been collected on the

*Professor of Pediatrics and Physiology Emeritus, University of Pennsylvania School of Medicine, Philadelphia, Pa.

Division of Neonatology, Barbara Bush Children’s Hospital at Maine Medical Center, Portland, Me.

e184 NeoReviews Vol.11 No.4 April 2010


Downloaded from http://neoreviews.aappublications.org. Provided by USUHS LRC on July 18, 2010
neurology hypoxic-ischemic encephalopathy

fetal cardiovascular and respiratory response to oxygen


limitations, giving rise to a better understanding and
management of neonatal deterioration induced by hyp-
oxia. Besides these physiologic studies, cellular and bio-
chemical mechanisms are being explored increasingly to
investigate the complex and interrelated biochemical
alterations that result in hypoxic brain injury and brain
cell death in the fetus and newborn. (4)(5) It is important
to recognize the factors that may determine the sus-
ceptibility of the developing brain to neonatal and peri-
natal hypoxia. These determinants include the lipid com-
position of the brain cell membrane, the rate of lipid
peroxidation, the presence of antioxidant defenses, the
development and modulation of the excitatory neuro-
transmitter receptors such as the NMDA receptor, and
the intracellular calcium influx mechanisms. In addition
to the developmental status of these cellular compo-
nents, the response of the potential mechanisms to hyp-
oxia determines the fate of the hypoxic brain cell in the
developing brain of the fetus and the newborn.

Steps of Posthypoxic Brain Injury


Energy Breakdown
Under normal conditions, oxidative phosphorylation
and the synthesis of high-energy phosphates such as
adenosine triphosphate (ATP) requires adequate oxygen
supply. Under anaerobic conditions, the metabolic cost
of ATP production is critically increased, leading to a
breakdown of energy balance that depletes brain cells of
high-energy compounds necessary for energy-dependent Figure 1. Effects of hypoxia leading to increased intranuclear
metabolic processes in neurons and glial cells. calcium. ATPⴝadenosine triphosphate, NMDAⴝN-methyl-D-
aspartate
Excitotoxic Mechanisms (Fig. 1)
The decrease in ATP concentrations leads to cell mem- ergy loss; generalized disruption of internal homeostasis
brane depolarization and a disruption of voltage- leading to eventual lysis of the nucleus, organelles, and
dependent ion channels, which allows excessive amounts plasma cell membranes; and the release of intracellular
of calcium to enter the cytosol, initiating the release of components that induce a local inflammatory response.
glutamate that activates NMDA receptors. The increased The result is edema and injury to neighboring cells. The
expression/activation of NMDA receptors further en- inflammatory response triggers expression of the cyto-
hances cellular calcium influx. The entire mechanism is kines interleukin-1-beta and tumor necrosis factor-alpha,
accelerated further by the dysfunction of the energy- which stimulate release of oxygen free radicals from
dependent reuptake of glutamate both in neurons and neutrophils that activate microglial cells.
in astrocytes in a vicious cycle. When calcium influx is Necrosis is only one of the mechanisms of cell death
unregulated, activation of phospholipases, proteases, and following hypoxia or ischemia to the brain. Programmed
endonucleases can result in nuclear, organelle, and cell cell death or apoptosis also appears to contribute to cell
membrane disruption. death following hypoxia-ischemia, especially cell death
that occurs days to weeks following the insult. (6)(7)
Cell Death (Fig. 2) Apoptosis is an active process that requires activation of a
Acute or long-term consequences of HIE are related genetic program and specific endonucleolytic digestion
either to necrosis or to apoptosis of neuronal cells. Ne- of nuclear DNA. In contrast to necrosis, programmed
crosis is characterized by passive cell swelling; rapid en- cell death is characterized by cell shrinkage, coarse chro-

NeoReviews Vol.11 No.4 April 2010 e185


Downloaded from http://neoreviews.aappublications.org. Provided by USUHS LRC on July 18, 2010
neurology hypoxic-ischemic encephalopathy

diated through its interaction with specific cell mem-


brane receptors, of which the NMDA, kainate, and
AMPA subtypes are the best characterized. (16)
The NMDA-type glutamate receptor is a predomi-
nant mediator of excitotoxicity in the immature brain
compared with the adult brain due to overexpression of
the receptor in the developing immature CNS. (17)
Within the developmental period, NMDA receptor-
mediated processes may depend not only on the ontog-
eny of the NMDA receptor, but also potential modifica-
tion by intracellular mechanisms.

Structure and Function of the NMDA Receptor


The activity of the NMDA receptor-ion channel complex
is regulated by a number of pharmacologically distinct
binding sites. The NMDA receptor possesses a neuro-
transmitter binding site, or recognition site, that binds
glutamate or NMDA; a coactivator site that binds gly-
cine; a channel site that binds MK-801; a voltage-
dependent magnesium-binding site; a polyamine site; an
ifenprodil site; and an inhibitory divalent cation site that
binds zinc. (17) Ligand binding studies indicate two
distinct binding sites or states associated with the gluta-
mate recognition site, one that preferentially binds ago-
Figure 2. Pathways of neuronal cell death following initial
nists and one that preferentially binds antagonists. (18)
energy failure. NMDAⴝN-methyl-D-aspartate. Reprinted
with permission from Marro PJ. The etiology and pharmaco-
The NMDA receptor is associated with a cation-
logic approach to hypoxic-ischemic encephalopathy in the selective ion channel that gates sodium, potassium, and
newborn. NeoReviews. 2002;3:e99. calcium ions. In the resting state, when blocked by
magnesium in a voltage-dependent manner, (19)(20)
blockade of the ion channel complex by glutamate or
matin aggregation with extensive nuclear DNA fragmen- NMDA is allowed, and agonist-dependent calcium in-
tation, nuclear pyknosis, and extrusion of membrane- flux occurs. The influx of calcium ions is believed to
bound cytoplasmic fragments or apoptotic bodies, but it initiate biochemical processes responsible for both
is not associated with lysis of the plasma membrane. NMDA receptor-induced plasticity in the developing
(8)(9) Studies in cell culture models have demonstrated brain and NMDA receptor-mediated excitotoxic cell
that hypoxia can trigger programmed cell death. (10) death. (15)(21)(22)
Programmed cell death, as assessed by the cleavage of Each of the regulatory sites of the NMDA receptor-
genomic DNA, also has been shown to occur in the brain ion channel complex is modified during brain develop-
following focal (6)(11)(12) and global ischemia. (7)(13) ment, which may play a role in altering the response of
The mechanism by which hypoxia causes DNA fragmen- the receptor during development and hypoxia.
tation has been studied extensively but is not well under-
stood. Mechanism of NMDA Receptor Modification
During Hypoxia
NMDA Receptors Brain tissue hypoxia modifies the NMDA receptor rec-
Glutamate is the primary excitatory amino acid neuro- ognition, coactivator, and ion channel sites. A decrease in
transmitter that contributes to a number of essential the apparent number of NMDA receptors and an in-
developmental processes such as synaptogenesis, synaptic crease in receptor affinity for MK-801 were observed in
plasticity, long-term potentiation, learning, and memory hypoxic fetal guinea pig and newborn piglet brains.
as well as neurodegeneration and hypoxia-induced in- (23)(24) In these same studies, glutamate- and glycine-
jury. (14)(15) The physiologic and pathologic effects of dependent activation of the NMDA receptor was de-
glutamate in the central nervous system (CNS) are me- creased and spermine-dependent and basal state receptor

e186 NeoReviews Vol.11 No.4 April 2010


Downloaded from http://neoreviews.aappublications.org. Provided by USUHS LRC on July 18, 2010
neurology hypoxic-ischemic encephalopathy

activation were increased during hypoxia. Hypoxia- nitric oxide synthase (NOS) pathway plays a critical role
induced modification of the recognition, coactivator, in the NO-mediated mechanism of hypoxia-induced
and modulatory sites of the NMDA receptor-ion channel modification of the NMDA receptor complex in the
complex is likely through nitric oxide (NO)-mediated newborn brain.
nitration. In neurons of the CNS, neuronal nitric oxide
synthase (nNOS) is colocalized with the NMDA recep- Free Radicals
tor, (25)(26) thereby favoring nitration of the receptor. Free radicals are molecular species that have unpaired
In addition, nNOS activity is decreased by phosphoryla- electrons in the outer orbit with a strong tendency to
tion and increased by dephosphorylation, (27)(28) a initiate chain reactions that result in membrane peroxi-
condition likely present during hypoxia. Dephosphory- dation, protein oxidation, nucleic acid oxidation, and cell
lation of the receptor also makes tyrosine sites available damage. Normally, more than 80% of the oxygen con-
for nitration by peroxynitrite, which is produced by NO sumed by the cell is reduced completely by cytochrome
and superoxide radicals, both of which are produced oxidase to water without production of oxygen free
during hypoxia. Thus, dephosphorylation during hyp- radicals. The remaining 10% to 20% undergoes other
oxia may facilitate peroxynitrite-mediated nitration of oxidation reduction reactions in the cytoplasm and mi-
tyrosine-inhibiting phosphorylation of proteins and al- tochondria that produce a superoxide anion radical.
teration of the NMDA receptor-ion channel complex.
(29) Mechanisms of Free Radical Generation During
In neurons of the CNS, nNOS is activated by calcium Hypoxia
influx through the NMDA receptor-ion channel, but There are a number of potential mechanisms of free
nNOS is not efficiently stimulated by activation of non- radical generation under hypoxic conditions. During
NMDA receptors that also induce calcium influx. (30) hypoxia, the increased accumulation of intracellular cal-
The synaptic localization of nNOS in the brain may be cium resulting from excessive activation of NMDA (32)
mediated by the postsynaptic density protein PSD-95. and non-NMDA receptors is crucial in hypoxia-induced
Recently, it was demonstrated that nNOS, PSD-95, and excitotoxicity. Increased intracellular calcium can initiate
NMDA receptor subunit NR2B from the brain coimmu- a number of biochemical events that could lead to free
noprecipitate and that the PSD-95 is sufficient to assem- radical generation and cell death (Table 1).
ble a tight tertiary complex with nNOS and the NR2B In addition to calcium mediation, other potential
subunit of the NMDA receptor. (31) mechanisms of free radical generation during hypoxia
In summary, results of these studies indicate that NO include: 1) reduction of electron transport chain com-
production in the brain is preferentially activated by ponents, including ubiquinone (a component that un-
calcium influx through the NMDA receptor-ion channel, dergoes auto-oxidation to produce free radicals); 2) in-
that there is a specific structural and functional link creased release of ferritin under the conditions of
between the NMDA receptor and nNOS, and that the decreased cellular high-energy compounds; and 3) in-

Table 1. Deleterious Effects of Increased Intracellular Calcium


Action Result
Activates phospholipases ● Phospholipid hydrolysis and cell membrane injury
● Generation of free radicals
Activates proteases ● Cytoskeletal disruption of microtubules
● Proteolysis of other cellular proteins
Activates nucleases ● Nuclear injury
Activates calcium-ATPase ● Further increase in cytosolic and nuclear calcium
● Consumes ATP at time of energy depletion
Enters mitochondrion and uncouples oxidative phosphorylation ● Decreased ATP production
Increases neurotransmitter release ● Activation of glutamate receptors and calcium influx
Activates protein that transforms xanthine dehydrogenase ● Free radical formation
to xanthine oxide
Activates nitric oxide synthase ● Generation of nitric oxide
ATP⫽adenosine triphosphate

NeoReviews Vol.11 No.4 April 2010 e187


Downloaded from http://neoreviews.aappublications.org. Provided by USUHS LRC on July 18, 2010
neurology hypoxic-ischemic encephalopathy

creased degradation of ATP during hypoxia, increasing The exact molecular mechanism of hypoxic mem-
the substrate for the xanthine oxidase reaction and lead- brane damage is not clear. However, it has been shown
ing to increased free radical generation. that peroxynitrite (formed by the reaction between su-
Increased free radical generation during hypoxia in peroxide anions and NO) can cause lipid peroxidation in
the cerebral cortex of newborn piglets has been docu- vitro. (40) Therefore, high concentrations of NO during
mented directly through spin trapping of free radicals hypoxia may result in increased production of peroxyni-
and measuring the resulting spin adduct signal with trite, causing lipid peroxidation.
electron spin resonance spectroscopy. The characteristics
of the spin adduct signal identify the free radical species Neuronal Nuclear Calcium Influx
present in the hypoxic tissue as predominantly an alkoxyl A number of critical nuclear functions, including regula-
radical, indicating that free radical-mediated lipid peroxi- tion of transcription factors, cell cycle regulation tran-
dation is an ongoing event during cerebral hypoxia, a scription, DNA replication, and nuclear envelope break-
mechanism of hypoxic neuronal injury. These studies not down, are controlled by intracellular calcium (Table 2).
only demonstrate increased free radical generation dur- (41) Furthermore, nuclear calcium signals potentially
ing hypoxia in the cerebral cortex of the fetus and the control a number of events leading to hypoxia-induced
newborn but reduced hypoxia-induced production of programmed cell death. Nuclear and cytosolic calcium
free radical species due to inhibitors of pathways of free signals are regulated differently, and the extranuclear
radical generation. calcium concentration determines the mode of calcium
entry into the nucleus. Increased intracellular calcium
Nitric Oxide Free Radicals and Neuronal Injury has been shown to be a primary mediator of activity-
Three major isoforms of NOS have been identified: dependent gene transcription under a number of exper-
constitutive neuronal, constitutive endothelial, and in- imental conditions. (42)(43)(44) Several factors may be
ducible macrophage isoforms. Following ischemia, NO involved in calcium-regulated gene expression and tran-
produced from neuronal NOS has toxic effects, but NO scription, including the site of calcium entry, the ampli-
produced from endothelial NOS has protective effects in tude and the spatial properties of the calcium signals, and
the brain. (33) Hypoxic brain injury is associated with the duration of the calcium signal. (45)(46)
the formation of NO. (34)(35) Although NO physiolog- Studies have shown that cerebral hypoxia results in
ically mediates cerebral vasodilation under normal con- increased nuclear calcium influx in neuronal nuclei of the
ditions, (36) recent studies suggest that NO, a gaseous cerebral cortex of newborn guinea pigs. (47) The nuclear
free radical, may react with superoxide anion to form calcium influx increased as a function of increased cere-
peroxynitrite and cause neurotoxicity. (37)(38)(39) bral tissue hypoxia. Cerebral hypoxia resulted in in-
NO is reported to cause neuronal damage through creased calcium/calmodulin kinase (CaM kinase) IV ac-
various mechanisms. Studies using the NOS inhibitor tivity, cAMP response element binding (CREB) protein
Nw-nitro-l-arginine (NNLA) demonstrated that free phosphorylation, activity of high-affinity Ca⫹⫹-ATPase,
radicals, corresponding to alkoxyl radicals, were induced and inositol triphosphate (IP3)-dependent calcium in-
by hypoxia but were inhibited by pretreatment with flux in neuronal nuclei of newborn piglets. In addition,
NNLA. NNLA also inhibited hypoxia-induced genera- NO donors increased neuronal nuclear calcium influx,
tion of conjugated dienes (products of lipid peroxida- (42) and hypoxia resulted in generation of NO free
tion) and preserved Na⫹, K⫹-ATPase activity (an index radicals and increased high-affinity Ca⫹⫹-ATPase activ-
of cellular membrane function). These data demon- ity.
strated that NOS generates free radicals during hypoxia During hypoxia, NO-mediated modification of the
via peroxynitrite production, presumably causing lipid
peroxidation and membrane dysfunction. The produc-
tion of secondarily formed lipid free radicals provides
strong evidence of peroxidative injury. This is particularly
Role of Calcium Inside
Table 2.

true for alkoxyl radicals, which are generated from lipid the Nucleus
peroxide by either iron or copper ions and can abstract
hydrogen atoms from polyunsaturated fatty acids, lead- ● Transcription of genes
● Regulation of the cell cycle
ing to further lipid peroxidation. This suggests that NO
● Replication of DNA
has an in vivo role in the generation of alkoxyl radicals, ● Breakdown of the nuclear envelope
leading to free radical-mediated lipid peroxidation.

e188 NeoReviews Vol.11 No.4 April 2010


Downloaded from http://neoreviews.aappublications.org. Provided by USUHS LRC on July 18, 2010
neurology hypoxic-ischemic encephalopathy

nuclear membrane high-affinity Ca⫹⫹-ATPase and IP3 way and the mitochondria-initiated pathway. The re-
receptor is a potential mechanism of increased intranu- cruitment and cleavage of procaspase-8 to produce the
clear calcium that leads to activation of calcium- active form of caspase-8 is a critical biochemical event in
dependent nuclear mechanisms and activates cascades of the death receptor-mediated apoptosis. Following its
hypoxic programmed cell death. activation, caspase-8 can activate downstream caspases
by direct cleavage or by indirectly cleaving the proapop-
Expression and Posttranslational totic protein and inducing cytochrome c (cyt c) release
Modification of Apoptotic Proteins from the mitochondria. In the mitochondria-initiated
The Bcl-2 Proteins pathway, caspase activation is triggered by formation
The Bcl-2 family of proteins (including Bax and Bcl- of an oligomeric apoptotic protease activation factor
2) control cell proliferation, differentiation, and pro- (Apaf-1)/cyt c complex. The resulting complex formed
grammed cell death during normal brain development. by the combination of Apaf-1, cyt c, Bax/Bcl-2, and
(48)(49) Bax and Bcl-2 are inducible genes found in the procaspase-9 is referred to as the apoptosome, which
developing and adult central and peripheral nervous leads to recruiting and activating procaspase-9, an up-
systems. (50)(51) The antiapoptotic protein Bcl-2 pre- stream caspase. Other, less-defined pathways of apopto-
vents apoptosis by forming a heterodimer with the pro- tic caspase activation that also are active in neurons act
apoptotic protein Bax and protects cells from pro- predominantly through the caspases-8 or -9. Following
grammed cell death following hypoxia. (48)(49) hypoxia in the cerebral cortex of newborn piglets, there is
Cerebral hypoxia results in increased expression of an increase in protein expression and activity of caspase-
Bax protein in neuronal nuclei of the cerebral cortex of 8 and caspase-9 (initiator caspases) as well as the expres-
newborn piglets. (52) Increased Bax protein in the mi- sion and activity of caspase-3 (executioner caspase).
tochondrial, cytosolic, and neuronal nuclear fractions
indicates increased expression of the protein rather than Nuclear Calcium Influx and Caspase-9 and
its translocation from mitochondria to cytosol during Caspase-3 Activation
hypoxia. The expression of antiapoptotic protein Bcl-2 Increased activity of casapse-9 and caspase-3 during
did not increase during hypoxia. Therefore, the ratio of hypoxia is mediated by nuclear calcium influx. Studies
proapoptotic protein Bax to antiapoptotic protein Bcl-2 investigating the role of nuclear calcium influx in caspase
increases in all compartments of the cell during hypoxia, activation during hypoxia were performed in newborn
which may lead to activation of the hypoxia-induced piglets. (55)(56) The calcium-ATPase inhibitor (cloni-
cascade of neuronal death. NOS inhibition prevented the dine) was administered to block nuclear calcium influx.
hypoxia-induced increased expression of proapoptotic Cerebral tissue hypoxia resulted in increased caspase-9
protein Bax, indicating that the hypoxia-induced in- activity, and pretreatment with clonidine prevented this
creased expression of Bax is mediated by NO. (53) hypoxia-induced increase in caspase-9 activity. In addi-
tion, hypoxia resulted in increased caspase-3 activity, a
Caspases-3, -8, and -9 consequence of caspase-9 activation, and pretreatment
Caspases are a unique family of cysteinyl-aspartate pro- with clonidine prevented this increase in caspase-3 activ-
teases that play an important role in the initiation and ity. These results demonstrate that hypoxia-induced in-
execution of apoptosis. (54) They are divided into two crease in caspase-3 activity is mediated by nuclear calcium
primary classes: Class I caspases have long prodomain influx.
and Class II caspases have short prodomain.
Class I caspases such as 8, 9, and 10 can autocatalyze NO and Caspase-9 and Caspase-3 Activation
their own activation and are activated in the early phase To delineate caspase activation during hypoxia further,
of apoptosis. These are termed the initiator caspases. the role of NO derived from neuronal NOS was studied
Class II caspases, such as 3, 6, and 7, require cleavage in newborn piglets, using a relatively selective inhibitor
by another protease and are responsible for the break- of nNOS, 7-NINA salt. (44) Following cerebral tissue
down of cells. These are termed the effector or the hypoxia, an increase in caspase-9 activity was seen, but
executioner caspases. Upstream caspase activation dur- pretreatment with the nNOS inhibitor prevented the
ing apoptosis leads to the activation of downstream hypoxia-induced increase in caspase-9 activity. Cerebral
caspases in a self-amplifying cascade. tissue hypoxia again resulted in increased caspase-3 activ-
Two pathways of caspase activation have been inves- ity, a consequence of caspase-9 activation, and pre-
tigated: the cell surface death receptor-mediated path- treatment with the nNOS inhibitor prevented this

NeoReviews Vol.11 No.4 April 2010 e189


Downloaded from http://neoreviews.aappublications.org. Provided by USUHS LRC on July 18, 2010
neurology hypoxic-ischemic encephalopathy

hypoxia-induced increase in caspase-3 activity. These Clinical Implications


results demonstrate that the hypoxia-induced increase in Understanding the very complex and interrelated mech-
caspase-9 is mediated by nNOS-derived NO. anisms of cell death after a hypoxic-ischemic result may
Caspase-9 can be activated during hypoxia by multi- serve as background for critical care of the newborn.
ple mechanisms that are dependent on generation of Hypoxia at the cellular level results from failure in oxygen
nNOS-derived NO and neuronal nuclear calcium influx. transport from the lung alveolar space to the mitochon-
NO increases calcium influx in synaptosomes as well as dria. To prevent neuronal cell death, any insufficiency or
neuronal nuclei. By increasing nuclear calcium influx, failure in the respiratory and circulatory systems must be
NO can increase expression of caspase-9 as well as pro- restored in a matter of minutes. Among the hazards in
apoptotic proteins. NO-mediated protein modification restoring oxygen supply is medically induced hyperoxia
also may alter its activation. Caspase-9 plays a significant (high FiO2), which may worsen the neuronal insult by
role in the hypoxia-induced programmed cell death in producing additional oxygen free radicals. An under-
the newborn brain, and caspase activation during hyp- standing of the temporal evolution of the posthypoxic
oxia in the newborn brain is mediated by both biochemical disturbances during and following hypoxic-
transcription-dependent and -independent mechanisms. ischemic insult may offer the opportunity for pharmaco-
(57) logic interventions at key steps of the biochemical events.

DNA Fragmentation American Board of Pediatrics Neonatal-Perinatal


It has been proposed that the cleavage of DNA at its Medicine Content Specification
intranucleosomal linkage region is produced by specific • Know the incidence, causes and
endonucleases that are dependent on calcium. (58)(59) pathophysiology, including cellular
Caspase-3, acting as cysteine protease, cleaves and inac- abnormalities, of acute perinatal asphyxia.
tivates nuclear enzymes, including poly-ADP-ribose-
polymerase (PARP), a DNA repair enzyme, and inhibitor
of caspase-activated DNase (ICAD). The caspase-
activated DNase then enters the nucleus and cleaves
genomic chromosomal DNA. (60)(61) This nuclear
References
1. Hill A, Volpe J. Hypoxic-ischemic cerebral injury in the new-
genomic DNA fragmentation correlates exponentially born. In: Swaiman KF, Ashwal S, eds. Pediatric Neurology: Princi-
with the degree of cerebral tissue hypoxia in newborn ples and Practice. St. Louis, Mo: Mosby; 1999:191–204
piglets (62) and is characteristic of cellular apoptosis. 2. Raichle ME. The pathophysiology of brain ischemia. Ann Neu-
rol. 1983;13:2–10
3. Piazza AJ. Postasphyxial management of the newborn. Clin
Perinatol. 1983;26:749 –765
Temporal Biochemical Changes 4. Vannucci RC. Experimental biology of cerebral hypoxia-isch-
The steps of posthypoxic neuronal injury evolve within emia: relation to perinatal brain damage. Pediatr Res. 1990;27:
hours (the necrotic process) and days (the apoptotic 317–326
process). Temporal biochemical changes and associated 5. Legido A, Katsetos CD, Mishra OP, et al. Perinatal hypoxia-
ischemia encephalopathy: current and future treatments. Int Pedi-
nuclear fragmentation have been assessed in the cerebral
atr. 2001;15:143–151
cortex of newborn guinea pigs following hypoxia. Initial 6. Linnik MD, Zobirst RH, Hatfield MD. Evidence supporting a
cellular injury may be followed by a failure of cellular role for programmed cell death in focal cerebral ischemia in rats.
repair mechanisms, leading to further delayed brain in- Strokes. 1993;24:2002–2008
jury. Neuronal nuclear calcium influx increases immedi- 7. Ferrer I, Tortosa A, Macaya A, et al. Evidence of nuclear DNA
fragmentation following hypoxia-ischemia in the infant rat brain,
ately following hypoxia and remains elevated through
and transient forebrain ischemia in the adult gerbil. Brain Pathol.
7 days of age. Similarly, nuclear Bax protein expression 1994;4:115–122
increases immediately following hypoxia and remains 8. Wylie AH, Kerr JFR, Currie AR. Cell death, the significance of
elevated through 7 days, but Bcl-2 protein remains sim- apoptosis. Int Rev Cytol. 1980;68:251–306
ilar to control during hypoxia. These biphasic temporal 9. Columbano A. Cell death: current difficulties in discriminating
apoptosis and necrosis in the context of pathological processes in
changes may reflect not only the primary hypoxic insult
vivo. J Cell Biochem. 1995;58:181–190
but also a secondary cellular damage due to recurrent 10. Rosenbaum DM, Michaelson M, Batter DK, et al. Evidence for
(continuing) free radical release during the reperfusion– hypoxia induced programmed cell death of cultured neurons. Ann
reoxygenation phase. Neurol. 1994;25:19 –33

e190 NeoReviews Vol.11 No.4 April 2010


Downloaded from http://neoreviews.aappublications.org. Provided by USUHS LRC on July 18, 2010
neurology hypoxic-ischemic encephalopathy

11. Dragunow M, Beiharz E, Sirimanne E, etal. Immediately early induced protein modification on tyrosine phosphorylation and deg-
gene protein expression in neurons undergoing delayed death, but radation. FEBS Lett. 1996;385:63– 66
not necrosis following hypoxic-ischemic injury to the young rat 30. Kiedrowski I, Costa E, Wroblewski JT. Glutamate receptor
brain. Brain Res Mol Brain Res. 1994;25:1933 agonist stimulate nitric oxide synthase in primary cultures of cere-
12. Gillardon F, Lenz C, Waschle KF. Altered expression of Bcl-2, bellar granule cells. J Neuroch. 1992;58:335–341
Bcl-X, Bax and c-Fos colocalizes with DNA fragmentation and 31. Christopherson KS, Hillier BJ, Lim WAS, et al. PSD-95 assem-
ischemic cell damage following middle cerebral artery occlusion in bles a ternary complex with the N-methyl-D-aspartic acid receptor
rats. Brain Res Mol Brain Res. 1996;40:254 –260 and bivalent neuronal NO synthase PDX domain. J Biol Chem.
13. Nitatori T, Sato N, Waguri S, et al. Delayed neuronal death in 1999;274:27467–27473
the CA1 pyramidal layer of the gerbil hippocampus following 32. Zanelli SA. NMDA receptor-mediated calcium influx in cere-
transient ischemia in apoptsis. J Neurosci. 1995;15:1001–1011 bral cortical synaptosomes of the hypoxic guinea pig fetus. Neuro-
14. Choi DW. Cerebral hypoxia: some new approaches and unan- chem Res. 1999;24:434 – 446
swered questions. J Neurosci. 1990;10:2493–2501 33. Huang Z. Effects of cerebral ischemia in mice deficient in
15. Rothman SM, Olney JW. Glutamate and the pathophysiology neuronal nitric oxide. Science. 1994;265:1883–1885
of hypoxic-ischemic brain damage. Anne Neurol. 1986;19:105–111 34. Beckman JS. The double-edged role of nitric oxide in brain
16. Monaghan DT, Bridges RJ, Cotman CW. The excitatory function and superoxide-mediated injury. J Dev Physiol. 1991;15:
amino acid receptors: their classes, pharmacology, and distinct 53–59
properties in the function of the central nervous system. Annu Rev 35. Cazevielle C. Superoxide and nitric oxide cooperation in
Pharmacol Toxicol. 1989;29:365– 402 hypoxia/reoxygenation-induced neuron injury. Free Radic Biol
17. Johnston MV. Neurotransmitters and vulnerability of the de- Med. 1993;14:359 –395
veloping brain. Brain Dev. 1995;17:301–306 36. Faraci FM. Role of endothelium-derived relaxing factor in
18. Monaghan DT, Olvenman HJ, Nguyen L, et al. Two classes of cerebral circulation: large arteries vs. microcirculation. Am J Physiol.
N-methyl-D-aspartate recognition sites: differential distribution 1991;261:H1038 –H1042
and differential regulation by glycine. Proc Natl Acad Sci USA. 37. Beckman JS. Apparent hydroxyl radical production by per-
1988;85:9836 –9840 oxynitrite: implications for endothelial injury from nitric oxide and
19. Nowak L, Bregetovski P, Ascher P, et al. Magnesium gates superoxide. Proc Natl Acad Sci USA. 1990;87:1620 –1624
glutamate-activated channels in mouse central neurons. Nature. 38. Dawson VL. Nitric oxide mediates glutamate neurotoxicity in
1984;307:462– 465 primary cortical cultures. Proc Natl Acad Sci USA. 1991;88:
20. Mayer ML, Westbrook GL, Guthrie PB. Voltage-dependent 6368 – 6371
block by Mg⫹⫹ of NMDA responses in spinal cord neurons. Na- 39. Hamada Y. Inhibitors of nitric oxide synthesis reduce hypoxic-
ture. 1984;309:261–263 ischemic brain damage in the neonatal rat. Pediatr Res. 1994;35:
21. Collingridge G. Synaptic plasticity. The role of NMDA recep- 10 –14
tors in learning and memory. Nature. 1987;330:604 – 605 40. Radi R. Peroxynitrite-induced membrane lipid peroxidation:
22. Tang YP, Shimizu E, Dube GR, et al. Genetic enhancement of the cytotoxic potential of superoxide and nitric oxide. Arch Biochem
learning and memory in mice. Nature. 1999;401:63– 69 Biophys. 1991;288:481– 487
23. Mishra OP, Delivoria-Papadopoulos M. NMDA receptor 41. Delivoria-Papadopoulos M, Akhter W, Mishra OP. Hypoxia-
modification of the fetal guinea pig brain during hypoxia. Neuro- induced Ca⫹⫹ -influx in cerebral cortical neuronal nuclei of new-
chem Res. 1992;17:1211–1216 born piglets. Neurosci Lett. 2003;342:119
24. Hoffman DJ, DiGiacomo JE, Marro PJ, et al. Hypoxia- 42. Ghosh A, Greenberg ME. Calcium signaling in neurons: mo-
induced modification of the N-methyl-D-aspartate (NMDA) recep- lecular and cellular consequences. Science. 1995;268:239 –247
tor in the brain of newborn piglets. Neurosci Lett. 1994;167: 43. Hardigham GE, Bading H. Nuclear calcium: a key regulator of
156 –160 gene expression. Biometals. 1998;11:345
25. Bhat GK, Mahesh VB, Lamar CA, et al. Histochemical local- 44. Chawla S, Bading H. CREB/CBP and SRE-interacting tran-
ization of nitric oxide neurons in the hypothalamus: association scriptional regulators are fast on-off switches: duration of calcium
with gonadotropin-releasing hormone neurons and co-localization transients specifies the magnitude of transcriptional responses.
with N-methyl-D-aspartate receptors. Neuroendocrinol Lett. 1997; J Neurochem. 2001;79:849 – 858
62:187–197 45. Handigham GE, Chawla S, Cruzaleguie S, et al. Control of
26. Aoki C, Rhee J, Lubin, M, et al. NMDA-R1 subunit of the recruitment and transcription-activating function of CBP deter-
cerebral cortex co-localizes with neuronal nitric oxide synthase at mines gene regulation by NMDA receptors and L-type calcium
pre and postsynaptic sites and in spines. Brain Res. 1997;750:25– 40 channels. Neuron. 1999;22:789
27. Bredt DS, Ferris CD, Snyder SH. Nitric oxide synthase regu- 46. Dometsch RE, Pajvani U, Fife R, et al. Signaling to the nucleus
latory sites. Phosphorylation by cyclic AMP-dependent protein by an L-type calcium channel-calmodulin complex through the
kinase, protein kinase C, and calcium/calmodulin protein kinase, MAP kinase pathway. Science. 2001;294:333
identification of flavin and calmodulin sites. J Biol Chem. 1992;267: 47. Mishra OP, Delivoria-Papadopoulos M. Hypoxia-induced
10976 –10981 generation of nitric oxide free radicals in the cerebral cortex of
28. Dawson TM, Steiner JP, Dawson VL, et al. Immunosuppres- newborn guinea pigs. Neurochem Res. 2000;25:1559
sant FK506 enhances phosphorylation of nitric oxide synthase and 48. Farlie PG, Dringen R, Rees SM, et al. Bcl-2 transgene expres-
protects against glutamate neurotoxicity. Proc Natl Acad Sci USA. sion can protect neurons against developmental and induced cell
1993;90:9808 –9812 death. Proc Natl Acad Sci USA. 1995;92:4397
29. Gow AJ, Duran D, Malcom S, et al. Effect of peroxynitrite- 49. Oltavi ZN, Milliman CM, Korsmeyer SJ. Bcl-2 heterodimer-

NeoReviews Vol.11 No.4 April 2010 e191


Downloaded from http://neoreviews.aappublications.org. Provided by USUHS LRC on July 18, 2010
neurology hypoxic-ischemic encephalopathy

izes in vivo with a conserved homolog, Bax, that accelerates pro- protein kinase IV activation and of cyclic AMP response element
grammed cell death. Cell. 1993;74:609 binding protein phosphorylation during hypoxia in the cerebral
50. Merry DE, Veis DJ, Hickey WF, et al. Bcl-2 protein expression cortex of newborn piglets. Brain Res. 2007;1150:40 – 45
is widespread in the developing nervous system and retained in the 57. Mishra OP, Delivoria-Papadopoulos M. Effect of neuronal
adult PNS. Development. 1994;120:301 nitric oxide synthase inhibition on caspase-9 activity during hypoxia
51. Reed JC. Mechanisms of Bcl-2 family protein function and in the cerebral cortex of newborn piglets. Neurosci Lett. 2006;401:
dysfunction in health and disease. Behring Inst Mitt. 1996;97:72 81– 85
52. Ravishankar S, Ashraf QM, Mishra OP, et al. Expression of Bax 58. Cohen JJ, Duke RC. Glucocorticoid activation of a calcium-
and Bcl-2 proteins during hypoxia in the cerebral cortical neuronal dependent endonuclease in thymocyte nuclei leads to cell death.
nuclei of newborn piglets: effect of administration of magnesium
J Immunol. 1984;32:38 – 42
sulfate. Brain Res. 2001;901:23
59. Ishida R, Akiyoshi H, Takahashi T. Isolation and purification
53. Zanelli SA, Ashraf QM, Mishra OP. Nitration is a mechanism
of calcium and magnesium dependent endonuclease from rat liver
of regulation of the NMDA receptor function during hypoxia.
nuclei. Biochem Biophys Res Commun. 1974;56:703–710
Neurosci. 2002;112869
54. Nicholson DW, Thornberry NA. Caspase killer proteases. 60. Hameed A, Olsen KJ, Lee MK, et al. Cytolysis by Ca-
Trends Biochem Sci. 1997;22:299 –306 permeable transmembrane channels: pore formation causes exten-
55. Delivoria-Papadopoulos M, Ashraf QM, Mishra OP. Effect of sive DNA degradation and cell lysis. J Exp Med. 1989;169:765–777
hypoxia on expression of apoptotic proteins in nuclear, mitochon- 61. Tominaga T, Kagure S, Narisawa K, et al. Endonuclease acti-
drial and cytosolic fractions of the cerebral cortex of newborn vation following focal ischemic injury in the rat brain. Brain Res.
piglets: the role of nuclear Ca⫹⫹ -influx. Neurochem Res. 2008;33: 1993;608:21–26
1196 –1204 62. Waseem W, Ashraf QM, Zanelli SA, et al. Effect of graded
56. Hornick K, Chang E, Zubrow AB, Mishra OP, Delivoria- hypoxia on cerebral cortical genomic DNA fragmentation in new-
Papadopoulos M. Mechanism of Ca(2⫹)/calmodulin-dependent born piglet. Biol Neonate. 2001;79:187–193

e192 NeoReviews Vol.11 No.4 April 2010


Downloaded from http://neoreviews.aappublications.org. Provided by USUHS LRC on July 18, 2010
neurology hypoxic-ischemic encephalopathy

NeoReviews Quiz
1. Under anaerobic conditions, the production of high-energy phosphates decreases, which leads to cell
membrane depolarization and intracellular calcium influx. Increased intracellular calcium has several
deleterious effects on cell structure and function. Of the following, cytoskeletal disruption of microtubules
is most likely the result of calcium-mediated activation of:
A. Nitric oxide synthase.
B. Nuclease.
C. Phospholipase.
D. Protease.
E. Xanthine oxidase.

2. The two mechanisms of cell death following hypoxia-ischemia of the brain are necrosis and apoptosis.
Whereas necrosis is more immediate in onset after the hypoxic-ischemic injury, apoptosis occurs days to
weeks following the insult. Of the following, the most distinguishing feature of apoptosis is:
A. Cell swelling.
B. Chromatin aggregation.
C. Local cytokine release.
D. Oxygen free radical formation.
E. Plasma membrane lysis.

3. N-methyl-D-aspartate (NMDA) cell membrane receptor is a predominant mediator of excitotoxicity in the


immature developing brain. This receptor has a number of pharmacologically distinct binding sites. Of the
following, the coactivator site of the NMDA receptor is most likely to bind to:
A. Glutamine.
B. Glycine.
C. Magnesium.
D. Mk-801.
E. Zinc.

4. Caspases are a family of cysteinyl-aspartate proteases that play an important role in the initiation and
execution of apoptosis. Caspases are of two classes: class I caspases initiate apoptosis and class II caspases
execute apoptosis. Caspase activation occurs through two pathways: the cell surface death receptor-
mediated pathway and mitochondria-initiated pathway. Of the following, the critical caspase in cell surface
death receptor-mediated apoptosis is:
A. Caspase-3.
B. Caspase-6.
C. Caspase-7.
D. Caspase-8.
E. Caspase-10.

NeoReviews Vol.11 No.4 April 2010 e193


Downloaded from http://neoreviews.aappublications.org. Provided by USUHS LRC on July 18, 2010
Biochemical Basis of Hypoxic-Ischemic Encephalopathy
Maria Delivoria-Papadopoulos and Peter J. Marro
NeoReviews 2010;11;e184-e193
DOI: 10.1542/neo.11-4-e184

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


& Services http://neoreviews.aappublications.org/cgi/content/full/neoreview
s;11/4/e184
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. Provided by USUHS LRC on July 18, 2010

Das könnte Ihnen auch gefallen