Birth asphyxia in term neonates affects 1–2/1000 live births and results in the development of hypoxic–ischaemic encephalopathy with devastating life-long consequences. The majority of neuronal cell death occurs with a delay, providing the potential of a treatment window within which to act. Currently, treatment options are limited to therapeutic hypothermia which is not universally successful. To identify new interventions, we need to understand the molecular mechanisms underlying the injury. Here, we provide an overview of the contribution of both oxidative stress and endoplasmic reticulum stress in the development of neonatal brain injury and identify current preclinical therapeutic strategies.

Term neonates (>37 weeks gestation) who develop hypoxic–ischaemic encephalopathy (HIE), neonatal encephalopathy or perinatal arterial stroke have a greater incidence of cerebral palsy, cognitive impairment, developmental delay and epilepsy [13]. Clinical MRI studies report that perinatal hypoxic–ischaemic brain injury is characterised by lesions in grey matter structures such as the basal ganglia, thalamus and cortex, and to a lesser extent infarctions in white matter, where increasing severity is predictive of poorer neurodevelopmental and motor outcome [46]. Currently, the only available treatment is therapeutic hypothermia, where if initiated within 6 h can improve neurological outcome [4,7]. The underlying mechanisms contributing to the development of perinatal brain injury have not been entirely elucidated.

Studies from small and large animal models have shown that with advancing gestation, the developing brain is increasingly vulnerable to hypoxia–ischaemia (HI), with term age-equivalent models showing greater vulnerability to neuronal cell death [8,9] in part due to their high metabolic demand [911]. Following HI, cells initially undergo impaired cerebral oxidative metabolism, swelling and extracellular accumulation of excitatory amino acids (primary phase) and transiently recover (latent phase) prior to a period of secondary energy failure (secondary phase). Over subsequent hours to days, the injurious mechanisms of the secondary phase lead to neuronal cell death. These include excitotoxicity, deterioration of mitochondrial function, inflammation, increased reactive oxygen species (ROS) production and activation of nitric oxide synthase (NOS) and accumulation of intracellular Ca2+ [1215]. We and others have identified mitochondrial dysfunction as the intracellular hub of these injury responses following HI in the immature brain [16].

Mitochondria are key determinants of cell fate having the ability to induce cell death. Delayed cell death that follows neonatal HI is morphologically characterised by either apoptotic (programmed), necrotic (uncontrolled) and more recently, necroptotic (programmed necrosis) phenotype [11,1621]. In comparison with the adult, the immature brain has a high expression of many pro-apoptotic proteins (including Bax, caspase-2, -3, -8 and -9) [22,23], and apoptosis occurs following mitochondrial outer membrane permeabilisation (MOMP). Briefly, Bax translocates from the cytosol to form Bax–Bak pores at the outer membrane which allows for the release of pro-apoptotic proteins. Cytochrome c, apoptosis-inducing factor (AIF), smac/Diablo and endonuclease G translocate from the mitochondria to the cytosol, and while each has a different downstream target, all ultimately lead to apoptotic cell death [2022,2426]. Necroptosis is linked with apoptosis as ligands such as tumour necrosis factor-alpha, TNF-related apoptosis-inducing ligand and Fas can activate death receptors in both instances. However, caspase-8 appears to be the checkpoint that determines whether a cell undergoes necroptosis (caspase-8-independent) or apoptosis [20,27]. Cell death is obviously the endpoint of the induction of these pathways, but in order to design intelligent intervention, a clear understanding of the mechanisms leading to the triggering of such signalling is required. Oxidative stress and endoplasmic reticulum (ER) stress responses are rapidly induced following HI and contribute to cellular injury and death in the immature brain. Here, we will briefly outline how these stresses are generated in the developing brain after HI and the potential for therapeutic intervention.

Oxidative stress occurs when the generation of ROS and reactive nitrogen species (RNS) exceeds the capability of endogenous antioxidant systems. Under normal physiological conditions, low concentrations of ROS and RNS can act as signalling molecules [28]. However, during reperfusion following the primary phase of HI, there is a rapid production of free radicals. Aberrant glutamate receptor activation results in calcium influx activating neuronal nitric oxide synthase (nNOS) to generate nitric oxide (NO; Figure 1). Superoxide (), formed from leakage of electrons from complexes I and III of the electron transport chain, interacts with NO to form peroxynitrite (ONOO), leading to generation of hydroxyl radicals (·OH); subsequent ·OH-mediated cellular impairment occurs through lipid peroxidation, protein oxidation, DNA damage and inhibition of mitochondrial function (specifically complex IV). Complexes I and IV are also targets for direct damage by ONOO (Figure 1). Excitotoxicity-mediated calcium influx also triggers the formation of through the action of NADPH oxidase (NOX, [29]). can be neutralised by superoxide dismutase (SOD), which converts into hydrogen peroxide (H2O2), subsequently converted into H2O by catalase. However, excessive build-up of H2O2 can lead to further generation of ·OH through the iron-based Fenton reaction.

Schematic diagram of the production of ROS and nitrogen species following HI.

Figure 1.
Schematic diagram of the production of ROS and nitrogen species following HI.

Increased intracellular calcium active NOS to generate NO. Superoxide () interacts with NO to form ONOO and subsequently leads to generation of ·OH. ONOO itself can target the mitochondrial respiratory complexes directly. SOD also converts into H2O2, where excessive build-up of H2O2 leads to an exacerbated generation of ·OH via the Fenton reaction. All of these reactive oxygen and nitrogen species contribute to lipid peroxidation, DNA damage and harmful protein oxidation in addition to influencing the release of pro-apoptotic proteins from the mitochondria to initiate cell death.

Figure 1.
Schematic diagram of the production of ROS and nitrogen species following HI.

Increased intracellular calcium active NOS to generate NO. Superoxide () interacts with NO to form ONOO and subsequently leads to generation of ·OH. ONOO itself can target the mitochondrial respiratory complexes directly. SOD also converts into H2O2, where excessive build-up of H2O2 leads to an exacerbated generation of ·OH via the Fenton reaction. All of these reactive oxygen and nitrogen species contribute to lipid peroxidation, DNA damage and harmful protein oxidation in addition to influencing the release of pro-apoptotic proteins from the mitochondria to initiate cell death.

Close modal

The immature brain has a high oxygen consumption and consists of a high concentration of free iron, water content and easily oxidised unsaturated fatty acids. In addition, low myelination and a low expression of antioxidant enzymes [SOD and glutathione peroxidase (GPx)] consequently lead to an underdeveloped antioxidant system, rendering it particularly vulnerable to oxidative damage, with both ROS and RNS strongly influencing excitotoxicity, cell death and mitochondrial impairment [3034].

Studies of neonates with HIE identified evidence of oxidative stress [35,36] as well as evidence suggesting that therapeutic hypothermia reduced lipid peroxidation among its many benefits [36]. It is therefore critical that mechanisms specific to immature brain are identified in order to develop interventions. NOS and mitochondrial electron leakage are believed to be the major contributors of ROS/RNS in the immature brain [37,38] Following HI in the neonatal rat, there is an increase in expression of hydroxyl radicals and free iron in ischaemic regions, and it was recently established that this was mediated in part by nitric oxide [39,40]. Both endothelial nitric oxide synthase and nNOS are significantly increased in the hours following HI [41,42]. Interestingly, the developmental expression pattern of nNOS is comparable to regions vulnerable to injury, and nNOS knockout mice show less injury in response to HI [4346]. Modulating expression of H2O2 has further implicated ROS as detrimental following neonatal HI. Detoxifying enzyme GPx1, which decreases H2O2 levels, has been shown to be protective to neonatal brain after HI [47], whereas overexpression of H2O2 which occurs in mice that overexpress Cu/Zn SOD1, required to produce H2O2, is associated with exacerbation of the injury after HI [48]. This could be due to the accumulation of H2O2 being exposed to high levels of free iron in the immature brain moving the Fenton reaction towards generation of hydroxyl radicals (Figure 1). The role of NOX is less clear in the immature brain. Superoxide production is rapidly increased in the immature mouse brain after HI, coinciding with an increase in the expression of NOX subunits. This increase is functionally coupled to phosphorylation of the NMDA receptor 2B (NR2B) subunit as mutation of this NR2B phosphorylation site reduces HI-induced up-regulation of expression of NOX2, suppresses superoxide formation and reduces cell death [29]. However, genetic ablation of NOX subunits directly has no effect on infarct size and may exacerbate brain damage in excitotoxicity models of immature injury [49]. Clearly, the complexity of these signalling pathways coupled with contributions from different neural cell types makes it difficult to determine the extent on NOX involvement.

Mitochondria are a major contributor to the generation of ROS but also sensitive to oxidative stress, which can culminate in cell death (Figure 1) [38,50]. The release of cytochrome c from the mitochondria is a crucial step in induction of the apoptotic pathway. For this to occur, it must be dissociated from the inner mitochondrial membrane where it is bound to cardiolipin (rich in polyunsaturated fatty acids). Numerous studies suggest that increased ROS generation promotes cytochrome c release from the inner mitochondrial membrane by acting as a catalyst in the oxidation of cardiolipin [5153]. Independent of this, NO, peroxynitrite and 4-hydroxynononeal, a product of lipid peroxidation, have been shown to enhance mitochondrial permeabilisation [54]. Downstream, once initiated, both caspase activation and necroptosis have been suggested to exacerbate production of ROS [55,56].

Given that induction of ROS and RNS is an early event in the development of injury, it is unsurprising that antioxidant therapies are being explored to combat neonatal HI-mediated brain injury. Among its many effects, melatonin acts to scavenge free radicals and to induce expression of antioxidants. Its neuroprotective action was promising in both small and large animal models of neonatal HI [57,58], and this has led to the initiation of clinical trials. However, translation from immature preclinical models to clinical trials is not always straightforward as a mixed cell population such as the brain can lead to differential responses. This was highlighted recently in trials of erythropoietin (EPO), where previous studies showed a high degree of neuroprotection in neonatal models of HI hypothesised to be due to suppression of oxidative stress [59,60]. Very recent studies identified that excessively high H2O2 expression resulted in EPO treatment worsening injury [61], and that differential effects were observed in astrocytes compared with microglia [62]. In a similar manner, allopurinol, a free radical scavenger, provided beneficial effects in animal models [63], which were difficult to reproduce consistently in clinical trials (reviewed in ref. [64]). Other antioxidants under consideration as neuroprotectants in immature brain after HI include the plant polyphenol resveratrol [65] and n-acetylcysteine [6669].

The ER functions to synthesise and regulate protein folding of transmembrane and secreted proteins as well as synthesising phospholipids and cholesterol, but, in addition, it is the site of the highest intracellular calcium concentrations [70]. Therefore, it is unsurprising that pathological alterations in a wide range of intracellular mechanisms culminate in ER stress, manifested as disturbed protein folding and accumulation of unfolded proteins in the ER lumen. To maintain cellular homeostasis, the ER triggers the unfolded protein response (UPR) [71,72] mediated by the chaperone protein glucose-regulated protein (GRP)78 also known as binding immunoglobulin protein. GRP78 is localised in the lumen of the ER and is required for numerous ER functions [73]. Normally, GRP78 binds to and inhibits the activation of the UPR sensors inositol-requiring enzyme (IRE)1, protein kinase RNA-like endoplasmic reticulum kinase (PERK) and activating transcription factor (ATF)6, transmembrane proteins, which possess GFP78-binding sites in their ER lumen portions (Figure 2A). However, accumulation of unfolded proteins results in the sequestering of GRP78 away from these UPR sensors leading to regulation of protein synthesis and increased degradation of unfolded proteins, either through autophagy or ER-activated degradation (ERAD).

The response of the ER during homeostasis and under mild and chronic stress.

Figure 2.
The response of the ER during homeostasis and under mild and chronic stress.

(A) During homeostasis when normal protein folding occurs, chaperone protein glucose-related protein (GRP)78 binds transmembrane proteins IRE1, ATF6 and PERK in the ER. (B) Following mild stress leading to misfolding of proteins, the ER initiates the UPR cascade, releasing GRP78 to chaperone proper protein folding. Activated transmembrane proteins damp down protein synthesis except for proteins required for the UPR to restore equilibrium. (C) Severe, prolonged stress to the ER can induce cell death that overwhelms the UPR and culminates in cell death through many mechanisms.

Figure 2.
The response of the ER during homeostasis and under mild and chronic stress.

(A) During homeostasis when normal protein folding occurs, chaperone protein glucose-related protein (GRP)78 binds transmembrane proteins IRE1, ATF6 and PERK in the ER. (B) Following mild stress leading to misfolding of proteins, the ER initiates the UPR cascade, releasing GRP78 to chaperone proper protein folding. Activated transmembrane proteins damp down protein synthesis except for proteins required for the UPR to restore equilibrium. (C) Severe, prolonged stress to the ER can induce cell death that overwhelms the UPR and culminates in cell death through many mechanisms.

Close modal

Once released from GRP78, IRE1 oligomerises, becomes phosphorylated and utilises its endoribonuclease domain to degrade RNA, reducing the burden on the ER. In addition, IRE1 splices and activates XBP1, which translocates to the nucleus, up-regulating expression of key proteins required to manage both the stress response (e.g. chaperones GRP78 and GRP94) and improved clearing of unfolded proteins (ERAD) [74]. In addition to degradation by ERAD, both PERK and IRE1 can trigger autophagy in response to ER stress. Oligomerised, phosphorylated PERK down-regulates general protein synthesis through phosphorylating and inactivating elongation initiation factor (eIF)2α. However, translation of selective mRNAs is up-regulated after eIF2α inhibition, e.g. ATF4, which can induce the expression of C/EBP-homologous protein (CHOP). PERK also phosphorylates NRF2 resulting in its activation and increased antioxidant response [75]. Finally, ATF6 translocates to the Golgi where it is cleaved, becoming an active transcription factor capable of up-regulating expression of chaperones, as well as CHOP and XBP1 (Figure 2B) [76].

Pathological signalling events triggered by birth asphyxia disrupt cellular homeostasis and as such lead to the induction of ER stress and the UPR [77]. Using a well-characterised rat model of neonatal HI, Puka-Sundvall et al. [19] demonstrated that there is a significant accumulation of calcium in the ER with clear increases from 30 min to 3 h following HI injury. In cortical neurones subjected to oxygen-glucose deprivation (an in vitro mimic of HI), phosphorylation of PERK, IRE1 and cleavage of ATF6 is observed to occur rapidly after injury [78]. Phosphorylation of PERK and eIF2α also occurs rapidly and transiently in vivo after neonatal HI in term-equivalent mice as well as an up-regulation of GRP78 and CHOP [7881].

Although the aim of the UPR is to restore proteostasis and cellular homeostasis, severe ER stress and prolonged activation of the above pathways can induce cell death (Figure 2C) [76]. Prolonged activation of PERK can result in expression of Noxa which induces Bax localisation to the mitochondrial outer membrane resulting in its permeabilisation and induction of apoptosis [82]. Bax-mediated mitochondrial outer membrane permeabilisation is a hallmark of secondary brain injury in neonatal HI [23,83]. Similarly, ATF6 will induce pro-apoptotic Bim expression [84], while IRE1 recruits TRAF2 and downstream JNK signalling to inhibit anti-apoptotic Bcl-2 protein expression [85]. Induction of death receptor expression is also a consequence of prolonged ER stress (Figure 2C) [86]. As all these pro- and anti-apoptotic effects are observed in the development of injury in neonatal HI [8790], it is tempting to speculate that targeting prolonged ER stress may represent an intervention point for the early inhibition of cell death mechanisms triggered after neonatal HI.

Although the mechanisms are not as clearly defined, prolonged ER stress can also induce the up-regulation of autophagy and phosphorylation of key signalling molecules in the necroptotic (regulated necrosis) cell death pathway. Inhibition of necroptosis is neuroprotective after HI in vivo [91], and ER stress is reported to function upstream of necroptosis in this model [92]. Prolonged ER stress can also result in cross-talk with autophagy when the ERAD degradation of proteins becomes overwhelmed [93]. In animal and cellular models of mitochondrial respiratory complex diseases, both ER stress and autophagy were recently found to be up-regulated as a consequence of disease-induced translation dysfunction and proteotoxic stress [94]. This elegant study by Peng et al. found that rather than targeting the complexes themselves, significant mitochondrial functional benefits were instead conferred by inhibiting the downstream consequences. In particular, low-dose cycloheximide and probucol (an anti-hyperlipidemic drug in clinical use) were very effective in preventing the induction of ER stress and the up-regulation of autophagy, promoting oxidative phosphorylation, cell survival in vitro and reduced disease severity in vivo [94]. Aberrant autophagy is also reported in the in vivo model of HI, although whether the pathway acts in a beneficial or deleterious manner is currently unclear [81,95,96].

Compared with the field of oxidative stress in neonatal HI (see above), there are significantly fewer neuroprotective interventions targeting ER stress. However, many recent studies have identified neuroprotective paradigms in in vivo rodent models of neonatal HI, which act through the prevention of prolonged ER stress (Table 1). None of these neuroprotectants targets ER stress uniquely (e.g. melatonin also act as a free radical scavenger), and it remains to be seen whether these potential therapies will offer synergistic efficacy with hypothermia, the current standard of care, or whether they represent ‘stand-alone’ treatments.

Table 1
Reducing ER stress is neuroprotective in rodent models of neonatal HI
TreatmentSpecies/deliveryOutcomeRef.
Basic fibroblast growth factor (bFGF) Rat (P7)/intranasal Reduced ER stress (ATF6, GRP78, XBP-1, ATF4 and CHOP)
Reduced infarct volume 
[80
Acidic fibroblast growth factor (aFGF) Rat (P7)/intranasal Reduced ER stress (ATF6, GRP78, XBP-1, ATF4 and CHOP)
Reduced infarct volume 
[100
Necrostatin-1 (Nec-1) Mice (P7)/intracerebroventricular Reduced ER stress (GRP78, PERK, peIF1a, XBP1, GADD34 and CHOP)
EM (organelle pathology) 
[92
Hydrogen-rich saline Mice (P7)/intraperitoneal Reduced ER stress (GRP78 and CHOP)
Reduced infarct volume 
[101
Notoginsenoside R1 (NGR1) Rats (P7)/intraperitoneal Reduced ER stress (GRP78, PERK, IRE1α, CHOP and BCL-2)
Reduced infarct volume 
[79
Melatonin Rats (P7)/intraperitoneal Reduced ER stress (GRP78, PERK, IRE1α, CHOP and XBP-1)
Reduced infarct volume 
[102
TreatmentSpecies/deliveryOutcomeRef.
Basic fibroblast growth factor (bFGF) Rat (P7)/intranasal Reduced ER stress (ATF6, GRP78, XBP-1, ATF4 and CHOP)
Reduced infarct volume 
[80
Acidic fibroblast growth factor (aFGF) Rat (P7)/intranasal Reduced ER stress (ATF6, GRP78, XBP-1, ATF4 and CHOP)
Reduced infarct volume 
[100
Necrostatin-1 (Nec-1) Mice (P7)/intracerebroventricular Reduced ER stress (GRP78, PERK, peIF1a, XBP1, GADD34 and CHOP)
EM (organelle pathology) 
[92
Hydrogen-rich saline Mice (P7)/intraperitoneal Reduced ER stress (GRP78 and CHOP)
Reduced infarct volume 
[101
Notoginsenoside R1 (NGR1) Rats (P7)/intraperitoneal Reduced ER stress (GRP78, PERK, IRE1α, CHOP and BCL-2)
Reduced infarct volume 
[79
Melatonin Rats (P7)/intraperitoneal Reduced ER stress (GRP78, PERK, IRE1α, CHOP and XBP-1)
Reduced infarct volume 
[102

The molecular mechanisms underlying the development of brain injury in neonates who have suffered birth asphyxia remain unclear and providing treatment options is a critical, unmet clinical need. The recent avalanche of preclinical data has identified many key pathways activated early in the development of the pathology and which may be amenable to therapeutic intervention. Here, we have distilled the recent evidence regarding induction of oxidative stress and ER stress signalling after HI injury in the immature brain. More is currently understood about oxidative stress than ER stress in the immature brain. Mice genetically modified to alter the expression of the ER transmembrane receptors may provide more specific clues as to the contribution of ER stress in the development of neonatal brain injury. However, it is impossible to consider these areas in isolation. Although due to their complexity it is hard to evaluate this cross-talk experimentally, there are a few examples emerging. For example, induction of ER stress in macrophages is reported to result in increased NOX expression and subsequent apoptosis is mediated by CHOP [97], and a member of the GPx family promotes the function of GRP78 [98]. More generally, ROS can also be produced in the ER as a by-product of protein folding, upsetting the critical redox balance within the ER lumen, inducing downstream oxidative stress responses and disrupting cellular homeostasis which feeds back to exacerbate ER stress further [99]. Identifying drugs which can tackle multiple targets within these pathways, e.g. melatonin, may prove to be the key in the development of potential interventions to ameliorate brain injury in these vulnerable neonates.

·OH

hydroxyl radicals

ATF

activating transcription factor

CHOP

C/EBP-homologous protein

eIF

elongation initiation factor

eNOS

endothelial nitric oxide synthase

EPO

erythropoietin

ER

endoplasmic reticulum

ERAD

ER-activated degradation

GPx

glutathione peroxidase

GRP

glucose-regulated protein

H2O2

hydrogen peroxide

HI

hypoxia–ischaemia

HIE

hypoxic–ischaemic encephalopathy

IRE

inositol-requiring enzyme

nNOS

neuronal nitric oxide synthase

NO

nitric oxide

NOS

nitric oxide synthase

NOX

NADPH oxidase

NR2B

NMDA receptor 2B

superoxide

ONOO

peroxynitrite

PERK

protein kinase RNA-like endoplasmic reticulum kinase

RNS

reactive nitrogen species

ROS

reactive oxygen species

SOD

superoxide dismutase

UPR

unfolded protein response

We gratefully acknowledge the support of the Department of Perinatal Imaging and Health and financial support from Wellcome Trust [WT094823], King's Health Partners, The Research Foundation of Cerebral Palsy Alliance [PG1715], Medical Research Council, Action Medical Research, Swedish Medical Research Council [VR 2015-02493], Hjärnfonden [Brain Foundation 2015-0004], Governmental Grants to University Hospitals in Sweden [ALF-GBG 426401] and the Wilhelm and Martina Lundgren Foundation.

The Authors declare that there are no competing interests associated with the manuscript.

1
Cowan
,
F.
,
Rutherford
,
M.
,
Groenendaal
,
F.
,
Eken
,
P.
,
Mercuri
,
E.
,
Bydder
,
G.M.
et al
(
2003
)
Origin and timing of brain lesions in term infants with neonatal encephalopathy
.
Lancet
361
,
736
742
doi:
2
Badawi
,
N.
,
Felix
,
J.F.
,
Kurinczuk
,
J.J.
,
Dixon
,
G.
,
Watson
,
L.
,
Keogh
,
J.M.
et al
(
2005
)
Cerebral palsy following term newborn encephalopathy: a population-based study
.
Dev. Med. Child Neurol.
47
,
293
298
doi:
3
Lee
,
J.
,
Croen
,
L.A.
,
Lindan
,
C.
,
Nash
,
K.B.
,
Yoshida
,
C.K.
,
Ferriero
,
D.M.
et al
(
2005
)
Predictors of outcome in perinatal arterial stroke: a population-based study
.
Ann. Neurol.
58
,
303
308
doi:
4
Rutherford
,
M.
,
Ramenghi
,
L.A.
,
Edwards
,
A.D.
,
Brocklehurst
,
P.
,
Halliday
,
H.
,
Levene
,
M.
et al
(
2010
)
Assessment of brain tissue injury after moderate hypothermia in neonates with hypoxic–ischaemic encephalopathy: a nested substudy of a randomised controlled trial
.
Lancet Neurol.
9
,
39
45
doi:
5
Alderliesten
,
T.
,
Nikkels
,
P.G.J.
,
Benders
,
M.J.N.L.
,
de Vries
,
L.S.
and
Groenendaal
,
F.
(
2013
)
Antemortem cranial MRI compared with postmortem histopathologic examination of the brain in term infants with neonatal encephalopathy following perinatal asphyxia
.
Arch. Dis. Child Fetal Neonatal Ed.
98
,
F304
F309
doi:
6
Rutherford
,
M.
,
Malamateniou
,
C.
,
McGuinness
,
A.
,
Allsop
,
J.
,
Biarge
,
M.M.
and
Counsell
,
S.
(
2010
)
Magnetic resonance imaging in hypoxic-ischaemic encephalopathy
.
Early Hum. Dev.
86
,
351
360
doi:
7
Azzopardi
,
D.
,
Strohm
,
B.
,
Marlow
,
N.
,
Brocklehurst
,
P.
,
Deierl
,
A.
,
Eddama
,
O.
et al
(
2014
)
Effects of hypothermia for perinatal asphyxia on childhood outcomes
.
N. Engl. J. Med.
371
,
140
149
doi:
8
Towfighi
,
J.
,
Mauger
,
D.
,
Vannucci
,
R.C.
and
Vannucci
,
S.J.
(
1997
)
Influence of age on the cerebral lesions in an immature rat model of cerebral hypoxia–ischemia: a light microscopic study
.
Dev. Brain Res.
100
,
149
160
doi:
9
Mallard
,
E.C.
,
Williams
,
C.E.
,
Johnston
,
B.M.
and
Gluckman
,
P.D.
(
1994
)
Increased vulnerability to neuronal damage after umbilical cord occlusion in fetal sheep with advancing gestation
.
Am. J. Obstet. Gynecol.
170
,
206
214
doi:
10
Folkerth
,
R.D.
(
2005
)
Neuropathologic substrate of cerebral palsy
.
J. Child Neurol.
20
,
940
949
doi:
11
Northington
,
F.J.
,
Chavez-Valdez
,
R.
and
Martin
,
L.J.
(
2011
)
Neuronal cell death in neonatal hypoxia-ischemia
.
Ann. Neurol.
69
,
743
758
doi:
12
Blumberg
,
R.M.
,
Cady
,
E.B.
,
Wigglesworth
,
J.S.
,
McKenzie
,
J.E.
and
Edwards
,
A.D.
(
1997
)
Relation between delayed impairment of cerebral energy metabolism and infarction following transient focal hypoxia-ischaemia in the developing brain
.
Exp. Brain Res.
113
,
130
137
doi:
13
Hagberg
,
H.
,
David Edwards
,
A.
and
Groenendaal
,
F.
(
2016
)
Perinatal brain damage: the term infant
.
Neurobiol. Dis.
92
,
102
112
doi:
14
Gilland
,
E.
,
Bona
,
E.
and
Hagber
,
H.
(
1998
)
Temporal changes of regional glucose use, blood flow, and microtubule-associated protein 2 immunostaining after hypoxia–ischemia in the immature rat brain
.
J. Cereb. Blood Flow Metab.
18
,
222
228
doi:
15
Lorek
,
A.
,
Takei
,
Y.
,
Cady
,
E.B.
,
Wyatt
,
J.S.
,
Penrice
,
J.
,
Edwards
,
A.D.
et al
(
1994
)
Delayed (‘secondary’) cerebral energy failure after acute hypoxia-ischemia in the newborn piglet: continuous 48-hour studies by phosphorus magnetic resonance spectroscopy
.
Pediatr. Res.
36
,
699
706
doi:
16
Hagberg
,
H.
,
Mallard
,
C.
,
Rousset
,
C.I.
and
Thornton
,
C.
(
2014
)
Mitochondria: hub of injury responses in the developing brain
.
Lancet Neurol.
13
,
217
232
doi:
17
Northington
,
F.J.
,
Zelaya
,
M.E.
,
O'Riordan
,
D.P.
,
Blomgren
,
K.
,
Flock
,
D.L.
,
Hagberg
,
H.
et al
(
2007
)
Failure to complete apoptosis following neonatal hypoxia–ischemia manifests as ‘continuum’ phenotype of cell death and occurs with multiple manifestations of mitochondrial dysfunction in rodent forebrain
.
Neuroscience
149
,
822
833
doi:
18
Portera-Cailliau
,
C.
,
Price
,
D.L.
and
Martin
,
L.J.
(
1997
)
Excitotoxic neuronal death in the immature brain is an apoptosis-necrosis morphological continuum
.
J. Comp. Neurol.
378
,
70
87
PMID:
[PubMed]
19
Puka-Sundvall
,
M.
,
Gajkowska
,
B.
,
Cholewinski
,
M.
,
Blomgren
,
K.
,
Lazarewicz
,
J.W.
and
Hagberg
,
H.
(
2000
)
Subcellular distribution of calcium and ultrastructural changes after cerebral hypoxia-ischemia in immature rats
.
Dev. Brain Res.
125
,
31
41
doi:
20
Thornton
,
C.
and
Hagberg
,
H.
(
2015
)
Role of mitochondria in apoptotic and necroptotic cell death in the developing brain
.
Clin. Chim. Acta
451
,
35
38
doi:
21
Thornton
,
C.
,
Rousset
,
C.I.
,
Kichev
,
A.
,
Miyakuni
,
Y.
,
Vontell
,
R.
,
Baburamani
,
A.A.
et al
(
2012
)
Molecular mechanisms of neonatal brain injury
.
Neurol. Res. Int.
2012
,
506320
doi:
22
Zhu
,
C.
,
Wang
,
X.
,
Xu
,
F.
,
Bahr
,
B.A.
,
Shibata
,
M.
,
Uchiyama
,
Y.
et al
(
2005
)
The influence of age on apoptotic and other mechanisms of cell death after cerebral hypoxia–ischemia
.
Cell Death Differ.
12
,
162
176
doi:
23
Wang
,
X.
,
Carlsson
,
Y.
,
Basso
,
E.
,
Zhu
,
C.
,
Rousset
,
C.I.
,
Rasola
,
A.
et al
(
2009
)
Developmental shift of cyclophilin D contribution to hypoxic-ischemic brain injury
.
J. Neurosci.
29
,
2588
2596
doi:
24
Wang
,
X.
,
Karlsson
,
J.-O.
,
Zhu
,
C.
,
Bahr
,
B.A.
,
Hagberg
,
H.
and
Blomgren
,
K.
(
2001
)
Caspase-3 activation after neonatal rat cerebral hypoxia-ischemia
.
Neonatology
79
,
172
179
doi:
25
Zhu
,
C.
,
Wang
,
X.
,
Deinum
,
J.
,
Huang
,
Z.
,
Gao
,
J.
,
Modjtahedi
,
N.
et al
(
2007
)
Cyclophilin A participates in the nuclear translocation of apoptosis-inducing factor in neurons after cerebral hypoxia-ischemia
.
J. Exp. Med.
204
,
1741
1748
doi:
26
Zhu
,
C.
,
Wang
,
X.
,
Huang
,
Z.
,
Qiu
,
L.
,
Xu
,
F.
,
Vahsen
,
N.
et al
(
2007
)
Apoptosis-inducing factor is a major contributor to neuronal loss induced by neonatal cerebral hypoxia-ischemia
.
Cell Death Differ.
14
,
775
784
doi:
27
Chavez-Valdez
,
R.
,
Martin
,
L.J.
and
Northington
,
F.J.
(
2012
)
Programmed necrosis: a prominent mechanism of cell death following neonatal brain injury
.
Neurol. Res. Int.
2012
,
257563
doi:
28
Weidinger
,
A.
and
Kozlov
,
A.V.
(
2015
)
Biological activities of reactive oxygen and nitrogen species: oxidative stress versus signal transduction
.
Biomolecules
5
,
472
484
doi:
29
Knox
,
R.
,
Brennan-Minnella
,
A.M.
,
Lu
,
F.
,
Yang
,
D.
,
Nakazawa
,
T.
,
Yamamoto
,
T.
et al
(
2014
)
NR2B phosphorylation at tyrosine 1472 contributes to brain injury in a rodent model of neonatal hypoxia-ischemia
.
Stroke
45
,
3040
3047
doi:
30
Perrone
,
S.
,
Negro
,
S.
,
Tataranno
,
M.L.
and
Buonocore
,
G.
(
2010
)
Oxidative stress and antioxidant strategies in newborns
.
J. Matern. Fetal Neonatal Med.
23
(
Suppl 3
),
63
65
doi:
31
Ikonomidou
,
C.
and
Kaindl
,
A.M.
(
2011
)
Neuronal death and oxidative stress in the developing brain
.
Antiox. Redox Signal.
14
,
1535
1550
doi:
32
Ferriero
,
D.M.
(
2001
)
Oxidant mechanisms in neonatal hypoxia-ischemia
.
Dev. Neurosci.
23
,
198
202
doi:
33
Ten
,
V.S.
and
Starkov
,
A.
(
2012
)
Hypoxic-ischemic injury in the developing brain: the role of reactive oxygen species originating in mitochondria
.
Neurol. Res. Int.
2012
,
542976
doi:
34
Mayurasakorn
,
K.
,
Niatsetskaya
,
Z.V.
,
Sosunov
,
S.A.
,
Williams
,
J.J.
,
Zirpoli
,
H.
,
Vlasakov
,
I.
et al
(
2016
)
DHA but not EPA emulsions preserve neurological and mitochondrial function after brain hypoxia-ischemia in neonatal mice
.
PLoS ONE
11
,
e0160870
doi:
35
Mutlu
,
M.
,
Sariaydin
,
M.
,
Aslan
,
Y.
,
Kader
,
Ş.
,
Dereci
,
S.
,
Kart
,
C.
et al
(
2016
)
Status of vitamin D, antioxidant enzymes, and antioxidant substances in neonates with neonatal hypoxic-ischemic encephalopathy
.
J. Matern. Fetal Neonatal Med.
29
,
2259
2263
doi:
36
Perrone
,
S.
,
Szabó
,
M.
,
Bellieni
,
C.V.
,
Longini
,
M.
,
Bangó
,
M.
,
Kelen
,
D.
et al
(
2010
)
Whole body hypothermia and oxidative stress in babies with hypoxic-ischemic brain injury
.
Pediatr. Neurol.
43
,
236
240
doi:
37
Zhao
,
M.
,
Zhu
,
P.
,
Fujino
,
M.
,
Zhuang
,
J.
,
Guo
,
H.
,
Sheikh
,
I.A.
et al
(
2016
)
Oxidative stress in hypoxic-ischemic encephalopathy: molecular mechanisms and therapeutic strategies
.
Int. J. Mol. Sci.
17
,
2078
doi:
38
Blomgren
,
K.
and
Hagberg
,
H.
(
2006
)
Free radicals, mitochondria, and hypoxia–ischemia in the developing brain
.
Free Radic. Biol. Med.
40
,
388
397
doi:
39
Palmer
,
C.
,
Menzies
,
S.L.
,
Roberts
,
R.L.
,
Pavlick
,
G.
and
Connor
,
J.R.
(
1999
)
Changes in iron histochemistry after hypoxic-ischemic brain injury in the neonatal rat
.
J. Neurosci. Res.
56
,
60
71
doi:
40
Lu
,
Q.
,
Harris
,
V.A.
,
Rafikov
,
R.
,
Sun
,
X.
,
Kumar
,
S.
and
Black
,
S.M.
(
2015
)
Nitric oxide induces hypoxia ischemic injury in the neonatal brain via the disruption of neuronal iron metabolism
.
Redox Biol.
6
,
112
121
doi:
41
van den Tweel
,
E.R.W.
,
Nijboer
,
C.
,
Kavelaars
,
A.
,
Heijnen
,
C.J.
,
Groenendaal
,
F.
and
van Bel
,
F.
(
2005
)
Expression of nitric oxide synthase isoforms and nitrotyrosine formation after hypoxia–ischemia in the neonatal rat brain
.
J. Neuroimmunol.
167
,
64
71
doi:
42
Ergenekon
,
E.
and
Gücüyener
,
K.
(
1998
)
Nitric oxide in developing brain
.
Eur. J. Paediatr. Neurol.
2
,
297
301
doi:
43
Black
,
S.M.
,
Bedolli
,
M.A.
,
Martinez
,
S.
,
Bristow
,
J.D.
,
Ferriero
,
D.M.
and
Soifer
,
S.J.
(
1995
)
Expression of neuronal nitric oxide synthase corresponds to regions of selective vulnerability to hypoxia-ischaemia in the developing rat brain
.
Neurobiol. Dis.
2
,
145
155
doi:
44
Ferriero
,
D.M.
,
Holtzman
,
D.M.
,
Black
,
S.M.
and
Sheldon
,
R.A.
(
1996
)
Neonatal mice lacking neuronal nitric oxide synthase are less vulnerable to hypoxic–ischemic injury
.
Neurobiol. Dis.
3
,
64
71
doi:
45
Muramatsu
,
K.
,
Sheldon
,
R.A.
,
Black
,
S.M.
,
Täuber
,
M.
and
Ferriero
,
D.M.
(
2000
)
Nitric oxide synthase activity and inhibition after neonatal hypoxia ischemia in the mouse brain
.
Dev. Brain Res.
123
,
119
127
doi:
46
Ferriero
,
D.M.
,
Sheldon
,
R.A.
,
Black
,
S.M.
and
Chuai
,
J.
(
1995
)
Selective destruction of nitric oxide synthase neurons with quisqualate reduces damage after hypoxia-ischemia in the neonatal rat
.
Pediatr. Res.
38
,
912
918
doi:
47
Sheldon
,
R.A.
,
Christen
,
S.
and
Ferriero
,
D.M.
(
2008
)
Genetic and pharmacologic manipulation of oxidative stress after neonatal hypoxia-ischemia
.
Int. J. Dev. Neurosci.
26
,
87
92
doi:
48
Ditelberg
,
J.S.
,
Sheldon
,
R.A.
,
Epstein
,
C.J.
and
Ferriero
,
D.M.
(
1996
)
Brain injury after perinatal hypoxia-ischemia is exacerbated in copper/zinc superoxide dismutase transgenic mice
.
Pediatr. Res.
39
,
204
208
doi:
49
Doverhag
,
C.
,
Keller
,
M.
,
Karlsson
,
A.
,
Hedtjarn
,
M.
,
Nilsson
,
U.
,
Kapeller
,
E.
et al
(
2008
)
Pharmacological and genetic inhibition of NADPH oxidase does not reduce brain damage in different models of perinatal brain injury in newborn mice
.
Neurobiol. Dis.
31
,
133
144
doi:
50
Turrens
,
J.F.
(
2003
)
Mitochondrial formation of reactive oxygen species
.
J. Physiol.
552
,
335
344
doi:
51
Petrosillo
,
G.
,
Ruggiero
,
F.M.
and
Paradies
,
G.
(
2003
)
Role of reactive oxygen species and cardiolipin in the release of cytochrome c from mitochondria
.
FASEB J.
17
,
2202
2208
doi:
52
Petrosillo
,
G.
,
Ruggiero
,
F.M.
,
Pistolese
,
M.
and
Paradies
,
G.
(
2001
)
Reactive oxygen species generated from the mitochondrial electron transport chain induce cytochrome c dissociation from beef-heart submitochondrial particles via cardiolipin peroxidation. Possible role in the apoptosis
.
FEBS Lett.
509
,
435
438
doi:
53
Kagan
,
V.E.
,
Tyurin
,
V.A.
,
Jiang
,
J.
,
Tyurina
,
Y.Y.
,
Ritov
,
V.B.
,
Amoscato
,
A.A.
et al
(
2005
)
Cytochrome c acts as a cardiolipin oxygenase required for release of proapoptotic factors
.
Nat. Chem. Biol.
1
,
223
232
doi:
54
Vieira
,
H.L.A.
,
Belzacq
,
A.-S.
,
Haouzi
,
D.
,
Bernassola
,
F.
,
Cohen
,
I.
,
Jacotot
,
E.
et al
(
2001
)
The adenine nucleotide translocator: a target of nitric oxide, peroxynitrite, and 4-hydroxynonenal
.
Oncogene
20
,
4305
4316
doi:
55
Dixon
,
S.J.
and
Stockwell
,
B.R.
(
2014
)
The role of iron and reactive oxygen species in cell death
.
Nat. Chem. Biol.
10
,
9
17
doi:
56
Ricci
,
J.-E.
,
Muñoz-Pinedo
,
C.
,
Fitzgerald
,
P.
,
Bailly-Maitre
,
B.
,
Perkins
,
G.A.
,
Yadava
,
N.
et al
(
2004
)
Disruption of mitochondrial function during apoptosis is mediated by caspase cleavage of the p75 subunit of complex I of the electron transport chain
.
Cell
117
,
773
786
doi:
57
Husson
,
I.
,
Mesplès
,
B.
,
Bac
,
P.
,
Vamecq
,
J.
,
Evrard
,
P.
and
Gressens
,
P.
(
2002
)
Melatoninergic neuroprotection of the murine periventricular white matter against neonatal excitotoxic challenge
.
Ann. Neurol.
51
,
82
92
doi:
58
Welin
,
A.-K.
,
Svedin
,
P.
,
Lapatto
,
R.
,
Sultan
,
B.
,
Hagberg
,
H.
,
Gressens
,
P.
et al
(
2007
)
Melatonin reduces inflammation and cell death in white matter in the mid-gestation fetal sheep following umbilical cord occlusion
.
Pediatr. Res.
61
,
153
158
doi:
59
Kellert
,
B.A.
,
McPherson
,
R.J.
and
Juul
,
S.E.
(
2007
)
A comparison of high-dose recombinant erythropoietin treatment regimens in brain-injured neonatal rats
.
Pediatr. Res.
61
,
451
455
doi:
60
Gonzalez
,
F.F.
,
McQuillen
,
P.
,
Mu
,
D.
,
Chang
,
Y.
,
Wendland
,
M.
,
Vexler
,
Z.
et al
(
2007
)
Erythropoietin enhances long-term neuroprotection and neurogenesis in neonatal stroke
.
Dev. Neurosci.
29
,
321
330
doi:
61
Sheldon
,
R.A.
,
Windsor
,
C.
,
Lee
,
B.S.
,
Arteaga Cabeza
,
O.
and
Ferriero
,
D.M.
(
2017
)
Erythropoietin treatment exacerbates moderate injury after hypoxia-ischemia in neonatal superoxide dismutase transgenic mice
.
Dev. Neurosci.
39
,
228
237
doi:
62
Pathipati
,
P.
and
Ferriero
,
D.M.
(
2017
)
The differential effects of erythropoietin exposure to oxidative stress on microglia and astrocytes in vitro
.
Dev. Neurosci.
39
,
310
322
doi:
63
Palmer
,
C.
,
Towfighi
,
J.
,
Roberts
,
R.L.
and
Heitjan
,
D.F.
(
1993
)
Allopurinol administered after inducing hypoxia-ischemia reduces brain injury in 7-day-old rats
.
Pediatr. Res.
33
,
405
411
doi:
64
Arteaga
,
O.
,
Álvarez
,
A.
,
Revuelta
,
M.
,
Santaolalla
,
F.
,
Urtasun
,
A.
and
Hilario
,
E.
(
2017
)
Role of antioxidants in neonatal hypoxic–ischemic brain injury: new therapeutic approaches
.
Int. J. Mol. Sci.
18
,
265
doi:
65
Pan
,
S.
,
Li
,
S.
,
Hu
,
Y.
,
Zhang
,
H.
,
Liu
,
Y.
,
Jiang
,
H.
et al
(
2016
)
Resveratrol post-treatment protects against neonatal brain injury after hypoxia-ischemia
.
Oncotarget
7
,
79247
79261
doi:
66
Nie
,
X.
,
Lowe
,
D.W.
,
Rollins
,
L.G.
,
Bentzley
,
J.
,
Fraser
,
J.L.
,
Martin
,
R.
et al
(
2016
)
Sex-specific effects of N-acetylcysteine in neonatal rats treated with hypothermia after severe hypoxia-ischemia
.
Neurosci. Res.
108
,
24
33
doi:
67
Lee
,
T.-F.
,
Tymafichuk
,
C.N.
,
Bigam
,
D.L.
and
Cheung
,
P.-Y.
(
2008
)
Effects of postresuscitation N-acetylcysteine on cerebral free radical production and perfusion during reoxygenation of hypoxic newborn piglets
.
Pediatr. Res.
64
,
256
261
doi:
68
Lee
,
T.-F.
,
Jantzie
,
L.L.
,
Todd
,
K.G.
and
Cheung
,
P.-Y.
(
2008
)
Postresuscitation N-acetylcysteine treatment reduces cerebral hydrogen peroxide in the hypoxic piglet brain
.
Intensive Care Med.
34
,
190
197
doi:
69
Kannan
,
S.
,
Dai
,
H.
,
Navath
,
R.S.
,
Balakrishnan
,
B.
,
Jyoti
,
A.
,
Janisse
,
J.
et al
(
2012
)
Dendrimer-based postnatal therapy for neuroinflammation and cerebral palsy in a rabbit model
.
Sci. Transl. Med.
4
,
130
146
doi:
70
Xu
,
C.
,
Bailly-Maitre
,
B.
and
Reed
,
J.C.
(
2005
)
Endoplasmic reticulum stress: cell life and death decisions
.
J. Clin. Invest.
115
,
2656
2664
doi:
71
Wang
,
M.
and
Kaufman
,
R.J.
(
2016
)
Protein misfolding in the endoplasmic reticulum as a conduit to human disease
.
Nature
529
,
326
335
doi:
72
Hetz
,
C.
,
Chevet
,
E.
and
Oakes
,
S.A.
(
2015
)
Proteostasis control by the unfolded protein response
.
Nat. Cell Biol.
17
,
829
838
doi:
73
Wang
,
M.
,
Wey
,
S.
,
Zhang
,
Y.
,
Ye
,
R.
and
Lee
,
A.S.
(
2009
)
Role of the unfolded protein response regulator GRP78/BiP in development, cancer, and neurological disorders
.
Antioxid. Redox Signal.
11
,
2307
2316
doi:
74
Chen
,
Y.
and
Brandizzi
,
F.
(
2013
)
IRE1: ER stress sensor and cell fate executor
.
Trends Cell Biol.
23
,
547
555
doi:
75
Liu
,
Z.
,
Lv
,
Y.
,
Zhao
,
N.
,
Guan
,
G.
and
Wang
,
J.
(
2015
)
Protein kinase R-like ER kinase and its role in endoplasmic reticulum stress-decided cell fate
.
Cell Death Dis.
6
,
e1822
doi:
76
Iurlaro
,
R.
and
Muñoz-Pinedo
,
C.
(
2016
)
Cell death induced by endoplasmic reticulum stress
.
FEBS J.
283
,
2640
2652
doi:
77
Bueter
,
W.
,
Dammann
,
O.
and
Leviton
,
A.
(
2009
)
Endoplasmic reticulum stress, inflammation, and perinatal brain damage
.
Pediatr. Res.
66
,
487
494
doi:
78
Badiola
,
N.
,
Penas
,
C.
,
Miñano-Molina
,
A.
,
Barneda-Zahonero
,
B.
,
Fado
,
R.
,
Sánchez-Opazo
,
G.
et al
(
2011
)
Induction of ER stress in response to oxygen-glucose deprivation of cortical cultures involves the activation of the PERK and IRE-1 pathways and of caspase-12
.
Cell Death Dis.
2
,
e149
doi:
79
Wang
,
Y.
,
Tu
,
L.
,
Li
,
Y.
,
Chen
,
D.
and
Wang
,
S.
(
2016
)
Notoginsenoside R1 protects against neonatal cerebral hypoxic-ischemic injury through estrogen receptor-dependent activation of endoplasmic reticulum stress pathways
.
J. Pharmacol. Exp. Ther.
357
,
591
605
doi:
80
Lin
,
Z.
,
Hu
,
Y.
,
Wang
,
Z.
,
Pan
,
S.
,
Zhang
,
H.
,
Ye
,
L.
et al
(
2017
)
Intranasal basic fibroblast growth factor attenuates endoplasmic reticulum stress and brain injury in neonatal hypoxic-ischaemic injury
.
Am. J. Transl. Res.
9
,
275
288
PMID:
[PubMed]
81
Carloni
,
S.
,
Albertini
,
M.C.
,
Galluzzi
,
L.
,
Buonocore
,
G.
,
Proietti
,
F.
and
Balduini
,
W.
(
2014
)
Increased autophagy reduces endoplasmic reticulum stress after neonatal hypoxia–ischemia: role of protein synthesis and autophagic pathways
.
Exp. Neurol.
255
,
103
112
doi:
82
Zhang
,
L.
,
Lopez
,
H.
,
George
,
N.M.
,
Liu
,
X.
,
Pang
,
X.
and
Luo
,
X.
(
2011
)
Selective involvement of BH3-only proteins and differential targets of Noxa in diverse apoptotic pathways
.
Cell Death Differ.
18
,
864
873
doi:
83
Wang
,
X.
,
Han
,
W.
,
Du
,
X.
,
Zhu
,
C.
,
Carlsson
,
Y.
,
Mallard
,
C.
et al
(
2010
)
Neuroprotective effect of Bax-inhibiting peptide on neonatal brain injury
.
Stroke
41
,
2050
2055
doi:
84
Puthalakath
,
H.
,
O'Reilly
,
L.A.
,
Gunn
,
P.
,
Lee
,
L.
,
Kelly
,
P.N.
,
Huntington
,
N.D.
et al
(
2007
)
ER stress triggers apoptosis by activating BH3-only protein Bim
.
Cell
129
,
1337
1349
doi:
85
Urano
,
F.
,
Wang
,
X.Z.
,
Bertolotti
,
A.
,
Zhang
,
Y.
,
Chung
,
P.
,
Harding
,
H.P.
et al
(
2000
)
Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1
.
Science
287
,
664
666
doi:
86
Martín-Pérez
,
R.
,
Niwa
,
M.
and
López-Rivas
,
A.
(
2012
)
ER stress sensitizes cells to TRAIL through down-regulation of FLIP and Mcl-1 and PERK-dependent up-regulation of TRAIL-R2
.
Apoptosis
17
,
349
363
doi:
87
Kichev
,
A.
,
Rousset
,
C.I.
,
Baburamani
,
A.A.
,
Levison
,
S.W.
,
Wood
,
T.L.
,
Gressens
,
P.
et al
(
2014
)
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) signaling and cell death in the immature central nervous system after hypoxia-ischemia and inflammation
.
J. Biol. Chem.
289
,
9430
9439
doi:
88
Nijboer
,
C.H.
,
van der Kooij
,
M.A.
,
van Bel
,
F.
,
Ohl
,
F.
,
Heijnen
,
C.J.
and
Kavelaars
,
A.
(
2010
)
Inhibition of the JNK/AP-1 pathway reduces neuronal death and improves behavioral outcome after neonatal hypoxic–ischemic brain injury
.
Brain Behav. Immun.
24
,
812
821
doi:
89
Nijboer
,
C.H.
,
Heijnen
,
C.J.
,
van der Kooij
,
M.A.
,
Zijlstra
,
J.
,
van Velthoven
,
C.T.J.
,
Culmsee
,
C.
et al
(
2011
)
Targeting the p53 pathway to protect the neonatal ischemic brain
.
Ann. Neurol.
70
,
255
264
doi:
90
Pirianov
,
G.
,
Brywe
,
K.G.
,
Mallard
,
C.
,
Edwards
,
A.D.
,
Flavell
,
R.A.
,
Hagberg
,
H.
et al
(
2007
)
Deletion of the c-Jun N-terminal kinase 3 gene protects neonatal mice against cerebral hypoxic–ischaemic injury
.
J. Cereb. Blood Flow Metab.
27
,
1022
1032
doi:
91
Chavez-Valdez
,
R.
,
Martin
,
L.J.
,
Flock
,
D.L.
and
Northington
,
F.J.
(
2012
)
Necrostatin-1 attenuates mitochondrial dysfunction in neurons and astrocytes following neonatal hypoxia–ischemia
.
Neuroscience
219
,
192
203
doi:
92
Chavez-Valdez
,
R.
,
Flock
,
D.L.
,
Martin
,
L.J.
and
Northington
,
F.J.
(
2016
)
Endoplasmic reticulum pathology and stress response in neurons precede programmed necrosis after neonatal hypoxia-ischemia
.
Int. J. Dev. Neurosci.
48
,
58
70
doi:
93
Høyer-Hansen
,
M.
and
Jäättelä
,
M.
(
2007
)
Connecting endoplasmic reticulum stress to autophagy by unfolded protein response and calcium
.
Cell Death Differ.
14
,
1576
1582
doi:
94
Peng
,
M.
,
Ostrovsky
,
J.
,
Kwon
,
Y.J.
,
Polyak
,
E.
,
Licata
,
J.
,
Tsukikawa
,
M.
et al
(
2015
)
Inhibiting cytosolic translation and autophagy improves health in mitochondrial disease
.
Hum. Mol. Genet.
24
,
4829
4847
doi:
95
Xie
,
C.
,
Ginet
,
V.
,
Sun
,
Y.
,
Koike
,
M.
,
Zhou
,
K.
,
Li
,
T.
et al
(
2016
)
Neuroprotection by selective neuronal deletion of Atg7 in neonatal brain injury
.
Autophagy
12
,
410
423
doi:
96
Xu
,
Y.
,
Tian
,
Y.
,
Tian
,
Y.
,
Li
,
X.
and
Zhao
,
P.
(
2016
)
Autophagy activation involved in hypoxic-ischemic brain injury induces cognitive and memory impairment in neonatal rats
.
J. Neurochem.
139
,
795
805
doi:
97
Li
,
G.
,
Scull
,
C.
,
Ozcan
,
L.
and
Tabas
,
I.
(
2010
)
NADPH oxidase links endoplasmic reticulum stress, oxidative stress, and PKR activation to induce apoptosis
.
J. Cell Biol.
191
,
1113
1125
doi:
98
Wei
,
P.-C.
,
Hsieh
,
Y.-H.
,
Su
,
M.-I.
,
Jiang
,
X.
,
Hsu
,
P.-H.
,
Lo
,
W.-T.
et al
(
2012
)
Loss of the oxidative stress sensor NPGPx compromises GRP78 chaperone activity and induces systemic disease
.
Mol. Cell
48
,
747
759
doi:
99
Zeeshan
,
H.M.
,
Lee
,
G.H.
,
Kim
,
H.-R.
and
Chae
,
H.-J.
(
2016
)
Endoplasmic reticulum stress and associated ROS
.
Int. J. Mol. Sci.
17
,
327
doi:
100
Hu
,
Y.
,
Wang
,
Z.
,
Pan
,
S.
,
Fang
,
M.
,
Jiang
,
H.
,
Mao
,
Y.
et al
(
2017
)
Inhibition of endoplasmic reticulum stress is involved in the neuroprotective effect of aFGF in neonatal hypoxic-ischaemic brain injury
.
Oncotarget
,
in press
doi:
101
Bai
,
X.
,
Liu
,
S.
,
Yuan
,
L.
,
Xie
,
Y.
,
Li
,
T.
,
Wang
,
L.
et al
(
2016
)
Hydrogen-rich saline mediates neuroprotection through the regulation of endoplasmic reticulum stress and autophagy under hypoxia-ischemia neonatal brain injury in mice
.
Brain Res.
1646
,
410
417
doi:
102
Carloni
,
S.
,
Albertini
,
M.C.
,
Galluzzi
,
L.
,
Buonocore
,
G.
,
Proietti
,
F.
and
Balduini
,
W.
(
2014
)
Melatonin reduces endoplasmic reticulum stress and preserves sirtuin 1 expression in neuronal cells of newborn rats after hypoxia-ischemia
.
J. Pineal Res.
57
,
192
199
doi:
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).