You are on page 1of 9

The Journal of Maternal-Fetal & Neonatal Medicine

ISSN: 1476-7058 (Print) 1476-4954 (Online) Journal homepage: http://www.tandfonline.com/loi/ijmf20

Neonatal hypoxic–ischemic encephalopathy:


emerging therapeutic strategies based on
pathophysiologic phases of the injury

Qiuli Wang, Hongyan Lv, Lixin Lu, Pengshun Ren & Lianxiang Li

To cite this article: Qiuli Wang, Hongyan Lv, Lixin Lu, Pengshun Ren & Lianxiang Li (2018):
Neonatal hypoxic–ischemic encephalopathy: emerging therapeutic strategies based on
pathophysiologic phases of the injury, The Journal of Maternal-Fetal & Neonatal Medicine, DOI:
10.1080/14767058.2018.1468881

To link to this article: https://doi.org/10.1080/14767058.2018.1468881

Accepted author version posted online: 22


Apr 2018.
Published online: 02 May 2018.

Submit your article to this journal

Article views: 41

View related articles

View Crossmark data

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


http://www.tandfonline.com/action/journalInformation?journalCode=ijmf20
THE JOURNAL OF MATERNAL-FETAL & NEONATAL MEDICINE
https://doi.org/10.1080/14767058.2018.1468881

REVIEW ARTICLE

Neonatal hypoxic–ischemic encephalopathy: emerging therapeutic strategies


based on pathophysiologic phases of the injury
Qiuli Wanga, Hongyan Lva,b, Lixin Luc, Pengshun Rena and Lianxiang Lib,d
a
Department of Neonatology, Handan Maternal and Child Health Care Hospital, Handan, PR China; bDepartment of Neonatal
Pathology, Handan Maternal and Child Health Care Hospital, Handan, PR China; cDepartment of Pediatrics, Handan 7th Hospital,
Handan, PR China; dDepartment of Neural Development and Neural Pathology, Hebei University of Engineering School of Medicine,
Handan, PR China

ABSTRACT ARTICLE HISTORY


Neonatal hypoxic ischemic encephalopathy (HIE) is an important cause of neonatal death and Received 5 February 2018
disability. At present, there is no unified standard and specialized treatment method for neonatal Accepted 20 April 2018
HIE. In clinical practice, we have found that a gap remains between preclinical medical research
KEYWORDS
and clinical application in the treatment of neonatal HIE. To promote an organic combination of
Cerebral hypoxic ischemic;
preclinical research and clinical application, we propose the different phases as intervention tar- erythropoietin; hypothermia;
gets, based on the pathophysiologic changes in phases I, II, and III of neonatal HIE; moreover, newborn; rehabilitation;
we suggest transformative medicine as a principle that may improve the therapeutic effect by stem cell transplantation;
blocking the progression of the disease to an irreversible stage. For instance, in phase I, mild therapeutics
hypothermia, free radical scavenger (erythropoietin, hydrogen-rich saline), excitatory amino acid
receptor blocker, and neuroprotective agents should be administered to neonates with moder-
ate/severe HIE; in phase II, following phase I treatment, anti-inflammatory agents, neuroprotec-
tive or nerve regeneration agents, and stem cell transplantation should be administered to
patients; in phase III, anti-inflammatory agents, neuroprotective or nerve regeneration agents,
and stem cell transplantation should be administered to patients. As soon as the patient’s condi-
tion has stabilized, acupuncture, massage, and rehabilitation training should be performed.
Following further study of stem cells, stem cell transplantation is expected to become the most
promising therapeutic candidate for treatment of severe neonatal HIE with its sequelae.

Introduction additionally, the intervention methods are inadequate,


since the intervention targets and critical time window
Neonatal hypoxic–ischemic encephalopathy (HIE) is a
are not fully known. Additionally, there is a certain
kind of brain injury caused by the severe reduction of
gap between preclinical medical research and its clin-
cerebral oxygen and/or blood supply in the perinatal
ical application.
period [1]. According to current reports, approximately
Animal experimentation indicates that pathophysio-
1 million newborns worldwide die from neonatal HIE
logical changes in neonatal HIE can be divided into
each. The incidence of neonatal HIE is 1–2/1000 live phases I, II, and III. There are significant differences in
births, of which 10–15% of the infants succumb and the pathophysiology and biochemistry of the brain tis-
25–30% suffer permanent neurological damage, sues in each of these phases. According to the patho-
including cerebral palsy, epilepsy, mental disorders, physiological changes of the different phases, a
vision and hearing impairment, and cognition and corresponding intervention is administered, which has
learning disabilities [2,3]. Thus, it places a great mental been closely designed based on the experimental
and economic burden on the family and has become pathology outcome. The time of hospitalization after
one of the primary global medical problems. In the onset of the injury for each patient is highly inconsist-
face of this challenge, a large number of scholars have ent in clinical practice, suggesting that the pathophysi-
conducted numerous animal experiments and clinical ology phases are also different within different
studies. Nonetheless, therapy for neonatal HIE remains patients. Without consideration of these factors, there
largely unsatisfactory, which partly arises from the lack is a degree of blindness in the therapeutic process,
of a unified standard treatment for newborn HIE; which, ultimately, prevents achievement of the

CONTACT Hongyan Lv 3581479478@qq.com Departments of Neonatology and Neonatal Pathology, Handan Maternal and Child Health Care
Hospital, No. 6, Li Ming Street, Peace Road, Handan City, Hebei Province, China
ß 2018 Informa UK Limited, trading as Taylor & Francis Group
2 Q. WANG ET AL.

desired results. Another problem arises when the inter- cell apoptosis begins. In addition, as inflammatory fac-
vention results of animal experiments are transformed tors become involved and brain injury is more sub-
into clinical practice, as there often remains a gap in stantial [8], phase II has a close relationship with the
treatment. If the preclinical medical research is organ- onset of epileptic seizures. Therefore, the treatment
ically combined with clinical applications, it is clearly strategies during phase II follow the treatments
beneficial to the patient. Therefore, this review is employed during phase I: patients should be treated
designed to describe the changes in pathophysiologic with anti-inflammatory agents, neuroprotective, or
phases I, II, and III of neonatal HIE; further, in the con- nerve regenerating agents (e.g. nerve growth factor),
text of transformative medicine, the intervention tar- and stem cell transplantation. (3) Injury repair or
gets in different periods are discussed, with the aim of chronic inflammation phase (phase III): this phase
achieving a better therapeutic effect. occurs >72 h after the onset of neonatal HIE, and lasts
for days or months. In accordance with the severity of
the disease, the duration of hypoxia–ischemia, and the
Intervention measures and pathophysiology
effects of prior therapeutic interventions, there are
phases of neonatal hypoxic–ischemic
generally two outcomes: one involves recovery, where
encephalopathy
the damaged brain tissue enters the repair process,
The pathological and biochemical changes of neonatal and the surviving neurons and glial cells begin to dif-
HIE constitute a cascading reaction process, where ferentiate, proliferate, and regenerate; in the other out-
apoptosis or necrosis of nerve cells is the final out- come, the injured tissue continues to deteriorate. Here,
come of brain damage. Animal experiments show that as inflammation continues to increase and persist, the
the pathophysiological process of neonatal HIE is div- damaged brain tissues lose the support of neuro-
ided into three phases as follows: (1). primary energy trophic factors. Surviving microglia and astrocytes con-
failure phase (phase I): this phase occurs 0–6 h after tinue to release harmful cytokines, which promote
the hypoxic–ischemic injury. Under normal circumstan- additional neuronal death and axonal injury, leading
ces, maintenance of the cerebral blood flow is to further aggravation of the injured brain tissues [9].
dependent on systemic blood pressure; however, due Therefore, if a patient continues to deteriorate upon
to insufficient blood supply in the placenta (such as reaching phase III, treatment should include anti-
umbilical cord around the neck, dystocia, placenta pre- inflammatory agents, neuroprotective, or nerve regen-
via), a variety of changes occur, including hypoxia–i- erating agents (e.g. nerve growth factor), and stem cell
schemia and acidosis in tissues and organs, reduced transplantation. Once the patient’s condition has stabi-
myocardial contractility, decreased arterial blood pres- lized, acupuncture, massage, and rehabilitation should
sure, and reduced cerebral blood flow [4]. Due to brain be administered to the patient as early as possible
tissue hypoxia–ischemia, glucose anaerobic fermenta- (Figure 1).
tion is enhanced in nerve cells, thereby reducing the
adenosine triphosphate (ATP) in the cells. This leads to
Evidence of the neuroprotective effects of
high energy storage depletion; ion pump dysfunction;
existing intervention measures
and Naþ, Ca2þþ transport hindrance; it also causes
accumulation of lactic acid, free radicals, and excitatory Free radical inhibitors and scavengers
amino acids, as well as mitochondrial dysfunction,
Melatonin
nerve cell edema, and eventual apoptosis (or cell
death) [5,6]. Therefore, the treatment strategies during Melatonin is an amine hormone secreted by the pineal
phase I utilize conventional methods: patients should gland, with strong anti-inflammatory and free radical-
be treated with hypothermia, free radical scavengers scavenging effects. Notably, melatonin can directly
(e.g. melatonin, erythropoietin, hydrogen-rich saline, or induce the production of glutathione peroxidase,
coenzyme Q10), excitatory amino acid receptor block- glutathione reductase, 6-phosphate glucose dehydro-
ing agents, and/or neuroprotective agents. (2) genase, and superoxide dismutase; moreover, it
Secondary energy failure phase (phase II): this phase increases the efficiency of mitochondrial electron
occurs 6–72 h after hypoxia–ischemia. Excitatory neuro- transport. Animal experiments have shown that mela-
transmitters and free radicals continue to be released, tonin can reduce the recruitment and activation of
mitochondrial dysfunction becomes more serious, and glial cells after neonatal hypoxic brain injury [10]; it
large phosphorus reserves are depleted [7]; as the can also promote the formation of myelin sheath and
phase progresses, cytochrome C escapes from mito- improve the survival of brain neurons [11].
chondria, the cascade reaction is activated, and nerve Importantly, melatonin is much more capable of
THE JOURNAL OF MATERNAL-FETAL & NEONATAL MEDICINE 3

Figure 1. Pathophysiology phases of neonatal hypoxic-ischemic encephalopathy and its treatment strategies.

scavenging hydroxyl radicals than mannitol, glutathi- neonates with moderate/or severe HIE, EPO (200 IU/kg)
one, or vitamin E. Recent clinical trials have shown can promote the early recovery of neonatal neuro-
that, for neonates with moderate/or severe HIE, mela- logical function [13] and improve infant fine motor
tonin therapy combined with mild hypothermia can and language skills, as well as the development of per-
reduce oxidative stress, thereby improving survival and ception [14]. The use of EPO (300 or 500 IU/kg) for
neurodevelopmental outcomes at 6 months of age neonates with HIE can improve intelligence and motor
[12]. Therefore, as melatonin is a safe, non-toxic, effect- development, thus reducing the incidence of cerebral
ive, and promising neuroprotective drug for neonatal palsy and the rate of disability [15]. Low doses of
HIE, it is worth popularizing. erythropoietin for neonatal HIE have been demon-
strated to be safe. Recently, a phase II EPO trial
Erythropoietin (EPO) showed that high doses of EPO (1000 IU/kg) for neo-
nates with HIE can alleviate the severity of brain injury
EPO is a 34-kDa glycoprotein hormone, which is widely
(as detected by magnetic resonance imaging) and
used in treatment of anemia, renal failure, cancer, pre-
improve motor function at 1 year of age [16]. EPO
maturity, and chronic inflammatory diseases. More
may serve as a neuroprotective drug for neonatal HIE;
recently, EPO has been shown to exhibit an obvious
however, the optimal dosage, the long-term effect of
neuroprotective effect. Many animal experiments have
nerve function improvement, and the therapeutic
demonstrated that EPO can promote neuron differenti-
mechanism of EPO still require further study.
ation and regeneration in cases of neonatal HIE; more-
over, it can inhibit nerve cell apoptosis, as well as
Hydrogen-rich saline
reduce and regulate the inflammatory response and
anti-oxidative damage, thereby promoting vascular Hydrogen is one of the most renowned medical gases,
regeneration and improving microcirculation. as it exhibits a powerful ability to scavenge free radi-
Additionally, EPO can prevent glutamic excitotoxicity cals, and can improve the activity of peroxidase dismu-
and brain edema. Our previous study showed that, for tase, enhance the activity of mitochondrial respiratory
4 Q. WANG ET AL.

chain enzyme, and provide an anti-apoptotic effect. In neurofascin 155 (NF 155), as well as related lipid rafts,
a rat model of cerebral ischemia, different concentra- thereby preventing damage to the myelin sheath [23].
tions of hydrogen-rich saline have been shown to Clinical trials have shown that the ganglioside therapy
reduce the infarct size and expression of inflammatory (20 mg/d) for neonates with HIE can significantly
factors [17], as well as to improve cognitive function shorten the recovery times of primitive reflexes,
[18]. Because the study of hydrogen as a therapy for muscle tension, and consciousness [24].
neonatal HIE is still in the initial stages, further investi-
gation is needed regarding the ideal dosage and form Nerve growth factor (NGF)
of hydrogen to be administered to infants; moreover, Animal experiments showed that, in neonatal HIE, NGF
the range of therapeutic time, standard application therapy could inhibit neuronal apoptosis, promote the
method, pharmacokinetics, and toxicology remain to activation and regeneration of endogenous neural
be determined. stem cells, and reduce the degeneration and necrosis
of neurons. NGF is effective in promoting the recovery
Anti-inflammatory treatment of nerve function and long-term nerve development
[25]; additionally, clinical trials have shown that NGF
Minocycline can significantly improve neurological function and
In vitro cell culture and in vivo animal experiments reduce mortality in neonates with HIE [26].
have shown that minocycline can protect neurons by
inhibiting the proliferation and activation of microglia, Therapeutic hypothermia
thereby reducing neuronal death [19,20]. If a patient is Hypothermia is regarded as a good therapeutic meas-
treated with a sufficient dose of minocycline within a ure for moderate/severe neonatal HIE. Currently, the
few hours to days after the onset of HIE, the patient neuroprotection mechanism of hypothermia is not
may experience a reduction in brain tissue damage clear; it may be related to a variety of factors: reduc-
caused by an excessive inflammatory reaction. tion of glutamate release, reduction of the metabolism
and energy consumption of brain tissue, reduction of
Glucocorticoid hormone acidosis in neurons and promotion of protein synthe-
Glucocorticoid can increase the use of glucose in the sis, reduction of the synthesis of leukotriene, reduction
brain, alleviate the inflammatory response, and inhibit of blood–brain barrier damage and brain edema,
apoptosis; thus, it has a significant ability to reduce inhibition of free radicals, inhibition of cell apoptosis,
HIE. Currently, glucocorticoid is recommended for pre- and/or inhibition of free radical generation and lipid
natal use, but cannot be used immediately after birth peroxide reaction.
[21]. Therefore, clinical data regarding glucocorticoid The best treatment window for hypothermia begins
therapy for neonatal HIE remain scarce. Notably, the within 6 h after birth and must be maintained for
ideal dosage of glucocorticoid hormone, the optimal 48–72 h; the patient’s rectal temperature must be
timing of glucocorticoid application, and the ability of maintained at 34.5–35  C. Most clinical trials have
glucocorticoid to improve prognosis in neonatal HIE, shown that hypothermia can reduce the mortality and
risk of neurodevelopmental disabilities in neonates
are key areas for future research.
with moderate/severe HIE [27–29]. However, some
reports indicate that, in neonates with HIE who have
Neuroprotection and neural regeneration agents undergone therapeutic hypothermia, 44–53% of
Gangliosides patients still died or were left with moderate or severe
neurological dysfunction [30]. There are also reports
Gangliosides serve as an important component of that whereas hypothermia for neonates with HIE
nerve tissue. Exogenous gangliosides exhibit a strong (defined in those studies as cooling to 33–35  C for
affinity for brain tissue; they can promote the regener- 72 h within the first 6 h of life) can improve the neuro-
ation and repair of damaged nerves. In a rat model, logical outcome in survivors, it does not significantly
ganglioside treatment (0.02 mg) can increase the acidic reduce the combined rate of death or severe disability
content of myelin basic protein and glial fibrillary [31]. There have also been some adverse reactions
acidic protein in brain tissue; further, it can reduce reported during hypothermia, including arrhythmias,
neurobehavioral abnormalities in neonatal rats [22]. In septic episodes, and epileptic attack [32]; in one
addition, gangliosides can repair the structure of mye- report, there was an increase in the risk of death in
lin sheath and axon lipid rafts that incorporate neonates [33]. Despite this contradictory data,
THE JOURNAL OF MATERNAL-FETAL & NEONATAL MEDICINE 5

Table 1. The summary of animal experiment outcome and clinical application of the stem cell transplantation for neonatal hypo-
xic–ischemic encephalopathy.
Receiving Delivery
Cell type subject Delivery time route Outcome Author [Reference]
NSCs
hFNSCs Rat 3 d after HIE CVD After stem cell transplantation, it is distribution in the Qu et al. [34]
damaged cortex and hippocampus, and stem cell
differentiate into neurons and astrocytes
hENSCs Rat 24 h after HIE F.D After stem cell transplantation, a lot of microglia cell Daadi et al. [35]
appear, and it can enhance the repair of endogen-
ous brain tissue, motor function is improved
ES-NPCs Rat 40 h after HIE DMC ES-NPCs can differentiate to cortical neurons with pyr- Shinoyama et al. [36]
amidal morphology, the deep layer-specific marker
is expressed, the motor function is improved
hENSCs Rat 24 h after HIE IND hNSCs extensively migrated into brain areas within Ji et al [37]
24 h after stem cell, delivery, hNSCs reduced the
up-regulation of cytosolic IL-1b, p-IjBa, and NF-jB
p65 level, neurological outcomes are improved
hFNSCs Human infant 75 d after HIE CVD twenty-eight days after stem cell transplantation, the Luan et al. [38]
patient’s myotonia is remarkable improved, the
patient’s intelligence the motor function
is improved
hNPCs Human newborn 4–20 d after HIE CVD Two days after transplantation, the sucking and swal- Luan et al. [39]
lowing reflex are appeared
gradually in all patients, muscular tension is also
improved, and convulsion
stopped. Six cases are followed up for 12 months,
four cases are normal
development in psychomotor cores of each scale
reached normal level,
two cases have cerebral palsy
hUCB-MCs
hUCB-MCs Rat 3 h after HIE PCD hUCB-MCs can promote sensorimotor reflexes of rats, Pimentel-Coelno Co,
the number of activated et al. [40]
microglial cells in the cerebral cortex is decreased,
neurological function is improved
hUCB-MCs Rat 24 h after HIE A.B Learning ability and long-term spatial memory are sig- Greggio et al. [42]
nificantly improved
hUCB-MCs Rat 7 d after HIE V.B hUCB-MCs can increase the neurotrophic factor, the Yasuhara et al. [44]
neurological behavior of rats are improved
HIE: neonatal hypoxic ischemic encephalopathy; NSCs: neural stem cells; hFNSCs: human fetal neural stem cells; hENSCs: human embryonic neural stem
cells; hNPCs: human neural precursor cells; ES-NPCs: embryonic stem cell-derived neural progenitor cells; hUCB-MCs: human umbilical cord blood mono-
nuclear cells; CVD: cerebral ventricle delivery; FD: forebrain deliver; IND: intranasally delivery; IHT: intrahippocampal delivery; ICD: intracardial delivery;
ICD: intracerebral delivery; PCD: peritoneal cavity delivery; VB: vein delivery; AB: artery delivery.

hypothermia is an indispensable method to treat neo- ventricle of HIE neonatal rats, the hNSCs can reach the
natal HIE; it minimizes the brain tissue damage in the areas of brain damage along the subventricle zone
acute phase of neonatal HIE and delays or curbs the and corpus callosum, especially in the cerebral cortex
development of the disease, thereby providing valu- and hippocampus; 85% of the transplanted cells form
able time to attempt other treatments. hNuc-NF or hNuc-tubulin positive cells in the cerebral
cortex, whereas 60% of the transplanted cells differen-
Stem cell transplantation tiate into hNuc-GFAP-positive cells [34]. Stem cells also
may facilitate endogenous brain repair [35]. When
Neural stem cells (NSCs) neural precursor cells (derived from embryonic stem
NSCs widely exist in the embryonic and adult nervous cells) are implanted into the deep motor cortex of HIE
system; importantly, they can differentiate into neu- newborn rats, neural precursor cells can differentiate
rons, astrocytes, and oligodendrocytes, both in vivo into cortical neurons and motor function is improved,
and in vitro. Exogenous stem cell transplantation for within 3 weeks post-implantation [36]. When hNSCs
neonatal HIE has aroused the interest of many scholars are implanted into the nasal cavity of HIE newborn
and shows promising preliminary results (Table 1). The rats, hNSCs become widely distributed in different
delivery routes for stem cell transplantation include brain regions; thus, multiple cytokines (LI-1b, P-Lkba,
intracerebral, intrathecal, arterial, intravenous, intraperi- NF-Kb, p65) exhibit decreased expression and neuro-
toneal, and intranasal approaches. When human logical function is improved [37]. The noninvasive
embryonic NSCs (hNSCs) are transplanted into the nasal cavity implantation provides a simple and easy
6 Q. WANG ET AL.

method to perform stem cell transplantation in the reduce neuronal apoptosis in the cerebral cortex and
future. The clinical application of NSCs has been pre- hippocampus of rats with hypoxic-ischemic brain
liminarily attempted in infants with HIE. Luan et al. injury [45], enhance learning and memory abilities,
[38] reported that hNSCs were transplanted into one and upregulate brain-derived neurotrophic factor
infant with neurological disability that was caused by (BDNF) and glial cell-derived neurotrophic factor (NGF)
severe HIE (implantation at 75 d after birth); 28 d after [46]. The therapeutic mechanism may be related to
stem cell transplantation, the infant’s myotonia, intelli- the opening of K-ATP channels, inhibition of c-Fos and
gence, and movement were remarkably improved, c-Jun immediate genes in the early hypoxic–ischemic
such that the infant became similar to normal infants stage, and inhibition of neuronal apoptosis.
of the same age. In another study, human neural pro- Acupuncture for cerebral palsy in children can improve
genitor cells were implanted in six cases of neonatal
fine motor skills by 67.7%, increase the grip index by
HIE; the second day after cell transplantation, all
87.1%, and improve the visual perception index [47]. It
patients’ sucking and swallowing reflexes appeared,
has been reported that the fine motor, gross motor,
convulsions stopped, and muscle tension was
language, and other aspects can be improved in cases
improved. Importantly, all patients were re-examined
of early neonatal brain injury, using traditional Chinese
at 12 months of age; four cases exhibited normal men-
medicine massage [48]. However, a comprehensive
tal motor development, whereas two cases presented
with cerebral palsy [39]. The above studies have pro- analysis indicated that the inclusion criteria and con-
vided very valuable experience for further research trasting experiment regarding acupuncture in the
and clinical applications. patient serve as defects in the design of the experi-
ment; thus, the authors of that analysis do not recom-
Cord blood mononuclear cells mend acupuncture for neonatal HIE [49]. The study
design regarding acupuncture and massage for neo-
Cord blood cell transplantation exhibits a strong natal HIE should be further improved, along with an
potential for nerve regeneration. Notably, these stem
increase in the clinical sample size, to correctly evalu-
cells are easier to obtain than other stem cells. Animal
ate its effect.
experiments have shown that cord blood mononuclear
cells can reduce the activity of microglia, inhibit neur-
onal cell death, and promote the recovery of sensori- Conclusion
motor reflex function [40]. Cord blood mononuclear
To improve the clinical treatment of neonatal HIE, this
cells can promote the differentiation of endogenous
review aimed to provide the context of transform-
neural stem cells into mature neurons to aid in the
ational medicine, and to describe the pathophysiologic
recovery and development of damaged neurons [41];
moreover, cord blood mononuclear cells significantly changes of different phases in neonatal HIE as key
improve learning ability and long-term spatial memory treatment targets for novel intervention strategies,
in rats [42,43] and can promote the up-regulation of according to the different phases.
neurotrophic factor in the brain after cell transplant-
ation [44]. Therefore, cord blood cell transplantation Disclosure statement
may be the most potent therapeutic candidate for
neonatal HIE, with wide application prospects. No potential conflict of interest was reported by the authors.
However, the optimal timing, dosage, and delivery of
stem cell transplantation for neonatal HIE will require Funding
further study.
This study is supported by Health and Family Planning
Commission of Hebei (No 20150033). Science and
Acupuncture and massage Technology Bureau of Handan, City, Hebei Province (No.
152810879-6); Science and Technology Department of Hebei
Acupuncture and massage are effective and unique
Province (No. 162777201).
methods for many neurological diseases with sequelae.
These methods are very helpful for promoting the
recovery of neurological function and reducing the References
degree of disability. In recent years, there have been [1] Mattiesen WR, Tauber SC, Gerber J, et al. Increased
an increasing number of preliminary reports regarding neurogenesis after hypoxic–ischemic encephalopathy
their usage in the treatment of neonatal HIE. Animal in humans is age related. Acta Neuropathol.
experimentation has shown that acupuncture can 2009;117(5):525–534.
THE JOURNAL OF MATERNAL-FETAL & NEONATAL MEDICINE 7

[2] Gulczyn ska E, Gadzinowski J. Therapeutic hypothermia [19] Filipovic R, Zecevic N. Neuroprotective role of mino-
for neonatal hypoxic-ischemic encephalopathy. cycline in co-cultures of human fetal neurons and
Ginekol Pol. 2012;83(3):214–218. microglia. Exp Neurol. 2008;211(1):41–51.
[3] Tagin MA, Woolcott CG, Vincer MJ, et al. Hypothermia [20] Ofek-Shlomai N, Berger I. Inflammatory injury to the
for neonatal hypoxic ischemic encephalopathy: an neonatal brain – what can we do? Front Pediatr.
updated systematic review and meta-analysis. Arch 2014;2:30.
Pediatr Adolesc Med. 2012;166(6):558–566. [21] Shinwell ES, Karplus M, Reich D, et al. Early postnatal
[4] Douglas-Escobar M, Weiss MD. Hypoxic-ischemic dexamethasone treatment and increased incidence of
encephalopathy: a review for the Clinician. JAMA cerebral palsy. Arch Dis Child Fetal Neonatal Ed.
Pediatr. 2015;169(4):397–403. 2000;83(3):F177–F181.
[5] Distefano G, Pratico  AD. Actualities on molecular [22] Rong X, Zhou W, Xiao-Wen C, et al. Ganglioside GM1
pathogenesis and repairing processes of cerebral reduces white matter damage in neonatal rats. Acta
damage in perinatal hypoxic-ischemic encephalop- Neurobiol Exp (Wars). 2013;73(3):379–386.
athy. Ital J Pediatr. 2010;36:63. [23] Zhang YP, Huang QL, Zhao CM, et al. GM1 improves
[6] Lai MC, Yang SN. Perinatal hypoxic-ischemic encephal- neurofascin155 association with lipid rafts and pre-
opathy. J Biomed Biotechnol. 2011;2011:609813. vents rat brain myelin injury after hypoxia–ischemia.
[7] Dixon BJ, Reis C, Ho WM, et al. Neuroprotective strat- Braz J Med Biol Res. 2011;44(6):553–561.
egies after neonatal hypoxic ischemic encephalop- [24] Chen JH. Clinical observation on curative effect of
athy. Int J Mol Sci. 2015;16(9):22368–22401. monosialoganglioside in treatment of neonatal hyp-
[8] Leonardo CC, Pennypacker KR. Neuroinflammation oxic-ischemic encephapathy. Qiqihar Yixueyuan
and MMPs: potential therapeutic targets in neonatal Xuebao. 2014;35(22):3300–3302.
hypoxic–ischemic injury. J Neuroinflamm. 2009;6:13. [25] Hu W, Gong SP, Lan Y, et al. Short term protective
[9] Juul SE, Ferriero DM. Pharmacologic neuroprotective effect and long term influence of neural growth factor
strategies in neonatal brain injury. Clin Perinatol. in neonatal rats following hypoxic ischemic brain
2014;41(1):119–131. damage. Zhongguo Ertong Baojian Zazhi. 2007;15(1):
[10] Balduini W, Carloni S, Perrone S, et al. The use of 54–56.
melatonin in hypoxic-ischemic brain damage: an [26] Guo L. Clinical effect of rat nerve growth factor in
experimental study. J Matern Fetal Neonatal Med. treatment of hypoxic–ischemic encephalopathy. Jilin
2012;25:119–124. Yixue Zazhi. 2015;36(7):1306–1130.
[11] Alonso-Alconada D, Alvarez A, Lacalle J, et al. [27] Hagmann CF, Brotschi B, Bernet V, et al. Hypothermia
Histological study of the protective effect of mela- for perinatal asphyxial encephalopathy. Swiss Med
tonin on neural cells after neonatal hypoxia–ischemia. Wkly. 2011;141:w13145.
Histol Histopathol. 2012;27(6):771–783. [28] Higgins RD, Raju T, Edwards AD, et al. Hypothermia
[12] Aly H, Elmahdy H, El-Dib M, et al. Melatonin use for and other treatment options for neonatal encephalop-
neuroprotection in perinatal asphyxia: a randomized athy: an executive summary of the Eunice Kennedy
controlled pilot study. J Perinatol. 2015;35(3):186–191. Shriver NICHD workshop. J Pediatr. 2011;159(5):
[13] Lv HY, Wu SJ, Wang QL, et al. Effect of erythropoietin 851–858.e1.
combined with hypothermia on serum Tau protein [29] Chalak LF, Sanchez PJ, Adams-Huet B, et al.
levels and neurodevelopmental outcome in neonates Biomarkers for severity of neonatal hypoxic-ischemic
with hypoxic–ischemic encephalopathy. Neural Regen encephalopathy and outcomes in newborns receiving
Res. 2017;12(10):1655–1663. hypothermia therapy. J Pediatr. 2014;164(3):
[14] Wang YJ, Pan KL, Zhao XL, et al. Therapeutic effects 468–474.e1.
of erythropoietin on hypoxic–ischemic encephalop- [30] Wu YW, Gonzalez FF. Erythropoietin: a novel therapy
athy in neonates. Zhongguo Dang Dai Er Ke Za Zhi. for hypoxic–ischaemic encephalopathy? Dev Med
2011;13(11):855–858. Child Neurol. 2015;57(Suppl 3):34–39.
[15] Zhu C, Kang W, Xu F, et al. Erythropoietin improved [31] Azzopardi DV, Strohm B, Edwards AD, et al. Moderate
neurologic outcomes in newborns with hypoxic– hypothermia to treat perinatal asphyxial encephalop-
ischemic encephalopathy. Pediatrics. 2009;124(2): athy. N Engl J Med. 2009;361(14):1349–1358.
e218–e226. [32] Eicher DJ, Wagner CL, Katikaneni LP, et al. Moderate
[16] Wu YW, Mathur AM, Chang T, et al. High-dose erythro- hypothermia in neonatal encephalopathy: efficacy
poietin and hypothermia for hypoxic–ischemic outcomes. Pediatr Neurol. 2005;32(1):11–17.
encephalopathy: a phase II trial. Pediatrics. 2016;137(6): [33] Shankaran S, Laptook AR, Pappas A, et al. Effect of
e20160191. DOI:10.1542/peds.2016-0191. depth and duration of cooling on deaths in the NICU
[17] Li J, Dong Y, Chen H, et al. Protective effects of hydro- among neonates with hypoxic ischemic encephalop-
gen-rich saline in a rat model of permanent focal cerebral athy: a randomized clinical trial. JAMA. 2014;312(24):
ischemia via reducing oxidative stress and inflammatory 2629–2639.
cytokines. Brain Res. 2012;1486:103–111. [34] Qu SQ, Luan Z, Yin GC, et al. Transplantation of
[18] Ge P, Zhao J, Li S, et al. Inhalation of hydrogen gas human fetal neural stem cells into cerebral ventricle
attenuates cognitive impairment in transient cerebral of the neonatal rat following hypoxic–ischemic injury:
ischemia via inhibition of oxidative stress. Neurol Res. survival, migration and differentiation. Zhonghua Er
2012;34(2):187–194. Ke Za Zhi. 2005;43(8):576–579.
8 Q. WANG ET AL.

[35] Daadi MM, Davis AS, Arac A, et al. Human neural [42] Greggio S, de Paula S, Azevedo PN, et al. Intra-arterial
stem cell grafts modify microglial response and transplantation of human umbilical cord blood mono-
enhance axonal sprouting in neonatal hypoxic–ische- nuclear cells in neonatal hypoxic–ischemic rats. Life
mic brain injury. Stroke. 2010;41(3):516–523. Sci. 2014;96(1–2):33–39.
[36] Shinoyama M, Ideguchi M, Kida H, et al. Cortical [43] de paula S, Greggio S, Marinowic DR, et al. The dos-
region-specific engraftment of embryonic stem cell- e–response effect of acute intravenous transplantation
derived neural progenitor cell restores axonal sprout- of human umbilical cord blood cells on brain damage
ing to a subcortical target and achieves motor and spatial memory deficits in neonatal hypoxia–
functional recovery in a mouse model of neonatal ischemia. Neuroscience. 2012;210:431–441.
hypoxic-ischemic brain injury. Front Cell Neurosci. [44] Yasuhara T, Hara K, Maki M, et al. Mannitol facilitates
2013;7:128. neurotrophic factor up-regulation and behavioural
[37] Ji G, Liu M, Zhao XF, et al. NF-kB signaling is involved recovery in neonatal hypoxic–ischaemic rats with
in the effects of intranasally engrafted human neural human umbilical cord blood grafts. J Cell Mol Med.
stem cells on neurofunctional improvements in neo-
2010;14(4):914–921.
natal rat hypoxic–ischemic encephalopathy. CNS
[45] Li XE, Yuan Q, Tang CZ, et al. Effect of acupuncture
Neurosci Ther. 2015;21(12):926–935.
intervention on 14-3-3 expression in cerebral cortex
[38] Luan Z, Yin GC, Hu XH, et al. Treatment of an infant
of hypoxic–ischemic brain damage rats. Zhen Ci Yan
with severe neonatal hypoxic–ischemic encephalop-
Jiu. 2014;39(6):466–471.
athy sequelae with transplantation of human neural
[46] Zhang Y, Lan R, Wang J, et al. Acupuncture reduced
stem cell into cerebral ventricle. Zhonghua Er Ke Za
apoptosis and up-regulated BDNF and GDNF expres-
Zhi. 2005;43(8):580–583; discussion 580.
[39] Luan Z, Liu WP, Qu SQ, et al. Treatment of newborns sion in hippocampus following hypoxia–ischemia
with severe injured brain with transplantation of in neonatal rats. J Ethnopharmacol. 2015;172:
human neural precursor cell. Zhonghua Er Ke Za Zhi. 124–132.
2011;49(6):445–449. [47] Shi BP, Li H, Bo HD, et al. Observation of the thera-
[40] Pimentel-coelho PM, Magalh~aes ES, Lopes LM, et al. peutic effect of acupuncture on hand functions in
Human cord blood transplantation in a neonatal rat children with encephalopathy. Zhongguo Zhenjiu.
model of hypoxic–ischemic brain damage: functional 2004;24(1):15–16.
outcome related to neuroprotection in the striatum. [48] Wei SX, Chen X. The curative effect of early interven-
Stem Cells Dev. 2010;19(3):351–358. tion of traditional Chinese medicine on treating the
[41] Huang HZ, Wen XH, Liu H, et al. Human umbilical prognosis of neonatal brain injury. Zhongguo Shiyong
cord blood mononuclear cell transplantation pro- Shenjing Jibing Zazhi. 2015;18(4):28–29.
motes long-term neurobehavioral functional develop- [49] Wong V, Cheuk DK, Chu V. Acupuncture for hypoxic
ment of newborn SD rats with hypoxic ischemic brain ischemic encephalopathy in neonates. Cochrane
injury. Zhonghua Er Ke Za Zhi. 2013;51(6):460–466. Database Syst Rev. 2013;1(1):CD007968.

You might also like