Abstract
Background
Hypoxic-ischemia (HI), infection/inflammation and reperfusion injury are pathogenic factors of encephalopathy of prematurity, which involves maturational/neurotrophic disturbances in oligodendrocyte progenitor cells (OPC) and neurons/axons. Mesenchymal stem cells (MSCs) might facilitate neuroserpin production, which is neurotrophic for OPC/neurons. This study investigated MSC effects on developmental disturbances after lipopolysaccharide (LPS)-sensitized HI/reperfusion (LHIR) injury and the relation to neuroserpin expression.
Methods
Postnatal day 2 (P2) rat pups received intraperitoneal LPS (5 µg/kg) injection followed by HI (unilateral common-carotid-artery ligation and 6.5% oxygen exposure for 90 min) and post-HI reperfusion (release of ligation). MSCs (5 × 104 cells) were injected into the left lateral ventricle at 24 h post-LHIR. Neurological tests and brain tissue examinations were performed between P5 and P56.
Results
After LHIR injury, MSC therapy significantly reduced cell death in subplate neurons, attenuated axonal damage, and facilitated synaptophysin synthesis in the cortex. It also alleviated OPC maturation arrest and preserved the complexity of myelinated axons in the white matter, leading to cognitive, motor and behavioral functional improvements. These beneficial effects were linked to restored neuroserpin expression in subplate neurons.
Conclusions
MSC therapy ameliorated developmental disturbances after LHIR injury through protection of neuroserpin expression, serving as a promising approach for treating encephalopathy of prematurity.
Impact
-
Neuroserpin is secreted by subplate neurons and may regulate the development of neurons and oligodendrocyte-axon contact for myelination in the premature brain.
-
LPS-sensitized hypoxic-ischemia/reperfusion (LHIR) injury caused the developmental disturbances of neurons/axons and oligodendrocytes, and lowered neuroserpin levels in a neonatal rat model simulating encephalopathy of prematurity.
-
Mesenchymal stem cell therapy alleviated the developmental disturbances after LHIR injury through protection of neuroserpin expression in subplate neurons, offering a new perspective on potential treatment for encephalopathy of prematurity.
Similar content being viewed by others
Introduction
Diffuse white matter injury is the most common form of encephalopathy of prematurity (EoP), leading to neurological deficits such as cognitive impairment, cerebral palsy, or autism spectrum disorder in extremely preterm (<28 weeks of gestation) survivors.1,2,3 Hypoxic ischemia (HI) and infection/inflammation are the two major pathogenic factors of EoP, and may aggravate brain injury through inflammation-sensitized HI, or exert cumulative effects to increase the risk of neurodevelopmental impairment.1,4,5 In addition, post-HI reperfusion with high oxygen concentration during resuscitation may trigger reactive oxygen/nitrogen species production to further damage the brain.1,6
White matter injury in preterm infants is often accompanied by neuronal/axonal diseases with reduction of regional brain volumes detected on magnetic resonance imaging.7,8 Experimental evidence also suggests that EoP may be a complex amalgam of primary destructive diseases and secondary developmental disturbances (or brain dysmaturation) involving pre-myelinating oligodendrocytes (pre-OLs), subplate neurons and axons.7,8 Pre-OLs are the target cells of damage in the white matter,1,9 while subplate neurons in the subcortical white matter play pivotal roles in cortical layer formation, axonal outgrowth, synaptic connections, and myelination in early brain development.10,11 Several experimental studies have demonstrated destructive contributors such as neuroinflammation and neurovascular damage in the premature brain,12,13,14 but few have investigated the developmental disturbances,15,16 which are potentially related to an insufficiency of neurotrophic factors.7
Neuroserpin is a serine protease inhibitor secreted by cortical plate and subplate neurons during early brain development, with its distribution concentrated in the subcortical area between 23 and 32 weeks of gestation,17 which fall within the developmental window of vulnerability for EoP.9 Neuroserpin plays key roles in not only neuronal differentiation, axonogenesis and synaptic refinement,18,19 but also oligodendrocyte-axon interactions for myelination through up-regulation of cell adhesion molecules such as N-cadherin.20,21,22 In addition to its neurotrophic roles, neuroserpin has shown protective effects against neuroinflammation and ischemia/reperfusion-induced injury in experimental studies.23,24,25 Very few studies have investigated the effects of neuroserpin on neonatal brain injury,26 and it remains unclear whether neuroserpin helps ameliorate developmental disturbances in the immature brain after inflammation-sensitized HI/reperfusion injury.
Displaying anti-inflammatory properties as well as providing trophic support with low immunogenicity, mesenchymal stem cells (MSCs) transplantation has emerged as a promising therapy for many neurological disorders,27 and was reported to be effective in animal models of HI and/or inflammation-induced neonatal brain injury.15,16,28,29 MSCs may facilitate neurotrophic factor production through paracrine secretion or up-regulation of endogenous synthesis.27,30 Using an animal model of EoP induced by low-dose lipopolysaccharide (LPS) and subthreshold HI/reperfusion in postnatal day 2 (P2) rat pups (brain maturation equivalent to 23–25 weeks’ gestation in human), this study tested the hypothesis that MSC therapy may alleviate the developmental disturbances after LPS-sensitized HI/reperfusion brain injury to improve neurocognitive, motor and behavioral function through protecting neuroserpin/N-cadherin signaling.
Methods
Animals
Pregnant Sprague-Dawley rats (12–14 days of gestation) were purchased from BioLASCO Co., Ltd Taiwan (Taipei, Taiwan) and housed under standard housing conditions with a 12-h light/dark cycle. After delivery, rat pups were cared for by their mothers. The Institutional Animal Care and Use Committee in Chi Mei Medical Center approved the study protocols, and the reports were written according to the ARRIVE checklist. A total of 120 pups were utilized in the study, and the sample size for each experiment was determined using GPower 3.1 software. During the experiments, rat pups that appeared cyanotic and apneic were removed immediately and provided with fresh air. If the pups did not recover and remained comatose for more than 3 h after the intervention, they were humanely euthanized via cervical dislocation. To reduce the impact of individual variations on the data, pups were eliminated from the study if their experiments had been interrupted. The elimination rate in this study was 15%.
A neonatal rat model of EoP induced by LPS-sensitized HI/reperfusion
The neonatal rat model of EoP employed in this study was adapted from our previously established protocols.4,12 P2 rat pups received an intraperitoneal injection of 5 µg/kg LPS (Escherichia coli O55:B5; L2880, Sigma-Aldrich, St Louis, MO) 3 h before HI. To prevent LPS-induced fluctuations in body temperature, the pups were returned to their dams and kept in an incubator, maintaining a stable temperature of 33–34 °C until HI induction. The HI model involved unilateral ligation of the right common carotid artery, followed by a hypoxic challenge. Under anesthesia with isoflurane (3–5%), pups received an incision on the neck to expose the right common carotid artery, and a temporary ligature was applied using 4-0 surgical silk. After surgery, the pups were allowed to recover for one hour in an incubator. Subsequently, they were placed in a hypoxia chamber maintained at 36–36.5 °C, with humidified 6.5% oxygen flowing at 3 L/min for 90 min. Following the hypoxic period, the ligature was removed, and the artery was gently compressed with forceps to facilitate post-HI reperfusion. The neck wound was sutured, and the pups were returned to their dam for recovery. In this model, rat pups undertaking temporary ligation had more neuroinflammation and brain injury than those receiving permanent ligation (Supplementary Fig. 1), suggesting the additional impact of post-HI reperfusion. The mortality rate in the neonatal rat model was ~10–12%.
Human mesenchymal stem cell culture
Commercially available human MSCs were purchased from Merck (# SCC034, Billerica, MA). The cells were cultured in low-glucose DMEM (#31600034, Gibco, Waltham, MA) with 10% FBS, 0.5% human fibroblast growth factor-basic (#GF003, Millipore, St Louis, MO) and 2 mM L-glutamine at 37 °C. MSCs were identified as CD90+/CD73+/CD105+ and CD34−/CD45− cells using FACS flow cytometry (Beckman Coulter, Indianapolis, IN) and monoclonal antibodies, and further verified by their ability of differentiation into the osteogenic, adipogenic or chondrogenic lineage, as described in our previous study (Supplementary Fig. 2).31
Experimental groups
The P2 rat pups were randomly assigned to three groups—sham control (without LPS, HI or reperfusion), LPS-sensitized HI with reperfusion (LHIR), and LHIR followed by MSC treatment (LHIR + MSC) at 24 h post-insult (P3). The MSCs (5 × 104 cells/5 μl) were intracerebrally injected into the left lateral ventricle under anesthesia according to the published method.32 Male and female pups were randomly allocated in different experimental groups. Rats in each group received sequential tests from P5 to P56 for performance of neurological function, and were sacrificed for brain tissue examinations on P5, P10, P16, and P23 (Supplementary Fig. 3).
Immunofluorescence staining
The rat brains were paraffinized and coronally sectioned (10-μm thick) from the genu of the corpus callosum to the end of the dorsal hippocampus. Four coronal sections, two at the level of the striatum (0.26 mm and 0.92 mm posterior to the bregma) and another two at the levels of the dorsal hippocampus (3.14 mm and 4.16 mm posterior to the bregma), were selected according to a rodent brain atlas and assessed for each brain.33
After blocking, the sections were incubated overnight at 4 °C with one or two of the following primary antibodies. Neuronal markers included anti-complexin 3 (subplate neuron marker; 1:200, #122302, Synaptic Systems, Göttingen, Germany), anti-doublecortin (1:200, #ab153668, abcam, Boston, MA), anti-amyloid precursor protein (axonal injury marker; 1:200, #ab32136, abcam), anti-synaptophysin (1:200, #ab32127, abcam), and anti-neuroserpin (1:200, #ab33077, abcam); while oligodendroglial markers comprised anti-A2B5 (1:100, #ab53521, abcam), anti-O4 (1:500, #MAB1326, R&D, Minneapolis, MN), and anti-myelin basic protein (MBP; 1:200, #MAB386, Sigma-Aldrich, Temecula, CA). Other markers included anti-ionized calcium binding adaptor molecule 1 (Iba-1; 1: 500, #019-19741, Wako, Osaka, Japan) and anti-TNF-α (1:200, #ab307164, abcam). The sections were then washed and incubated with Alexa Fluor 594 or 488 secondary antibodies (1:200; Invitrogen, Waltham, MA). The slides were photographed for red (Alexa Fluor 594) and green (Alexa Fluor 488) fluorescence with a fluorescent microscope.
The quantification of fluorescence image areas was conducted using an image software (Image-Pro Plus 6.0; Media Cybernetics, Bethesda, MD). Measurements were performed at 100× (0.579 mm2) or 200× (0.145 mm2) magnification per visual field. Three visual fields in the medial, middle and lateral areas of each hemisphere per section and four sections per brain were analyzed.12 The ratios of the antigen-positive area to the cross-sectional area were averaged and compared in the same hemispheres between groups.
Coherency of myelinated axons
Coherency of myelinated axons, as an inverse measure of complexity of white matter microstructure, was assessed using the “OrientationJ measure” function of ImageJ plugin OrientationJ in the selected whole micrograph.34
Western blot analysis
The brain tissue was homogenized in cold lysis buffer and the protein concentrations were determined using a Bio-Rad Protein Assay kit (Bio-Rad Laboratories, Hercules, CA). Samples (50 μg) were separated using 10% SDS-PAGE and blotted onto polyvinylidene fluoride membranes. After blocking, the membranes were incubated with primary antibodies at 4 °C overnight. Immunoreactivity was detected by horseradish-conjugated secondary antibodies and visualized by enhanced chemiluminescence. The primary antibodies used were anti-neuroserpin (1:1000, #ab269277, abcam), anti-N-cadherin (1:1000, #ab18203, abcam), and anti-β-actin (1:5000; Invitrogen). The band signals were quantified using an imaging software (ImagePro Plus 6.0; Media Cybernetics, Bethesda, MD).
Neurobehavioral, neurocognitive, and neuromotor function assessment
Surface righting reflex
P5 pups were placed on their backs, and the time spent for turnover was recorded.35
Negative geotaxis
P9 pups were placed on a 15° inclined surface and facing toward the bottom of a slope. The pups would turn around and start to climb the slope, and the time spent for completing the 180° turnaround was recorded.35
Wire hanging test
P10 pups were placed on an iron wire net. The net was then turned upside down so that the pups hung on the net. The time from suspension to fall was recorded.35
Pouncing and pinning (social play behavior)
P28 rats were habituated to the test chamber for 10 min on 2 consecutive days. On the third day, the rats were secluded for 2.5 h and put into the test chamber together with nonfamiliar, weight- and sex-matched rats of the same experimental group. For 15 min, social play behavior was videotaped for pouncing (trying to rub the nape of other rats with the nose) and pinning (standing on another rat while the back of the other rat is lying on the floor) using a video recording device.14
Repetitive self-grooming (repetitive behavior)
On P35, rats were placed in a transparent cage containing only fresh bedding material for 20 min. The time spent grooming the hair was recorded using a video recording device.14
Rotarod test (neuromotor function)
The rotarod consists of a circular rod turning at a constant or increasing speed. P35 rats were placed on the rotating rod, and the time and the maximal turning speed staying on the rod were recorded.
Open field test (exploratory behavior)
The P49 rats were placed in a circular field with a diameter of 1 m for 5 min, and software was used to track their motor behavior automatically. The circular field was divided into two parts: the outer edge (12.5 cm) and the inner area (75 cm in diameter). The number of times that animals left the outer zone (sheltered by the walls of the arena) and entered the inner zone within 5 min was recorded.
Passive avoidance task (learning and memory)
The latency to refrain from crossing into the punished compartment served as an index of the ability to avoid and allowed memory to be assessed in P56 rats. The apparatus (Shuttlebox-Passive Avoidance, Accuscan Instrument, Inc., Columbus, OH) consisted of a rectangular chamber divided into two compartments by an automatic guillotine door. An overhead stimulus light illuminated one compartment, and the other was black. A rat was placed in the illuminated chamber during the pre-training trial. Once the rat had completely entered the dark compartment, the door was closed and a mild electric shock was applied to the floor grid for 3 s. A retention test was conducted 48 h after the pre-training trial to assess the retention of passive avoidance performance. The rat was placed back in the lighted chamber, and the time it took to enter the dark room with four paws was measured.
Statistical analysis
Statistical analyses were performed using GraphPad Prism 8 software (GraphPad Software Inc., CA). Data are expressed as the mean ± standard deviation (SD). The immunofluorescence staining data with non-normal distribution were analyzed using the Kruskal-Wallis test and Dunn’s post-hoc test. One-way analysis of variance with Tukey’s post hoc test was used to analyze the Western blotting and behavioral performance. P values < 0.05 were considered statistically significant. No outliers were excluded from the statistical analysis. The person charged with outcome measurements of staining, Western blotting, or neurological function was blind to treatments.
Results
MSC therapy attenuated neuroinflammation, neuronal death, axonal damage, and synaptic disruption after LPS-sensitized HI/reperfusion (LHIR) injury
At post-insult 72 h (P5), the LHIR group showed increased activation of microglia with elevated pro-inflammatory cytokine TNF-α expression, whereas MSC treatment significantly attenuated neuroinflammation (Fig. 1a–d, p < 0.01). Compared to the sham group, there was significant decreases in the numbers of complexin-3-positive subplate neurons (Fig. 1e–h) with increased cell death (Fig. 1i–l) in the LHIR group. MSC treatment reduced cell death and preserved most subplate neurons (Fig. 1e–l, all p < 0.01).
In the neonatal rodent brain, the maturation of cortical plate neurons is completed by P7, followed by axonal outgrowth and synaptic formation reaching a mature morphology at the end of the first month.36 On P23, we observed significant decreases in NeuN-positive mature neurons in both the ipsilateral (Fig. 2a–d, p < 0.01) and contralateral (Supplementary Fig. 4a–d, p < 0.01) cortices in the LHIR group. In contrast, the LHIR + MSC and sham groups did not show differences in cell numbers. The LHIR group also displayed axonal injury with significant accumulation of amyloid precursor protein in axons (Fig. 2e–h), and reduced synaptic formation with lower synaptophysin expression (Fig. 2i–l) compared to the sham group (all p < 0.01); whereas MSC treatment ameliorated axonal injury and restored synaptic connections (Fig. 2e–l, p < 0.01).
MSC therapy alleviated the maturation arrest of oligodendrocyte progenitors and maintained the complexity of myelinated axons after LHIR injury
During the lineage progression of rodent oligodendrocyte, A2B5-positive oligodendrocyte precursor cells (OPCs) appear first in the embryonic stage and gradually decline after birth, followed by O4-positive pre-OLs from P2 onward, and finally MBP-positive mature oligodendrocytes predominate from P14.9,37 At post-insult 72 h (P5) and 8 days (P10) in this study, the LHIR group had significantly increased A2B5-positive and decreased O4-positive cells compared to the sham group (p < 0.01), while MSC treatment normalized the developmental sequence of oligodendrocyte lineage (Fig. 3a–h on P5; 3i-p on P10). On P16 and P23, significant hypomyelination with decreased MBP expression occurred in the LHIR group (p < 0.01), whereas MSC treatment preserved MBP expression (Fig. 4a–d on P16, e-h on P23). On P23, the LHIR group also displayed increased coherency of myelinated axons in the ipsilateral (Fig. 4i–l, p < 0.01) and contralateral (Supplementary Fig. 4e–h, p < 0.01) hemispheres, indicating reduced complexity with less axonal outgrowth and arborization. The LHIR + MSC group restored axonal complexity to levels similar to those in the sham group.
MSC therapy improved neurofunctional impairment after LHIR injury
In the early postnatal period (P5-P10), rat pups in the LHIR group spent more time on righting reflex and negative geotaxis, and had less hanging time in wire hanging test, while pups in the LHIR + MSC group had significantly better performance (Fig. 5a–c, all p < 0.05). After the first month of life (P28-P56), rats in the LHIR + MSC group had better neuromotor and cognitive function than those in the LHIR group, with longer time walking on the rolling wheel and faster rotating speeds in the Rotarod test, and lower frequencies of erroneous entry into the dark compartment in the passive avoidance task (Fig. 5d–f, all p < 0.05). Rats in the LHIR group exhibited autistic-like behavior including a longer duration of repetitive self-grooming, reduced pinning/pouncing for social interactions, and a lower frequency of exploratory behavior in the open field test (Fig. 5g–i, all p < 0.01); while rats in the LHIR + MSC and the sham control groups had comparable performance in behavior. There were no sexual differences in the performance of open field test and passive avoidance task, which were performed between P49 and P56 at the ages corresponding to sexual maturity (Supplementary Fig. 5).
MSC treatment restored neuroserpin/N-cadherin expression in neuronal cells after LHIR injury
We further examined the potential roles of neuroserpin in the protective mechanisms of MSC therapy. In P5 sham control rats, both subplate neurons (positive for complexin-3) and neuron progenitor cells (positive for doublecortin) displayed neuroserpin expression (Fig. 6a, e). After LHIR injury, there were significantly decreased numbers of neuronal cells with neuroserpin expression, which were restored after MSC treatment (Fig. 6b–d, f–h, all p < 0.05). There were few O4-positive oligodendrocytes with neuroserpin expression in each group, especially in the LHIR group (p < 0.05); while the LHIR + MSC and the sham groups had similar cell numbers of neuroserpin-expressing oligodendrocytes (Fig. 6i–l). Western blotting further demonstrated that MSC therapy restored not only neuroserpin but also N-cadherin expression in the cortex after LHIR injury (Fig. 6m–p, all p < 0.05), implicating a potential neuroserpin/N-cadherin signaling pathway.
Discussion
Using a neonatal rat model to simulate EoP, this study investigated the therapeutic effects of MSC transplantation on the developmental disturbances of neurons/axons and oligodendrocyte in relation to neuroserpin expression. Following an injury induced by LPS-sensitized HI/reperfusion, we found microglial activation followed by increased cell death in subplate neurons, reduced mature neurons, axonal destruction, and decreased synaptic formation. The damage to neurons/axons might lower neuroserpin levels in the cortex, along with arrested maturation of oligodendrocyte progenitors leading to hypomyelination in the white matter. The maturational disturbances during early brain development resulted in not only cognitive and neuromotor functional impairments but also autistic-like behaviors, reflecting neurodevelopmental disorders seen in children born extremely preterm. MSC treatment mitigated neuronal death, protected against injury in both the cortex and white matter, improved neurological and behavioral outcomes, and elevated neuroserpin and N-cadherin levels in neurons. These findings suggest that neuroserpin/N-cadherin signaling might play critical roles in the reparative mechanisms of MSC therapy (Supplementary Fig. 6).
The pathogenic mechanism of EoP is a combination of a destructive injury and the subsequent trophic/maturational (i.e., developmental) disturbances.7,8 Most experimental studies of preterm brain injury focused on the destructive component, which was induced by exposure to HI, or to neurotoxin such as LPS or pro-inflammatory cytokines in neonatal animals.38,39,40,41 Our previous studies have found that subthreshold HI was sensitized by low-dose LPS to trigger significant injuries in the neonatal rat brain, characterized by microglia-mediated neuroinflammation and neurovascular damage.12,13 This study further explored the developmental disturbances and neurobehavioral outcomes using the modified model of LPS-sensitized HI brain injury followed by post-HI reperfusion, which occurs frequently in preterm neonates but is rarely considered in previous studies.
Subplate neurons reach their culmination size during the peak period for occurrence of EoP (23–32 weeks of gestation), and serve as a platform for axonal guidance and synaptic contacts linking the development of cerebral cortex, subcortical white matter and deep nuclei.10,11,42 An animal study has shown that circuit changes in the subplate neurons after HI in P1 rat pups led to altered brain development.43 Our study found increased cell death with decreased numbers of subplate neurons at 72 h (P5) after LHIR injury. Damage to the subplate neurons potentially induces degeneration of axons and their originating neurons through retrograde or anterograde effects, and axonal degeneration further results in hypomyelination.7,44,45 Our findings provided evidence of neuronal degeneration, axonal injury, and impaired synaptic formation at post-insult 21 days (P23).
During the lineage progression of oligodendrocytes, O4-positive pre-OLs predominate in the P2 rodent brain, which coincides with the developmental window of vulnerability for EoP in the human brain.37,46 Pre-OLs contact axons extending to and from the hub of subplate neurons for myelination, and are selectively vulnerable to HI and reperfusion-induced oxidative stress.47,48 The damaged pre-OLs are shown to undergo cell death or maturation arrest leading to hypomyelination,49,50,51 or trigger axonal degeneration leading to impaired neuronal development.7,45,52 After LHIR on P2, we found increased A2B5-positive OPCs and decreased O4-positive pre-OLs on P5, followed by diminished MBP expression and reduced complexity of myelinated axons on P23. Our findings implicated that hypomyelination was attributed to axonal injury and early maturation arrest with failure of OPC differentiation into pre-OLs.
This study verified the hypothesis that MSC therapy ameliorated the developmental disturbances and improved neurobehavioral outcomes after LHIR injury through protection of neuroserpin/N-cadherin signaling in subplate neurons and neuronal progenitor cells. Neuroserpin is expressed in the neocortex, hippocampus and limbic system that are responsible for learning, memory and social behavior, and reaches a peak level in the rodent brain during the first week of life.53,54 Although neuroserpin is an inhibitor of tissue plasminogen activators, it is shown in animal studies to attenuate destructive brain injury such as ischemic stroke via non-enzymatic effects.18,25 Experimental studies also implicate neuroserpin is involved in regulating critical cellular functions through crosstalk with downstream signaling networks.18,22 During the process of neural circuit formation that occurred in the platform of subplate neurons, neuroserpin might not only exert anti-apoptotic effects on neural progenitor cells, but also promote axonal outgrowth, synaptic connections, and oligodendrocyte-axon contact for myelination through regulation of cell adhesion molecules such as N-cadherin.18,19,20,21,22 Lack or dysregulation of neuroserpin leads to neuropsychiatric disorders such as cognitive impairment, social ability deficits, and defective exploratory behavior.18,53,55 Despite restoring neuroserpin expression correlates with improved brain histology and neurobehavioral outcomes in this study, it does not establish a direct causal relationship, and how MSC therapy normalized neuroserpin expression after LHIR injury remains unclear. Further studies are needed to elucidate the underlying mechanisms and the causative links between neuroserpin expression and brain repair.
There are limitations in this study. First, this study used an intracerebroventricular route for MSC transplantation, which is not feasible in clinical practice for infants born very preterm, despite the superior therapeutic efficacy provided by targeted delivery and better paracrine potency of MSCs compared to other routes.56,57 Our findings of intracerebral MSC effects might also be observed with systemic intravenous administration, which has shown therapeutic effects comparable to those via the intracerebroventricular route in animal studies of preterm brain injury.58,59 Since the paracrine action, rather than direct regeneration, is regarded as the primary protective mechanism of MSC transplantation, strategies to enhance the paracrine potency, such as preconditioning or genetic engineering of MSCs, may improve the therapeutic efficacy.59 Second, the MSC dose used in this study has limited clinical translatability. The determination of the MSC dose in this study was modified from a previous study of intraventricular hemorrhage (IVH) in P5 Sprague–Dawley rat, which showed that injecting 1 × 105 MSCs into the lateral ventricles on P6 improved severe IVH.60 Considering the treatment time point in our study is on P3 when the ventricular size is smaller than that on P6, we administered half the dose used in that study (5 × 104 MSCs) for treatment. The optimal dose of MSC transplantation needs to be determined according to the specific brain injury site and brain disorders with the specific source, timing and route of administration in different species,59 especially safety in the clinical context. Third, pharmacological approaches or genetically modified mice might be used to verify the potential mechanisms of MSC effects through neuroserpin/N-cadherin signaling. Fourth, although sex-related differences are largely attributed to hormonal variations after sexual maturity (~40–60 days of age in rats),61 there is growing evidence suggesting that sexual differences should be considered in neonatal rodent models during the first month of life.62 Despite no observed differences in brain damage and outcome performance between male and female rats in this study, we cannot completely exclude the possibility of sex-related differences. Future studies may investigate the potential effects of sex on the underlying mechanisms of EoP.
In conclusion, using a novel model of LPS-sensitized HI/reperfusion injury based on the pathogenesis of EoP, this animal study demonstrated the developmental disturbances in neurons and oligodendrocytes and the subsequent impairment of neurobehavioral functions, and uncovered the roles of neuroserpin in brain repair mechanisms of MSC therapy. Future research may investigate the therapeutic potential of MSC-conditioned medium or MSC-derived extracellular vesicles/exosomes for better clinical application, and explore other reparative mechanisms of MSC secretome for developmental disturbances in the premature brain.
Data availability
The data that support the findings of this study are available from the corresponding author upon reasonable request.
References
Volpe, J. J., Kinney, H. C., Jensen, F. E. & Rosenberg, P. A. The developing oligodendrocyte: key cellular target in brain injury in the premature infant. Int. J. Devl. Neurosci. 29, 423–440 (2011).
Johnson, S. & Marlow, N. Early and long-term outcome of infants born extremely preterm. Arch. Dis. Child. 102, 97–102 (2017).
Cainelli, E., Arrigoni, F. & Vedovelli, L. White matter injury and neurodevelopmental disabilities: a cross-disease (dis)connection. Prog. Neurobiol. 193, 101845 (2020).
Wang, L. W., Chang, Y. C., Lin, C. Y., Hong, J. S. & Huang, C. C. Low-dose LPS selectively sensitizes hypoxic ischemia-induced white matter injury in the immature brain. Pediatr. Res. 68, 41–47 (2010).
Wang, L. W., Lin, Y. C., Wang, S. T., Yeh, T. F. & Huang, C. C. Hypoxic/ischemic and infectious events have cumulative effects on the risk of cerebral palsy in very-low-birth-weight preterm infants. Neonatology 106, 209–215 (2014).
Martini, S., Austin, T., Aceti, A., Faldella, G. & Corvaglia, L. Free radicals and neonatal encephalopathy: mechanisms of injury, biomarkers, and antioxidant treatment perspectives. Pediatr. Res. 87, 823–833 (2020).
Volpe, J. J. Brain injury in premature infants: a complex amalgam of destructive and developmental disturbances. Lancet Neurol. 8, 110–124 (2009).
Inder, T. E., Volpe, J. J. & Anderson, P. J. Defining the neurologic consequences of preterm birth. N. Engl. J. Med. 389, 441–453 (2023).
Back, S. A. White matter injury in the preterm infant: pathology and mechanisms. Acta Neuropathol. 134, 331–349 (2017).
Hoerder-Suabedissen, A. & Molnar, Z. Development, evolution and pathology of neocortical subplate neurons. Nat. Rev. Neurosci. 16, 133–146 (2015).
Kostovic, I. & Judas, M. The development of the subplate and thalamocortical connections in the human foetal brain. Acta Paediatr. 99, 1119–1127 (2010).
Wang, L. W., Tu, Y. F., Huang, C. C. & Ho, C. J. JNK signaling is the shared pathway linking neuroinflammation, blood–brain barrier disruption, and oligodendroglial apoptosis in the white matter injury of the immature brain. J. Neuroinflamm. 9, 175 (2012).
Wang, L. W. et al. TNFR1-JNK signaling is the shared pathway of neuroinflammation and neurovascular damage after LPS-sensitized hypoxic-ischemic injury in the immature brain. J. Neuroinflamm. 11, 215 (2014).
van Tilborg, E. et al. Combined fetal inflammation and postnatal hypoxia causes myelin deficits and autism-like behavior in a rat model of diffuse white matter injury. Glia 66, 78–93 (2018).
Vaes, J. E. G. et al. Intranasal mesenchymal stem cell therapy to boost myelination after encephalopathy of prematurity. Glia 69, 655–680 (2021).
Vaes, J. E. G. et al. Regenerative therapies to restore interneuron disturbances in experimental models of encephalopathy of prematurity. Int. J. Mol. Sci. 22, 211 (2021).
Adorjan, I. et al. Neuroserpin expression during human brain development and in adult brain revealed by immunohistochemistry and single cell RNA sequencing. J. Anat. 235, 543–554 (2019).
Godinez, A. et al. Neuroserpin, a crucial regulator for axogenesis, synaptic modelling and cell-cell interactions in the pathophysiology of neurological disease. Cell. Mol. Life Sci. 79, 172 (2022).
D’Acunto, E. et al. Neuroserpin: structure, function, physiology and pathology. Cell. Mol. Life Sci. 78, 6409–6430 (2021).
Chooi, W. H. & Chew, S. Y. Modulation of cell-cell interactions for neural tissue engineering: Potential therapeutic applications of cell adhesion molecules in nerve regeneration. Biomaterials 197, 327–344 (2019).
Chen, M. et al. N-cadherin is involved in neuronal activity-dependent regulation of myelinating capacity of zebrafish individual oligodendrocytes in vivo. Mol. Neurobiol. 54, 6917–6930 (2017).
Lee, T. W., Coates, L. C. & Birch, N. P. Neuroserpin regulates N-cadherin-mediated cell adhesion independently of its activity as an inhibitor of tissue plasminogen activator. J. Neurosci. Res. 86, 1243–1253 (2008).
Yang, X. et al. Neuroserpin protects rat neurons and microglia-mediated inflammatory response against oxygen-glucose deprivation and reoxygenation treatments in an in vitro study. Cell. Physiol. Biochem. 38, 1472–1482 (2016).
Gu, R. P. et al. Retina is protected by neuroserpin from ischemic/reperfusion-induced injury independent of tissue-type plasminogen activator. PLoS One 10, e0130440 (2015).
Ding, S. et al. The neuroprotective role of neuroserpin in ischemic and hemorrhagic stroke. Curr. Neuropharmacol. 19, 1367–1378 (2021).
Ma, J. et al. Effect of neuroserpin in a neonatal hypoxic-ischemic injury model ex vivo. Biol. Res. 45, 357–362 (2012).
Vaes, J. E. G. et al. The potential of stem cell therapy to repair white matter injury in preterm infants: lessons learned from experimental models. Front. Physiol. 10, 540 (2019).
Morioka, C. et al. Neuroprotective effects of human umbilical cord-derived mesenchymal stem cells on periventricular leukomalacia-like brain injury in neonatal rats. Inflamm. Regen. 37, 1 (2017).
Zhu, L. H. et al. Improvement of human umbilical cord mesenchymal stem cell transplantation on glial cell and behavioral function in a neonatal model of periventricular white matter damage. Brain Res. 1563, 13–21 (2014).
Keshtkar, S., Azarpira, N. & Ghahremani, M. H. Mesenchymal stem cell-derived extracellular vesicles: novel frontiers in regenerative medicine. Stem Cell Res. Ther. 9, 63 (2018).
Chang, C. et al. Hypoxic preconditioning enhances the therapeutic potential of the secretome from cultured human mesenchymal stem cells in experimental traumatic brain injury. Clin. Sci. 124, 165–176 (2013).
Kawasaki H., et al. Intracerebroventricular and intravascular injection of viral particles and fluorescent microbeads into the neonatal brain. J. Vis. Exp. 113, https://doi.org/10.3791/54164-v (2016).
Paxinos, G. & Watson, C. The Mouse Brain in Stereotaxic Coordinates (Academic, New York, 2001).
van Tilborg, E. et al. A quantitative method for microstructural analysis of myelinated axons in the injured rodent brain. Sci. Rep. 7, 16492 (2017).
Wu, Z., Xue, H., Zhang, Y. & Zhao, P. Dexmedetomidine alleviates neurobehavioral impairments and myelination deficits following lipopolysaccharide exposure in early postnatal rats. Life Sci. 263, 118556 (2020).
Farhy-Tselnicker, I. & Allen, N. J. Astrocytes, neurons, synapses: a tripartite view on cortical circuit development. Neural Dev. 13, 7 (2018).
Craig, A. et al. Quantitative analysis of perinatal rodent oligodendrocyte lineage progression and its correlation with human. Exp. Neurol. 181, 231–240 (2003).
Albertsson, A. M. et al. The effect of osteopontin and osteopontin-derived peptides on preterm brain injury. J. Neuroinflamm. 11, 197 (2014).
Jantzie, L. L. et al. Chloride cotransporter NKCC1 inhibitor bumetanide protects against white matter injury in a rodent model of periventricular leukomalacia. Pediatr. Res. 77, 554–562 (2015).
Pang, Y. et al. IGF-1 can either protect against or increase LPS-induced damage in the developing rat brain. Pediatr. Res. 67, 579–584 (2010).
Aden, U. et al. Systemic inflammation sensitizes the neonatal brain to excitotoxicity through a pro-/anti-inflammatory imbalance: key role of TNF-alpha pathway and protection by etanercept. Brain Behav. Immun. 24, 747–758 (2010).
Ohtaka-Maruyama, C. et al. Synaptic transmission from subplate neurons controls radial migration of neocortical neurons. Science 360, 313–317 (2018).
Sheikh, A. et al. Neonatal hypoxia-ischemia causes functional circuit changes in subplate neurons. Cereb. Cortex 29, 765–776 (2019).
Haynes, R. L. et al. Diffuse axonal injury in periventricular leukomalacia as determined by apoptotic marker fractin. Pediatr. Res. 63, 656–661 (2008).
Piaton, G., Gould, R. M. & Lubetzki, C. Axon-oligodendrocyte interactions during developmental myelination, demyelination and repair. J. Neurochem. 114, 1243–1260 (2010).
van Tilborg, E. Origin and dynamics of oligodendrocytes in the developing brain: Implications for perinatal white matter injury. Glia 66, 221–238 (2018).
Back, S. A. et al. Selective vulnerability of late oligodendrocyte progenitors to hypoxia–Ischemia. J. Neurosci. 22, 455–463 (2002).
Back, S. A. et al. Selective vulnerability of preterm white matter to oxidative damage defined by F2-isoprostanes. Ann. Neurol. 58, 108–120 (2005).
van Tilborg, E. Impaired oligodendrocyte maturation in preterm infants: Potential therapeutic targets. Prog. Neurobiol. 136, 28–49 (2016).
Segovia, K. N. et al. Arrested oligodendrocyte lineage maturation in chronic perinatal white matter injury. Ann. Neurol. 63, 520–530 (2008).
Buser, J. R. et al. Arrested preoligodendrocyte maturation contributes to myelination failure in premature infants. Ann. Neurol. 71, 93–109 (2012).
Huang, S. et al. New insights into the roles of oligodendrocytes regulation in ischemic stroke recovery. Neurobiol. Dis. 184, 106200 (2023).
Reumann, R. et al. The serine protease inhibitor neuroserpin is required for normal synaptic plasticity and regulates learning and social behavior. Learn. Mem. 24, 650–659 (2017).
Krueger, S. R. et al. Expression of neuroserpin, an inhibitor of tissue plasminogen activator, in the developing and adult nervous system of the mouse. J. Neurosci. 17, 8984–8996 (1997).
Madani, R. et al. Impaired explorative behavior and neophobia in genetically modified mice lacking or overexpressing the extracellular serine protease inhibitor neuroserpin. Mol. Cell. Neurosci. 23, 473–494 (2003).
Park, W. S., Ahn, S. Y., Sung, S. I., Ahn, J. Y. & Chang, Y. S. Strategies to enhance paracrine potency of transplanted mesenchymal stem cells in intractable neonatal disorders. Pediatr. Res. 83, 214–222 (2018).
Sarmah, D. et al. Mesenchymal stem cell therapy in ischemic stroke: a meta-analysis of preclinical studies. Clin. Pharmacol. Ther. 103, 990–998 (2018).
Ahn, S. Y. et al. Optimal route for mesenchymal stem cells transplantation after severe intraventricular hemorrhage in newborn rats. PLoS One 10, e0132919 (2015).
Ahn, S. Y., Chang, Y. S. & Park, W. S. Stem cells for neonatal brain injury–lessons from the bench. Semin. Perinatol. 47, 151726 (2023).
Ahn, S. Y. et al. Pivotal role of brain-derived neurotrophic factor secreted by mesenchymal stem cells in severe intraventricular hemorrhage in newborn rats. Cell Transpl. 26, 145–156 (2017).
Kohn, D. F., Clifford, C. B. Biology and diseases of rats. In: Fox, J. G., Anderson, L. C., Loew, F. M., Quimby, F. W., editors. Laboratory Animal Medicine. 2nd ed. (Academic Press, 2002).
Dieu-Lugon, B. L. et al. Why considering sexual differences is necessary when studying encephalopathy of prematurity through rodent models. Eur. J. Neurosci. 52, 2560–2574 (2020).
Acknowledgements
We are thankful to Mr. Chung-Han Wang for his experimental and technical assistance. This work was supported by grants from the Ministry of Science and Technology (MOST 108-2314-B384-005-MY3), and Chi Mei Medical Center (CMNDMC11102) in Taiwan.
Author information
Authors and Affiliations
Contributions
Each author has met the Pediatric Research authorship requirements and completed the following criteria: (1) substantial contributions to conception and design, acquisition of data, or analysis and interpretation of data; (2) drafting the article or revising it critically for important intellectual content; and (3) final approval of the version to be published.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
Ethics approval
All animal experiments were approved and carried out in accordance with the Institutional Animal Care and Use Committee at the Chi Mei Medical Center, Tainan, Taiwan (Approval No. 108101805).
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary information
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Wang, LW., Hsiung, CW., Chang, CP. et al. Neuroserpin normalization by mesenchymal stem cell therapy after encephalopathy of prematurity in neonatal rats. Pediatr Res (2024). https://doi.org/10.1038/s41390-024-03412-z
Received:
Revised:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41390-024-03412-z