Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.888
Filter
1.
Front Biosci (Landmark Ed) ; 29(9): 329, 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39344311

ABSTRACT

BACKGROUND: Hypoxic-ischemic brain damage (HIBD) is a prevalent brain injury with high mortality and morbidity. It results from hypoxia and ischemia of the brain due to various perinatal factors. A previous study showed that knockdown of programmed cell death factor 4 (PDCD4) could reduce infarction injury resulting from ischemia/reperfusion injury. However, exact mechanism by which PDCD4 acts in HIBD is not yet understood. Our aim in present investigation was to investigate the function and mechanism of PDCD4 in alleviating HIBD. METHODS: An HIBD model was developed using neonatal rats. After 48 h of modeling, short-term neurological function was evaluated and the brain tissue removed for assessment of cerebral infarct volume and brain water content (BWC). A cell model of oxygen glucose deprivation/reoxygenation (OGD/R) was also constructed. Overexpression or knockdown of insulin-like growth factor 2 mRNA binding protein 3 (IGF2BP3) or PDCD4 was performed in pretreated cells. RESULTS: The geotaxis reflex time, cerebral infarct volume, and BWC all increased after HIBD in this neonatal rat model. Additionally, the levels of PDCD4 and of the N6-Methyladenosine (m6A) reader protein IGF2BP3 were increased in HIBD rats and OGD/R-stimulated pheochromocytoma (PC12) cells relative to controls. Moreover, OGD/R-stimulated pheochromocytoma PC12 cells showed decreased cell viability, increased apoptosis, and elevated Interleukin 6 (IL-6), Interleukin 1 ß (IL-1ß), and tumor necrosis factor-α (TNF-α) contents. These features were reversed after knocking down IGF2BP3. The interaction between IGF2BP3 protein and PDCD4 mRNA was confirmed by RNA immunoprecipitation and RNA pull-down assays. Furthermore, knockdown of IGF2BP3 in OGD/R-stimulated PC12 cells reduced cell damage via down-regulation of PDCD4. Finally, the IGF2BP3/PDCD4 axis alleviated OGD/R-induced cell injury in primary cortical neurons (PCNs). CONCLUSIONS: PDCD4 and m6A reader protein IGF2BP3 were up-regulated in an HIBD neonatal rat model. Knockdown of IGF2BP3 in OGD/R-stimulated PC12 cells or PCNs alleviated cell damage through reducing PDCD4.


Subject(s)
Apoptosis Regulatory Proteins , Down-Regulation , Gene Knockdown Techniques , Hypoxia-Ischemia, Brain , RNA-Binding Proteins , Rats, Sprague-Dawley , Animals , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/genetics , Hypoxia-Ischemia, Brain/pathology , Rats , PC12 Cells , Animals, Newborn , Disease Models, Animal , Male
2.
Int J Mol Sci ; 25(17)2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39273353

ABSTRACT

Cerebral palsy (CP) is a common neurodevelopmental disorder characterized by pronounced motor dysfunction and resulting in physical disability. Neural precursor cells (NPCs) have shown therapeutic promise in mouse models of hypoxic-ischemic (HI) perinatal brain injury, which mirror hemiplegic CP. Constraint-induced movement therapy (CIMT) enhances the functional use of the impaired limb and has emerged as a beneficial intervention for hemiplegic CP. However, the precise mechanisms and optimal application of CIMT remain poorly understood. The potential synergy between a regenerative approach using NPCs and a rehabilitation strategy using CIMT has not been explored. We employed the Rice-Vannucci HI model on C57Bl/6 mice at postnatal day (PND) 7, effectively replicating the clinical and neuroanatomical characteristics of hemiplegic CP. NPCs were transplanted in the corpus callosum (CC) at PND21, which is the age corresponding to a 2-year-old child from a developmental perspective and until which CP is often not formally diagnosed, followed or not by Botulinum toxin injections in the unaffected forelimb muscles at PND23, 26, 29 and 32 to apply CIMT. Both interventions led to enhanced CC myelination and significant functional recovery (as shown by rearing and gait analysis testing), through the recruitment of endogenous oligodendrocytes. The combinatorial treatment indicated a synergistic effect, as shown by newly recruited oligodendrocytes and functional recovery. This work demonstrates the mechanistic effects of CIMT and NPC transplantation and advocates for their combined therapeutic potential in addressing hemiplegic CP.


Subject(s)
Disease Models, Animal , Hypoxia-Ischemia, Brain , Mice, Inbred C57BL , Neural Stem Cells , Recovery of Function , Animals , Neural Stem Cells/transplantation , Mice , Hypoxia-Ischemia, Brain/therapy , Hypoxia-Ischemia, Brain/pathology , Cerebral Palsy/therapy , Corpus Callosum , Exercise Therapy/methods , Male , Female
3.
J Am Heart Assoc ; 13(19): e036054, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39319465

ABSTRACT

BACKGROUND: Melatonin has been shown to be neuroprotective in different animal models of neonatal hypoxic-ischemic brain injury. However, its exact molecular mechanism of action remains unknown. Our aim was to prove melatonin's short- and long-term neuroprotection and investigate its role on the AMPK (AMP-activated protein kinase)/mTOR (mammalian target of rapamycin) pathway following neonatal hypoxic-ischemic brain injury. METHODS AND RESULTS: Seven-day-old Wistar rat pups were exposed to hypoxia-ischemia, followed by melatonin or vehicle treatment. Detailed analysis of the AMPK/mTOR/autophagy pathway, short- and long-term neuroprotection, myelination, and oligodendrogenesis was performed at different time points. At 7 days after hypoxia-ischemia, melatonin-treated animals showed a significant decrease in tissue loss, increased oligodendrogenesis, and myelination. Long-term neurobehavioral results showed significant motor improvement following melatonin treatment. Molecular pathway analysis showed a decrease in the AMPK expression, with a significant increase at mTOR's downstream substrates, and a significant decrease at the autophagy marker levels in the melatonin group compared with the vehicle group. CONCLUSIONS: Melatonin treatment reduced brain area loss and promoted oligodendrogenesis with a clear improvement of motor function. We found that melatonin associated neuroprotection is regulated via the AMPK/mTOR/autophagy pathway. Considering the beneficial effects of melatonin and the results of our study, melatonin seems to be an optimal candidate for the treatment of newborns with hypoxic-ischemic brain injury in high- as well as in low- and middle-income countries.


Subject(s)
AMP-Activated Protein Kinases , Animals, Newborn , Autophagy , Disease Models, Animal , Hypoxia-Ischemia, Brain , Melatonin , Neuroprotective Agents , Rats, Wistar , Signal Transduction , TOR Serine-Threonine Kinases , Animals , Melatonin/pharmacology , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/drug therapy , Hypoxia-Ischemia, Brain/pathology , TOR Serine-Threonine Kinases/metabolism , Neuroprotective Agents/pharmacology , Signal Transduction/drug effects , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/drug effects , Autophagy/drug effects , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Oligodendroglia/pathology , Brain/drug effects , Brain/metabolism , Brain/pathology , Rats , Behavior, Animal/drug effects
4.
Biomed Pharmacother ; 179: 117338, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39278187

ABSTRACT

A recent study has introduced a recombinant fusion protein, consisting of the extracellular domain (ECD) of p75 and the Fc fragment of human immunoglobulin IgG1 (p75ECD-Fc), as a multifaceted agent within the nervous system. This research aimed to assess the effects of p75ECD-Fc on neuronal growth and the restoration of neurological functions in rats afflicted with neonatal hypoxic-ischemic encephalopathy (NHIE). In vitro analyses revealed that 1 µM p75ECD-Fc treatment markedly increased cell viability and facilitated neurite outgrowth in neurons exposed to oxygen-glucose deprivation (OGD). Subsequent in vivo studies determined that a dose of 78.6 µg/3 µl of p75ECD-Fc significantly mitigated brain damage and both acute and long-term neurological impairments, outperforming the therapeutic efficacy of hypothermia, as evidenced through behavioral assessments. Additionally, in vivo immunostaining showed that p75ECD-Fc administration enhanced neuronal survival and regeneration, and reduced astrocytosis and microglia activation in the cortex and hippocampus of NHIE rats. A noteworthy shift from A1 to A2 astrocyte phenotypes and from M1 to M2 microglia phenotypes was observed after p75ECD-Fc treatment. Furthermore, a co-expression of the p75 neurotrophin receptor (p75NTR) and Nestin was identified, with an overexpression of Nestin alleviating the neurological dysfunction induced by NHIE. Mechanistically, the neuroprotective effects of p75ECD-Fc, particularly its inhibition of neuronal apoptosis post-OGD, may be attributed to Nestin. Taken together, these results highlight the neuroprotective and anti-inflammatory effects of p75ECD-Fc treatment through the modulation of glial cell phenotypes and the Nestin-mediated inhibition of neuronal apoptosis, positioning it as a viable therapeutic approach for NHIE.


Subject(s)
Animals, Newborn , Apoptosis , Hypoxia-Ischemia, Brain , Immunoglobulin Fc Fragments , Nestin , Rats, Sprague-Dawley , Animals , Hypoxia-Ischemia, Brain/drug therapy , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/metabolism , Apoptosis/drug effects , Nestin/metabolism , Immunoglobulin Fc Fragments/pharmacology , Rats , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/pharmacology , Recombinant Fusion Proteins/pharmacology , Male , Cell Survival/drug effects , Microglia/drug effects , Microglia/pathology , Microglia/metabolism , Humans , Receptors, Nerve Growth Factor/metabolism , Disease Models, Animal
5.
Cells ; 13(18)2024 Sep 14.
Article in English | MEDLINE | ID: mdl-39329733

ABSTRACT

Neonatal hypoxic-ischemic encephalopathy (HIE) occurs in 1.5 per 1000 live births, leaving affected children with long-term motor and cognitive deficits. Few animal models of HIE incorporate maternal immune activation (MIA) despite the significant risk MIA poses to HIE incidence and diagnosis. Our non-invasive model of HIE pairs late gestation MIA with postnatal hypoxia. HIE pups exhibited a trend toward smaller overall brain size and delays in the ontogeny of several developmental milestones. In adulthood, HIE animals had reduced strength and gait deficits, but no difference in speed. Surprisingly, HIE animals performed better on the rotarod, an assessment of motor coordination. There was significant upregulation of inflammatory genes in microglia 24 h after hypoxia. Single-cell RNA sequencing (scRNAseq) revealed two microglia subclusters of interest following HIE. Pseudobulk analysis revealed increased microglia motility gene expression and upregulation of epigenetic machinery and neurodevelopmental genes in macrophages following HIE. No sex differences were found in any measures. These results support a two-hit noninvasive model pairing MIA and hypoxia as a model for HIE in humans. This model results in a milder phenotype compared to established HIE models; however, HIE is a clinically heterogeneous injury resulting in a variety of outcomes in humans. The pathways identified in our model of HIE may reveal novel targets for therapy for neonates with HIE.


Subject(s)
Animals, Newborn , Disease Models, Animal , Hypoxia-Ischemia, Brain , Inflammation , Microglia , Monocytes , Animals , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/genetics , Hypoxia-Ischemia, Brain/metabolism , Mice , Inflammation/pathology , Inflammation/genetics , Monocytes/metabolism , Female , Microglia/metabolism , Microglia/pathology , Male , Developmental Disabilities/genetics , Developmental Disabilities/pathology , Mice, Inbred C57BL , Motor Disorders/genetics , Motor Disorders/pathology
6.
Niger J Clin Pract ; 27(8): 1027-1032, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39212441

ABSTRACT

BACKGROUND: Prolonged perinatal asphyxia (PA) may cause hypoxic-ischemic damage to the brain. The aim of this study was to investigate the brain diffusion changes of patients with PA and examine the relationship with brain damage. METHODS: This retrospective study included 55 patients diagnosed with PA, separated into mild and severe PA groups. For the evaluation of brain damage in all the study neonates, brain and diffusion MRI scans were performed using a 3T device. The scans were taken between 5 and 10 days postnatal, after completion of hypothermia treatment, in accordance with the standard clinical protocol of our institution. Apparent diffusion coefficient (ADC) values of the lentiform nucleus, thalamus, frontal white matter, and posterior limbs of the internal capsule were measured. Minitab package programs and SPSS version 20.0 software were used for statistical analysis and graphic drawing. Spearman's rank correlation analysis was used. RESULTS: The bilateral lentiform nucleus, thalamus, frontal white matter, and posterior limbs of the internal capsule ADC values were significantly higher in the severe PA group than in the mild PA group. CONCLUSIONS: In neonates with severe perinatal asphyxia, brain damage can be evaluated on diffusion-weighted imaging (DWI) of the cerebral deep white matter and basal ganglia. DWI, imaging with conventional brain MRI comes to the fore in clinical importance in PA patients.


Subject(s)
Asphyxia Neonatorum , Diffusion Magnetic Resonance Imaging , Humans , Retrospective Studies , Diffusion Magnetic Resonance Imaging/methods , Infant, Newborn , Female , Male , Brain/diagnostic imaging , Brain/pathology , Hypoxia-Ischemia, Brain/diagnostic imaging , Hypoxia-Ischemia, Brain/pathology , Severity of Illness Index , Hypothermia, Induced/methods
7.
Int J Mol Sci ; 25(15)2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39125778

ABSTRACT

Neonatal hypoxic-ischemic encephalopathy is the most common cause of long-term disability in term neonates, and white matter injury is the primary cause of cerebral palsy. Therapies that focus on the neuroprotection of myelination and oligodendrocyte proliferation could potentially ameliorate long-lasting neurological impairments after hypoxic-ischemic encephalopathy. Clemastine, a histamine H1 antagonist, has been shown to exert neuroprotective effects in multiple sclerosis and spinal cord injury by promoting oligodendrogenesis and re-myelination. In this study, we demonstrated the neuroprotective effects of clemastine in our rat model of neonatal hypoxic-ischemic brain injury. Animals received a single intraperitoneal injection of either vehicle or clemastine (10 mg/kg) for 6 consecutive days. Our results showed a significant reduction in white matter loss after treatment, with a clear effect of clemastine on oligodendrocytes, showing a significant increase in the number of Olig2+ cells. We characterized the MAPK/ERK pathway as a potential mechanistic pathway underlying the neuroprotective effects of clemastine. Altogether, our results demonstrate that clemastine is a potential compound for the treatment of hypoxic-ischemic encephalopathy, with a clear neuroprotective effect on white matter injury by promoting oligodendrogenesis.


Subject(s)
Animals, Newborn , Cell Proliferation , Clemastine , Disease Models, Animal , Hypoxia-Ischemia, Brain , MAP Kinase Signaling System , Neuroprotective Agents , Oligodendroglia , Animals , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Hypoxia-Ischemia, Brain/drug therapy , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Rats , Clemastine/pharmacology , MAP Kinase Signaling System/drug effects , Cell Proliferation/drug effects , Rats, Sprague-Dawley
8.
Neurobiol Dis ; 200: 106629, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39111704

ABSTRACT

Hypoxic-ischemic encephalopathy (HIE) in neonates causes mortality and neurologic morbidity, including poor cognition with a complex neuropathology. Injury to the cholinergic basal forebrain and its rich innervation of cerebral cortex may also drive cognitive pathology. It is uncertain whether genes associated with adult cognition-related neurodegeneration worsen outcomes after neonatal HIE. We hypothesized that neocortical damage caused by neonatal HI in mice is ushered by persistent cholinergic innervation and interneuron (IN) pathology that correlates with cognitive outcome and is exacerbated by genes linked to Alzheimer's disease. We subjected non-transgenic (nTg) C57Bl6 mice and mice transgenically (Tg) expressing human mutant amyloid precursor protein (APP-Swedish variant) and mutant presenilin (PS1-ΔE9) to the Rice-Vannucci HI model on postnatal day 10 (P10). nTg and Tg mice with sham procedure were controls. Visual discrimination (VD) was tested for cognition. Cortical and hippocampal cholinergic axonal and IN pathology and Aß plaques, identified by immunohistochemistry for choline acetyltransferase (ChAT) and 6E10 antibody respectively, were counted at P210. Simple ChAT+ axonal swellings were present in all sham and HI groups; Tg mice had more than their nTg counterparts, but HI did not affect the number of axonal swellings in APP/PS1 Tg mice. In contrast, complex ChAT+ neuritic clusters (NC) occurred only in Tg mice; HI increased that burden. The abundance of ChAT+ clusters in specific regions correlated with decreased VD. The frequency of attritional ChAT+ INs in the entorhinal cortex (EC) was increased in Tg shams relative to their nTg counterparts, but HI obviated this difference. Cholinergic IN pathology in EC correlated with NC number. The Aß deposition in APP/PS1 Tg mice was not exacerbated by HI, nor did it correlate with other metrics. Adult APP/PS1 Tg mice have significant cortical cholinergic axon and EC ChAT+ IN pathologies; some pathology was exacerbated by neonatal HI and correlated with VD. Mechanisms of neonatal HI induced cognitive deficits and cortical neuropathology may be modulated by genetic risk, perhaps accounting for some of the variability in outcomes.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor , Animals, Newborn , Cholinergic Neurons , Mice, Inbred C57BL , Mice, Transgenic , Neocortex , Animals , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Mice , Neocortex/metabolism , Neocortex/pathology , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Cholinergic Neurons/pathology , Cholinergic Neurons/metabolism , Presenilin-1/genetics , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/genetics , Brain Injuries/pathology , Brain Injuries/metabolism , Brain Injuries/genetics , Choline O-Acetyltransferase/metabolism , Choline O-Acetyltransferase/genetics , Humans , Male , Disease Models, Animal
9.
Crit Rev Eukaryot Gene Expr ; 34(8): 47-57, 2024.
Article in English | MEDLINE | ID: mdl-39180207

ABSTRACT

FTO alpha-ketoglutarate dependent dioxygenase (FTO) is aberrantly expressed in brain disorders. However, the roles of FTO in neonatal hypoxic-ischemic brain injury (HIE) are still unclear. This study aims to investigate the potential of FTO in neonatal HIE. Oxygen-glucose deprivation (OGD) was used to establish HIE in vitro. mRNA levels were detected by real-time reverse transcriptase-quantitative polymerase chain reaction (RT-qPCR). Protein expression was detected by Western blot. The levels of malondialdehyde (MDA), superoxide dismutase (SOD), ferrous iron (Fe2+) and glutathione (GSH) was detected by specific kit. m6A sites were analyzed using SRAMP and further verify by methylated RNA immunoprecipitation (MeRIP) assay. Cell proliferation was determined by Cell Counting Kit-8 (CCK-8) assay. Cell death was determined by propidium iodide (PI) staining. FTO was downregulated in patients with neonatal HIE and OGD-treated neurons. Moreover, FTO mRNA expression was decreased in ferroptosis inducer, especially ferric ammonium citrate (FAC). However, overexpression of FTO inhibited the ferroptosis of neurons. Moreover, FTO-mediated N6-methyladenosine (m6A) modification of ferritin heavy chain 1 (FTH1) suppressed its mRNA expression and stability, inhibiting its protein expression. However, overexpression of FTH1 abrogated the effects of FTO and promoted the ferroptosis of neurons. In summary, FTO functions as a protective role in neonatal HIE via inhibiting FTH1 signaling. Thence, targeting may be a promising strategy for FTO neonatal HIE.


Subject(s)
Adenosine , Alpha-Ketoglutarate-Dependent Dioxygenase FTO , Ferroptosis , Hypoxia-Ischemia, Brain , Neurons , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Humans , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/genetics , Hypoxia-Ischemia, Brain/pathology , Ferroptosis/genetics , Neurons/metabolism , Adenosine/analogs & derivatives , Adenosine/metabolism , Infant, Newborn , Ferritins , Oxidoreductases
10.
Int J Mol Sci ; 25(16)2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39201562

ABSTRACT

Neonatal hypoxic-ischemic encephalopathy (HIE) is a critical condition characterized by significant brain damage due to insufficient blood flow and oxygen delivery at birth, leading to high rates of neonatal mortality and long-term neurological deficits worldwide. 2,3-Diphosphoglyceric acid (2,3-DPG), a small molecule metabolite prevalent in erythrocytes, plays an important role in regulating oxygen delivery, but its potential neuroprotective role in hypoxic-ischemic brain damage (HIBD) has yet to be fully elucidated. Our research reveals that the administration of 2,3-DPG effectively reduces neuron damage caused by hypoxia-ischemia (HI) both in vitro and in vivo. We observed a notable decrease in HI-induced neuronal cell apoptosis, attributed to the downregulation of Bax and cleaved-caspase 3, alongside an upregulation of Bcl-2 expression. Furthermore, 2,3-DPG significantly alleviates oxidative stress and mitochondrial damage induced by oxygen-glucose deprivation/reperfusion (OGD/R). The administration of 2,3-DPG in rats subjected to HIBD resulted in a marked reduction in brain edema and infarct volume, achieved through the suppression of neuronal apoptosis and neuroinflammation. Using RNA-seq analysis, we validated that 2,3-DPG offers protection against neuronal apoptosis under HI conditions by modulating the p38 MAPK pathway. These insights indicated that 2,3-DPG might act as a promising novel therapeutic candidate for HIE.


Subject(s)
Apoptosis , Hypoxia-Ischemia, Brain , p38 Mitogen-Activated Protein Kinases , Animals , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/drug therapy , Hypoxia-Ischemia, Brain/pathology , p38 Mitogen-Activated Protein Kinases/metabolism , Rats , Apoptosis/drug effects , Oxidative Stress/drug effects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neurons/metabolism , Neurons/drug effects , Neurons/pathology , Rats, Sprague-Dawley , Male , Mitochondria/metabolism , Mitochondria/drug effects
11.
Neuroreport ; 35(14): 904-908, 2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39166416

ABSTRACT

We investigated differences in subcortical white matter according to the presence disorders of consciousness (DOC) in patients with hypoxic-ischemic brain injury (HI-BI), using tract-based spatial statistics (TBSS). Thirty-two consecutive patients with HI-BI were recruited. The patients were assigned in group A [preserved consciousness (Glasgow Coma Scale: 15 and Coma Recovery Scale-revised (CRS-R): 23, 9 patients)] or group B [DOC present (Glasgow Coma Scale <15 and CRS-R < 23, 20 patients)]. Voxel-wise statistical analysis of fractional anisotropy data was performed by using TBSS as implemented in the FMRIB Software Library. We calculated mean fractional anisotropy values across the white matter skeleton and within 48 regions of interest (ROIs) based on intersections between the skeleton and the probabilistic Johns Hopkins University white matter atlases. Among the 48 ROIs examined, the fractional anisotropy values of two ROIs (the left superior corona radiata, and left tapetum) were significantly lower in group B than in group A ( P  < 0.05). No significant differences were observed, however, in the other 46 ROIs ( P  > 0.05). Our results suggest that abnormalities of the superior corona radiata and tapetum may be critical for DOC presence in patients with HI-BI.


Subject(s)
Consciousness Disorders , Hypoxia-Ischemia, Brain , White Matter , Humans , Hypoxia-Ischemia, Brain/diagnostic imaging , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/complications , Female , White Matter/diagnostic imaging , White Matter/pathology , Male , Middle Aged , Consciousness Disorders/diagnostic imaging , Consciousness Disorders/etiology , Consciousness Disorders/pathology , Adult , Diffusion Tensor Imaging/methods , Aged , Anisotropy , Young Adult
12.
Exp Neurol ; 381: 114929, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39168170

ABSTRACT

Neonatal hypoxia-ischemia (HI) is one of the main causes of mortality and long-term disabilities in newborns, and the only clinical approach to treat this condition is therapeutic hypothermia, which shows some limitations. Thus, putative neuroprotective agents have been tested in animal models of HI. Lactate is a preferential metabolic substrate of the neonatal brain and has already been shown to produce beneficial neuroprotective outcomes in neonatal animals exposed to HI. Here, we administered lactate as a treatment in neonatal rats previously exposed to HI and evaluated the impact of this treatment in adulthood. Seven-day-old (P7) male and female Wistar rats underwent permanent common right carotid occlusion combined with an exposition to a hypoxic atmosphere (8% oxygen) for 60 min. Animals were assigned to one of four experimental groups: HI, HI+LAC, SHAM, SHAM+LAC. Lactate was administered intraperitoneally 30 min and 2 h after hypoxia in HI+LAC and SHAM+LAC groups, whereas HI and SHAM groups received vehicle. Animals were tested in the behavioral tasks of negative geotaxis and righting reflex (P8), cylinder test (P24), and the modified neurological severity score was calculated (P25). Open field (OF), and novel object recognition (NOR) were evaluated in adulthood. Animals were killed at P60, and the brains were harvested and processed to evaluate the volume of brain injury. Our results showed that lactate administration reduced the volume of brain lesion and improved sensorimotor and cognitive behaviors in neonatal, juvenile, and adult life in HI animals from both sexes. Thus, lactate administration might be considered as a potential neuroprotective strategy for the treatment of neonatal HI, which is a prevalent disorder affecting newborns.


Subject(s)
Animals, Newborn , Hypoxia-Ischemia, Brain , Lactic Acid , Neuroprotective Agents , Rats, Wistar , Animals , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/metabolism , Rats , Female , Male , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Lactic Acid/metabolism , Disease Models, Animal , Recognition, Psychology/drug effects , Exploratory Behavior/drug effects
13.
Immun Inflamm Dis ; 12(8): e70000, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39172048

ABSTRACT

BACKGROUND: Oxidative stress (OS) plays a major role in the progress of hypoxic-ischemic brain damage (HIBD). This study aimed to investigate OS-related genes and their underlying molecular mechanisms in neonatal HIBD. METHODS: Microarray data sets were acquired from the Gene Expression Omnibus (GEO) database to screen the differentially expressed genes (DEGs) between control samples and HIBD samples. OS-related genes were drawn from GeneCards and OS-DEGs in HIBD were obtained by intersecting with the DEGs. Subsequently, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA) were conducted to determine the underlying mechanisms and functions of OS-DEGs in HIBD. Moreover, the hub genes were screened using the protein-protein interaction network and identified in the GSE144456 data set. CIBERSORT was then performed to evaluate the expression of immunocytes in each sample and perform a correlation analysis of the optimal OS-DEGs and immunocytes. Finally, quantitative reverse transcription polymerase chain reaction (RT-qPCR) and immunohistochemistry were performed to validate the expression levels of the optimal OS-DEGs. RESULTS: In total, 93 OS-DEGs were identified. GO, KEGG, and GSEA enrichment analyses indicated that these genes were predominantly enriched in OS and inflammation. Four OS-related biomarker genes (Jun, Fos, Tlr2, and Atf3) were identified and verified. CIBERSORT analysis revealed the dysregulation of six types of immune cells in the HIBD group. Moreover, 47 drugs that might target four OS-related biomarker genes were screened. Eventually, RT-qPCR and immunohistochemistry results for rat samples further validated the expression levels of Fos, Tlr2, and Atf3. CONCLUSIONS: Fos, Tlr2 and Atf3 are potential OS-related biomarkers of HIBD progression. The mechanisms of OS are associated with those of neonatal HIBD.


Subject(s)
Computational Biology , Hypoxia-Ischemia, Brain , Oxidative Stress , Protein Interaction Maps , Computational Biology/methods , Hypoxia-Ischemia, Brain/genetics , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Animals , Gene Expression Profiling , Humans , Rats , Gene Ontology , Gene Regulatory Networks , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Databases, Genetic , Gene Expression Regulation
14.
ACS Chem Neurosci ; 15(15): 2811-2821, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39058922

ABSTRACT

Neonatal hypoxic-ischemic (HI) brain insult is a major cause of neonatal mortality and morbidity. To assess the underlying pathological mechanisms, we mapped the spatiotemporal changes in polyamine, amino acid, and neurotransmitter levels, following HI insult (by the Rice-Vannucci method) in the brains of seven-day-old rat pups. Matrix-assisted laser desorption/ionization mass spectrometry imaging of chemically modified small-molecule metabolites by 4-(anthracen-9-yl)-2-fluoro-1-methylpyridin-1-ium iodide revealed critical HI-related metabolomic changes of 22 metabolites in 14 rat brain subregions, much earlier than light microscopy detected signs of neuronal damage. For the first time, we demonstrated excessive polyamine oxidation and accumulation of 3-aminopropanal in HI neonatal brains, which was later accompanied by neuronal apoptosis enhanced by increases in glycine and norepinephrine in critically affected brain regions. Specifically, putrescine, cadaverine, and 3-aminopropanal increased significantly as early as 12 h postinsult, mainly in motor and somatosensory cortex, hippocampus, and midbrain, followed by an increase in norepinephrine 24 h postinsult, which was predominant in the caudate putamen, the region most vulnerable to HI. The decrease of γ-aminobutyric acid (GABA) and the continuous dysregulation of the GABAergic system together with low taurine levels up to 36 h sustained progressive neurodegenerative cellular processes. The molecular alterations presented here at the subregional rat brain level provided unprecedented insight into early metabolomic changes in HI-insulted neonatal brains, which may further aid in the identification of novel therapeutic targets for the treatment of neonatal HI encephalopathy.


Subject(s)
Animals, Newborn , Brain , Hypoxia-Ischemia, Brain , Neurotransmitter Agents , Polyamines , Animals , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Polyamines/metabolism , Brain/metabolism , Neurotransmitter Agents/metabolism , Rats , Rats, Sprague-Dawley , Neurons/metabolism , Metabolomics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods
15.
J Cell Mol Med ; 28(13): e18505, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39001579

ABSTRACT

Hypoxia-ischaemia (HI) can induce the death of cerebrovascular constituent cells through oxidative stress. Hydrogen is a powerful antioxidant which can activate the antioxidant system. A hypoxia-ischaemia brain damage (HIBD) model was established in 7-day-old SD rats. Rats were treated with different doses of hydrogen-rich water (HRW), and brain pericyte oxidative stress damage, cerebrovascular function and brain tissue damage were assessed. Meanwhile, in vitro-cultured pericytes were subjected to oxygen-glucose deprivation and treated with different concentrations of HRW. Oxidative injury was measured and the molecular mechanism of how HRW alleviated oxidative injury of pericytes was also examined. The results showed that HRW significantly attenuated HI-induced oxidative stress in the brain pericytes of neonatal rats, partly through the Nrf2-HO-1 pathway, further improving cerebrovascular function and reducing brain injury and dysfunction. Furthermore, HRW is superior to a single-cell death inhibitor for apoptosis, ferroptosis, parthanatos, necroptosis and autophagy and can better inhibit HI-induced pericyte death. The liver and kidney functions of rats were not affected by present used HRW dose. This study elucidates the role and mechanism of hydrogen in treating HIBD from the perspective of pericytes, providing new theoretical evidence and mechanistic references for the clinical application of hydrogen in neonatal HIE.


Subject(s)
Animals, Newborn , Brain , Hydrogen , Hypoxia-Ischemia, Brain , Oxidative Stress , Pericytes , Rats, Sprague-Dawley , Animals , Pericytes/drug effects , Pericytes/metabolism , Hydrogen/pharmacology , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/drug therapy , Rats , Oxidative Stress/drug effects , Brain/pathology , Brain/drug effects , Brain/metabolism , NF-E2-Related Factor 2/metabolism , Apoptosis/drug effects , Disease Models, Animal , Antioxidants/pharmacology
16.
J Neuroimmune Pharmacol ; 19(1): 34, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38949694

ABSTRACT

Amorfrutin B is a selective PPARγ modulator that we demonstrated to be a promising neuroprotective compound in cellular models of stroke and perinatal asphyxia. Although neuronal mechanisms of amorfrutin B-evoked neuroprotection have been identified, none of them reflects the actions of the compound on microglia, which play a pivotal role in brain response to hypoxia/ischemia. Here, we provide evidence for amorfrutin B-induced effects on human microglia subjected to hypoxia/ischemia; the compound counteracts inflammation, and influences mitochondrial status and proliferation potential in a PPARγ-dependent manner. Post-treatment with amorfrutin B decreased the IBA1 fluorescence intensity, reduced caspase-1 activity, and downregulated IL1B/IL-1ß and TNFA but not IL10/IL-10 expression, which was upregulated. Amorfrutin B also stimulated PPARγ signaling, as evidenced by increased mRNA and/or protein levels of PPARγ and PGC1α. In addition, amorfrutin B reversed the hypoxia/ischemia-evoked effects on mitochondria-related parameters, such as mitochondrial membrane potential, BCL2/BCL2 expression and metabolic activity, which were correlated with diminished proliferation potential of microglia. Interestingly, the inhibitory effect of amorfrutin B on the proliferation potential and mitochondrial function of microglia is opposite to the stimulatory effect of amorfrutin B on mouse neuronal survival, as evidenced by increased neuronal viability and reduced neurodegeneration. In summary, this study showed for the first time that amorfrutin B compromises hypoxia/ischemia-induced activation of human microglia in a PPARγ-dependent manner, which involves inhibiting inflammation, normalizing mitochondrial status, and controlling proliferation potential. These data extend the protective potential of amorfrutin B in the pharmacotherapy of hypoxic/ischemic brain injury, targeting not only neurons but also activated microglia.


Subject(s)
Cell Proliferation , Hypoxia-Ischemia, Brain , Microglia , Mitochondria , PPAR gamma , PPAR gamma/metabolism , Humans , Microglia/drug effects , Microglia/metabolism , Cell Proliferation/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Hypoxia-Ischemia, Brain/drug therapy , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Inflammation/metabolism , Inflammation/drug therapy , Cells, Cultured , Neuroprotective Agents/pharmacology
17.
Int J Dev Neurosci ; 84(6): 594-604, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38940222

ABSTRACT

Pyroptosis is an inflammation-associated programmed cell death, and neuroinflammation is strongly associated with severe neurological deficits in neonatal hypoxic-ischemic encephalopathy (HIE). Ethyl pyruvate (EP), a known anti-inflammatory agent, has shown promise in the treatment of hypoxic-ischemic brain damage (HIBD) rats; nevertheless, the therapeutic mechanism of EP and its capacity to suppress neuronal pyroptosis in HIBD rats remain unclear. In both the neonatal Rice-Vannucci rat model and the OGD/R model, this study examined alterations in the NLRP3/Caspase-1/GSDMD classical pyroptosis pathway in hippocampal neurons during HIE and the potential inhibitory impact of ethyl pyruvate on this pathway. We used HE staining, immunofluorescence double staining, transmission electron microscopy, and western blot to demonstrate that EP effectively inhibited hippocampal neuronal pyroptosis and attenuated the activation of the NLRP3/Caspase-1/GSDMD signaling pathway in HIBD rats, which resulted in a reduction of neuroinflammation and facilitated neural recovery. The results suggest that EP may be a promising neuroprotective agent for treating HIE.


Subject(s)
Caspase 1 , Hypoxia-Ischemia, Brain , NLR Family, Pyrin Domain-Containing 3 Protein , Neurons , Pyroptosis , Pyruvates , Rats, Sprague-Dawley , Animals , Rats , Animals, Newborn , Caspase 1/metabolism , Gasdermins , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/metabolism , Hypoxia-Ischemia, Brain/drug therapy , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/metabolism , Neurons/drug effects , Neurons/pathology , Neurons/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Phosphate-Binding Proteins/metabolism , Pyroptosis/drug effects , Pyroptosis/physiology , Pyruvates/pharmacology , Pyruvates/therapeutic use , Signal Transduction/drug effects
18.
Biochem Biophys Res Commun ; 726: 150259, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-38909535

ABSTRACT

Hypoxic-ischemic brain damage (HIBD) in the perinatal period is an important cause of cerebral damage and long-term neurological sequelae, and can place much pressure on families and society. Our previous study demonstrated that miRNA-326 reduces neuronal apoptosis by up-regulating the δ-opioid receptor (DOR) under oxygen-glucose deprivation in vitro. In the present study, we aimed to explore the neuroprotective effects of the miRNA-326/DOR axis by inhibiting apoptosis in HIBD using neonatal miRNA-326 knockout mice. Neonatal C57BL/6 mice, neonatal miRNA-326 knockout mice, and neonatal miRNA-326 knockout mice intraperitoneally injected with the DOR inhibitor naltrindole were treated with hypoxic-ischemia (HI). Neurological deficit scores, magnetic resonance imaging, terminal deoxynucleotidyl transferase-mediated uridine 5'-triphosphate-biotin nick end labeling, and Caspase-3, Bax, and B-cell lymphoma 2 (Bcl-2) expression were evaluated on day 2 after HI. Neurobehavioral analyses were performed on days 2 and 28 after HI. Additionally, the Morris water maze test was conducted on days 28. Compared with HI-treated neonatal C57BL/6 mice, HI-treated neonatal miRNA-326 knockout mice had higher neurological deficit scores, smaller cerebral infarction areas, and improved motor function, reaction ability, and long-term spatial learning and memory. These effects were likely the result of inhibiting apoptosis; the DOR inhibitor reversed these neuroprotective effects. Our findings indicate that miRNA-326 knockout plays a neuroprotective effect in neonatal HIBD by inhibiting apoptosis via the target gene DOR.


Subject(s)
Animals, Newborn , Apoptosis , Hypoxia-Ischemia, Brain , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs , Receptors, Opioid, delta , Animals , Male , Mice , Apoptosis/genetics , Hypoxia-Ischemia, Brain/genetics , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Neuroprotective Agents/pharmacology , Receptors, Opioid, delta/genetics , Receptors, Opioid, delta/metabolism
19.
Neuroscience ; 554: 83-95, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-38944149

ABSTRACT

Neonatal hypoxic-ischemic brain damage (HIBD) can lead to mortality and severe neurological dysfunction. Emodin is a natural anthraquinone derivative that is easy to obtain and has good neuroprotective effects. This study aimed to investigate the neuroprotective effect of emodin on neonatal mouse HIBD. The modified Rice-Vannucci method was used to induce HIBD in mouse pups. Eighty postnatal 7-day (P7) C57BL/6 neonatal mice were randomly divided into the sham group (sham), vehicle group (vehicle), and emodin group (emodin). TTC staining and whole-brain morphology were used to evaluate the infarct volume and morphology of the brain tissue. The condition of the neurons was observed through Nissl staining, HE staining, FJC staining, immunofluorescence and Western blot for NeuN, IBA-1, and GFAP. The physiological status of the mice was evaluated using weight measurements. The neural function of the mice was assessed using the negative geotaxis test, righting reflex test, and grip test. TUNEL staining was used to detect apoptosis in brain cells. Finally, Western blot and immunofluorescence were used to detect the expression levels of apoptosis-related proteins, such as P53, cleaved caspase-3, Bax and Bcl-2, in the brain. Experiments have shown that emodin can reduce the cerebral infarct volume, brain oedema, neuronal apoptosis, and degeneration and improve the reconstruction of brain tissue morphology, neuronal morphology, physiological conditions, and neural function. Additionally, emodin inhibited the expression of proapoptotic proteins such as P53, Bax and cleaved caspase-3 and promoted the expression of the antiapoptotic protein Bcl-2. Emodin attenuates HIBD by inhibiting neuronal apoptosis in neonatal mice.


Subject(s)
Animals, Newborn , Apoptosis , Brain , Emodin , Hypoxia-Ischemia, Brain , Mice, Inbred C57BL , Neurons , Neuroprotective Agents , Animals , Emodin/pharmacology , Apoptosis/drug effects , Hypoxia-Ischemia, Brain/drug therapy , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/metabolism , Neurons/drug effects , Neurons/pathology , Neurons/metabolism , Neuroprotective Agents/pharmacology , Brain/drug effects , Brain/pathology , Brain/metabolism , Mice , Disease Models, Animal
20.
Immun Inflamm Dis ; 12(6): e1320, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38888378

ABSTRACT

BACKGROUND: At present, neonatal hypoxic-ischemic encephalopathy (HIE), especially moderate to severe HIE, is a challenging disease for neonatologists to treat, and new alternative/complementary treatments are urgently needed. The neuroinflammatory cascade triggered by hypoxia-ischemia (HI) insult is one of the core pathological mechanisms of HIE. Early inhibition of neuroinflammation provides long-term neuroprotection. Plant-derived monomers have impressive anti-inflammatory effects. Aloesin (ALO) has been shown to have significant anti-inflammatory and antioxidant effects in diseases such as ulcerative colitis, but its role in HIE is unclear. To this end, we conducted a series of experiments to explore the potential mechanism of ALO in preventing and treating brain damage caused by HI insult. MATERIALS AND METHODS: Hypoxic-ischemic brain damage (HIBD) was induced in 7-day-old Institute of Cancer Research (ICR) mice, which were then treated with 20 mg/kg ALO. The neuroprotective effects of ALO on HIBD and the underlying mechanism were evaluated through neurobehavioral testing, infarct size measurement, apoptosis detection, protein and messenger RNA level determination, immunofluorescence, and molecular docking. RESULTS: ALO alleviated the long-term neurobehavioral deficits caused by HI insult; reduced the extent of cerebral infarction; inhibited cell apoptosis; decreased the levels of the inflammatory factors interleukin (IL)-1ß, IL-6, and tumor necrosis factor-α; activated microglia and astrocytes; and downregulated the protein expression of members in the TLR4 signaling pathway. In addition, molecular docking showed that ALO can bind stably to TLR4. CONCLUSION: ALO ameliorated HIBD in neonatal mice by inhibiting the neuroinflammatory response mediated by TLR4 signaling.


Subject(s)
Animals, Newborn , Hypoxia-Ischemia, Brain , Neuroinflammatory Diseases , Neuroprotective Agents , Toll-Like Receptor 4 , Animals , Toll-Like Receptor 4/metabolism , Hypoxia-Ischemia, Brain/drug therapy , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Mice , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/metabolism , Mice, Inbred ICR , Disease Models, Animal , Signal Transduction/drug effects , Apoptosis/drug effects , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Molecular Docking Simulation
SELECTION OF CITATIONS
SEARCH DETAIL