Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (34)

Search Parameters:
Keywords = unilateral brain injury

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 2443 KiB  
Article
Contralateral Structure and Molecular Response to Severe Unilateral Brain Injury
by Xixian Liao, Xiaojian Xu, Ming Li, Runfa Tian, Yuan Zhuang and Guoyi Gao
Brain Sci. 2025, 15(8), 837; https://doi.org/10.3390/brainsci15080837 - 5 Aug 2025
Viewed by 471
Abstract
Background: Severe damage to one side of the brain often leads to adverse consequences and can also cause widespread changes throughout the brain, especially in the contralateral area. Studying molecular changes in the contralateral cerebral hemisphere, especially with regard to genetic regulation, [...] Read more.
Background: Severe damage to one side of the brain often leads to adverse consequences and can also cause widespread changes throughout the brain, especially in the contralateral area. Studying molecular changes in the contralateral cerebral hemisphere, especially with regard to genetic regulation, can help discover potential treatment strategies to promote recovery after severe brain trauma on one side. Methods: In our study, the right motor cortex was surgically removed to simulate severe unilateral brain injury, and changes in glial cells and synaptic structure in the contralateral cortex were subsequently assessed through immunohistological, morphological, and Western blot analyses. We conducted transcriptomic studies to explore changes in gene expression levels associated with the inflammatory response. Results: Seven days after corticotomy, levels of reactive astrocytes and hypertrophic microglia increased significantly in the experimental group, while synapsin-1 and PSD-95 levels in the contralateral motor cortex increased. These molecular changes are associated with structural changes, including destruction of dendritic structures and the encapsulation of astrocytes by synapses. Genome-wide transcriptome analysis showed a significant increase in gene pathways involved in inflammatory responses, synaptic activity, and nerve fiber regeneration in the contralateral cortex after corticorectomy. Key transcription factors such as NF-κB1, Rela, STAT3 and Jun were identified as potential regulators of these contralateral changes. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) confirmed that the mRNA expression levels of Cacna1c, Tgfb1 and Slc2a1 genes related to STAT3, JUN, and NF-κB regulation significantly increased in the contralateral cortex of the experimental group. Conclusions: After unilateral brain damage occurs, changes in the contralateral cerebral hemisphere are closely related to processes involving inflammation and synaptic function. Full article
Show Figures

Figure 1

19 pages, 1176 KiB  
Article
Temporal Changes in Mitochondria-Centric Excitotoxic Responses Following Severe Penetrating Traumatic Brain Injury
by Hiren R. Modi, Sudeep Musyaju, Anke H. Scultetus and Jignesh D. Pandya
Biomedicines 2025, 13(7), 1520; https://doi.org/10.3390/biomedicines13071520 - 21 Jun 2025
Viewed by 475
Abstract
Background/Objectives: Traumatic brain injury (TBI) remains a significant and urgent medical concern for the US military. TBI triggers excitotoxic responses immediately, involving mitochondrial dysfunction characterized by loss of calcium (Ca2+) cycling, membrane damage and increased cell death. However, a comprehensive understanding [...] Read more.
Background/Objectives: Traumatic brain injury (TBI) remains a significant and urgent medical concern for the US military. TBI triggers excitotoxic responses immediately, involving mitochondrial dysfunction characterized by loss of calcium (Ca2+) cycling, membrane damage and increased cell death. However, a comprehensive understanding of mitochondria-centric excitotoxic responses over time has yet to be fully demonstrated after severe TBI. The current study evaluated mitochondria-centric time course responses between 30 min and 2 weeks (seven time points) after penetrating TBI (pTBI). Methods: Anesthetized adult male Sprague-Dawley rats were subjected to either 10% unilateral pTBI or Sham craniectomy. Animals were euthanized at various time points, and mitochondria were isolated from the injury core. Results: Post-injury mitochondrial Ca2+ homeostasis was significantly compromised in pTBI compared to the Sham group. In parallel, mitochondrial membrane integrity markers, including cytochrome c (Cyt C) and voltage-dependent anion channel (VDAC), showed significant reduction over time post-pTBI. Apoptosis-responsive markers, such as glyceraldehyde 3-phosphate dehydrogenase (GAPDH) and B-cell lymphoma 2 (Bcl-2), exhibited elevated responses over time post-pTBI. Conclusions: Our results demonstrate profound insights into elevated excitotoxic mitochondrial damage after severe TBI. This time course study uncovers novel mitochondrial targets involved in TBI excitotoxicity and offers mitigation opportunities to alleviate excitotoxic responses after penetrating TBI. Full article
Show Figures

Figure 1

11 pages, 1048 KiB  
Case Report
Preservation of Bilateral Corticospinal Projections from Injured Hemisphere After Perinatal Stroke
by Cameron P. Casey, Ellen N. Sutter, Alina Grimaldo, Kellie M. Collins, Jose Guerrero-Gonzalez, Ryan M. McAdams, Douglas C. Dean and Bernadette T. Gillick
Brain Sci. 2025, 15(1), 82; https://doi.org/10.3390/brainsci15010082 - 17 Jan 2025
Viewed by 1238
Abstract
Background: Perinatal brain injury is a leading cause of developmental disabilities, including cerebral palsy. However, further work is needed to understand early brain development in the presence of brain injury. In this case report, we examine the longitudinal neuromotor development of a term [...] Read more.
Background: Perinatal brain injury is a leading cause of developmental disabilities, including cerebral palsy. However, further work is needed to understand early brain development in the presence of brain injury. In this case report, we examine the longitudinal neuromotor development of a term infant following a significant loss of right-hemispheric brain tissue due to a unilateral ischemic stroke. Our analysis focuses on the integrity and development of the corticospinal tract (CST) from the lesioned hemisphere. This case provides a unique opportunity to evaluate CST development after loss of the majority of the motor cortex. Methods: Evaluations were conducted when the infant was 4 (Visit-1), 18 (Visit 2), and 25 (Visit 3) months old. Assessments included magnetic resonance imaging (MRI) to characterize the lesion and quantify CST structural integrity, single-pulse transcranial magnetic stimulation (spTMS) to evaluate CST functional circuitry, and neuromotor assessments. Results: At Visit 1, bilateral CSTs were identified through diffusion-weighted MRI (dMRI) despite an estimated loss of 92.7% (7.3% retained) of age-typical motor cortex from the right hemisphere. Both hemispheres exhibited bilateral motor-evoked potential in response to stimulation with spTMS, which remained when reassessed at Visits 2 and 3. Longitudinal MRI showed distinct developmental trajectories of CST integrity in each hemisphere, with the lesioned hemisphere exhibiting initial increases in integrity between Visits 1 and 2 followed by a decrease in integrity between Visits 2 and 3. The non-lesioned hemisphere showed increased integrity from Visit 1 to Visit 2, which remained stable at Visit 3. Motor assessments at all visits indicated a high risk of cerebral palsy. Conclusions: This report highlights the utility of MRI and spTMS in studying neuromotor development. The findings reveal preserved functional bilateral CST circuitry despite majority loss of the right-hemispheric motor cortex as well as distinct developmental trajectories in CST integrity between hemispheres. These results underscore the potential for neural plasticity after perinatal brain injury. Clinical Trials Registration: NCT05013736. Full article
Show Figures

Figure 1

8 pages, 984 KiB  
Article
Motor Learning Deficits in a Neonatal Mouse Model of Hypoxic-Ischemic Injury
by Maria Marlicz, Weronika Matysik, Emily Zucker, Sarah Lee, Hannah Mulhern and Jennifer Burnsed
Children 2025, 12(1), 27; https://doi.org/10.3390/children12010027 - 27 Dec 2024
Viewed by 992
Abstract
Background/Objectives: Motor deficits following neonatal brain injury, from cerebral palsy to subtle deficits in motor planning, are common yet underreported. Rodent models of motor deficits in neonatal hypoxia–ischemia (HI) allow improved understanding of the underlying mechanisms and neuroprotective strategies. Our goal was to [...] Read more.
Background/Objectives: Motor deficits following neonatal brain injury, from cerebral palsy to subtle deficits in motor planning, are common yet underreported. Rodent models of motor deficits in neonatal hypoxia–ischemia (HI) allow improved understanding of the underlying mechanisms and neuroprotective strategies. Our goal was to test motor performance and learning in a mouse model of neonatal HI. Methods: We induced HI in postnatal day (p)10 C57/Bl6 mice through unilateral carotid ligation followed by 60 min of 8% oxygen exposure, or a sham procedure. At p30, we assessed complex motor performance and learning using the accelerating rotarod and complex running wheel tasks. Results: In the rotarod task, HI mice performed worse than sham mice, with shorter latencies to fall (n = 6 sham, 9 HI; day 1, p = 0.033; day 2, p = 0.013; day 3, p = 0.023). Sham mice demonstrated improved performance across days (p = 0.005), and HI mice did not (p = 0.44). During the simple running wheel task, we observed no difference in wheel rotation and speed between groups (n = 5/group; day 1, p = 0.67; day 4, p = 0.53). However, when navigating a wheel with a random pattern of spokes removed (complex task), HI mice took longer than sham mice to reach a plateau in performance (n = 5/group; day 1, p = 0.02; day 4, p = 0.77). Conclusions: Our findings demonstrate that young adult mice exposed to HI exhibit significant deficits and delayed learning in complex motor performance compared to sham mice. HI mice do not show deficits in gross motor performance; however, more subtle impairments are present in complex motor performance and learning. This HI model exhibits subtle motor deficits relevant to findings in humans and may be a useful tool in testing further neuroprotective strategies. Full article
Show Figures

Figure 1

9 pages, 1240 KiB  
Article
Electrophysiological Screening to Assess Foot Drop Syndrome in Severe Acquired Brain Injury in Rehabilitative Settings
by Francesco Piccione, Antonio Cerasa, Paolo Tonin, Simone Carozzo, Rocco Salvatore Calabrò, Stefano Masiero and Lucia Francesca Lucca
Biomedicines 2024, 12(4), 878; https://doi.org/10.3390/biomedicines12040878 - 16 Apr 2024
Cited by 1 | Viewed by 2173
Abstract
Background: Foot drop syndrome (FDS), characterized by severe weakness and atrophy of the dorsiflexion muscles of the feet, is commonly found in patients with severe acquired brain injury (ABI). If the syndrome is unilateral, the cause is often a peroneal neuropathy (PN), due [...] Read more.
Background: Foot drop syndrome (FDS), characterized by severe weakness and atrophy of the dorsiflexion muscles of the feet, is commonly found in patients with severe acquired brain injury (ABI). If the syndrome is unilateral, the cause is often a peroneal neuropathy (PN), due to compression of the nervous trunk on the neck of the fibula at the knee level; less frequently, the cause is a previous or concomitant lumbar radiculopathy. Bilateral syndromes are caused by polyneuropathies and myopathies. Central causes, due to brain or spinal injury, mimic this syndrome but are usually accompanied by other symptoms, such as spasticity. Critical illness polyneuropathy (CIP) and myopathy (CIM), isolated or in combination (critical illness polyneuromyopathy, CIPNM), have been shown to constitute an important cause of FDS in patients with ABI. Assessing the causes of FDS in the intensive rehabilitation unit (IRU) has several limitations, which include the complexity of the electrophysiological tests, limited availability of neurophysiology consultants, and the severe disturbance in consciousness and lack of cooperation from patients. Objectives: We sought to propose a simplified electrophysiological screening that identifies FDS causes, particularly PN and CIPNM, to help clinicians to recognize the significant clinical predictors of poor outcomes in severe ABI at admission to IRU. Methods: This prospective, single-center study included 20 severe ABI patients with FDS (11 females/9 males, mean age 55.10 + 16.26; CRS-R= 11.90 + 6.32; LCF: 3.30 + 1.30; DRS: 21.45 + 3.33), with prolonged rehabilitation treatment (≥2 months). We applied direct tibialis anterior muscle stimulation (DMS) associated with peroneal nerve motor conduction evaluation, across the fibular head (NCS), to identify CIP and/or CIM and to exclude demyelinating or compressive unilateral PN. Results: At admission to IRU, simplified electrophysiological screening reported four unilateral PN, four CIP and six CIM with a CIPNM overall prevalence estimate of about 50%. After 2 months, the CIPNM group showed significantly poorer outcomes compared to other ABI patients without CIPNM, as demonstrated by the lower probability of achieving endotracheal-tube weaning (20% versus 90%) and lower CRS-R and DRS scores. Due to the subacute rehabilitation setting of our study, it was not possible to evaluate the motor results of recovery of the standing position, functional walking and balance, impaired by the presence of unilateral PN. Conclusions: The implementation of the proposed simplified electrophysiological screening may enable the early identification of unilateral PN or CIPNM in severe ABI patients, thereby contributing to better functional prognosis in rehabilitative settings. Full article
Show Figures

Figure 1

20 pages, 18479 KiB  
Article
Pulmonary Effects of Traumatic Brain Injury in Mice: A Gene Set Enrichment Analysis
by Wei-Hung Chan, Shih-Ming Huang and Yi-Lin Chiu
Int. J. Mol. Sci. 2024, 25(5), 3018; https://doi.org/10.3390/ijms25053018 - 5 Mar 2024
Cited by 2 | Viewed by 2622
Abstract
Acute lung injury occurs in 20–25% of cases following traumatic brain injury (TBI). We investigated changes in lung transcriptome expression post-TBI using animal models and bioinformatics. Employing unilateral controlled cortical impact for TBI, we conducted microarray analysis after lung acquisition, followed by gene [...] Read more.
Acute lung injury occurs in 20–25% of cases following traumatic brain injury (TBI). We investigated changes in lung transcriptome expression post-TBI using animal models and bioinformatics. Employing unilateral controlled cortical impact for TBI, we conducted microarray analysis after lung acquisition, followed by gene set enrichment analysis of differentially expressed genes. Our findings indicate significant upregulation of inflammation-related genes and downregulation of nervous system genes. There was enhanced infiltration of adaptive immune cells, evidenced by positive enrichment in Lung-Th1, CD4, and CD8 T cells. Analysis using the Tabula Sapiens database revealed enrichment in lung-adventitial cells, pericytes, myofibroblasts, and fibroblasts, indicating potential effects on lung vasculature and fibrosis. Gene set enrichment analysis linked TBI to lung diseases, notably idiopathic pulmonary hypertension. A Venn diagram overlap analysis identified a common set of 20 genes, with FOSL2 showing the most significant fold change. Additionally, we observed a significant increase in ADRA1AIL6 production post-TBI using the L1000 library. Our study highlights the impact of brain trauma on lung injury, revealing crucial gene expression changes related to immune cell infiltration, cytokine production, and potential alterations in lung vasculature and fibrosis, along with a specific spectrum of disease influence. Full article
Show Figures

Figure 1

14 pages, 7655 KiB  
Article
An Improved Surgical Approach for Complete Interhemispheric Corpus Callosotomy Combined with Extended Frontoparietal Craniotomy in Mice
by Ilja Jelisejevs, Jolanta Upite, Shivan Kalnins and Baiba Jansone
Biomedicines 2023, 11(7), 1782; https://doi.org/10.3390/biomedicines11071782 - 21 Jun 2023
Cited by 1 | Viewed by 3807
Abstract
Callosotomy is an invasive method that is used to study the role of interhemispheric functional connectivity in the brain. This surgical approach is technically demanding to perform in small laboratory animals, such as rodents, due to several methodological challenges. To date, there exist [...] Read more.
Callosotomy is an invasive method that is used to study the role of interhemispheric functional connectivity in the brain. This surgical approach is technically demanding to perform in small laboratory animals, such as rodents, due to several methodological challenges. To date, there exist two main approaches for transecting the corpus callosum (CC) in rodents: trephine hole(s) or unilateral craniotomy, which cause damage to the cerebral cortex or the injury of large vessels, and may lead to intracranial hemorrhage and animal death. This study presents an improved surgical approach for complete corpus callosotomy in mice using an interhemispheric approach combined with bilateral and extended craniotomy across the midline. This study demonstrated that bilateral and extended craniotomy provided the visual space required for hemisphere and sinus retraction, thus keeping large blood vessels and surrounding brain structures intact under the surgical microscope using standardized surgical instruments. We also emphasized the importance of good post-operative care leading to an increase in overall animal survival following experimentation. This optimized surgical approach avoids extracallosal tissue and medium- to large-sized cerebral blood vessel damage in mice, which can provide higher study reproducibility/validity among animals when revealing the role of the CC in various neurological pathologies. Full article
(This article belongs to the Section Neurobiology and Clinical Neuroscience)
Show Figures

Figure 1

19 pages, 4703 KiB  
Article
Atypical Neurogenesis, Astrogliosis, and Excessive Hilar Interneuron Loss Are Associated with the Development of Post-Traumatic Epilepsy
by Erwin Kristobal Gudenschwager-Basso, Oleksii Shandra, Troy Volanth, Dipan C. Patel, Colin Kelly, Jack L. Browning, Xiaoran Wei, Elizabeth A. Harris, Dzenis Mahmutovic, Alexandra M. Kaloss, Fernanda Guilhaume Correa, Jeremy Decker, Biswajit Maharathi, Stefanie Robel, Harald Sontheimer, Pamela J. VandeVord, Michelle L. Olsen and Michelle H. Theus
Cells 2023, 12(9), 1248; https://doi.org/10.3390/cells12091248 - 25 Apr 2023
Cited by 11 | Viewed by 4172
Abstract
Background: Traumatic brain injury (TBI) remains a significant risk factor for post-traumatic epilepsy (PTE). The pathophysiological mechanisms underlying the injury-induced epileptogenesis are under investigation. The dentate gyrus—a structure that is highly susceptible to injury—has been implicated in the evolution of seizure development. Methods: [...] Read more.
Background: Traumatic brain injury (TBI) remains a significant risk factor for post-traumatic epilepsy (PTE). The pathophysiological mechanisms underlying the injury-induced epileptogenesis are under investigation. The dentate gyrus—a structure that is highly susceptible to injury—has been implicated in the evolution of seizure development. Methods: Utilizing the murine unilateral focal control cortical impact (CCI) injury, we evaluated seizure onset using 24/7 EEG video analysis at 2–4 months post-injury. Cellular changes in the dentate gyrus and hilus of the hippocampus were quantified by unbiased stereology and Imaris image analysis to evaluate Prox1-positive cell migration, astrocyte branching, and morphology, as well as neuronal loss at four months post-injury. Isolation of region-specific astrocytes and RNA-Seq were performed to determine differential gene expression in animals that developed post-traumatic epilepsy (PTE+) vs. those animals that did not (PTE), which may be associated with epileptogenesis. Results: CCI injury resulted in 37% PTE incidence, which increased with injury severity and hippocampal damage. Histological assessments uncovered a significant loss of hilar interneurons that coincided with aberrant migration of Prox1-positive granule cells and reduced astroglial branching in PTE+ compared to PTE mice. We uniquely identified Cst3 as a PTE+-specific gene signature in astrocytes across all brain regions, which showed increased astroglial expression in the PTE+ hilus. Conclusions: These findings suggest that epileptogenesis may emerge following TBI due to distinct aberrant cellular remodeling events and key molecular changes in the dentate gyrus of the hippocampus. Full article
(This article belongs to the Special Issue Advances in Neurogenesis: 2nd Edition)
Show Figures

Graphical abstract

19 pages, 4894 KiB  
Article
Upregulation of Sarcolemmal Hemichannels and Inflammatory Transcripts with Neuromuscular Junction Instability during Lower Limb Unloading in Humans
by Giuseppe Sirago, Julián Candia, Martino V. Franchi, Fabio Sarto, Elena Monti, Luana Toniolo, Carlo Reggiani, Emiliana Giacomello, Sandra Zampieri, Lisa M. Hartnell, Giuseppe De Vito, Marco Sandri, Luigi Ferrucci and Marco V. Narici
Biology 2023, 12(3), 431; https://doi.org/10.3390/biology12030431 - 10 Mar 2023
Cited by 7 | Viewed by 2802
Abstract
Human skeletal muscle atrophy and a disproportionate force loss occur within a few days of unloading in space and on Earth, but the underlying mechanisms are not fully understood. Disruption of neuromuscular junction homeostasis has been proposed as one of the possible causes. [...] Read more.
Human skeletal muscle atrophy and a disproportionate force loss occur within a few days of unloading in space and on Earth, but the underlying mechanisms are not fully understood. Disruption of neuromuscular junction homeostasis has been proposed as one of the possible causes. Here, we investigated the potential mechanisms involved in this neuromuscular disruption induced by a 10-day unilateral lower limb suspension (ULLS) in humans. Specifically, we investigated hemichannels’ upregulation, neuromuscular junction and axonal damage, neurotrophins’ receptor downregulation and inflammatory transcriptional signatures. Biomarkers were evaluated at local and systemic levels. At the sarcolemmal level, changes were found to be associated with an increased expression of connexin 43 and pannexin-1. Upregulation of the inflammatory transcripts revealed by deep transcriptomics was found after 10 days of ULLS. The destabilisation of the neuromuscular junction was not accompanied by changes in the secretion of the brain-derived neurotrophic factor and neurotrophin-4, while their receptor, BDNF/NT growth factors receptor (TrkB), decreased. Furthermore, at 5 days of ULLS, there was already a significant upregulation of the serum neurofilament light chain concentration, an established clinical biomarker of axonal injury. At 10 days of ULLS, other biomarkers of early denervation processes appeared. Hence, short periods of muscle unloading induce sarcolemmal hemichannels upregulation, inflammatory transcripts upregulation, neuromuscular junction instability and axonal damage. Full article
(This article belongs to the Section Physiology)
Show Figures

Figure 1

10 pages, 1243 KiB  
Brief Report
A Single Episode of Cortical Spreading Depolarization Increases mRNA Levels of Proinflammatory Cytokines, Calcitonin Gene-Related Peptide and Pannexin-1 Channels in the Cerebral Cortex
by Maria N. Volobueva, Elena M. Suleymanova, Maria P. Smirnova, Alexey P. Bolshakov and Lyudmila V. Vinogradova
Int. J. Mol. Sci. 2023, 24(1), 85; https://doi.org/10.3390/ijms24010085 - 21 Dec 2022
Cited by 18 | Viewed by 2336
Abstract
Cortical spreading depolarization (CSD) is the neuronal correlate of migraine aura and the reliable consequence of acute brain injury. The role of CSD in triggering headaches that follow migraine aura and brain injury remains to be uncertain. We examined whether a single CSD [...] Read more.
Cortical spreading depolarization (CSD) is the neuronal correlate of migraine aura and the reliable consequence of acute brain injury. The role of CSD in triggering headaches that follow migraine aura and brain injury remains to be uncertain. We examined whether a single CSD occurring in awake animals modified the expression of proinflammatory cytokines (Il1b, TNF, and Il6) and endogenous mediators of nociception/neuroinflammation-pannexin 1 (Panx1) channel and calcitonin gene-related peptide (CGRP), transforming growth factor beta (TGFb) in the cortex. Unilateral microinjury of the somatosensory cortex triggering a single CSD was produced in awake Wistar rats. Three hours later, tissue samples from the lesioned cortex, intact ipsilesional cortex invaded by CSD, and homologous areas of the contralateral sham-treated cortex were harvested and analyzed using qPCR. Three hours post-injury, intact CSD-exposed cortexes increased TNF, Il1b, Panx1, and CGRP mRNA levels. The strongest upregulation of proinflammatory cytokines was observed at the injury site, while CGRP and Panx1 were upregulated more strongly in the intact cortexes invaded by CSD. A single CSD is sufficient to produce low-grade parenchymal neuroinflammation with simultaneous overexpression of Panx1 and CGRP. The CSD-induced molecular changes may contribute to pathogenic mechanisms of migraine pain and post-injury headache. Full article
(This article belongs to the Special Issue Molecular Research of Migraine: From Pathogenesis to Treatment)
Show Figures

Figure 1

18 pages, 2439 KiB  
Article
Effects of Three Different Doses of Inter-Alpha Inhibitor Proteins on Severe Hypoxia–Ischemia-Related Brain Injury in Neonatal Rats
by Liam M. Koehn, Kevin Nguyen, Xiaodi Chen, Andre Santoso, Richard Tucker, Yow-Pin Lim and Barbara S. Stonestreet
Int. J. Mol. Sci. 2022, 23(21), 13473; https://doi.org/10.3390/ijms232113473 - 3 Nov 2022
Cited by 7 | Viewed by 2817
Abstract
Hypoxia–ischemia (HI)-related brain injury is an important cause of morbidity and long-standing disability in newborns. We have previously shown that human plasma-derived inter-alpha inhibitor proteins (hIAIPs) attenuate HI-related brain injury in neonatal rats. The optimal dose of hIAIPs for their neuroprotective effects and [...] Read more.
Hypoxia–ischemia (HI)-related brain injury is an important cause of morbidity and long-standing disability in newborns. We have previously shown that human plasma-derived inter-alpha inhibitor proteins (hIAIPs) attenuate HI-related brain injury in neonatal rats. The optimal dose of hIAIPs for their neuroprotective effects and improvement in behavioral outcomes remains to be determined. We examined the efficacy of 30, 60, or 90 mg/kg of hIAIPs administered to neonatal rats after exposure to HI for 2 h. Postnatal day 7 (P7) Wistar rats were exposed to either sham-surgery or unilateral HI (right carotid artery ligation, 2 h of 8% O2) brain injury. A placebo, 30, 60, or 90 mg/kg of hIAIPs were injected intraperitoneally at 0, 24 and 48 h after HI (n = 9–10/sex). We carried out the following behavioral analyses: P8 (righting reflex), P9 (negative geotaxis) and P10 (open-field task). Rats were humanely killed on P10 and their brains were stained with cresyl violet. Male extension/contraction responses and female righting reflex times were higher in the HI placebo groups than the sham groups. Female open-field exploration was lower in the HI placebo group than the sham group. hIAIPs attenuated these behavioral deficits. However, the magnitude of the responses did not vary by hIAIP dose. hIAIPs reduced male brain infarct volumes in a manner that correlated with improved behavioral outcomes. Increasing the hIAIP dose from 30 to 90 mg/kg did not further accentuate the hIAIP-related decreases in infarct volumes. We conclude that larger doses of hIAIPs did not provide additional benefits over the 30 mg/kg dose for behavior tasks or reductions in infarct volumes in neonatal rats after exposure to severe HI. Full article
(This article belongs to the Section Molecular Neurobiology)
Show Figures

Figure 1

22 pages, 3820 KiB  
Article
Dendrimer-Conjugated nSMase2 Inhibitor Reduces Tau Propagation in Mice
by Carolyn Tallon, Benjamin J. Bell, Anjali Sharma, Arindom Pal, Medhinee M. Malvankar, Ajit G. Thomas, Seung-Wan Yoo, Kristen R. Hollinger, Kaleem Coleman, Elizabeth L. Wilkinson, Sujatha Kannan, Norman J. Haughey, Rangaramanujam M. Kannan, Rana Rais and Barbara S. Slusher
Pharmaceutics 2022, 14(10), 2066; https://doi.org/10.3390/pharmaceutics14102066 - 28 Sep 2022
Cited by 14 | Viewed by 3367
Abstract
Alzheimer’s disease (AD) is characterized by the progressive accumulation of amyloid-β and hyperphosphorylated tau (pTau), which can spread throughout the brain via extracellular vesicles (EVs). Membrane ceramide enrichment regulated by the enzyme neutral sphingomyelinase 2 (nSMase2) is a critical component of at least [...] Read more.
Alzheimer’s disease (AD) is characterized by the progressive accumulation of amyloid-β and hyperphosphorylated tau (pTau), which can spread throughout the brain via extracellular vesicles (EVs). Membrane ceramide enrichment regulated by the enzyme neutral sphingomyelinase 2 (nSMase2) is a critical component of at least one EV biogenesis pathway. Our group recently identified 2,6-Dimethoxy-4-(5-Phenyl-4-Thiophen-2-yl-1H-Imidazol-2-yl)-Phenol (DPTIP), the most potent (30 nM) and selective inhibitor of nSMase2 reported to date. However, DPTIP exhibits poor oral pharmacokinetics (PK), modest brain penetration, and rapid clearance, limiting its clinical translation. To enhance its PK properties, we conjugated DPTIP to a hydroxyl-PAMAM dendrimer delivery system, creating dendrimer-DPTIP (D-DPTIP). In an acute brain injury model, orally administered D-DPTIP significantly reduced the intra-striatal IL-1β-induced increase in plasma EVs up to 72 h post-dose, while oral DPTIP had a limited effect. In a mouse tau propagation model, where a mutant hTau (P301L/S320F) containing adeno-associated virus was unilaterally seeded into the hippocampus, oral D-DPTIP (dosed 3× weekly) significantly inhibited brain nSMase2 activity and blocked the spread of pTau to the contralateral hippocampus. These data demonstrate that dendrimer conjugation of DPTIP improves its PK properties, resulting in significant inhibition of EV propagation of pTau in mice. Dendrimer-based delivery of DPTIP has the potential to be an exciting new therapeutic for AD. Full article
(This article belongs to the Special Issue Recent Advances in Dendrimer Nanomedicine)
Show Figures

Figure 1

10 pages, 813 KiB  
Brief Report
Unilateral Cervical Vagotomy Modulates Immune Cell Profiles and the Response to a Traumatic Brain Injury
by M. Karen Newell-Rogers, Amanda Duong, Rizwan Nazarali, Richard P. Tobin, Susannah K. Rogers and Lee A. Shapiro
Int. J. Mol. Sci. 2022, 23(17), 9851; https://doi.org/10.3390/ijms23179851 - 30 Aug 2022
Cited by 7 | Viewed by 2945
Abstract
TBI induces splenic B and T cell expansion that contributes to neuroinflammation and neurodegeneration. The vagus nerve, the longest of the cranial nerves, is the predominant parasympathetic pathway allowing the central nervous system (CNS) control over peripheral organs, including regulation of inflammatory responses. [...] Read more.
TBI induces splenic B and T cell expansion that contributes to neuroinflammation and neurodegeneration. The vagus nerve, the longest of the cranial nerves, is the predominant parasympathetic pathway allowing the central nervous system (CNS) control over peripheral organs, including regulation of inflammatory responses. One way this is accomplished is by vagus innervation of the celiac ganglion, from which the splenic nerve innervates the spleen. This splenic innervation enables modulation of the splenic immune response, including splenocyte selection, activation, and downstream signaling. Considering that the left and right vagus nerves have distinct courses, it is possible that they differentially influence the splenic immune response following a CNS injury. To test this possibility, immune cell subsets were profiled and quantified following either a left or a right unilateral vagotomy. Both unilateral vagotomies caused similar effects with respect to the percentage of B cells and in the decreased percentage of macrophages and T cells following vagotomy. We next tested the hypothesis that a left unilateral vagotomy would modulate the splenic immune response to a traumatic brain injury (TBI). Mice received a left cervical vagotomy or a sham vagotomy 3 days prior to a fluid percussion injury (FPI), a well-characterized mouse model of TBI that consistently elicits an immune and neuroimmune response. Flow cytometric analysis showed that vagotomy prior to FPI resulted in fewer CLIP+ B cells, and CD4+, CD25+, and CD8+ T cells. Vagotomy followed by FPI also resulted in an altered distribution of CD11bhigh and CD11blow macrophages. Thus, transduction of immune signals from the CNS to the periphery via the vagus nerve can be targeted to modulate the immune response following TBI. Full article
Show Figures

Figure 1

17 pages, 4461 KiB  
Article
Impact of Hypoxia-Ischemia on Neurogenesis and Structural and Functional Outcomes in a Mild–Moderate Neonatal Hypoxia-Ischemia Brain Injury Model
by Anne Ehlting, Margit Zweyer, Elke Maes, Yvonne Schleehuber, Hardik Doshi, Hemmen Sabir and Maria Eugenia Bernis
Life 2022, 12(8), 1164; https://doi.org/10.3390/life12081164 - 30 Jul 2022
Cited by 19 | Viewed by 3835
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a common type of brain injury caused by a lack of oxygen and blood flow to the brain during the perinatal period. The incidence of HIE is approximately 2–3 cases per 1000 live births in high-income settings; while in [...] Read more.
Hypoxic-ischemic encephalopathy (HIE) is a common type of brain injury caused by a lack of oxygen and blood flow to the brain during the perinatal period. The incidence of HIE is approximately 2–3 cases per 1000 live births in high-income settings; while in low- and middle-income countries, the incidence is 3–10-fold higher. Therapeutic hypothermia (TH) is the current standard treatment for neonates affected by moderate–severe HIE. However, more than 50% of all infants with suspected HIE have mild encephalopathy, and these infants are not treated with TH because of their lower risk of adverse outcomes. Despite this, several analyses of pooled data provide increasing evidence that infants who initially have mild encephalopathy may present signs of more significant brain injury later in life. The purpose of this study was to expand our knowledge about the effect of mild–moderate hypoxia-ischemia (HI) at the cellular, structural, and functional levels. An established rat model of mild–moderate HI was used, where postnatal day (P) 7 rats were exposed to unilateral permanent occlusion of the left carotid artery and 90 min of 8% hypoxia, followed by TH or normothermia (NT) treatment. The extent of injury was assessed using histology (P14 and P42) and MRI (P11 and P32), as well as with short-term and long-term behavioral tests. Neurogenesis was assessed by BrdU staining. We showed that mild–moderate HI leads to a progressive loss of brain tissue, pathological changes in MRI scans, as well as an impairment of long-term motor function. At P14, the median area loss assessed by histology for HI animals was 20% (p < 0.05), corresponding to mild–moderate brain injury, increasing to 55% (p < 0.05) at P42. The data assessed by MRI corroborated our results. HI led to a decrease in neurogenesis, especially in the hippocampus and the lateral ventricle at early time points, with a delayed partial recovery. TH was not neuroprotective at early time points following mild–moderate HI, but prevented the increase in brain damage over time. Additionally, rats treated with TH showed better long-term motor function. Altogether, our results bring more light to the understanding of pathophysiology following mild-moderate HI. We showed that, in the context of mild-moderate HI, TH failed to be significantly neuroprotective. However, animals treated with TH showed a significant improvement in motor, but not cognitive long-term function. These results are in line with what is observed in some cases where neonates with mild HIE are at risk of neurodevelopmental deficits in infancy or childhood. Whether TH should be used as a preventive treatment to reduce adverse outcomes in mild-HIE remains of active interest, and more research has to be carried out in order to address this question. Full article
Show Figures

Figure 1

23 pages, 5802 KiB  
Article
Analysis of Givinostat/ITF2357 Treatment in a Rat Model of Neonatal Hypoxic-Ischemic Brain Damage
by Paulina Pawelec, Joanna Sypecka, Teresa Zalewska and Malgorzata Ziemka-Nalecz
Int. J. Mol. Sci. 2022, 23(15), 8287; https://doi.org/10.3390/ijms23158287 - 27 Jul 2022
Cited by 3 | Viewed by 2913
Abstract
The histone deacetylase inhibitor (HDACi) Givinostat/ITF2357 provides neuroprotection in adult models of brain injury; however, its action after neonatal hypoxia-ischemia (HI) is still undefined. The aim of our study was to test the hypothesis that the mechanism of Givinostat is associated with the [...] Read more.
The histone deacetylase inhibitor (HDACi) Givinostat/ITF2357 provides neuroprotection in adult models of brain injury; however, its action after neonatal hypoxia-ischemia (HI) is still undefined. The aim of our study was to test the hypothesis that the mechanism of Givinostat is associated with the alleviation of inflammation. For this purpose, we analyzed the microglial response and the effect on molecular mediators (chemokines/cytokines) that are crucial for inducing cerebral damage after neonatal hypoxia-ischemia. Seven-day-old rat pups were subjected to unilateral carotid artery ligation followed by 60 min of hypoxia (7.6% O2). Givinostat (10 mg/kg b/w) was administered in a 5-day regimen. The effects of Givinostat on HI-induced inflammation (cytokine, chemokine and microglial activation and polarization) were assessed with a Luminex assay, immunohistochemistry and Western blot. Givinostat treatment did not modulate the microglial response specific for HI injury. After Givinostat administration, the investigated chemokines and cytokines remained at the level induced by HI. The only immunosuppressive effect of Givinostat may be associated with the decrease in MIP-1α. Neonatal hypoxia-ischemia produces an inflammatory response by activating the proinflammatory M1 phenotype of microglia, disrupting the microglia–neuron (CX3CL1/CX3CR1) axis and elevating numerous proinflammatory cytokines/chemokines. Givinostat/ITF2357 did not prevent an inflammatory reaction after HI. Full article
Show Figures

Figure 1

Back to TopTop