Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 10.397
1.
Toxins (Basel) ; 16(4)2024 Apr 10.
Article En | MEDLINE | ID: mdl-38668612

Accidents caused by Bothrops jararaca (Bj) snakes result in several local and systemic manifestations, with pain being a fundamental characteristic. The inflammatory process responsible for hyperalgesia induced by Bj venom (Bjv) has been studied; however, the specific roles played by the peripheral and central nervous systems in this phenomenon remain unclear. To clarify this, we induced hyperalgesia in rats using Bjv and collected tissues from dorsal root ganglia (DRGs) and spinal cord (SC) at 2 and 4 h post-induction. Samples were labeled for Iba-1 (macrophage and microglia), GFAP (satellite cells and astrocytes), EGR1 (neurons), and NK1 receptors. Additionally, we investigated the impact of minocycline, an inhibitor of microglia, and GR82334 antagonist on Bjv-induced hyperalgesia. Our findings reveal an increase in Iba1 in DRG at 2 h and EGR1 at 4 h. In the SC, markers for microglia, astrocytes, neurons, and NK1 receptors exhibited increased expression after 2 h, with EGR1 continuing to rise at 4 h. Minocycline and GR82334 inhibited venom-induced hyperalgesia, highlighting the crucial roles of microglia and NK1 receptors in this phenomenon. Our results suggest that the hyperalgesic effects of Bjv involve the participation of microglial and astrocytic cells, in addition to the activation of NK1 receptors.


Bothrops , Crotalid Venoms , Ganglia, Spinal , Hyperalgesia , Receptors, Neurokinin-1 , Animals , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Crotalid Venoms/toxicity , Male , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Receptors, Neurokinin-1/metabolism , Minocycline/pharmacology , Spinal Cord/drug effects , Spinal Cord/metabolism , Early Growth Response Protein 1/metabolism , Early Growth Response Protein 1/genetics , Microglia/drug effects , Microglia/metabolism , Neuroglia/drug effects , Neuroglia/metabolism , Rats , Glial Fibrillary Acidic Protein/metabolism , Calcium-Binding Proteins/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Microfilament Proteins/metabolism , Neurokinin-1 Receptor Antagonists/pharmacology , Rats, Sprague-Dawley
2.
Nat Commun ; 15(1): 2866, 2024 Apr 03.
Article En | MEDLINE | ID: mdl-38570482

Traumatic brain injury leads to a highly orchestrated immune- and glial cell response partially responsible for long-lasting disability and the development of secondary neurodegenerative diseases. A holistic understanding of the mechanisms controlling the responses of specific cell types and their crosstalk is required to develop an efficient strategy for better regeneration. Here, we combine spatial and single-cell transcriptomics to chart the transcriptomic signature of the injured male murine cerebral cortex, and identify specific states of different glial cells contributing to this signature. Interestingly, distinct glial cells share a large fraction of injury-regulated genes, including inflammatory programs downstream of the innate immune-associated pathways Cxcr3 and Tlr1/2. Systemic manipulation of these pathways decreases the reactivity state of glial cells associated with poor regeneration. The functional relevance of the discovered shared signature of glial cells highlights the importance of our resource enabling comprehensive analysis of early events after brain injury.


Brain Injuries , Wounds, Stab , Animals , Mice , Male , Glial Fibrillary Acidic Protein/metabolism , Neuroglia/metabolism , Brain Injuries/metabolism , Cerebral Cortex/metabolism , Wounds, Stab/complications , Wounds, Stab/metabolism
3.
Zhongguo Dang Dai Er Ke Za Zhi ; 26(4): 394-402, 2024 Apr 15.
Article Zh | MEDLINE | ID: mdl-38660904

OBJECTIVES: To compare the repair effects of different doses of human umbilical cord mesenchymal stem cells (hUC-MSCs) on white matter injury (WMI) in neonatal rats. METHODS: Two-day-old Sprague-Dawley neonatal rats were randomly divided into five groups: sham operation group, WMI group, and hUC-MSCs groups (low dose, medium dose, and high dose), with 24 rats in each group. Twenty-four hours after successful establishment of the neonatal rat white matter injury model, the WMI group was injected with sterile PBS via the lateral ventricle, while the hUC-MSCs groups received injections of hUC-MSCs at different doses. At 14 and 21 days post-modeling, hematoxylin and eosin staining was used to observe pathological changes in the tissues around the lateral ventricles. Real-time quantitative polymerase chain reaction was used to detect the quantitative expression of myelin basic protein (MBP) and glial fibrillary acidic protein (GFAP) mRNA in the brain tissue. Immunohistochemistry was employed to observe the expression levels of GFAP and neuron-specific nuclear protein (NeuN) in the tissues around the lateral ventricles. TUNEL staining was used to observe cell apoptosis in the tissues around the lateral ventricles. At 21 days post-modeling, the Morris water maze test was used to observe the spatial learning and memory capabilities of the neonatal rats. RESULTS: At 14 and 21 days post-modeling, numerous cells with nuclear shrinkage and rupture, as well as disordered arrangement of nerve fibers, were observed in the tissues around the lateral ventricles of the WMI group and the low dose group. Compared with the WMI group, the medium and high dose groups showed alleviated pathological changes; the arrangement of nerve fibers in the medium dose group was relatively more orderly compared with the high dose group. Compared with the WMI group, there was no significant difference in the expression levels of MBP and GFAP mRNA in the low dose group (P>0.05), while the expression levels of MBP mRNA increased and GFAP mRNA decreased in the medium and high dose groups. The expression level of MBP mRNA in the medium dose group was higher than that in the high dose group, and the expression level of GFAP mRNA in the medium dose group was lower than that in the high dose group (P<0.05). Compared with the WMI group, there was no significant difference in the protein expression of GFAP and NeuN in the low dose group (P>0.05), while the expression of NeuN protein increased and GFAP protein decreased in the medium and high dose groups. The expression of NeuN protein in the medium dose group was higher than that in the high dose group, and the expression of GFAP protein in the medium dose group was lower than that in the high dose group (P<0.05). Compared with the WMI group, there was no significant difference in the number of apoptotic cells in the low dose group (P>0.05), while the number of apoptotic cells in the medium and high dose groups was less than that in the WMI group, and the number of apoptotic cells in the medium dose group was less than that in the high dose group (P<0.05). Compared with the WMI group, there was no significant difference in the escape latency time in the low dose group (P>0.05); starting from the third day of the latency period, the escape latency time in the medium dose group was less than that in the WMI group (P<0.05). The medium and high dose groups crossed the platform more times than the WMI group (P<0.05). CONCLUSIONS: Low dose hUC-MSCs may yield unsatisfactory repair effects on WMI in neonatal rats, while medium and high doses of hUC-MSCs have significant repair effects, with the medium dose demonstrating superior efficacy.


Animals, Newborn , Mesenchymal Stem Cell Transplantation , Rats, Sprague-Dawley , Umbilical Cord , White Matter , Animals , Rats , Humans , Umbilical Cord/cytology , White Matter/pathology , White Matter/metabolism , Glial Fibrillary Acidic Protein/metabolism , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/analysis , Mesenchymal Stem Cells , Myelin Basic Protein/genetics , Myelin Basic Protein/analysis , Myelin Basic Protein/metabolism , Male , Apoptosis , Female , RNA, Messenger/analysis , RNA, Messenger/metabolism
4.
J Neuropathol Exp Neurol ; 83(5): 345-356, 2024 Apr 19.
Article En | MEDLINE | ID: mdl-38441171

Identification of central nervous system injury is a critical part of perinatal autopsies; however, injury is not always easily identifiable due to autolysis and immaturity of the developing brain. Here, the role of immunohistochemical stains in the identification of perinatal brain injury was investigated. Blinded semiquantitative scoring of injury was performed on sections of frontal lobe from 76 cases (51 liveborn and 25 stillborn) using H&E, GFAP, Iba-1, and ß-APP stains. Digital image analysis was used to quantify GFAP and Iba-1 staining. Commonly observed pathologies included diffuse white matter gliosis (DWMG) and white matter necrosis (WMN). DWMG scores were very similar on H&E and GFAP stains for liveborn subjects. For stillborn subjects, DWMG scores were significantly higher on GFAP stain than H&E. ß-APP was needed for identification of WMN in 71.4% of stillborn subjects compared to 15.4% of liveborn subjects. Diffuse staining for Iba-1 within cortex and white matter was positively correlated with subject age. Staining quantification on digital image analysis was highly correlated to semiquantitative scoring. Overall, GFAP and ß-APP stains were most helpful in identifying white matter injury not seen on H&E in stillborn subjects. Immunostains may therefore be warranted as an integral part of stillborn brain autopsies.


Brain Injuries , White Matter , Pregnancy , Female , Humans , Brain/pathology , Brain Injuries/pathology , White Matter/pathology , Frontal Lobe/metabolism , Cerebral Cortex/pathology , Glial Fibrillary Acidic Protein/metabolism
5.
Cytokine ; 177: 156565, 2024 05.
Article En | MEDLINE | ID: mdl-38442443

BACKGROUND: Perinatal hypoxia triggers the release of cytokines and chemokines by neurons, astrocytes and microglia. In response to hypoxia-ischemia resting/ramified microglia proliferate and undergo activation, producing proinflammatory molecules. The brain damage extension seems to be related to both the severity of hypoxia and the balance between pro and anti-inflammatory response and can be explored with neuroimaging. AIMS: The aim of this preliminary study was to explore possible relationships between plasma levels of inflammatory cytokines/chemokines and the severe brain damage detectable by Magnetic Resonance Imaging (MRI), performed during the hospitalization. METHODS: In 10 full terms neonates with hypoxic ischemic encephalopathy (HIE) undergoing therapeutic hypothermia (TH), divided into cases and controls, according to MRI results, we measured and compared the plasma levels of CCL2/MCP-1, CXCL8, GFAP, IFN y, IL-10, IL-18, IL-6, CCL3, ENOLASE2, GM-CSF, IL-1b, IL-12p70, IL-33, TNFα, collected at four different time points during TH (24, 25-48, 49-72 h of life, and 7-10 days from birth). Five of enrolled babies had pathological brain MRI (cases) and 5 had a normal MRI examination (controls). Cytokines were measured by Magnetic Luminex Assay. MRI images were classified according to Barkovich's score. RESULTS: Mean levels of all cytokines and molecules at time T1 were not significantly different in the two groups. Comparing samples paired by day of collection, the greatest differences between cases and controls were found at times T2 and T3, during TH. At T4, levels tended to get closer again (except for IL-6, IL10 and IL18). Infants with worse MRI showed higher plasmatic GFAP levels than those with normal MRI, while their IL-18 was lower. The mean levels of CCL3MIP1alpha, GMCSF, IL1BETA overlapped throughout the observation period in both groups. CONCLUSION: In a small number of infants with worse brain MRI, we found higher levels of GFAP and of IL-10 at T4 and a trend toward low IL-18 levels than in infants with normal MRI, considered early biomarker of brain damage and a predictor of adverse outcome, respectively. The greatest, although not significant, difference between the levels of molecules was found in cases and controls at time points T2 and T3, during TH.


Brain Injuries , Hypoxia-Ischemia, Brain , Infant, Newborn , Infant , Female , Pregnancy , Humans , Hypoxia-Ischemia, Brain/diagnostic imaging , Cytokines/metabolism , Interleukin-10/metabolism , Interleukin-18/metabolism , Glial Fibrillary Acidic Protein/metabolism , Interleukin-6/metabolism , Brain/metabolism , Magnetic Resonance Imaging/methods , Chemokines/metabolism , Neuroimaging
6.
Cells ; 13(6)2024 Mar 13.
Article En | MEDLINE | ID: mdl-38534343

The role of RNA Binding Motif Protein 8a (RBM8A), an exon junction complex (EJC) component, in neurodevelopmental disorders has been increasingly studied for its crucial role in regulating multiple levels of gene expression. It regulates mRNA splicing, translation, and mRNA degradation and influences embryonic development. RBM8A protein is expressed in both neurons and astrocytes, but little is known about RBM8A's specific role in glial fibrillary acid protein (GFAP)-positive astrocytes. To address the role of RBM8A in astrocytes, we generated a conditional heterozygous knockout (KO) mouse line of Rbm8a in astrocytes using a GFAP-cre line. We confirmed a decreased expression of RBM8A in astrocytes of heterozygous conditional KO mice via RT-PCR and Sanger sequencing, as well as qRT-PCR, immunohistochemistry, and Western blot. Interestingly, these mice exhibit significantly increased movement and mobility, alongside sex-specific altered anxiety in the open field test (OFT) and elevated plus maze (OPM) tests. These tests, along with the rotarod test, suggest that these mice have normal motor coordination but hyperactive phenotypes. In addition, the haploinsufficiency of Rbm8a in astrocytes leads to a sex-specific change in astrocyte density in the dentate gyrus. This study further reveals the contribution of Rbm8a deletion to CNS pathology, generating more insights via the glial lens of an Rbm8a model of neurodevelopmental disorder.


Astrocytes , Neurons , Male , Female , Mice , Animals , Astrocytes/metabolism , Glial Fibrillary Acidic Protein/metabolism , Neurons/metabolism , Mice, Knockout , Exons , Locomotion , RNA-Binding Proteins/metabolism
7.
Sci Rep ; 14(1): 6429, 2024 03 18.
Article En | MEDLINE | ID: mdl-38499607

Given the huge impact of the COVID-19 pandemic, it appears of paramount importance to assess the cognitive effects on the population returning to work after COVID-19 resolution. Serum levels of neurofilament light chain (sNfL) and glial fibrillary acidic protein (sGFAP) represent promising biomarkers of neuro-axonal damage and astrocytic activation. In this cohort study, we explored the association between sNfL and sGFAP concentrations and cognitive performance in a group of 147 adult workers with a previous asymptomatic SARS-CoV-2 infection or mild COVID-19, one week and, in 49 of them, ten months after SARS-Cov2 negativization and compared them to a group of 82 age and BMI-matched healthy controls (HCs). sNfL and sGFAP concentrations were assessed using SimoaTM assay Neurology 2-Plex B Kit. COVID-19 patients were interviewed one-on-one by trained physicians and had to complete a list of questionnaires, including the Cognitive Failure Questionnaire (CFQ). At the first assessment (T0), sNfL and sGFAP levels were significantly higher in COVID-19 patients than in HCs (p < 0.001 for both). The eleven COVID-19 patients with cognitive impairment had significantly higher levels of sNfL and sGFAP than the others (p = 0.005 for both). At the subsequent follow-up (T1), sNfL and sGFAP levels showed a significant decrease (median sNfL 18.3 pg/mL; median sGFAP 77.2 pg/mL), although they were still higher than HCs (median sNfL 7.2 pg/mL, median sGFAP 63.5 pg/mL). Our results suggest an ongoing damage involving neurons and astrocytes after SARS-Cov2 negativization, which reduce after ten months even if still evident compared to HCs.


COVID-19 , Multiple Sclerosis , Adult , Humans , Biomarkers , Cohort Studies , COVID-19/metabolism , Glial Fibrillary Acidic Protein/metabolism , Intermediate Filaments/metabolism , Multiple Sclerosis/metabolism , Neurofilament Proteins , Pandemics , RNA, Viral/metabolism , SARS-CoV-2
8.
Curr Opin Cell Biol ; 87: 102340, 2024 Apr.
Article En | MEDLINE | ID: mdl-38401182

Glial fibrillary acidic protein (GFAP) is an intermediate filament (IF) protein expressed in specific types of glial cells in the nervous system. The expression of GFAP is highly regulated during brain development and in neurological diseases. The presence of distinct GFAP-isoforms in various cell types, developmental stages, and diseases indicates that GFAP (post-)transcriptional regulation has a role in glial cell physiology and pathology. GFAP-isoforms differ in sub-cellular localisation, IF-network assembly properties, and IF-dynamics which results in distinct molecular interactions and mechanical properties of the IF-network. Therefore, GFAP (post-)transcriptional regulation is likely a mechanism by which radial glia, astrocytes, and glioma cells can modulate cellular function.


Astrocytes , Intermediate Filaments , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Intermediate Filaments/metabolism , Astrocytes/metabolism , Astrocytes/pathology , Protein Isoforms/genetics , Gene Expression Regulation
9.
Brain Struct Funct ; 229(3): 695-703, 2024 Apr.
Article En | MEDLINE | ID: mdl-38308043

The pathophysiology of Fronto Temporal Dementia (FTD) remains poorly understood, specifically the role of astroglia. Our aim was to explore the hypothesis of astrocytic alterations as a component for FTD pathophysiology. We performed an in-depth tri-dimensional (3-D) anatomical and morphometric study of glial fibrillary acidic protein (GFAP)-positive and glutamine synthetase (GS)-positive astrocytes in the human entorhinal cortex (EC) of FTD patients. The studies at this level in the different types of human dementia are scarce. We observed a prominent astrocyte atrophy of GFAP-positive astrocytes and co-expressing GFAP/GS astrocytes, characterised by a decrease in area and volume, whilst minor changes in GS-positive astrocytes in FTD compared to non-dementia controls (ND) samples. This study evidences the importance of astrocyte atrophy and dysfunction in human EC. We hypothesise that FTD is not only a neuropathological disease, but also a gliopathological disease having a major relevance in the understanding the astrocyte role in FTD pathological processes and development.


Entorhinal Cortex , Frontotemporal Dementia , Humans , Entorhinal Cortex/pathology , Astrocytes/metabolism , Frontotemporal Dementia/pathology , Atrophy/pathology , Glial Fibrillary Acidic Protein/metabolism
10.
Cell Biochem Funct ; 42(2): e3958, 2024 Mar.
Article En | MEDLINE | ID: mdl-38396357

Iron accumulation in the brain causes oxidative stress, blood-brain barrier (BBB) breakdown, and neurodegeneration. We examined the preventive effects of acetylated oligopeptides (AOP) from whey protein on iron-induced hippocampal damage compared to N-acetyl cysteine (NAC). This 5-week study used 40 male albino rats. At the start, all rats received 150 mg/kg/day of oral NAC for a week. The 40 animals were then randomly divided into four groups: Group I (control) received a normal diet; Group II (iron overload) received 60 mg/kg/day intraperitoneal iron dextran 5 days a week for 4 weeks; Group III (NAC group) received 150 mg/kg/day NAC and iron dextran; and Group IV (AOP group) received 150 mg/kg/day AOP and iron dextran. Enzyme-linked immunosorbent assay, spectrophotometry, and qRT-PCR were used to measure MMP-9, tissue inhibitor metalloproteinase-1 (TIMP-1), MDA, reduced glutathione (GSH) levels, and nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) gene expression. Histopathological and immunohistochemical detection of nestin, claudin, caspase, and GFAP was also done. MMP-9, TIMP-1, MDA, caspase, and GFAP rose in the iron overload group, while GSH, Nrf2, HO-1, nestin, and claudin decreased. The NAC and AOP administrations improved iron overload-induced biochemical and histological alterations. We found that AOP and NAC can protect the brain hippocampus from iron overload, improve BBB disruption, and provide neuroprotection with mostly no significant difference from healthy controls.


Acetylcysteine , Iron Overload , Oligopeptides , Animals , Male , Rats , Acetylcysteine/pharmacology , Acetylcysteine/metabolism , Caspases/metabolism , Claudins/genetics , Dentate Gyrus/metabolism , Dentate Gyrus/pathology , Dextrans/metabolism , Dextrans/pharmacology , Down-Regulation , Glutathione/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Iron/metabolism , Iron/pharmacology , Iron Overload/complications , Iron Overload/drug therapy , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/pharmacology , Nestin/genetics , Nestin/metabolism , Nestin/pharmacology , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-1/pharmacology , Up-Regulation , Oligopeptides/pharmacology , Heme Oxygenase-1/drug effects , Glial Fibrillary Acidic Protein/drug effects , Glial Fibrillary Acidic Protein/metabolism
11.
Neurosci Lett ; 824: 137674, 2024 Feb 28.
Article En | MEDLINE | ID: mdl-38355005

Adult neural stem cells (NSCs) located in the two canonical neurogenic niches, the subventricular zone (SVZ) and the subgranular zone (SGZ), express the glial fibrillary acidic protein (GFAP). Recently, proliferative activity has been described in the hypothalamus although the characterization of hypothalamic neural stem/progenitor cells (NSPCs) is still uncertain. We therefore investigated whether hypothalamic GFAP-positive cells, as in the SVZ and SGZ, also have neurogenic potential. We used a transgenic mouse line expressing green fluorescent protein (GFP) under the control of the GFAP promoter. GFAP-GFP expressing cells are localized in the ependymal layer as well as in the parenchyma of the mediobasal hypothalamus (MBH) and express Sox2, a marker for NSCs. Interestingly, no sexual dimorphism was observed in the numbers of GFP + and GFP-Sox2 + cells. After cells sorting, these cells were able to generate neurospheres in vitro and give rise to neurons, astrocytes and oligodendrocytes. Taken together, these results show that hypothalamic GFAP-expressing cells form a population of NSPCs.


Neural Stem Cells , Mice , Animals , Cell Lineage , Glial Fibrillary Acidic Protein/metabolism , Cell Differentiation/physiology , Neural Stem Cells/metabolism , Mice, Transgenic , Hypothalamus/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism
12.
Acta Neuropathol ; 147(1): 31, 2024 Feb 03.
Article En | MEDLINE | ID: mdl-38310187

Anti-glial fibrillary acidic protein (GFAP) meningoencephalomyelitis (autoimmune GFAP astrocytopathy) is a new autoimmune central nervous system (CNS) disease diagnosable by the presence of anti-GFAP autoantibodies in the cerebrospinal fluid and presents as meningoencephalomyelitis in the majority of patients. Only few neuropathological reports are available and little is known about the pathogenic mechanisms. We performed a histopathological study of two autopsies and nine CNS biopsies of patients with anti-GFAP autoantibodies and found predominantly a lymphocytic and in one autopsy case a granulomatous inflammatory phenotype. Inflammatory infiltrates were composed of B and T cells, including tissue-resident memory T cells. Although obvious astrocytic damage was absent in the GFAP-staining, we found cytotoxic T cell-mediated reactions reflected by the presence of CD8+/perforin+/granzyme A/B+ cells, polarized towards astrocytes. MHC-class-I was upregulated in reactive astrocytes of all biopsies and two autopsies but not in healthy controls. Importantly, we observed a prominent immunoreactivity of astrocytes with the complement factor C4d. Finally, we provided insight into an early phase of GFAP autoimmunity in an autopsy of a pug dog encephalitis that was characterized by marked meningoencephalitis with selective astrocytic damage with loss of GFAP and AQP4 in the lesions.Our histopathological findings indicate that a cytotoxic T cell-mediated immune reaction is present in GFAP autoimmunity. Complement C4d deposition on astrocytes could either represent the cause or consequence of astrocytic reactivity. Selective astrocytic damage is prominent in the early phase of GFAP autoimmunity in a canine autopsy case, but mild or absent in subacute and chronic stages in human disease, probably due to the high regeneration potential of astrocytes. The lymphocytic and granulomatous phenotypes might reflect different stages of lesion development or patient-specific modifications of the immune response. Future studies will be necessary to investigate possible implications of pathological subtypes for clinical disease course and therapeutic strategies.


Autoimmune Diseases of the Nervous System , Encephalomyelitis , Meningoencephalitis , Humans , Animals , Dogs , Glial Fibrillary Acidic Protein/metabolism , Encephalomyelitis/pathology , Astrocytes/pathology , Autoimmune Diseases of the Nervous System/cerebrospinal fluid , Autoimmune Diseases of the Nervous System/therapy , Meningoencephalitis/cerebrospinal fluid , Meningoencephalitis/pathology , Autoantibodies
13.
J Neurovirol ; 30(1): 1-21, 2024 Feb.
Article En | MEDLINE | ID: mdl-38280928

Opioid overdose deaths have dramatically increased by 781% from 1999 to 2021. In the setting of HIV, opioid drug abuse exacerbates neurotoxic effects of HIV in the brain, as opioids enhance viral replication, promote neuronal dysfunction and injury, and dysregulate an already compromised inflammatory response. Despite the rise in fentanyl abuse and the close association between opioid abuse and HIV infection, the interactive comorbidity between fentanyl abuse and HIV has yet to be examined in vivo. The HIV-1 Tat-transgenic mouse model was used to understand the interactive effects between fentanyl and HIV. Tat is an essential protein produced during HIV that drives the transcription of new virions and exerts neurotoxic effects within the brain. The Tat-transgenic mouse model uses a glial fibrillary acidic protein (GFAP)-driven tetracycline promoter which limits Tat production to the brain and this model is well used for examining mechanisms related to neuroHIV. After 7 days of fentanyl exposure, brains were harvested. Tight junction proteins, the vascular cell adhesion molecule, and platelet-derived growth factor receptor-ß were measured to examine the integrity of the blood brain barrier. The immune response was assessed using a mouse-specific multiplex chemokine assay. For the first time in vivo, we demonstrate that fentanyl by itself can severely disrupt the blood-brain barrier and dysregulate the immune response. In addition, we reveal associations between inflammatory markers and tight junction proteins at the blood-brain barrier.


Blood-Brain Barrier , Fentanyl , HIV-1 , Mice, Transgenic , Neuroinflammatory Diseases , tat Gene Products, Human Immunodeficiency Virus , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Blood-Brain Barrier/virology , Mice , Fentanyl/pharmacology , HIV-1/drug effects , HIV-1/genetics , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism , Neuroinflammatory Diseases/genetics , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/virology , HIV Infections/virology , HIV Infections/genetics , HIV Infections/pathology , HIV Infections/drug therapy , Disease Models, Animal , Analgesics, Opioid/pharmacology , Analgesics, Opioid/adverse effects , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Tight Junction Proteins/metabolism , Tight Junction Proteins/genetics , Humans , Brain/drug effects , Brain/virology , Brain/metabolism , Brain/pathology , Opioid-Related Disorders/genetics , Opioid-Related Disorders/pathology , Opioid-Related Disorders/metabolism
14.
Exp Neurol ; 374: 114700, 2024 Apr.
Article En | MEDLINE | ID: mdl-38272160

Wound healing after closed-head injury is a significant medical issue. However, conventional models of focal traumatic brain injury, such as fluid percussion injury and controlled cortical impact, employ mechanical impacts on the exposed cerebral cortex after craniotomy. These animal models are inappropriate for studying gliosis, as craniotomy itself induces gliosis. To address this, we developed a closed-head injury model and named "photo injury", which employs intense light illumination through a thinned-skull cranial window. Our prior work demonstrated that the gliosis of focal cerebral lesion after the photo injury does not encompass artificial gliosis and comprises two distinct reactive astrocyte subpopulations. The reactive astrocytes accumulated in the perilesional recovery area actively proliferate and express Nestin, a neural stem cell marker, while those in distal regions do not exhibit these traits. The present study investigated the role of perilesional reactive astrocytes (PRAs) in wound healing using the ablation of reactive astrocytes by the conditional knockout of Stat3. The extensive and non-selective ablation of reactive astrocytes in Nestin-Cre:Stat3f/f mice resulted in an exacerbation of injury, marked by increased inflammation and BBB disruption. On the other hand, GFAP-CreERT2:Stat3f/f mice exhibited the partial and selective ablation of the PRAs, while their exacerbation of injury was at the same extent as in Nestin-Cre:Stat3f/f mice. The comparison of these two mouse strains indicates that the PRAs are an essential astrocyte component for wound healing after closed-head injury, and their anti-inflammatory and regenerative functions are significantly affected even by incomplete accumulation. In addition, the reporter gene expression in the PRAs by GFAP-CreERT2 indicated a substantial elimination of these cells and an absence of differentiation into other cell types, despite Nestin expression, after wound healing. Thus, the accumulation and subsequent elimination of PRA are proposed as promising diagnostic and therapeutic avenues to bolster wound healing after closed-head injury.


Brain Injuries , Head Injuries, Closed , Mice , Animals , Astrocytes/metabolism , Nestin/metabolism , Gliosis/pathology , Glial Fibrillary Acidic Protein/metabolism , Wound Healing , Brain Injuries/metabolism , Head Injuries, Closed/pathology , Inflammation/metabolism
15.
Brain ; 147(2): 698-716, 2024 Feb 01.
Article En | MEDLINE | ID: mdl-37955589

Alexander disease (AxD) is an intractable neurodegenerative disorder caused by GFAP mutations. It is a primary astrocyte disease with a pathological hallmark of Rosenthal fibres within astrocytes. AxD astrocytes show several abnormal phenotypes. Our previous study showed that AxD astrocytes in model mice exhibit aberrant Ca2+ signals that induce AxD aetiology. Here, we show that microglia have unique phenotypes with morphological and functional alterations, which are related to the pathogenesis of AxD. Immunohistochemical studies of 60TM mice (AxD model) showed that AxD microglia exhibited highly ramified morphology. Functional changes in microglia were assessed by Ca2+ imaging using hippocampal brain slices from Iba1-GCaMP6-60TM mice and two-photon microscopy. We found that AxD microglia showed aberrant Ca2+ signals, with high frequency Ca2+ signals in both the processes and cell bodies. These microglial Ca2+ signals were inhibited by pharmacological blockade or genetic knockdown of P2Y12 receptors but not by tetrodotoxin, indicating that these signals are independent of neuronal activity but dependent on extracellular ATP from non-neuronal cells. Our single-cell RNA sequencing data showed that the expression level of Entpd2, an astrocyte-specific gene encoding the ATP-degrading enzyme NTPDase2, was lower in AxD astrocytes than in wild-type astrocytes. In situ ATP imaging using the adeno-associated virus vector GfaABC1D ATP1.0 showed that exogenously applied ATP was present longer in 60TM mice than in wild-type mice. Thus, the increased ATP level caused by the decrease in its metabolizing enzyme in astrocytes could be responsible for the enhancement of microglial Ca2+ signals. To determine whether these P2Y12 receptor-mediated Ca2+ signals in AxD microglia play a significant role in the pathological mechanism, a P2Y12 receptor antagonist, clopidogrel, was administered. Clopidogrel significantly exacerbated pathological markers in AxD model mice and attenuated the morphological features of microglia, suggesting that microglia play a protective role against AxD pathology via P2Y12 receptors. Taken together, we demonstrated that microglia sense AxD astrocyte dysfunction via P2Y12 receptors as an increase in extracellular ATP and alter their morphology and Ca2+ signalling, thereby protecting against AxD pathology. Although AxD is a primary astrocyte disease, our study may facilitate understanding of the role of microglia as a disease modifier, which may contribute to the clinical diversity of AxD.


Alexander Disease , Mice , Animals , Alexander Disease/metabolism , Alexander Disease/pathology , Glial Fibrillary Acidic Protein/metabolism , Astrocytes/metabolism , Microglia/metabolism , Clopidogrel/metabolism , Calcium/metabolism , Disease Progression , Adenosine Triphosphate/metabolism
16.
J Neurosci Methods ; 402: 110025, 2024 Feb.
Article En | MEDLINE | ID: mdl-38036185

BACKGROUND: Immunohistochemical upregulation of glial fibrillary acidic protein (GFAP) is commonly used to detect astrogliosis in tissue sections and includes measurement of intensity and/or distribution of staining. There remains a lack of standard objective measures when diagnosing astrogliosis and its severity. NEW METHOD: Aim was to test a novel semi-quantitative assessment of GFAP which we term reactivity (R)-score, on its reproducibility and sensitivity to measure astrogliosis. The R-score, which is based on the proportion of astrocytes seen at each level of reactivity, was compared to 3 other commonly employed quantification methods in research: (1) thresholding, (2) point-counting, and (3) qualitative grading. Sub-regions of the hippocampus, medulla, and cerebellum were studied in piglet, and 4 human cases with clinically reported astrogliosis. Intra-assay coefficient of variation (CV) and percentage agreement cut-offs of ≤ 20% and ≥ 75% were used respectively to compare amongst the methods, with outcome measures being reproducibility across serial and non-serial sections, resilience to changes in experimental conditions, and inter- and intra-rater concordance. RESULTS: Averaged across 3 brain regions, the intra-assay coefficient of variation (CV) was 5% for R-score, with inter and intra-rater kappa scores being 0.99 and 0.95 respectively. COMPARISON WITH EXISTING METHODS AND CONCLUSIONS: Based on CV values, the R-score was superior to thresholding (CV of 51%) and point-counting (CV of 16%), with the qualitative grade being found to be on par (percentage agreement 95%). Given the ease, reproducibility and selectivity of the R-score, we propose its validity in future research purposes and clinical application.


Brain , Gliosis , Animals , Humans , Swine , Gliosis/metabolism , Immunohistochemistry , Glial Fibrillary Acidic Protein/metabolism , Reproducibility of Results , Brain/metabolism , Astrocytes/metabolism
17.
J Neurol ; 271(1): 1-23, 2024 Jan.
Article En | MEDLINE | ID: mdl-38015300

OBJECTIVE: This study aimed to synthesize the existing evidence on biomarkers related to coronavirus disease 2019 (COVID-19) patients who presented neurological events. METHODS: A systematic review of observational studies (any design) following PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines and the Cochrane Collaboration recommendations was performed (PROSPERO: CRD42021266995). Searches were conducted in PubMed and Scopus (updated April 2023). The methodological quality of nonrandomized studies was assessed using the Newcastle‒Ottawa Scale (NOS). An evidence gap map was built considering the reported biomarkers and NOS results. RESULTS: Nine specific markers of glial activation and neuronal injury were mapped from 35 studies published between 2020 and 2023. A total of 2,237 adult patients were evaluated in the included studies, especially during the acute phase of COVID-19. Neurofilament light chain (NfL) and glial fibrillary acidic protein (GFAP) biomarkers were the most frequently assessed (n = 27 studies, 77%, and n = 14 studies, 40%, respectively). Although these biomarkers were found to be correlated with disease severity and worse outcomes in the acute phase in several studies (p < 0.05), they were not necessarily associated with neurological events. Overall, 12 studies (34%) were judged as having low methodological quality, 9 (26%) had moderate quality, and 9 (26%) had high quality. CONCLUSIONS: Different neurological biomarkers in neurosymptomatic COVID-19 patients were identified in observational studies. Although the evidence is still scarce and conflicting for some biomarkers, well-designed longitudinal studies should further explore the pathophysiological role of NfL, GFAP, and tau protein and their potential use for COVID-19 diagnosis and management.


COVID-19 , Adult , Humans , COVID-19/complications , COVID-19 Testing , Evidence Gaps , Biomarkers/metabolism , Neurons/metabolism , Glial Fibrillary Acidic Protein/metabolism
18.
World Neurosurg ; 181: 96-106, 2024 Jan.
Article En | MEDLINE | ID: mdl-37852475

Nanofibers made by electrospinning have been used as bridging materials in animal models to regenerate nerves after spinal cord injury (SCI). In this meta-analysis study, we investigated the effect of these nanofibers on the motor function of animals after SCI. An extensive search in databases was performed. After primary and secondary screening, data included functional behavior, expression of glial fibrillary acidic protein, neurofilament-200 (NF-200), and ß-tubulin III were taken from the articles. The quality control of the articles, statistical analysis, and subgroup analysis were performed. The results from 14 articles and 16 separate experiments showed that electrospun nanofibers used alone could improve motor behavior and reduce glial injury after SCI.


Nanofibers , Spinal Cord Injuries , Rats , Animals , Rats, Sprague-Dawley , Recovery of Function , Nanofibers/therapeutic use , Spinal Cord Injuries/complications , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Spinal Cord
19.
Alzheimers Dement ; 20(1): 483-493, 2024 Jan.
Article En | MEDLINE | ID: mdl-37690071

INTRODUCTION: We studied how biomarkers of reactive astrogliosis mediate the pathogenic cascade in the earliest Alzheimer's disease (AD) stages. METHODS: We performed path analysis on data from 384 cognitively unimpaired individuals from the ALzheimer and FAmilies (ALFA)+ study using structural equation modeling to quantify the relationships between biomarkers of reactive astrogliosis and the AD pathological cascade. RESULTS: Cerebrospinal fluid (CSF) amyloid beta (Aß)42/40 was associated with Aß aggregation on positron emission tomography (PET) and with CSF p-tau181 , which was in turn directly associated with CSF neurofilament light (NfL). Plasma glial fibrillary acidic protein (GFAP) mediated the relationship between CSF Aß42/40 and Aß-PET, and CSF YKL-40 partly explained the association between Aß-PET, p-tau181 , and NfL. DISCUSSION: Our results suggest that reactive astrogliosis, as indicated by different fluid biomarkers, influences the pathogenic cascade during the preclinical stage of AD. While plasma GFAP mediates the early association between soluble and insoluble Aß, CSF YKL-40 mediates the latter association between Aß and downstream Aß-induced tau pathology and tau-induced neuronal injury. HIGHLIGHTS: Lower CSF Aß42/40 was directly linked to higher plasma GFAP concentrations. Plasma GFAP partially explained the relationship between soluble Aß and insoluble Aß. CSF YKL-40 mediated Aß-induced tau phosphorylation and tau-induced neuronal injury.


Alzheimer Disease , Humans , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Astrocytes/metabolism , Biomarkers/cerebrospinal fluid , Chitinase-3-Like Protein 1 , Glial Fibrillary Acidic Protein/metabolism , Gliosis/pathology , Inflammation , Intermediate Filaments/metabolism , Intermediate Filaments/pathology , tau Proteins/cerebrospinal fluid
20.
Acta Histochem ; 126(1): 152131, 2024 Jan.
Article En | MEDLINE | ID: mdl-38159478

The study of astrocytes and its role in the development and evolution of neurodegenerative diseases, including Alzheimer's disease (AD) is essential to fully understand their aetiology. The aim if this study is to deepen into the concept of the heterogeneity of astrocyte subpopulations in the EC and in particular the identification of differentially functioning astrocyte subpopulations that respond differently to AD progression. S100ß protein belongs to group of small calcium regulators of cell membrane channels and pumps that are expressed by astrocytes and is hypothesised to play and have a relevant role in AD development. We analysed the selective differentiation of S100ß-positive astrocytes into Glutamine synthetase (GS) and Glial fibrillary acidic protein (GFAP)-positive sub-groups in the entorhinal cortex (EC) of AD triple transgenic animal model (3xTg-AD). EC is the brain region earliest affected in humans AD but also in this closest animal model regarding their pathology and time course. We observed no changes in the number of S100ß-positive astrocytes between 1 and 18 months of age in the EC of 3xTg-AD mice. However, we identified relevant morphological changes in S100ß/GFAP positive astrocytes showing a significant reduction in their surface and volume whilst an increase in number and percentage. Furthermore, the percentage of S100ß/GS positive astrocyte population was also increased in 18 months old 3xTg-AD mice compared to the non-Tg mice. Our findings reveal the presence of differentially controlled astrocyte populations that respond differently to AD progression in the EC of 3xTg-AD mice. These results highpoints the major astrocytic role together with its early and marked affection in AD and arguing in favour of its importance in neurogenerative diseases and potential target for new therapeutic approaches.


Alzheimer Disease , Animals , Humans , Infant , Mice , Alzheimer Disease/genetics , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Astrocytes/metabolism , Disease Models, Animal , Entorhinal Cortex/metabolism , Entorhinal Cortex/pathology , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Glutamate-Ammonia Ligase/genetics , Glutamate-Ammonia Ligase/metabolism , Mice, Transgenic
...