Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 640
1.
J Integr Neurosci ; 21(1): 38, 2022 Jan 28.
Article En | MEDLINE | ID: mdl-35164474

Sepsis-associated encephalopathy is a common brain diseases, presenting severe diffuse brain dysfunction. The umbilical cord mesenchymal stem cells have been reported to have protective role for treating diseases, while its role in sepsis-associated encephalopathy remained elusive. This brief report investigated the therapeutic effect of umbilical cord mesenchymal stem cells on sepsis-associated encephalopathy in mice model and uncovering the underlying mechanism. The sepsis-associated encephalopathy mice were injected with 3 mg/kg lipopolysaccharide. An enzyme-linked immunosorbent assay was carried out to determine the production of inflammatory cytokines. Morris water maze test was used to evaluate mice's neurological dysfunction. Cell apoptosis and tissue injury of the cerebral cortex were assessed using terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay and HE staining. Evans Blue leakage detection was used to examine the blood-brain barrier integrity. The protein levels were determined using Western blot. Results showed that the productions of inflammatory cytokines including interleukin 6 (IL-6), interleukin-1ß (IL-1ß), tumor necrosis factor α (TNF-α), and high mobility group box protein 1 (HMGB1) and activated NF-κB were increased in sepsis-associated encephalopathy mice, which were decreased by umbilical cord mesenchymal stem cells treatment. Besides, umbilical cord mesenchymal stem cells inhibited lipopolysaccharide-induced cell apoptosis and neuron injury of the cerebral cortex in sepsis-associated encephalopathy mice. Moreover, cognitive dysfunction was observed in sepsis-associated encephalopathy mice, which was alleviated by umbilical cord mesenchymal stem cells. Furthermore, umbilical cord mesenchymal stem cells activated PI3K/AKT signaling pathway. In conclusion, umbilical cord mesenchymal stem cells alleviated inflammation, cell apoptosis and neuron injury of the cerebral cortex, and cognitive dysfunction in sepsis-associated encephalopathy animal model in a PI3K/AKT dependent pathway, making them to be a promising therapeutic strategy for treating sepsis-associated encephalopathy.


Cerebral Cortex , Cognitive Dysfunction/therapy , Mesenchymal Stem Cell Transplantation , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sepsis-Associated Encephalopathy/immunology , Sepsis-Associated Encephalopathy/therapy , Umbilical Cord , Animals , Behavior, Animal/physiology , Cerebral Cortex/immunology , Cerebral Cortex/pathology , Cognitive Dysfunction/etiology , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred C57BL , Sepsis-Associated Encephalopathy/complications , Signal Transduction/physiology
2.
Brain Res Bull ; 181: 46-54, 2022 04.
Article En | MEDLINE | ID: mdl-35077842

The aim of the current study was to determine the effects of cerebral contusion injury with purinergic adenosine triphosphate Y1 (P2Y1) receptor blockers on postinjury inflammatory responses. Adenosine triphosphate (ATP) is released into the extracellular space in several in vivo models, including traumatic brain injury. Released ATP triggers neuroinflammation via activation of microglial cells. P2Y1 receptor blockers were reported to suppress extracellular ATP elevation in several disease models through inhibition of cellular ATP release. In addition to the beneficial effects of inflammation, excess inflammatory reactions cause secondary damage and aggravate outcomes. Here, we assessed the effect of the selective P2Y1 receptor blocker MRS2179 on its potential to prevent posttraumatic inflammation in a rat cerebral contusion model. Cerebral contusion injury was induced in the rat cerebral cortex. Either MRS2179 or artificial cerebral spinal fluid as a control was administered in situ into the center of contused tissue via a subcutaneously implanted osmotic pump. Galectin 3, a marker of microglia and proinflammatory cytokines, was measured 1, 3 and 7 days following injury. Another group of rats was assessed for behavioral performance up to 28 days after injury, including the beam walk test, neurological response test and plus maze test. The Galectin 3 levels in the cortex around the contusion cavity and in the cortex far from the contusion cavity were significantly suppressed by MRS2179 administration on postinjury Days 1 and 3 (p < 0.05). However, administration of MRS2179 failed to improve behavioral outcome. Administration of MRS2179 successfully suppressed microglial activation in a traumatic brain injury model, which will be a potent treatment option in the future. Further study is required to conclude its therapeutic effects.


Adenosine Diphosphate/analogs & derivatives , Anti-Inflammatory Agents/pharmacology , Brain Injuries, Traumatic/drug therapy , Cerebral Cortex/drug effects , Cerebral Cortex/injuries , Galectin 3/drug effects , Neuroinflammatory Diseases/drug therapy , Purinergic P2Y Receptor Antagonists/pharmacology , Adenosine Diphosphate/pharmacology , Animals , Behavior, Animal/drug effects , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/immunology , Cerebral Cortex/immunology , Disease Models, Animal , Microglia/drug effects , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/immunology , Rats
3.
Front Immunol ; 12: 766178, 2021.
Article En | MEDLINE | ID: mdl-34721438

Neuroinflammation is a key process in the pathogenesis of subarachnoid hemorrhage (SAH) and contributes to poor outcome in patients. The purpose of this study is to explore the effect of triggering receptor expressed on myeloid cells 1 (TREM1) in the SAH, as well as its potential mechanism. In our study, plasma levels of soluble TREM1 was increased significantly after SAH and correlated to SAH severity and serum C-reactiveprotein. TREM1 inhibitory peptide LP17 alleviated the neurological deficits, attenuated brain water content, and reduced neuronal damage after SAH. Meanwhile, TREM1 inhibitory peptide decreased neuroinflammation (evidenced by the decreased levels of markers including IL-6, IL-1ß, TNF-α) by attenuating proinflammatory subtype transition of microglia (evidenced by the decreased levels of markers including CD68, CD16, CD86) and decreasing the formation of neutrophil extracellular traps (evidenced by the decreased levels of markers including CitH3, MPO, and NE). Further mechanistic study identified that TREM1 can activate downstream proinflammatory pathways through interacting with spleen tyrosine kinase (SYK). In conclusion, inhibition of TREM1 alleviates neuroinflammation by attenuating proinflammatory subtype transition of microglia and decreasing the formation of neutrophil extracellular traps through interacting with SYK after SAH. TREM1 may be a a promising therapeutic target for SAH.


Extracellular Traps/immunology , Microglia/immunology , Neuroinflammatory Diseases/immunology , Subarachnoid Hemorrhage/immunology , Syk Kinase/immunology , Triggering Receptor Expressed on Myeloid Cells-1/immunology , Animals , Antigens, CD/genetics , Cerebral Cortex/immunology , Cytokines/genetics , Humans , Male , Mice, Inbred C57BL , Neuroinflammatory Diseases/blood , Subarachnoid Hemorrhage/blood , Triggering Receptor Expressed on Myeloid Cells-1/blood
4.
Nat Commun ; 12(1): 5659, 2021 09 27.
Article En | MEDLINE | ID: mdl-34580300

Early Alzheimer's disease (AD) pathology can be found in cortical biopsies taken during shunt placement for Normal Pressure Hydrocephalus. This represents an opportunity to study early AD pathology in living patients. Here we report RNA-seq data on 106 cortical biopsies from this patient population. A restricted set of genes correlate with AD pathology in these biopsies, and co-expression network analysis demonstrates an evolution from microglial homeostasis to a disease-associated microglial phenotype in conjunction with increasing AD pathologic burden, along with a subset of additional astrocytic and neuronal genes that accompany these changes. Further analysis demonstrates that these correlations are driven by patients that report mild cognitive symptoms, despite similar levels of biopsy ß-amyloid and tau pathology in comparison to patients who report no cognitive symptoms. Taken together, these findings highlight a restricted set of microglial and non-microglial genes that correlate with early AD pathology in the setting of subjective cognitive decline.


Alzheimer Disease/complications , Cerebral Cortex/pathology , Cognitive Dysfunction/immunology , Gene Regulatory Networks/immunology , Hydrocephalus, Normal Pressure/immunology , Age of Onset , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Astrocytes/immunology , Astrocytes/pathology , Biopsy , Cerebral Cortex/cytology , Cerebral Cortex/immunology , Cognitive Dysfunction/diagnosis , Cognitive Dysfunction/genetics , Cognitive Dysfunction/pathology , Female , Humans , Hydrocephalus, Normal Pressure/genetics , Hydrocephalus, Normal Pressure/pathology , Hydrocephalus, Normal Pressure/surgery , Male , Microglia/immunology , Microglia/pathology , Neuropsychological Tests , RNA-Seq , Retrospective Studies
5.
Biochim Biophys Acta Mol Basis Dis ; 1867(11): 166230, 2021 11 01.
Article En | MEDLINE | ID: mdl-34358627

Orexin-A (OXA) is a neuropeptide with neuroprotective effect by reducing cerebral ischemia/reperfusion injury (CIRI). Inflammation and apoptosis mediated by astrocyte activation are the key pathological mechanisms for CIRI. We thus attempted to confirm neuroprotective effects of OXA on astrocytic inflammation and apoptosis in CIRI and clarify the relative mechanisms. A middle cerebral artery occlusion and reperfusion (MCAO/R) rat model and U251 glioma cells model subjected to oxygen glucose deprivation and reperfusion (OGD/R) were established, with or without OXA treatment. Neurological deficit score was determined, and cerebral infarct volume was evaluated by 2,3,5-triphenyltetrazolium chloride (TTC) staining. Western Blot was used to detect the expressions of NF-κB p65, p-p65, p-ERK, p-p38, GFAP, OX1R, IL-1ß, TNF-α, IL-6, iNOS, Bcl-2, Bax, CytC, cleaved caspase-9 and cleaved caspase-3 in vivo and in vitro. Pro-inflammatory cytokines in cell supernatant IL-1ß, TNF-α and IL-6 were determined by ELISA. Hoechst 33342 staining was used to detect the apoptosis of astrocyte. Immunofluorescent staining was performed to assess the nuclear translocation of p65 and the expression of GFAP. The results showed that OXA significantly improved neurological deficit score and decreased the volume of infarct area in brain. OXA decreased inflammatory mediators, inhibited astrocyte activation and nuclear translocation of NF-κB and phosphorylation of NF-κB, MAPK/ERK and MAPK/p38. Besides, OXA suppressed apoptosis via upregulating the ratio of Bcl-2/Bax and downregulating cytochrome C, cleaved-caspase-9 and cleaved caspase-3. Overall, it was concluded that OXA exerts neuroprotective effect during CIRI through attenuating astrocytes apoptosis, astrocytes activation and pro-inflammatory cytokines production, by Inhibiting OX1R-mediated NF-κB, MAPK/ERK and MAPK/p38 signaling pathways. The progress in our study is helpful to elucidate the molecular mechanisms of OXA neuroprotection, which could lead to the development of new treatment strategies for ischemic stroke.


Astrocytes/pathology , Infarction, Middle Cerebral Artery/complications , Orexins/metabolism , Reperfusion Injury/immunology , Animals , Apoptosis/immunology , Astrocytes/immunology , Cell Line, Tumor , Cerebral Cortex/blood supply , Cerebral Cortex/cytology , Cerebral Cortex/immunology , Cerebral Cortex/pathology , Disease Models, Animal , Humans , Infarction, Middle Cerebral Artery/immunology , Infarction, Middle Cerebral Artery/pathology , MAP Kinase Signaling System/immunology , Male , NF-kappa B/metabolism , Orexin Receptors/metabolism , Orexins/administration & dosage , Rats , Reperfusion Injury/pathology
6.
Cells ; 10(7)2021 07 06.
Article En | MEDLINE | ID: mdl-34359880

An imbalance of TNF signalling in the inflammatory milieu generated by meningeal immune cell infiltrates in the subarachnoid space in multiple sclerosis (MS), and its animal model may lead to increased cortical pathology. In order to explore whether this feature may be present from the early stages of MS and may be associated with the clinical outcome, the protein levels of TNF, sTNF-R1 and sTNF-R2 were assayed in CSF collected from 122 treatment-naïve MS patients and 36 subjects with other neurological conditions at diagnosis. Potential correlations with other CSF cytokines/chemokines and with clinical and imaging parameters at diagnosis (T0) and after 2 years of follow-up (T24) were evaluated. Significantly increased levels of TNF (fold change: 7.739; p < 0.001), sTNF-R1 (fold change: 1.693; p < 0.001) and sTNF-R2 (fold change: 2.189; p < 0.001) were detected in CSF of MS patients compared to the control group at T0. Increased TNF levels in CSF were significantly (p < 0.01) associated with increased EDSS change (r = 0.43), relapses (r = 0.48) and the appearance of white matter lesions (r = 0.49). CSF levels of TNFR1 were associated with cortical lesion volume (r = 0.41) at T0, as well as with new cortical lesions (r = 0.56), whilst no correlation could be found between TNFR2 levels in CSF and clinical or MRI features. Combined correlation and pathway analysis (ingenuity) of the CSF protein pattern associated with TNF expression (encompassing elevated levels of BAFF, IFN-γ, IL-1ß, IL-10, IL-8, IL-16, CCL21, haptoglobin and fibrinogen) showed a particular relationship to the interaction between innate and adaptive immune response. The CSF sTNF-R1-associated pattern (encompassing high levels of CXCL13, TWEAK, LIGHT, IL-35, osteopontin, pentraxin-3, sCD163 and chitinase-3-L1) was mainly related to altered T cell and B cell signalling. Finally, the CSF TNFR2-associated pattern (encompassing high CSF levels of IFN-ß, IFN-λ2, sIL-6Rα) was linked to Th cell differentiation and regulatory cytokine signalling. In conclusion, dysregulation of TNF and TNF-R1/2 pathways associates with specific clinical/MRI profiles and can be identified at a very early stage in MS patients, at the time of diagnosis, contributing to the prediction of the disease outcome.


Multiple Sclerosis/diagnostic imaging , Multiple Sclerosis/genetics , Receptors, Tumor Necrosis Factor, Type II/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Tumor Necrosis Factor-alpha/genetics , Adaptive Immunity , Adult , Antigens, CD/cerebrospinal fluid , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/cerebrospinal fluid , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/immunology , B-Lymphocytes/immunology , B-Lymphocytes/pathology , C-Reactive Protein/cerebrospinal fluid , C-Reactive Protein/genetics , C-Reactive Protein/immunology , Case-Control Studies , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/immunology , Cerebral Cortex/pathology , Chemokine CXCL13/cerebrospinal fluid , Chemokine CXCL13/genetics , Chemokine CXCL13/immunology , Chitinase-3-Like Protein 1/cerebrospinal fluid , Chitinase-3-Like Protein 1/genetics , Chitinase-3-Like Protein 1/immunology , Cytokine TWEAK/cerebrospinal fluid , Cytokine TWEAK/genetics , Cytokine TWEAK/immunology , Early Diagnosis , Female , Gene Expression Regulation , Humans , Immunity, Innate , Interleukins/cerebrospinal fluid , Interleukins/genetics , Interleukins/immunology , Magnetic Resonance Imaging , Male , Meninges/diagnostic imaging , Meninges/immunology , Meninges/pathology , Multiple Sclerosis/cerebrospinal fluid , Multiple Sclerosis/pathology , Osteopontin/cerebrospinal fluid , Osteopontin/genetics , Osteopontin/immunology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Receptors, Tumor Necrosis Factor, Type I/cerebrospinal fluid , Receptors, Tumor Necrosis Factor, Type I/immunology , Receptors, Tumor Necrosis Factor, Type II/cerebrospinal fluid , Receptors, Tumor Necrosis Factor, Type II/immunology
7.
Int J Mol Sci ; 22(13)2021 Jun 24.
Article En | MEDLINE | ID: mdl-34202695

Chrysomycin A (Chr-A), an antibiotic chrysomycin, was discovered in 1955 and is used to treat cancer and tuberculosis. In the present study, the anti-neuroinflammatory effects and possible mechanism of Chr-A in BALB/c mice and in BV2 microglia cells stimulated by lipopolysaccharide (LPS) were investigated. Firstly, the cortex tissues of mice were analyzed by RNA-seq transcriptome to identify differentially expressed genes (DEGs) regulated by Chr-A in LPS-stimulated mice. Inflammatory cytokines and inflammatory proteins were detected by enzyme-linked immunosorbent assay and Western blot. In RNAseq detection, 639 differential up-regulated genes between the control group and LPS model group and 113 differential down-regulated genes between the LPS model group and Chr-A treatment group were found, and 70 overlapping genes were identified as key genes for Chr-A against neuroinflammation. Subsequent GO biological process enrichment analysis showed that the anti-neuroinflammatory effect of Chr-A might be related to the response to cytokine, cellular response to cytokine stimulus, and regulation of immune system process. The significant signaling pathways of KEGG enrichment analysis were mainly involved in TNF signaling pathway, cytokine-cytokine receptor interaction, NF-κB signaling pathway, IL-17 signaling pathway and NOD-like receptor signaling pathway. Our results of in vivo or in vitro experiments showed that the levels of pro-inflammatory factors including NO, IL-6, IL-1ß, IL-17, TNF-α, MCP-1, CXCL12, GM-CSF and COX2 in the LPS-stimulated group were higher than those in the control group, while Chr-A reversed those conditions. Furthermore, the Western blot analysis showed that its anti-neuroinflammation appeared to be related to the down-regulation of NLRP3/cleaved caspase-1 signaling pathway. The current findings provide new insights into the activity and molecular mechanisms of Chr-A for the treatment of neuroinflammation.


Aminoglycosides/pharmacology , Caspase 1/metabolism , Microglia/drug effects , Microglia/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neurogenic Inflammation/metabolism , Signal Transduction/drug effects , Aminoglycosides/chemistry , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/immunology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation/drug effects , Lipopolysaccharides/immunology , Mice , Microglia/immunology , Molecular Structure , Neurogenic Inflammation/etiology , Proteolysis , Transcriptome
8.
Cells ; 10(6)2021 06 19.
Article En | MEDLINE | ID: mdl-34205372

Pain is an unpleasant sensation that alerts one to the presence of obnoxious stimuli or sensations. These stimuli are transferred by sensory neurons to the dorsal root ganglia-spinal cord and finally to the brain. Glial cells in the peripheral nervous system, astrocytes in the brain, dorsal root ganglia, and immune cells all contribute to the development, maintenance, and resolution of pain. Both innate and adaptive immune responses modulate pain perception and behavior. Neutrophils, microglial, and T cell activation, essential components of the innate and adaptive immune responses, can play both excitatory and inhibitory roles and are involved in the transition from acute to chronic pain. Immune responses may also exacerbate pain perception by modulating the function of the cortical-limbic brain regions involved in behavioral and emotional responses. The link between an emotional state and pain perception is larger than what is widely acknowledged. In positive psychological states, perception of pain along with other somatic symptoms decreases, whereas in negative psychological states, these symptoms may worsen. Sex differences in mechanisms of pain perception are not well studied. In this review, we highlight what is known, controversies, and the gaps in this field.


Cerebral Cortex/immunology , Limbic System/immunology , Microglia/immunology , Neurons/immunology , Pain/immunology , Animals , Astrocytes/immunology , Astrocytes/pathology , Cerebral Cortex/pathology , Humans , Limbic System/pathology , Lymphocyte Activation , Microglia/pathology , Neurons/pathology , Neutrophil Activation , Neutrophils/immunology , Neutrophils/pathology , Pain/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
9.
J Immunol Res ; 2021: 5317662, 2021.
Article En | MEDLINE | ID: mdl-34327243

Zika virus (ZIKV) has attracted the wide global attention due to its causal link to microcephaly. In this study, two amino acid (aa) mutation (E143K and R3394K) were identified at the fourth generation (named ZKC2P4) during the serial passage of ZIKV-Asian lineage ZKC2/2016 strain in the newborn mouse brain, while another seven aa deletions in envelope (E) protein were detected in ZKC2P6. ZKC2P6 is a novel nonglycosylated E protein Asian ZIKV we first identified and provides the first direct supporting evidence that glycosylation motif could be lost during the passage in neonatal mice. To study the impact of E protein glycosylation ablation, we compared the pathogenicity of ZKC2P6 with that of ZKC2P4. The results showed that the loss of E protein glycosylation accelerated the disease progression, as evidenced by an earlier weight loss and death, a thinner cerebral cortex, and more serious tissue lesions and inflammation/necrosis. Furthermore, ZKC2P6 exhibited a greater ability to replicate and caused severer cell apoptosis than that of ZKC2P4. Therefore, the ablation of E glycosylation generally enhances the neurovirulence of ZIKV and cell apoptosis in newborn mice.


Cerebral Cortex/pathology , Viral Envelope Proteins/metabolism , Zika Virus Infection/immunology , Zika Virus/pathogenicity , Animals , Animals, Newborn , Apoptosis , Cell Line, Tumor , Cerebral Cortex/immunology , Cerebral Cortex/virology , Chlorocebus aethiops , Disease Models, Animal , Disease Progression , Glycosylation , Humans , Mice , Sequence Deletion , Vero Cells , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Virus Replication/immunology , Zika Virus/genetics , Zika Virus/immunology , Zika Virus/metabolism , Zika Virus Infection/pathology , Zika Virus Infection/virology
10.
Int Immunopharmacol ; 98: 107689, 2021 Sep.
Article En | MEDLINE | ID: mdl-34153666

BACKGROUND: Heparin, a commonly used anticoagulant, has been found to improve cerebral ischemia-reperfusion injury (CIR-CA) following cardiopulmonary resuscitation (CPR). Here, we aimed to explore the role of pleiotrophin (PTN)/syndecan-3 pathway in heparin therapy for CIR-CA. MATERIALS AND METHODS: The CA-CPR model was constructed in Sprague-Dawley (SD) rats, which were treated with low molecular weight heparin, and the neurological changes and brain histopathological changes were evaluated. For in-vitro experiments, the ischemic injury model of primary neurons was established by oxygen and glucose deprivation (OGD), and the neuron regeneration was detected via the Cell counting Kit-8 (CCK8) method, flow cytometry and microscopy. CREB antagonist (KG-501), ERK antagonist (PD98059) and si-PTN were used respectively to inhibit the expression of CREB, ERK and PTN in cells, so as to explore the role of heparin in regulating neuronal regeneration. RESULTS: Compared with the sham rats, the neurological deficits and cerebral edema of CA-CPR rats were significantly improved after heparin treatment. Heparin also attenuated OGD-mediated neuronal apoptosis and promoted neurite outgrowth in vitro. Moreover, heparin attenuated CA-CPR-mediated neuronal apoptosis and microglial neuroinflammation. In terms of the mechanism, heparin upregulated the expression of ERK, CREB, NF200, BDNF, NGF, PTN and syndecan-3 in the rat brains. Inhibition of ERK, CREB and interference with PTN expression notably weakened the heparin-mediated neuroprotective effects and restrained the expression of ERK/CREB and PTN/syndecan-3 pathway. CONCLUSION: Heparin attenuates the secondary brain injury induced by CA-CPR through regulating the ERK/CREB-mediated PTN/syndecan-3 pathway.


Brain Ischemia/drug therapy , Heart Arrest/complications , Heparin/administration & dosage , Neuroprotective Agents/administration & dosage , Reperfusion Injury/prevention & control , Animals , Apoptosis , Brain Ischemia/etiology , Brain Ischemia/pathology , Cardiopulmonary Resuscitation/adverse effects , Carrier Proteins/metabolism , Cells, Cultured , Cerebral Cortex/blood supply , Cerebral Cortex/drug effects , Cerebral Cortex/immunology , Cerebral Cortex/pathology , Cyclic AMP Response Element-Binding Protein/metabolism , Cytokines/metabolism , Disease Models, Animal , Heart Arrest/therapy , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Male , Microglia/drug effects , Microglia/immunology , Microglia/pathology , Nerve Regeneration/drug effects , Neurons/drug effects , Neurons/immunology , Neurons/pathology , Primary Cell Culture , Rats , Reperfusion Injury/etiology , Reperfusion Injury/pathology , Syndecan-3/metabolism
11.
Front Immunol ; 12: 582768, 2021.
Article En | MEDLINE | ID: mdl-34177880

Background: The presence of fluid attenuated inversion recovery (FLAIR)-hyperintense lesions in anti-myelin oligodendrocyte glycoprotein (MOG) antibody-associated cerebral cortical encephalitis with seizures (FLAMCES) was recently reported. However, the clinical characteristics and outcome of this rare clinico-radiographic syndrome remain unclear. Methods: The present study reported two new cases. In addition, cases in the literature were systematically reviewed to investigate the clinical symptoms, magnetic resonance imaging (MRI) abnormalities, treatments and prognosis for this rare clinico-radiographic syndrome. Results: A total of 21 cases were identified during a literature review, with a mean patient age at onset of 26.8 years. The primary clinicopathological characteristics included seizures (100%), headache (71.4%), fever (52.3%) and other cortical symptoms associated with the encephalitis location (61.9%). The common seizure types were focal to bilateral tonic-clonic seizures (28.6%) and unknown-onset tonic-clonic seizures (38.1%). The cortical abnormalities on MRI FLAIR imaging were commonly located in the frontal (58.8%), parietal (70.6%) and temporal (64.7%) lobes. In addition, pleocytosis in the cerebrospinal fluid was reported in the majority of the patients (95.2%). All patients received a treatment regimen of corticosteroids and 9 patients received anti-epileptic drugs. Clinical improvement was achieved in all patients; however, one-third of the patients reported relapse following recovery from cortical encephalitis. Conclusions: FLAMCES is a rare phenotype of MOG-associated disease. Thus, the wider recognition of this rare syndrome may enable timely diagnosis and the development of suitable treatment regimens.


Autoantibodies/metabolism , Cerebral Cortex/pathology , Cerebrospinal Fluid/immunology , Encephalitis/diagnosis , Immune Complex Diseases/diagnosis , Adrenal Cortex Hormones/therapeutic use , Adult , Anticonvulsants/therapeutic use , Cerebral Cortex/immunology , Encephalitis/drug therapy , Female , Headache , Humans , Immune Complex Diseases/drug therapy , Leukocytosis , Magnetic Resonance Imaging , Male , Middle Aged , Myelin-Oligodendrocyte Glycoprotein , Seizures , Young Adult
12.
Can J Physiol Pharmacol ; 99(9): 847-856, 2021 Sep.
Article En | MEDLINE | ID: mdl-34161158

This study explored the potential effects of mild hyperhomocysteinemia (HHcy) on the blood-brain barrier (BBB) and neuroinflammation. Seven-week-old male wild-type C57BL/6 mice were fed normal mouse chow (the control group) or a methionine-enriched diet (the HHcy group) for 14 weeks. Mice in the HHcy group exhibited a slight increase in serum Hcy levels (13.56 ± 0.61 µmol/L). Activation of the ERK signaling pathway, up-regulation of matrix metalloproteinase-9 (MMP-9), and degradation of tight junction proteins (occludin and claudin-5) were observed in both the cerebral cortex and hippocampus of mice with mild HHcy. However, microglia were not activated and the levels of tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß) were not changed in either the cerebral cortex or hippocampus of mice with mild HHcy. Moreover, the signaling activity of STAT3 also did not differ significantly between the two groups. These findings demonstrate that the BBB is highly vulnerable to homocysteine insult. Even a slight increase in serum homocysteine levels up-regulates MMP-9 expression and disrupts the BBB integrity. Meanwhile, microglia activation or the STAT3 pathway might not contribute to the effects of mild HHcy on the brain.


Blood-Brain Barrier/physiopathology , Cerebral Cortex/immunology , Hippocampus/immunology , Hyperhomocysteinemia/physiopathology , Neuroinflammatory Diseases/etiology , Animals , Cytokines/analysis , Homocysteine/blood , MAP Kinase Signaling System/physiology , Male , Matrix Metalloproteinase 9/analysis , Mice , Mice, Inbred C57BL , STAT3 Transcription Factor/physiology , Signal Transduction/physiology
13.
Exp Cell Res ; 405(1): 112635, 2021 08 01.
Article En | MEDLINE | ID: mdl-34051241

Exosomes have been shown to have therapeutic potential for cerebral ischemic diseases. In this study, we investigated the neuroprotective effects of normoxic and hypoxic bone marrow mesenchymal stromal cells-derived exosomes (N-BM-MSCs-Exo and H-BM-MSCs-Exo, respectively) on oxygen-glucose deprivation (OGD) injury in mouse neuroblastoma N2a cells and rat primary cortical neurons. The proportions of dead cells in N2a and primary cortical neurons after OGD injury were significantly increased, and N-BM-MSCs-Exo (40 µg/ml) could reduce the ratios, noteworthily, the protective effects of H-BM-MSCs-Exo (40 µg/ml) were more potent. Western blotting analysis indicated that N-BM-MSCs-Exo decreased the expression of NLRP3, ASC, Caspase-1, GSDMD-N, cleaved IL-1ß and IL-18 in N2a cells. However, H-BM-MSCs-Exo (40 µg/ml) was more powerful in inhibiting the expression of these proteins in comparison with N-BM-MSCs-Exo. Similar results were obtained in primary cortical neurons. Immunofluorescence assays showed that after N-BM-MSCs-Exo and H-BM-MSCs-Exo treatment, the co-localization of NLRP3, ASC, Caspase-1 and the GSDMD translocation from the nucleus to the cytoplasm and membrane after OGD injury were reduced in N2a cells and primary cortical neurons, and H-BM-MSCs-Exo had a more obvious effect. In addition, N-BM-MSCs-Exo and H-BM-MSCs-Exo significantly reduced lactate dehydrogenase (LDH) release and the IL-18 levels in cell culture medium in N2a cells and primary cortical neurons. Once again H-BM-MSCs-Exo induced these effects more potently than N-BM-MSCs-Exo. All of these results demonstrated that N-BM-MSCs-Exo and H-BM-MSCs-Exo have significant neuroprotective effects against NLRP3 inflammasome-mediated pyroptosis. H-BM-MSCs-Exo has a more pronounced protective effect than N-BM-MSCs-Exo and may be used to ameliorate the progression of cerebral ischemia and hypoxia injury in patients.


Exosomes/physiology , Hypoxia/physiopathology , Mesenchymal Stem Cells/cytology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neuroblastoma/prevention & control , Neurons/cytology , Pyroptosis , Animals , Cerebral Cortex/cytology , Cerebral Cortex/immunology , Cerebral Cortex/injuries , Cerebral Cortex/metabolism , Glucose/deficiency , Inflammasomes/physiology , Male , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Neuroblastoma/etiology , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neurons/immunology , Neurons/metabolism , Neurons/pathology , Oxygen/metabolism , Rats , Rats, Sprague-Dawley , Recovery of Function , Signal Transduction
14.
Neurosci Lett ; 754: 135869, 2021 05 29.
Article En | MEDLINE | ID: mdl-33857550

Intracellular Aß (iAß) expression, extracellular Aß (eAß) plaque formation and microglial reactivity are characteristic neuropathological events of Alzheimer's disease (AD) and have been detected in several transgenic mouse models of this disease. In this work we decided to investigate the early (2-7 months of age) development of these phenomena at both regional and cellular levels in 5XFAD mice, a severe transgenic mouse model of AD. We demonstrated that 1) Aß pathology develops in many but not all brain regions, 2) iAß is transient and almost always followed by eAß in grey matter regions, and the respective levels are roughly proportional, and 3) in about 1/3 of the grey matter regions with Aß pathology and in several white matter regions, eAß plaques can appear where no iAß-positive structures were detected. We also showed that male and female mice share a similar regional and cellular pattern of Aß pathology development that is more prominent in females. Early iAß is associated to the activation of microglia, while subsequent formation of eAß plaques is associated with markedly increased density of microglial cells that acquire a characteristic clustered phenotype. Present analysis is relevant to set a reference for pathophysiological studies and to define specific targets for the test of therapeutic interventions in this widely used AD transgenic model.


Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Cerebral Cortex/pathology , Plaque, Amyloid/pathology , Alzheimer Disease/genetics , Alzheimer Disease/immunology , Amyloid beta-Peptides/analysis , Animals , Cerebral Cortex/cytology , Cerebral Cortex/immunology , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Transgenic , Microglia/immunology , Microglia/metabolism , Microglia/pathology , Plaque, Amyloid/genetics , Plaque, Amyloid/immunology , Sex Factors
15.
J Neuroimmunol ; 355: 577549, 2021 06 15.
Article En | MEDLINE | ID: mdl-33839521

The pathophysiology of neurological diseases related to potassium-channel dysfunction such as epilepsy is increasingly linked to immune system modulation. However, there are limited reports of which interleukin-4 (IL-4) can act on the neuroinflammatory response after seizure. Hence, we evaluated the effect of IL-4 in murine model of neuroexcitotoxcity using kaliotoxin (KTx), a potassium-channel blocker. Results showed that IL-4 treatment can significantly reduce the neuronal death induced by KTx. Probably by decreasing mitochondria swelling, reversing oxidative damage and enhancing Bcl-2 expression. Furthermore, IL-4 treatment significantly reduced TNF-α expression and enhanced GFAP and IL-10 expressions in the brain. IL-4 can be neuroprotective in epileptogenesis.


Cerebral Cortex/drug effects , Epilepsy/prevention & control , Immunologic Factors/administration & dosage , Interleukin-4/administration & dosage , Neuroprotective Agents/administration & dosage , Potassium Channel Blockers/toxicity , Animals , Cerebral Cortex/immunology , Cerebral Cortex/pathology , Epilepsy/chemically induced , Epilepsy/immunology , Epilepsy/pathology , Injections, Intraperitoneal , Injections, Intraventricular , Mice , Scorpion Venoms/toxicity
16.
Neurology ; 96(14): e1865-e1875, 2021 04 06.
Article En | MEDLINE | ID: mdl-33737372

OBJECTIVES: To explore in vivo innate immune cell activation as a function of the distance from ventricular CSF in patients with multiple sclerosis (MS) using [18F]-DPA714 PET and to investigate its relationship with periventricular microstructural damage, evaluated by magnetization transfer ratio (MTR), and with trajectories of disability worsening. METHODS: Thirty-seven patients with MS and 19 healthy controls underwent MRI and [18F]-DPA714 TSPO dynamic PET, from which individual maps of voxels characterized by innate immune cell activation (DPA+) were generated. White matter (WM) was divided in 3-mm-thick concentric rings radiating from the ventricular surface toward the cortex, and the percentage of DPA+ voxels and mean MTR were extracted from each ring. Two-year trajectories of disability worsening were collected to identify patients with and without recent disability worsening. RESULTS: The percentage of DPA+ voxels was higher in patients compared to controls in the periventricular WM (p = 6.10e-6) and declined with increasing distance from ventricular surface, with a steeper gradient in patients compared to controls (p = 0.001). This gradient was found in both periventricular lesions and normal-appearing WM. In the total WM, it correlated with a gradient of microstructural tissue damage measured by MTR (r s = -0.65, p = 1.0e-3). Compared to clinically stable patients, patients with disability worsening were characterized by a higher percentage of DPA+ voxels in the periventricular normal-appearing WM (p = 0.025). CONCLUSIONS: Our results demonstrate that in MS the innate immune cell activation predominates in periventricular regions and is associated with microstructural damage and disability worsening. This could result from the diffusion of proinflammatory CSF-derived factors into surrounding tissues.


Cerebral Cortex/immunology , Cerebral Cortex/pathology , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , White Matter/immunology , White Matter/pathology , Adult , Cerebral Ventricles/immunology , Cerebral Ventricles/pathology , Female , Humans , Image Interpretation, Computer-Assisted , Male , Middle Aged , Positron-Emission Tomography
17.
Int J Mol Sci ; 22(4)2021 Feb 09.
Article En | MEDLINE | ID: mdl-33572157

Platelet-rich plasma (PRP) is a biologic therapy that promotes healing responses across multiple medical fields, including the central nervous system (CNS). The efficacy of this therapy depends on several factors such as the donor's health status and age. This work aims to prove the effect of PRP on cellular models of the CNS, considering the differences between PRP from young and elderly donors. Two different PRP pools were prepared from donors 65‒85 and 20‒25 years old. The cellular and molecular composition of both PRPs were analyzed. Subsequently, the cellular response was evaluated in CNS in vitro models, studying proliferation, neurogenesis, synaptogenesis, and inflammation. While no differences in the cellular composition of PRPs were found, the molecular composition of the Young PRP showed lower levels of inflammatory molecules such as CCL-11, as well as the presence of other factors not found in Aged PRP (GDF-11). Although both PRPs had effects in terms of reducing neural progenitor cell apoptosis, stabilizing neuronal synapses, and decreasing inflammation in the microglia, the effect of the Young PRP was more pronounced. In conclusion, the molecular composition of the PRP, conditioned by the age of the donors, affects the magnitude of the biological response.


Cerebral Cortex/immunology , Inflammation Mediators/metabolism , Microglia/immunology , Platelet-Rich Plasma/immunology , Adult , Age Factors , Aged , Aged, 80 and over , Aging/immunology , Animals , Apoptosis/immunology , Cell Line, Tumor , Cell Proliferation , Cerebral Cortex/cytology , Chemokine CCL11/metabolism , Female , Humans , Male , Mice , Microglia/cytology , Neural Stem Cells/immunology , Neurogenesis/immunology , Neurons/immunology , Platelet-Rich Plasma/cytology , Platelet-Rich Plasma/metabolism , Primary Cell Culture , Rats , Synapses/immunology , Young Adult
18.
J Clin Invest ; 131(1)2021 01 04.
Article En | MEDLINE | ID: mdl-33393505

Human herpes simplex virus 1 (HSV-1) encephalitis can be caused by inborn errors of the TLR3 pathway, resulting in impairment of CNS cell-intrinsic antiviral immunity. Deficiencies of the TLR3 pathway impair cell-intrinsic immunity to vesicular stomatitis virus (VSV) and HSV-1 in fibroblasts, and to HSV-1 in cortical but not trigeminal neurons. The underlying molecular mechanism is thought to involve impaired IFN-α/ß induction by the TLR3 recognition of dsRNA viral intermediates or by-products. However, we show here that human TLR3 controls constitutive levels of IFNB mRNA and secreted bioactive IFN-ß protein, and thereby also controls constitutive mRNA levels for IFN-stimulated genes (ISGs) in fibroblasts. Tlr3-/- mouse embryonic fibroblasts also have lower basal ISG levels. Moreover, human TLR3 controls basal levels of IFN-ß secretion and ISG mRNA in induced pluripotent stem cell-derived cortical neurons. Consistently, TLR3-deficient human fibroblasts and cortical neurons are vulnerable not only to both VSV and HSV-1, but also to several other families of viruses. The mechanism by which TLR3 restricts viral growth in human fibroblasts and cortical neurons in vitro and, by inference, by which the human CNS prevents infection by HSV-1 in vivo, is therefore based on the control of early viral infection by basal IFN-ß immunity.


Cerebral Cortex/immunology , Fibroblasts/immunology , Herpesvirus 1, Human/immunology , Interferon-beta/immunology , Neurons/immunology , Toll-Like Receptor 3/immunology , Vesiculovirus/immunology , Animals , Cell Line , Cerebral Cortex/pathology , Cerebral Cortex/virology , Fibroblasts/pathology , Fibroblasts/virology , Humans , Interferon-beta/genetics , Mice , Mice, Knockout , Neurons/pathology , Neurons/virology , Toll-Like Receptor 3/genetics
19.
Behav Brain Res ; 397: 112945, 2021 01 15.
Article En | MEDLINE | ID: mdl-33022354

Alzheimer's disease (AD) is the most prevalent form of dementia with a complex pathophysiology not fully elucidated but with limited pharmacological treatment. The Usnic acid (UA) is a lichen secondary metabolite found in two enantiomeric forms: (R)-(+)-UA or (S)-(-)-UA, with antioxidant and anti-inflammatory potential. Thus, given the role of neuroinflammation and oxidative injury in the AD, this study aimed to investigate experimentally the cognitive enhancing and anti-neuroinflammatory effects of UA enantiomers. First, the interactions of UA on acetylcholinesterase (AChE) was assessed by molecular docking and its inhibitory capability on AChE was assessed in vitro. In vivo trials investigated the effects of UA enantiomers in mice exposed to Aß1-42 peptide (400 pmol/mice) intracerebroventricularly (i.c.v.). For this, mice were treated orally during 24 days with (R)-(+)-UA or (S)-(-)-UA at 25, 50, or 100 mg/kg, vehicle, or donepezil (2 mg/kg). Animals were submitted to the novel object recognized, Morris water maze, and inhibitory-avoidance task to assess the cognitive deficits. Additionally, UA antioxidant capacity and neuroinflammatory biomarkers were measured at the cortex and hippocampus from mice. Our results indicated that UA enantiomers evoked complex-receptor interaction with AChE like galantamine in silico. Also, UA enantiomers improved the learning and memory of the animals and in parallel decreased the myeloperoxidase activity and the lipid hydroperoxides (LOOH) on the cortex and hippocampus and reduced the IL-1ß levels on the hippocampus. In summary, UA restored the cognitive deficits, as well as the signs of LOOH and neuroinflammation induced by Aß1-42 administration in mice.


Acetylcholinesterase/drug effects , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/pharmacology , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Benzofurans/pharmacology , Cerebral Cortex/drug effects , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/drug therapy , Inflammation/drug therapy , Nootropic Agents/pharmacology , Peptide Fragments/pharmacology , Amyloid beta-Peptides/administration & dosage , Animals , Anti-Inflammatory Agents/administration & dosage , Antioxidants/administration & dosage , Behavior, Animal/drug effects , Benzofurans/administration & dosage , Cerebral Cortex/immunology , Disease Models, Animal , Female , Hippocampus/drug effects , Hippocampus/immunology , Inflammation/chemically induced , Injections, Intraventricular , Interleukin-1beta/drug effects , Mice , Molecular Docking Simulation , Nootropic Agents/administration & dosage , Peptide Fragments/administration & dosage
20.
Gastroenterology ; 160(4): 1315-1329.e13, 2021 03.
Article En | MEDLINE | ID: mdl-33227282

BACKGROUND & AIMS: Hepatic encephalopathy (HE) is a serious neurologic complication in patients with liver cirrhosis. Very little is known about the role of the meningeal lymphatic system in HE. We tested our hypothesis that enhancement of meningeal lymphatic drainage could decrease neuroinflammation and ameliorate HE. METHODS: A 4-week bile duct ligation model was used to develop cirrhosis with HE in rats. Brain inflammation in patients with HE was evaluated by using archived GSE41919. The motor function of rats was assessed by the rotarod test. Adeno-associated virus 8-vascular endothelial growth factor C (AAV8-VEGF-C) was injected into the cisterna magna of HE rats 1 day after surgery to induce meningeal lymphangiogenesis. RESULTS: Cirrhotic rats with HE showed significantly increased microglia activation in the middle region of the cortex (P < .001) as well as increased neuroinflammation, as indicated by significant increases in interleukin 1ß, interferon γ, tumor necrosis factor α, and ionized calcium binding adaptor molecule 1 (Iba1) expression levels in at least 1 of the 3 regions of the cortex. Motor function was also impaired in rats with HE (P < .05). Human brains of patients with cirrhosis with HE also exhibited up-regulation of proinflammatory genes (NFKB1, IbA1, TNF-α, and IL1ß) (n = 6). AAV8-VEGF-C injection significantly increased meningeal lymphangiogenesis (P = .035) and tracer dye uptake in the anterior and middle regions of the cortex (P = .006 and .003, respectively), their corresponding meninges (P = .086 and .006, respectively), and the draining lymph nodes (P = .02). Furthermore, AAV8-VEGF-C decreased microglia activation (P < .001) and neuroinflammation and ameliorated motor dysfunction (P = .024). CONCLUSIONS: Promoting meningeal lymphatic drainage and enhancing waste clearance improves HE. Manipulation of meningeal lymphangiogenesis could be a new therapeutic strategy for the treatment of HE.


Glymphatic System/pathology , Hepatic Encephalopathy/immunology , Liver Cirrhosis/complications , Motor Disorders/immunology , Vascular Endothelial Growth Factor C/metabolism , Animals , Cell Line , Cerebral Cortex/immunology , Cerebral Cortex/pathology , Cisterna Magna/immunology , Cisterna Magna/pathology , Dependovirus/genetics , Disease Models, Animal , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Glymphatic System/immunology , Hepatic Encephalopathy/pathology , Humans , Liver Cirrhosis/immunology , Lymphangiogenesis/immunology , Male , Microglia/immunology , Microglia/pathology , Motor Disorders/pathology , Rats , Vascular Endothelial Growth Factor C/genetics
...