Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Int J Med Sci ; 19(1): 142-151, 2022.
Article in English | MEDLINE | ID: mdl-34975308

ABSTRACT

Background: Toll-like receptor 4 (TLR4) is implicated in neonatal hypoxic-ischemic brain damage (HIBD), but the underlying mechanism is unclear. Hypothesis: We hypothesized that TLR4 mediates brain damage after hypoxic ischemia (HI) by inducing abnormal neuroimmune responses, including activation of immune cells and expression disorder of immune factors, while early inhibition of TLR4 can alleviate the neuroimmune dysfunction. Method: Postnatal day 7 rats were randomized into control, HI, and HI+TAK-242 (TAK-242) groups. The HIBD model was developed using the Rice-Vannucci method (the left side was the ipsilateral side of HI). TAK-242 (0.5 mg/kg) was given to rat pups in the TAK-242 group at 30 min before modeling. Immunofluorescence, immunohistochemistry, and western blotting were used to determine the TLR4 expression; the number of Iba-1+, GFAP+, CD161+, MPO+, and CD3+ cells; ICAM-1 and C3a expression; and interleukin (IL)-1ß, tumor necrosis factor (TNF)-α, and IL-10 expression in the hippocampal CA1 region. Result: Significantly increased TLR4 expression was observed in the left hippocampus, and was alleviated by TAK-242. The significant increases in Iba-1+, MPO+, and CD161+ cells at 24 h and 7 days after HI and in GFAP+ and CD3+ T cells at 7 days after HI were also counteracted by TAK-242, but no significant differences were observed among groups at 24 h after HI. ICAM-1 expression increased 24 h after HI, while C3a expression decreased; TAK-242 also alleviated these changes. TNF-α and IL-1ß expression increased, while IL-10 expression decreased at 24 h and 7 days after HI; TAK-242 counteracted the increased TNF-α and IL-1ß expression at 24 h and the changes in IL-1ß and IL-10 at 7 days, but induced no significant differences in IL-10 expression at 24 h and TNF-α expression at 7 days. Conclusion: Early TLR4 inhibition can alleviate hippocampal immune dysfunction after neonatal HIBD.


Subject(s)
Hippocampus/immunology , Hypoxia-Ischemia, Brain/immunology , Toll-Like Receptor 4/physiology , Animals , Animals, Newborn , CA1 Region, Hippocampal/immunology , CA1 Region, Hippocampal/metabolism , CD3 Complex , Cytokines/metabolism , Female , Hippocampus/metabolism , Hypoxia-Ischemia, Brain/metabolism , Intercellular Adhesion Molecule-1/metabolism , Killer Cells, Natural/metabolism , Male , Models, Animal , NK Cell Lectin-Like Receptor Subfamily B , Neutrophils/enzymology , Peroxidase , Random Allocation , Rats , Sulfonamides/pharmacology , T-Lymphocytes/metabolism , Toll-Like Receptor 4/antagonists & inhibitors
2.
Behav Brain Res ; 390: 112683, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32442548

ABSTRACT

Inflammatory pain is commonly associated with cognitive impairment. However, its molecular mechanisms are poorly understood. Thus, this study was conducted to investigate the molecular mechanisms of behavioral changes associated with inflammatory pain. Briefly, 36 Wistar rats were randomly divided into two main groups: CFA group treated with 100 µL of Complete Freunds' Adjuvant (CFA) and CFA + Minocycline group treated with 100 µL of CFA+40 mg/kg/day of minocycline). After that, each group was divided into three subgroups based on different time points of the study. The pain was induced using CFA and subsequent behavioral changes (i.e., hyperalgesia and learning and spatial memory) were analyzed by the Morris Water Maze (MWM) task and Radiant Heat. Then, the cellular and molecular changes were assessed using Western Blotting, Immunohistochemistry, and Terminal deoxynucleotidyl transferase dUTP Nick End Labeling (TUNEL) techniques. Results of the study indicated that CFA-induced pain impaired spatial learning and memory functions. Studying the cellular changes showed that persistent inflammatory pain increased the microglial activity in CA1 and Dentate Gyrus (DG) regions. Furthermore, an increase was observed in the percentage of TUNEL-positive cells. Also, pro-Brain-Derived Neurotrophic Factor (BDNF)/BDNF ratio, Caspase3, and Receptor-Interacting Protein kinase 3 (RIP3) levels increased in the rats' hippocampus following induction of persistent inflammatory pain. These changes were reversed following the cessation of pain as well as the injection of minocycline. Taking together, the results of the current study for the first time revealed that an increase in the microglia dependent proBDNF/BDNF ratio following persistent inflammatory pain leads to cell death of the CA1 and DG neurons that subsequently causes a cognitive deficit in the learning and spatial memory functions.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , CA1 Region, Hippocampal , Cognitive Dysfunction/etiology , Dentate Gyrus , Inflammation/complications , Microglia , Nociceptive Pain/complications , Spatial Memory , Animals , Anti-Bacterial Agents/administration & dosage , Behavior, Animal/physiology , CA1 Region, Hippocampal/immunology , CA1 Region, Hippocampal/physiopathology , Cell Death/physiology , Cognitive Dysfunction/immunology , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/physiopathology , Dentate Gyrus/immunology , Dentate Gyrus/physiopathology , Freund's Adjuvant/administration & dosage , Inflammation/immunology , Inflammation/metabolism , Male , Microglia/immunology , Microglia/metabolism , Minocycline/administration & dosage , Nociceptive Pain/immunology , Nociceptive Pain/metabolism , Rats , Rats, Wistar , Spatial Memory/physiology
3.
Biochem Biophys Res Commun ; 518(3): 486-491, 2019 10 20.
Article in English | MEDLINE | ID: mdl-31434608

ABSTRACT

It has been shown that pro-inflammatory cytokines preferentially attenuate long-term potentiation (LTP), at the same time the effect of anti-inflammatory cytokines on synaptic plasticity has not been fully studied yet. Here we studied the effect of two anti-inflammatory cytokines - interleukin-10 (IL-10) and transforming growth factor-ß1 (TGF-ß1) on long-term potentiation. It was found that exogenously added IL-10 as well as TGF-ß1 were able to effectively facilitate LTP evoked with ether high frequency or theta burst stimulation protocols in CA1 area of hippocampus. Effectiveness of IL-10 and TGF-ß1 on LTP varied depending on the concentration of used cytokine and type of tetanic stimulation protocol used for LTP induction. Overall the positive effect of studied cytokines on LTP was associated with their ability to increase basal synaptic strength at Schaffer collateral - CA1 synapse. At the same time IL-10 and TGF-ß1 did not have any effect on short-term plasticity. Our results provide new evidence upon the modulatory effects that anti-inflammatory cytokines exert on synaptic plasticity further highlighting their potency as modulators of neuronal function.


Subject(s)
CA1 Region, Hippocampal/physiology , Interleukin-10/immunology , Long-Term Potentiation , Neuronal Plasticity , Transforming Growth Factor beta1/immunology , Animals , CA1 Region, Hippocampal/immunology , Male , Rats, Wistar
4.
Transl Psychiatry ; 8(1): 32, 2018 01 31.
Article in English | MEDLINE | ID: mdl-29382825

ABSTRACT

In an aging society, Alzheimer's disease (AD) exerts an increasingly serious health and economic burden. Current treatments provide inadequate symptomatic relief as several distinct pathological processes are thought to underlie the decline of cognitive and neural function seen in AD. This suggests that the efficacy of treatment requires a multitargeted approach. In this context, palmitoylethanolamide (PEA) provides a novel potential adjunct therapy that can be incorporated into a multitargeted treatment strategy. We used young (6-month-old) and adult (12-month-old) 3×Tg-AD mice that received ultramicronized PEA (um-PEA) for 3 months via a subcutaneous delivery system. Mice were tested with a range of cognitive and noncognitive tasks, scanned with magnetic resonance imaging/magnetic resonance spectroscopy (MRI/MRS), and neurochemical release was assessed by microdialysis. Potential neuropathological mechanisms were assessed postmortem by western blot, reverse transcription-polymerase chain reaction (RT-PCR), and immunofluorescence. Our data demonstrate that um-PEA improves learning and memory, and ameliorates both the depressive and anhedonia-like phenotype of 3×Tg-AD mice. Moreover, it reduces Aß formation, the phosphorylation of tau proteins, and promotes neuronal survival in the CA1 subregion of the hippocampus. Finally, um-PEA normalizes astrocytic function, rebalances glutamatergic transmission, and restrains neuroinflammation. The efficacy of um-PEA is particularly potent in younger mice, suggesting its potential as an early treatment. These data demonstrate that um-PEA is a novel and effective promising treatment for AD with the potential to be integrated into a multitargeted treatment strategy in combination with other drugs. Um-PEA is already registered for human use. This, in combination with our data, suggests the potential to rapidly proceed to clinical use.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/deficiency , Anti-Inflammatory Agents/pharmacology , CA1 Region, Hippocampal/drug effects , Cognitive Dysfunction/drug therapy , Ethanolamines/pharmacology , Inflammation/drug therapy , Learning/drug effects , Memory Disorders/drug therapy , Neuroprotective Agents/pharmacology , Palmitic Acids/pharmacology , tau Proteins/drug effects , Age Factors , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amides , Animals , Anti-Inflammatory Agents/administration & dosage , Behavior, Animal/drug effects , CA1 Region, Hippocampal/immunology , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/physiopathology , Cognitive Dysfunction/immunology , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/physiopathology , Disease Models, Animal , Ethanolamines/administration & dosage , Inflammation/immunology , Inflammation/metabolism , Inflammation/physiopathology , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Male , Memory Disorders/immunology , Memory Disorders/metabolism , Memory Disorders/physiopathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Microdialysis , Neuroprotective Agents/administration & dosage , Palmitic Acids/administration & dosage
5.
Mol Med Rep ; 16(2): 1627-1634, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28627606

ABSTRACT

Ischemic preconditioning (IPC) is induced by exposure to brief durations of transient ischemia, which results in ischemic tolerance to a subsequent longer or lethal period of ischemia. In the present study, the effects of IPC (2 min of transient cerebral ischemia) were examined on immunoreactivity of platelet­derived growth factor (PDGF)­BB and on neuroprotection in the gerbil hippocampal CA1 region following lethal transient cerebral ischemia (LTCI; 5 min of transient cerebral ischemia). IPC was subjected to a 2­min sublethal ischemia and a LTCI was given 5­min transient ischemia. The animals in all of the groups were given recovery times of 1, 2 and 5 days and change in PDGF­BB immunoreactivity was examined as was the neuronal damage/death in the hippocampus induced by LTCI. LTCI induced a significant loss of pyramidal neurons in the hippocampal CA1 region 5 days after LTCI, and significantly decreased PDGF­BB immunoreactivity in the CA1 pyramidal neurons from day 1 after LTCI. Conversely, IPC effectively protected the CA1 pyramidal neurons from LTCI and increased PDGF­BB immunoreactivity in the CA1 pyramidal neurons post­LTCI. In conclusion, the results demonstrated that LTCI significantly altered PDGF­BB immunoreactivity in pyramidal neurons in the hippocampal CA1 region, whereas IPC increased the immunoreactivity. These findings indicated that PDGF­BB may be associated with IPC­mediated neuroprotection.


Subject(s)
CA1 Region, Hippocampal/metabolism , Gerbillinae/metabolism , Ischemic Attack, Transient/metabolism , Proto-Oncogene Proteins c-sis/metabolism , Animals , Becaplermin , CA1 Region, Hippocampal/immunology , CA1 Region, Hippocampal/pathology , Cell Death/physiology , Hippocampus/metabolism , Hippocampus/pathology , Immunohistochemistry , Ischemic Attack, Transient/pathology , Ischemic Preconditioning/methods , Locomotion , Male , Neuroprotection , Proto-Oncogene Proteins c-sis/immunology , Pyramidal Cells/immunology , Pyramidal Cells/metabolism , Pyramidal Cells/pathology
6.
Mol Med Rep ; 15(6): 4148-4154, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28487994

ABSTRACT

Activated M1 microglia secrete proinflammatory cytokines into damaged brain areas. The present study examined activated M1 microglial morphology and expression in the hippocampal Cornu Ammonis (CA) 1 region, which is vulnerable to transient ischemia. Transient cerebral ischemia was performed for 5 min in gerbils, and neuronal death in the CA1 region following transient cerebral ischemia was confirmed using cresyl violet staining, neuronal nuclear antigen immunohistochemistry and Fluoro­Jade B histofluorescent staining. In addition, CA1 regions were stained for cluster of differentiation (CD) 74, a marker for activated M1 microglia and a ligand for macrophage migration inhibitory factor In sham­operated animals, no CD74 immunoreactivity was observed in the hippocampal CA1 region. CD74 immunoreactivity was not observed in the hippocampal CA1 region until 3 days post­ischemic insult; however, elevated CD74 immunoreactivity was detected in the CA1 region from 5 days post­ischemia. Double immunofluorescence staining for CD74 and ionized calcium­binding adapter molecule 1, a marker for M1 microglial cells, confirmed the expression of CD74 on this microglial subtype. These results indicated that M1 microglia are activated late in the hippocampal CA1 region following ischemic stroke. Therefore, optimizing the timing of therapeutic intervention may reduce activated M1 microglial-induced neuronal damage.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/metabolism , CA1 Region, Hippocampal/metabolism , Histocompatibility Antigens Class II/metabolism , Ischemic Attack, Transient/metabolism , Microglia/metabolism , Animals , CA1 Region, Hippocampal/immunology , CA1 Region, Hippocampal/pathology , Cell Death , Disease Models, Animal , Gerbillinae , Immunohistochemistry , Ischemic Attack, Transient/immunology , Ischemic Attack, Transient/pathology , Male , Microglia/immunology , Protein Binding , Protein Transport , Pyramidal Cells/metabolism , Pyramidal Cells/pathology
7.
Behav Brain Res ; 322(Pt A): 167-176, 2017 03 30.
Article in English | MEDLINE | ID: mdl-28115220

ABSTRACT

Several animal studies demonstrated that the volatile anesthetic isoflurane could influence the blood-brain barrier (BBB) integrity, which involved the cognitive impairment. Increasing evidence has also shown that the receptor for advanced glycation end-products (RAGE) played a major role in maintaining the integrity of BBB. The present study aimed to determine whether the RAGE-specific antibody protects against BBB disruption and cognitive impairment induced by isoflurane exposure in aged rats. 108 aged rats were randomly divided into four groups: (1) control group (Control); (2) 4h of 2% isoflurane exposure group (ISO); (3) RAGE antibody (20µL, 2.5µg/µL) treated+4h of 2% isoflurane exposure group (anti-RAGE+ISO); (4) RAGE antibody (20µL, 2.5µg/µL) treated group (anti-RAGE). The isoflurane anesthesia resulted in the upregulation of hippocampal RAGE expression, disruption of BBB integrity, neuroinflammation, and beta-amyloid (Aß) accumulation in aged rats. In addition, significant cognitive deficits in the Morris water maze test was also observed. The antibody pretreatment resulted in significant improvements in BBB integrity. Furthermore, the expression of RAGE and proinflammatory mediators, as well as, Aß accumulation were attenuated. Moreover, the antibody administration attenuated the isoflurane-induced cognitive impairment in aged rats. These results demonstrate that RAGE signaling is involved in BBB damage after isoflurane exposure. Thus, the RAGE antibody represents a novel therapeutic intervention to prevent isoflurane-induced cognitive impairment.


Subject(s)
Antibodies/administration & dosage , Blood-Brain Barrier/drug effects , Cognitive Dysfunction/drug therapy , Isoflurane/toxicity , Nootropic Agents/administration & dosage , Receptor for Advanced Glycation End Products/antagonists & inhibitors , Receptor for Advanced Glycation End Products/immunology , Aging/drug effects , Aging/metabolism , Aging/pathology , Aging/psychology , Amyloid beta-Peptides/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/immunology , CA1 Region, Hippocampal/pathology , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Disease Models, Animal , Male , Random Allocation , Rats, Sprague-Dawley , Receptor for Advanced Glycation End Products/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
Oxid Med Cell Longev ; 2016: 3974648, 2016.
Article in English | MEDLINE | ID: mdl-28003864

ABSTRACT

We investigated possible interaction between an arbovirus infection and the ME7 induced mice prion disease. C57BL/6, females, 6-week-old, were submitted to a bilateral intrahippocampal injection of ME7 prion strain (ME7) or normal brain homogenate (NBH). After injections, animals were organized into two groups: NBH (n = 26) and ME7 (n = 29). At 15th week after injections (wpi), animals were challenged intranasally with a suspension of Piry arbovirus 0.001% or with NBH. Behavioral changes in ME7 animals appeared in burrowing activity at 14 wpi. Hyperactivity on open field test, errors on rod bridge, and time reduction in inverted screen were detected at 15th, 19th, and 20th wpi respectively. Burrowing was more sensitive to earlier hippocampus dysfunction. However, Piry-infection did not significantly affect the already ongoing burrowing decline in the ME7-treated mice. After behavioral tests, brains were processed for IBA1, protease-resistant form of PrP, and Piry virus antigens. Although virus infection in isolation did not change the number of microglia in CA1, virus infection in prion diseased mice (at 17th wpi) induced changes in number and morphology of microglia in a laminar-dependent way. We suggest that virus infection exacerbates microglial inflammatory response to a greater degree in prion-infected mice, and this is not necessarily correlated with hippocampal-dependent behavioral deficits.


Subject(s)
Arboviruses/pathogenicity , CA1 Region, Hippocampal/virology , Coinfection , Encephalitis, Arbovirus/complications , Microglia/virology , Prion Diseases/complications , Animals , Antigens, Viral/immunology , Arboviruses/immunology , Behavior, Animal , CA1 Region, Hippocampal/immunology , CA1 Region, Hippocampal/pathology , CA1 Region, Hippocampal/physiopathology , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Disease Progression , Encephalitis, Arbovirus/immunology , Encephalitis, Arbovirus/pathology , Encephalitis, Arbovirus/psychology , Female , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Microglia/immunology , Microglia/pathology , Motor Activity , Nerve Degeneration , Prion Diseases/pathology , Prion Diseases/psychology , Time Factors
9.
Ann Neurol ; 80(3): 388-400, 2016 09.
Article in English | MEDLINE | ID: mdl-27399303

ABSTRACT

OBJECTIVE: To demonstrate that ephrin-B2 (the ligand of EphB2 receptor) antagonizes the pathogenic effects of patients' N-methyl-D-aspartate receptor (NMDAR) antibodies on memory and synaptic plasticity. METHODS: One hundred twenty-two C57BL/6J mice infused with cerebrospinal fluid (CSF) from patients with anti-NMDAR encephalitis or controls, with or without ephrin-B2, were investigated. CSF was infused through ventricular catheters connected to subcutaneous osmotic pumps over 14 days. Memory, behavioral tasks, locomotor activity, presence of human antibodies specifically bound to hippocampal NMDAR, and antibody effects on the density of cell-surface and synaptic NMDAR and EphB2 were examined at different time points using reported techniques. Short- and long-term synaptic plasticity were determined in acute brain sections; the Schaffer collateral pathway was stimulated and the field excitatory postsynaptic potentials were recorded in the CA1 region of the hippocampus. RESULTS: Mice infused with patients' CSF, but not control CSF, developed progressive memory deficit and depressive-like behavior along with deposits of NMDAR antibodies in the hippocampus. These findings were associated with a decrease of the density of cell-surface and synaptic NMDAR and EphB2, and marked impairment of long-term synaptic plasticity without altering short-term plasticity. Administration of ephrin-B2 prevented the pathogenic effects of the antibodies in all the investigated paradigms assessing memory, depressive-like behavior, density of cell-surface and synaptic NMDAR and EphB2, and long-term synaptic plasticity. INTERPRETATION: Administration of ephrin-B2 prevents the pathogenic effects of anti-NMDAR encephalitis antibodies on memory and behavior, levels of cell-surface NMDAR, and synaptic plasticity. These findings reveal a strategy beyond immunotherapy to antagonize patients' antibody effects. Ann Neurol 2016;80:388-400.


Subject(s)
Anti-N-Methyl-D-Aspartate Receptor Encephalitis/drug therapy , Antibodies/drug effects , CA1 Region, Hippocampal/drug effects , Depression/prevention & control , Ephrin-B2/pharmacology , Memory Disorders/prevention & control , Neuronal Plasticity/drug effects , Animals , Anti-N-Methyl-D-Aspartate Receptor Encephalitis/cerebrospinal fluid , Anti-N-Methyl-D-Aspartate Receptor Encephalitis/immunology , Antibodies/immunology , Behavior, Animal , CA1 Region, Hippocampal/immunology , Depression/etiology , Depression/immunology , Disease Models, Animal , Humans , Male , Memory Disorders/etiology , Memory Disorders/immunology , Mice , Mice, Inbred C57BL , Neuronal Plasticity/immunology , Receptor, EphB2
10.
Bull Exp Biol Med ; 161(2): 316-9, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27383167

ABSTRACT

We evaluated the effect of hippocampal injection of lentiviral particles p156-CMV-EGFP on behavior, learning, and microglial Iba1(+) cells activation in mice. Testing in the open field and elevated plus-maze revealed higher anxiety levels in lentiviral-injected mice in comparison with animals injected with vehicle. At the same time, lentivirus injection did not change learning and memory of mice in the hippocampal-dependent fear conditioning task. Microglia density in lentivirus-injected mice was significantly higher than in vehicle-injected mice. Thus, hippocampal injection of lentiviral particles with minimum content of transgenes produced evident inflammation process, changed anxiety level of experimental animals, but had no effect on hippocampal-dependent learning and memory.


Subject(s)
CA1 Region, Hippocampal/virology , Dentate Gyrus/virology , Lentivirus/immunology , Neurons/virology , Transduction, Genetic , Animals , CA1 Region, Hippocampal/immunology , Cognition , Dentate Gyrus/immunology , Lentivirus/genetics , Male , Maze Learning , Mice, Inbred C57BL , Neurons/immunology
11.
Science ; 352(6286): 712-716, 2016 May 06.
Article in English | MEDLINE | ID: mdl-27033548

ABSTRACT

Synapse loss in Alzheimer's disease (AD) correlates with cognitive decline. Involvement of microglia and complement in AD has been attributed to neuroinflammation, prominent late in disease. Here we show in mouse models that complement and microglia mediate synaptic loss early in AD. C1q, the initiating protein of the classical complement cascade, is increased and associated with synapses before overt plaque deposition. Inhibition of C1q, C3, or the microglial complement receptor CR3 reduces the number of phagocytic microglia, as well as the extent of early synapse loss. C1q is necessary for the toxic effects of soluble ß-amyloid (Aß) oligomers on synapses and hippocampal long-term potentiation. Finally, microglia in adult brains engulf synaptic material in a CR3-dependent process when exposed to soluble Aß oligomers. Together, these findings suggest that the complement-dependent pathway and microglia that prune excess synapses in development are inappropriately activated and mediate synapse loss in AD.


Subject(s)
Alzheimer Disease/immunology , Alzheimer Disease/pathology , Complement C1q/immunology , Microglia/immunology , Phagocytosis/immunology , Synapses/immunology , Synapses/pathology , Amyloid beta-Peptides/immunology , Animals , CA1 Region, Hippocampal/immunology , CA1 Region, Hippocampal/pathology , CA1 Region, Hippocampal/physiopathology , Cognition Disorders/immunology , Cognition Disorders/pathology , Complement C1q/genetics , Complement Pathway, Classical/immunology , Disease Models, Animal , Disks Large Homolog 4 Protein , Guanylate Kinases/immunology , Long-Term Potentiation , Macrophage-1 Antigen/genetics , Macrophage-1 Antigen/immunology , Membrane Proteins/immunology , Mice , Mice, Knockout , Plaque, Amyloid/immunology , Synaptophysin/immunology , Up-Regulation
12.
Int J Environ Res Public Health ; 11(11): 11438-49, 2014 Nov 05.
Article in English | MEDLINE | ID: mdl-25380458

ABSTRACT

Crotamine is one of the main constituents of the venom of the South American rattlesnake Crotalus durissus terrificus. Here we sought to investigate the inflammatory and toxicological effects induced by the intrahippocampal administration of crotamine isolated from Crotalus whole venom. Adult rats received an intrahippocampal infusion of crotamine or vehicle and were euthanized 24 h or 21 days after infusion. Plasma and brain tissue were collected for biochemical analysis. Complete blood count, creatinine, urea, glutamic oxaloacetic transaminase (GOT), glutamic pyruvic transaminase (GPT), creatine-kinase (CK), creatine kinase-muscle B (CK-MB) and oxidative parameters (assessed by DNA damage and micronucleus frequency in leukocytes, lipid peroxidation and protein carbonyls in plasma and brain) were quantified. Unpaired and paired t-tests were used for comparisons between saline and crotamine groups, and within groups (24 h vs. 21 days), respectively. After 24 h crotamine infusion promoted an increase of urea, GOT, GPT, CK, and platelets values (p ≤ 0.01), while red blood cells, hematocrit and leukocytes values decreased (p ≤ 0.01). Additionally, 21 days after infusion crotamine group showed increased creatinine, leukocytes, TBARS (plasma and brain), carbonyl (plasma and brain) and micronucleus compared to the saline-group (p ≤ 0.01). Our findings show that crotamine infusion alter hematological parameters and cardiac markers, as well as oxidative parameters, not only in the brain, but also in the blood, indicating a systemic pro-inflammatory and toxicological activity. A further scientific attempt in terms of preserving the beneficial activity over toxicity is required.


Subject(s)
Brain/drug effects , CA1 Region, Hippocampal/drug effects , Crotalid Venoms/pharmacology , Crotalus , Animals , Blood Cell Count , Blood Chemical Analysis , CA1 Region, Hippocampal/immunology , Crotalid Venoms/administration & dosage , Crotalid Venoms/adverse effects , Infusions, Intraventricular , Male , Rats , Rats, Wistar
13.
Brain Behav Immun ; 41: 59-64, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24886967

ABSTRACT

Postmortem and positron emission tomography studies have indicated the pathophysiological involvement of microglial cells in schizophrenia. We hypothesized that the microglial production of quinolinic acid (QUIN), an endogenous N-methyl-d-aspartate receptor (NMDAR) agonist, may be linked to the previously described glutamatergic deficits in the hippocampus of schizophrenia patients. We performed a semi-quantitative assessment of QUIN-immunoreactive microglial cells in schizophrenia patients and matched controls in the CA1, CA2/3, and dentate gyrus (DG) area of the posterior hippocampal formation. Complementary immunostaining of the commonly used microglial surface marker HLA-DR was performed in adjacent histological sections. Fewer QUIN-immunoreactive microglial cells were observed in the CA1 hippocampal subregion of schizophrenia patients compared to controls (left p=0.028, right p=0.018). No significant diagnosis-dependent changes were observed in the CA2/3 and DG regions. These results were controlled for potential confounds by age, duration of disease, autolysis time, psychotropic medication, and hippocampal volume. No diagnosis-related differences were observed for the overall density of microglial cells (HLA-DR expression). Our findings suggest that reduced microglial QUIN content in the hippocampal CA1 region is associated with schizophrenia. We hypothesize that this association may contribute to impaired glutamatergic neurotransmission in the hippocampus of schizophrenia patients.


Subject(s)
CA1 Region, Hippocampal/chemistry , Excitatory Amino Acid Agonists/analysis , Microglia/chemistry , Quinolinic Acid/analysis , Receptors, N-Methyl-D-Aspartate/agonists , Schizophrenia/metabolism , Adult , CA1 Region, Hippocampal/immunology , CA1 Region, Hippocampal/pathology , Cell Count , Female , Glutamic Acid/physiology , HLA-DR Antigens/analysis , Hippocampus/chemistry , Hippocampus/immunology , Hippocampus/pathology , Humans , Male , Microglia/immunology , Middle Aged , Neuroimmunomodulation/physiology , Organ Specificity , Schizophrenia/immunology , Schizophrenia/pathology , Synaptic Transmission
14.
Glia ; 62(7): 1041-52, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24648328

ABSTRACT

The expansion of the microglial population is one of the hallmarks of numerous brain disorders. The addition of circulating progenitors to the pool of brain macrophages can contribute to the progression of brain disease and needs to be precisely defined to better understand the evolution of the glial and inflammatory reactions in the brain. We have analyzed the degree of infiltration/recruitment of circulating monocytes to the microglial pool, in a prion disease model of chronic neurodegeneration. Our results indicate a minimal/absent level of CCR2-dependent recruitment of circulating monocytes, local proliferation of microglia is the main driving force maintaining the amplification of the population. A deficiency in CCR2, and thus the absence of recruitment of circulating monocytes, does not impact microglial dynamics, the inflammatory profile or the temporal behavioral course of prion disease. However, the lack of CCR2 has unexpected effects including the failure to recruit perivascular macrophages in diseased but not healthy CNS and a small reduction in microglia proliferation. These data define the composition of the CNS-resident macrophage populations in prion disease and will help to understand the dynamics of the CNS innate immune response during chronic neurodegeneration.


Subject(s)
CA1 Region, Hippocampal/immunology , Macrophages/immunology , Microglia/immunology , Monocytes/immunology , Prion Diseases/immunology , Receptors, CCR2/metabolism , Animals , Antigens, CD34/metabolism , Behavior, Animal/physiology , CA1 Region, Hippocampal/blood supply , Cell Proliferation , Chronic Disease , Disease Models, Animal , Disease Progression , Female , Mice, Inbred C57BL , Mice, Knockout , Neurodegenerative Diseases/immunology , Receptors, CCR2/genetics
15.
PLoS One ; 8(8): e70927, 2013.
Article in English | MEDLINE | ID: mdl-23940669

ABSTRACT

OBJECTIVE: Microglia are among the first immune cells to respond to ischemic insults. Triggering of this inflammatory response may involve the microglial purinergic GPCR, P2Y12, activation via extracellular release of nucleotides from injured cells. It is also the inhibitory target of the widely used antiplatelet drug, clopidogrel. Thus, inhibiting this GPCR in microglia should inhibit microglial mediated neurotoxicity following ischemic brain injury. METHODS: Experimental cerebral ischemia was induced, in vitro with oxygen-glucose deprivation (OGD), or in vivo via bilateral common carotid artery occlusion (BCCAO). Genetic knock-down in vitro via siRNA, or in vivo P2Y12 transgenic mice (P2Y12-/- or P2Y12+/-), or in vivo treatment with clopidogrel, were used to manipulate the receptor. Neuron death, microglial activation, and microglial migration were assessed. RESULTS: The addition of microglia to neuron-astrocyte cultures increases neurotoxicity following OGD, which is mitigated by microglial P2Y12 deficiency (P<0.05). Wildtype microglia form clusters around these neurons following injury, which is also prevented in P2Y12 deficient microglia (P<0.01). P2Y12 knock-out microglia migrated less than WT controls in response to OGD-conditioned neuronal supernatant. P2Y12 (+/-) or clopidogrel treated mice subjected to global cerebral ischemia suffered less neuronal injury (P<0.01, P<0.001) compared to wild-type littermates or placebo treated controls. There were also fewer microglia surrounding areas of injury, and less activation of the pro-inflammatory transcription factor, nuclear factor Kappa B (NFkB). INTERPRETATION: P2Y12 participates in ischemia related inflammation by mediating microglial migration and potentiation of neurotoxicity. These data also suggest an additional anti-inflammatory, neuroprotective benefit of clopidogrel.


Subject(s)
Brain Ischemia/metabolism , Microglia/physiology , Receptors, Purinergic P2Y12/deficiency , Animals , Apoptosis , Astrocytes/physiology , Brain Ischemia/immunology , Brain Ischemia/pathology , CA1 Region, Hippocampal/immunology , CA1 Region, Hippocampal/pathology , Cell Hypoxia , Cell Movement , Cell Survival , Cells, Cultured , Clopidogrel , Coculture Techniques , Gene Knockdown Techniques , Glucose/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Neurons/physiology , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y12/genetics , Ticlopidine/analogs & derivatives , Ticlopidine/pharmacology
16.
Cell Death Dis ; 4: e667, 2013 Jun 13.
Article in English | MEDLINE | ID: mdl-23764848

ABSTRACT

We previously demonstrated that overexpression of RanBP9 led to enhanced Aß generation in a variety of cell lines and primary neuronal cultures, and subsequently, we confirmed increased amyloid plaque burden in a mouse model of Alzheimer's disease (AD). In the present study, we found striking reduction of spinophilin protein levels when RanBP9 is overexpressed. At 12 months of age, we found spinophilin levels reduced by 70% (P<0.001) in the cortex of APΔE9/RanBP9 mice compared with that in wild-type (WT) controls. In the hippocampus, the spinophilin levels were reduced by 45% (P<0.01) in the APΔE9/RanBP9 mice. Spinophilin immunoreactivity was also reduced by 22% (P<0.01) and 12% (P<0.05) in the cortex of APΔE9/RanBP9 and APΔE9 mice, respectively. In the hippocampus, the reductions were 27% (P<0.001) and 14% (P<0.001) in the APΔE9/RanBP9 and APΔE9 mice, respectively. However, in the cerebellum, spinophilin levels were not altered in either APΔE9 or APΔE9/RanBP9 mice. Additionally, synaptosomal functional integrity was reduced under basal conditions by 39% (P<0.001) in the APΔE9/RanBP9 mice and ~23% (P<0.001) in the APΔE9 mice compared with that in WT controls. Under ATP- and KCl-stimulated conditions, we observed higher mitochondrial activity in the WT and APΔE9 mice, but lower in the APΔE9/RanBP9 mice. Significantly, we confirmed the inverse relationship between RanBP9-N60 and spinophilin in the synaptosomes of Alzheimer's brains. More importantly, both APΔE9 and APΔE9/RanBP9 mice showed impaired learning and memory skills compared to WT controls. These data suggest that RanBP9 might play a crucial role in the loss of spines and synapses in AD.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Alzheimer Disease/metabolism , Cytoskeletal Proteins/physiology , Microfilament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/physiology , Synaptosomes/metabolism , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Animals , CA1 Region, Hippocampal/immunology , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , Cerebellum/metabolism , Cerebellum/pathology , Cerebral Cortex/immunology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Maze Learning , Mice , Mice, Transgenic , Microglia/immunology , Microglia/metabolism , Mitochondria/metabolism , Synaptosomes/physiology
17.
J Neurol Sci ; 326(1-2): 40-7, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23357314

ABSTRACT

It has been reported that the young were much more resistant to transient cerebral ischemia than in the adult. In the present study, we examined that about 90% of CA1 pyramidal cells in the adult gerbil hippocampus died at 4days after ischemia-reperfusion; however, in the young hippocampus, about 56% of them died at 7days after ischemia-reperfusion. We compared immunoreactivities and levels of calcium binding proteins (CBPs), such as calbindin 28k (CB-D28k), calretinin (CR) and parvalbumin (PV). The immunoreactivities and protein levels of all the CBPs in the young sham were higher than those in the adult sham. In the adult, the immunoreactivities and protein levels of all the CBPs were markedly decreased at 4days after ischemia-reperfusion, however, in the young, they were apparently maintained. At 7days after ischemia-reperfusion, they were decreased in the young, however, they were much higher than those in the adult. In brief, the immunoreactivities and levels of CBPs were not decreased in the ischemic CA1 region of the young 4days after transient cerebral ischemia. This finding indicates that the longer maintenance of CBPs may contribute to a less and more delayed neuronal death/damage in the young.


Subject(s)
CA1 Region, Hippocampal/immunology , Calcium-Binding Proteins/immunology , Calcium-Binding Proteins/metabolism , Ischemic Attack, Transient/immunology , Age Factors , Animals , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , Gerbillinae , Ischemic Attack, Transient/metabolism , Ischemic Attack, Transient/pathology , Male , Protein Binding/immunology
18.
J Neurol Sci ; 325(1-2): 108-14, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23287813

ABSTRACT

Young gerbils are much more resistant to transient cerebral ischemia than the adult. In the present study, we observed that about 90% of CA1 pyramidal cells in the adult hippocampus died 4days post-ischemia; however, about 56% of them in the young hippocampus died at 7days post-ischemia. To compare excitotoxicity between them, we carried out immunoreactivities of NMDA receptor 1 (NMDAR1) and NMDAR2A/B in the hippocampal CA1 region (CA1) induced by 5min of transient cerebral ischemia in the young and adult gerbils. Their immunoreactivities and protein levels in the young sham-group were much lower than those in the adult sham-group. Four days after ischemia-reperfusion, they were significantly decreased in the adult ischemia-group; however, in the young ischemia-group, they were much higher than those in the adult. Seven days after ischemia-reperfusion, NMDAR1 immunoreactivity and its level in the young were much higher than those in the adult; NMDAR2A/B immunoreactivity and its level in the young were lower than in the adult. In brief, the immunoreactivities of NMDARs were not decreased in the ischemic CA1 region of the young 4days after transient cerebral ischemia. This finding indicates that longer maintenance of NMDARs may contribute to less and more delayed neuronal death/damage in the young CA1.


Subject(s)
Aging/metabolism , CA1 Region, Hippocampal/metabolism , Ischemic Attack, Transient/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Aging/immunology , Aging/pathology , Animals , CA1 Region, Hippocampal/immunology , CA1 Region, Hippocampal/pathology , Gerbillinae , Ischemic Attack, Transient/immunology , Ischemic Attack, Transient/pathology , Male
19.
Br J Nutr ; 109(1): 50-6, 2013 Jan 14.
Article in English | MEDLINE | ID: mdl-23068715

ABSTRACT

Proinflammatory cytokines play a central role in depression-like behaviour and apoptosis in the limbic system after myocardial infarction (MI). A PUFA n-3 diet or the combination of Lactobacillus helveticus R0052 and Bifidobacterium longum R0175 probiotics, when given before the ischaemic period, reduce circulating proinflammatory cytokines as well as apoptosis in the limbic system. The present study was designed to determine if the same nutritional interventions maintain their beneficial effects when started after the onset of the reperfusion period and attenuate depression-like behaviour observed after MI. MI was induced by the occlusion of the left anterior descending coronary artery for 40 min in rats. After the onset of reperfusion, animals were fed with a high- or low-PUFA n-3 diet, combined or not with one billion live bacteria of L. helveticus and B. longum. At 3 d post-MI, caspase-3 enzymatic activities and terminal 2'-deoxyuridine, 5'-triphosphate (dUTP) nick-end labelling (TUNEL)-positive cells were decreased in the CA1, dentate gyrus (DG) and amygdala with the high-PUFA n-3 diet, as compared to the three other diets. Probiotics attenuated caspase-3 activity and TUNEL-positive cells in the DG and the medial amygdala. At 2 weeks post-MI, depression-like behaviour was observed in the low-PUFA n-3 diet without probiotics-group, and this behaviour was attenuated with the high-PUFA n-3 diet or/and probiotics. These results indicate that a high-PUFA n-3 diet or the administration of probiotics, starting after the onset of reperfusion, are beneficial to attenuate apoptosis in the limbic system and post-MI depression in the rat.


Subject(s)
Depression/prevention & control , Fatty Acids, Omega-3/therapeutic use , Lactobacillus/immunology , Limbic System/metabolism , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/prevention & control , Probiotics/therapeutic use , Amygdala/immunology , Amygdala/metabolism , Amygdala/pathology , Animals , Apoptosis , Behavior, Animal , CA1 Region, Hippocampal/immunology , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , Cytokines/blood , Dentate Gyrus/immunology , Dentate Gyrus/metabolism , Dentate Gyrus/pathology , Depression/etiology , Lactobacillus/growth & development , Limbic System/immunology , Limbic System/pathology , Male , Myocardial Infarction/psychology , Myocardial Reperfusion Injury/immunology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Neurons/immunology , Neurons/metabolism , Neurons/pathology , Random Allocation , Rats , Rats, Sprague-Dawley , Time Factors
20.
Exp Neurol ; 232(2): 329-32, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21945008

ABSTRACT

The aim of this study was to investigate the comparative effects of transforming growth factor ß1 (TGF-ß1) and interleukin-10 (IL-10) on the repeated brief hypoxia-induced alterations in the activity of hippocampal slice CA1 pyramidal neurons. The method of field potentials measurement in CA1 region of hippocampal slices was used. The principal results of our work are summarized as follow. 1. TGF-ß1 reduces the depressive effect of brief hypoxia on the population spike amplitude more effectively than IL-10. 2. During TGF-ß1 exposure (in contrast to IL-10), three 3-min hypoxic episodes do not induce the rapid hypoxic preconditioning. 3. TGF-ß1 and IL-10 equally abolish posthypoxic hyperexcitability induced by repeated brief episodes of hypoxia in CA1 pyramidal neurons. These findings indicated that TGF-ß1 and IL-10 are able to evoke anti-hypoxic effect and abolish the development of posthypoxic hyperexcitability induced by repeated brief hypoxic episodes in hippocampal CA1 pyramidal neurons. Our results also demonstrated that TGF-ß1 reduced the effectiveness of hypoxia to depress neuronal activity more effectively than IL-10. We suggest that the present findings allow to explain the certain neuroprotective mechanisms of IL-10 and TGF-beta1 in the early phase of hypoxia and indicate that a therapeutic anti-inflammatory approach using these substances can provide neuroprotection in the brain hypoxic conditions.


Subject(s)
Anti-Inflammatory Agents/pharmacology , CA1 Region, Hippocampal , Hypoxia, Brain , Interleukin-10/pharmacology , Transforming Growth Factor beta1/pharmacology , Animals , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/immunology , CA1 Region, Hippocampal/physiopathology , Hypoxia, Brain/drug therapy , Hypoxia, Brain/immunology , Hypoxia, Brain/physiopathology , Ischemic Preconditioning/methods , Male , Membrane Potentials/drug effects , Organ Culture Techniques , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...