Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Neurochem Res ; 44(12): 2832-2842, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31691882

ABSTRACT

Sepsis-associated encephalopathy (SAE) is a potentially irreversible acute cognitive dysfunction with unclear mechanism. Striatal-enriched protein tyrosine phosphatase (STEP) is a brain-specific phosphatase which normally opposes synaptic strengthening by regulating key signaling molecules involved in synaptic plasticity and neuronal function. Thus, we hypothesized that abnormal STEP signaling pathway was involved in sepsis-induced cognitive impairment evoked by lipopolysaccharides (LPS) injection. The levels of STEP, phosphorylation of GluN2B (pGluN2B), the kinases extracellular signal-regulated kinase 1/2 (pERK), cAMP-response element binding protein (CREB), synaptophysin, brain derived neurotrophic factor (BDNF), and post-synaptic density protein 95 (PSD95) in the hippocampus, prefrontal cortex, and striatum were determined at the indicated time points. In the present study, we found that STEP levels were significantly increased in the hippocampus, prefrontal cortex, and striatum following LPS injection, which might resulted from the disruption of the ubiquitin-proteasome system. Notably, a STEP inhibitor TC-2153 treatment alleviated sepsis-induced memory impairment by increasing phosphorylation of GluN2B and ERK1/2, CREB/BDNF, and PSD95. In summary, our results support the key role of STEP in sepsis-induced memory impairment in a mouse model of SAE, whereas inhibition of STEP may provide a novel therapeutic approach for this disorder and possible other neurodegenerative diseases.


Subject(s)
Memory Disorders/physiopathology , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Sepsis-Associated Encephalopathy/physiopathology , Signal Transduction/physiology , Animals , Benzothiepins/pharmacology , Brain-Derived Neurotrophic Factor/chemistry , Brain-Derived Neurotrophic Factor/metabolism , Corpus Striatum/metabolism , Cyclic AMP Response Element-Binding Protein/chemistry , Cyclic AMP Response Element-Binding Protein/metabolism , Disks Large Homolog 4 Protein/chemistry , Disks Large Homolog 4 Protein/metabolism , Hippocampus/metabolism , Lipopolysaccharides , Male , Memory/drug effects , Memory/physiology , Memory Disorders/chemically induced , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/chemistry , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/chemistry , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects , Prefrontal Cortex/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/metabolism , Sepsis-Associated Encephalopathy/chemically induced , Signal Transduction/drug effects
2.
Front Cell Neurosci ; 13: 293, 2019.
Article in English | MEDLINE | ID: mdl-31354429

ABSTRACT

Sepsis-associated encephalopathy induces cognitive dysfunction via mechanisms that commonly involve neuroinflammation and synaptic plasticity impairment of the hippocampus. The ß2-adrenoceptor (ß2-AR) is a G-protein coupled receptor that regulates immune response and synaptic plasticity, whereas its dysfunction has been implicated in various neurodegenerative diseases. Thus, we hypothesized abnormal ß2-AR signaling is involved in sepsis-induced cognitive impairment. In the present study, C57BL/6 mice were subjected to cecal ligation and puncture (CLP) to mimic the clinical human sepsis-associated encephalopathy. The levels of hippocampal ß2-AR, proinflammatory cytokines tumor necrosis factor (TNF-α), interleukin-1ß (IL-1ß), IL-6, cAMP-response element binding protein (CREB), brain derived neurotrophic factor (BDNF), post-synaptic density protein 95 (PSD95), and NMDA receptor 2 B subtypes (GluN2B) were determined at 6, 12, 24 h and 7 and 16 days after CLP. For the interventional study, mice were treated with ß2-AR agonist clenbuterol in two ways: early treatment (immediately following CLP) and delayed treatment (on the 8th day following CLP). Neurobehavioral performances were assessed by open field and fear conditioning tests. Here, we found that hippocampal ß2-AR expression was significantly decreased starting from 12 h and persisted until 16 days following CLP. Besides, sepsis mice also exhibited increasing neuroinflammation, down-regulated CREB/BDNF, decreasing PSD95 and GluN2B expression, and displayed hippocampus-dependent cognitive impairments. Notably, early clenbuterol treatment alleviated sepsis-induced cognitive deficits by polarizing microglia toward an anti-inflammatory phenotype, reducing proinflammatory cytokines including IL-1ß, TNF-α, and up-regulating CREB/BDNF, PSD95, and GluN2B. Intriguingly, delayed clenbuterol treatment also improved cognitive impairments by normalization of hippocampal CREB/BDNF, PSD95, and GluN2B. In summary, our results support the beneficial effects of both early and delayed clenbuterol treatment, which suggests that activation of ß2-AR has a translational value in sepsis-associated organ dysfunction including cognitive impairments.

3.
Int Immunopharmacol ; 70: 446-458, 2019 May.
Article in English | MEDLINE | ID: mdl-30856395

ABSTRACT

Postintensive care syndrome (PICS) is defined as a new or worsening impairment in cognition, mental health, and physical function after critical illness. However, there is still a lack of a clinically relevant animal model. Thus, development of a PICS model is essential for understanding the mechanism underlying PICS and screening treatment methods for this neuropsychiatric disorder. The purpose of this study was to establish a clinically relevant PICS model based on the two-hit concept, in which lipopolysaccharide (LPS, 3 mg/kg) injection was served as the first hit and subsequent modified chronic unpredictable stress as the second hit. In order to pharmacologically verify the proposed model of PICS, we studied the effectiveness of fluoxetine to reverse the behavioral and molecular abnormalities in this model. In the present study, body- and adrenal weight changes proved our model was effective, as reflected by body weight loss, increased adrenals weight, and a significantly increased level of plasma corticosterone. Moreover, our PICS model displayed reproducible anxiety- and depression like behavior and cognitive impairments. Neurobiological investigations revealed a significant up-regulation of the microglial marker CD68 and pro-inflammatory cytokine IL-6 in the hippocampus of stressed mice. Notably, chronic treatment with fluoxetine for three weeks reversed most of the affected parameters. In summary, we believe that we have developed a new model of PICS that is clinically relevant, which could advance the mechanism research and the development of therapeutic strategies.


Subject(s)
Critical Illness/rehabilitation , Disease Models, Animal , Hippocampus/metabolism , Lipopolysaccharides/immunology , Stress, Physiological/physiology , Subacute Care/methods , Animals , Anxiety/drug therapy , Chronic Disease , Cognition Disorders/drug therapy , Corticosterone/blood , Critical Care , Fluoxetine/therapeutic use , Humans , Interleukin-6/metabolism , Male , Mice , Mice, Inbred C57BL , Selective Serotonin Reuptake Inhibitors/therapeutic use , Syndrome
4.
Inflammation ; 42(1): 306-318, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30276509

ABSTRACT

Sepsis-associated encephalopathy (SAE) is a common complication that leads to long-term cognitive impairments and increased mortality in sepsis survivors. The mechanisms underlying this complication remain unclear and an effective intervention is lacking. Accumulating evidence suggests the nucleotide-binding domain-like receptor protein3 (NLRP3)/caspase-1 pathway is involved in several neurodegenerative diseases. Thus, we hypothesized that the NLRP3/caspase-1 pathway is involved in NLRP3-mediated pyroptosis, maturation and release of inflammatory cytokines, and cognitive deficits in SAE. We used the NLRP3 inhibitor MCC950 and the caspase-1 inhibitor Ac-YVAD-CMK to study the role of the NLRP3/caspase-1 pathway in pyroptosis and cognitive deficits in a mouse model of SAE. Mice were randomly assigned to one of six groups: sham+saline, sham+MCC950, sham+Ac-YVAD-CMK, cecal ligation and puncture (CLP)+saline, CLP+MCC950, and CLP+Ac-YVAD-CMK. Surviving mice underwent behavioral tests or had hippocampal tissues collected for histochemical analysis and biochemical assays. Our results show that CLP-induced hippocampus-dependent memory deficits are accompanied by increased NLRP3 and caspase-1 positive cells, and augmented protein levels of NLRP3, caspase-1, gasdermin-D, and pro-inflammatory cytokines in the hippocampus. In addition, administration of MCC950 or Ac-YVAD-CMK rescues cognitive deficits and ameliorates increased hippocampal NLRP3-mediated neuronal pyroptosis and pro-inflammatory cytokines. Our results suggest that the NLRP3/caspase-1 pathway-induced pyroptosis mediates cognitive deficits in a mouse model of SAE.


Subject(s)
Caspase 1/metabolism , Cognition Disorders/etiology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis , Sepsis-Associated Encephalopathy/complications , Animals , Cytokines/metabolism , Hippocampus/metabolism , Hippocampus/physiopathology , Mice
5.
Neurosci Lett ; 662: 361-367, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29102784

ABSTRACT

Sepsis impairs learning and memory function, yet marked interindividual variability exists in the degree to which sepsis compromises learning and memory function. Thus, testing resilience versus susceptibility to systemic inflammation induced-memory impairment and the underlying mechanism is needed. In the present study, we firstly used lipopolysaccharide (LPS) to induce memory impairment, and then evaluated cognitive function on days 4-7 after the first LPS challenge. Subjects' scores on both behavioral measures were subjected to a hierarchical cluster analysis, identifying two clusters that differed notably on the Y-maze and fear conditioning tests. This analysis divided these subjects into two groups, one cluster (13 of 34 subjects) displayed impaired working and associative memory, named "Susceptive". The remaining cluster (21 of 34 subjects) showed normal memory, named "Resilient". We have also included another group receiving normal saline to serve as the control group. The three groups underwent a battery of biochemical detections. In addition, we investigated whether the individual differences would disappear between the "Resilient" and "Susceptive" groups by using microglia inhibitor minocycline. We showed that as compared with the "Resilient" or control group, the "Susceptive" group was accompanied by increased tumor necrosis factor-alpha, interleukin-1beta (IL-1ß), IL-6, and biomarkers of microglia activation ionized calcium binding adaptor molecule-1 and cluster of differentiation 68. Notably, after decreasing the activation of microglia, the differences in cognitive function between the "Resilient" and "Susceptive" groups disappeared. Collectively, our study suggests that individual differences in the brain are associated with resilience versus susceptibility to LPS-induced memory impairment.


Subject(s)
Brain/physiopathology , Disease Resistance/physiology , Microglia/metabolism , Sepsis-Associated Encephalopathy/physiopathology , Animals , Brain/metabolism , Inflammation/chemically induced , Inflammation/physiopathology , Lipopolysaccharides/toxicity , Male , Memory Disorders/etiology , Memory Disorders/metabolism , Memory Disorders/physiopathology , Mice, Inbred C57BL , Sepsis-Associated Encephalopathy/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL
...