Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
2.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(6): 481-487, 2024 Jun.
Article in Chinese | MEDLINE | ID: mdl-38952086

ABSTRACT

Objective To elucidate the role of chaperone-mediated autophagy (CMA) in alleviating emotional dysfunction in mice with sepsis-associated encephalopathy (SAE). Methods The SAE mouse model was established by cecal ligation and perforation (CLP). The severity of sepsis was assessed using the sepsis severity score (MSS). Emotional function in SAE mice was assessed by the open-field test and elevated plus-maze. The expression levels of cognitive heat shock cognate protein 70 (HSC70), lysosomal-associated membrane protein 2A (LAMP2A) and high mobility group box 1 protein B1 (HMGB1) were detected using Western blotting. Co-localization of LAMP2A in the hippocampal neurons was observed by immunofluorescence. The release of inflammatory factors interleukin 6 (IL-6) and tumor necrosis factor α (TNF-α) was measured using ELISA. Following 12 hours post-CLP, mice were orally administered resveratrol at a dose of 30 mg/kg once daily until day 14. Results The mortality rate of CLP mice was 45.83% 24 days post CLP, and all surviving mice exhibited emotional disturbances. 24 hours after CLP, a significant decrease in HSC70 and LAMP2A expression in hippocampal neurons was observed, indicating impaired CMA activity. Meanwhile, HMGB1 and inflammatory cytokines (IL-6 and TNF-α) levels increased. After resveratrol treatment, an increase of HSC70 and LAMP2A expression, and a decrease of HMGB1 expression and inflammatory cytokine release were observed, suggesting enhanced CMA activity and reduced neuroinflammation. Behavioral tests showed that emotional dysfunction was improved in SAE mice after resveratrol treatment. Conclusion CMA activity of hippocampal neurons in SAE mice is significantly reduced, leading to emotional dysfunction. Resveratrol can alleviate neuroinflammation and emotional dysfunction in SAE mice by promoting CMA and inhibiting the expression of HMGB1 and the release of inflammatory factors.


Subject(s)
Chaperone-Mediated Autophagy , HMGB1 Protein , Resveratrol , Sepsis-Associated Encephalopathy , Animals , Mice , Sepsis-Associated Encephalopathy/drug therapy , Sepsis-Associated Encephalopathy/physiopathology , Sepsis-Associated Encephalopathy/metabolism , Male , Resveratrol/pharmacology , HMGB1 Protein/metabolism , Chaperone-Mediated Autophagy/drug effects , Tumor Necrosis Factor-alpha/metabolism , Lysosomal-Associated Membrane Protein 2/metabolism , Lysosomal-Associated Membrane Protein 2/genetics , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/metabolism , Hippocampus/metabolism , Hippocampus/drug effects , Interleukin-6/metabolism , Stilbenes/pharmacology , HSC70 Heat-Shock Proteins/metabolism , Sepsis/complications , Sepsis/drug therapy , Sepsis/metabolism , Sepsis/physiopathology , Mice, Inbred C57BL , Disease Models, Animal
3.
Sheng Li Xue Bao ; 76(2): 289-300, 2024 Apr 25.
Article in Chinese | MEDLINE | ID: mdl-38658377

ABSTRACT

Sepsis-associated encephalopathy (SAE) refers to diffuse brain dysfunction caused by sepsis, which is characterized by decreased attention, directional impairment, being prone to irritation, and in severe cases the patient will experience drowsiness and coma. The pathogenesis of SAE mainly includes neuroinflammation, damage of blood-brain barrier, cerebral vascular dysfunction, and neurometabolic changes, among which neuroinflammation is the core pathological process. Microglia are considered to be important immune cells of the central nervous system and play an important role in neuroinflammation. This article systematically describes the role of microglia in the development of SAE, and discusses the phenotype and related signaling pathways of microglia, in order to clarify the role of microglia in SAE and provide a theoretical basis for clinical treatment of SAE.


Subject(s)
Microglia , Sepsis-Associated Encephalopathy , Humans , Sepsis-Associated Encephalopathy/physiopathology , Sepsis-Associated Encephalopathy/metabolism , Sepsis-Associated Encephalopathy/etiology , Microglia/metabolism , Microglia/physiology , Animals , Blood-Brain Barrier/metabolism , Signal Transduction , Sepsis/complications , Sepsis/physiopathology , Neuroinflammatory Diseases/etiology
4.
Neurobiol Dis ; 195: 106499, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38588753

ABSTRACT

The gut-brain axis is a bidirectional communication network linking the gut and the brain, overseeing digestive functions, emotional responses, body immunity, brain development, and overall health. Substantial research highlights a connection between disruptions of the gut-brain axis and various psychiatric and neurological conditions, including depression and Alzheimer's disease. Given the impact of the gut-brain axis on behavior, cognition, and brain diseases, some studies have started to pay attention to the role of the axis in sepsis-associated encephalopathy (SAE), where cognitive impairment is the primary manifestation. SAE emerges as the primary and earliest form of organ dysfunction following sepsis, potentially leading to acute cognitive impairment and long-term cognitive decline in patients. Notably, the neuronal damage in SAE does not stem directly from the central nervous system (CNS) infection but rather from an infection occurring outside the brain. The gut-brain axis is posited as a pivotal factor in this process. This review will delve into the gut-brain axis, exploring four crucial pathways through which inflammatory signals are transmitted and elevate the incidence of SAE. These pathways encompass the vagus nerve pathway, the neuroendocrine pathway involving the hypothalamic-pituitary-adrenal (HPA) axis and serotonin (5-HT) regulation, the neuroimmune pathway, and the microbial regulation. These pathways can operate independently or collaboratively on the CNS to modulate brain activity. Understanding how the gut affects and regulates the CNS could offer the potential to identify novel targets for preventing and treating this condition, ultimately enhancing the prognosis for individuals with SAE.


Subject(s)
Brain-Gut Axis , Brain , Sepsis-Associated Encephalopathy , Humans , Brain-Gut Axis/physiology , Sepsis-Associated Encephalopathy/physiopathology , Sepsis-Associated Encephalopathy/metabolism , Animals , Brain/physiopathology , Brain/metabolism , Gastrointestinal Microbiome/physiology , Hypothalamo-Hypophyseal System/physiopathology , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/physiopathology , Pituitary-Adrenal System/metabolism , Sepsis/physiopathology , Sepsis/complications
5.
Intern Emerg Med ; 19(4): 983-991, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38480612

ABSTRACT

Sepsis-associated encephalopathy (SAE) is defined as a dysfunction of the central nervous system experienced during sepsis with variable clinical features. The study aims to identify the prognostic role of urinary ketone bodies in relation to clinical outcomes in patients with SAE. The Medical Information Mart for Intensive Care III (MIMIC-III) database was used to conduct a retrospective cohort study. We recruited 427 patients with SAE admitted to the intensive care unit (ICU) from the MIMIC-III database. Patients with SAE were divided into a survival group (380 patients) and a non-survival group (47 patients). We used the Wilcoxon signed-rank test and the multivariate logistic regression analysis to analyze the relationship between the level of urinary ketone bodies and the clinical prognosis in patients with SAE. The primary outcome was the relationship between urinary ketone body levels and 28-day mortality of SAE. The secondary outcomes were the relationship between urinary ketone body levels and length of ICU stays, Simplified Acute Physiology Score II, Sequential Organ Failure Assessment (SOFA), Glasgow Coma Scale, mechanical ventilation, renal replacement therapy, and the use of vasopressors. The 28-day mortality of patients with SAE was 11.0%. Urinary ketone body levels were not significantly associated with the 28-day mortality of patients with SAE. Urinary ketone body levels were associated with SOFA score and the use of vasopressors in patients with SAE. The SOFA score was an independent risk factor for the 28-day mortality in patients with SAE. Urinary ketone body levels were significantly associated with SOFA score and the use of vasopressors in patients with SAE. Furthermore, the SOFA score can predict the prognosis of short-term outcomes of patients with SAE. Therefore, we should closely monitor the changes of urinary ketone bodies and SOFA score and intervene in time.


Subject(s)
Ketone Bodies , Sepsis-Associated Encephalopathy , Humans , Retrospective Studies , Female , Male , Ketone Bodies/urine , Prognosis , Middle Aged , Aged , Sepsis-Associated Encephalopathy/urine , Sepsis-Associated Encephalopathy/physiopathology , Sepsis-Associated Encephalopathy/complications , Intensive Care Units/statistics & numerical data , Cohort Studies , Organ Dysfunction Scores , Biomarkers/urine
6.
Inflammation ; 44(6): 2143-2150, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34291398

ABSTRACT

Sepsis is a life-threatening clinical condition caused by a dysregulated host response to infection. Sepsis-associated encephalopathy (SAE) is a common but poorly understood neurological complication of sepsis, which is associated with increased morbidity and mortality. SAE clinical presentation may range from mild confusion and delirium to severe cognitive impairment and deep coma. Important mechanisms associated with SAE include excessive microglial activation, impaired endothelial barrier function, and blood-brain barrier (BBB) dysfunction. Endotoxemia and pro-inflammatory cytokines produced systemically during sepsis lead to microglial and brain endothelial cell activation, tight junction downregulation, and increased leukocyte recruitment. The resulting neuroinflammation and BBB dysfunction exacerbate SAE pathology and aggravate sepsis-induced brain dysfunction. In this mini-review, recent literature surrounding some of the mediators of BBB dysfunction during sepsis is summarized. Modulation of microglial activation, endothelial cell dysfunction, and the consequent prevention of BBB permeability represent relevant therapeutic targets that may significantly impact SAE outcomes.


Subject(s)
Blood-Brain Barrier/metabolism , Capillary Permeability , Endothelial Cells/metabolism , Microglia/metabolism , Neuroinflammatory Diseases/metabolism , Sepsis-Associated Encephalopathy/metabolism , Animals , Blood-Brain Barrier/pathology , Blood-Brain Barrier/physiopathology , Cytokines/metabolism , Endothelial Cells/pathology , Endotoxins/metabolism , Humans , Inflammation Mediators/metabolism , Microglia/pathology , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/physiopathology , Sepsis-Associated Encephalopathy/pathology , Sepsis-Associated Encephalopathy/physiopathology , Signal Transduction
7.
Life Sci ; 277: 119490, 2021 Jul 15.
Article in English | MEDLINE | ID: mdl-33862114

ABSTRACT

AIMS: Sepsis-associated encephalopathy (SAE) is one of the most common complications of sepsis, and it might lead to long-term cognitive dysfunction and disability. This study aimed to explore the role of S100 calcium binding protein B (S100B)/RAGE/ceramide signaling pathway in SAE. MAIN METHODS: FPS-ZM1 (an inhibitor of RAGE), myriocin and GW4869 (an inhibitor of ceramide) were used to explore the role of S100B/RAGE/ceramide in acute brain injury and long-term cognitive impairment in sepsis. In addition, Mdivi-1 (inhibitor of Drp1) and Drp1 siRNA were utilized to assess the effects of C2-ceramide on neuronal mitochondria, and to explore the specific underlying mechanism in C2 ceramide-induced death of HT22 mouse hippocampal neuronal cells. KEY FINDINGS: Western blot analysis showed that sepsis significantly up-regulated S100B and RAGE. Nissl staining and Morris water maze (MWM) test revealed that inhibition of RAGE with FPS-ZM1 markedly attenuated cecal ligation and puncture (CLP)-induced brain damage and cognitive dysfunction. Furthermore, FPS-ZM1 relieved sepsis-induced C2-ceramide accumulation and abnormal mitochondrial dynamics. Moreover, inhibition of ceramide also showed similar protective effects both in vivo and in vitro. Furthermore, Mdivi-1 and Drp1 siRNA significantly reduced C2-ceramide-induced neuronal mitochondrial fragmentation and cell apoptosis in vitro. SIGNIFICANCE: This study confirmed that S100B regulates mitochondrial dynamics through RAGE/ceramide pathway, in addition to the role of this pathway in acute brain injury and long-term cognitive impairment during sepsis.


Subject(s)
Ceramides/metabolism , Receptor for Advanced Glycation End Products/metabolism , S100 Calcium Binding Protein beta Subunit/metabolism , Sepsis-Associated Encephalopathy/metabolism , Animals , Apoptosis/drug effects , Brain/metabolism , Brain Diseases/complications , Brain Diseases/metabolism , Brain Injuries/metabolism , Cognitive Dysfunction/metabolism , Disease Models, Animal , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Neurons/metabolism , Sepsis/complications , Sepsis-Associated Encephalopathy/complications , Sepsis-Associated Encephalopathy/physiopathology , Signal Transduction
8.
Mol Neurobiol ; 58(6): 2770-2779, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33495934

ABSTRACT

Sepsis is an organ dysfunction caused by an uncontrolled inflammatory response from the host to an infection. Sepsis is the main cause of morbidity and mortality in intensive care units (ICU) worldwide. One of the first organs to suffer from injuries resulting from sepsis is the brain. The central nervous system (CNS) is particularly vulnerable to damage, mediated by inflammatory and oxidative processes, which can cause the sepsis-associated encephalopathy (SAE), being reported in up to 70% of septic patients. This review aims to bring a summary of the main pathophysiological changes and dysfunctions in SAE, and the main focuses of current experimental studies for new treatments and therapies. The pathophysiology of SAE is complex and multifactorial, combining intertwined processes, and is promoted by countless alterations and dysfunctions resulting from sepsis, such as inflammation, neuroinflammation, oxidative stress, reduced brain metabolism, and injuries to the integrity of the blood-brain barrier (BBB). The treatment is limited once its cause is not completely understood. The patient's sedation is far to provide an adequate treatment to this complex condition. Studies and experimental advances are important for a better understanding of its pathophysiology and for the development of new treatments, medicines, and therapies for the treatment of SAE and to reduce its effects during and after sepsis.


Subject(s)
Sepsis-Associated Encephalopathy/pathology , Sepsis-Associated Encephalopathy/physiopathology , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Humans , Models, Biological , Oxidative Stress
9.
Neurotherapeutics ; 18(2): 1393-1413, 2021 04.
Article in English | MEDLINE | ID: mdl-33410107

ABSTRACT

Lower sepsis mortality rates imply that more patients are discharged from the hospital, but sepsis survivors often experience sequelae, such as functional disability, cognitive impairment, and psychiatric morbidity. Nevertheless, the mechanisms underlying these long-term disabilities are not fully understood. Considering the extensive use of animal models in the study of the pathogenesis of neuropsychiatric disorders, it seems adopting this approach to improve our knowledge of postseptic psychiatric symptoms is a logical approach. With the purpose of gathering and summarizing the main findings of studies using animal models of sepsis-induced psychiatric symptoms, we performed a systematic review of the literature on this topic. Thus, 140 references were reviewed, and most of the published studies suggested a time-dependent recovery from behavior alterations, despite the fact that some molecular alterations persist in the brain. This review reveals that animal models can be used to understand the mechanisms that underlie anxiety and depression in animals recovering from sepsis.


Subject(s)
Behavior, Animal , Brain/physiopathology , Mental Disorders/physiopathology , Sepsis-Associated Encephalopathy/physiopathology , Sepsis/physiopathology , Survivors/psychology , Anhedonia/physiology , Animals , Anxiety/physiopathology , Anxiety/psychology , Anxiety Disorders/physiopathology , Anxiety Disorders/psychology , Critical Illness/psychology , Depression/physiopathology , Depression/psychology , Depressive Disorder/physiopathology , Depressive Disorder/psychology , Disease Models, Animal , Escape Reaction/physiology , Exploratory Behavior/physiology , Locomotion/physiology , Mental Disorders/psychology , Quality of Life , Sepsis/psychology , Sepsis-Associated Encephalopathy/psychology , Stress Disorders, Post-Traumatic/physiopathology , Stress Disorders, Post-Traumatic/psychology
10.
Expert Rev Anti Infect Ther ; 19(2): 215-231, 2021 02.
Article in English | MEDLINE | ID: mdl-32808580

ABSTRACT

INTRODUCTION: Sepsis-associated encephalopathy (SAE) and septic encephalitis (SE) are associated with increased mortality, long-term cognitive impairment, and focal neurological deficits. AREAS COVERED: The PUBMED database was searched 2016-2020. The clinical manifestation of SAE is delirium, SE additionally is characterized by focal neurological symptoms. SAE is caused by inflammation with endothelial/microglial activation, increase of permeability of the blood-brain-barrier, hypoxia, imbalance of neurotransmitters, glial activation, axonal, and neuronal loss. Septic-embolic (SEE) and septic-metastatic encephalitis (SME) are characterized by focal ischemia (SEE) and small abscesses (SME). The continuum between SAE, SME, and SEE is documented by imaging techniques and autopsies. The backbone of treatment is rapid optimum antibiotic therapy. Experimental approaches focus on modulation of inflammation, stabilization of the blood-brain barrier, and restoration of membrane/mitochondrial function. EXPERT OPINION: The most promising diagnostic approaches are new imaging techniques. The most important measure to fight delirium remains establishment of daily structure and adequate sensory stimuli. Dexmedetomidine and melatonin appear to reduce the frequency of delirium, their efficacy in SAE and SE remains to be established. Drugs already licensed for other indications or available as food supplements which may be effective in SAE are statins, L-DOPA/benserazide, ß-hydroxybutyrate, palmitoylethanolamide, and tetracyclines or other bactericidal non-lytic antibiotics.


Subject(s)
Encephalitis/etiology , Sepsis-Associated Encephalopathy/therapy , Sepsis/complications , Animals , Anti-Bacterial Agents/administration & dosage , Blood-Brain Barrier/pathology , Cognitive Dysfunction/etiology , Cognitive Dysfunction/therapy , Encephalitis/physiopathology , Encephalitis/therapy , Humans , Mitochondria/pathology , Sepsis/physiopathology , Sepsis/therapy , Sepsis-Associated Encephalopathy/diagnostic imaging , Sepsis-Associated Encephalopathy/physiopathology
11.
Brain Res ; 1751: 147174, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33172595

ABSTRACT

Endogenous toxicity caused by systemic inflammation as well as by acute liver failure triggers a wide range of dysfunctional disorders in the brain ranging from delirium and acute psychosis to coma. Astrocytes, the main homeostatic cells of the central nervous system (CNS), play a key role in pathophysiology of neurotoxic insults. We examined the cecal ligation and puncture (CLP) and acetaminophen-induced liver failure (AILF) of Wistar rats, and analysed ultrastructure of astrocytes in the brain cortex and subcortical white matter of sensorimotor zone with transmission electron microscopy. Both models showed significant similarities in reactive changes of astroglial endosomal machinery. In survived animals (with relative prevalence in the CLP-model), at 24 h after intervention we found an increase in number of multivesicular bodies (MVBs) in astroglial perikarya and astroglial processes. In particular, the number of MVBs substantially (3 times of control values) increased in the perivascular astroglial endfeet. Increased number of MVBs in astrocytes was associated with the lesser degree of intracellular oedema and with signs of compensated oedematous tissue changes. In deceased animals, up to 24 h after intervention, single MVBs were localised mainly in astroglial perikarya, and their number was not significantly changed compared to control. Activation of astroglial endosomal-exosomal machinery in both models reflects the uniform pattern of reactive changes of astroglia in these two systemic conditions and may represent activation of astroglial defence in sepsis-associated encephalopathy (SAE) and acute hepatic encephalopathy (AHE). Our data highlight the special role of astroglial adaptive activity in the counterbalancing of an impaired brain homeostasis under action of endogenous toxins. Accumulation of MVBs in astrocytic processes indicates the activation of their intercellular and gliovascular interactions through endo- and exocytosis in SAE and AHE.


Subject(s)
Astrocytes/physiology , Multivesicular Bodies/physiology , Neurotoxicity Syndromes/physiopathology , Acute Disease , Animals , Astrocytes/metabolism , Brain/physiopathology , Brain Diseases/metabolism , Brain Diseases/physiopathology , Central Nervous System/cytology , Male , Multivesicular Bodies/metabolism , Neurons/metabolism , Neurotoxicity Syndromes/metabolism , Rats , Rats, Wistar , Sensorimotor Cortex/physiopathology , Sepsis/physiopathology , Sepsis-Associated Encephalopathy/physiopathology
12.
J Neuroinflammation ; 17(1): 292, 2020 Oct 07.
Article in English | MEDLINE | ID: mdl-33028343

ABSTRACT

BACKGROUND: Sepsis-associated encephalopathy (SAE) is an early and frequent event of infection-induced systemic inflammatory response syndrome. Phosphoinositide 3-kinase γ (PI3Kγ) is linked to neuroinflammation and inflammation-related microglial activity. In homeotherms, variations in ambient temperature (Ta) outside the thermoneutral zone lead to thermoregulatory responses, mainly driven by a gradually increasing sympathetic activity, and may affect disease severity. We hypothesized that thermoregulatory response to hypothermia (reduced Ta) aggravates SAE in PI3Kγ-dependent manner. METHODS: Experiments were performed in wild-type, PI3Kγ knockout, and PI3Kγ kinase-dead mice, which were kept at neutral (30 ± 0.5 °C) or moderately lowered (26 ± 0.5 °C) Ta. Mice were exposed to lipopolysaccharide (LPS, 10 µg/g, from Escherichia coli serotype 055:B5, single intraperitoneal injection)-evoked systemic inflammatory response (SIR) and monitored 24 h for thermoregulatory response and blood-brain barrier integrity. Primary microglial cells and brain tissue derived from treated mice were analyzed for inflammatory responses and related cell functions. Comparisons between groups were made with one-way or two-way analysis of variance, as appropriate. Post hoc comparisons were made with the Holm-Sidak test or t tests with Bonferroni's correction for adjustments of multiple comparisons. Data not following normal distribution was tested with Kruskal-Wallis test followed by Dunn's multiple comparisons test. RESULTS: We show that a moderate reduction of ambient temperature triggers enhanced hypothermia of mice undergoing LPS-induced systemic inflammation by aggravated SAE. PI3Kγ deficiency enhances blood-brain barrier injury and upregulation of matrix metalloproteinases (MMPs) as well as an impaired microglial phagocytic activity. CONCLUSIONS: Thermoregulatory adaptation in response to ambient temperatures below the thermoneutral range exacerbates LPS-induced blood-brain barrier injury and neuroinflammation. PI3Kγ serves a protective role in suppressing release of MMPs, maintaining microglial motility and reinforcing phagocytosis leading to improved brain tissue integrity. Thus, preclinical research targeting severe brain inflammation responses is seriously biased when basic physiological prerequisites of mammal species such as preferred ambient temperature are ignored.


Subject(s)
Body Temperature Regulation/physiology , Class Ib Phosphatidylinositol 3-Kinase/deficiency , Lipopolysaccharides/toxicity , Sepsis-Associated Encephalopathy/enzymology , Sepsis-Associated Encephalopathy/physiopathology , Animals , Animals, Newborn , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/enzymology , Blood-Brain Barrier/physiopathology , Body Temperature/drug effects , Body Temperature/physiology , Body Temperature Regulation/drug effects , Cells, Cultured , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sepsis-Associated Encephalopathy/chemically induced
13.
Medicina (Kaunas) ; 56(5)2020 May 18.
Article in English | MEDLINE | ID: mdl-32443606

ABSTRACT

Sepsis is a potentially life-threatening condition caused by a systemic dysregulated host response to infection. The brain is particularly susceptible to the effects of sepsis with clinical manifestations ranging from mild confusion to a deep comatose state. Sepsis-associated delirium (SAD) is a cerebral manifestation commonly occurring in patients with sepsis and is thought to occur due to a combination of neuroinflammation and disturbances in cerebral perfusion, the blood brain barrier (BBB) and neurotransmission. The neurological impairment associated with SAD can persist for months or even longer, after the initial septic episode has subsided which may impair the rehabilitation potential of sepsis survivors. Early identification and treatment of the underlying sepsis is key in the management of SAD as once present it can be difficult to control. Through the regular use of validated screening tools for delirium, cases of SAD can be identified early; this allows potentially aggravating factors to be addressed promptly. The usefulness of biomarkers, neuroimaging and electroencephalopathy (EEG) in the diagnosis of SAD remains controversial. The Society of Critical Care Medicine (SCCM) guidelines advise against the use of medications to treat delirium unless distressing symptoms are present or it is hindering the patient's ability to wean from organ support.


Subject(s)
Delirium/diagnosis , Delirium/etiology , Sepsis-Associated Encephalopathy/etiology , Sepsis/complications , Biomarkers/analysis , Biomarkers/blood , Delirium/blood , Humans , Sepsis/blood , Sepsis-Associated Encephalopathy/physiopathology
14.
Brain Res ; 1737: 146780, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32205148

ABSTRACT

The prevention and treatment of sepsis associated encephalopathy (SAE) remains challenging in clinic. Besides the anti-infection treatments and goal-directed supportive treatments, no specific method is reported for the prevention and treatment of SAE. This study tried to investigate the effects and underlying mechanisms of small dose of L-dopa/Benserazide hydrochloride (L-DA) on SAE. We found that L-DA administration (i.p.) at early stage of sepsis, but not at late stage, improved learning and memory of sepsis surviving mice in Cecal ligation and perforation (CLP) model. Corresponding to the improvement of learning and memory in CLP model, L-DA administration limited neuroinflammation, improved neuroplasticity, reversed sepsis-induced decrease of hippocampal dopamine level, but had no obvious effects on the survival and body weight recovery. Further studies showed that specific inhibitors of dopamine D1 or D2 receptors both partly reduced the protective effect of L-DA on the learning and memory of lipopolysaccharides (LPS) treated mice. D1 receptor specific inhibitor significantly blocked the anti-neuroinflammation effects of L-DA in LPS treated mice, but D2 receptor inhibitor did not. All these suggest that L-DA administration could prevent and treat SAE via dopamine D1 and D2 receptors. Dopamine D1 receptor is a potential target of anti-neuroinflammation.


Subject(s)
Benserazide/therapeutic use , Cognitive Dysfunction/drug therapy , Levodopa/therapeutic use , Neuroimmunomodulation/drug effects , Animals , Brain/physiopathology , Cognitive Dysfunction/physiopathology , Disease Models, Animal , Drug Combinations , Hippocampus/physiopathology , Male , Mice , Mice, Inbred C57BL , Neuroimmunomodulation/physiology , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Sepsis/complications , Sepsis-Associated Encephalopathy/physiopathology
15.
Pak J Pharm Sci ; 33(6): 2567-2577, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33867332

ABSTRACT

Ginsenoside (Rg1) has biological effects including anti-oxidation, anti-inflammation, neuroprotection and neural function improvement, but with few studies in sepsis-associated encephalopathy (SAE). This study thus evaluated Ginsenoside in alleviating SAE, suppressing oxidative stress (OS) or neuronal apoptosis. SAE mouse model was generated and were assigned into SAE, SAE + LD-Rg1, and SAE + HD-Rg1 groups to measure neural apoptosis by flow cytometry. Contents of malondialdehyde (MDA), superoxide dismutase (SOD), GSH-Px and caspase-3 were quantified, and mouse neural reflex function was evaluated. Expression of Nrf2, HO-1 was measured. Mouse neuron MN-c and microglia BV2 were co-cultured in control, LPS, LPS+Rg1 (20µM) and LPS+Rg1 (40µM) groups. Iba-1 expression of BV2 cells was measured by flow cytometry. Contents of tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), and IL-6 were quantified. Apoptosis of MN-c cells was measured by flow cytometry, and reactive oxygen species (ROS) content was measured by DCFH-DA staining. SAE mice had elevated caspase-3 activity, cell apoptosis, MDA content, and decreased SOD, GSH-Px activity or neural reflex score comparing to Sham group. Rg1 treatment suppressed caspase-3 activity, apoptotic rate or MDA content, recovered SOD activity, neural reflex score, and expression of Nrf2 and HO-1. LPS treatment elevated Iba-1 expression and release of inflammatory cytokines TNF-α, IL-1ß and IL-6, induced MN-c apoptosis or ROS production, and enhanced Nrf2 and HO-1 expression. Rg1 treatment remarkably inhibited LPS-induced response or cell apoptosis. Ginsenoside can alleviate SAE damage via up-regulating Nrf2 and HO-1 to enhance anti-OS potency and to reduce neural cell apoptosis.


Subject(s)
Apoptosis/drug effects , Brain/drug effects , Ginsenosides/pharmacology , Oxidative Stress/drug effects , Sepsis-Associated Encephalopathy/drug therapy , Animals , Brain/metabolism , Brain/pathology , Caspase 3/metabolism , Cell Line , Cytokines/metabolism , Disease Models, Animal , Heme Oxygenase-1/metabolism , Male , Membrane Proteins/metabolism , Mice, Inbred C57BL , Microglia/drug effects , NF-E2-Related Factor 2/metabolism , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Sepsis-Associated Encephalopathy/pathology , Sepsis-Associated Encephalopathy/physiopathology
16.
J Intensive Care Med ; 35(12): 1453-1464, 2020 Dec.
Article in English | MEDLINE | ID: mdl-30760173

ABSTRACT

BACKGROUND: Impaired cerebral autoregulation and cerebral hypoperfusion may play a critical role in the high morbidity and mortality in patients with sepsis-associated encephalopathy (SAE). Bedside assessment of cerebral autoregulation may help individualize hemodynamic targets that optimize brain perfusion. We hypothesize that near-infrared spectroscopy (NIRS)-derived cerebral oximetry can identify blood pressure ranges that enhance autoregulation in patients with SAE and that disturbances in autoregulation are associated with severity of encephalopathy. METHODS: Adult patients with acute encephalopathy directly attributable to sepsis were followed using NIRS-based multimodal monitoring for 12 consecutive hours. We used the correlation in time between regional cerebral oxygen saturation and mean arterial pressure (MAP) to determine the cerebral oximetry index (COx) as a measure of cerebral autoregulation. Autoregulation curves were constructed for each patient with averaged COx values sorted by MAP in 3 sequential 4-hour periods; the optimal pressure (MAPOPT), defined as the MAP associated with most robust autoregulation (lowest COx), was identified in each period. Severity of encephalopathy was measured with Glasgow coma scale (GCS). RESULTS: Six patients with extracranial sepsis met the stringent criteria specified, including no pharmacological sedation or neurologic premorbidity. Optimal MAP was identified in all patients and ranged from 55 to 115 mmHg. Additionally, MAPOPT varied within individual patients over time during monitoring. Disturbed autoregulation, based on COx, was associated with worse neurologic status (GCS < 13) both with and without controlling for age and severity of sepsis (adjusted odds ratio [OR]: 2.11; 95% confidence interval [CI]: 1.77-2.52; P < .001; OR: 2.97; 95% CI: 1.63-5.43; P < .001). CONCLUSIONS: In this high-fidelity group of patients with SAE, continuous, NIRS-based monitoring can identify blood pressure ranges that improve autoregulation. This is important given the association between cerebral autoregulatory function and severity of encephalopathy. Individualizing blood pressure goals using bedside autoregulation monitoring may better preserve cerebral perfusion in SAE than current practice.


Subject(s)
Cerebrovascular Circulation , Sepsis-Associated Encephalopathy , Aged , Blood Pressure/physiology , Cerebrovascular Circulation/physiology , Female , Homeostasis , Humans , Male , Middle Aged , Oximetry , Sepsis-Associated Encephalopathy/physiopathology , Spectroscopy, Near-Infrared
17.
Neurocrit Care ; 32(2): 459-468, 2020 04.
Article in English | MEDLINE | ID: mdl-31187433

ABSTRACT

BACKGROUND: Sepsis-induced brain dysfunction (SIBD) is often encountered in sepsis patients and is related to increased morbidity. No specific tests are available for SIBD, and neuroimaging findings are often normal. In this study, our aim was to analyze the diagnostic value of volumetric analysis of the brain structures and to find out its significance as a prognostic measure. METHODS: In this prospective observational study, brain magnetic resonance imaging (MRI) sections of 25 consecutively enrolled SIBD patients (17 with encephalopathy and 8 with coma) and 22 healthy controls underwent volumetric evaluation by an automated segmentation method. RESULTS: Ten SIBD patients had normal MRI, and 15 patients showed brain lesions or atrophy. The most prominent volume reduction was found in cerebral and cerebellar white matter, cerebral cortex, hippocampus, and amygdala, whereas deep gray matter regions and cerebellar cortex were relatively less affected. SIBD patients with normal MRI showed significantly reduced volumes in hippocampus and cerebral white matter. Caudate nuclei, putamen, and thalamus showed lower volume values in non-survivor SIBD patients, and left putamen and right thalamus showed a more pronounced volume reduction in coma patients. CONCLUSIONS: Volumetric analysis of the brain appears to be a sensitive measure of volumetric changes in SIBD. Volume reduction in specific deep gray matter regions might be an indicator of unfavorable outcome.


Subject(s)
Brain/diagnostic imaging , Coma/diagnostic imaging , Sepsis-Associated Encephalopathy/diagnostic imaging , Sepsis/physiopathology , Amygdala/diagnostic imaging , Amygdala/pathology , Atrophy , Brain/pathology , Case-Control Studies , Caudate Nucleus/diagnostic imaging , Caudate Nucleus/pathology , Cerebellar Cortex/diagnostic imaging , Cerebellar Cortex/pathology , Cerebellum/diagnostic imaging , Cerebellum/pathology , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/pathology , Cerebral Infarction/diagnostic imaging , Coma/etiology , Coma/physiopathology , Female , Gray Matter/diagnostic imaging , Gray Matter/pathology , Hippocampus/diagnostic imaging , Hippocampus/pathology , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Organ Size , Posterior Leukoencephalopathy Syndrome/diagnostic imaging , Putamen/diagnostic imaging , Putamen/pathology , Sepsis/complications , Sepsis-Associated Encephalopathy/etiology , Sepsis-Associated Encephalopathy/physiopathology , Thalamus/diagnostic imaging , Thalamus/pathology , White Matter/diagnostic imaging , White Matter/pathology
18.
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
19.
Brain Behav Immun ; 80: 859-870, 2019 08.
Article in English | MEDLINE | ID: mdl-31145977

ABSTRACT

Sepsis-associated encephalopathy (SAE) manifested clinically in acute and long-term cognitive impairments and associated with increased morbidity and mortality worldwide. The potential pathological changes of SAE are complex and remain to be elucidated. Pyroptosis, a novel programmed cell death, is executed by caspase-1-cleaved GSDMD N-terminal (GSDMD-NT) and we investigated it in peripheral blood immunocytes of septic patients previously. Here, a caspase-1 inhibitor VX765 was treated with CLP-induced septic mice. Novel object recognition test indicated that VX765 treatment reversed cognitive dysfunction in septic mice. Elevated plus maze, tail suspension test and open field test revealed that depressive-like behaviors of septic mice were relieved. Inhibited caspase-1 suppressed the expressions of GSDMD and its cleavage form GSDMD-NT, and reduced pyroptosis in brain at day 1 and day 7 after sepsis. Meantime, inhibited caspase-1 mitigated the expressions of IL-1ß, MCP-1 and TNF-α in serum and brain, diminished microglia activation in septic mice, and reduced sepsis-induced brain-blood barrier disruption and ultrastructure damages in brain as well. Inhibited caspase-1 protected the synapse plasticity and preserved long-term potential, which may be the possible mechanism of cognitive functions protective effects of septic mice. In conclusion, caspase-1 inhibition exerts brain-protective effects against SAE and cognitive impairments in a mouse model of sepsis.


Subject(s)
Cognitive Dysfunction/physiopathology , Pyroptosis/drug effects , Sepsis-Associated Encephalopathy/metabolism , Animals , Apoptosis/drug effects , Brain/metabolism , Brain Diseases/metabolism , Brain Diseases/physiopathology , Caspase 1/metabolism , Caspase Inhibitors/pharmacology , Dipeptides/pharmacology , Hippocampus/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lipopolysaccharides/pharmacology , Macrophage Activation , Male , Mice , Mice, Inbred BALB C , Phosphate-Binding Proteins/metabolism , Pyroptosis/physiology , Sepsis/complications , Sepsis/metabolism , Sepsis/physiopathology , Sepsis-Associated Encephalopathy/physiopathology , Synapses/metabolism , para-Aminobenzoates/pharmacology
20.
Brain Res ; 1719: 40-48, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31075263

ABSTRACT

Sepsis-associated encephalopathy (SAE) is a common and serious complication of sepsis, which is thought to be caused by neuroinflammation. In our previous study, ubiquitin-specific protease 8 (USP8), was reported to regulate inflammation in vitro. In the current study, we investigated whether increased USP8 expression would ameliorate the cognitive and motor impairments induced by cecal ligation and puncture (CLP) in mice, a model of SAE. Male adult mice were randomly divided into four groups: control, sham, CLP, and CLP + USP8 groups. The CLP + USP8 mice showed reduced weight loss on day 4 post-CLP, with a slight increase noted on day 7. The mortality rate in the CLP group was 70% 48 h after CLP; however, USP8 significantly improved survival after CLP. USP8 modulated the neurobehavioral scores in CLP mice. Our results also indicate that USP8 attenuated the CLP-induced cognitive and motor impairments, based on the performance of mice in the Morris water maze (MWM), pole-climbing, and wire suspension tests. USP8 suppressed the release of pro-inflammatory mediators, including prostaglandin E2(PGE2) in the serum and nitric oxide (NO) in brain tissue, as well as levels of inducible NO synthase (iNOS) and cyclooxygenase-2 (COX-2) in brain tissue. Immunofluorescence experiments revealed that USP8 inhibited CLP-induced increases in microglial size and density in the hippocampus, and protected hippocampal neurons. Our findings indicate that neuroinflammation occurs in the brains of CLP mice, and that USP8 exerts protective effects against CLP-induced neuroinflammation and cognitive and motor impairments, which may aid in the development of novel therapeutic strategies for SAE.


Subject(s)
Endopeptidases/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Sepsis-Associated Encephalopathy/physiopathology , Ubiquitin Thiolesterase/metabolism , Animals , Brain/metabolism , Cecum , Cognition/drug effects , Cognitive Dysfunction/physiopathology , Disease Models, Animal , Endopeptidases/physiology , Endosomal Sorting Complexes Required for Transport/physiology , Hippocampus/metabolism , Inflammation/metabolism , Inhibition, Psychological , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , Motor Activity/drug effects , Neuroimmunomodulation/physiology , Nitric Oxide Synthase Type II/metabolism , Sepsis/complications , Ubiquitin Thiolesterase/physiology
SELECTION OF CITATIONS
SEARCH DETAIL