Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 217
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Neuroinflammation ; 21(1): 140, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38807233

ABSTRACT

BACKGROUND: Perihematomal edema (PHE) after post-intracerebral hemorrhage (ICH) has complex pathophysiological mechanisms that are poorly understood. The complicated immune response in the post-ICH brain constitutes a crucial component of PHE pathophysiology. In this study, we aimed to characterize the transcriptional profiles of immune cell populations in human PHE tissue and explore the microscopic differences between different types of immune cells. METHODS: 9 patients with basal ganglia intracerebral hemorrhage (hematoma volume 50-100 ml) were enrolled in this study. A multi-stage profile was developed, comprising Group1 (n = 3, 0-6 h post-ICH, G1), Group2 (n = 3, 6-24 h post-ICH, G2), and Group3 (n = 3, 24-48 h post-ICH, G3). A minimal quantity of edematous tissue surrounding the hematoma was preserved during hematoma evacuation. Single cell RNA sequencing (scRNA-seq) was used to map immune cell populations within comprehensively resected PHE samples collected from patients at different stages after ICH. RESULTS: We established, for the first time, a comprehensive landscape of diverse immune cell populations in human PHE tissue at a single-cell level. Our study identified 12 microglia subsets and 5 neutrophil subsets in human PHE tissue. What's more, we discovered that the secreted phosphoprotein-1 (SPP1) pathway served as the basis for self-communication between microglia subclusters during the progression of PHE. Additionally, we traced the trajectory branches of different neutrophil subtypes. Finally, we also demonstrated that microglia-produced osteopontin (OPN) could regulate the immune environment in PHE tissue by interacting with CD44-positive cells. CONCLUSIONS: As a result of our research, we have gained valuable insight into the immune-microenvironment within PHE tissue, which could potentially be used to develop novel treatment modalities for ICH.


Subject(s)
Brain Edema , Cerebral Hemorrhage , Disease Progression , Sequence Analysis, RNA , Single-Cell Analysis , Humans , Brain Edema/immunology , Brain Edema/pathology , Brain Edema/genetics , Brain Edema/metabolism , Brain Edema/etiology , Cerebral Hemorrhage/immunology , Cerebral Hemorrhage/pathology , Cerebral Hemorrhage/genetics , Male , Female , Middle Aged , Sequence Analysis, RNA/methods , Aged , Hematoma/pathology , Hematoma/immunology , Hematoma/genetics
2.
Blood ; 137(19): 2699-2703, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33649761

ABSTRACT

Acute platelet transfusion after intracerebral hemorrhage (ICH) given in efforts to reverse antiplatelet medication effects and prevent ongoing bleeding does not appear to improve outcome and may be associated with harm. Although the underlying mechanisms are unclear, the influence of ABO-incompatible platelet transfusions on ICH outcomes has not been investigated. We hypothesized that patients with ICH who receive ABO-incompatible platelet transfusions would have worse platelet recovery (using absolute count increment [ACI]) and neurological outcomes (mortality and poor modified Rankin Scale [mRS 4-6]) than those receiving ABO-compatible transfusions. In a single-center cohort of consecutively admitted patients with ICH, we identified 125 patients receiving acute platelet transfusions, of whom 47 (38%) received an ABO-incompatible transfusion. Using quantile regression, we identified an association of ABO-incompatible platelet transfusion with lower platelet recovery (ACI, 2 × 103cells per µL vs 15 × 103cells per µL; adjusted coefficient ß, -19; 95% confidence interval [CI], -35.55 to -4.44; P = .01). ABO-incompatible platelet transfusion was also associated with increased odds of mortality (adjusted odds ratio [OR], 2.59; 95% CI, 1.00-6.73; P = .05) and poor mRS (adjusted OR, 3.61; 95% CI, 0.97-13.42; P = .06); however, these estimates were imprecise. Together, these findings suggest the importance of ABO compatibility for platelet transfusions for ICH, but further investigation into the mechanism(s) underlying these observations is required.


Subject(s)
ABO Blood-Group System/immunology , Blood Group Incompatibility , Cerebral Hemorrhage/therapy , Platelet Transfusion , Aged , Brain Damage, Chronic/etiology , Cerebral Hemorrhage/complications , Cerebral Hemorrhage/immunology , Cerebral Hemorrhage/mortality , Female , Hematoma/etiology , Hematoma/prevention & control , Hospital Mortality , Humans , Male , Middle Aged , Platelet Aggregation Inhibitors/adverse effects , Platelet Count , Platelet Transfusion/adverse effects , Prospective Studies , Treatment Outcome
3.
Stroke ; 52(11): 3613-3623, 2021 11.
Article in English | MEDLINE | ID: mdl-34281379

ABSTRACT

Background and Purpose: Spontaneous intracerebral hemorrhage (sICH) is a common form of hemorrhagic stroke, with high mortality and morbidity. Pathophysiological mechanisms in sICH are poorly understood and treatments limited. Neuroinflammation driven by microglial-macrophage activation contributes to brain damage post-sICH. We aim to test the hypothesis that an anti-inflammatory (repair) process occurs in parallel with neuroinflammation in clinical sICH. Methods: We performed quantitative analysis of immunohistochemical markers for microglia and macrophages (Iba1, CD68, TMEM119, CD163, and CD206) in brain tissue biospecimens 1 to 12 days post-sICH and matched control cases. In a parallel, prospective group of patients, we assayed circulating inflammatory markers (CRP [C-reactive protein], total white cell, and monocyte count) over 1 to 12 days following sICH. Results: In 27 supratentorial sICH cases (n=27, median [interquartile range] age: 59 [52­80.5], 14F/13M) all microglia-macrophage markers increased post-sICH, relative to control brains. Anti-inflammatory markers (CD163 and CD206) were elevated alongside proinflammatory markers (CD68 and TMEM119). CD163 increased progressively post-sICH (15.0-fold increase at 7­12 days, P<0.001). CD206 increased at 3 to 5 days (5.2-fold, P<0.001) then returned to control levels at 7 to 12 days. The parenchymal immune response combined brain-derived microglia (TMEM119 positive) and invading monocyte-derived macrophages (CD206 positive). In a prospective sICH patient cohort (n=26, age 74 [66­79], National Institutes of Health Stroke Scale on admission: 8 [4­17]; 14F/12M) blood CRP concentration and monocyte density (but not white blood cell) increased post-sICH. CRP increased from 0 to 2 to 3 to 5 days (8.3-fold, P=0.020) then declined at 7 to 12 days. Monocytes increased from 0 to 2 to 3 to 5 days (1.8-fold, P<0.001) then declined at 7 to 12 days. Conclusions: An anti-inflammatory pathway, enlisting native microglia and blood monocytes, occurs alongside neuroinflammation post-sICH. This novel pathway offers therapeutic targets and a window of opportunity (3­5 days post-sICH) for delivery of therapeutics via invading monocytes.


Subject(s)
Cerebral Hemorrhage/immunology , Hemorrhagic Stroke/immunology , Immunity, Innate/immunology , Neuroinflammatory Diseases/immunology , Adult , Aged , Aged, 80 and over , Cerebral Hemorrhage/pathology , Female , Hemorrhagic Stroke/pathology , Humans , Macrophages/immunology , Male , Microglia/immunology , Middle Aged , Neuroinflammatory Diseases/pathology
4.
J Neurophysiol ; 125(6): 2025-2033, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33909508

ABSTRACT

Stroke is the second leading cause of death worldwide, estimated that one-sixth of the world population will suffer it once in their life. The most common type of this medical condition is the ischemic stroke (IS), produced by a thrombotic or embolic occlusion of a major cerebral artery or its branches, leading to the formation of a complex infarct region caused by oxidative stress, excitotoxicity, and endothelial dysfunction. Nowadays, the immediate treatment for IS involves thrombolytic agents or mechanical thrombectomy, depending on the integrity of the blood-brain barrier (BBB). A common stroke complication is the hemorrhagic transformation (HT), which consists of bleeding into the ischemic brain area. Currently, better treatments for IS are urgently needed. As such, the neurohormone melatonin has been proposed as a good candidate due to its antioxidant, anti-inflammatory, and neuroprotective effects, particularly against lipid peroxidation and oxidative stress during brain ischemia. Here, we proposed to develop intravenous or intranasal melatonin nanoformulation to specifically target the brain in patients with stroke. Nowadays, the challenge is to find a formulation able to cross the barriers and reach the target organ in an effective dose to generate the pharmacological effect. In this review, we discuss the current literature about stroke pathophysiology, melatonin properties, and its potential use in nanoformulations as a novel therapeutic approach for ischemic stroke.


Subject(s)
Blood-Brain Barrier , Cerebral Hemorrhage/drug therapy , Melatonin/administration & dosage , Nanoparticles/administration & dosage , Neuroprotective Agents/administration & dosage , Stroke/drug therapy , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiopathology , Cerebral Hemorrhage/etiology , Cerebral Hemorrhage/immunology , Cerebral Hemorrhage/metabolism , Humans , Stroke/complications , Stroke/immunology , Stroke/metabolism
5.
J Neuroinflammation ; 18(1): 160, 2021 Jul 18.
Article in English | MEDLINE | ID: mdl-34275493

ABSTRACT

BACKGROUND: Germinal matrix hemorrhage (GMH) is defined by the rupture of immature blood vessels in the germinal matrix, where subsequent hemorrhage enters the subependymal zone and the cerebral lateral ventricles. The consequent blood clot has been identified as the causative factor of secondary brain injury, which triggers a series of complex parallel and sequential harmful mechanisms, including neuroinflammation. The orphan G-protein-coupled receptor 40 (GPR40), a free fatty acid (FFA) receptor 1, has been shown to exert anti-inflammatory effects when activated and improved outcomes in animal models of stroke. We aimed to investigate the anti-inflammatory effects of GPR40 and its underlying mechanisms after GMH. METHODS: GMH model was induced in 7-day-old rat pups by an intraparenchymal injection of bacterial collagenase. GPR40 agonist, GW9508, was administered intranasally 1 h, 25 h, and 49 h after GMH induction. CRISPR targeting GPR40, PAK4, and KDM6B were administered through intracerebroventricular injection 48 h before GMH induction. Neurologic scores, microglia polarization, and brain morphology were evaluated by negative geotaxis, right reflex, rotarod test, foot fault test, Morris water maze, immunofluorescence staining, Western blots, and nissl staining respectfully. RESULTS: The results demonstrated that GW9508 improved neurological and morphological outcomes after GMH in the short (24 h, 48 h, 72h) and long-term (days 21-27). However, the neuroprotective effects of treatment were abolished by GW1100, a selective GPR40 antagonist. GW9508 treatment increased populations of M2 microglia and decreased M1 microglia in periventricular areas 24 h after GMH induction. GW9508 upregulated the phosphorylation of PAK4, CREB, and protein level of KDM6B, CD206, IL-10, which was also met with the downregulation of inflammatory markers IL-1ß and TNF-α. The mechanism study demonstrated that the knockdown of GPR40, PAK4, and KDM6B reversed the neuroprotective effects brought on by GW9508. This evidence suggests that GPR40/PAK4/CREB/KDM6B signaling pathway in microglia plays a role in the attenuation of neuroinflammation after GMH. CONCLUSIONS: In conclusion, the present study demonstrates that the activation of GPR40 attenuated GMH-induced neuroinflammation through the activation of the PAK4/CREB/KDM6B signaling pathway, and M2 microglia may be a major mediator of this effect. Thus, GPR40 may serve as a potential target in the reduction of the inflammatory response following GMH, thereby improving neurological outcomes in the short- and long-term.


Subject(s)
Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/pathology , Microglia/metabolism , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Animals , Animals, Newborn , Behavior Rating Scale , Cerebral Hemorrhage/immunology , Cytokines , Disease Models, Animal , Fluorescent Antibody Technique , Jumonji Domain-Containing Histone Demethylases/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction
6.
FASEB J ; 34(2): 2774-2791, 2020 02.
Article in English | MEDLINE | ID: mdl-31912591

ABSTRACT

Immunologic changes in the hematoma of patients with intracerebral hemorrhage (ICH) and the contribution of these changes to prognosis are unknown. We collected the blood samples and hematoma fluid from 35 patients with acute ICH (<30 hours from symptom onset) and 55 age-matched healthy controls. Using flow cytometry and ELISA, we found that the percentages of granulocytes, regulatory T cells, helper T (Th) 17 cells, and dendritic cells were higher in the peripheral blood of patients with ICH than in healthy controls, whereas the percentages of lymphocytes, M1-like macrophages, and M2-like macrophages were lower. Levels of IL-6, IL-17, IL-23, TNF-α, IL-4, IL-10, and TGF-ß were higher in the peripheral blood of patients with ICH. The absolute counts of white blood cells, lymphocytes, monocytes, and granulocytes in the hematoma tended to be greater at 12-30 hours than they were within 12 hours after ICH, but the percentage of Th cells decreased in peripheral blood. Increased levels of IL-10 in the serum and hematoma, and a reduction in M1-like macrophages in hematoma were independently associated with favorable outcome on day 90. These results indicate that immunocytes present in the hematoma may participate in the acute-phase inflammatory response after ICH.


Subject(s)
Cerebral Hemorrhage/immunology , Hematoma/immunology , Inflammation/immunology , Macrophages/immunology , Adult , Aged , Aged, 80 and over , Cerebral Hemorrhage/diagnosis , Female , Hematoma/diagnosis , Humans , Interleukin-17/blood , Male , Middle Aged , Monocytes/immunology , Prognosis
7.
Brain ; 143(5): 1297-1314, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31919518

ABSTRACT

The prognosis of intracerebral haemorrhage continues to be devastating despite much research into this condition. A prominent feature of intracerebral haemorrhage is neuroinflammation, particularly the excessive representation of pro-inflammatory CNS-intrinsic microglia and monocyte-derived macrophages that infiltrate from the circulation. The pro-inflammatory microglia/macrophages produce injury-enhancing factors, including inflammatory cytokines, matrix metalloproteinases and reactive oxygen species. Conversely, the regulatory microglia/macrophages with potential reparative and anti-inflammatory roles are outcompeted in the early stages after intracerebral haemorrhage, and their beneficial roles appear to be overwhelmed by pro-inflammatory microglia/macrophages. In this review, we describe the activation of microglia/macrophages following intracerebral haemorrhage in animal models and clinical subjects, and consider their multiple mechanisms of cellular injury after haemorrhage. We review strategies and medications aimed at suppressing the pro-inflammatory activities of microglia/macrophages, and those directed at elevating the regulatory properties of these myeloid cells after intracerebral haemorrhage. We consider the translational potential of these medications from preclinical models to clinical use after intracerebral haemorrhage injury, and suggest that several approaches still lack the experimental support necessary for use in humans. Nonetheless, the preclinical data support the use of deactivator or inhibitor of pro-inflammatory microglia/macrophages, whilst enhancing the regulatory phenotype, as part of the therapeutic approach to improve the prognosis of intracerebral haemorrhage.


Subject(s)
Cerebral Hemorrhage/immunology , Inflammation/immunology , Macrophages/immunology , Microglia/immunology , Animals , Humans , Phenotype
8.
Int J Med Sci ; 18(3): 639-645, 2021.
Article in English | MEDLINE | ID: mdl-33437198

ABSTRACT

Intracerebral hemorrhage (ICH) represents a common acute cerebrovascular event that imparts high rates of disability. The microglia-mediated inflammatory response is a critical factor in determining cerebral damage post-ICH. Clemastine (CLM) is a histamine receptor H1 (HRH1) antagonist that has been shown to modulate the inflammatory response. However, the effects of CLM on ICH and the underlying mechanism remain to be determined. This investigation reveals that CLM resulted in reduction of cerebral hematoma volume, decreased cerebral edema and lower rates of neuronal apoptosis as well as improved behavioral scores in an acute ICH murine model. CLM treatment was noted to decrease pro-inflammatory effectors and increased anti-inflammatory effectors post-ICH. In addition, CLM reduced the deleterious effects of activated microglia on neurons in a transwell co-culture system. Our findings show that CLM likely mediates its therapeutic effect through inhibition of microglia-induced inflammatory response and apoptosis, thereby enhancing restoration of neuronal function.


Subject(s)
Brain Edema/drug therapy , Cerebral Hemorrhage/drug therapy , Clemastine/pharmacology , Inflammation Mediators/metabolism , Neuroprotective Agents/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/immunology , Brain Edema/immunology , Brain Edema/pathology , Cells, Cultured , Cerebral Hemorrhage/complications , Cerebral Hemorrhage/immunology , Cerebral Hemorrhage/pathology , Clemastine/therapeutic use , Coculture Techniques , Disease Models, Animal , Male , Mice , Microglia/drug effects , Microglia/immunology , Microglia/pathology , Neurons/drug effects , Neurons/immunology , Neurons/pathology , Neuroprotective Agents/therapeutic use , Primary Cell Culture , Stereotaxic Techniques
9.
J Endocrinol Invest ; 44(12): 2685-2698, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34024028

ABSTRACT

OBJECTIVES: Intracerebral hemorrhage (ICH) induced by diabetes results in further brain injury and nerve cell death. Bone marrow mesenchymal stem cell (BMSC) transplantation contributes to attenuating neurological deficits after ICH. This study investigated the mechanism of extracellular vesicles (EVs) derived from BMSCs in reducing neuroinflammation after diabetic ICH. METHODS: BMSC-EVs were isolated and identified. The rat model of db/db-ICH was established and the model rats were administered with EVs. miR-183-5p expression in brain tissues of db/db-ICH rats was detected. The brain injury of db/db-ICH rats was evaluated by measuring neurobehavioral score, brain water content and inflammatory factors. BV2 cells were cultured in vitro to establish high-glucose (HG)-Hemin-BV2 cell model. The levels of reactive oxygen species (ROS) and inflammatory factors in BV2 cells were measured, and BV2 cell viability and apoptosis were assessed. The targeting relationship between miR-183-5p and PDCD4 was predicted and verified. The activation of PDCD4/NLRP3 pathway in rat brain tissues and BV2 cells was detected. RESULTS: miR-183-5p expression was reduced in db/db-ICH rats brain tissues. BMSC-EVs ameliorated cranial nerve function, decreased brain water content and repressed inflammatory response by carrying miR-183-5p. BMSC-EVs mitigated HG-Hemin-BV2 cell injury, reduced ROS level and suppressed inflammatory response. miR-183-5p targeted PDCD4. PDCD4 promoted BV2 cell inflammation by activating the NLRP3 pathway. BMSC-EVs inhibited HG-Hemin-BV2 cell inflammation through the miR-183-5p/PDCD4/NLRP3 pathway, and inhibition of miR-183-5p reversed the protective effect of EVs. CONCLUSION: BMSC-EVs carried miR-183-5p into db/db-ICH rat brain tissues and repressed the NLRP3 pathway by targeting PDCD4, thus alleviating neuroinflammation after diabetic ICH.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Cerebral Hemorrhage , Diabetes Complications , Extracellular Vesicles/transplantation , MicroRNAs/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Brain/immunology , Brain/metabolism , Cells, Cultured , Cerebral Hemorrhage/etiology , Cerebral Hemorrhage/immunology , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/therapy , Diabetes Complications/immunology , Diabetes Complications/metabolism , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/ultrastructure , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/therapy , Protective Factors , Rats , Signal Transduction
10.
Stroke ; 51(3): 967-974, 2020 03.
Article in English | MEDLINE | ID: mdl-32019481

ABSTRACT

Background and Purpose- Microglia are among the first cells to respond to intracerebral hemorrhage (ICH), but the mechanisms that underlie their activity following ICH remain unclear. IL (interleukin)-15 is a proinflammatory cytokine that orchestrates homeostasis and the intensity of the immune response following central nervous system inflammatory events. The goal of this study was to investigate the role of IL-15 in ICH injury. Methods- Using brain slices of patients with ICH, we determined the presence and cellular source of IL-15. A transgenic mouse line with targeted expression of IL-15 in astrocytes was generated to determine the role of astrocytic IL-15 in ICH. The expression of IL-15 was controlled by a glial fibrillary acidic protein promoter (GFAP-IL-15tg). ICH was induced by intraparenchymal injection of collagenase or autologous blood. Results- In patients with ICH and wild-type mice subjected to experimental ICH, we found a significant upregulation of IL-15 in astrocytes. In GFAP-IL-15tg mice, we found that astrocyte-targeted expression of IL-15 exacerbated brain edema and neurological deficits following ICH. This aggravated ICH injury in GFAP-IL-15tg mice is accompanied by increased microglial accumulation in close proximity to astrocytes in perihematomal tissues. Additionally, microglial expression of CD86, IL-1ß, and TNF-α is markedly increased in GFAP-IL-15tg mice following ICH. Furthermore, depletion of microglia using a colony stimulating factor 1 receptor inhibitor diminishes the exacerbation of ICH injury in GFAP-IL-15tg mice. Conclusions- Our findings identify IL-15 as a mediator of the crosstalk between astrocytes and microglia that exacerbates brain injury following ICH.


Subject(s)
Astrocytes/immunology , Brain Injuries/immunology , Cerebral Hemorrhage/immunology , Interleukin-15/immunology , Microglia/immunology , Aged , Aged, 80 and over , Animals , Astrocytes/pathology , Brain Injuries/etiology , Brain Injuries/genetics , Brain Injuries/pathology , Cerebral Hemorrhage/complications , Cerebral Hemorrhage/genetics , Cerebral Hemorrhage/pathology , Female , Humans , Interleukin-15/genetics , Male , Mice , Mice, Transgenic , Microglia/pathology
11.
FASEB J ; 33(8): 9616-9626, 2019 08.
Article in English | MEDLINE | ID: mdl-31145859

ABSTRACT

Delayed cognitive decline commonly occurs following intracerebral hemorrhage (ICH), but the mechanisms underlying this phenomenon remain obscure. We therefore investigated the potential mechanisms responsible for impaired cognitive function in a mouse collagenase model of ICH. Following recovery of motor and sensory deficits in the chronic phase of ICH, we noted significant cognitive impairment, which was assessed by the Morris water maze. This finding was accompanied by reduced dendrite spine density of ipsilateral hippocampal CA1 neurons. Reduced synaptic plasticity, manifested by impaired long-term potentiation in hippocampal neurons, was also evident in both ipsilateral and contralateral hemispheres, suggesting that ICH also induces functional alterations in distal brain regions remote from the site of injury. In addition, the accumulation of microglia, infiltration of peripheral immune cells, and generation of reactive oxygen species were observed in both contralateral and ipsilateral hemispheres up to 5 wk post-ICH. Furthermore, depletion of microglia using PLX3397, which inhibits colony stimulating factor 1 receptor, ameliorated this delayed cognitive impairment. Collectively, these results suggest that persistent and diffuse brain inflammation may contribute to cognitive impairment in the chronic stage of ICH recovery.-Shi, E., Shi, K., Qiu, S., Sheth, K. N., Lawton, M. T., Ducruet, A. F. Chronic inflammation, cognitive impairment, and distal brain region alteration following intracerebral hemorrhage.


Subject(s)
Cerebral Hemorrhage/immunology , Cognitive Dysfunction/immunology , Inflammation/immunology , Aminopyridines/pharmacology , Animals , Brain/metabolism , Cerebral Hemorrhage/metabolism , Cognition/drug effects , Cognitive Dysfunction/metabolism , Disease Models, Animal , Fingolimod Hydrochloride/pharmacology , Flow Cytometry , Hippocampus/metabolism , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , Neuroimaging , Neuronal Plasticity/drug effects , Pyrroles/pharmacology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism
12.
J Stroke Cerebrovasc Dis ; 29(9): 105057, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32807462

ABSTRACT

BACKGROUND AND AIMS: In experimental models, enhanced inflammation contributes to secondary brain injury in spontaneous intracerebral hemorrhage (ICH). Several inflammatory markers have investigated in humans with inconclusive results. Here, we report the relationship between Systemic Immune-Inflammation (SII) Index and outcome. METHODS: We reviewed the medical records of 239 supratentorial spontaneous ICH patients. Patients were dichotomized based on modified Rankin Scale (mRS) at discharge in good (mRS 0-3) and poor (mRS 4-6) outcome. Demographic, clinical, laboratory and imaging data at admission were compared for both groups. SII index was calculated as [(Platelet counts x Absolute Neutrophil Counts (ANC)/Absolute Lymphocyte Counts (ALC))/1000]. Logistic regression analyses were performed to determine the association between markers of inflammation (ANC, ALC, Platelets, SII index) and outcome adjusting for baseline differences. RESULTS: Sixty-two percent of patients had poor outcome (median [IQR] age= 60 [52-71] years). Patients with poor outcome had lower Glasgow coma scale, larger hematoma volumes, and higher incidence of diabetes and intraventricular extension (p<0.05 for each variable). In univariate analysis, ANC and SII index were independently associated with poor outcome (p<0.05). In multivariate analysis, only SII index remained significantly associated with poor outcome (OR=1.34, 95% CI=1.04-1.72, p=0.02). ROC analysis showed that adjusted SII index is a good discriminator for poor outcome (AUC=0.89, 95% CI=0.84-0.93; P <0.0001), with the best cut-off value being 0.73 (Sensitivity 95%, Specificity 71%). CONCLUSIONS: In patients with supratentorial spontaneous ICH early SII index is an independent predictor of poor outcome at time of hospital discharge.


Subject(s)
Blood Platelets , Cerebral Hemorrhage/diagnosis , Inflammation/diagnosis , Lymphocytes , Neutrophils , Aged , Cerebral Hemorrhage/blood , Cerebral Hemorrhage/immunology , Cerebral Hemorrhage/therapy , Female , Health Status , Humans , Inflammation/blood , Inflammation/immunology , Inflammation/therapy , Lymphocyte Count , Male , Middle Aged , Patient Discharge , Platelet Count , Predictive Value of Tests , Prognosis , Recovery of Function , Reproducibility of Results , Retrospective Studies
13.
J Stroke Cerebrovasc Dis ; 29(8): 104861, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32430234

ABSTRACT

OBJECTIVE: After an intracerebral hemorrhage, there is an immunological reaction, the specific mechanism of which is not fully understood, that seems to contribute to secondary brain injury. In this study, we investigated alterations of inflammatory markers in the blood and clinical outcome after an intracerebral hemorrhage. METHODS: Between July 2013 and February 2016, we performed a prospective study for which we recruited patients who had suffered an intracerebral hemorrhage. Using various scoring scales we evaluated the neurological state upon admission and discharge, and at one and three months following the ICH. During the hospital stay, various inflammatory markers were examined in blood samples. RESULTS: Out of 132 screened patients, 27 were included (48.2% male, mean age 68 years). We found significantly elevated serum concentrations of interleukin-6 (p=0.006) at the time of admission and throughout days three and five. There were also elevated c-reactive protein and granulocyte-colony stimulating factor concentrations found. The concentrations of these immune parameters showed significant monotonic relationships. The ROC analyses showed a better discrimination for mortality with regard to the percentage of T helper cells than with regard to the ICH volume alone. CONCLUSION: Our results may be regarded as preliminary evidence of the occurrence of inflammation after intracerebral hemorrhage. If there is a relationship between inflammation and clinical outcome remains speculative.


Subject(s)
Cerebral Hemorrhage/blood , Inflammation Mediators/blood , Aged , Biomarkers/blood , C-Reactive Protein/metabolism , Cerebral Hemorrhage/diagnosis , Cerebral Hemorrhage/immunology , Cerebral Hemorrhage/therapy , Disability Evaluation , Female , Granulocyte Colony-Stimulating Factor/blood , Humans , Interleukin-6/blood , Male , Patient Admission , Patient Discharge , Prospective Studies , Recovery of Function , Risk Factors , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Time Factors , Treatment Outcome , Up-Regulation
14.
J Neurosci ; 38(40): 8707-8722, 2018 10 03.
Article in English | MEDLINE | ID: mdl-30201775

ABSTRACT

Microcirculatory damage is a common complication for those with vascular risk factors, such as diabetes. To resolve vascular insults, the brain's immune cells (microglia) must rapidly envelop the site of injury. Currently, it is unknown whether Type 1 diabetes, a condition associated with chronic immune system dysfunction, alters microglial responses to damage and what mechanisms are responsible. Using in vivo two-photon microscopy in adult male mice, we show that microglial envelopment of laser-induced cerebral microbleeds is diminished in a hyperglycemic mouse model of Type 1 diabetes, which could not be fully rescued with chronic insulin treatment. Microglia were important for vessel repair because reduced microglial accumulation in diabetic mice or near-complete depletion in healthy controls was associated with greater secondary leakage of the damaged vessel. Broadly suppressing inflammation with dexamethasone in diabetic mice but not healthy controls, significantly enhanced microglial responses to microbleeds and attenuated secondary vessel leakage. These enhancements were associated with changes in IFN-γ signaling because dexamethasone suppressed abnormally high levels of IFN-γ protein levels in brain and blood serum of diabetic mice. Further, blocking IFN-γ in diabetic mice with neutralizing antibodies restored normal microglial chemotaxic responses and purinoceptor P2ry12 gene expression, as well as mitigated secondary leakage. These results suggest that abnormal IFN-γ signaling disrupts microglial function in the diabetic brain, and that immunotherapies targeting IFN-γ can stimulate microglial repair of damaged vessels.SIGNIFICANCE STATEMENT Although Type 1 diabetes is an established risk factor for vascular complications, such as microbleeds, and is known to hinder wound healing in the body, no study has examined how diabetes impacts the brain's innate immune reparative response (involving cells called microglia) to vascular injury. Here we show that microglial responses to brain microbleeds were diminished in diabetic animals, which also exacerbated secondary leakage from damaged vessels. These impairments were related to abnormally high levels of the proinflammatory cytokine IFN-γ because reducing IFN-γ with immunosuppressant drugs or blocking antibodies helped restore normal microglial responses and repair of damaged vessels. These data highlight the use of IFN-γ modulating therapeutics to enhance vascular repair in at-risk populations.


Subject(s)
Cerebral Cortex/immunology , Cerebral Hemorrhage/immunology , Diabetes Mellitus, Type 1/immunology , Interferon-gamma/immunology , Microglia/immunology , Animals , Cerebral Cortex/blood supply , Cerebral Hemorrhage/complications , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/complications , Disease Models, Animal , Encephalitis/blood , Encephalitis/complications , Encephalitis/immunology , Male , Mice, Inbred C57BL , Optical Imaging
15.
J Neuroinflammation ; 16(1): 56, 2019 Mar 05.
Article in English | MEDLINE | ID: mdl-30836997

ABSTRACT

BACKGROUND: Intracerebral hemorrhage (ICH) has a high morbidity and mortality. The peripheral immune system and cross-talk between peripheral blood and brain have been implicated in the ICH immune response. Thus, we delineated the gene networks associated with human ICH in the peripheral blood transcriptome. We also compared the differentially expressed genes in blood following ICH to a prior human study of perihematomal brain tissue. METHODS: We performed peripheral blood whole-transcriptome analysis of ICH and matched vascular risk factor control subjects (n = 66). Gene co-expression network analysis identified groups of co-expressed genes (modules) associated with ICH and their most interconnected genes (hubs). Mixed-effects regression identified differentially expressed genes in ICH compared to controls. RESULTS: Of seven ICH-associated modules, six were enriched with cell-specific genes: one neutrophil module, one neutrophil plus monocyte module, one T cell module, one Natural Killer cell module, and two erythroblast modules. The neutrophil/monocyte modules were enriched in inflammatory/immune pathways; the T cell module in T cell receptor signaling genes; and the Natural Killer cell module in genes regulating alternative splicing, epigenetic, and post-translational modifications. One erythroblast module was enriched in autophagy pathways implicated in experimental ICH, and NRF2 signaling implicated in hematoma clearance. Many hub genes or module members, such as IARS, mTOR, S1PR1, LCK, FYN, SKAP1, ITK, AMBRA1, NLRC4, IL6R, IL17RA, GAB2, MXD1, PIK3CD, NUMB, MAPK14, DDX24, EVL, TDP1, ATG3, WDFY3, GSK3B, STAT3, STX3, CSF3R, PIP4K2A, ANXA3, DGAT2, LRP10, FLOT2, ANK1, CR1, SLC4A1, and DYSF, have been implicated in neuroinflammation, cell death, transcriptional regulation, and some as experimental ICH therapeutic targets. Gene-level analysis revealed 1225 genes (FDR p < 0.05, fold-change > |1.2|) have altered expression in ICH in peripheral blood. There was significant overlap of the 1225 genes with dysregulated genes in human perihematomal brain tissue (p = 7 × 10-3). Overlapping genes were enriched for neutrophil-specific genes (p = 6.4 × 10-08) involved in interleukin, neuroinflammation, apoptosis, and PPAR signaling. CONCLUSIONS: This study delineates key processes underlying ICH pathophysiology, complements experimental ICH findings, and the hub genes significantly expand the list of novel ICH therapeutic targets. The overlap between blood and brain gene responses underscores the importance of examining blood-brain interactions in human ICH.


Subject(s)
Autophagy/physiology , Cerebral Hemorrhage , Cytokines/metabolism , Gene Expression Regulation/physiology , Gene Regulatory Networks , Signal Transduction/physiology , Case-Control Studies , Cerebral Hemorrhage/genetics , Cerebral Hemorrhage/immunology , Cerebral Hemorrhage/pathology , Cytokines/genetics , Female , Gene Expression Profiling , Humans , Immune System , Male , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Transcriptome/physiology
16.
FASEB J ; 32(1): 220-229, 2018 01.
Article in English | MEDLINE | ID: mdl-28877956

ABSTRACT

Severe brain injury significantly influences immune responses; however, the levels at which this influence occurs and which neurogenic pathways are involved are not well defined. Here, we used MRI to measure spleen volume and tissue diffusion changes in patients with intracerebral hemorrhage (ICH). We observed increased capillary exchange and spleen shrinkage by d 3 post-ICH, with recovery by d 14. The extent of spleen shrinkage was associated with brain hematoma size, and a reduced progression of perihematomal edema was observed in the presence of severe spleen shrinkage. At the cellular level, lymphopenia was present in patients with ICH at admission and persisted up to 14 d. Lymphopenia did not parallel the observed spleen alteration. In addition, patients with ICH with infection had significant deficiencies of T and NK cells and poor functional outcomes. Finally, in mouse models of ICH, spleen shrinkage could be related to innervations from adrenergic input and the hypothalamus-pituitary-adrenal (HPA) axis. In sum, the profound impact of ICH on the immune system involves the coordinated actions of sympathetic innervation and the HPA axis, which modulate spleen shrinkage and cellular immunity.-Zhang, J., Shi, K., Li, Z., Li, M., Han, Y., Wang, L., Zhang, Z., Yu, C., Zhang, F., Song, L., Dong, J.-F., La Cava, A., Sheth, K. N., Shi, F.-D. Organ- and cell-specific immune responses are associated with the outcomes of intracerebral hemorrhage.


Subject(s)
Cerebral Hemorrhage/immunology , Aged , Animals , Brain Edema/diagnostic imaging , Cerebral Hemorrhage/diagnostic imaging , Disease Models, Animal , Disease Progression , Female , Hematoma/diagnostic imaging , Humans , Hypothalamo-Hypophyseal System/immunology , Immunity, Cellular , Killer Cells, Natural/immunology , Lymphopenia/immunology , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Middle Aged , Neuroimmunomodulation , Pituitary-Adrenal System/immunology , Spleen/diagnostic imaging , Spleen/immunology , T-Lymphocytes/immunology
17.
J Immunol ; 198(2): 820-831, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27986908

ABSTRACT

Reducing excessive inflammation is beneficial for the recovery from intracerebral hemorrhage (ICH). Here, the roles and mechanisms of A20 (TNFAIP3), an important endogenous anti-inflammatory factor, are examined in ICH. A20 expression in the PBMCs of ICH patients and an ICH mouse model was detected, and the correlation between A20 expression and neurologic deficits was analyzed. A20 expression was increased in PBMCs and was negatively related to the modified Rankin Scale score. A20 expression was also increased in mouse perihematomal tissues. A20-/- and A20-overexpressing mice were generated to further analyze A20 function. Compared with wild-type (WT) mice, A20-/- and A20-overexpressing mice showed significant increases and decreases, respectively, in hematoma volume, neurologic deficit score, mortality, neuronal degeneration, and proinflammatory factors. Moreover, WT-A20-/- parabiosis was established to explore the role of A20 in peripheral blood in ICH injury. ICH-induced damage, including brain edema, neurologic deficit score, proinflammatory factors, and neuronal apoptosis, was reduced in A20-/- parabionts compared with A20-/- mice. Finally, the interactions between TRAF6 and Ubc13 and UbcH5c were increased in A20-/- mice compared with WT mice; the opposite occurred in A20-overexpressing mice. Enhanced IκBα degradation and NF-κB activation were observed in A20-/- mice, but the results were reversed in A20-overexpressing mice. These results suggested that A20 is involved in regulating ICH-induced inflammatory injury in both the central and peripheral system and that A20 reduces ICH-induced inflammation by regulating TRAF6 polyubiquitination. Targeting A20 may thus be a promising therapeutic strategy for ICH.


Subject(s)
Cerebral Hemorrhage/pathology , Inflammation/metabolism , TNF Receptor-Associated Factor 6/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Adult , Aged , Animals , Apoptosis/physiology , Blotting, Western , Cerebral Hemorrhage/immunology , Cerebral Hemorrhage/metabolism , Electrophoretic Mobility Shift Assay , Female , Fluorescent Antibody Technique , Gene Knockdown Techniques , Humans , Immunoprecipitation , Inflammation/immunology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Real-Time Polymerase Chain Reaction , Tumor Necrosis Factor alpha-Induced Protein 3/immunology , Ubiquitination
18.
Immunopharmacol Immunotoxicol ; 41(1): 7-15, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30702002

ABSTRACT

The secondary inflammatory injury following intracerebral hemorrhage (ICH) results in increased morbidity and mortality. White blood cells have been implicated as critical mediators of this inflammatory injury. Currently, no medications have been clinically proven to ameliorate or beneficially modulate inflammation, or to improve outcomes by any mechanism, following ICH. However, other neuroinflammatory conditions, such as multiple sclerosis, have approved pharmacologic therapies that modulate the inflammatory response and minimize the damage caused by inflammatory cells. Thus, there is substantial interest in existing therapies for neuroinflammation and their potential applicability to other acute neurological diseases such as ICH. In this review, we examined the mechanism of action of twelve currently approved medications for multiple sclerosis: alemtuzumab, daclizumab, dimethyl fumarate, fingolimod, glatiramer acetate, interferon beta-1a, interferon beta-1b, mitoxantrone, natalizumab, ocrelizumab, rituximab, teriflunomide. We analyzed the existing literature pertaining to the effects of these medications on various leukocytes and also with emphasis on mechanisms of action during the acute period following initiation of therapy. As a result, we provide a valuable summary of the current body of knowledge regarding these therapies and evidence that supports or refutes their likely promise for treating neuroinflammation following ICH.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Cerebral Hemorrhage/drug therapy , Immunologic Factors/therapeutic use , Models, Theoretical , Multiple Sclerosis/drug therapy , Anti-Inflammatory Agents/administration & dosage , Cerebral Hemorrhage/immunology , Drug Approval , Humans , Immunologic Factors/administration & dosage , Multiple Sclerosis/immunology , Off-Label Use
19.
Bull Exp Biol Med ; 166(6): 714-718, 2019 Apr.
Article in English | MEDLINE | ID: mdl-31020580

ABSTRACT

Specific features of neurological deficit and changes in the cellular composition of tracheal lymphoid structures during the immediate stage (day 1) of hemorrhagic stroke were studied in rats with various behavioral parameters. Modeling of hemorrhage in the left caudate nucleus of the brain was followed by the development of motor disturbances in the forelimb use asymmetry test and corner rotation paradigm. These animals preferred to use the left forelimb (ipsilateral to the side of hemorrhage) to lean on the cylinder wall. The frequency of using the right forelimb or both forelimbs was reduced under these conditions. The number of left-sided rotations increased, while the percentage of right-sided rotations decreased. The observed changes were accompanied by immune dysfunction. It was manifested in the depletion of lymphoid aggregates of the tracheal wall in lymphocytes and plasma cells. The severity of abnormal neurological symptoms and disturbances in immune homeostasis during the immediate stage of hemorrhagic stroke was greater in behaviorally passive rats than in active specimens.


Subject(s)
Caudate Nucleus/physiopathology , Cerebral Hemorrhage/physiopathology , Lymphoid Tissue/physiopathology , Stroke/physiopathology , Trachea/physiopathology , Animals , Behavior, Animal , Caudate Nucleus/immunology , Caudate Nucleus/pathology , Cerebral Hemorrhage/immunology , Cerebral Hemorrhage/pathology , Disease Models, Animal , Functional Laterality , Immunity, Innate , Lymphocyte Count , Lymphocytes/immunology , Lymphocytes/pathology , Lymphoid Tissue/immunology , Lymphoid Tissue/pathology , Male , Motor Activity , Rats , Rats, Wistar , Stroke/immunology , Stroke/pathology , Trachea/immunology , Trachea/pathology
20.
Neurocrit Care ; 28(3): 288-295, 2018 06.
Article in English | MEDLINE | ID: mdl-28620846

ABSTRACT

Mast cells are first responders to intracerebral hemorrhage. They release potent mediators that can disrupt the blood-brain barrier promoting injury, vasogenic edema formation, and hematoma exacerbation. Also, mast cells recruit other inflammatory cells that maintain and amplify brain damage. Given their early role in the cascade of events in intracerebral hemorrhage, mast cells may offer an alternative target for antichemotactic interventions.


Subject(s)
Blood-Brain Barrier , Brain Diseases , Cerebral Hemorrhage , Inflammation , Mast Cells , Animals , Blood-Brain Barrier/immunology , Blood-Brain Barrier/metabolism , Brain Diseases/immunology , Brain Diseases/metabolism , Cerebral Hemorrhage/immunology , Cerebral Hemorrhage/metabolism , Humans , Inflammation/immunology , Inflammation/metabolism , Mast Cells/immunology , Mast Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL