Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 29
1.
J Neuroinflammation ; 21(1): 106, 2024 Apr 24.
Article En | MEDLINE | ID: mdl-38658922

BACKGROUND: Intracerebral hemorrhage (ICH) is a devastating neurological disease causing severe sensorimotor dysfunction and cognitive decline, yet there is no effective treatment strategy to alleviate outcomes of these patients. The Mas axis-mediated neuroprotection is involved in the pathology of various neurological diseases, however, the role of the Mas receptor in the setting of ICH remains to be elucidated. METHODS: C57BL/6 mice were used to establish the ICH model by injection of collagenase into mice striatum. The Mas receptor agonist AVE0991 was administered intranasally (0.9 mg/kg) after ICH. Using a combination of behavioral tests, Western blots, immunofluorescence staining, hematoma volume, brain edema, quantitative-PCR, TUNEL staining, Fluoro-Jade C staining, Nissl staining, and pharmacological methods, we examined the impact of intranasal application of AVE0991 on hematoma absorption and neurological outcomes following ICH and investigated the underlying mechanism. RESULTS: Mas receptor was found to be significantly expressed in activated microglia/macrophages, and the peak expression of Mas receptor in microglia/macrophages was observed at approximately 3-5 days, followed by a subsequent decline. Activation of Mas by AVE0991 post-treatment promoted hematoma absorption, reduced brain edema, and improved both short- and long-term neurological functions in ICH mice. Moreover, AVE0991 treatment effectively attenuated neuronal apoptosis, inhibited neutrophil infiltration, and reduced the release of inflammatory cytokines in perihematomal areas after ICH. Mechanistically, AVE0991 post-treatment significantly promoted the transformation of microglia/macrophages towards an anti-inflammatory, phagocytic, and reparative phenotype, and this functional phenotypic transition of microglia/macrophages by Mas activation was abolished by both Mas inhibitor A779 and Nrf2 inhibitor ML385. Furthermore, hematoma clearance and neuroprotective effects of AVE0991 treatment were reversed after microglia depletion in ICH. CONCLUSIONS: Mas activation can promote hematoma absorption, ameliorate neurological deficits, alleviate neuron apoptosis, reduced neuroinflammation, and regulate the function and phenotype of microglia/macrophages via Akt/Nrf2 signaling pathway after ICH. Thus, intranasal application of Mas agonist ACE0991 may provide promising strategy for clinical treatment of ICH patients.


Hematoma , Hemorrhagic Stroke , Mice, Inbred C57BL , Receptors, G-Protein-Coupled , Recovery of Function , Animals , Mice , Hematoma/drug therapy , Hematoma/pathology , Hematoma/metabolism , Male , Hemorrhagic Stroke/pathology , Hemorrhagic Stroke/drug therapy , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Recovery of Function/drug effects , Recovery of Function/physiology , Proto-Oncogene Proteins/metabolism , Brain Edema/etiology , Brain Edema/metabolism , Brain Edema/drug therapy , Microglia/drug effects , Microglia/metabolism
2.
Exp Neurol ; 374: 114692, 2024 Apr.
Article En | MEDLINE | ID: mdl-38244885

Using cell grafting to direct glia-based repair mechanisms in adult CNS injuries represents a potential therapeutic strategy for supporting functional neural parenchymal repair. However, glia repair directed by neural progenitor cell (NPC) grafts is dramatically altered by increasing lesion size, severity, and mode of injury. To address this, we studied the interplay between astrocyte differentiation and cell proliferation of NPC in vitro to generate proliferating immature astrocytes (ImA) using hysteretic conditioning. ImA maintain proliferation rates at comparable levels to NPC but showed robust immature astrocyte marker expression including Gfap and Vimentin. ImA demonstrated enhanced resistance to myofibroblast-like phenotypic transformations upon exposure to serum enriched environments in vitro compared to NPC and were more effective at scratch wound closure in vitro compared to quiescent astrocytes. Glia repair directed by ImA at acute ischemic striatal stroke lesions was equivalent to NPC but better than quiescent astrocyte grafts. While ischemic injury environments supported enhanced survival of grafts compared to healthy striatum, hemorrhagic lesions were hostile towards both NPC and ImA grafts leading to poor survival and ineffective modulation of natural wound repair processes. Our findings demonstrate that lesion environments, rather than transcriptional pre-graft states, determine the survival, cell-fate, and glia repair competency of cell grafts applied to acute CNS injuries.


Hemorrhagic Stroke , Neural Stem Cells , Stroke , Humans , Astrocytes/metabolism , Neurons/metabolism , Hemorrhagic Stroke/metabolism , Hemorrhagic Stroke/pathology , Neural Stem Cells/pathology , Stroke/surgery , Stroke/metabolism , Cell Differentiation
3.
J Neuroinflammation ; 20(1): 82, 2023 Mar 21.
Article En | MEDLINE | ID: mdl-36944982

BACKGROUND: Central post-stroke pain (CPSP) is an intractable and disabling central neuropathic pain that severely affects patients' lives, well-being, and socialization abilities. However, CPSP has been poorly studied mechanistically and its treatment remains challenging. Here, we used a rat model of CPSP induced by thalamic hemorrhage to investigate its underlying mechanisms and the effect of stellate ganglion block (SGB) on CPSP and emotional comorbidities. METHODS: Thalamic hemorrhage was produced by injecting collagenase IV into the ventral-posterolateral nucleus (VPL) of the right thalamus. The up-and-down method with von Frey hairs was used to measure the mechanical allodynia. Behavioral tests were carried out to examine depressive and anxiety-like behaviors including the open field test (OFT), elevated plus maze test (EPMT), novelty-suppressed feeding test (NSFT), and forced swim test (FST). The peri-thalamic lesion tissues were collected for immunofluorescence, western blotting, and enzyme-linked immunosorbent assay (ELISA). Genetic knockdown of thalamic hypoxia-inducible factor-1α (HIF-1α) and NOD-like receptor thermal protein domain associated protein 3 (NLRP3) with microinjection of HIF-1α siRNA and NLRP3 siRNA into the VPL of thalamus were performed 3 days before collagenase injection into the same regions. Microinjection of lificiguat (YC-1) and MCC950 into the VPL of thalamus were administrated 30 min before the collagenase injection in order to inhibited HIF-1α and NLRP3 pharmacologically. Repetitive right SGB was performed daily for 5 days and laser speckle contrast imaging (LSCI) was conducted to examine cerebral blood flow. RESULTS: Thalamic hemorrhage caused persistent mechanical allodynia and anxiety- and depression-like behaviors. Accompanying the persistent mechanical allodynia, the expression of HIF-1α and NLRP3, as well as the activities of microglia and astrocytes in the peri-thalamic lesion sites, were significantly increased. Genetic knockdown of thalamic HIF-1α and NLRP3 significantly attenuated mechanical allodynia and anxiety- and depression-like behaviors following thalamic hemorrhage. Further studies revealed that intra-thalamic injection of YC-1, or MCC950 significantly suppressed the activation of microglia and astrocytes, the release of pro-inflammatory cytokines, the upregulation of malondialdehyde (MDA), and the downregulation of superoxide dismutase (SOD), as well as mechanical allodynia and anxiety- and depression-like behaviors following thalamic hemorrhage. In addition, repetitive ipsilateral SGB significantly restored the upregulated HIF-1α/NLRP3 signaling and the hyperactivated microglia and astrocytes following thalamic hemorrhage. The enhanced expression of pro-inflammatory cytokines and the oxidative stress in the peri-thalamic lesion sites were also reversed by SGB. Moreover, LSCI showed that repetitive SGB significantly increased cerebral blood flow following thalamic hemorrhage. Most strikingly, SGB not only prevented, but also reversed the development of mechanical allodynia and anxiety- and depression-like behaviors induced by thalamic hemorrhage. However, pharmacological activation of thalamic HIF-1α and NLRP3 with specific agonists significantly eliminated the therapeutic effects of SGB on mechanical allodynia and anxiety- and depression-like behaviors following thalamic hemorrhage. CONCLUSION: This study demonstrated for the first time that SGB could improve CPSP with comorbid anxiety and depression by increasing cerebral blood flow and inhibiting HIF-1α/NLRP3 inflammatory signaling.


Hemorrhagic Stroke , Neuralgia , Stroke , Rats , Animals , Hyperalgesia/drug therapy , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Hemorrhagic Stroke/complications , Hemorrhagic Stroke/pathology , Depression/etiology , Depression/therapy , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Stellate Ganglion/metabolism , Stellate Ganglion/pathology , Rats, Sprague-Dawley , Stroke/pathology , Thalamus/metabolism , Cerebral Hemorrhage/pathology , Neuralgia/metabolism , Anxiety , Collagenases/metabolism , Cytokines/metabolism
4.
Mol Neurobiol ; 59(5): 3040-3051, 2022 May.
Article En | MEDLINE | ID: mdl-35258849

As one form of stroke, intracerebral hemorrhage (ICH) is a fatal cerebrovascular disease, which has high morbidity and mortality and lacks effective medical treatment. Increased infiltration of inflammatory cytokines coupled with pyroptotic cell death is involved in the pathophysiological process of ICH. However, little is known about whether concomitant fracture patients have the same progression of inflammation and pyroptosis. Hence, we respectively established the mouse ICH model and ICH with bilateral tibial fracture model (MI) to explore the potential cross-talk between the above two injuries. We found that MI obviously reversed the expressions of pyroptosis-associated proteins, which were remarkably up-regulated at the acute phase after ICH. Similar results were observed in neuronal expressions via double immunostaining. Furthermore, brain edema was also significantly alleviated in mice who suffered MI, when compared with ICH alone. To better clarify the potential mechanisms that mediated this cross-talk, recombinant mouse interleukin-13 (IL-13) was used to investigate its effect on pyroptosis in the mouse MI model, in which a lower level of IL-13 was observed. Remarkably, IL-13 administration re-awakened cell death, which was mirrored by the re-upregulation of pyroptosis-associated proteins and PI-positive cell counts. The results of hemorrhage volume and behavioral tests further confirmed its critical role in regulating neurological functions. Besides, the IL-13-treated MI group showed poor outcomes of fracture healing. To sum up, our research indicates that controlling the IL-13 content in the acute phase would be a promising target in influencing the outcomes of brain injury and fracture, and meanwhile, provides new evidence in repairing compound injuries in clinics.


Hemorrhagic Stroke , Interleukin-13 , Tibial Fractures , Animals , Cerebral Hemorrhage/complications , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/pathology , Disease Models, Animal , Hemorrhagic Stroke/pathology , Humans , Interleukin-13/pharmacology , Mice , Pyroptosis/drug effects , Tibial Fractures/complications , Tibial Fractures/metabolism , Tibial Fractures/pathology
5.
Cerebrovasc Dis ; 51(4): 461-472, 2022.
Article En | MEDLINE | ID: mdl-34983048

INTRODUCTION: Stroke is characterized by deleterious oxidative stress. Selenoprotein enzymes are essential endogenous antioxidants, and detailed insight into their role after stroke could define new therapeutic treatments. This systematic review aimed to elucidate how blood selenoprotein concentration and activity change in the acute phase of stroke. METHODS: We searched PubMed, EMBASE, and Medline databases for studies measuring serial blood selenoprotein concentration or activity in acute stroke patients or in stroke patients compared to non-stroke controls. Meta-analyses of studies stratified by the type of stroke, blood compartment, and type of selenoprotein measurement were conducted. RESULTS: Eighteen studies and data from 941 stroke patients and 708 non-stroke controls were included in this review. Glutathione peroxidase (GPx) was the only identified selenoprotein, and its activity was most frequently measured. Results from 12 studies and 693 patients showed that compared to non-stroke controls in acute ischaemic stroke patients, the GPx activity increased in haemolysate (standardized mean difference [SMD]: 0.27, 95% CI: 0.07-0.47) but decreased in plasma (mean difference [MD]: -1.08 U/L, 95% CI: -1.94 to -0.22) and serum (SMD: -0.54, 95% CI: -0.91 to -0.17). From 4 identified studies in 106 acute haemorrhagic stroke patients, the GPx activity decreased in haemolysate (SMD: -0.40, 95% CI: -0.68 to -0.13) and remained unchanged in plasma (MD: -0.10 U/L, 95% CI: -0.81 to 0.61) and serum (MD: -5.00 U/mL, 95% CI: -36.17 to 26.17) compared to non-stroke controls. Results from studies assessing the GPx activity in the haemolysate compartment were inconsistent and characterized by high heterogeneity. CONCLUSIONS: Our results suggest a reduction of the blood GPx activity in acute ischaemic stroke patients, a lack of evidence regarding a role for GPx in haemorrhagic stroke patients, and insufficient evidence for other selenoproteins.


Brain Ischemia , Hemorrhagic Stroke , Ischemic Stroke , Selenoproteins , Antioxidants , Brain Ischemia/diagnosis , Brain Ischemia/pathology , Glutathione Peroxidase , Hemorrhagic Stroke/diagnosis , Hemorrhagic Stroke/pathology , Humans , Ischemic Stroke/diagnosis , Ischemic Stroke/pathology , Selenium , Selenoproteins/metabolism
6.
Microvasc Res ; 140: 104300, 2022 03.
Article En | MEDLINE | ID: mdl-34953822

Capillary density rarefaction and endothelial dysfunction contribute to chronic hypoperfusion and cerebral small vessel disease. Previous animal experiments revealed spatiotemporal microvascular remodeling directing post-stroke brain reorganization. We hypothesized that microcirculatory changes during acute cerebrovascular events could be reflected systemically and visualized sublingually. In a prospective observational trial in vivo sublingual sidestream darkfield videomicroscopy was performed in twenty-one patients with either acute stroke (n = 13 ischemic, n = 1 ischemic with hemorrhagic transformation and n = 2 hemorrhagic stroke) or transitory ischemic attacks (n = 5) within 24 h after hospital admission and compared to an age- and sex-matched control group. Repetitive measurements were performed on the third day and after one week. Functional and perfused total capillary density was rarefied in the overall patient group (3060 vs 3717 µm/mm2, p = 0.001 and 5263 vs 6550 µm/mm2, p = 0.002, respectively) and in patients with ischemic strokes (2897 vs. 3717 µm/mm2, p < 0.001 and 5263 vs. 6550 µm/mm2, p = 0.006, respectively) when compared to healthy controls. The perfused boundary region (PBR), which was measured as an inverse indicator of glycocalyx thickness, was markedly related to red blood cell (RBC) filling percentage (regarded as an estimate of microvessel perfusion) in the overall patient group (r = -0.843, p < 0.001), in patients with ischemic strokes (r = -0.82, p = 0.001) as well as in healthy volunteers (r = -0.845, p < 0.001). In addition, there were significant associations between platelet count or platelet aggregation values (as measured by whole blood impedance aggregometry) and microvascular parameters in the overall patient collective, as well as in patients with ischemic strokes. In conclusion, cerebrovascular events are associated with altered systemic microvascular perfusion.


Capillaries/pathology , Hemorrhagic Stroke/pathology , Ischemic Attack, Transient/pathology , Ischemic Stroke/pathology , Microvascular Rarefaction , Mouth Floor/blood supply , Aged , Aged, 80 and over , Capillaries/physiopathology , Female , Hemorrhagic Stroke/diagnostic imaging , Hemorrhagic Stroke/physiopathology , Humans , Ischemic Attack, Transient/diagnostic imaging , Ischemic Attack, Transient/physiopathology , Ischemic Stroke/diagnostic imaging , Ischemic Stroke/physiopathology , Male , Microcirculation , Microscopy, Video , Middle Aged , Platelet Aggregation , Prospective Studies
7.
Int J Mol Sci ; 22(18)2021 Sep 13.
Article En | MEDLINE | ID: mdl-34576050

Intracerebral hemorrhage (ICH) occurs when brain blood vessels rupture, causing inflammation and cell death. 2-Fucosyllactose (2FL), a human milk oligosaccharide, has potent antiapoptotic and anti-inflammatory effects. The purpose of this study was to examine the protective effect of 2FL in cellular and rodent models of ICH. Hemin was added to a primary rat cortical neuronal and BV2 microglia coculture to simulate ICH in vitro. IBA1 and MAP2 immunoreactivities were used to determine inflammation and neuronal survival. Hemin significantly increased IBA1, while it reduced MAP2 immunoreactivity. 2FL significantly antagonized both responses. The protective effect of 2FL was next examined in a rat ICH model. Intracerebral administration of type VII collagenase reduced open-field locomotor activity. Early post-treatment with 2FL significantly improved locomotor activity. Brain tissues were collected for immunohistochemistry and qRT-PCR analysis. 2FL reduced IBA1 and CD4 immunoreactivity in the lesioned striatum. 2FL downregulated the expression of ER stress markers (PERK and CHOP), while it upregulated M2 macrophage markers (CD206 and TGFß) in the lesioned brain. Taken together, our data support that 2FL has a neuroprotective effect against ICH through the inhibition of neuroinflammation and ER stress. 2FL may have clinical implications for the treatment of ICH.


Calcium-Binding Proteins/genetics , Hemorrhagic Stroke/drug therapy , Microfilament Proteins/genetics , Microtubule-Associated Proteins/genetics , Trisaccharides/pharmacology , Animals , Cell Line , Coculture Techniques , Collagenases/toxicity , Disease Models, Animal , Gene Expression Regulation , Hemin/toxicity , Hemorrhagic Stroke/chemically induced , Hemorrhagic Stroke/genetics , Hemorrhagic Stroke/pathology , Humans , Locomotion/drug effects , Microglia/drug effects , Microglia/pathology , Milk, Human/chemistry , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Oligosaccharides/chemistry , Oligosaccharides/pharmacology , Rats , Trisaccharides/chemistry
8.
Sci Rep ; 11(1): 15364, 2021 07 28.
Article En | MEDLINE | ID: mdl-34321571

Sodium-glucose cotransporter 2 (SGLT2) inhibitors have shown impressive effects in reducing major vascular events in several randomized controlled trials (RCTs). The purpose of this study was to perform a meta-analysis to evaluate the effect of SGLT2 inhibitors on the risk of stroke and its subtypes. All data from prospective RCTs up to 20 October 2020 involving SGLT2 inhibitors that reported stroke events as the primary endpoint or safety in subjects with type 2 diabetes were subjected to meta-analysis. Five eligible RCTs (EMPA-REG, CANVAS, DECLARE-TIMI 58, CREDENCE and VERTIS CV) involving 46,969 participants were included. Pooled analysis of the RCTs showed no significant effect of SGLT2 inhibitors on total stroke [risk ratio (RR) = 0.95; 95% confidence interval (CI) 0.79-1.13, P = 0.585]. Subgroup analysis indicated that SGLT2 inhibitors had no significant effect against fatal stroke, non-fatal stroke, ischemic stroke or transient ischemic attack. When only hemorrhagic stroke was included, SGLT2 inhibitors were associated with a significant 50% reduction compared with placebo (RR = 0.49, 95% CI 0.30-0.82, P = 0.007). This meta-analysis shows that SGLT2 inhibitors have a neutral effect on the risk of stroke and its subtypes but a potential protective effect against hemorrhagic stroke.


Diabetes Mellitus, Type 2/drug therapy , Hemorrhagic Stroke/drug therapy , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Sodium-Glucose Transporter 2/genetics , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Hemorrhagic Stroke/genetics , Hemorrhagic Stroke/pathology , Hemorrhagic Stroke/prevention & control , Humans , Randomized Controlled Trials as Topic , Risk Assessment , Sodium-Glucose Transporter 2 Inhibitors/adverse effects
9.
Stroke ; 52(11): 3613-3623, 2021 11.
Article En | MEDLINE | ID: mdl-34281379

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.


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
10.
J Cereb Blood Flow Metab ; 41(12): 3232-3247, 2021 12.
Article En | MEDLINE | ID: mdl-34187233

Hemorrhagic stroke is a leading cause of death. The causes of intracerebral hemorrhage (ICH)-induced brain damage are thought to include lysis of red blood cells, hemin release and iron overload. These mechanisms, however, have not proven very amenable to therapeutic intervention, and so other mechanistic targets are being sought. Here we report that accumulation of endogenously formed zinc protoporphyrin (ZnPP) also critically contributes to ICH-induced brain damage. ICH caused a significant accumulation of ZnPP in brain tissue surrounding hematoma, as evidenced by fluorescence microscopy of ZnPP, and further confirmed by fluorescence spectroscopy and supercritical fluid chromatography-mass spectrometry. ZnPP formation was dependent upon both ICH-induced hypoxia and an increase in free zinc accumulation. Notably, inhibiting ferrochelatase, which catalyzes insertion of zinc into protoporphyrin, greatly decreased ICH-induced endogenous ZnPP generation. Moreover, a significant decrease in brain damage was observed upon ferrochelatase inhibition, suggesting that endogenous ZnPP contributes to the damage in ICH. Our findings reveal a novel mechanism of ICH-induced brain damage through ferrochelatase-mediated formation of ZnPP in ICH tissue. Since ferrochelatase can be readily inhibited by small molecules, such as protein kinase inhibitors, this may provide a promising new and druggable target for ICH therapy.


Brain Injuries/metabolism , Hemorrhagic Stroke/metabolism , Protoporphyrins/metabolism , Animals , Brain Injuries/pathology , Hemorrhagic Stroke/pathology , Male , Mice
11.
Gene ; 790: 145696, 2021 Jul 20.
Article En | MEDLINE | ID: mdl-33964377

BACKGROUND AND AIMS: Hemorrhagic stroke (HS) results in significant mortality and disability worldwide. Angiotensin Converting Enzyme (ACE) is responsible for blood pressure regulation and vascular homeostasis. Our objective was to conduct a comprehensive meta-analysis for ascertaining the association of ACE I/D polymorphism with HS since a number of studies depicted inconclusive evidence. METHODS: Literature search was performed till July 10, 2020 in PubMed, EMBASE, Cochrane, Chinese National Knowledge Information and Google Scholar databases with keywords: ('Angiotensin Converting Enzyme' OR 'ACE') AND ('Single Nucleotide polymorphisms' OR 'SNP') AND ('Hemorrhagic stroke or 'HS'). Pooled Odds Ratio (OR) and 95% Confidence Interval (CI) were determined for gene-disease association using either fixed (when I2 < 50%) or random effect (when I2 > 50%) models. Risk of bias in studies was assessed using funnel plots and sensitivity analyses. Statistical analysis was performed using STATA version 13.0 software. RESULTS: A total of 53 studies having 5186 HS and 7347 healthy control subjects were included in our meta-analysis. Pooled analyses showed that ACE I/D gene polymorphism had significant association with risk of HS in overall study population [(dominant model: OR = 1.29, 95% CI = 1.12-1.50 & recessive model: OR = 1.79, 95% CI = 1.46-2.20)]. Population subgroup analyses further revealed significant relationship of ACE I/D polymorphism with ICH in Asians (recessive: OR 1.97, 95% CI = 1.57-2.47) but not in Caucasians (recessive: OR 1.02, 95% CI = 0.76-1.36). CONCLUSION: This meta-analysis suggests that ACE I/D polymorphism may lead to risk of HS and can be a potential biomarker for HS susceptibility especially in Asian population.


Hemorrhagic Stroke/etiology , INDEL Mutation , Peptidyl-Dipeptidase A/genetics , Hemorrhagic Stroke/pathology , Humans
12.
Mayo Clin Proc ; 96(6): 1639-1654, 2021 06.
Article En | MEDLINE | ID: mdl-33952393

Spontaneous intracerebral hemorrhage (ICH) is a medical emergency and is disproportionately associated with higher mortality and long-term disability compared with ischemic stroke. The phrase "time is brain" was derived for patients with large vessel occlusion ischemic stroke in which approximately 1.9 million neurons are lost every minute. Similarly, this statement holds true for ICH patients due to a high volume of neurons that are damaged at initial onset and during hematoma expansion. Most cases of spontaneous ICH pathophysiologically stem from chronic hypertension and rupture of small perforating vessels off of larger cerebral arteries supplying deep brain structures, with cerebral amyloid angiopathy being another cause for lobar hemorrhages in older patients. Optimal ICH medical management strategies include timely diagnosis, aggressive blood pressure control, correction of underlying coagulopathy defects if present, treatment of cerebral edema, and continuous assessment for possible surgical intervention. Current strategies in the surgical management of ICH include newly developed minimally invasive techniques for hematoma evacuation, with the goal of mitigating injury to fiber tracts while accessing the clot. We review evidence-based medical and surgical management of spontaneous ICH with the overall goal of reducing neurologic injury and optimizing functional outcome.


Hemorrhagic Stroke/diagnosis , Hemorrhagic Stroke/mortality , Hemorrhagic Stroke/pathology , Hemorrhagic Stroke/therapy , Humans , Thrombolytic Therapy/methods , Treatment Outcome
13.
Sci Rep ; 11(1): 10071, 2021 05 12.
Article En | MEDLINE | ID: mdl-33980906

We research into the clinical, biochemical and neuroimaging factors associated with the outcome of stroke patients to generate a predictive model using machine learning techniques for prediction of mortality and morbidity 3-months after admission. The dataset consisted of patients with ischemic stroke (IS) and non-traumatic intracerebral hemorrhage (ICH) admitted to Stroke Unit of a European Tertiary Hospital prospectively registered. We identified the main variables for machine learning Random Forest (RF), generating a predictive model that can estimate patient mortality/morbidity according to the following groups: (1) IS + ICH, (2) IS, and (3) ICH. A total of 6022 patients were included: 4922 (mean age 71.9 ± 13.8 years) with IS and 1100 (mean age 73.3 ± 13.1 years) with ICH. NIHSS at 24, 48 h and axillary temperature at admission were the most important variables to consider for evolution of patients at 3-months. IS + ICH group was the most stable for mortality prediction [0.904 ± 0.025 of area under the receiver operating characteristics curve (AUC)]. IS group presented similar results, although variability between experiments was slightly higher (0.909 ± 0.032 of AUC). ICH group was the one in which RF had more problems to make adequate predictions (0.9837 vs. 0.7104 of AUC). There were no major differences between IS and IS + ICH groups according to morbidity prediction (0.738 and 0.755 of AUC) but, after checking normality with a Shapiro Wilk test with the null hypothesis that the data follow a normal distribution, it was rejected with W = 0.93546 (p-value < 2.2e-16). Conditions required for a parametric test do not hold, and we performed a paired Wilcoxon Test assuming the null hypothesis that all the groups have the same performance. The null hypothesis was rejected with a value < 2.2e-16, so there are statistical differences between IS and ICH groups. In conclusion, machine learning algorithms RF can be effectively used in stroke patients for long-term outcome prediction of mortality and morbidity.


Algorithms , Cerebral Hemorrhage/mortality , Hemorrhagic Stroke/mortality , Hospitalization/statistics & numerical data , Ischemic Stroke/mortality , Severity of Illness Index , Aged , Cerebral Hemorrhage/pathology , Cerebral Hemorrhage/therapy , Female , Follow-Up Studies , Hemorrhagic Stroke/pathology , Hemorrhagic Stroke/therapy , Humans , Ischemic Stroke/pathology , Ischemic Stroke/therapy , Machine Learning , Male , Prognosis , Prospective Studies , ROC Curve , Survival Rate
14.
J Pediatr Hematol Oncol ; 43(3): e438-e444, 2021 04 01.
Article En | MEDLINE | ID: mdl-32011561

Pediatric stroke is an event caused by disturbance of cerebral circulation that occurs in individuals between 28 days and 18 years of age. Although an uncommon event, pediatric stroke still carries significant morbidity and mortality. Unlike adults, causes of pediatric stroke are various and include vascular, infectious, hematologic, neoplastic, and toxic etiologies. Clinical presentation of nontraumatic intracerebral hemorrhages in older children is similar to adults, however in neonates and infants signs and symptoms can be more subtle, especially with smaller hemorrhages. Management of nontraumatic intracerebral hemorrhage consists of stabilizing the patient, management of the hemorrhage itself, and reduction of the rebleeding risk. Even so, when child reaches a medical care, morbidity and mortality rates are still high. We described a case series of pediatric patients with intracerebral nontraumatic hemorrhagic stroke from different etiologies. Although increasingly recognized, such situations are still poorly described in children and our report offers a good overview on this topic.


Cerebral Hemorrhage/pathology , Hemorrhagic Stroke/pathology , Cerebral Hemorrhage/diagnostic imaging , Cerebral Hemorrhage/etiology , Cerebral Hemorrhage/therapy , Child , Child, Preschool , Disease Management , Female , Hemorrhagic Stroke/diagnostic imaging , Hemorrhagic Stroke/etiology , Hemorrhagic Stroke/therapy , Humans , Infant , Male , Tomography, X-Ray Computed
15.
Naunyn Schmiedebergs Arch Pharmacol ; 394(1): 73-84, 2021 01.
Article En | MEDLINE | ID: mdl-32808069

PURPOSE: Intracranial hemorrhage (ICH) is a devastating disease with high mortality and morbidity. After ICH, iron released from the hematoma plays a crucial role in secondary brain injury. Deferasirox (DFR) is a trivalent iron chelator, which was approved to treat iron overload syndrome after transfusion. The aim of the present study was to investigate the protective effects of DFR in both in vitro and in vivo ICH models. METHODS: Using a hemin-induced SH-SY5Y cell damage model, we performed an intracellular bivalent iron (Fe2+) accumulation assay, cell death assay, oxidative stress assessments, and Western blotting analysis. Moreover, the effects of DFR intraventricular administration on hematoma, neurological deficits, and histological alteration were evaluated in an in vivo ICH mouse model by collagenase. RESULTS: DFR significantly suppressed the intracellular Fe2+ accumulation and cell death caused by hemin exposure. These effects were related to the suppression of both reactive oxygen species and lipid peroxidation over-production. In Western blotting analysis, hemin increased the expression of ferritin (an iron storage protein), LC3 and p62 (autophagy-related markers), phosphorylated p38 (a stress response protein), and cleaved-caspase3 and cleaved-poly (adenosine diphosphate ribose) polymerase (PARP) (apoptosis-related makers). However, DFR suppressed the increase of these proteins. In addition, DFR attenuated the neurological deficits until 7 days after ICH without affecting hematoma and injury area. Furthermore, DFR also suppressed microglia/macrophage activation in peri-hematoma area at 3 days after ICH. CONCLUSION: These findings indicate that DFR might be a useful therapeutic agent for the therapy of ICH.


Antioxidants/therapeutic use , Deferasirox/therapeutic use , Hemorrhagic Stroke/drug therapy , Iron Chelating Agents/therapeutic use , Neuroprotective Agents/therapeutic use , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Brain/drug effects , Brain/pathology , Cell Death/drug effects , Cell Line, Tumor , Deferasirox/pharmacology , Disease Models, Animal , Hemorrhagic Stroke/pathology , Humans , Iron/metabolism , Iron Chelating Agents/pharmacology , Male , Mice , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Reactive Oxygen Species/metabolism
16.
Sci Rep ; 10(1): 22013, 2020 12 16.
Article En | MEDLINE | ID: mdl-33328490

High intracranial pressure (ICP) can impede cerebral blood flow resulting in secondary injury or death following severe stroke. Compensatory mechanisms include reduced cerebral blood and cerebrospinal fluid volumes, but these often fail to prevent raised ICP. Serendipitous observations in intracerebral hemorrhage (ICH) suggest that neurons far removed from a hematoma may shrink as an ICP compliance mechanism. Here, we sought to critically test this observation. We tracked the timing of distal tissue shrinkage (e.g. CA1) after collagenase-induced striatal ICH in rat; cell volume and density alterations (42% volume reduction, 34% density increase; p < 0.0001) were highest day one post-stroke, and rebounded over a week across brain regions. Similar effects were seen in the filament model of middle cerebral artery occlusion (22% volume reduction, 22% density increase; p ≤ 0.007), but not with the Vannucci-Rice model of hypoxic-ischemic encephalopathy (2.5% volume increase, 14% density increase; p ≥ 0.05). Concerningly, this 'tissue compliance' appears to cause sub-lethal damage, as revealed by electron microscopy after ICH. Our data challenge the long-held assumption that 'healthy' brain tissue outside the injured area maintains its volume. Given the magnitude of these effects, we posit that 'tissue compliance' is an important mechanism invoked after severe strokes.


Cerebral Hemorrhage/pathology , Hemorrhagic Stroke/pathology , Ischemic Stroke/pathology , Models, Biological , Animals , Astrocytes/pathology , CA1 Region, Hippocampal/pathology , CA1 Region, Hippocampal/ultrastructure , Cell Size , Male , Neurons/pathology , Rats, Sprague-Dawley
17.
Stroke Vasc Neurol ; 5(4): 388-395, 2020 Dec.
Article En | MEDLINE | ID: mdl-33376200

Spontaneous intracerebral haemorrhage (ICH) is a devastating type of stroke with high mortality and morbidity and for which no effective treatments are available to date. Much experimental and clinical research have been performed to explore its mechanisms regard the subsequent inflammatory cascade and to seek the potential therapeutic strategies. The aim of this review is to discuss insights from clinical settings that have led to the development of numerous animal models of ICH. Some of the current and future challenges for clinicians to understand ICH are also surveyed.


Cerebral Hemorrhage , Hemorrhagic Stroke , Animals , Cerebral Hemorrhage/etiology , Cerebral Hemorrhage/pathology , Cerebral Hemorrhage/physiopathology , Cerebral Hemorrhage/therapy , Disease Models, Animal , Disease Progression , Hemorrhagic Stroke/etiology , Hemorrhagic Stroke/pathology , Hemorrhagic Stroke/physiopathology , Hemorrhagic Stroke/therapy , Humans , Prognosis
18.
Proc Natl Acad Sci U S A ; 117(51): 32679-32690, 2020 12 22.
Article En | MEDLINE | ID: mdl-33293423

Intracerebral hemorrhage (ICH) is a devastating form of stroke affecting millions of people worldwide. Parenchymal hematoma triggers a series of reactions leading to primary and secondary brain injuries and permanent neurological deficits. Microglia and macrophages carry out hematoma clearance, thereby facilitating functional recovery after ICH. Here, we elucidate a pivotal role for the interleukin (IL)-4)/signal transducer and activator of transcription 6 (STAT6) axis in promoting long-term recovery in both blood- and collagenase-injection mouse models of ICH, through modulation of microglia/macrophage functions. In both ICH models, STAT6 was activated in microglia/macrophages (i.e., enhanced expression of phospho-STAT6 in Iba1+ cells). Intranasal delivery of IL-4 nanoparticles after ICH hastened STAT6 activation and facilitated hematoma resolution. IL-4 treatment improved long-term functional recovery in young and aged male and young female mice. In contrast, STAT6 knockout (KO) mice exhibited worse outcomes than WT mice in both ICH models and were less responsive to IL-4 treatment. The construction of bone marrow chimera mice demonstrated that STAT6 KO in either the CNS or periphery exacerbated ICH outcomes. STAT6 KO impaired the capacity of phagocytes to engulf red blood cells in the ICH brain and in primary cultures. Transcriptional analyses identified lower level of IL-1 receptor-like 1 (ST2) expression in microglia/macrophages of STAT6 KO mice after ICH. ST2 KO diminished the beneficial effects of IL-4 after ICH. Collectively, these data confirm the importance of IL-4/STAT6/ST2 signaling in hematoma resolution and functional recovery after ICH. Intranasal IL-4 treatment warrants further investigation as a clinically feasible therapy for ICH.


Cerebral Hemorrhage/metabolism , Hematoma/metabolism , Hemorrhagic Stroke/metabolism , Interleukin-4/metabolism , STAT6 Transcription Factor/metabolism , Animals , Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/pathology , Disease Models, Animal , Female , Hematoma/drug therapy , Hematoma/pathology , Hemorrhagic Stroke/drug therapy , Hemorrhagic Stroke/pathology , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-4/administration & dosage , Interleukin-4/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Morris Water Maze Test/drug effects , Phagocytosis/drug effects , Phagocytosis/physiology , Rotarod Performance Test , STAT6 Transcription Factor/genetics , Signal Transduction
19.
Sci Rep ; 10(1): 18170, 2020 10 23.
Article En | MEDLINE | ID: mdl-33097782

Stroke is caused by obstructed blood flow (ischaemia) or unrestricted bleeding in the brain (haemorrhage). Global brain ischaemia occurs after restricted cerebral blood flow e.g. during cardiac arrest. Following ischaemic injury, restoration of blood flow causes ischaemia-reperfusion (I/R) injury which worsens outcome. Secondary injury mechanisms after any stroke are similar, and encompass inflammation, endothelial dysfunction, blood-brain barrier (BBB) damage and apoptosis. We developed a new model of transient global forebrain I/R injury (dual carotid artery ligation; DCAL) and compared the manifestations of this injury with those in a conventional I/R injury model (middle-cerebral artery occlusion; MCAo) and with intracerebral haemorrhage (ICH; collagenase model). MRI revealed that DCAL produced smaller bilateral lesions predominantly localised to the striatum, whereas MCAo produced larger focal corticostriatal lesions. After global forebrain ischaemia mice had worse overall neurological scores, although quantitative locomotor assessment showed MCAo and ICH had significantly worsened mobility. BBB breakdown was highest in the DCAL model while apoptotic activity was highest after ICH. VCAM-1 upregulation was specific to ischaemic models only. Differential transcriptional upregulation of pro-inflammatory chemokines and cytokines and TLRs was seen in the three models. Our findings offer a unique insight into the similarities and differences in how biological processes are regulated after different types of stroke. They also establish a platform for analysis of therapies such as endothelial protective and anti-inflammatory agents that can be applied to all types of stroke.


Cerebrovascular Circulation/physiology , Hemorrhagic Stroke/pathology , Ischemic Stroke/pathology , Prosencephalon/blood supply , Reperfusion Injury/pathology , Animals , Anti-Inflammatory Agents/therapeutic use , Apoptosis/immunology , Blood-Brain Barrier/diagnostic imaging , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Carotid Arteries/physiopathology , Cerebrovascular Circulation/drug effects , Collagenases/administration & dosage , Collagenases/adverse effects , Cytokines/genetics , Cytokines/immunology , Disease Models, Animal , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Hemorrhagic Stroke/drug therapy , Hemorrhagic Stroke/immunology , Hemorrhagic Stroke/physiopathology , Humans , Ischemic Stroke/drug therapy , Ischemic Stroke/immunology , Ischemic Stroke/physiopathology , Ligation , Locomotion/physiology , Magnetic Resonance Imaging , Male , Mice , Middle Cerebral Artery/physiopathology , Prosencephalon/diagnostic imaging , Prosencephalon/drug effects , Prosencephalon/pathology , Protective Agents/therapeutic use , Reperfusion Injury/drug therapy , Reperfusion Injury/immunology , Reperfusion Injury/physiopathology , Toll-Like Receptors/genetics , Transcriptional Activation/immunology
20.
JCI Insight ; 5(20)2020 10 15.
Article En | MEDLINE | ID: mdl-33055425

Thalamic pain, a type of central poststroke pain, frequently occurs following ischemia/hemorrhage in the thalamus. Current treatment of this disorder is often ineffective, at least in part due to largely unknown mechanisms that underlie thalamic pain genesis. Here, we report that hemorrhage caused by microinjection of type IV collagenase or autologous whole blood into unilateral ventral posterior lateral nucleus and ventral posterior medial nucleus of the thalamus increased the expression of Fgr, a member of the Src family nonreceptor tyrosine kinases, at both mRNA and protein levels in thalamic microglia. Pharmacological inhibition or genetic knockdown of thalamic Fgr attenuated the hemorrhage-induced thalamic injury on the ipsilateral side and the development and maintenance of mechanical, heat, and cold pain hypersensitivities on the contralateral side. Mechanistically, the increased Fgr participated in hemorrhage-induced microglial activation and subsequent production of TNF-α likely through activation of both NF-κB and ERK1/2 pathways in thalamic microglia. Our findings suggest that Fgr is a key player in thalamic pain and a potential target for the therapeutic management of this disorder.


Hemorrhagic Stroke/genetics , Hyperalgesia/genetics , Neuralgia/genetics , Pain Measurement/methods , Proto-Oncogene Proteins/genetics , src-Family Kinases/genetics , Animals , Collagenases/toxicity , Disease Models, Animal , Hemorrhagic Stroke/chemically induced , Hemorrhagic Stroke/pathology , Humans , Hyperalgesia/chemically induced , Hyperalgesia/pathology , MAP Kinase Signaling System/genetics , Mice , Mice, Knockout , NF-kappa B/genetics , Neuralgia/chemically induced , Neuralgia/pathology , Thalamus/drug effects , Thalamus/metabolism , Thalamus/pathology
...