Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Stroke ; 55(3): 747-756, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38288607

ABSTRACT

BACKGROUND: Intravenous injection of alteplase, a recombinant tPA (tissue-type plasminogen activator) as a thrombolytic agent has revolutionized ischemic stroke management. However, tPA is a more complex enzyme than expected, being for instance able to promote thrombolysis, but at the same time, also able to influence neuronal survival and to affect the integrity of the blood-brain barrier. Accordingly, the respective impact of endogenous tPA expressed/present in the brain parenchyma versus in the circulation during stroke remains debated. METHODS: To address this issue, we used mice with constitutive deletion of tPA (tPANull [tPA-deficient mice]) or conditional deletion of endothelial tPA (VECad [vascular endothelial-Cadherin-Cre-recombinase]-Cre∆tPA). We also developed parabioses between tPANull and wild-type mice (tPAWT), anticipating that a tPAWT donor would restore levels of tPA to normal ones, in the circulation but not in the brain parenchyma of a tPANull recipient. Stroke outcomes were investigated by magnetic resonance imaging in a thrombo-embolic or a thrombotic stroke model, induced by local thrombin injection or FeCl3 application on the endothelium, respectively. RESULTS: First, our data show that endothelial tPA, released into the circulation after stroke onset, plays an overall beneficial role following thrombo-embolic stroke. Accordingly, after 24 hours, tPANull/tPANull parabionts displayed less spontaneous recanalization and reperfusion and larger infarcts compared with tPAWT/tPAWT littermates. However, when associated to tPAWT littermates, tPANull mice had similar perfusion deficits, but less severe brain infarcts. In the thrombotic stroke model, homo- and hetero-typic parabionts did not differ in the extent of brain damages and did not differentially recanalize and reperfuse. CONCLUSIONS: Together, our data reveal that during thromboembolic stroke, endogenous circulating tPA from endothelial cells sustains a spontaneous recanalization and reperfusion of the tissue, thus, limiting the extension of ischemic lesions. In this context, the impact of endogenous parenchymal tPA is limited.


Subject(s)
Stroke , Thrombotic Stroke , Animals , Mice , Disease Models, Animal , Endothelial Cells , Endothelium , Mice, Knockout , Stroke/diagnostic imaging , Stroke/pathology , Tissue Plasminogen Activator/genetics , Tissue Plasminogen Activator/metabolism
2.
Brain Behav Immun ; 119: 381-393, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38604270

ABSTRACT

INTRODUCTION: Multiple sclerosis (MS) is an autoimmune disease of the central nervous system. Recent evidence suggests that lymphocyte trafficking in the intestines could play a key role in its etiology. Nevertheless, it is not clear how intestinal tissue is involved in the disease onset nor its evolution. In the present study, we aimed to evaluate intestinal inflammation dynamic throughout the disease course and its potential impact on disease progression. METHODS: We used tissue immunophenotyping (immunohistofluorescence and flow cytometry) and a recently described molecular magnetic resonance imaging (MRI) method targeting mucosal addressin cell adhesion molecule-1 (MAdCAM-1) to assess intestinal inflammation in vivo in two distinct animal models of MS (Experimental Autoimmune Encephalomyelitis - EAE) at several time points of disease progression. RESULTS: We report a positive correlation between disease severity and MAdCAM-1 MRI signal in two EAE models. Moreover, high MAdCAM-1 MRI signal during the asymptomatic phase is associated with a delayed disease onset in progressive EAE and to a lower risk of conversion to a secondary-progressive form in relapsing-remitting EAE. During disease evolution, in line with a bi-directional immune communication between the gut and the central nervous system, we observed a decrease in T-CD4+ and B lymphocytes in the ileum concomitantly with their increase in the spinal cord. CONCLUSION: Altogether, these data unveil a crosstalk between intestinal and central inflammation in EAE and support the use of molecular MRI of intestinal MAdCAM-1 as a new biomarker for prognostic in MS patients.


Subject(s)
Biomarkers , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental , Magnetic Resonance Imaging , Mice, Inbred C57BL , Mucoproteins , Multiple Sclerosis , Animals , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/diagnostic imaging , Multiple Sclerosis/diagnostic imaging , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Magnetic Resonance Imaging/methods , Mice , Biomarkers/metabolism , Mucoproteins/metabolism , Female , Prognosis , Disease Progression , Cell Adhesion Molecules/metabolism , Intestines/diagnostic imaging , Intestines/pathology , Immunoglobulins/metabolism , Inflammation/metabolism , Inflammation/diagnostic imaging , Intestinal Mucosa/metabolism , Intestinal Mucosa/diagnostic imaging
3.
J Cardiovasc Pharmacol ; 83(6): 580-587, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38467037

ABSTRACT

ABSTRACT: Multimers of von Willebrand factor play a critical role in various processes inducing morbidity and mortality in cardiovascular-risk patients. With the ability to reduce von Willebrand factor multimers, N-acetylcysteine (NAC) could reduce mortality in patients undergoing coronary catheterization or cardiac surgery. However, its impact in perioperative period has never been studied so far in regard of its potential cardiovascular benefits. Then, 4 databases were searched for randomized controlled trials that compared in-hospital mortality between an experimental group, with NAC, and a control group without NAC, in patients undergoing coronary catheterization or cardiac surgery. The primary efficacy outcome was in-hospital mortality. Secondary outcomes were the occurrence of thrombotic events, major cardiovascular events, myocardial infarction, and contrast-induced nephropathy. The safety outcome was occurrence of hemorrhagic events. Nineteen studies totaling 3718 patients were included. Pooled analysis demonstrated a reduction of in-hospital mortality associated with NAC: odds ratio, 0.60; 95% confidence interval, 0.39-0.92; P = 0.02. The occurrence of secondary outcomes was not significantly reduced with NAC except for contrast-induced nephropathy. No difference was reported for hemorrhagic events. Subgroup analyses revealed a life-saving effect of NAC in a dose-dependent manner with reduction of in-hospital mortality for the NAC high-dose group, but not for the NAC standard-dose (<3500-mg) group. In conclusion, without being able to conclude on the nature of the mechanism involved, our review suggests a benefit of NAC in cardiovascular-risk patients in perioperative period in terms of mortality and supports prospective confirmatory studies.


Subject(s)
Acetylcysteine , Cardiac Catheterization , Cardiac Surgical Procedures , Hospital Mortality , Humans , Cardiac Catheterization/adverse effects , Cardiac Catheterization/mortality , Acetylcysteine/adverse effects , Acetylcysteine/therapeutic use , Acetylcysteine/administration & dosage , Cardiac Surgical Procedures/adverse effects , Cardiac Surgical Procedures/mortality , Treatment Outcome , Risk Factors , Risk Assessment , Female , Randomized Controlled Trials as Topic , Male , Aged , Middle Aged
4.
Proc Natl Acad Sci U S A ; 114(23): 6116-6121, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28533365

ABSTRACT

New strategies for detecting disease activity in multiple sclerosis are being investigated to ameliorate diagnosis and follow-up of patients. Today, although magnetic resonance imaging (MRI) is widely used to diagnose and monitor multiple sclerosis, no imaging tools exist to predict the evolution of disease and the efficacy of therapeutic strategies. Here, we show that molecular MRI targeting the endothelial adhesion molecule P-selectin unmasks the pathological events that take place in the spinal cord of mice subjected to chronic or relapsing experimental autoimmune encephalomyelitis. This approach provides a quantitative spatiotemporal follow-up of disease course in relation to clinical manifestations. Moreover, it predicts relapse in asymptomatic mice and remission in symptomatic animals. Future molecular MRI targeting P-selectin may be used to improve diagnosis, follow-up of treatment, and management of relapse/remission cycles in multiple sclerosis patients by providing information currently inaccessible through conventional MRI techniques.


Subject(s)
Magnetic Resonance Imaging/methods , Multiple Sclerosis/diagnostic imaging , Multiple Sclerosis/pathology , Animals , Blood-Brain Barrier/diagnostic imaging , Brain/pathology , Contrast Media , Disease Models, Animal , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/pathology , Male , Mice , Mice, Inbred C57BL , Multiple Sclerosis/metabolism , P-Selectin/metabolism , Recurrence , Spinal Cord/pathology
5.
Circulation ; 136(7): 646-660, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28487393

ABSTRACT

BACKGROUND: Platelet cross-linking during arterial thrombosis involves von Willebrand Factor (VWF) multimers. Therefore, proteolysis of VWF appears promising to disaggregate platelet-rich thrombi and restore vessel patency in acute thrombotic disorders such as ischemic stroke, acute coronary syndrome, or acute limb ischemia. N-Acetylcysteine (NAC, a clinically approved mucolytic drug) can reduce intrachain disulfide bonds in large polymeric proteins. In the present study, we postulated that NAC might cleave the VWF multimers inside occlusive thrombi, thereby leading to their dissolution and arterial recanalization. METHODS: Experimental models of thrombotic stroke induced by either intra-arterial thrombin injection or ferric chloride application followed by measurement of cerebral blood flow using a combination of laser Doppler flowmetry and MRI were performed to uncover the effects of NAC on arterial thrombi. To investigate the effect of NAC on larger vessels, we also performed ferric chloride-induced carotid artery thrombosis. In vitro experiments were performed to study the molecular bases of NAC thrombolytic effect, including platelet aggregometry, platelet-rich thrombi lysis assays, thromboelastography (ROTEM), and high-shear VWF string formation using microfluidic devices. We also investigated the putative prohemorrhagic effect of NAC in a mouse model of intracranial hemorrhage induced by in situ collagenase type VII injection. RESULTS: We demonstrated that intravenous NAC administration promotes lysis of arterial thrombi that are resistant to conventional approaches such as recombinant tissue-type plasminogen activator, direct thrombin inhibitors, and antiplatelet treatments. Through in vitro and in vivo experiments, we provide evidence that the molecular target underlying the thrombolytic effects of NAC is principally the VWF that cross-link platelets in arterial thrombi. Coadministration of NAC and a nonpeptidic GpIIb/IIIa inhibitor further improved its thrombolytic efficacy, essentially by accelerating thrombus dissolution and preventing rethrombosis. Thus, in a new large-vessel thromboembolic stroke model in mice, this cotreatment significantly improved ischemic lesion size and neurological outcome. It is important to note that NAC did not worsen hemorrhagic stroke outcome, suggesting that it exerts thrombolytic effects without significantly impairing normal hemostasis. CONCLUSIONS: We provide evidence that NAC is an effective and safe alternative to currently available antithrombotic agents to restore vessel patency after arterial occlusion.


Subject(s)
Acetylcysteine/therapeutic use , Fibrinolytic Agents/therapeutic use , Infarction, Middle Cerebral Artery/drug therapy , Thromboembolism/drug therapy , Acetylcysteine/pharmacology , Animals , Blood Platelets/cytology , Blood Platelets/metabolism , Chlorides/toxicity , Disease Models, Animal , Ferric Compounds/toxicity , Fibrinolytic Agents/pharmacology , Infarction, Middle Cerebral Artery/etiology , Male , Mice , Platelet Aggregation/drug effects , Ristocetin/pharmacology , Thromboembolism/chemically induced , Thrombosis/prevention & control , Tissue Plasminogen Activator/therapeutic use , von Willebrand Factor/chemistry , von Willebrand Factor/metabolism
6.
Stroke ; 49(12): 3071-3074, 2018 12.
Article in English | MEDLINE | ID: mdl-30571423

ABSTRACT

Background and Purpose- In experimental models of ischemic stroke, abrupt reperfusion is associated with secondary brain damages, responsible for up to 70% of the final lesion size. Whether this remains true in humans is unknown. Methods- Using data from the ASTER randomized trial (Aspiration vs Stent Retriever for Successful Revascularization), we investigated the effect of complete reperfusion (defined as a modified Thrombolysis In Cerebral Infarction 3) after endovascular thrombectomy on early lesion growth as assessed by diffusion-weighted imaging at baseline and 1 day after reperfusion. Results- Among 381 patients included in the trial, 35 achieved complete reperfusion, benefited from both baseline and day 1 diffusion-weighted imaging, lacked significant hemorrhagic transformation, and were, therefore, included in the present study. We found that the median growth of the ischemic lesion between baseline and day 1 was only 0.9 mL after complete reperfusion, representing <4% of the mean lesion size. The actual lesion growth occurring after reperfusion is probably even smaller because this lesion growth occurred, at least in part, between baseline imaging and complete reperfusion, as demonstrated by a statistically significant positive correlation between imaging-to-reperfusion time and lesion growth ( R2=0.116; P=0.048). Conclusions- There is no significant lesion growth after complete reperfusion in most patients. This important discrepancy between clinical and preclinical pathophysiologies should be considered during preclinical evaluation of neuroprotective strategies.


Subject(s)
Brain Ischemia/surgery , Endovascular Procedures , Postoperative Complications/epidemiology , Reperfusion Injury/epidemiology , Stroke/surgery , Thrombectomy , Aged , Aged, 80 and over , Diffusion Magnetic Resonance Imaging , Disease Progression , Female , Humans , Male , Middle Aged , Postoperative Complications/diagnostic imaging , Reperfusion Injury/diagnostic imaging
7.
Blood ; 128(20): 2423-2434, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27531677

ABSTRACT

Hyperfibrinolysis is a systemic condition occurring in various clinical disorders such as trauma, liver cirrhosis, and leukemia. Apart from increased bleeding tendency, the pathophysiological consequences of hyperfibrinolysis remain largely unknown. Our aim was to develop an experimental model of hyperfibrinolysis and to study its effects on the homeostasis of the blood-brain barrier (BBB). We induced a sustained hyperfibrinolytic state in mice by hydrodynamic transfection of a plasmid encoding for tissue-type plasminogen activator (tPA). As revealed by near-infrared fluorescence imaging, hyperfibrinolytic mice presented a significant increase in BBB permeability. Using a set of deletion variants of tPA and pharmacological approaches, we demonstrated that this effect was independent of N-methyl-D-aspartate receptor, low-density lipoprotein-related protein, protease-activated receptor-1, or matrix metalloproteinases. In contrast, we provide evidence that hyperfibrinolysis-induced BBB leakage is dependent on plasmin-mediated generation of bradykinin and subsequent activation of bradykinin B2 receptors. Accordingly, this effect was prevented by icatibant, a clinically available B2 receptor antagonist. In agreement with these preclinical data, bradykinin generation was also observed in humans in a context of acute pharmacological hyperfibrinolysis. Altogether, these results suggest that B2 receptor blockade may be a promising strategy to prevent the deleterious effects of hyperfibrinolysis on the homeostasis of the BBB.


Subject(s)
Blood-Brain Barrier/metabolism , Bradykinin/physiology , Capillary Permeability/physiology , Fibrinolysin/physiology , Fibrinolysis/physiology , Animals , Blood-Brain Barrier/drug effects , Bradykinin/metabolism , Bradykinin B2 Receptor Antagonists/pharmacology , Brain/drug effects , Brain/metabolism , Capillary Permeability/drug effects , Capillary Permeability/genetics , Fibrinolysin/metabolism , Fibrinolysis/drug effects , Fibrinolysis/genetics , Hydrodynamics , Mice , Mice, Transgenic , Receptor, Bradykinin B2/genetics , Receptor, Bradykinin B2/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Tissue Plasminogen Activator/genetics , Tissue Plasminogen Activator/metabolism
8.
Brain ; 140(1): 146-157, 2017 01.
Article in English | MEDLINE | ID: mdl-28031221

ABSTRACT

SEE SUN ET AL DOI101093/AWW306 FOR A SCIENTIFIC COMMENTARY ON THIS ARTICLE: About 20% of patients with ischaemic stroke have a preceding transient ischaemic attack, which is clinically defined as focal neurological symptoms of ischaemic origin resolving spontaneously. Failure to diagnose transient ischaemic attack is a wasted opportunity to prevent recurrent disabling stroke. Unfortunately, diagnosis can be difficult, due to numerous mimics, and to the absence of a specific test. New diagnostic tools are thus needed, in particular for radiologically silent cases, which correspond to the recommended tissue-based definition of transient ischaemic attack. As endothelial activation is a hallmark of cerebrovascular events, we postulated that this may also be true for transient ischaemic attack, and that it would be clinically relevant to develop non-invasive in vivo imaging to detect this endothelial activation. Using transcriptional and immunohistological analyses for adhesion molecules in a mouse model, we identified brain endothelial P-selectin as a potential biomarker for transient ischaemic attack. We thus developed ultra-sensitive molecular magnetic resonance imaging using antibody-based microparticles of iron oxide targeting P-selectin. This highly sensitive imaging strategy unmasked activated endothelial cells after experimental transient ischaemic attack and allowed discriminating transient ischaemic attack from epilepsy and migraine, two important transient ischaemic attack mimics. We provide preclinical evidence that combining conventional magnetic resonance imaging with molecular magnetic resonance imaging targeting P-selectin might aid in the diagnosis of transient ischaemic attack.


Subject(s)
Ischemic Attack, Transient/metabolism , Magnetic Resonance Imaging/methods , Molecular Imaging/methods , P-Selectin/metabolism , Stroke/metabolism , Animals , Biomarkers/metabolism , Disease Models, Animal , Endothelial Cells , Ischemic Attack, Transient/diagnostic imaging , Male , Mice , Stroke/diagnostic imaging
9.
Cereb Cortex ; 27(10): 4783-4796, 2017 10 01.
Article in English | MEDLINE | ID: mdl-27613436

ABSTRACT

In humans, spatial cognition and navigation impairments are a frequent situation during physiological and pathological aging, leading to a dramatic deterioration in the quality of life. Despite the discovery of neurons with location-specific activity in rodents, that is, place cells in the hippocampus and later on grid cells in the entorhinal cortex (EC), the molecular mechanisms underlying spatial cognition are still poorly known. Our present data bring together in an unusual combination 2 molecules of primary biological importance: a major neuronal excitatory receptor, N-methyl-D-aspartate receptor (NMDAR), and an extracellular protease, tissue plasminogen activator (tPA), in the control of spatial navigation. By using tPA-deficient mice and a structure-selective pharmacological approach, we demonstrate that the tPA-dependent NMDAR signaling potentiation in the EC plays a key and selective role in the encoding and the subsequent use of distant landmarks during spatial learning. We also demonstrate that this novel function of tPA in the EC is reduced during aging. Overall, these results argue for the concept that encoding of proximal versus distal landmarks is mediated not only by different anatomical pathways but also by different molecular mechanisms, with the tPA-dependent potentiation of NMDAR signaling in the EC that plays an important role.


Subject(s)
Entorhinal Cortex/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Aging , Animals , Calcium/metabolism , Female , Hippocampus/metabolism , Male , Mice, Knockout , Neurons/metabolism , Signal Transduction/physiology , Tissue Plasminogen Activator/deficiency , Tissue Plasminogen Activator/metabolism
10.
Stroke ; 48(9): 2574-2582, 2017 09.
Article in English | MEDLINE | ID: mdl-28754830

ABSTRACT

BACKGROUND AND PURPOSE: Although the mechanisms that contribute to intracranial aneurysm (IA) formation and rupture are not totally elucidated, inflammation and matrix remodeling are incriminated. Because tPA (tissue-type plasminogen activator) controls both inflammatory and matrix remodeling processes, we hypothesized that tPA could be involved in the pathophysiology of IA. METHODS: Immunofluorescence analyses of tPA and its main substrate within the aneurysmal wall of murine and human samples were performed. We then compared the formation and rupture of IAs in wild-type, tPA-deficient and type 1 plasminogen activator inhibitor-deficient mice subjected to a model of elastase-induced IA. The specific contribution of vascular versus global tPA was investigated by performing hepatic hydrodynamic transfection of a cDNA encoding for tPA in tPA-deficient mice. The formation and rupture of IAs were monitored by magnetic resonance imaging tracking for 28 days. RESULTS: Immunofluorescence revealed increased expression of tPA within the aneurysmal wall. The number of aneurysms and their symptomatic ruptures were significantly lower in tPA-deficient than in wild-type mice. Conversely, they were higher in plasminogen activator inhibitor-deficient mice. The wild-type phenotype could be restored in tPA-deficient mice by selectively increasing circulating levels of tPA via hepatic hydrodynamic transfection of a cDNA encoding for tPA. CONCLUSIONS: Altogether, this preclinical study demonstrates that the tPA present in the blood stream is a key player of the formation of IAs. Thus, tPA should be considered as a possible new target for the prevention of IAs formation and rupture.


Subject(s)
Aneurysm, Ruptured/metabolism , Intracranial Aneurysm/metabolism , Tissue Plasminogen Activator/metabolism , Adult , Aneurysm, Ruptured/diagnostic imaging , Animals , Female , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Intracranial Aneurysm/diagnostic imaging , Magnetic Resonance Imaging , Mice , Mice, Knockout , Plasminogen Activator Inhibitor 1/genetics , Rupture, Spontaneous , Tissue Plasminogen Activator/genetics
11.
Blood ; 125(8): 1325-32, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25540192

ABSTRACT

Circulating thrombin-activatable fibrinolysis inhibitor (TAFI) and plasminogen activator inhibitor-1 (PAI-1) are causal factors for thrombolytic failure. Therefore, we evaluated an antibody-engineered bispecific inhibitor against TAFI and PAI-1 (heterodimer diabody, Db-TCK26D6x33H1F7) in several mouse models of thrombosis and stroke. Prophylactic administration of the diabody (0.8 mg/kg) in a thromboplastin-induced model of thromboembolism led to decreased lung fibrin deposition. In a model of cerebral ischemia and reperfusion, diabody administration (0.8 mg/kg, 1 hour postocclusion) led to a mitigated cerebral injury with a 2.3-fold reduced lesion and improved functional outcomes. In a mouse model of thrombin-induced middle cerebral artery occlusion, the efficacy of the diabody was compared to the standard thrombolytic treatment with recombinant tissue-type plasminogen activator (tPA). Early administration of diabody (0.8 mg/kg) caused a twofold decrease in brain lesion size, whereas that of tPA (10 mg/kg) had a much smaller effect. Delayed administration of diabody or tPA had no effect on lesion size, whereas the combined administration of diabody with tPA caused a 1.7-fold decrease in lesion size. In contrast to tPA, the diabody did not increase accumulative bleeding. In conclusion, administration of a bispecific inhibitor against TAFI and PAI-1 results in a prominent profibrinolytic effect in mice without increased bleeding.


Subject(s)
Antibodies, Bispecific/therapeutic use , Fibrinolytic Agents/therapeutic use , Histone Acetyltransferases/immunology , Serpin E2/immunology , Stroke/therapy , TATA-Binding Protein Associated Factors/immunology , Therapies, Investigational/methods , Transcription Factor TFIID/immunology , Venous Thromboembolism/therapy , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/metabolism , Disease Models, Animal , Female , Immunotherapy , Male , Mice , Mice, Inbred C57BL , Protein Multimerization , Stroke/pathology , Venous Thromboembolism/pathology
12.
Blood ; 123(21): 3354-63, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24553181

ABSTRACT

Interactions between platelet glycoprotein (Gp) IIb/IIIa and plasma proteins mediate platelet cross-linking in arterial thrombi. However, GpIIb/IIIa inhibitors fail to disperse platelet aggregates after myocardial infarction or ischemic stroke. These results suggest that stability of occlusive thrombi involves additional and as-yet-unidentified mechanisms. In the present study, we investigated the mechanisms driving platelet cross-linking during occlusive thrombus formation. Using computational fluid dynamic simulations and in vivo thrombosis models, we demonstrated that the inner structure of occlusive thrombi is heterogeneous and primarily determined by the rheological conditions that prevailed during thrombus growth. Unlike the first steps of thrombus formation, which are GpIIb/IIIa-dependent, our findings reveal that closure of the arterial lumen is mediated by GpIbα-von Willebrand Factor (VWF) interactions. Accordingly, disruption of platelet cross-linking using GpIbα-VWF inhibitors restored vessel patency and improved outcome in a mouse model of ischemic stroke, although the thrombi were resistant to fibrinolysis or traditional antithrombotic agents. Overall, our study demonstrates that disruption of GpIbα-VWF interactions restores vessel patency after occlusive thrombosis by specifically disaggregating the external layer of occlusive thrombi, which is constituted of platelet aggregates formed under very high shear rates.


Subject(s)
Blood Platelets/pathology , Platelet Glycoprotein GPIb-IX Complex/metabolism , Thrombosis/metabolism , Thrombosis/pathology , von Willebrand Factor/metabolism , Animals , Benzofurans , Blood Platelets/metabolism , Blood Vessels/metabolism , Blood Vessels/pathology , Hemorheology , Male , Mice , Platelet Aggregation , Protein Interaction Maps , Quinolines
13.
Stroke ; 46(6): 1641-50, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25922513

ABSTRACT

BACKGROUND AND PURPOSE: Tissue-type plasminogen activator (tPA) is the only acute treatment for ischemic stroke. Unfortunately, the benefit of tPA-driven thrombolysis is not systematic, and understanding the reasons for this is mandatory. The balance between beneficial and detrimental effects of tPA might explain the limited overall efficiency of thrombolysis. Here, we investigated whether this balance could be influenced by excessive alcohol intake. METHODS: We used a murine model of thromboembolic stroke, coupled to an array of biochemical assays, near-infrared or magnetic resonance imaging scans, 2-photon microscopy, hydrodynamic transfections, and immunohistological techniques. RESULTS: We found that 6 weeks of alcohol consumption (10% in drinking water) worsens ischemic lesions and cancels the beneficial effects of tPA-induced thrombolysis. We accumulate in vivo and in vitro evidence showing that this aggravation is correlated with a decrease in lipoprotein receptor-related protein 1-mediated hepatic clearance of tPA in alcohol-exposed mice. CONCLUSIONS: An efficient liver-driven clearance of tPA might influence the safety of thrombolysis after stroke.


Subject(s)
Alcohol Drinking/adverse effects , Brain Ischemia/drug therapy , Liver/metabolism , Stroke/drug therapy , Thrombolytic Therapy , Tissue Plasminogen Activator/pharmacokinetics , Alcohol Drinking/pathology , Animals , Brain Ischemia/metabolism , Brain Ischemia/pathology , Disease Models, Animal , Liver/pathology , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Mice , Stroke/metabolism , Stroke/pathology , Tissue Plasminogen Activator/pharmacology
15.
Stroke ; 45(10): 3092-6, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25190438

ABSTRACT

BACKGROUND AND PURPOSE: The aim of the present study was to investigate the impact of different stroke subtypes on the glymphatic system using MRI. METHODS: We first improved and characterized an in vivo protocol to measure the perfusion of the glymphatic system using MRI after minimally invasive injection of a gadolinium chelate within the cisterna magna. Then, the integrity of the glymphatic system was evaluated in 4 stroke models in mice including subarachnoid hemorrhage (SAH), intracerebral hemorrhage, carotid ligature, and embolic ischemic stroke. RESULTS: We were able to reliably evaluate the glymphatic system function using MRI. Moreover, we provided evidence that the glymphatic system was severely impaired after SAH and in the acute phase of ischemic stroke, but was not altered after carotid ligature or in case of intracerebral hemorrhage. Notably, this alteration in glymphatic perfusion reduced brain clearance rate of low-molecular-weight compounds. Interestingly, glymphatic perfusion after SAH can be improved by intracerebroventricular injection of tissue-type plasminogen activator. Moreover, spontaneous arterial recanalization was associated with restoration of the glymphatic function after embolic ischemic stroke. CONCLUSIONS: SAH and acute ischemic stroke significantly impair the glymphatic system perfusion. In these contexts, injection of tissue-type plasminogen activator either intracerebroventricularly to clear perivascular spaces (for SAH) or intravenously to restore arterial patency (for ischemic stroke) may improve glymphatic function.


Subject(s)
Magnetic Resonance Imaging/methods , Stroke/cerebrospinal fluid , Subarachnoid Hemorrhage/cerebrospinal fluid , Animals , Cerebrospinal Fluid/physiology , Contrast Media , Disease Models, Animal , Image Interpretation, Computer-Assisted , Mice , Stroke/pathology , Subarachnoid Hemorrhage/pathology
16.
J Neuroinflammation ; 11: 154, 2014 Sep 16.
Article in English | MEDLINE | ID: mdl-25220760

ABSTRACT

BACKGROUND: Plasminogen activation is a ubiquitous source of fibrinolytic and proteolytic activity. Besides its role in prevention of thrombosis, plasminogen is involved in inflammatory reactions in the central nervous system. Plasminogen has been detected in the cerebrospinal fluid (CSF) of patients with inflammatory diseases; however, its origin remains controversial, as the blood-CSF barrier may restrict its diffusion from blood. METHODS: We investigated the origin of plasminogen in CSF using Alexa Fluor 488-labelled rat plasminogen injected into rats with systemic inflammation and blood-CSF barrier dysfunction provoked by lipopolysaccharide (LPS). Near-infrared fluorescence imaging and immunohistochemistry fluorescence microscopy were used to identify plasminogen in brain structures, its concentration and functionality were determined by Western blotting and a chromogenic substrate assay, respectively. In parallel, plasminogen was investigated in CSF from patients with Guillain-Barré syndrome (n = 15), multiple sclerosis (n = 19) and noninflammatory neurological diseases (n = 8). RESULTS: Endogenous rat plasminogen was detected in higher amounts in the CSF and urine of LPS-treated animals as compared to controls. In LPS-primed rats, circulating Alexa Fluor 488-labelled rat plasminogen was abundantly localized in the choroid plexus, CSF and urine. Plasminogen in human CSF was higher in Guillain-Barré syndrome (median = 1.28 ng/µl (interquartile range (IQR) = 0.66 to 1.59)) as compared to multiple sclerosis (median = 0.3 ng/µl (IQR = 0.16 to 0.61)) and to noninflammatory neurological diseases (median = 0.27 ng/µl (IQR = 0.18 to 0.35)). CONCLUSIONS: Our findings demonstrate that plasminogen is transported from circulating blood into the CSF of rats via the choroid plexus during inflammation. Our data suggest that a similar mechanism may explain the high CSF concentrations of plasminogen detected in patients with inflammation-derived CSF barrier impairment.


Subject(s)
Blood-Brain Barrier/physiology , Inflammation/blood , Inflammation/cerebrospinal fluid , Plasminogen/cerebrospinal fluid , Animals , Blotting, Western , Humans , Male , Microscopy, Fluorescence , Rats , Rats, Wistar
17.
Nat Commun ; 15(1): 5070, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38871729

ABSTRACT

In acute ischemic stroke, even when successful recanalization is obtained, downstream microcirculation may still be obstructed by microvascular thrombosis, which is associated with compromised brain reperfusion and cognitive decline. Identifying these microthrombi through non-invasive methods remains challenging. We developed the PHySIOMIC (Polydopamine Hybridized Self-assembled Iron Oxide Mussel Inspired Clusters), a MRI-based contrast agent that unmasks these microthrombi. In a mouse model of thromboembolic ischemic stroke, our findings demonstrate that the PHySIOMIC generate a distinct hypointense signal on T2*-weighted MRI in the presence of microthrombi, that correlates with the lesion areas observed 24 hours post-stroke. Our microfluidic studies reveal the role of fibrinogen in the protein corona for the thrombosis targeting properties. Finally, we observe the biodegradation and biocompatibility of these particles. This work demonstrates that the PHySIOMIC particles offer an innovative and valuable tool for non-invasive in vivo diagnosis and monitoring of microthrombi, using MRI during ischemic stroke.


Subject(s)
Contrast Media , Disease Models, Animal , Ferric Compounds , Indoles , Magnetic Resonance Imaging , Polymers , Thrombosis , Animals , Polymers/chemistry , Magnetic Resonance Imaging/methods , Indoles/chemistry , Mice , Contrast Media/chemistry , Ferric Compounds/chemistry , Thrombosis/diagnostic imaging , Male , Stroke/diagnostic imaging , Humans , Fibrinogen/metabolism , Ischemic Stroke/diagnostic imaging , Mice, Inbred C57BL , Protein Corona/chemistry , Protein Corona/metabolism , Brain/diagnostic imaging , Brain/metabolism , Brain/pathology
19.
Arterioscler Thromb Vasc Biol ; 32(6): 1477-87, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22492089

ABSTRACT

OBJECTIVE: Thrombin induces CD40 ligand (CD40L) and matrix metalloproteinases (MMPs) under inflammatory/prothrombotic conditions. Thrombin and CD40L could modulate endothelial MMP-10 expression in vitro and in vivo. METHODS AND RESULTS: Human endothelial cells were stimulated with thrombin (0.1-10 U/mL), CD40L (0.25-1 µg/mL), or their combination (thrombin/CD40L) to assess MMP-10 expression and microparticle generation. Thrombin/CD40L elicited higher MMP-10 mRNA (5-fold; P<0.001) and protein levels (4.5-fold; P<0.001) than either stimulus alone. This effect was mimicked by a protease-activated receptor-1 agonist and antagonized by hirudin, a-protease-activated receptor-1, α-CD40L, and α-CD40 antibodies. The synergistic effect was dependent on p38 mitogen-activated protein kinase and c-Jun N-terminal kinase-1 pathways. Thrombin also upregulated the expression of CD40 in endothelial cell surface increasing its availability, thereby favoring its synergistic effects with CD40L. In mice, thrombin/CD40L further increased the aortic MMP-10 expression. Septic patients with systemic inflammation and enhanced thrombin generation (n=60) exhibited increased MMP-10 and soluble CD40L levels associated with adverse clinical outcome. Endothelial and systemic activation by thrombin/CD40L and lipopolysaccharide also increased microparticles harboring MMP-10 and CD40L. CONCLUSIONS: Thrombin/CD40L elicited a strong synergistic effect on endothelial MMP-10 expression and microparticles containing MMP-10 in vitro and in vivo, which may represent a new link between inflammation/thrombosis with prognostic implications.


Subject(s)
CD40 Ligand/metabolism , Cell-Derived Microparticles/enzymology , Endothelial Cells/enzymology , Matrix Metalloproteinase 10/metabolism , Sepsis/enzymology , Thrombin/metabolism , Adult , Aged , Aged, 80 and over , Animals , Antibodies/pharmacology , Blood Coagulation , CD40 Antigens/antagonists & inhibitors , CD40 Antigens/metabolism , CD40 Ligand/antagonists & inhibitors , CD40 Ligand/blood , Case-Control Studies , Cell-Derived Microparticles/pathology , Cells, Cultured , Disease Models, Animal , Disseminated Intravascular Coagulation/enzymology , Disseminated Intravascular Coagulation/pathology , Endothelial Cells/drug effects , Endothelial Cells/pathology , Endotoxemia/enzymology , Endotoxemia/genetics , Endotoxemia/pathology , Female , Gene Expression Regulation, Enzymologic , Hirudins/pharmacology , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Lipopolysaccharides/pharmacology , Male , Matrix Metalloproteinase 10/blood , Matrix Metalloproteinase 10/deficiency , Matrix Metalloproteinase 10/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Mitogen-Activated Protein Kinase 8/antagonists & inhibitors , Mitogen-Activated Protein Kinase 8/metabolism , Multivariate Analysis , Peptides/pharmacology , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/metabolism , Receptor, PAR-1/agonists , Receptor, PAR-1/antagonists & inhibitors , Receptor, PAR-1/metabolism , Risk Assessment , Risk Factors , Sepsis/mortality , Sepsis/pathology , Signal Transduction , Spain
20.
Fluids Barriers CNS ; 20(1): 11, 2023 Feb 03.
Article in English | MEDLINE | ID: mdl-36737775

ABSTRACT

BACKGROUND: Regulation of cerebral blood flow (CBF) directly influence brain functions and dysfunctions and involves complex mechanisms, including neurovascular coupling (NVC). It was suggested that the serine protease tissue-type plasminogen activator (tPA) could control CNV induced by whisker stimulation in rodents, through its action on N-methyl-D-Aspartate receptors (NMDARs). However, the origin of tPA and the location and mechanism of its action on NMDARs in relation to CNV remained debated. METHODS: Here, we answered these issues using tPANull mice, conditional deletions of either endothelial tPA (VECad-CreΔtPA) or endothelial GluN1 subunit of NMDARs (VECad-CreΔGluN1), parabioses between wild-type and tPANull mice, hydrodynamic transfection-induced deletion of liver tPA, hepatectomy and pharmacological approaches. RESULTS: We thus demonstrate that physiological concentrations of vascular tPA, achieved by the bradykinin type 2 receptors-dependent production and release of tPA from liver endothelial cells, promote NVC, through a mechanism dependent on brain endothelial NMDARs. CONCLUSIONS: These data highlight a new mechanism of regulation of NVC involving both endothelial tPA and NMDARs.


Subject(s)
Neurovascular Coupling , Tissue Plasminogen Activator , Mice , Animals , N-Methylaspartate/pharmacology , Endothelial Cells/metabolism , Brain/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Mice, Knockout , Liver/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL