Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 39
1.
JMIR Form Res ; 8: e55064, 2024 May 08.
Article En | MEDLINE | ID: mdl-38717803

BACKGROUND: Health organizations face the critical task of executing and overseeing comprehensive health care. To address the challenges associated with this task, evidence-based dashboards have emerged as valuable tools. Since 2016, the regional health organizations of Quebec, Canada, have been responsible for ensuring implementation of the Quebec Alzheimer Plan (QAP), a provincial plan that aims to reinforce the capacity of primary care services to detect, diagnose, and treat persons with dementia. Despite the provincial scope of the QAP, the diverse material and human resources across regions introduce variability in the interest, utility, and specific needs associated with these dashboards. OBJECTIVE: The aim of this study was to assess the interest and utility of dashboards to support the QAP implementation, as well as to determine the needs for improving these aspects according to the perspectives of various types of professionals involved across regions. METHODS: An evaluative study using qualitative methods was conducted within a collaborative research approach involving different stakeholders, including the ministerial advisor and the four project managers responsible for supporting the implementation of the QAP, as well as researchers/scientific advisors. To support these organizations, we developed tailored, 2-page paper dashboards, detailing quantitative data on the prevalence of dementia, the use of health services by persons with dementia, and achievements and challenges of the QAP implementation in each organization's jurisdiction. We then conducted 23 focus groups with the managers and leading clinicians involved in the implementation of the QAP of each regional health organization. Real-time notes were taken using a structured observation grid. Content analysis was conducted according to different regions (organizations with university mandates or nearby organizations, labeled "university/peripheral"; organizations for which only part of the territory is in rural areas, labeled "mixed"; and organizations in remote or isolated areas, labeled "remote/isolated") and according to different types of participants (managers, leading clinicians, and other participants). RESULTS: Participants from organizations in all regions expressed interest in these dashboards and found them useful in several ways. However, they highlighted the need for indicators on orphan patients and other health care providers. Differences between regions were observed, particularly in the interest in continuity of care in university/peripheral regions and the need for diagnostic tools adapted to the culture in remote/isolated regions. CONCLUSIONS: These dashboards support the implementation of an Alzheimer Plan and contribute to the emergence of a learning health care system culture. This project allows each region to increase its monitoring capacity for the implementation of the QAP and facilitates reflection among individuals locally carrying out the implementation. The perspectives expressed will guide the preparation of the next iteration of the dashboards.

2.
Fluids Barriers CNS ; 20(1): 11, 2023 Feb 03.
Article En | MEDLINE | ID: mdl-36737775

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.


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
3.
BMC Biol ; 20(1): 218, 2022 10 05.
Article En | MEDLINE | ID: mdl-36199089

BACKGROUND: Perineuronal nets (PNNs) are specialized extracellular matrix structures mainly found around fast-spiking parvalbumin (FS-PV) interneurons. In the adult, their degradation alters FS-PV-driven functions, such as brain plasticity and memory, and altered PNN structures have been found in neurodevelopmental and central nervous system disorders such as Alzheimer's disease, leading to interest in identifying targets able to modify or participate in PNN metabolism. The serine protease tissue-type plasminogen activator (tPA) plays multifaceted roles in brain pathophysiology. However, its cellular expression profile in the brain remains unclear and a possible role in matrix plasticity through PNN remodeling has never been investigated. RESULT: By combining a GFP reporter approach, immunohistology, electrophysiology, and single-cell RT-PCR, we discovered that cortical FS-PV interneurons are a source of tPA in vivo. We found that mice specifically lacking tPA in FS-PV interneurons display denser PNNs in the somatosensory cortex, suggesting a role for tPA from FS-PV interneurons in PNN remodeling. In vitro analyses in primary cultures of mouse interneurons also showed that tPA converts plasminogen into active plasmin, which in turn, directly degrades aggrecan, a major structural chondroitin sulfate proteoglycan (CSPG) in PNNs. CONCLUSIONS: We demonstrate that tPA released from FS-PV interneurons in the central nervous system reduces PNN density through CSPG degradation. The discovery of this tPA-dependent PNN remodeling opens interesting insights into the control of brain plasticity.


Parvalbumins , Tissue Plasminogen Activator , Aggrecans/metabolism , Animals , Chondroitin Sulfate Proteoglycans/metabolism , Extracellular Matrix/metabolism , Fibrinolysin/metabolism , Interneurons/physiology , Mice , Parvalbumins/metabolism , Plasminogen/metabolism , Tissue Plasminogen Activator/metabolism
4.
Mol Psychiatry ; 27(4): 2197-2205, 2022 04.
Article En | MEDLINE | ID: mdl-35145231

Tissue plasminogen activator (tPA) is a serine protease expressed in several brain regions and reported to be involved in the control of emotional and cognitive functions. Nevertheless, little is known about the structure-function relationships of these tPA-dependent behaviors. Here, by using a new model of constitutive tPA-deficient mice (tPAnull), we first show that tPA controls locomotor activity, spatial cognition and anxiety. To investigate the brain structures involved in these tPA-dependent behavioral phenotypes, we next generated tPAflox mice allowing conditional tPA deletion (cKO) following stereotaxic injections of adeno-associated virus driving Cre-recombinase expression (AAV-Cre-GFP). We demonstrate that tPA removal in the dentate gyrus of the hippocampus induces hyperactivity and partial spatial memory deficits. Moreover, the deletion of tPA in the central nucleus of the amygdala, but not in the basolateral nucleus, induces hyperactivity and reduced anxiety-like level. Importantly, we prove that these behaviors depend on the tPA present in the adult brain and not on neurodevelopmental disorders. Also, interestingly, our data show that tPA from Protein kinase-C delta-positive (PKCδ) GABAergic interneurons of the lateral/ capsular part of adult mouse central amygdala controls emotional functions through neuronal activation of the medial central amygdala. Together, our study brings new data about the critical central role of tPA in behavioral modulations in adult mice.


Central Amygdaloid Nucleus , Protein Kinase C-delta/metabolism , Animals , Anxiety , Anxiety Disorders , Central Amygdaloid Nucleus/metabolism , GABAergic Neurons/metabolism , Mice , Mice, Inbred C57BL , Tissue Plasminogen Activator/genetics , Tissue Plasminogen Activator/metabolism
5.
Front Cardiovasc Med ; 8: 752769, 2021.
Article En | MEDLINE | ID: mdl-34869659

Delayed cerebral ischemia (DCI) is one of the main prognosis factors for disability after aneurysmal subarachnoid hemorrhage (SAH). The lack of a consensual definition for DCI had limited investigation and care in human until 2010, when a multidisciplinary research expert group proposed to define DCI as the occurrence of cerebral infarction (identified on imaging or histology) associated with clinical deterioration. We performed a systematic review to assess whether preclinical models of SAH meet this definition, focusing on the combination of noninvasive imaging and neurological deficits. To this aim, we searched in PUBMED database and included all rodent SAH models that considered cerebral ischemia and/or neurological outcome and/or vasospasm. Seventy-eight publications were included. Eight different methods were performed to induce SAH, with blood injection in the cisterna magna being the most widely used (n = 39, 50%). Vasospasm was the most investigated SAH-related complication (n = 52, 67%) compared to cerebral ischemia (n = 30, 38%), which was never investigated with imaging. Neurological deficits were also explored (n = 19, 24%). This systematic review shows that no preclinical SAH model meets the 2010 clinical definition of DCI, highlighting the inconsistencies between preclinical and clinical standards. In order to enhance research and favor translation to humans, pertinent SAH animal models reproducing DCI are urgently needed.

6.
BMC Geriatr ; 21(1): 73, 2021 01 22.
Article En | MEDLINE | ID: mdl-33482740

BACKGROUND: The "Program of Research on the Integration of Services for the Maintenance of Autonomy" (PRISMA-7) and "Emergency room evaluation and recommendations" (ER2) are both clinical tools used in Québec Emergency Departments (EDs) for screening of older ED users at higher risk of poor outcomes, such as prolonged length of stay (LOS) in EDs and in hospital. The study aimed to: 1) examine whether the PRISMA-7 and ER2 risk levels were associated with length of stays in ED and hospital, as well as hospital admission; and 2) compare the criteria performance (i.e., sensitivity, specificity, positive predictive value, negative predictive value, likelihood ratios and area under receiver operating characteristic curve) of the PRISMA-7 and ER2 high-risk levels for these three ED adverse events in Québec older patients visiting ED on a stretcher. METHODS: A total of 1905 older patients who visited the ED of the Jewish General Hospital (Montreal, Québec, Canada) on stretchers were recruited in this prospective observational cohort. Upon their ED arrival, PRISMA-7 and ER2 were performed. The outcomes were LOS in ED and in hospital, and hospital admission. RESULTS: The PRISMA-7 and ER2 risk levels were associated with length of stay in ED and hospital as well as with hospital admission. Prolonged stays and higher hospitalization rates were associated with high-risk levels, whereas those in low-risk level groups had significantly shorter LOS and a lower rate of hospital admission (P < 0.006). While performance measures were poor for both assessment tools, ER2 had a greater prognostic testing accuracy compared with PRISMA-7. CONCLUSION: PRISMA-7 and ER2 were both associated with incidental short-term ED adverse events but their overall prognostic testing accuracy was low, suggesting that they cannot be used as prognostic tools for this purpose.


Emergency Service, Hospital , Geriatric Assessment , Aged , Canada , Humans , Length of Stay , Prognosis , Prospective Studies , Quebec/epidemiology
7.
Sci Rep ; 10(1): 19577, 2020 11 11.
Article En | MEDLINE | ID: mdl-33177650

Histone methyltransferase EZH2 is upregulated during osteoarthritis (OA), which is the most widespread rheumatic disease worldwide, and a leading cause of disability. This study aimed to assess the impact of EZH2 inhibition on cartilage degradation, inflammation and functional disability. In vitro, gain and loss of EZH2 function were performed in human articular OA chondrocytes stimulated with IL-1ß. In vivo, the effects of EZH2 inhibition were investigated on medial meniscectomy (MMX) OA mouse model. The tissue alterations were assayed by histology and the functional disabilities of the mice by actimetry and running wheel. In vitro, EZH2 overexpression exacerbated the action of IL-1ß in chondrocytes increasing the expression of genes involved in inflammation, pain (NO, PGE2, IL6, NGF) and catabolism (MMPs), whereas EZH2 inhibition by a pharmacological inhibitor, EPZ-6438, reduced IL-1ß effects. Ex vivo, EZH2 inhibition decreased IL-1ß-induced degradation of cartilage. In vivo, intra-articular injections of the EZH2 inhibitor reduced cartilage degradation and improved motor functions of OA mice. This study demonstrates that the pharmacological inhibition of the histone methyl-transferase EZH2 slows the progression of osteoarthritis and improves motor functions in an experimental OA model, suggesting that EZH2 could be an effective target for the treatment of OA by reducing catabolism, inflammation and pain.


Cartilage, Articular/pathology , Enhancer of Zeste Homolog 2 Protein/genetics , Osteoarthritis/pathology , Aged , Aged, 80 and over , Animals , Benzamides/pharmacology , Biphenyl Compounds/pharmacology , Cartilage, Articular/drug effects , Chondrocytes/drug effects , Chondrocytes/physiology , Disease Models, Animal , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Expression Regulation , Humans , Interleukin-1beta/pharmacology , Male , Mice, Inbred C57BL , Middle Aged , Morpholines/pharmacology , Nerve Growth Factor/metabolism , Organ Culture Techniques , Pyridones/pharmacology
8.
Brain ; 143(10): 2957-2972, 2020 10 01.
Article En | MEDLINE | ID: mdl-32893288

Anti-N-methyl-d-aspartate receptor (NMDAR) encephalitis is a neuropsychiatric disease characterized by an antibody-mediated autoimmune response against NMDAR. Recent studies have shown that anti-NMDAR antibodies are involved in the pathophysiology of the disease. However, the upstream immune and inflammatory processes responsible for this pathogenic response are still poorly understood. Here, we immunized mice against the region of NMDA receptor containing the N368/G369 amino acids, previously implicated in a pathogenic response. This paradigm induced encephalopathy characterized by blood-brain barrier opening, periventricular T2-MRI hyperintensities and IgG deposits into the brain parenchyma. Two weeks after immunization, mice developed clinical symptoms reminiscent of encephalitis: anxiety- and depressive-like behaviours, spatial memory impairment (without motor disorders) and increased sensitivity to seizures. This response occurred independently of overt T-cell recruitment. However, it was associated with B220+ (B cell) infiltration towards the ventricles, where they differentiated into CD138+ cells (plasmocytes). Interestingly, these B cells originated from peripheral lymphoid organs (spleen and cervical lymphoid nodes). Finally, blocking the B-cell response using a depleting cocktail of antibodies reduced the severity of symptoms in encephalitis mice. This study demonstrates that the B-cell response can lead to an autoimmune reaction against NMDAR that drives encephalitis-like behavioural impairments. It also provides a relevant platform for dissecting encephalitogenic mechanisms in an animal model, and enables the testing of therapeutic strategies targeting the immune system in anti-NMDAR encephalitis.


Autoantibodies/blood , B-Lymphocytes/metabolism , Encephalitis/blood , Hashimoto Disease/blood , Nerve Tissue Proteins/toxicity , Animals , Autoantibodies/immunology , B-Lymphocytes/immunology , Encephalitis/chemically induced , Encephalitis/immunology , Hashimoto Disease/chemically induced , Hashimoto Disease/immunology , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/immunology , Receptors, N-Methyl-D-Aspartate/immunology
9.
J Neurosci ; 40(8): 1778-1787, 2020 02 19.
Article En | MEDLINE | ID: mdl-31953371

Increase in blood-brain barrier (BBB) permeability is a crucial step in neuroinflammatory processes. We previously showed that N Methyl D Aspartate Receptor (NMDARs), expressed on cerebral endothelial cells forming the BBB, regulate immune cell infiltration across this barrier in the mouse. Here, we describe the mechanism responsible for the action of NMDARs on BBB permeabilization. We report that mouse CNS endothelial NMDARs display the regulatory GluN3A subunit. This composition confers to NMDARs' unconventional properties: these receptors do not induce Ca2+ influx but rather show nonionotropic properties. In inflammatory conditions, costimulation of human brain endothelial cells by NMDA agonists (NMDA or glycine) and the serine protease tissue plasminogen activator, previously shown to potentiate NMDAR activity, induces metabotropic signaling via the Rho/ROCK pathway. This pathway leads to an increase in permeability via phosphorylation of myosin light chain and subsequent shrinkage of human brain endothelial cells. Together, these data draw a link between NMDARs and the cytoskeleton in brain endothelial cells that regulates BBB permeability in inflammatory conditions.SIGNIFICANCE STATEMENT The authors describe how NMDARs expressed on endothelial cells regulate blood-brain barrier function via myosin light chain phosphorylation and increase in permeability. They report that these non-neuronal NMDARs display distinct structural, functional, and pharmacological features than their neuronal counterparts.


Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Myosins/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism , Animals , Blood-Brain Barrier/drug effects , Cell Line , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Endothelial Cells/drug effects , Excitatory Amino Acid Agonists/pharmacology , Male , Mice , N-Methylaspartate/pharmacology , Neurons/drug effects , Neurons/metabolism , Permeability , Phosphorylation/drug effects , Receptors, N-Methyl-D-Aspartate/agonists , Signal Transduction/drug effects , Signal Transduction/physiology , Tissue Plasminogen Activator/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
10.
Int Forum Allergy Rhinol ; 10(3): 395-404, 2020 03.
Article En | MEDLINE | ID: mdl-31721464

BACKGROUND: Despite the development of anterior skull base surgery, the anatomy of the nasal bone and anterior cribriform plate remains unclear. A recent study confirmed 2 distinct foramina in the anterior part of cribriform plate: the ethmoidal slit (ES) and the cribroethmoidal foramen (CF). The aim of this study was to specify their content, their anatomic relationship to the frontal sinus and skull base, and their potential value in skull base surgery. METHODS: Dissections were performed on 36 cadaver heads. Macro- and microscopic examinations were carried out. Microcomputed tomography scans contrasted with osmium were performed to identify vessels and nerves. Histology with neural, meningeal, or luteinizing hormone-releasing hormone immunomarkers was performed on the content of the foramina. Finally, endonasal surgical dissections were carried out. RESULTS: The ES and the CF were observed in all cases. They measured a mean of 4.2 and 1.6 mm, respectively. The ES contained dura mater, arachnoid tissues, lymphatics, and the terminal nerve. The CF contained the anterior ethmoidal nerve and artery. This foramen continued forward with the cribroethmoidal groove, which measured a mean of 2.5 mm. This groove was under the frontal sinus and in front of the skull base. We also described a "cribroethmoidal canal" and a "nasal bone foramen." CONCLUSION: The clinical applications of this new anatomic description concern both cribriform plate and frontal sinus surgeries. Identifying the terminal nerve passing through the ES is a step forward in understanding pheromone recognition in humans.


Ethmoid Bone/blood supply , Ethmoid Bone/innervation , Nasal Bone/anatomy & histology , Aged , Cadaver , Dissection , Ethmoid Bone/diagnostic imaging , Female , Humans , Male , Nasal Bone/diagnostic imaging , Natural Orifice Endoscopic Surgery , Ophthalmic Artery , Ophthalmic Nerve , Skull Base/anatomy & histology , Skull Base/diagnostic imaging , Skull Base/surgery
11.
Dev Neurobiol ; 78(9): 851-858, 2018 09.
Article En | MEDLINE | ID: mdl-30027587

Solute transport through the brain is of major importance for the clearance of toxic molecules and metabolites, and it plays key roles in the pathophysiology of the central nervous system. This solute transport notably depends on the cerebrospinal fluid (CSF) flow, which circulates in the subarachnoid spaces, the ventricles and the perivascular spaces. We hypothesized that the CSF flow may be different in the perinatal period compared to the adult period. Using in vivo magnetic resonance imaging (MRI) and near-infrared fluorescence imaging (NIRF), we assessed the dynamic of the CSF flow in rodents at different ages. By injecting a contrast agent into the CSF, we first used MRI to demonstrate that CSF flow gradually increases with age, with the adult pattern observed at P90. This observation was confirmed by NIRF, which revealed an increased CSF flow in P90 rats when compared with P4 rats not only at the surface of the brain but also deep in the brain structures. Lastly, we evaluated the exit routes of the CSF from the brain. We demonstrated that indocyanine green injected directly into the striatum spread throughout the parenchyma in adult rats, whereas it stayed at the injection point in P4 rats. Moreover, the ability of CSF to exit through the nasal mucosa was increased in the adult rodents. Our results provide evidence that the perinatal brain has nonoptimal CSF flow and exit and, thus, may have impaired clean-up capacity. © 2018 Wiley Periodicals, Inc. Develop Neurobiol, 2018.


Animals, Newborn/cerebrospinal fluid , Biological Transport/physiology , Brain/metabolism , Cerebral Ventricles/physiology , Cerebrospinal Fluid/physiology , Adult , Animals , Humans , Magnetic Resonance Imaging/methods , Mice , Rats, Wistar
12.
Glia ; 65(12): 1961-1975, 2017 12.
Article En | MEDLINE | ID: mdl-28850711

Myelination is a late developmental process regulated by a set of inhibitory and stimulatory factors, including extracellular matrix components. Accordingly, chondroitin sulfate proteoglycans (CSPGs) act as negative regulators of myelination processes. A disintegrin and metalloproteinase with thrombospondin motifs type 4 (ADAMTS-4) is an extracellular protease capable of degrading CSPGs. Although exogenous ADAMTS-4 has been proven to be beneficial in several models of central nervous system (CNS) injuries, the physiological functions of endogenous ADAMTS-4 remain poorly understood. We first used Adamts4/LacZ reporter mice to reveal that ADAMTS-4 is strongly expressed in the CNS, especially in the white matter, with a cellular profile restricted to mature oligodendrocytes. Interestingly, we evidenced an abnormal myelination in Adamts4-/- mice, characterized by a higher diameter of myelinated axons with a shifting g-ratio. Accordingly, lack of ADAMTS-4 is accompanied by motor deficits and disturbed nervous electrical activity. In conclusion, we demonstrate that ADAMTS-4 is a new marker of mature oligodendrocytes contributing to the myelination processes and thus to the control of motor capacities.


ADAMTS4 Protein/metabolism , Movement Disorders/genetics , Oligodendroglia/metabolism , ADAMTS4 Protein/genetics , Animals , Animals, Newborn , Calcium-Binding Proteins/metabolism , Corpus Callosum/metabolism , Corpus Callosum/pathology , Corpus Callosum/ultrastructure , Disease Models, Animal , Evoked Potentials, Somatosensory/genetics , Evoked Potentials, Somatosensory/physiology , Gait Disorders, Neurologic/etiology , Locomotion/genetics , Locomotion/physiology , Male , Mice , Mice, Transgenic , Microfilament Proteins/metabolism , Microscopy, Electron , Movement Disorders/physiopathology , Myelin Basic Protein/metabolism , Nerve Tissue Proteins/metabolism , Oligodendroglia/pathology , Oligodendroglia/ultrastructure , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Statistics, Nonparametric , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
13.
Stroke ; 48(9): 2574-2582, 2017 09.
Article En | MEDLINE | ID: mdl-28754830

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.


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
14.
Mol Neurodegener ; 12(1): 20, 2017 02 23.
Article En | MEDLINE | ID: mdl-28231842

BACKGROUND: The ability of oligodendrocyte progenitor cells (OPCs) to give raise to myelin forming cells during developmental myelination, normal adult physiology and post-lesion remyelination in white matter depends on factors which govern their proliferation, migration and differentiation. Tissue plasminogen activator (tPA) is a serine protease expressed in the central nervous system (CNS), where it regulates cell fate. In particular, tPA has been reported to protect oligodendrocytes from apoptosis and to facilitate the migration of neurons. Here, we investigated whether tPA can also participate in the migration of OPCs during CNS development and during remyelination after focal white matter lesion. METHODS: OPC migration was estimated by immunohistological analysis in spinal cord and corpus callosum during development in mice embryos (E13 to P0) and after white matter lesion induced by the stereotactic injection of lysolecithin in adult mice (1 to 21 days post injection). Migration was compared in these conditions between wild type and tPA knock-out animals. The action of tPA was further investigated in an in vitro chemokinesis assay. RESULTS: OPC migration along vessels is delayed in tPA knock-out mice during development and during remyelination. tPA enhances OPC migration via an effect dependent on the activation of epidermal growth factor receptor. CONCLUSION: Endogenous tPA facilitates the migration of OPCs during development and during remyelination after white matter lesion by the virtue of its epidermal growth factor-like domain.


Cell Differentiation/drug effects , Central Nervous System/growth & development , Neural Stem Cells/drug effects , Oligodendroglia/drug effects , Tissue Plasminogen Activator/pharmacology , Animals , Brain Injuries/pathology , Cell Movement/drug effects , Central Nervous System/drug effects , Corpus Callosum/drug effects , Corpus Callosum/pathology , Embryo, Mammalian , Epidermal Growth Factor , Imaging, Three-Dimensional , Immunoblotting , Immunohistochemistry , Magnetic Resonance Imaging , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Sheath/drug effects , Neural Stem Cells/cytology , Oligodendroglia/cytology , White Matter/drug effects
15.
Cereb Cortex ; 27(10): 4783-4796, 2017 10 01.
Article En | MEDLINE | ID: mdl-27613436

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.


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
16.
Cell Death Dis ; 7(11): e2466, 2016 11 10.
Article En | MEDLINE | ID: mdl-27831563

N-methyl-d-aspartate receptors (NMDARs) are ion channels whose synaptic versus extrasynaptic localization critically influences their functions. This distribution of NMDARs is highly dependent on their lateral diffusion at the cell membrane. Each obligatory subunit of NMDARs (GluN1 and GluN2) contains two extracellular clamshell-like domains with an agonist-binding domain and a distal N-terminal domain (NTD). To date, the roles and dynamics of the NTD of the GluN1 subunit in NMDAR allosteric signaling remain poorly understood. Using single nanoparticle tracking in mouse neurons, we demonstrate that the extracellular neuronal protease tissue-type plasminogen activator (tPA), well known to have a role in the synaptic plasticity and neuronal survival, leads to a selective increase of the surface dynamics and subsequent diffusion of extrasynaptic NMDARs. This process explains the previously reported ability of tPA to promote NMDAR-mediated calcium influx. In parallel, we developed a monoclonal antibody capable of specifically blocking the interaction of tPA with the NTD of the GluN1 subunit of NMDAR. Using this original approach, we demonstrate that the tPA binds the NTD of the GluN1 subunit at a lysine in position 178. Accordingly, when applied to mouse neurons, our selected antibody (named Glunomab) leads to a selective reduction of the tPA-mediated surface dynamics of extrasynaptic NMDARs, subsequent signaling and neurotoxicity, both in vitro and in vivo. Altogether, we demonstrate that the tPA is a ligand of the NTD of the obligatory GluN1 subunit of NMDAR acting as a modulator of their dynamic distribution at the neuronal surface and subsequent signaling.


Cell Membrane/metabolism , Neurons/cytology , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Tissue Plasminogen Activator/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Calcium/metabolism , Cell Death/drug effects , Cell Membrane/drug effects , Diffusion , Fibrinolysin/pharmacology , HEK293 Cells , Humans , Lysine/metabolism , Male , Mice, Inbred BALB C , Neurons/drug effects , Neurotoxins/toxicity , Protein Domains , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/chemistry , Signal Transduction/drug effects , Synapses/drug effects
17.
Brain ; 139(Pt 9): 2406-19, 2016 09.
Article En | MEDLINE | ID: mdl-27435092

Multiple sclerosis is among the most common causes of neurological disability in young adults. Here we provide the preclinical proof of concept of the benefit of a novel strategy of treatment for multiple sclerosis targeting neuroendothelial N-methyl-D-aspartate glutamate receptors. We designed a monoclonal antibody against N-methyl-D-aspartate receptors, which targets a regulatory site of the GluN1 subunit of N-methyl-D-aspartate receptor sensitive to the protease tissue plasminogen activator. This antibody reverted the effect of tissue plasminogen activator on N-methyl-D-aspartate receptor function without affecting basal N-methyl-D-aspartate receptor activity (n = 21, P < 0.01). This antibody bound N-methyl-D-aspartate receptors on the luminal surface of neurovascular endothelium in human tissues and in mouse, at the vicinity of tight junctions of the blood-spinal cord barrier. Noteworthy, it reduced human leucocyte transmigration in an in vitro model of the blood-brain barrier (n = 12, P < 0.05). When injected during the effector phase of MOG-induced experimental autoimmune encephalomyelitis (n = 24), it blocked the progression of neurological impairments, reducing cumulative clinical score (P < 0.001) and mean peak score (P < 0.001). This effect was observed in wild-type animals but not in tissue plasminogen activator knock-out animals (n = 10). This therapeutic effect was associated to a preservation of the blood-spinal cord barrier (n = 6, P < 0.001), leading to reduced leucocyte infiltration (n = 6, P < 0.001). Overall, this study unveils a critical function of endothelial N-methyl-D-aspartate receptor in multiple sclerosis, and highlights the therapeutic potential of strategies targeting the protease-regulated site of N-methyl-D-aspartate receptor.


Blood-Brain Barrier/metabolism , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Excitatory Amino Acid Antagonists/pharmacology , Nerve Tissue Proteins/drug effects , Receptors, N-Methyl-D-Aspartate/drug effects , Tissue Plasminogen Activator/metabolism , Animals , Endothelial Cells , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
18.
Mol Neurobiol ; 53(7): 5000-12, 2016 09.
Article En | MEDLINE | ID: mdl-26377106

Although the extracellular serine protease tissue plasminogen activator (tPA) is involved in pathophysiological processes such as learning and memory, anxiety, epilepsy, stroke, and Alzheimer's disease, information about its regional, cellular, and subcellular distribution in vivo is lacking. In the present study, we observed, in healthy mice and rats, the presence of tPA in endothelial cells, oligodendrocytes, mastocytes, and ependymocytes, but not in pericytes, microglial cells, and astrocytes. Moreover, blockage of the axo-dendritic transport unmasked tPA expression in neurons of cortical and hippocampal areas. Interestingly, combined electrophysiological recordings, single-cell reverse transcription polymerase chain reaction (RT-PCR), and immunohistological analyses revealed that the presence of tPA is restricted to subsets of excitatory pyramidal glutamatergic neurons. We further evidenced that tPA is stored in synaptobrevin-2-positive glutamatergic synaptic vesicles. Based on all these data, we propose the existence of tPA-ergic neurons in the mature brain.


Excitatory Postsynaptic Potentials/physiology , Glutamic Acid/metabolism , Pyramidal Cells/metabolism , Synaptic Vesicles/metabolism , Tissue Plasminogen Activator/biosynthesis , Animals , Cells, Cultured , Gene Expression , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Rats , Rats, Wistar , Tissue Plasminogen Activator/genetics
19.
Neurobiol Dis ; 66: 28-42, 2014 Jun.
Article En | MEDLINE | ID: mdl-24576594

Although tissue plasminogen activator (tPA) is known to promote neuronal remodeling in the CNS, no mechanism of how this plastic function takes place has been reported so far. We provide here in vitro and in vivo demonstrations that this serine protease neutralizes inhibitory chondroitin sulfate proteoglycans (CSPGs) by promoting their degradation via the direct activation of endogenous type 4 disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS-4). Accordingly, in a model of compression-induced spinal cord injury (SCI) in rats, we found that administration of either tPA or its downstream effector ADAMTS-4 restores the tPA-dependent activity lost after the SCI and thereby, reduces content of CSPGs in the spinal cord, a cascade of events leading to an improved axonal regeneration/sprouting and eventually long term functional recovery. This is the first study to reveal a tPA-ADAMTS-4 axis and its function in the CNS. It also raises the prospect of exploiting such cooperation as a therapeutic tool for enhancing recovery after acute CNS injuries.


ADAM Proteins/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Neuronal Plasticity/drug effects , Neuroprotective Agents/pharmacology , Procollagen N-Endopeptidase/metabolism , Spinal Cord Injuries/drug therapy , Tissue Plasminogen Activator/pharmacology , ADAMTS4 Protein , Animals , Axons/drug effects , Axons/physiology , Cells, Cultured , Female , Neurites/drug effects , Neurites/physiology , Neurocan , Neuropeptides/pharmacology , Plasminogen Activator Inhibitor 1/pharmacology , Rats , Rats, Wistar , Recovery of Function , Serine Proteinase Inhibitors/pharmacology , Serpins/pharmacology , Spinal Cord/drug effects , Spinal Cord/physiopathology , Spinal Cord Compression/drug therapy , Spinal Cord Compression/physiopathology , Spinal Cord Injuries/physiopathology , Tissue Plasminogen Activator/antagonists & inhibitors , Neuroserpin
20.
Blood ; 123(21): 3354-63, 2014 May 22.
Article En | MEDLINE | ID: mdl-24553181

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.


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
...