Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Int J Mol Sci ; 24(6)2023 Mar 20.
Article in English | MEDLINE | ID: mdl-36982947

ABSTRACT

Inhaled nitric oxide (iNO) is a therapy used in neonates with pulmonary hypertension. Some evidence of its neuroprotective properties has been reported in both mature and immature brains subjected to injury. NO is a key mediator of the VEGF pathway, and angiogenesis may be involved in the reduced vulnerability to injury of white matter and the cortex conferred by iNO. Here, we report the effect of iNO on angiogenesis in the developing brain and its potential effectors. We found that iNO promotes angiogenesis in the developing white matter and cortex during a critical window in P14 rat pups. This shift in the developmental program of brain angiogenesis was not related to a regulation of NO synthases by exogenous NO exposure, nor the VEGF pathway or other angiogenic factors. The effects of iNO on brain angiogenesis were found to be mimicked by circulating nitrate/nitrite, suggesting that these carriers may play a role in transporting NO to the brain. Finally, our data show that the soluble guanylate cyclase/cGMP signaling pathway is likely to be involved in the pro-angiogenetic effect of iNO through thrombospondin-1, a glycoprotein of the extracellular matrix, inhibiting soluble guanylate cyclase through CD42 and CD36. In conclusion, this study provides new insights into the biological basis of the effect of iNO in the developing brain.


Subject(s)
Nitric Oxide , Rodentia , Animals , Rats , Nitric Oxide/metabolism , Animals, Newborn , Rodentia/metabolism , Soluble Guanylyl Cyclase/metabolism , Vascular Endothelial Growth Factor A/metabolism , Brain/metabolism , Administration, Inhalation
2.
Transl Stroke Res ; 13(4): 512-527, 2022 08.
Article in English | MEDLINE | ID: mdl-34797519

ABSTRACT

Enhancing the collateral blood supply during the acute phase of cerebral ischemia may limit both the extension of the core infarct, by rescuing the penumbra area, and the degree of disability. Many imaging techniques have been applied to rodents in preclinical studies, to evaluate the magnitude of collateral blood flow and the time course of responses during the early phase of ischemic stroke. The collateral supply follows several different routes at the base of the brain (the circle of Willis) and its surface (leptomeningeal or pial arteries), corresponding to the proximal and distal collateral pathways, respectively. In this review, we describe and illustrate the cerebral collateral systems and their modifications following pre-Willis or post-Willis occlusion in rodents. We also review the potential pharmaceutical agents for stimulating the collateral blood supply tested to date. The time taken to establish a collateral blood flow supply through the leptomeningeal anastomoses differs between young and adult animals and between different species and genetic backgrounds. Caution is required when transposing preclinical findings to humans, and clinical trials must be performed to check the added value of pharmacological agents for stimulating the collateral blood supply at appropriate time points. However, collateral recruitment appears to be a rapid, beneficial, endogenous mechanism that can be stimulated shortly after artery occlusion. It should be considered a treatment target for use in addition to recanalization strategies.


Subject(s)
Brain Ischemia , Stroke , Animals , Cerebral Infarction , Cerebrovascular Circulation/physiology , Collateral Circulation/physiology
3.
Curr Issues Mol Biol ; 43(1): 301-312, 2021 Jun 07.
Article in English | MEDLINE | ID: mdl-34200155

ABSTRACT

The poly(ADP-ribose) polymerase inhibitor PJ34 has recently been reported to increase cerebral blood flow, via the endothelial NO synthase, in the naive mouse brain throughout life. We addressed here the benefits of PJ34 after neonatal ischemia on hemodynamics and components of the neurovascular unit including the blood-brain barrier (BBB), microglia, and astrocytes. Nine-day-old mice were subjected to permanent MCA occlusion (pMCAo), and treated with either PBS or PJ34 (10 mg/kg). Mean blood-flow velocities (mBFV) were measured in both internal carotid arteries (ICA) and basilar trunk (BT) using Doppler-ultrasonography. BBB opening was assessed through somatostatin-receptor type-2 internalization and immunohistochemistry at 24 and 48 h. Lesion areas were measured 8 days after ischemia. In PBS-treated mice, pMCAo involved a drop in mBFV in the left ICA (p < 0.001 vs. basal), whereas mBFV remained stable in both right ICA and BT. PJ34 prevented this drop in the left ICA (NS vs. basal) and increased mBFV in the right ICA (p = 0.0038 vs. basal). No modification was observed in the BT. In contrast to PBS, BBB disruption extent and astrocyte demise were reduced in PJ34 mice only in the rostral brain at 48 h and 8 days post-pMCAo, respectively. Accordingly, 8 days after pMCAo, affected areas were reduced in the rostral brain (Bregma +0.86 and +0.14 mm), whereas total tissue loss was not reduced after PJ34 (4.0 ± 3.1%) vs. PBS (5.8 ± 3.4%). These results show that PJ34 reduced BBB permeability, astrocyte demise, and tissue loss (particularly in the rostral territories), suggesting that collateral supply mainly proceeds from the anterior ICA's branches in the ischemic neonatal mouse brain.


Subject(s)
Brain/drug effects , Phenanthrenes/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Stroke/drug therapy , Animals , Animals, Newborn , Astrocytes/cytology , Astrocytes/metabolism , Blood-Brain Barrier , Brain/metabolism , Brain Ischemia/pathology , Carotid Artery, Internal/pathology , Female , Hemodynamics , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , Permeability , Phenotype , Stroke/physiopathology , Ultrasonography, Doppler
4.
Circ Res ; 128(3): 363-382, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33301355

ABSTRACT

RATIONALE: Cerebrovascular function is critical for brain health, and endogenous vascular protective pathways may provide therapeutic targets for neurological disorders. S1P (Sphingosine 1-phosphate) signaling coordinates vascular functions in other organs, and S1P1 (S1P receptor-1) modulators including fingolimod show promise for the treatment of ischemic and hemorrhagic stroke. However, S1P1 also coordinates lymphocyte trafficking, and lymphocytes are currently viewed as the principal therapeutic target for S1P1 modulation in stroke. OBJECTIVE: To address roles and mechanisms of engagement of endothelial cell S1P1 in the naive and ischemic brain and its potential as a target for cerebrovascular therapy. METHODS AND RESULTS: Using spatial modulation of S1P provision and signaling, we demonstrate a critical vascular protective role for endothelial S1P1 in the mouse brain. With an S1P1 signaling reporter, we reveal that abluminal polarization shields S1P1 from circulating endogenous and synthetic ligands after maturation of the blood-neural barrier, restricting homeostatic signaling to a subset of arteriolar endothelial cells. S1P1 signaling sustains hallmark endothelial functions in the naive brain and expands during ischemia by engagement of cell-autonomous S1P provision. Disrupting this pathway by endothelial cell-selective deficiency in S1P production, export, or the S1P1 receptor substantially exacerbates brain injury in permanent and transient models of ischemic stroke. By contrast, profound lymphopenia induced by loss of lymphocyte S1P1 provides modest protection only in the context of reperfusion. In the ischemic brain, endothelial cell S1P1 supports blood-brain barrier function, microvascular patency, and the rerouting of blood to hypoperfused brain tissue through collateral anastomoses. Boosting these functions by supplemental pharmacological engagement of the endothelial receptor pool with a blood-brain barrier penetrating S1P1-selective agonist can further reduce cortical infarct expansion in a therapeutically relevant time frame and independent of reperfusion. CONCLUSIONS: This study provides genetic evidence to support a pivotal role for the endothelium in maintaining perfusion and microvascular patency in the ischemic penumbra that is coordinated by S1P signaling and can be harnessed for neuroprotection with blood-brain barrier-penetrating S1P1 agonists.


Subject(s)
Blood-Brain Barrier/metabolism , Cerebral Arteries/metabolism , Endothelial Cells/metabolism , Infarction, Middle Cerebral Artery/metabolism , Ischemic Attack, Transient/metabolism , Ischemic Stroke/metabolism , Lysophospholipids/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Sphingosine/analogs & derivatives , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Blood-Brain Barrier/physiopathology , Cerebral Arteries/drug effects , Cerebral Arteries/pathology , Cerebral Arteries/physiopathology , Cerebrovascular Circulation , Disease Models, Animal , Endothelial Cells/pathology , Female , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Infarction, Middle Cerebral Artery/prevention & control , Ischemic Attack, Transient/pathology , Ischemic Attack, Transient/physiopathology , Ischemic Attack, Transient/prevention & control , Ischemic Stroke/pathology , Ischemic Stroke/physiopathology , Ischemic Stroke/prevention & control , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Microcirculation , Neuroprotective Agents/pharmacology , Signal Transduction , Sphingosine/metabolism , Sphingosine-1-Phosphate Receptors/agonists , Sphingosine-1-Phosphate Receptors/genetics , Vascular Patency
5.
Int J Mol Sci ; 21(18)2020 Sep 08.
Article in English | MEDLINE | ID: mdl-32911782

ABSTRACT

The poly(ADP-ribose) polymerase (PARP) inhibitor PJ34 has been reported to improve endothelial dysfunction in the peripheral system. We addressed the role of PJ34 on the vascular tone and vasoreactivity during development in the mouse brain. Blood flows were measured in the basilar trunk using ultrasonography. Cerebral vasoreactivity or vasodilation reserve was estimated as a percentage increase in mean blood flow velocities (mBFV) recorded under normoxia-hypercapnia in control and after PJ34 administration. Non-selective and selective eNOS and nNOS inhibitors were used to evaluate the role of NO-pathway into the hemodynamic effects of PJ34. PJ34 increased mBFVs from 15.8 ± 1.6 to 19.1 ± 1.9 cm/s (p = 0.0043) in neonatal, from 14.6 ± 1.4 to 16.1 ± 0.9 cm/s (p = 0.0049) in adult, and from 15.7 ± 1.7 to 17.5 ± 2.0 cm/s (p = 0.0024) in aged mice 48 h after administration. These PJ34 values were similar to those measured in age-matched control mice under normoxia-hypercapnia. This recruitment was mediated through the activation of constitutive NO synthases in both the neonatal (38.2 ± 6.7 nmol/min/mg protein) and adult (31.5 ± 4.4 nmol/min/mg protein) brain, as compared to age-matched control brain (6.9 ± 0.4 and 6.3 ± 0.7 nmol/min/mg protein), respectively. In addition, quite selective eNOS inhibitor was able to inhibit the recruitment. PJ34 by itself is able to increase cerebral blood flow through the NO-pathway activation at least over 48 h after a single administration.


Subject(s)
Nitric Oxide/metabolism , Phenanthrenes/metabolism , Phenanthrenes/pharmacology , Age Factors , Animals , Animals, Newborn/metabolism , Brain/embryology , Brain/metabolism , Cerebrovascular Circulation/drug effects , Cerebrovascular Circulation/physiology , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Vasodilator Agents/metabolism , Vasodilator Agents/pharmacology
6.
Int J Mol Sci ; 20(15)2019 Aug 04.
Article in English | MEDLINE | ID: mdl-31382688

ABSTRACT

We recently reported that neonatal ischemia induces microglia/macrophage activation three days post-ischemia. We also found that female mice sustained smaller infarcts than males three months post-ischemia. The objective of our current study was to examine whether differential acute neuroinflammatory response and infiltrated immune cells occurs between male and females after three days post-ischemia. Permanent middle cerebral artery occlusion was induced in male and female postnatal 9-day-old (P9) mice, and mice were sacrificed three days after ischemia. Brains were analyzed for mRNA transcription after microglia magnetic cell sorting to evaluate M1 and M2 markers. FACS analysis was performed to assess myeloid infiltration and microglial expression of CX3 chemokine receptor 1 (CX3CR1). Inflammatory cytokine expression and microglia/macrophage activation were analyzed via in situ hybridization combined with immunofluorescence techniques. Lesion volume and cell death were measured. An increase in microglia/macrophages occurred in male versus female mice. The cells exhibited amoeboid morphology, and TNFα and ptgs2 (Cox-2) genes were more expressed in males. More myeloid cell infiltration was found in male versus female brains. However, we did not observe sex-dependent differences in the injured volume or cell death density. Our data show that sex differences in the acute microglial and immune responses to neonatal ischemia are likely both gene- and region-specific.


Subject(s)
Brain Ischemia/immunology , Immunity, Innate/genetics , Inflammation/immunology , Stroke/immunology , Animals , Animals, Newborn/immunology , Brain/immunology , Brain/pathology , Brain Ischemia/genetics , Brain Ischemia/pathology , Disease Models, Animal , Female , Infarction, Middle Cerebral Artery , Inflammation/genetics , Inflammation/pathology , Macrophage Activation/genetics , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Mice , Sex Characteristics , Stroke/genetics , Stroke/pathology
8.
Brain Behav Immun ; 80: 315-327, 2019 08.
Article in English | MEDLINE | ID: mdl-30953775

ABSTRACT

Stroke is currently the second leading cause of death in industrialized countries and the second cause of dementia after Alzheimer's disease. Diabetes is an independent risk factor for stroke that exacerbates the severity of lesions, disability and cognitive decline. There is increasing evidence that sustained brain inflammation may account for this long-term prejudicial outcome in diabetic patients in particular. We sought to demonstrate that experimental permanent middle cerebral artery occlusion (pMCAo) in the diabetic mouse aggravates stroke, induces cognitive decline, and is associated with exacerbated brain inflammation, and that these effects can be alleviated and/or prevented by the immunomodulator, glatiramer acetate (GA). Male diabetic C57Bl6 mice (streptozotocin IP) subjected to permanent middle cerebral artery occlusion (pMCAo), were treated by the immunomodulator, GA (Copaxone®) (1 mg/kg daily, sc) until 3 or 7 days post stroke. Infarct volume, brain pro- and anti-inflammatory mediators, microglial/macrophage density, and neurogenesis were monitored during the first week post stroke. Neurological sensorimotor deficit, spatial memory and brain deposits of Aß40 and Aß42 were assessed until six weeks post stroke. In diabetic mice with pMCAo, proinflammatory mediators (IL-1ß, MCP1, TNFα and CD68) were significantly higher than in non-diabetic mice. In GA-treated mice, the infarct volume was reduced by 30% at D3 and by 40% at D7 post stroke (P < 0.05), sensorimotor recovery was accelerated as early as D3, and long-term memory loss was prevented. Moreover, proinflammatory mediators significantly decreased between D3 (COX2) and D7 (CD32, TNFα, IL-1ß), and neurogenesis was significantly increased at D7. Moreover, GA abrogates the accumulation of insoluble Aß40. This work is the first one to evidence that the immunomodulatory drug GA reduces infarct volume and proinflammatory mediators, enhances early neurogenesis, accelerates sensorimotor recovery, and prevents long-term memory loss in diabetic mice with pMCAo.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Brain Infarction/immunology , Diabetes Complications , Glatiramer Acetate/administration & dosage , Memory Disorders/immunology , Neuroprotective Agents/administration & dosage , Stroke/complications , Animals , Brain/drug effects , Brain/immunology , Brain Infarction/complications , Brain Infarction/prevention & control , Diabetes Complications/immunology , Encephalitis/etiology , Encephalitis/immunology , Inflammation Mediators/immunology , Male , Memory Disorders/etiology , Memory Disorders/prevention & control , Mice, Inbred C57BL , Microglia/drug effects , Neurogenesis/drug effects , Stroke/immunology
9.
Glia ; 67(2): 345-359, 2019 02.
Article in English | MEDLINE | ID: mdl-30506969

ABSTRACT

Prematurity and fetal growth restriction (FGR) are frequent conditions associated with adverse neurocognitive outcomes. We have previously identified early deregulation of genes controlling neuroinflammation as a putative mechanism linking FGR and abnormal trajectory of the developing brain. While the oxytocin system was also found to be impaired following adverse perinatal events, its role in the modulation of neuroinflammation in the developing brain is still unknown. We used a double-hit rat model of perinatal brain injury induced by gestational low protein diet (LPD) and potentiated by postnatal injections of subliminal doses of interleukin-1ß (IL1ß) and a zebrafish model of neuroinflammation. Effects of the treatment with carbetocin, a selective, long lasting, and brain diffusible oxytocin receptor agonist, have been assessed using a combination of histological, molecular, and functional tools in vivo and in vitro. In the double-hit model, white matter inflammation, deficient myelination, and behavioral deficits have been observed and the oxytocin system was impaired. Early postnatal supplementation with carbetocin alleviated microglial activation at both transcriptional and cellular levels and provided long-term neuroprotection. The central anti-inflammatory effects of carbetocin have been shown in vivo in rat pups and in a zebrafish model of early-life neuroinflammation and reproduced in vitro on stimulated sorted primary microglial cell cultures from rats subjected to LPD. Carbetocin treatment was associated with beneficial effects on myelination, long-term intrinsic brain connectivity and behavior. Targeting oxytocin signaling in the developing brain may be an effective approach to prevent neuroinflammation - induced brain damage of perinatal origin.


Subject(s)
Brain Injuries/drug therapy , Brain/pathology , Microglia/drug effects , Receptors, Oxytocin/metabolism , Animals , Animals, Genetically Modified , Animals, Newborn , Brain Injuries/chemically induced , Brain Injuries/pathology , Cells, Cultured , Computational Biology , Diet, Protein-Restricted/adverse effects , Disease Models, Animal , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Interleukin-1beta , Lipopolysaccharides/toxicity , Oxytocics/therapeutic use , Oxytocin/analogs & derivatives , Oxytocin/therapeutic use , Peptide Fragments , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/physiopathology , RNA, Messenger/metabolism , Zebrafish
10.
Int J Mol Sci ; 19(10)2018 Sep 30.
Article in English | MEDLINE | ID: mdl-30274381

ABSTRACT

While arterial reflow after a stroke represents an important challenge for better outcomes, it is also very important that sudden recanalization does not produce local oxidative and nitrogen species, deleterious for the brain and more particularly the immature brain. Our objective was to determine whether a supply in prostaglandin (Pg) E1 (Alprostadil), via its action on arterial pressure, might progressively improve cerebral reperfusion in a neonatal stroke model. Arterial blood flow was measured using ultrasonography. Rate-limiting and Pg terminal synthesizing enzymes were evaluated using reverse-transcriptase polymerase chain reaction. Our data suggests that a supply in PgE1 might delay and improve the ipsilateral reperfusion by decreasing thromboxane A synthase-1 gene, the density of reactive astrocytes and lesion volume.


Subject(s)
Alprostadil/therapeutic use , Collateral Circulation/drug effects , Stroke/drug therapy , Stroke/physiopathology , Alprostadil/pharmacology , Animals , Animals, Newborn , Brain Ischemia/drug therapy , Brain Ischemia/enzymology , Brain Ischemia/genetics , Brain Ischemia/physiopathology , Cerebrovascular Circulation/drug effects , Disease Models, Animal , Female , Male , Rats, Wistar , Stroke/complications , Stroke/enzymology , Thromboxane-A Synthase/genetics , Thromboxane-A Synthase/metabolism
11.
Front Neurol ; 9: 650, 2018.
Article in English | MEDLINE | ID: mdl-30131764

ABSTRACT

Neonatal hypoxia-ischemia (HI) and ischemia are a common cause of neonatal brain injury resulting in cerebral palsy with subsequent learning disabilities and epilepsy. Recent data suggest a higher incidence of focal ischemia-reperfusion located in the middle cerebral artery (MCA) territory in near-term and newborn babies. Pre-clinical studies in the field of cerebral palsy research used, and still today, the classical HI model in the P7 rat originally described by Rice et al. (1). At the end of the 90s, we designed a new model of focal ischemia in the P7 rat to explore the short and long-term pathophysiology of neonatal arterial ischemic stroke, particularly the phenomenon of reperfusion injury and its sequelae (reported in 1998). Cerebral blood-flow and cell death/damage correlates have been fully characterized. Pharmacologic manipulations have been applied to the model to test therapeutic targets. The model has proven useful for the study of seizure occurrence, a clinical hallmark for neonatal ischemia in babies. Main pre-clinical findings obtained within these 20 last years are discussed associated to clinical pattern of neonatal brain damage.

12.
Brain Behav Immun ; 73: 375-389, 2018 10.
Article in English | MEDLINE | ID: mdl-29852289

ABSTRACT

Neonatal acute ischemic stroke is a cause of neonatal brain injury that occurs more frequently in males, resulting in associated neurobehavioral disorders. The bases for these sex differences are poorly understood but might include the number, morphology and activation of microglia in the developing brain when subjected to stroke. Interestingly, poly (ADP-ribose) polymerase (PARP) inhibition preferentially protects males against neonatal ischemia. This study aims to examine the effects of PJ34, a PARP inhibitor, on microglial phenotypes at 3 and 8 days and on neurobehavioral disorders in adulthood for both male and female P9 mice subjected to permanent middle cerebral artery occlusion (pMCAo). PJ34 significantly reduced the lesion size by 78% and reduced the density of CX3CR1gfp-labeled microglial cells by 46% when examined 3 days after pMCAo in male but not in female mice. Eight days after pMCAo, the number of Iba1+/Cox-2+ cells did not differ between male and female mice in the cortical peri-infarct region. In the amygdala, Iba1+/Cox-2+ (M1-like) cell numbers were significantly decreased in PJ34-treated males but not in females. Conversely, Iba1+/Arg-1+ (M2-like) and Arg-1+/Cox-2+ (Mtransitional) cell numbers were significantly increased in PJ34-treated females. Regarding neurobehavioral disorders during adulthood, pMCAo induced a motor coordination deficit and a spatial learning deficit in female mice only. PJ34 prevented MBP fibers, motor coordination and learning disorders during adulthood in female mice. Our data show significant sex differences in the effects of PARP inhibition on microglia phenotypes following neonatal ischemia, associated with improved behavior and myelination during adulthood in females only. Our findings suggest that modulating microglial phenotypes may play key roles in behavior disorders and white matter injury following neonatal stroke.


Subject(s)
Brain Ischemia/pathology , Microglia/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/metabolism , Animals , Animals, Newborn , Brain Injuries/complications , Brain Ischemia/metabolism , Disease Models, Animal , Female , Infarction, Middle Cerebral Artery/physiopathology , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Neurons/drug effects , Phenanthrenes/metabolism , Phenanthrenes/pharmacology , Phenotype , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly (ADP-Ribose) Polymerase-1/metabolism , Sex Factors , Stroke/pathology
13.
Int J Mol Sci ; 19(1)2017 Dec 26.
Article in English | MEDLINE | ID: mdl-29278365

ABSTRACT

Cohort studies have demonstrated a higher vulnerability in males towards ischemic and/or hypoxic-ischemic injury in infants born near- or full-term. Male sex was also associated with limited brain repair following neonatal stroke and hypoxia-ischemia, leading to increased incidence of long-term cognitive deficits compared to females with similar brain injury. As a result, the design of pre-clinical experiments considering sex as an important variable was supported and investigated because neuroprotective strategies to reduce brain injury demonstrated sexual dimorphism. While the mechanisms underlining these differences between boys and girls remain unclear, several biological processes are recognized to play a key role in long-term neurodevelopmental outcomes: gonadal hormones across developmental stages, vulnerability to oxidative stress, modulation of cell death, and regulation of microglial activation. This review summarizes the current evidence for sex differences in neonatal hypoxic-ischemic and/or ischemic brain injury, considering the major pathways known to be involved in cognitive and behavioral deficits associated with damages of the developing brain.


Subject(s)
Brain/pathology , Hypoxia-Ischemia, Brain/epidemiology , Hypoxia-Ischemia, Brain/pathology , Animals , Brain/growth & development , Brain/metabolism , Brain/physiopathology , Cell Death , Disease Models, Animal , Female , Gonadal Hormones/metabolism , Humans , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/physiopathology , Infant, Newborn , Male , Microglia/metabolism , Microglia/pathology , Oxidative Stress , Sex Characteristics , Sex Factors
14.
J Cereb Blood Flow Metab ; 37(9): 3091-3096, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28695754

ABSTRACT

Our objective was to determine whether controlled reflow on one side and/or the other side after bilateral carotid occlusion release could reduce cell death in focal ischemic P14 rats. Arterial blood flow was measured using ultrasonography. Cell death, inflammation and nitrotyrosine were measured using immunofluorescence. When reflow was first induced in the contralateral side, we observed improved outcome markers compared with those when reflow was first induced in the ipsilateral side and/or simultaneous reflow was induced in both sides. Our data suggest that progressive rerouting of arterial flow through the circle of Willis toward the ischemic site reduced cell death.


Subject(s)
Brain Ischemia/therapy , Cerebral Arteries/physiopathology , Cerebrovascular Circulation/physiology , Reperfusion Injury/physiopathology , Reperfusion/methods , Animals , Brain Ischemia/diagnostic imaging , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cell Death , Cerebral Arteries/pathology , Cyclooxygenase 2/genetics , Disease Models, Animal , Prostaglandin-E Synthases/genetics , Rats, Wistar , Reperfusion Injury/diagnostic imaging , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Treatment Outcome , Tyrosine/analogs & derivatives , Tyrosine/biosynthesis , Ultrasonography
15.
Oncotarget ; 8(30): 48531-48532, 2017 Jul 25.
Article in English | MEDLINE | ID: mdl-28562317
16.
Neurobiol Dis ; 99: 145-153, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28042096

ABSTRACT

Perinatal arterial stroke is the most frequent form of cerebral infarction in children. Neonatal seizures are the most frequent symptom during the neonatal period. The current management of perinatal stroke is based on supportive care. It is currently unknown if treatment of the seizures modifies the outcome, and no clinical studies have focused on seizures during neonatal stroke. We studied the effect of phenobarbital and levetiracetam on an ischemic-reperfusion stroke model in P7 rats using prolonged electroencephalographic recordings and a histologic analysis of the brain (24h after injury). The following two types of epileptic events were observed: 1) bursts of high amplitude spikes during ischemia and the first hours of reperfusion and 2) organized seizures consisting in discharges of a 1-2Hz spike-and-wave. Both phenobarbital and levetiracetam decreased the total duration of the bursts of high amplitude spikes. Phenobarbital also delayed the start of seizures without changing the total duration of epileptic discharges. The markedly limited efficacy of the antiepileptic drugs studied in our neonatal stroke rat model is frequently observed in human neonatal seizures. Both drugs did not modify the stroke volume, which suggests that the modification of the quantity of bursts of high amplitude spikes does not influence the infarct size. In the absence of a reduction in seizure burden by the antiepileptic drugs, we increased the seizure burden and stroke volume by combining our neonatal stroke model with a lithium-pilocarpine-induced status epilepticus. Our data suggest that the reduction of burst of spikes did not influence the stroke volume. The presence of organized seizure with a pattern close to what is observed in human newborns seems related to the presence of the infarct. Further research is required to determine the relationship between seizure burden and infarct volume.


Subject(s)
Anticonvulsants/pharmacology , Brain Ischemia/drug therapy , Brain/drug effects , Epilepsy/drug therapy , Reperfusion Injury/drug therapy , Stroke/drug therapy , Animals , Animals, Newborn , Brain/diagnostic imaging , Brain/pathology , Brain/physiopathology , Brain Ischemia/diagnostic imaging , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Disease Models, Animal , Epilepsy/diagnostic imaging , Epilepsy/pathology , Epilepsy/physiopathology , Female , Levetiracetam , Lithium Compounds , Male , Phenobarbital/pharmacology , Pilocarpine , Piracetam/analogs & derivatives , Piracetam/pharmacology , Random Allocation , Rats, Wistar , Reperfusion Injury/diagnostic imaging , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Stroke/diagnostic imaging , Stroke/pathology , Stroke/physiopathology
17.
J Cereb Blood Flow Metab ; 37(2): 396-412, 2017 Feb.
Article in English | MEDLINE | ID: mdl-26762506

ABSTRACT

Insulin-like growth factors control numerous processes, namely somatic growth, metabolism and stress resistance, connecting this pathway to aging and age-related diseases. Insulin-like growth factor signaling also impacts on neurogenesis, neuronal survival and structural plasticity. Recent reports demonstrated that diminished insulin-like growth factor signaling confers increased stress resistance in brain and other tissues. To better understand the role of neuronal insulin-like growth factor signaling in neuroprotection, we inactivated insulin-like growth factor type-1-receptor in forebrain neurons using conditional Cre-LoxP-mediated gene targeting. We found that brain structure and function, including memory performance, were preserved in insulin-like growth factor receptor mutants, and that certain characteristics improved, notably synaptic transmission in hippocampal neurons. To reveal stress-related roles of insulin-like growth factor signaling, we challenged the brain using a stroke-like insult. Importantly, when charged with hypoxia-ischemia, mutant brains were broadly protected from cell damage, neuroinflammation and cerebral edema. We also found that in mice with insulin-like growth factor receptor knockout specifically in forebrain neurons, a substantial systemic upregulation of growth hormone and insulin-like growth factor-I occurred, which was associated with significant somatic overgrowth. Collectively, we found strong evidence that blocking neuronal insulin-like growth factor signaling increases peripheral somatotropic tone and simultaneously protects the brain against hypoxic-ischemic injury, findings that may contribute to developing new therapeutic concepts preventing the disabling consequences of stroke.


Subject(s)
Gene Deletion , Growth Hormone/metabolism , Neuroprotection , Prosencephalon/pathology , Receptor, IGF Type 1/genetics , Stroke/genetics , Stroke/pathology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Neurons/pathology , Prosencephalon/metabolism , Stroke/metabolism , Up-Regulation
18.
Stroke ; 47(12): 3048-3052, 2016 12.
Article in English | MEDLINE | ID: mdl-27834752

ABSTRACT

BACKGROUND AND PURPOSE: We previously showed that the selective neuronal nitric oxide synthase inhibitor 7-nitroindazole (7-NI) increases cerebral microcirculation in a juvenile ischemic rat model. We address the roles of cyclooxygenase (COX)-elaborated prostaglandins in collateral recruitment and blood supply. METHODS: Fourteen-day-old rats were subjected to ischemia-reperfusion and treated with either PBS or 7-NI (25 mg/kg) at the reperfusion onset. Six-keto-prostaglandin F1α was measured using ELISA. COX-1 and COX-2 and prostaglandin terminal synthesizing enzymes were evaluated using reverse-transcriptase polymerase chain reaction and immunofluorescence. Microvascular blood flow indexes (artery diameter and capillaries number) were measured using sidestream dark-field videomicroscopy in PBS- and 7-NI-treated ischemic rats in the absence or presence of the COX-2 inhibitor NS-398 (5 mg/kg). Cell death was measured with the TUNEL (terminal transferase dUTP nick end labeling) assay and cleaved-caspase-3 immunostaining. RESULTS: Six-keto-prostaglandin F1α and COX-2, associated with a prostaglandin E synthase, were significantly increased in PBS- and 7-NI-treated animals 15 minutes and 1 hour after ischemia-reperfusion, respectively. In contrast and as compared with PBS, 7-NI significantly decreased prostacyclin synthase and cytosolic prostaglandins E synthase mRNA. Selective COX-2 inhibition significantly decreased blood flow indexes and significantly reversed the effects of 7-NI, including the number of TUNEL+- and cleaved-caspase-3+-nuclei. CONCLUSIONS: These results show that the juvenile rat brains mostly respond to ischemia by a COX-2-dependent prostaglandins production and suggest that the transcriptional responses observed under 7-NI facilitate and reorient COX-2-dependent prostaglandins production.


Subject(s)
6-Ketoprostaglandin F1 alpha/metabolism , Cerebrovascular Circulation , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2/metabolism , Microcirculation , Prostaglandin-E Synthases/metabolism , Reperfusion Injury/metabolism , Animals , Disease Models, Animal , Rats
19.
Glia ; 64(12): 2306-2320, 2016 12.
Article in English | MEDLINE | ID: mdl-27687291

ABSTRACT

Fetal growth restriction (FGR) is a major complication of human pregnancy, frequently resulting from placental vascular diseases and prenatal malnutrition, and is associated with adverse neurocognitive outcomes throughout life. However, the mechanisms linking poor fetal growth and neurocognitive impairment are unclear. Here, we aimed to correlate changes in gene expression induced by FGR in rats and abnormal cerebral white matter maturation, brain microstructure, and cortical connectivity in vivo. We investigated a model of FGR induced by low-protein-diet malnutrition between embryonic day 0 and birth using an interdisciplinary approach combining advanced brain imaging, in vivo connectivity, microarray analysis of sorted oligodendroglial and microglial cells and histology. We show that myelination and brain function are both significantly altered in our model of FGR. These alterations, detected first in the white matter on magnetic resonance imaging significantly reduced cortical connectivity as assessed by ultrafast ultrasound imaging. Fetal growth retardation was found associated with white matter dysmaturation as shown by the immunohistochemical profiles and microarrays analyses. Strikingly, transcriptomic and gene network analyses reveal not only a myelination deficit in growth-restricted pups, but also the extensive deregulation of genes controlling neuroinflammation and the cell cycle in both oligodendrocytes and microglia. Our findings shed new light on the cellular and gene regulatory mechanisms mediating brain structural and functional defects in malnutrition-induced FGR, and suggest, for the first time, a neuroinflammatory basis for the poor neurocognitive outcome observed in growth-restricted human infants. GLIA 2016;64:2306-2320.


Subject(s)
Brain Injuries/etiology , Brain Injuries/pathology , Fetal Growth Retardation/physiopathology , Microglia/metabolism , Oligodendroglia/metabolism , Transcriptome/physiology , Adenomatous Polyposis Coli Protein/metabolism , Animals , Animals, Newborn , Antigens/metabolism , Antigens, CD/metabolism , Brain/diagnostic imaging , Brain/drug effects , Brain Injuries/diagnostic imaging , Cytokines/metabolism , Female , Gene Expression/physiology , Lipopolysaccharides/pharmacology , Myelin Basic Protein/metabolism , Neural Pathways/diagnostic imaging , Neural Pathways/drug effects , Oligodendrocyte Transcription Factor 2/metabolism , Pregnancy , Proteoglycans/metabolism , Rats , Rats, Sprague-Dawley
20.
Exp Neurol ; 282: 56-65, 2016 08.
Article in English | MEDLINE | ID: mdl-27222132

ABSTRACT

White-matter injury is the most common cause of the adverse neurodevelopmental outcomes observed in preterm infants. Only few options exist to prevent perinatal brain injury associated to preterm delivery. 17ß-estradiol (E2) is the predominant estrogen in circulation and has been shown to be neuroprotective in vitro and in vivo. However, while E2 has been found to modulate inflammation in adult models of brain damage, how estrogens influence glial cells response in the developing brain needs further investigations. Using a model of ibotenate-induced brain injury, we have refined the effects of E2 in the developing brain. E2 provides significant neuroprotection both in the cortical plate and the white matter in neonatal rats subjected to excitotoxic insult mimicking white matter and cortical damages frequently observed in very preterm infants. E2 promotes significant changes in microglial phenotypes balance in response to brain injury and the acceleration of oligodendrocyte maturation. Maturational effects of E2 on myelination process were observed both in vivo and in vitro. Altogether, these data demonstrate that response of glial cells to E2 could be responsible for its neuroprotective properties in neonatal excitotoxic brain injury.


Subject(s)
Estradiol/therapeutic use , Leukoencephalopathies/therapy , Neuroglia/drug effects , Neuroprotective Agents/therapeutic use , Adenomatous Polyposis Coli Protein/metabolism , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Embryo, Mammalian , Estradiol/pharmacology , Excitatory Amino Acid Agonists/toxicity , Ibotenic Acid/toxicity , Leukoencephalopathies/chemically induced , Myelin Basic Protein/metabolism , Nerve Tissue Proteins/metabolism , Neuroglia/physiology , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Oligodendrocyte Transcription Factor 2 , Plant Lectins/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...