Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Sci Rep ; 13(1): 15841, 2023 09 22.
Article in English | MEDLINE | ID: mdl-37740008

ABSTRACT

Despite efforts to identify modulatory neuroprotective mechanisms of damaging ischemic stroke cascade signaling, a void remains on an effective potential therapeutic. The present study defines neuroprotection by very long-chain polyunsaturated fatty acid (VLC-PUFA) Elovanoid (ELV) precursors C-32:6 and C-34:6 delivered intranasally following experimental ischemic stroke. We demonstrate that these precursors improved neurological deficit, decreased T2WI lesion volume, and increased SMI-71 positive blood vessels and NeuN positive neurons, indicating blood-brain barrier (BBB) protection and neurogenesis modulated by the free fatty acids (FFAs) C-32:6 and C-34:6. Gene expression revealed increased anti-inflammatory and pro-homeostatic genes and decreases in expression of pro-inflammatory genes in the subcortex. Additionally, the FFAs elicit a comprehensive downregulation of inflammatory microglia/monocyte-derived macrophages and astrocyte-associated genes in the subcortical region. Functional analysis reveals inhibition of immune-related pathways and production of upstream molecules related to detrimental signaling events in post-stroke acute and subacute phases.


Subject(s)
Ischemic Stroke , Stroke , Humans , Fatty Acids, Nonesterified , Neuroprotection , Stroke/genetics , Astrocytes
2.
Cell Mol Neurobiol ; 43(7): 3555-3573, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37270727

ABSTRACT

Neuroprotection to attenuate or block the ischemic cascade and salvage neuronal damage has been extensively explored for treating ischemic stroke. However, despite increasing knowledge of the physiologic, mechanistic, and imaging characterizations of the ischemic penumbra, no effective neuroprotective therapy has been found. This study focuses on the neuroprotective bioactivity of docosanoid mediators: Neuroprotectin D1 (NPD1), Resolvin D1 (RvD1), and their combination in experimental stroke. Molecular targets of NPD1 and RvD1 are defined by following dose-response and therapeutic window. We demonstrated that treatment with NPD1, RvD1, and combination therapy provides high-grade neurobehavioral recovery and decreases ischemic core and penumbra volumes even when administered up to 6 h after stroke. The expression of the following genes was salient: (a) Cd163, an anti-inflammatory stroke-associated gene, was the most differentially expressed gene by NPD1+RvD1, displaying more than a 123-fold upregulation in the ipsilesional penumbra (Lisi et al., Neurosci Lett 645:106-112, 2017); (b) 100-fold upregulation takes place in astrocyte gene PTX3, a key regulator of neurogenesis and angiogenesis after cerebral ischemia (. Rodriguez-Grande et al., J Neuroinflammation 12:15, 2015); and (c) Tmem119 and P2y12, two markers of homeostatic microglia, were found to be enhanced by ten- and fivefold, respectively (Walker et al. Int J Mol Sci 21:678, 2020). Overall, we uncovered that protection after middle cerebral artery occlusion (MCAo) by the lipid mediators elicits expression of microglia and astrocyte-specific genes (Tmem119, Fcrls, Osmr, Msr1, Cd68, Cd163, Amigo2, Thbs1, and Tm4sf1) likely participating in enhancing homeostatic microglia, modulating neuroinflammation, promoting DAMP clearance, activating NPC differentiation and maturation, synapse integrity and contributing to cell survival.


Subject(s)
Brain Ischemia , Ischemic Stroke , Stroke , Humans , Ischemic Stroke/metabolism , Microglia/metabolism , Astrocytes/metabolism , Stroke/drug therapy , Brain Ischemia/metabolism
3.
Med Res Arch ; 11(1)2023 Jan.
Article in English | MEDLINE | ID: mdl-36777192

ABSTRACT

Despite displaying efficacy in experimental stroke studies, neuroprotection has failed in clinical trials. The translational difficulties include a limited methodological agreement between preclinical and clinical studies and the heterogeneity of stroke in humans compared to standardized strokes in animal models. Promising neuroprotective approaches based on a deeper understanding of the complex pathophysiology of ischemic stroke, such as blocking pro-inflammatory pathways plus pro-survival mediators, are now evaluated in preclinical studies. Combinatorial therapy has become increasingly attractive in recent years as recognizing the complexity of stroke progression becomes evident. The paper aimed to test the hypothesis that blocking pro-inflammatory platelet-activating factor receptor (PAF-R) with LAU-0901 plus administering a selected docosanoid, aspirin-triggered neuroprotectin D1 (AT-NPD1), which activates cell-survival pathways after middle cerebral artery occlusion (MCAo), would lead to neurological recovery. We have demonstrated that LAU-0901 plus AT-NPD1 treatment affords high-grade neuroprotection in MCAo, equaling or exceeding that afforded by LAU-0901 or AT-NPD1 alone at considerably moderate doses, and it has a broad therapeutic window extending to 6 hours after stroke onset.

4.
Cell Mol Neurobiol ; 43(3): 1077-1096, 2023 Apr.
Article in English | MEDLINE | ID: mdl-35622188

ABSTRACT

Wnt5a triggers inflammatory responses and damage via NFkB/p65 in retinal pigment epithelial (RPE) cells undergoing uncompensated oxidative stress (UOS) and in experimental ischemic stroke. We found that Wnt5a-Clathrin-mediated uptake leads to NFkB/p65 activation and that Wnt5a is secreted in an exosome-independent fashion. We uncovered that docosahexaenoic acid (DHA) and its derivative, Neuroprotectin D1 (NPD1), upregulate c-Rel expression that, as a result, blunts Wnt5a abundance by competing with NFkB/p65 on the Wnt5a promoter A. Wnt5a increases in ischemic stroke penumbra and blood, while DHA reduces Wnt5a abundance with concomitant neuroprotection. Peptide inhibitor of Wnt5a binding, Box5, is also neuroprotective. DHA-decreased Wnt5a expression is concurrent with a drop in NFkB-driven inflammatory cytokine expression, revealing mechanisms after stroke, as in RPE cells exposed to UOS. Limiting the Wnt5a activity via Box5 reduces stroke size, suggesting neuroprotection pertinent to onset and progression of retinal degenerations and stroke consequences. NPD1 disrupts Wnt5a feedback loop at two sites: (1) decreasing FZD5, thus Wnt5a internalization, and (2) by enhancing cREL activity, which competes with p65/NFkB downstream endocytosis. As a result, Wnt5a expression is reduced, and so is its inflammatory signaling in RPE cells and neurons in ischemic stroke.


Subject(s)
Ischemic Stroke , Stroke , Humans , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/therapeutic use , Neuroprotection , Stroke/drug therapy , Stroke/metabolism , Wnt-5a Protein , Frizzled Receptors/metabolism
6.
J Stroke Cerebrovasc Dis ; 31(8): 106585, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35717719

ABSTRACT

OBJECTIVE: We tested the hypothesis that blocking pro-inflammatory platelet-activating factor receptor (PAFR) with LAU-0901 (LAU) plus administering a selected docosanoid, aspirin-triggered neuroprotectin D1 (AT-NPD1), which activates cell-survival pathways after middle cerebral artery occlusion (MCAo), would lead to neurological recovery. Dose-response and therapeutic window were investigated. MATERIALS AND METHODS: Male SD rats were subjected to 2 hours of MCAo. Behavior testing (days 1-7) and ex vivo MRI on day 7 were conducted. In dose-response, rats were treated with LAU (45 and 60 mg/kg; IP), AT-NPD1 (111, 222, 333 µg/kg; IV), LAU+AT-NPD1 (LAU at 3 hours and AT-NPD1 at 3.15 hours) or vehicle. In the therapeutic window, vehicle, LAU (60 mg/kg), AT-NPD1 (222 µg/kg), and LAU+AT-NPD1 were administered at 3, 4, 5, and 6 hours after onset of MCAo. RESULTS: LAU and AT-NPD1 treatments alone improved behavior by 40-42% and 20-30%, respectively, and LAU+AT-NPD1 by 40% compared to the vehicle group. T2-weighted imaging (T2WI) volumes were reduced with all doses of LAU and AT-NPD1 by 73-90% and 67-83% and LAU+AT-NPD1 by 94% compared to vehicle. In the therapeutic window, LAU+AT-NPD1, when administered at 3, 4, 5, and 6 hours, improved behavior by 50, 56, 33, and 26% and reduced T2WI volumes by 93, 90, 82, and 84% compared to vehicle. CONCLUSIONS: We have shown here for the first time that LAU plus AT-NPD1 treatment affords high-grade neuroprotection in MCAo, equaling or exceeding that afforded by LAU or AT-NPD1 alone at considerably moderate doses. It has a broad therapeutic window extending to 6 hours after stroke onset.


Subject(s)
Ischemic Stroke , Neuroprotective Agents , Stroke , Animals , Aspirin/therapeutic use , Infarction, Middle Cerebral Artery/diagnostic imaging , Infarction, Middle Cerebral Artery/drug therapy , Male , Neuroprotection , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Rats , Rats, Sprague-Dawley , Stroke/diagnostic imaging , Stroke/drug therapy , Stroke/etiology
7.
Nutr Rev ; 80(5): 1001-1012, 2022 04 08.
Article in English | MEDLINE | ID: mdl-34406390

ABSTRACT

Apolipoprotein E plays a crucial role in cholesterol metabolism. The immunomodulatory functions of the human polymorphic APOE gene have gained particular interest because APOE4, a well-recognized risk factor for late-onset Alzheimer's disease, has also been recently linked to increased risk of COVID-19 infection severity in a large UK biobank study. Although much is known about apoE functions in the nervous system, much less is known about APOE polymorphism effects on malnutrition and enteric infections and the consequences for later development in underprivileged environments. In this review, recent findings are summarized of apoE's effects on intestinal function in health and disease and the role of APOE4 in protecting against infection and malnutrition in children living in unfavorable settings, where poor sanitation and hygiene prevail, is highlighted. The potential impact of APOE4 on later development also is discussed and gaps in knowledge are identified that need to be addressed to protect children's development under adverse environments.


Subject(s)
Apolipoprotein E4 , Chronic Disease , Malnutrition , Alzheimer Disease , Apolipoprotein E4/genetics , Apolipoprotein E4/metabolism , Apolipoproteins E/genetics , Child , Humans , Malnutrition/complications
8.
J Pharmacol Exp Ther ; 380(1): 47-53, 2022 01.
Article in English | MEDLINE | ID: mdl-34728560

ABSTRACT

Ruthenium compounds, nitric oxide donors in biologic systems, have emerged as a promising therapeutic alternative to conventional drugs in anticancer chemotherapy and as a potential neuroprotective agent with fewer cytotoxic effects. This minireview summarizes promising studies with ruthenium complexes and their roles in cancer, neuroinflammation, neurovascular, and neurodegenerative diseases. The up-to-date evidence supports that ruthenium-based compounds have beneficial effects against gliomas and other types of brain cancers, reduce motor symptoms in models of cerebral ischemia-reperfusion, and may act in the control of nociceptive and inflammatory events, such as those seen in early Alzheimer's disease. More studies are needed to fill many current knowledge gaps about the intricate and complex biologic effects and therapeutic-related mechanisms of ruthenium, stimulating further research. SIGNIFICANCE STATEMENT: This minireview summarizes studies addressing the role of ruthenium compounds on neurological illnesses, focusing on brain cancer and neurovascular and neurodegenerative diseases. No such review is available in the literature.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/therapeutic use , Nitric Oxide Donors/therapeutic use , Ruthenium Compounds/therapeutic use , Animals , Brain Neoplasms/metabolism , Humans , Neurodegenerative Diseases/metabolism
9.
Front Pharmacol ; 12: 746470, 2021.
Article in English | MEDLINE | ID: mdl-34630114

ABSTRACT

Glioblastoma multiforme (GBM) is an aggressive, highly proliferative, invasive brain tumor with a poor prognosis and low survival rate. The current standard of care for GBM is chemotherapy combined with radiation following surgical intervention, altogether with limited efficacy, since survival averages 18 months. Improvement in treatment outcomes for patients with GBM requires a multifaceted approach due to the dysregulation of numerous signaling pathways. Recently emerging therapies to precisely modulate tumor angiogenesis, inflammation, and oxidative stress are gaining attention as potential options to combat GBM. Using a mouse model of GBM, this study aims to investigate Avastin (suppressor of vascular endothelial growth factor and anti-angiogenetic treatment), LAU-0901 (a platelet-activating factor receptor antagonist that blocks pro-inflammatory signaling), Elovanoid; ELV, a novel pro-homeostatic lipid mediator that protects neural cell integrity and their combination as an alternative treatment for GBM. Female athymic nude mice were anesthetized with ketamine/xylazine, and luciferase-modified U87MG tumor cells were stereotactically injected into the right striatum. On post-implantation day 13, mice received one of the following: LAU-0901, ELV, Avastin, and all three compounds in combination. Bioluminescent imaging (BLI) was performed on days 13, 20, and 30 post-implantation. Mice were perfused for ex vivo MRI on day 30. Bioluminescent intracranial tumor growth percentage was reduced by treatments with LAU-0901 (43%), Avastin (77%), or ELV (86%), individually, by day 30 compared to saline treatment. In combination, LAU-0901/Avastin, ELV/LAU-0901, or ELV/Avastin had a synergistic effect in decreasing tumor growth by 72, 92, and 96%, respectively. Additionally, tumor reduction was confirmed by MRI on day 30, which shows a decrease in tumor volume by treatments with LAU-0901 (37%), Avastin (67%), or ELV (81.5%), individually, by day 30 compared to saline treatment. In combination, LAU-0901/Avastin, ELV/LAU-0901, or ELV/Avastin had a synergistic effect in decreasing tumor growth by 69, 78.7, and 88.6%, respectively. We concluded that LAU-0901 and ELV combined with Avastin exert a better inhibitive effect in GBM progression than monotherapy. To our knowledge, this is the first study that demonstrates the efficacy of these novel therapeutic regimens in a model of GBM and may provide the basis for future therapeutics in GBM patients.

10.
Cancer Metastasis Rev ; 40(3): 643-647, 2021 09.
Article in English | MEDLINE | ID: mdl-34519960

ABSTRACT

Glioblastoma multiforme (GBM) is the most invasive type of glial tumor with poor overall survival, despite advances in surgical resection, chemotherapy, and radiation. One of the main challenges in treating GBM is related to the tumor's location, complex and heterogeneous biology, and high invasiveness. To meet the demand for oxygen and nutrients, growing tumors induce new blood vessels growth. Antibodies directed against vascular endothelial growth factor (VEGF), which promotes angiogenesis, have been developed to limit tumor growth. Bevacizumab (Avastin), an anti-VEGF monoclonal antibody, is the first approved angiogenesis inhibitor with therapeutic promise. However, it has limited efficacy, likely due to adaptive mutations in GBM, leading to overall survival compared to the standard of care in GBM patients. Molecular connections between angiogenesis, inflammation, oxidative stress pathways, and the development of gliomas have been recognized. Improvement in treatment outcomes for patients with GBM requires a multifaceted approach due to the converging dysregulation of signaling pathways. While most GBM clinical trials focus on "anti-angiogenic" modalities, stimulating inflammation resolution is a novel host-centric therapeutic avenue. The selective therapeutic possibilities for targeting the tumor microenvironment, specifically angiogenic and inflammatory pathways expand. So, a combination of agents aiming to interfere with several mechanisms might be beneficial to improve outcomes. Our approach might also be combined with other therapies to enhance sustained effectiveness. Here, we discuss Suramab (anti-angiogenic), LAU-0901 (a platelet-activating factor receptor antagonist), Elovanoid (ELV; a novel lipid mediator), and their combination as potential alternatives to contain GBM growth and invasiveness.


Subject(s)
Brain Neoplasms , Glioblastoma , Angiogenesis Inhibitors/therapeutic use , Bevacizumab/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Homeostasis , Humans , Neovascularization, Pathologic/drug therapy , Tumor Microenvironment , Vascular Endothelial Growth Factor A/therapeutic use
11.
CNS Neurosci Ther ; 26(11): 1155-1167, 2020 11.
Article in English | MEDLINE | ID: mdl-32757264

ABSTRACT

AIMS: Mesencephalic astrocyte-derived neurotrophic factor (MANF) is a secretory neurotrophic factor protein that promotes repair after neuronal injury. The microglia cell surface receptor (triggering receptor expressed on myeloid cells-2; TREM2) regulates the production of pro- and antiinflammatory mediators after stroke. Here, we study MANF and TREM2 expression after middle cerebral artery occlusion (MCAo) and explore if docosahexaenoic acid (DHA) treatment exerts a potentiating effect. METHODS: We used 2 hours of the MCAo model in rats and intravenously administered DHA or vehicle at 3 hours after the onset of MCAo. Neurobehavioral assessment was performed on days 1, 3, 7, and 14; MANF and TREM2 expression was measured by immunohistochemistry and Western blotting. RESULTS: MANF was upregulated in neurons and astrocytes on days 1, 7, and 14, and TREM2 was expressed on macrophages in the ischemic penumbra and dentate gyrus (DG) on days 7 and 14. DHA improved neurobehavioral recovery, attenuated infarct size on days 7 and 14, increased MANF and decreased TREM2 expression in ischemic core, penumbra, DG, and enhanced neurogenesis on Day 14. CONCLUSION: MANF and TREM2 protein abundance is robustly increased after MCAo, and DHA treatment potentiated MANF abundance, decreased TREM2 expression, improved neurobehavioral recovery, reduced infarction, and provided enhanced neuroprotection.


Subject(s)
Brain Ischemia/metabolism , Docosahexaenoic Acids/administration & dosage , Ischemic Stroke/metabolism , Membrane Glycoproteins/biosynthesis , Nerve Growth Factors/biosynthesis , Neurogenesis/drug effects , Receptors, Immunologic/biosynthesis , Administration, Intravenous , Animals , Brain Ischemia/drug therapy , Cerebral Infarction/drug therapy , Cerebral Infarction/metabolism , Ischemic Stroke/drug therapy , Male , Membrane Glycoproteins/antagonists & inhibitors , Nerve Growth Factors/agonists , Neurogenesis/physiology , Rats , Rats, Sprague-Dawley , Receptors, Immunologic/antagonists & inhibitors
12.
Brain Circ ; 6(4): 260-268, 2020.
Article in English | MEDLINE | ID: mdl-33506149

ABSTRACT

OBJECTIVE: Acute ischemic stroke triggers complex neurovascular, neuroinflammatory, and synaptic alterations. This study explores whether blocking pro-inflammatory platelet-activating factor receptor (PAF-R) plus selected docosanoids after middle cerebral artery occlusion (MCAo) would lead to neurological recovery. The following small molecules were investigated: (a) LAU-0901, a PAF-R antagonist that blocks pro-inflammatory signaling; and (b) derivatives of docosahexaenoic acid (DHA), neuroprotectin D1 (NPD1), and aspirin-triggered NPD1 (AT-NPD1), which activates cell survival pathways and are exert potent anti-inflammatory activity in the brain. MATERIALS AND METHODS: Sprague-Dawley rats received 2 h MCAo and LAU-0901 (30 or 60 mg/kg, 2 h after stroke), NPD1, and AT-NPD1 (333 µg/kg), DHA (5 mg/kg), and their combination were administered intravenous at 3 h after stroke. Behavior testing and ex vivo magnetic resonance imaging were conducted on day 3 or 14 to assess lesion characteristics and lipidomic analysis on day 1. Series 1 (LAU-0901 + NPD1, 14d), Series 2 (LAU-0901 + AT-NPD1, 3d), and Series 3 (LAU-0901 + DHA, 1d). RESULTS: All combinatory groups improved behavior compared to NPD1, AT-NPD1, or DHA treatments alone. Total lesion volumes were reduced with LAU-0901 + NPD1 by 62% and LAU-0901 + AT-NPD1 by 90% treatments versus vehicle groups. LAU-0901 and LAU-0901 + DHA increased the production of vasoactive lipid mediators (prostaglandins: PGE2, PGF2- α, 6-keto-PGF1- α, and PGD2) as well an inflammatory regulating mediator hydroxyoctadecadienoic acid. In contrast, LAU-0901 and LAU-0901 + DHA decreased the production of 12-hydroxyeicosatetraenoic acid, a pro-inflammatory mediator. CONCLUSION: Combination therapy with LAU-0901 and selected docosanoids is more effective than the single therapy, affording synergistic neuroprotection, with restored pro-homeostatic lipid mediators and improved neurological recovery. Altogether, our findings support the combinatory therapy as the basis for future therapeutics for ischemic stroke.

13.
Mol Neurobiol ; 55(8): 7090-7106, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29858774

ABSTRACT

Docosahexaenoic acid (DHA) and neuroprotectin D1 (NPD1) are neuroprotective after experimental ischemic stroke. To explore underlying mechanisms, SD rats underwent 2 h of middle cerebral artery occlusion (MCAo) and treated with DHA (5 mg/kg, IV) or NPD1 (5 µg/per rat, ICV) and vehicles 1 h after. Neuro-behavioral assessments was conducted on days 1, 2, and 3, and on week 1, 2, 3, or 4. BrdU was injected on days 4, 5, and 6, immunohistochemistry was performed on week 2 or 4, MRI on day 7, and lipidomic analysis at 4 and 5 h after onset of stroke. DHA improved short- and long-term behavioral functions and reduced cortical, subcortical, and total infarct volumes (by 42, 47, and 31%, respectively) after 2 weeks and reduced tissue loss by 50% after 4 weeks. DHA increased the number of BrdU+/Ki-67+, BrdU+/DCX+, and BrdU+/NeuN+ cells in the cortex, subventricular zone, and dentate gyrus and potentiated NPD1 synthesis in the penumbra at 5 h after MCAo. NPD1 improved behavior, reduced lesion volumes, protected ischemic penumbra, increased NeuN, GFAP, SMI-71-positive cells and vessels, axonal regeneration in the penumbra, and attenuated blood-brain barrier (BBB) after MCAo. We conclude that docosanoid administration increases neurogenesis and angiogenesis, activates NPD1 synthesis in the penumbra, and diminishes BBB permeability, which correlates to long-term neurobehavioral recovery after experimental ischemic stroke.


Subject(s)
Behavior, Animal , Blood-Brain Barrier/pathology , Brain Ischemia/pathology , Fatty Acids/pharmacology , Neovascularization, Physiologic , Neurogenesis , Stroke/pathology , Animals , Axons/pathology , Brain Ischemia/complications , Docosahexaenoic Acids/metabolism , Doublecortin Protein , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/pathology , Male , Neovascularization, Physiologic/drug effects , Nerve Regeneration/drug effects , Neurogenesis/drug effects , Permeability , Rats, Sprague-Dawley , Stroke/complications , Survival Analysis
14.
Sci Adv ; 3(9): e1700735, 2017 09.
Article in English | MEDLINE | ID: mdl-28959727

ABSTRACT

We report the characterization of a novel class of lipid mediators termed elovanoids (ELVs) (ELV-N32 and ELV-N34), which are dihydroxylated derivatives of 32:6n3 and 34:6n3, respectively. The precursors of ELVs are made by elongation of a 22:6n3 fatty acid and catalyzed by ELOVL4 (elongation of very-long-chain fatty acids-4). The structure and stereochemistry of ELVs were established using synthetic compounds produced by stereocontrolled total synthesis. We report that ELV-mediated protection is induced in neuronal cultures undergoing either oxygen/glucose deprivation or N-methyl-d-aspartate receptor-mediated excitotoxicity, as well as in experimental ischemic stroke. The methyl ester or sodium salt of ELV-N32 and ELV-N34 resulted in reduced infarct volumes, promoted cell survival, and diminished neurovascular unit disruption when administered 1 hour following 2 hours of ischemia by middle cerebral artery occlusion. Together, our data reveal a novel prohomeostatic and neuroprotective lipid-signaling mechanism aiming to sustain neural cell integrity.


Subject(s)
Homeostasis/drug effects , Neurons/drug effects , Neurons/physiology , Neuroprotective Agents/pharmacology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Biomarkers , Blood-Brain Barrier/metabolism , Cell Survival/drug effects , Cells, Cultured , Female , Magnetic Resonance Imaging , Molecular Structure , Neuroprotective Agents/chemistry , Pregnancy , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Rats , Receptors, N-Methyl-D-Aspartate/metabolism , Stereoisomerism , Stroke/drug therapy , Stroke/etiology , Stroke/metabolism , Stroke/physiopathology
15.
Cell Death Differ ; 24(6): 1091-1099, 2017 06.
Article in English | MEDLINE | ID: mdl-28430183

ABSTRACT

Ring finger protein 146 (Iduna) facilitates DNA repair and protects against cell death induced by NMDA receptor-mediated glutamate excitotoxicity or by cerebral ischemia. Neuroprotectin D1 (NPD1), a docosahexaenoic acid (DHA)-derived lipid mediator, promotes cell survival under uncompensated oxidative stress (UOS). Our data demonstrate that NPD1 potently upregulates Iduna expression and provides remarkable cell protection against UOS. Iduna, which was increased by the lipid mediator, requires the presence of the poly(ADP-ribose) (PAR) sites. Moreover, astrocytes and neurons in the penumbra display an enhanced abundance of Iduna, followed by remarkable neurological protection when DHA, a precursor of NPD1, is systemically administered 1 h after 2 h of ischemic stroke. These findings provide a conceptual advancement for survival of neural cells undergoing challenges to homeostasis because a lipid mediator, made 'on demand,' modulates the abundance of a critically important protein for cell survival.


Subject(s)
Brain Ischemia/metabolism , Docosahexaenoic Acids/metabolism , Oxidative Stress , Signal Transduction , Stroke/metabolism , Ubiquitin-Protein Ligases/metabolism , Brain Ischemia/physiopathology , Cell Line , Gene Expression Regulation , Humans , Neurons/metabolism , Neurons/physiology , Stroke/physiopathology , Ubiquitin-Protein Ligases/genetics , Up-Regulation
17.
Sci Rep ; 6: 30298, 2016 07 22.
Article in English | MEDLINE | ID: mdl-27444269

ABSTRACT

Temporal lobe epilepsy or limbic epilepsy lacks effective therapies due to a void in understanding the cellular and molecular mechanisms that set in motion aberrant neuronal network formations during the course of limbic epileptogenesis (LE). Here we show in in vivo rodent models of LE that the phospholipid mediator platelet-activating factor (PAF) increases in LE and that PAF receptor (PAF-r) ablation mitigates its progression. Synthetic PAF-r antagonists, when administered intraperitoneally in LE, re-establish hippocampal dendritic spine density and prevent formation of dysmorphic dendritic spines. Concomitantly, hippocampal interictal spikes, aberrant oscillations, and neuronal hyper-excitability, evaluated 15-16 weeks after LE using multi-array silicon probe electrodes implanted in the dorsal hippocampus, are reduced in PAF-r antagonist-treated mice. We suggest that over-activation of PAF-r signaling induces aberrant neuronal plasticity in LE and leads to chronic dysfunctional neuronal circuitry that mediates epilepsy.


Subject(s)
Epilepsy, Temporal Lobe/drug therapy , Epilepsy, Temporal Lobe/metabolism , Platelet Activating Factor/metabolism , Platelet Membrane Glycoproteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Blood Platelets/metabolism , Dendritic Spines/metabolism , Dendritic Spines/pathology , Disease Models, Animal , Epilepsy, Temporal Lobe/genetics , Epilepsy, Temporal Lobe/pathology , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Humans , Limbic Lobe/metabolism , Limbic Lobe/pathology , Mice , Neuronal Plasticity/drug effects , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Platelet Activating Factor/genetics , Platelet Membrane Glycoproteins/antagonists & inhibitors , Platelet Membrane Glycoproteins/genetics , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics
18.
Exp Transl Stroke Med ; 7(1): 3, 2015.
Article in English | MEDLINE | ID: mdl-25642315

ABSTRACT

BACKGROUND: Ischemic brain injury disrupts the blood-brain barrier (BBB) and then triggers a cascade of events, leading to edema formation, secondary brain injury and poor neurological outcomes. Recently, we have shown that docosahexaenoic acid (DHA) improves functional and histological outcomes following experimental stroke. However, little is known about the effect of DHA on BBB dysfunction after cerebral ischemia-reperfusion injury. The present study was designed to determine whether DHA protects against BBB disruption after focal cerebral ischemia in rats. METHODS: Physiologically-controlled SD rats received 2 h middle cerebral artery occlusion (MCAo). DHA (5 mg/kg) or vehicle (saline) was administered I.V. at 3 h after onset of MCAo. Fluorometric quantitation of Evans Blue dye (EB) was performed in eight brain regions at 6 h, 24 h or 72 h after MCAo. Fluorescein isothiocynate (FITC) - dextran leakage and histopathology was evaluated on day 3 after stroke. RESULTS: Physiological variables were stable and showed no significant differences between groups. DHA improved neurological deficits at 24 h, 48 h and 72 h and decreased EB extravasation in the ischemic hemisphere at 6 h (by 30%), 24 h (by 48%) and 72 h (by 38%). In addition, EB extravasation was decreased by DHA in the cortex and total hemisphere as well. FITC-dextran leakage was reduced by DHA treatment on day 3 by 68% compared to the saline group. DHA treatment attenuated cortical (by 50%) and total infarct volume (by 38%) compared to vehicle-treated rats on day 3 after stroke. CONCLUSIONS: DHA therapy diminishes BBB damage accompanied with the acceleration of behavioral recovery and attenuation of the infarct volume. It is reasonable to propose that DHA has the potential for treating focal ischemic stroke in the clinical setting.

19.
J Neurotrauma ; 32(14): 1101-8, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25669448

ABSTRACT

Recent studies show that myosin light chain kinase (MLCK) plays a pivotal role in development of cerebral edema, a known complication following traumatic brain injury (TBI) in children and a contributing factor to worsened neurologic recovery. Interferon-stimulated gene 15 (ISG15) is upregulated after cerebral ischemia and is neuroprotective. The significant role of ISG15 after TBI has not been studied. Postnatal Day (PND) 21 and PND24 mice were subjected to lateral closed-skull injury with impact depth of 2.0 or 2.25 mm. Behavior was examined at 7 d using two-object novel recognition and Wire Hang tests. Mice were sacrificed at 6 h, 12 h, 24 h, 48 h, 72 h, and 7 d. ISG15 and MLCK were analyzed by Western blot and immunohistochemistry, blood-brain barrier (BBB) disruption with Evans Blue (EB), and cerebral edema with wet/dry weights. EB extravasation and edema peaked at 72 h in both ages. PND21 mice had more severe neurological deficits, compared with PND24 mice. PND24 mice showed peak ISG15 expression at 6 h, and PND21 mice at 72 h. MLCK peaked in both age groups at 12 h and co-localized with ISG15 on immunohistochemistry and co-immunoprecipitation. These studies provide evidence, ISG15 is elevated following TBI in mice, preceding MLCK elevation, development of BBB disruption, and cerebral edema.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Edema/metabolism , Brain Injuries/metabolism , Cytokines/metabolism , Animals , Blood-Brain Barrier/pathology , Brain Edema/genetics , Brain Edema/pathology , Brain Injuries/genetics , Brain Injuries/pathology , Cytokines/genetics , Mice , Myosin-Light-Chain Kinase/metabolism , Ubiquitins/genetics , Ubiquitins/metabolism , Up-Regulation
20.
J Neurol Sci ; 338(1-2): 135-41, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24433927

ABSTRACT

Recently we demonstrated that docosahexaenoic acid (DHA) is highly neuroprotective when animals were allowed to survive during one week. This study was conducted to establish whether the neuroprotection induced by DHA persists with chronic survival. Sprague-Dawley rats underwent 2h of middle cerebral artery occlusion (MCAo) and treated with DHA or saline at 3h after MCAo. Animals received neurobehavioral examination (composite neuroscore, rota-rod, beam walking and Y maze tests) followed by ex vivo magnetic resonance imaging and histopathology at 3 weeks. DHA improved composite neurologic score beginning on day 1 by 20%, which persisted throughout weeks 1-3 by 24-41% compared to the saline-treated group. DHA prolonged the latency in rota-rod on weeks 2-3 by 162-178%, enhanced balance performance in the beam walking test on weeks 1 and 2 by 42-51%, and decreased the number of entries in the Y maze test by 51% and spontaneous alteration by 53% on week 2 compared to the saline-treated group. DHA treatment reduced tissue loss (computed from T2-weighted images) by 24% and total and cortical infarct volumes by 46% and 54% compared to the saline-treated group. These results show that DHA confers enduring ischemic neuroprotection.


Subject(s)
Docosahexaenoic Acids/therapeutic use , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/physiopathology , Neuroprotective Agents/therapeutic use , Analysis of Variance , Animals , Disease Models, Animal , Infarction, Middle Cerebral Artery/pathology , Magnetic Resonance Imaging , Male , Maze Learning/drug effects , Motor Activity/drug effects , Neurologic Examination , Psychomotor Performance/drug effects , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL