Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
CNS Neurol Disord Drug Targets ; 16(3): 326-338, 2017.
Article in English | MEDLINE | ID: mdl-28042770

ABSTRACT

Central nervous system diseases are major health issues and are often associated with disability or death. Most central nervous system disorders are characterized by high levels of oxidative stress. Nuclear factor erythroid 2 related factor (Nrf2) is known for its ability to regulate the expression of a series of enzymes with antioxidative, prosurvival, and detoxification effects. Under basal conditions, Nrf2 forms a complex with Kelch-like ECH associated protein 1, leading to Nrf2 inactivation via ubiquitination and degradation. However, following exposure of Keap1 to oxidative stress, Nrf2 is released from Keap1, activated, and translocated into the nucleus. Upon nuclear entry, Nrf2 binds to antioxidant response elements (ARE), thereby inducing the expression of genes such as glutathione s-transferase, heme oxygenase 1, and NADPH quinine oxidoreductase 1. Many dietary phytochemicals have been reported to activate the protective Nrf2/ARE pathway. Here, we review the preventive and protective effects of dietary Nrf2 activators against CNS diseases, including stroke, traumatic brain injury, Alzheimer's disease, and Parkinson's disease.


Subject(s)
Antioxidants/administration & dosage , Central Nervous System Diseases/prevention & control , Central Nervous System Diseases/therapy , Dietary Supplements , NF-E2-Related Factor 2/metabolism , Animals , Humans
2.
Neurobiol Dis ; 91: 37-46, 2016 07.
Article in English | MEDLINE | ID: mdl-26921472

ABSTRACT

Omega-3 polyunsaturated fatty acids (n-3 PUFAs) have been shown to protect the neonatal brain against hypoxic/ischemic (H/I) injury. However, the mechanism of n-3 PUFA-afforded neuroprotection is not well understood. One major determinant of H/I vulnerability is the permeability of the blood-brain barrier (BBB). Therefore, we examined the effects of n-3 PUFAs on BBB integrity after neonatal H/I. Female rats were fed a diet with or without n-3 PUFA enrichment from day 2 of pregnancy to 14days after parturition. H/I was introduced in 7day-old offspring. We observed relatively rapid BBB penetration of the small molecule cadaverine (640Da) at 4h post-H/I and a delayed penetration of larger dextrans (3kD-40kD) 24-48h after injury. Surprisingly, the neonatal BBB was impermeable to Evans Blue or 70kD dextran leakage for up to 48h post-H/I, despite evidence of IgG extravasation at this time. As expected, n-3 PUFAs ameliorated H/I-induced BBB damage, as shown by reductions in tracer efflux and IgG extravasation, preservation of BBB ultrastructure, and enhanced tight junction protein expression. Furthermore, n-3 PUFAs prevented the elevation in matrix metalloproteinase (MMP) activity in the brain and blood after H/I. Thus, n-3 PUFAs may protect neonates against BBB damage by blunting MMPs activation after H/I.


Subject(s)
Blood-Brain Barrier/drug effects , Brain/drug effects , Fatty Acids, Omega-3/pharmacology , Hypoxia-Ischemia, Brain/metabolism , Animals , Animals, Newborn , Blood-Brain Barrier/metabolism , Brain/metabolism , Disease Models, Animal , Fatty Acids, Omega-3/metabolism , Female , Hypoxia-Ischemia, Brain/drug therapy , Rats, Sprague-Dawley
3.
Stroke ; 46(10): 2943-50, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26374481

ABSTRACT

BACKGROUND AND PURPOSE: Omega-3 polyunsaturated fatty acids (n-3 PUFAs) attenuate neonatal hypoxic/ischemic (H/I) brain damage, but the underlying mechanisms are not fully understood. This study tested the hypothesis that n-3 PUFAs enhance Akt-dependent prosurvival signaling by promoting the biosynthesis of phosphatidylserine in neuronal cell membranes. METHODS: Dietary n-3 PUFA supplementation was initiated on the second day of pregnancy in dams. H/I was induced in 7-day-old rat pups by ipsilateral common carotid artery occlusion followed by hypoxia (8% oxygen for 2.5 hours). Neurological outcomes, brain tissue loss, cell death, and the activation of signaling events were assessed after H/I. The effects of n-3 PUFAs (docosahexaenoic acid and eicosapentaenoic acid) on oxygen-glucose deprivation-induced cell death and the underlying mechanism of protection were also examined in primary cortical neuron cultures. RESULTS: n-3 PUFAs reduced brain tissue loss at 7 days after H/I and improved neurological outcomes, whereas inhibition of PI3K/Akt signaling by LY294002 partially abrogated this neuroprotective effect. Docosahexaenoic acid/eicosapentaenoic acid also prevented ischemic neuronal death through the Akt prosurvival pathway in vitro. Furthermore, docosahexaenoic acid/eicosapentaenoic acid increased the production of phosphatidylserine, the major membrane-bound phospholipids, after ischemia both in vitro and in vivo. A reduction in membrane phosphatidylserine by shRNA-mediated knockdown of phosphatidylserine synthetase-1 attenuated Akt activation and neuronal survival after docosahexaenoic acid/eicosapentaenoic acid treatment in the oxygen-glucose deprivation model. CONCLUSIONS: n-3 PUFAs robustly protect against H/I-induced brain damage in neonates by activating Akt prosurvival pathway in compromised neurons. In addition, n-3 PUFAs promote the formation of membrane phosphatidylserine, thereby promoting Akt activity and improving cellular survival.


Subject(s)
Brain/drug effects , Fatty Acids, Omega-3/pharmacology , Hypoxia-Ischemia, Brain/metabolism , Neurons/drug effects , Phosphatidylserines/biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , Animals , Animals, Newborn , Brain/pathology , CDPdiacylglycerol-Serine O-Phosphatidyltransferase/genetics , Cell Death/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Gene Knockdown Techniques , Hypoxia-Ischemia, Brain/pathology , In Vitro Techniques , Neurons/metabolism , Phosphoinositide-3 Kinase Inhibitors , RNA, Small Interfering , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
4.
Exp Neurol ; 272: 170-80, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25771800

ABSTRACT

Stroke is a devastating neurological disease with no satisfactory therapies to preserve long-term neurological function, perhaps due to the sole emphasis on neuronal survival in most preclinical studies. Recent studies have revealed the importance of protecting multiple cell types in the injured brain, such as oligodendrocytes and components of the neurovascular unit, before long-lasting recovery of function can be achieved. For example, revascularization in the ischemic penumbra is critical to provide various neurotrophic factors that enhance the survival and activity of neurons and other progenitor cells, such as oligodendrocyte precursor cells. In the present study, we hypothesized that chronic dietary supplementation with fish oil promotes post-stroke angiogenesis, neurogenesis, and oligodendrogenesis, thereby leading to long-term functional improvements. Mice received dietary supplementation with n-3 PUFA-enriched fish oil for three months before and up to one month after stroke. As expected, dietary n-3 PUFAs significantly increased levels of n-3 PUFAs in the brain and improved long-term behavioral outcomes after cerebral ischemia. n-3 PUFAs also robustly improved revascularization and angiogenesis and boosted the survival of NeuN/BrdU labeled newborn neurons up to 35days after stroke injury. Furthermore, these pro-neurogenic effects were accompanied by robust oligodendrogenesis. Thus, this is the first study to demonstrate that chronic dietary intake of n-3 PUFAs is an effective prophylactic measure not only to protect against ischemic injury for the long term but also to actively promote neurovascular restorative dynamics and brain repair.


Subject(s)
Cerebrovascular Circulation/physiology , Dietary Supplements , Fatty Acids, Omega-3/therapeutic use , Infarction, Middle Cerebral Artery/complications , Nervous System Diseases/diet therapy , Nervous System Diseases/etiology , Analysis of Variance , Animals , Bromodeoxyuridine/metabolism , Cell Proliferation , Disease Models, Animal , Doublecortin Domain Proteins , Fatty Acids/metabolism , Male , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Motor Activity/physiology , Neurogenesis/physiology , Neuropeptides/metabolism , Phosphopyruvate Hydratase/metabolism , Psychomotor Performance/physiology , Time Factors
5.
Sci Rep ; 4: 7458, 2014 Dec 12.
Article in English | MEDLINE | ID: mdl-25500548

ABSTRACT

Microglia represent rational but challenging targets for improving white matter integrity because of their dualistic protective and toxic roles. The present study examines the effect of Omega-3 polyunsaturated fatty acids (n-3 PUFAs) on microglial responses to myelin pathology in primary cultures and in the cuprizone mouse model of multiple sclerosis (MS), a devastating demyelination disease. Docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), the two main forms of n-3 PUFAs in the brain, inhibited the release of nitric oxide and tumor necrosis factor-α from primary microglia upon IFN-γ and myelin stimulation. DHA and EPA also enhanced myelin phagocytosis in vitro. Therefore, n-3 PUFAs can inhibit inflammation while at the same time enhancing beneficial immune responses such as microglial phagocytosis. In vivo studies demonstrated that n-3 PUFA supplementation reduced cuprizone-induced demyelination and improved motor and cognitive function. The positive effects of n-3 PUFAs were accompanied by a shift in microglial polarization toward the beneficial M2 phenotype both in vitro and in vivo. These results suggest that n-3 PUFAs may be clinically useful as immunomodulatory agents for demyelinating diseases through a novel mechanism involving microglial phenotype switching.


Subject(s)
Docosahexaenoic Acids/administration & dosage , Eicosapentaenoic Acid/administration & dosage , Microglia/physiology , Multiple Sclerosis/drug therapy , Neuroprotective Agents/administration & dosage , Administration, Oral , Animals , Cells, Cultured , Dietary Supplements , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Male , Mice, Inbred C57BL , Microglia/drug effects , Multiple Sclerosis/pathology , Neuroprotective Agents/pharmacology , Primary Cell Culture
6.
Neurobiol Dis ; 68: 91-103, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24794156

ABSTRACT

Stroke is a devastating neurological disorder and one of the leading causes of death and serious disability. After cerebral ischemia, revascularization in the ischemic boundary zone provides nutritive blood flow as well as various growth factors to promote the survival and activity of neurons and neural progenitor cells. Enhancement of angiogenesis and the resulting improvement of cerebral microcirculation are key restorative mechanisms and represent an important therapeutic strategy for ischemic stroke. In the present study, we tested the hypothesis that post-stroke angiogenesis would be enhanced by omega-3 polyunsaturated fatty acids (n-3 PUFAs), a major component of dietary fish oil. To this end, we found that transgenic fat-1 mice that overproduce n-3 PUFAs exhibited long-term behavioral and histological protection against transient focal cerebral ischemia (tFCI). Importantly, fat-1 transgenic mice also exhibited robust improvements in revascularization and angiogenesis compared to wild type littermates, suggesting a potential role for n-3 fatty acids in post-stroke cerebrovascular remodeling. Mechanistically, n-3 PUFAs induced upregulation of angiopoietin 2 (Ang 2) in astrocytes after tFCI and stimulated extracellular Ang 2 release from cultured astrocytes after oxygen and glucose deprivation. Ang 2 facilitated endothelial proliferation and barrier formation in vitro by potentiating the effects of VEGF on phospholipase Cγ1 and Src signaling. Consistent with these findings, blockade of Src activity in post-stroke fat-1 mice impaired n-3 PUFA-induced angiogenesis and exacerbated long-term neurological outcomes. Taken together, our findings strongly suggest that n-3 PUFA supplementation is a potential angiogenic treatment capable of augmenting brain repair and improving long-term functional recovery after cerebral ischemia.


Subject(s)
Fatty Acids, Omega-3/therapeutic use , Neovascularization, Physiologic/drug effects , Neuroprotective Agents/therapeutic use , Stroke/drug therapy , Animals , Caenorhabditis elegans Proteins/genetics , Cell Proliferation/drug effects , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Coculture Techniques , Disease Models, Animal , Fatty Acid Desaturases/genetics , Glucose/deficiency , Hypoxia/pathology , Ischemic Attack, Transient/complications , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nervous System Diseases/etiology , Nervous System Diseases/prevention & control , Reaction Time/drug effects , Reaction Time/physiology , Stroke/complications , Stroke/etiology , Stroke/pathology , Time Factors
7.
J Neurosci ; 34(5): 1903-15, 2014 Jan 29.
Article in English | MEDLINE | ID: mdl-24478369

ABSTRACT

Ischemic stroke is a debilitating clinical disorder that affects millions of people, yet lacks effective neuroprotective treatments. Fish oil is known to exert beneficial effects against cerebral ischemia. However, the underlying protective mechanisms are not fully understood. The present study tests the hypothesis that omega-3 polyunsaturated fatty acids (n-3 PUFAs) attenuate ischemic neuronal injury by activating nuclear factor E2-related factor 2 (Nrf2) and upregulating heme oxygenase-1 (HO-1) in both in vitro and in vivo models. We observed that pretreatment of rat primary neurons with docosahexaenoic acid (DHA) significantly reduced neuronal death following oxygen-glucose deprivation. This protection was associated with increased Nrf2 activation and HO-1 upregulation. Inhibition of HO-1 activity with tin protoporphyrin IX attenuated the protective effects of DHA. Further studies showed that 4-hydroxy-2E-hexenal (4-HHE), an end-product of peroxidation of n-3 PUFAs, was a more potent Nrf2 inducer than 4-hydroxy-2E-nonenal derived from n-6 PUFAs. In an in vivo setting, transgenic mice overexpressing fatty acid metabolism-1, an enzyme that converts n-6 PUFAs to n-3 PUFAs, were remarkably resistant to focal cerebral ischemia compared with their wild-type littermates. Regular mice fed with a fish oil-enhanced diet also demonstrated significant resistance to ischemia compared with mice fed with a regular diet. As expected, the protection was associated with HO-1 upregulation, Nrf2 activation, and 4-HHE generation. Together, our data demonstrate that n-3 PUFAs are highly effective in protecting the brain, and that the protective mechanisms involve Nrf2 activation and HO-1 upregulation by 4-HHE. Further investigation of n-3 PUFA neuroprotective mechanisms may accelerate the development of stroke therapies.


Subject(s)
Brain Injuries/prevention & control , Brain Ischemia/metabolism , Fatty Acids, Omega-3/therapeutic use , Heme Oxygenase-1/metabolism , NF-E2-Related Factor 2/metabolism , Up-Regulation/drug effects , Aldehydes/pharmacology , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Brain Injuries/etiology , Brain Ischemia/complications , Brain Ischemia/physiopathology , Cadherins/genetics , Cell Death/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Disease Models, Animal , Docosahexaenoic Acids/pharmacology , Dose-Response Relationship, Drug , Embryo, Mammalian , Fatty Acids, Omega-3/pharmacology , Female , Glucose/deficiency , Hypoxia/prevention & control , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Neurons/drug effects , Rats , Time Factors
8.
J Cereb Blood Flow Metab ; 33(9): 1474-84, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23801244

ABSTRACT

Dietary supplementation with omega-3 (ω-3) fatty acids is a safe, economical mean of preventive medicine that has shown protection against several neurologic disorders. The present study tested the hypothesis that this method is protective against controlled cortical impact (CCI). Indeed, mice fed with ω-3 polyunsaturated fatty acid (PUFA)-enriched diet for 2 months exhibited attenuated short and long-term behavioral deficits due to CCI. Although ω-3 PUFAs did not decrease cortical lesion volume, these fatty acids did protect against hippocampal neuronal loss after CCI and reduced pro-inflammatory response. Interestingly, ω-3 PUFAs prevented the loss of myelin basic protein (MPB), preserved the integrity of the myelin sheath, and maintained the nerve fiber conductivity in the CCI model. ω-3 PUFAs also directly protected oligodendrocyte cultures from excitotoxicity and blunted the microglial activation-induced death of oligodendrocytes in microglia/oligodendrocyte cocultures. In sum, ω-3 PUFAs elicit multifaceted protection against behavioral dysfunction, hippocampal neuronal loss, inflammation, and loss of myelination and impulse conductivity. The present report is the first demonstration that ω-3 PUFAs protect against white matter injury in vivo and in vitro. The protective impact of ω-3 PUFAs supports the clinical use of this dietary supplement as a prophylaxis against traumatic brain injury and other nervous system disorders.


Subject(s)
Behavior, Animal/drug effects , Brain Injuries , Cerebral Cortex , Dietary Supplements , Fatty Acids, Omega-3/pharmacology , Hippocampus , Animals , Brain Injuries/drug therapy , Brain Injuries/metabolism , Brain Injuries/pathology , Brain Injuries/physiopathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Male , Mice , Myelin Basic Protein/metabolism , Neurons/metabolism , Neurons/pathology , Oligodendroglia/metabolism , Oligodendroglia/pathology
9.
Neurochem Int ; 61(3): 356-68, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22691629

ABSTRACT

Postmortem tissue from patients with neurodegeneration exhibits protein-misfolding stress and reduced proteasome activity. This hallmark burden of proteotoxic stress has led to the term "proteinopathies" for neurodegenerative diseases. Proteinopathies may also be exacerbated by previous insults, according to the two hit hypothesis of accelerated neurodegeneration. In order to model the response to two successive insults in a high-throughput fashion, we exposed the neuronal cell line N2a to two hits of the proteasome inhibitor MG132 and performed three unbiased viability assays. MG132 toxicity was synergistically exacerbated following sequential hits provided the first hit was high enough to be toxic. This accelerated viability loss was apparent by (1) a nuclear and cytoplasmic stain (DRAQ5+Sapphire), (2) immunocytochemistry for a cytoskeletal marker (α-tubulin), and (3) ATP levels (Cell Titer Glo). Ubiquitin-conjugated proteins were raised by toxic, but not subtoxic MG132, and were thus correlated with toxicity exacerbation at higher doses. We hypothesized that levels of autophagic and antioxidant defenses would be reduced with toxic, but not subtoxic MG132, explaining their differential impact on a second hit. However, proteins involved in chaperone-mediated autophagy were raised by toxic MG132, not reduced. Furthermore, inhibiting autophagy enhanced the toxicity of both subtoxic and toxic MG132 as well as of dual hits, suggesting that autophagic removal of cellular debris protected against proteasome inhibition. Two toxic hits of MG132 synergistically decreased the antioxidant glutathione. The glutathione precursor N-acetyl cysteine reversed this glutathione loss and prevented the toxic response to dual hits by all three assays. Dietary supplementation with N-acetyl cysteine benefits Alzheimer's patients and is currently undergoing clinical trials in Parkinson's disease. The present report is the first demonstration that this versatile compound is protective against synergistic loss of viability as well as of glutathione following unrelenting, sequential hits of proteotoxic stress as may occur in the diseased brain.


Subject(s)
Acetylcysteine/pharmacology , Neurons/drug effects , Autophagy , Blotting, Western , Cell Line, Tumor , Humans , Immunohistochemistry , Neurons/cytology
SELECTION OF CITATIONS
SEARCH DETAIL