Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters

Therapeutic Methods and Therapies TCIM
Database
Language
Affiliation country
Publication year range
1.
J Neurotrauma ; 38(18): 2622-2632, 2021 09 15.
Article in English | MEDLINE | ID: mdl-33913741

ABSTRACT

Repeated mild traumatic brain injury (TBI) can cause persistent neuropathological effects and is a major risk factor for chronic traumatic encephalopathy. PUFAs (n-3 polyunsaturated fatty acids) were shown to improve acute TBI outcomes in single-injury models in most cases. In this study, we demonstrate positive effects of dietary n-3 PUFA on long-term neuropathological and functional outcome in a clinically relevant model of repeated mild TBI using the Closed-Head Impact Model of Engineered Rotational Acceleration (CHIMERA). Adult mice, reared on n-3 PUFA adequate (higher n-3 PUFA) or deficient (lower n-3 PUFA) diets, were given a mild CHIMERA daily for 3 consecutive days. At 2 months after injury, visual function and spatial memory were evaluated. Glia cell activation was assessed by immunostaining using antibodies of ionized calcium-binding adaptor molecule 1 and glial fibrillary acidic protein, and axonal damage was examined using silver staining. Repeated CHIMERA (rCHIMERA)-induced gliosis was significantly suppressed in the optic tract, corpus callosum, and hippocampus of mice fed the n-3 PUFA adequate diet compared to the deficient diet group. Considerable axonal damage was detected in the optic tract after rCHIMERA, but the adequate diet group displayed less axonal damage compared to the deficient diet group. rCHIMERA induced a drastic reduction in N1 amplitude of the visual evoked potential in both diet groups and the a-wave amplitude of the electroretinogram in the deficient diet group. However, reduction of N1 and a-wave amplitude were less severe in the adequate diet group. The Morris water maze probe test indicated a significant decrease in the number of platform crossings in the deficient diet group compared to the adequate group. In summary, dietary n-3 PUFA can attenuate persistent glial cell activation and axonal damage and improve deficits in visual function and spatial memory after repeated mild TBI. These data support the neuroprotective potential of a higher n-3 PUFA diet in ameliorating the adverse outcome of repeated mild TBI.


Subject(s)
Brain Concussion/drug therapy , Brain Concussion/psychology , Diet , Fatty Acids, Omega-3/therapeutic use , Nervous System Diseases/etiology , Animals , Axons/pathology , Fatty Acids, Omega-3/metabolism , Female , Immunohistochemistry , Macrophage Activation , Male , Mice, Inbred C57BL , Nervous System Diseases/pathology , Nervous System Diseases/psychology , Neuroglia/drug effects , Optic Tract/pathology , Pregnancy , Recurrence , Spatial Memory , Treatment Outcome , Vision, Ocular
2.
J Neurosci Res ; 98(11): 2232-2244, 2020 11.
Article in English | MEDLINE | ID: mdl-32840025

ABSTRACT

Previous studies suggest that long-term supplementation and dietary intake of omega-3 polyunsaturated fatty acids (PUFAs) may have neuroprotective effects following brain injury. The objective of this study was to investigate potential neuroprotective effects of omega-3 PUFAs on white matter following closed-head trauma. The closed-head injury model of engineered rotational acceleration (CHIMERA) produces a reproducible injury in the optic tract and brachium of the superior colliculus in mice. Damage is detectable using diffusion tensor imaging (DTI) metrics, particularly fractional anisotropy (FA), with sensitivity comparable to histology. We acquired in vivo (n = 38) and ex vivo (n = 41) DTI data in mice divided into sham and CHIMERA groups with two dietary groups: one deficient in omega-3 PUFAs and one adequate in omega-3 PUFAs. We examined injury effects (reduction in FA) and neuroprotection (FA reduction modulated by diet) in the optic tract and brachium. We verified that diet did not affect FA in sham animals. In injured animals, we found significantly reduced FA in the optic tract and brachium (~10% reduction, p < 0.001), and Bayes factor analysis showed strong evidence to reject the null hypothesis. However, Bayes factor analysis showed substantial evidence to accept the null hypothesis of no diet-related FA differences in injured animals in the in vivo and ex vivo samples. Our results indicate no neuroprotective effect from adequate dietary omega-3 PUFA intake on white matter damage following traumatic brain injury. Since damage from CHIMERA mainly affects white matter, our results do not necessarily contradict previous findings showing omega-3 PUFA-mediated neuroprotection in gray matter.


Subject(s)
Brain Injuries, Traumatic/diagnostic imaging , Diet , Fatty Acids, Omega-3/therapeutic use , Neuroprotective Agents/therapeutic use , White Matter/diagnostic imaging , White Matter/injuries , Animals , Bayes Theorem , Diffusion Tensor Imaging , Gray Matter/pathology , Head Injuries, Closed/diagnostic imaging , Male , Mice , Mice, Inbred C57BL , Optic Tract/diagnostic imaging , Optic Tract/injuries , Superior Colliculi/diagnostic imaging , Superior Colliculi/injuries
3.
J Nutr ; 147(9): 1624-1630, 2017 09.
Article in English | MEDLINE | ID: mdl-28768838

ABSTRACT

Background: Premature infants are deprived of prenatal accumulation of brain docosahexaenoic acid [DHA (22:6n-3)], an omega-3 fatty acid [ω-3 FA (n-3 FA)] important for proper development of cognitive function. The resulting brain DHA deficit can be reversed by ω-3 FA supplementation.Objective: The objective was to test whether there is a critical period for providing ω-3 FA to correct cognitive deficits caused by developmental ω-3 FA deprivation in mice.Methods: Twelve timed-pregnant mice [embryonic day 14 (E14), C57/BL6NCr] were fed an ω-3 FA-deficient diet containing 0.04% α-linolenic acid [ALA (18:3n-3)], and their offspring were fed the same deficient diet (Def group) or changed to an ω-3 FA-adequate diet containing 3.1% ALA at 3 wk, 2 mo, or 4 mo of age. In parallel, 3 E14 pregnant mice were fed the adequate diet and their offspring were fed the same diet (Adeq group) throughout the experiment. Brain FA composition, learning and memory, and hippocampal synaptic protein expression were evaluated at 6 mo by gas chromatography, the Morris water maze test, and western blot analysis, respectively.Results: Maternal dietary ω-3 FA deprivation decreased DHA by >50% in the brain of their offspring at 3 wk of age. The Def group showed significantly worse learning and memory at 6 mo than those groups fed the adequate diet. These pups also had decreased hippocampal expression of postsynaptic density protein 95 (43% of Adeq group), Homer protein homolog 1 (21% of Adeq group), and synaptosome-associated protein of 25 kDa (64% of Adeq group). Changing mice to the adequate diet at 3 wk, 2 mo, or 4 mo of age restored brain DHA to the age-matched adequate concentration. However, deficits in hippocampal synaptic protein expression and spatial learning and memory were normalized only when the diet was changed at 3 wk.Conclusion: Developmental deprivation of brain DHA by dietary ω-3 FA depletion in mice may have a lasting impact on cognitive function if not corrected at an early age.


Subject(s)
Brain/drug effects , Cognition/drug effects , Deficiency Diseases/drug therapy , Docosahexaenoic Acids/administration & dosage , Infant, Premature , Maze Learning/drug effects , Memory/drug effects , Animals , Animals, Newborn , Brain/growth & development , Brain/metabolism , Deficiency Diseases/complications , Diet , Dietary Fats/administration & dosage , Dietary Fats/pharmacology , Dietary Fats/therapeutic use , Disks Large Homolog 4 Protein , Docosahexaenoic Acids/deficiency , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/therapeutic use , Female , Guanylate Kinases/metabolism , Homer Scaffolding Proteins/metabolism , Humans , Infant , Infant, Premature/growth & development , Infant, Premature/metabolism , Maternal Nutritional Physiological Phenomena , Membrane Proteins/metabolism , Mice , Pregnancy , Synaptosomal-Associated Protein 25/metabolism , Time Factors
4.
Adv Neurobiol ; 12: 41-56, 2016.
Article in English | MEDLINE | ID: mdl-27651247

ABSTRACT

With increasing life expectancy as a result of better quality of life and improved health care, the incidence of aging related diseases and disorders is heading toward epidemic proportions. Dementia, a spectrum of neurological diseases associated with aging, is an increasingly prevalent disease. No cure exists yet for dementia; however, there are many potential candidates for treatment of dementia that merit more exploration. Polyphenols, which constitute one such class of compounds, are dietary agents that are globally found in commonly consumed food. Many processes that are associated with the pathophysiology of dementia can be modulated by polyphenols. Polyphenolic compounds can alleviate oxidative stress by acting as direct scavengers of free radicals and clearing superoxide and hydroxyl radicals and by increasing the level of antioxidant enzymes such as glutathione peroxidase. They also chelate metal ions to prevent free radical formation. Polyphenols can also combat inflammation by affecting transcription factors such as NF-κB. Some polyphenols may have the potential to inhibit excitotoxicity by regulating intracellular calcium ion concentration, inhibiting glutamate receptors and increasing glutamate reuptake at the synapse. The cognitive decline in dementia due to decreased availability of acetylcholine can also be countered by polyphenols that inhibit acetyl-cholinesterase activity. Taken together, these findings suggest that increasing the consumption of polyphenol rich food may alleviate the effects of dementia. Moreover, their effects on controlling multiple mechanisms that are associated with dementia may also prevent or slow down the onset and progress of this devastating disease.


Subject(s)
Dementia/diet therapy , Diet , Polyphenols/therapeutic use , Antioxidants/therapeutic use , Humans , Quality of Life
5.
J Neuroinflammation ; 13(1): 253, 2016 Sep 23.
Article in English | MEDLINE | ID: mdl-27663791

ABSTRACT

BACKGROUND: Adequate consumption of polyunsaturated fatty acids (PUFA) is vital for normal development and functioning of the central nervous system. The long-chain n-3 PUFAs docosahexaenoic acid (DHA) and eicosapentaenoic acid are anti-inflammatory and neuroprotective in the models of central nervous system injury including traumatic brain injury (TBI). In the present study, we tested whether a higher brain DHA status in a mouse model on an adequate dietary α-linolenic acid (ALA) leads to reduced neuroinflammation and improved spontaneous recovery after TBI in comparison to a moderately lowered brain DHA status that can occur in humans. METHODS: Mice reared on diets with differing ALA content were injured by a single cortical contusion impact. Change in the expression of inflammatory cytokines was measured, and cellular changes occurring after injury were analyzed by immunostaining for macrophage/microglia and astrocytes. Behavioral studies included rotarod and beam walk tests and contextual fear conditioning. RESULTS: Marginal supply (0.04 %) of ALA as the sole dietary source of n-3 PUFA from early gestation produced reduction of brain DHA by 35 % in adult offspring mice in comparison to the mice on adequate ALA diet (3.1 %). The DHA-depleted group showed significantly increased TBI-induced expression of pro-inflammatory cytokines TNF-α, IL-1ß, and IL-6 in the brain as well as slower functional recovery from motor deficits compared to the adequate ALA group. Despite the reduction of pro-inflammatory cytokine expression, adequate ALA diet did not significantly alter either microglia/macrophage density around the contusion site or the relative M1/M2 phenotype. However, the glial fibrillary acidic protein immunoreactivity was reduced in the injured cerebral cortex of the mice on adequate ALA diet, indicating that astrocyte activation may have contributed to the observed differences in cellular and behavioral responses to TBI. CONCLUSIONS: Increasing the brain DHA level even from a moderately DHA-depleted state can reduce neuroinflammation and improve functional recovery after TBI, suggesting possible improvement of functional outcome by increasing dietary n-3 PUFA in human TBI.

6.
Neurobiol Aging ; 41: 73-85, 2016 May.
Article in English | MEDLINE | ID: mdl-27103520

ABSTRACT

Aging has been related to diminished cognitive function, which could be a result of ineffective synaptic function. We have previously shown that synaptic plasma membrane proteins supporting synaptic integrity and neurotransmission were downregulated in docosahexaenoic acid (DHA)-deprived brains, suggesting an important role of DHA in synaptic function. In this study, we demonstrate aging-induced synaptic proteome changes and DHA-dependent mitigation of such changes using mass spectrometry-based protein quantitation combined with western blot or messenger RNA analysis. We found significant reduction of 15 synaptic plasma membrane proteins in aging brains including fodrin-α, synaptopodin, postsynaptic density protein 95, synaptic vesicle glycoprotein 2B, synaptosomal-associated protein 25, synaptosomal-associated protein-α, N-methyl-D-aspartate receptor subunit epsilon-2 precursor, AMPA2, AP2, VGluT1, munc18-1, dynamin-1, vesicle-associated membrane protein 2, rab3A, and EAAT1, most of which are involved in synaptic transmission. Notably, the first 9 proteins were further reduced when brain DHA was depleted by diet, indicating that DHA plays an important role in sustaining these synaptic proteins downregulated during aging. Reduction of 2 of these proteins was reversed by raising the brain DHA level by supplementing aged animals with an omega-3 fatty acid sufficient diet for 2 months. The recognition memory compromised in DHA-depleted animals was also improved. Our results suggest a potential role of DHA in alleviating aging-associated cognitive decline by offsetting the loss of neurotransmission-regulating synaptic proteins involved in synaptic function.


Subject(s)
Aging/genetics , Aging/metabolism , Brain/metabolism , Docosahexaenoic Acids/metabolism , Docosahexaenoic Acids/physiology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Proteome , Synapses/physiology , Synaptic Transmission/genetics , Synaptic Transmission/physiology , Aging/psychology , Animals , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Down-Regulation , Fatty Acids, Omega-3/administration & dosage , Female , Male , Membrane Proteins/physiology , Memory , Mice, Inbred C57BL , Pregnancy , Recognition, Psychology
7.
PLoS One ; 9(1): e86472, 2014.
Article in English | MEDLINE | ID: mdl-24475126

ABSTRACT

Omega-3 fatty acids are crucial for proper development and function of the brain where docosahexaenoic acid (DHA), the primary omega-3 fatty acid in the brain, is retained avidly by the neuronal membranes. We investigated the effect of DHA depletion in the brain on the outcome of traumatic brain injury (TBI). Pregnant mice were put on an omega-3 fatty acid adequate or deficient diet from gestation day 14 and the pups were raised on the respective diets. Continuation of this dietary regime for three generations resulted in approximately 70% loss of DHA in the brain. Controlled cortical impact was delivered to both groups of mice to produce severe TBI and the functional recovery was compared. Compared to the omega-3 adequate mice, the DHA depleted mice exhibited significantly slower recovery from motor deficits evaluated by the rotarod and the beam walk tests. Furthermore, the DHA deficient mice showed greater anxiety-like behavior tested in the open field test as well as cognitive deficits evaluated by the novel object recognition test. The level of alpha spectrin II breakdown products, the markers of TBI, was significantly elevated in the deficient mouse cortices, indicating that the injury is greater in the deficient brains. This observation was further supported by the reduction of NeuN positive cells around the site of injury in the deficient mice, indicating exacerbated neuronal death after injury. These results suggest an important influence of the brain DHA status on TBI outcome.


Subject(s)
Biomarkers/metabolism , Brain Injuries/metabolism , Brain Injuries/physiopathology , Brain/metabolism , Docosahexaenoic Acids/deficiency , Analysis of Variance , Animals , Anxiety/etiology , Anxiety/physiopathology , Blotting, Western , Chromatography, Gas , DNA-Binding Proteins , Fatty Acids/metabolism , Female , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Pregnancy , Psychomotor Performance/physiology , Rotarod Performance Test , Spectrin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL