51
|
Belkouch M, Hachem M, Elgot A, Lo Van A, Picq M, Guichardant M, Lagarde M, Bernoud-Hubac N. The pleiotropic effects of omega-3 docosahexaenoic acid on the hallmarks of Alzheimer's disease. J Nutr Biochem 2016; 38:1-11. [PMID: 27825512 DOI: 10.1016/j.jnutbio.2016.03.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 12/14/2015] [Accepted: 03/03/2016] [Indexed: 12/17/2022]
Abstract
Among omega-3 polyunsaturated fatty acids (PUFAs), docosahexaenoic acid (DHA, 22:6n-3) is important for adequate brain development and cognition. DHA is highly concentrated in the brain and plays an essential role in brain functioning. DHA, one of the major constituents in fish fats, readily crosses the blood-brain barrier from blood to the brain. Its critical role was further supported by its reduced levels in the brain of Alzheimer's disease (AD) patients. This agrees with a potential role of DHA in memory, learning and cognitive processes. Since there is yet no cure for dementia such as AD, there is growing interest in the role of DHA-supplemented diet in the prevention of AD pathogenesis. Accordingly, animal, epidemiological, preclinical and clinical studies indicated that DHA has neuroprotective effects in a number of neurodegenerative conditions including AD. The beneficial effects of this key omega-3 fatty acid supplementation may depend on the stage of disease progression, other dietary mediators and the apolipoprotein ApoE genotype. Herein, our review investigates, from animal and cell culture studies, the molecular mechanisms involved in the neuroprotective potential of DHA with emphasis on AD.
Collapse
Affiliation(s)
- Mounir Belkouch
- Université de Lyon, UMR INSERM 1060, UMR INRA 1397, IMBL/INSA-Lyon, Cardiovasculaire, Métabolisme, Diabétologie et Nutrition Laboratory, Bât Louis Pasteur, INSA, Villeurbanne, France.
| | - Mayssa Hachem
- Université de Lyon, UMR INSERM 1060, UMR INRA 1397, IMBL/INSA-Lyon, Cardiovasculaire, Métabolisme, Diabétologie et Nutrition Laboratory, Bât Louis Pasteur, INSA, Villeurbanne, France
| | - Abdeljalil Elgot
- Laboratoire des Sciences et Technologies de la Santé, Unité des Sciences Biomédicales, Institut Supérieur des Sciences de la Santé, Université Hassan 1er, Settat, Morocco
| | - Amanda Lo Van
- Université de Lyon, UMR INSERM 1060, UMR INRA 1397, IMBL/INSA-Lyon, Cardiovasculaire, Métabolisme, Diabétologie et Nutrition Laboratory, Bât Louis Pasteur, INSA, Villeurbanne, France
| | - Madeleine Picq
- Université de Lyon, UMR INSERM 1060, UMR INRA 1397, IMBL/INSA-Lyon, Cardiovasculaire, Métabolisme, Diabétologie et Nutrition Laboratory, Bât Louis Pasteur, INSA, Villeurbanne, France
| | - Michel Guichardant
- Université de Lyon, UMR INSERM 1060, UMR INRA 1397, IMBL/INSA-Lyon, Cardiovasculaire, Métabolisme, Diabétologie et Nutrition Laboratory, Bât Louis Pasteur, INSA, Villeurbanne, France
| | - Michel Lagarde
- Université de Lyon, UMR INSERM 1060, UMR INRA 1397, IMBL/INSA-Lyon, Cardiovasculaire, Métabolisme, Diabétologie et Nutrition Laboratory, Bât Louis Pasteur, INSA, Villeurbanne, France
| | - Nathalie Bernoud-Hubac
- Université de Lyon, UMR INSERM 1060, UMR INRA 1397, IMBL/INSA-Lyon, Cardiovasculaire, Métabolisme, Diabétologie et Nutrition Laboratory, Bât Louis Pasteur, INSA, Villeurbanne, France
| |
Collapse
|
52
|
Effects of pre-natal alcohol exposure on hippocampal synaptic plasticity: Sex, age and methodological considerations. Neurosci Biobehav Rev 2016; 64:12-34. [PMID: 26906760 DOI: 10.1016/j.neubiorev.2016.02.014] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 01/14/2016] [Accepted: 02/18/2016] [Indexed: 12/28/2022]
Abstract
The consumption of alcohol during gestation is detrimental to the developing central nervous system (CNS). The severity of structural and functional brain alterations associated with alcohol intake depends on many factors including the timing and duration of alcohol consumption. The hippocampal formation, a brain region implicated in learning and memory, is highly susceptible to the effects of developmental alcohol exposure. Some of the observed effects of alcohol on learning and memory may be due to changes at the synaptic level, as this teratogen has been repeatedly shown to interfere with hippocampal synaptic plasticity. At the molecular level alcohol interferes with receptor proteins and can disrupt hormones that are important for neuronal signaling and synaptic plasticity. In this review we examine the consequences of prenatal and early postnatal alcohol exposure on hippocampal synaptic plasticity and highlight the numerous factors that can modulate the effects of alcohol. We also discuss some potential mechanisms responsible for these changes as well as emerging therapeutic avenues that are beginning to be explored.
Collapse
|
53
|
Hoffman DR, Hughbanks-Wheaton DK, Spencer R, Fish GE, Pearson NS, Wang YZ, Klein M, Takacs A, Locke KG, Birch DG. Docosahexaenoic Acid Slows Visual Field Progression in X-Linked Retinitis Pigmentosa: Ancillary Outcomes of the DHAX Trial. Invest Ophthalmol Vis Sci 2016; 56:6646-53. [PMID: 26469750 DOI: 10.1167/iovs.15-17786] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Docosahexaenoic acid (DHA) was supplemented in a single-site, placebo-controlled, randomized clinical trial designed to slow vision loss associated with X-linked retinitis pigmentosa (XLRP); the DHAX Trial. We previously reported no significant differences between supplemented and placebo groups in intent-to-treat analysis of primary ERG outcomes. Assessed herein are hypothesis-generating measures of ancillary visual function outcomes in participants fully adhering to trial protocol. METHODS Male participants with XLRP (range, 7-31 years) received 30 mg DHA/kg/d (n = 29) or placebo (n = 22) for 4 years. Visual outcomes were measured annually and red blood cell (RBC) DHA determined every 6 months. RESULTS Oral DHA supplementation increased mean RBC-DHA levels by 4-fold (P < 0.0001) over placebo. No group differences in progression were found for visual acuity (P = 0.11), shape discrimination (P = 0.18), or fundus appearance (P = 0.70). Optical coherence tomography (OCT) became available during year 2 of the trial; no group differences were seen in ellipsoid zone constriction (P = 0.87) over 2 years. Yearly rates of progression were reduced for dark-adapted thresholds (P = 0.06) and visual field sensitivity for foveal, macular, peripheral, total, and ellipsoid zone regions by DHA supplementation (P = 0.039, P = 0.031, P < 0.0001, P < 0.0001, and P = 0.033). Rates of visual field sensitivity decline were dependent on RBC-DHA (P = 0.046 to <0.0001). CONCLUSIONS Supplementation of DHA significantly elevated blood DHA levels and reduced the rate of progression in final dark-adapted thresholds and visual field sensitivity. From the relationship between RBC-DHA and the rate of field sensitivity loss, we can extrapolate that an RBC-DHA level of 17% could minimize the decline in field sensitivity. (ClinicalTrials.gov number, NCT00100230.)
Collapse
Affiliation(s)
- Dennis R Hoffman
- Retina Foundation of the Southwest, Dallas, Texas, United States 2Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Dianna K Hughbanks-Wheaton
- Retina Foundation of the Southwest, Dallas, Texas, United States 2Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Rand Spencer
- Texas Retina Associates, Dallas, Texas, United States
| | - Gary E Fish
- Texas Retina Associates, Dallas, Texas, United States
| | - N Shirlene Pearson
- Center for Teaching Excellence, Southern Methodist University, Dallas, Texas, United States
| | - Yi-Zhong Wang
- Retina Foundation of the Southwest, Dallas, Texas, United States 2Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Martin Klein
- Retina Foundation of the Southwest, Dallas, Texas, United States
| | - Alison Takacs
- Retina Foundation of the Southwest, Dallas, Texas, United States
| | - Kirsten G Locke
- Retina Foundation of the Southwest, Dallas, Texas, United States
| | - David G Birch
- Retina Foundation of the Southwest, Dallas, Texas, United States 2Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| |
Collapse
|
54
|
Alashmali SM, Hopperton KE, Bazinet RP. Lowering dietary n-6 polyunsaturated fatty acids: interaction with brain arachidonic and docosahexaenoic acids. Curr Opin Lipidol 2016; 27:54-66. [PMID: 26709472 DOI: 10.1097/mol.0000000000000255] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE OF REVIEW Arachidonic (ARA) and docosahexaenoic (DHA) acids are the most abundant polyunsaturated fatty acids (PUFA) in the brain, where they have many biological effects, including on inflammation, cell-signaling, appetite regulation, and blood flow. The Western diet contains a high ratio of n-6: n-3 PUFA. Although interest in lowering this ratio has largely focused on increasing intake of n-3 PUFA, few studies have examined lowering dietary n-6 PUFA. This review will evaluate the effect of lowering dietary n-6 PUFA on levels and metabolism of ARA and DHA in animal models and in humans, with a primary focus on the brain. RECENT FINDINGS In animal models, lowering dietary ARA or linoleic acid generally lowers levels of brain ARA and raises DHA. Lowering dietary n-6 PUFA can also modulate the levels of ARA and DHA metabolizing enzymes, as well as their associated bioactive mediators. Human studies examining changes in plasma fatty acid composition following n-6 PUFA lowering demonstrate no changes in levels of ARA and DHA, though there is evidence of alterations in their respective bioactive mediators. SUMMARY Lowering dietary n-6 PUFA, in animal models, can alter the levels and metabolism of ARA and DHA in the brain, but it remains to be determined whether these changes are clinically meaningful.
Collapse
Affiliation(s)
- Shoug M Alashmali
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada *Shoug M. Alashmali and Kathryn E. Hopperton contributed equally to the writing of this article
| | | | | |
Collapse
|
55
|
Simón MV, Agnolazza DL, German OL, Garelli A, Politi LE, Agbaga MP, Anderson RE, Rotstein NP. Synthesis of docosahexaenoic acid from eicosapentaenoic acid in retina neurons protects photoreceptors from oxidative stress. J Neurochem 2016; 136:931-46. [PMID: 26662863 DOI: 10.1111/jnc.13487] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 11/28/2015] [Accepted: 12/02/2015] [Indexed: 12/27/2022]
Abstract
Oxidative stress is involved in activating photoreceptor death in several retinal degenerations. Docosahexaenoic acid (DHA), the major polyunsaturated fatty acid in the retina, protects cultured retina photoreceptors from apoptosis induced by oxidative stress and promotes photoreceptor differentiation. Here, we investigated whether eicosapentaenoic acid (EPA), a metabolic precursor to DHA, had similar effects and whether retinal neurons could metabolize EPA to DHA. Adding EPA to rat retina neuronal cultures increased opsin expression and protected photoreceptors from apoptosis induced by the oxidants paraquat and hydrogen peroxide (H2 O2 ). Palmitic, oleic, and arachidonic acids had no protective effect, showing the specificity for DHA. We found that EPA supplementation significantly increased DHA percentage in retinal neurons, but not EPA percentage. Photoreceptors and glial cells expressed Δ6 desaturase (FADS2), which introduces the last double bond in DHA biosynthetic pathway. Pre-treatment of neuronal cultures with CP-24879 hydrochloride, a Δ5/Δ6 desaturase inhibitor, prevented EPA-induced increase in DHA percentage and completely blocked EPA protection and its effect on photoreceptor differentiation. These results suggest that EPA promoted photoreceptor differentiation and rescued photoreceptors from oxidative stress-induced apoptosis through its elongation and desaturation to DHA. Our data show, for the first time, that isolated retinal neurons can synthesize DHA in culture. Docosahexaenoic acid (DHA), the major polyunsaturated fatty acid in retina photoreceptors, and its precursor, eicosapentaenoic acid (EPA) have multiple beneficial effects. Here, we show that retina neurons in vitro express the desaturase FADS2 and can synthesize DHA from EPA. Moreover, addition of EPA to these cultures protects photoreceptors from oxidative stress and promotes their differentiation through its metabolization to DHA.
Collapse
Affiliation(s)
- María Victoria Simón
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Department of Biology, Biochemistry and Pharmacy, Univ Nacional del Sur (UNS)-CONICET, Bahía Blanca, Buenos Aires, Argentina
| | - Daniela L Agnolazza
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Department of Biology, Biochemistry and Pharmacy, Univ Nacional del Sur (UNS)-CONICET, Bahía Blanca, Buenos Aires, Argentina
| | - Olga Lorena German
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Department of Biology, Biochemistry and Pharmacy, Univ Nacional del Sur (UNS)-CONICET, Bahía Blanca, Buenos Aires, Argentina
| | - Andrés Garelli
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Department of Biology, Biochemistry and Pharmacy, Univ Nacional del Sur (UNS)-CONICET, Bahía Blanca, Buenos Aires, Argentina
| | - Luis E Politi
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Department of Biology, Biochemistry and Pharmacy, Univ Nacional del Sur (UNS)-CONICET, Bahía Blanca, Buenos Aires, Argentina
| | - Martin-Paul Agbaga
- Cell Biology, Univ of Oklahoma Hlth Sci Ctr, Oklahoma City, Oklahoma, USA.,Univ of Oklahoma Hlth Sci Ctr, Oklahoma City, Oklahoma, USA
| | - Robert E Anderson
- Univ of Oklahoma Hlth Sci Ctr, Oklahoma City, Oklahoma, USA.,Ophthalmology/Cell Biology, Univ of Oklahoma Hlth Sci Ctr, Oklahoma City, Oklahoma, USA
| | - Nora P Rotstein
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Department of Biology, Biochemistry and Pharmacy, Univ Nacional del Sur (UNS)-CONICET, Bahía Blanca, Buenos Aires, Argentina
| |
Collapse
|
56
|
Wang H, Liang S, Wang M, Gao J, Sun C, Wang J, Xia W, Wu S, Sumner SJ, Zhang F, Sun C, Wu L. Potential serum biomarkers from a metabolomics study of autism. J Psychiatry Neurosci 2016; 41:27-37. [PMID: 26395811 PMCID: PMC4688025 DOI: 10.1503/jpn.140009] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Early detection and diagnosis are very important for autism. Current diagnosis of autism relies mainly on some observational questionnaires and interview tools that may involve a great variability. We performed a metabolomics analysis of serum to identify potential biomarkers for the early diagnosis and clinical evaluation of autism. METHODS We analyzed a discovery cohort of patients with autism and participants without autism in the Chinese Han population using ultra-performance liquid chromatography quadrupole time-of-flight tandem mass spectrometry (UPLC/Q-TOF MS/MS) to detect metabolic changes in serum associated with autism. The potential metabolite candidates for biomarkers were individually validated in an additional independent cohort of cases and controls. We built a multiple logistic regression model to evaluate the validated biomarkers. RESULTS We included 73 patients and 63 controls in the discovery cohort and 100 cases and 100 controls in the validation cohort. Metabolomic analysis of serum in the discovery stage identified 17 metabolites, 11 of which were validated in an independent cohort. A multiple logistic regression model built on the 11 validated metabolites fit well in both cohorts. The model consistently showed that autism was associated with 2 particular metabolites: sphingosine 1-phosphate and docosahexaenoic acid. LIMITATIONS While autism is diagnosed predominantly in boys, we were unable to perform the analysis by sex owing to difficulty recruiting enough female patients. Other limitations include the need to perform test-retest assessment within the same individual and the relatively small sample size. CONCLUSION Two metabolites have potential as biomarkers for the clinical diagnosis and evaluation of autism.
Collapse
Affiliation(s)
- Han Wang
- From the Department of Child and Adolescent Health, School of Public Health, Harbin Medical University, Harbin, Heilongjiang, China (Wang, Liang, Gao, Sun, Wang, Xia, Wu); the Center for Endemic Disease Control, China Center for Disease Control and Prevention and Harbin Medical University, Harbin, Heilongjiang, China (Wang); the Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, Heilongjiang, China (Wang, Sun); the Department of Nursing, Daqing Campus, Harbin Medical University, Daqing, Heilongjiang, China (Gao); the Advanced Analytic Division, SAS Institute, Inc, Cary, North Carolina, USA (Wu); Systems and Translational Sciences, Research Triangle Institute, Research Triangle Park, North Carolina, USA (Sumner); and Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland, USA (Zhang)
| | - Shuang Liang
- From the Department of Child and Adolescent Health, School of Public Health, Harbin Medical University, Harbin, Heilongjiang, China (Wang, Liang, Gao, Sun, Wang, Xia, Wu); the Center for Endemic Disease Control, China Center for Disease Control and Prevention and Harbin Medical University, Harbin, Heilongjiang, China (Wang); the Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, Heilongjiang, China (Wang, Sun); the Department of Nursing, Daqing Campus, Harbin Medical University, Daqing, Heilongjiang, China (Gao); the Advanced Analytic Division, SAS Institute, Inc, Cary, North Carolina, USA (Wu); Systems and Translational Sciences, Research Triangle Institute, Research Triangle Park, North Carolina, USA (Sumner); and Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland, USA (Zhang)
| | - Maoqing Wang
- From the Department of Child and Adolescent Health, School of Public Health, Harbin Medical University, Harbin, Heilongjiang, China (Wang, Liang, Gao, Sun, Wang, Xia, Wu); the Center for Endemic Disease Control, China Center for Disease Control and Prevention and Harbin Medical University, Harbin, Heilongjiang, China (Wang); the Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, Heilongjiang, China (Wang, Sun); the Department of Nursing, Daqing Campus, Harbin Medical University, Daqing, Heilongjiang, China (Gao); the Advanced Analytic Division, SAS Institute, Inc, Cary, North Carolina, USA (Wu); Systems and Translational Sciences, Research Triangle Institute, Research Triangle Park, North Carolina, USA (Sumner); and Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland, USA (Zhang)
| | - Jingquan Gao
- From the Department of Child and Adolescent Health, School of Public Health, Harbin Medical University, Harbin, Heilongjiang, China (Wang, Liang, Gao, Sun, Wang, Xia, Wu); the Center for Endemic Disease Control, China Center for Disease Control and Prevention and Harbin Medical University, Harbin, Heilongjiang, China (Wang); the Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, Heilongjiang, China (Wang, Sun); the Department of Nursing, Daqing Campus, Harbin Medical University, Daqing, Heilongjiang, China (Gao); the Advanced Analytic Division, SAS Institute, Inc, Cary, North Carolina, USA (Wu); Systems and Translational Sciences, Research Triangle Institute, Research Triangle Park, North Carolina, USA (Sumner); and Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland, USA (Zhang)
| | - Caihong Sun
- From the Department of Child and Adolescent Health, School of Public Health, Harbin Medical University, Harbin, Heilongjiang, China (Wang, Liang, Gao, Sun, Wang, Xia, Wu); the Center for Endemic Disease Control, China Center for Disease Control and Prevention and Harbin Medical University, Harbin, Heilongjiang, China (Wang); the Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, Heilongjiang, China (Wang, Sun); the Department of Nursing, Daqing Campus, Harbin Medical University, Daqing, Heilongjiang, China (Gao); the Advanced Analytic Division, SAS Institute, Inc, Cary, North Carolina, USA (Wu); Systems and Translational Sciences, Research Triangle Institute, Research Triangle Park, North Carolina, USA (Sumner); and Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland, USA (Zhang)
| | - Jia Wang
- From the Department of Child and Adolescent Health, School of Public Health, Harbin Medical University, Harbin, Heilongjiang, China (Wang, Liang, Gao, Sun, Wang, Xia, Wu); the Center for Endemic Disease Control, China Center for Disease Control and Prevention and Harbin Medical University, Harbin, Heilongjiang, China (Wang); the Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, Heilongjiang, China (Wang, Sun); the Department of Nursing, Daqing Campus, Harbin Medical University, Daqing, Heilongjiang, China (Gao); the Advanced Analytic Division, SAS Institute, Inc, Cary, North Carolina, USA (Wu); Systems and Translational Sciences, Research Triangle Institute, Research Triangle Park, North Carolina, USA (Sumner); and Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland, USA (Zhang)
| | - Wei Xia
- From the Department of Child and Adolescent Health, School of Public Health, Harbin Medical University, Harbin, Heilongjiang, China (Wang, Liang, Gao, Sun, Wang, Xia, Wu); the Center for Endemic Disease Control, China Center for Disease Control and Prevention and Harbin Medical University, Harbin, Heilongjiang, China (Wang); the Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, Heilongjiang, China (Wang, Sun); the Department of Nursing, Daqing Campus, Harbin Medical University, Daqing, Heilongjiang, China (Gao); the Advanced Analytic Division, SAS Institute, Inc, Cary, North Carolina, USA (Wu); Systems and Translational Sciences, Research Triangle Institute, Research Triangle Park, North Carolina, USA (Sumner); and Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland, USA (Zhang)
| | - Shiying Wu
- From the Department of Child and Adolescent Health, School of Public Health, Harbin Medical University, Harbin, Heilongjiang, China (Wang, Liang, Gao, Sun, Wang, Xia, Wu); the Center for Endemic Disease Control, China Center for Disease Control and Prevention and Harbin Medical University, Harbin, Heilongjiang, China (Wang); the Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, Heilongjiang, China (Wang, Sun); the Department of Nursing, Daqing Campus, Harbin Medical University, Daqing, Heilongjiang, China (Gao); the Advanced Analytic Division, SAS Institute, Inc, Cary, North Carolina, USA (Wu); Systems and Translational Sciences, Research Triangle Institute, Research Triangle Park, North Carolina, USA (Sumner); and Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland, USA (Zhang)
| | - Susan J Sumner
- From the Department of Child and Adolescent Health, School of Public Health, Harbin Medical University, Harbin, Heilongjiang, China (Wang, Liang, Gao, Sun, Wang, Xia, Wu); the Center for Endemic Disease Control, China Center for Disease Control and Prevention and Harbin Medical University, Harbin, Heilongjiang, China (Wang); the Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, Heilongjiang, China (Wang, Sun); the Department of Nursing, Daqing Campus, Harbin Medical University, Daqing, Heilongjiang, China (Gao); the Advanced Analytic Division, SAS Institute, Inc, Cary, North Carolina, USA (Wu); Systems and Translational Sciences, Research Triangle Institute, Research Triangle Park, North Carolina, USA (Sumner); and Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland, USA (Zhang)
| | - Fengyu Zhang
- From the Department of Child and Adolescent Health, School of Public Health, Harbin Medical University, Harbin, Heilongjiang, China (Wang, Liang, Gao, Sun, Wang, Xia, Wu); the Center for Endemic Disease Control, China Center for Disease Control and Prevention and Harbin Medical University, Harbin, Heilongjiang, China (Wang); the Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, Heilongjiang, China (Wang, Sun); the Department of Nursing, Daqing Campus, Harbin Medical University, Daqing, Heilongjiang, China (Gao); the Advanced Analytic Division, SAS Institute, Inc, Cary, North Carolina, USA (Wu); Systems and Translational Sciences, Research Triangle Institute, Research Triangle Park, North Carolina, USA (Sumner); and Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland, USA (Zhang)
| | - Changhao Sun
- From the Department of Child and Adolescent Health, School of Public Health, Harbin Medical University, Harbin, Heilongjiang, China (Wang, Liang, Gao, Sun, Wang, Xia, Wu); the Center for Endemic Disease Control, China Center for Disease Control and Prevention and Harbin Medical University, Harbin, Heilongjiang, China (Wang); the Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, Heilongjiang, China (Wang, Sun); the Department of Nursing, Daqing Campus, Harbin Medical University, Daqing, Heilongjiang, China (Gao); the Advanced Analytic Division, SAS Institute, Inc, Cary, North Carolina, USA (Wu); Systems and Translational Sciences, Research Triangle Institute, Research Triangle Park, North Carolina, USA (Sumner); and Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland, USA (Zhang)
| | - Lijie Wu
- From the Department of Child and Adolescent Health, School of Public Health, Harbin Medical University, Harbin, Heilongjiang, China (Wang, Liang, Gao, Sun, Wang, Xia, Wu); the Center for Endemic Disease Control, China Center for Disease Control and Prevention and Harbin Medical University, Harbin, Heilongjiang, China (Wang); the Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, Heilongjiang, China (Wang, Sun); the Department of Nursing, Daqing Campus, Harbin Medical University, Daqing, Heilongjiang, China (Gao); the Advanced Analytic Division, SAS Institute, Inc, Cary, North Carolina, USA (Wu); Systems and Translational Sciences, Research Triangle Institute, Research Triangle Park, North Carolina, USA (Sumner); and Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland, USA (Zhang)
| |
Collapse
|
57
|
Zhang W, Liu J, Hu X, Li P, Leak RK, Gao Y, Chen J. n-3 Polyunsaturated Fatty Acids Reduce Neonatal Hypoxic/Ischemic Brain Injury by Promoting Phosphatidylserine Formation and Akt Signaling. Stroke 2015; 46:2943-50. [PMID: 26374481 DOI: 10.1161/strokeaha.115.010815] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 08/13/2015] [Indexed: 01/24/2023]
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.
Collapse
Affiliation(s)
- Wenting Zhang
- From the State Key Laboratory of Medical Neurobiology, Institute of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China (W.Z., J.L., X.H., P.L., Y.G., J.C.); Center of Cerebrovascular Disease Research, Department of Neurology, University of Pittsburgh School of Medicine, PA (X.H., J.C.); Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA (R.K.L.); Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA (X.H., J.C.).
| | - Jia Liu
- From the State Key Laboratory of Medical Neurobiology, Institute of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China (W.Z., J.L., X.H., P.L., Y.G., J.C.); Center of Cerebrovascular Disease Research, Department of Neurology, University of Pittsburgh School of Medicine, PA (X.H., J.C.); Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA (R.K.L.); Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA (X.H., J.C.)
| | - Xiaoming Hu
- From the State Key Laboratory of Medical Neurobiology, Institute of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China (W.Z., J.L., X.H., P.L., Y.G., J.C.); Center of Cerebrovascular Disease Research, Department of Neurology, University of Pittsburgh School of Medicine, PA (X.H., J.C.); Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA (R.K.L.); Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA (X.H., J.C.)
| | - Peiying Li
- From the State Key Laboratory of Medical Neurobiology, Institute of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China (W.Z., J.L., X.H., P.L., Y.G., J.C.); Center of Cerebrovascular Disease Research, Department of Neurology, University of Pittsburgh School of Medicine, PA (X.H., J.C.); Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA (R.K.L.); Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA (X.H., J.C.)
| | - Rehana K Leak
- From the State Key Laboratory of Medical Neurobiology, Institute of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China (W.Z., J.L., X.H., P.L., Y.G., J.C.); Center of Cerebrovascular Disease Research, Department of Neurology, University of Pittsburgh School of Medicine, PA (X.H., J.C.); Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA (R.K.L.); Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA (X.H., J.C.)
| | - Yanqin Gao
- From the State Key Laboratory of Medical Neurobiology, Institute of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China (W.Z., J.L., X.H., P.L., Y.G., J.C.); Center of Cerebrovascular Disease Research, Department of Neurology, University of Pittsburgh School of Medicine, PA (X.H., J.C.); Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA (R.K.L.); Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA (X.H., J.C.)
| | - Jun Chen
- From the State Key Laboratory of Medical Neurobiology, Institute of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China (W.Z., J.L., X.H., P.L., Y.G., J.C.); Center of Cerebrovascular Disease Research, Department of Neurology, University of Pittsburgh School of Medicine, PA (X.H., J.C.); Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA (R.K.L.); Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA (X.H., J.C.).
| |
Collapse
|
58
|
German OL, Agnolazza DL, Politi LE, Rotstein NP. Light, lipids and photoreceptor survival: live or let die? Photochem Photobiol Sci 2015. [PMID: 26204250 DOI: 10.1039/c5pp00194c] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Due to its constant exposure to light and its high oxygen consumption the retina is highly sensitive to oxidative damage, which is a common factor in inducing the death of photoreceptors after light damage or in inherited retinal degenerations. The high content of docosahexaenoic acid (DHA), the major polyunsaturated fatty acid in the retina, has been suggested to contribute to this sensitivity. DHA is crucial for developing and preserving normal visual function. However, further roles of DHA in the retina are still controversial. Current data support that it can tilt the scale either towards degeneration or survival of retinal cells. DHA peroxidation products can be deleterious to the retina and might lead to retinal degeneration. However, DHA has also been shown to act as, or to be the source of, a survival molecule that protects photoreceptors and retinal pigment epithelium cells from oxidative damage. We have established that DHA protects photoreceptors from oxidative stress-induced apoptosis and promotes their differentiation in vitro. DHA activates the retinoid X receptor (RXR) and the ERK/MAPK pathway, thus regulating the expression of anti and pro-apoptotic proteins. It also orchestrates a diversity of signaling pathways, modulating enzymatic pathways that control the sphingolipid metabolism and activate antioxidant defense mechanisms to promote photoreceptor survival and development. A deeper comprehension of DHA signaling pathways and context-dependent behavior is required to understand its dual functions in retinal physiology.
Collapse
Affiliation(s)
- Olga Lorena German
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Universidad Nacional del Sur (UNS)-CONICET, Bahía Blanca, Buenos Aires, Argentina.
| | | | | | | |
Collapse
|
59
|
Liu F, Rainosek SW, Frisch-Daiello JL, Patterson TA, Paule MG, Slikker W, Wang C, Han X. Potential Adverse Effects of Prolonged Sevoflurane Exposure on Developing Monkey Brain: From Abnormal Lipid Metabolism to Neuronal Damage. Toxicol Sci 2015. [PMID: 26206149 DOI: 10.1093/toxsci/kfv150] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Sevoflurane is a volatile anesthetic that has been widely used in general anesthesia, yet its safety in pediatric use is a public concern. This study sought to evaluate whether prolonged exposure of infant monkeys to a clinically relevant concentration of sevoflurane is associated with any adverse effects on the developing brain. Infant monkeys were exposed to 2.5% sevoflurane for 9 h, and frontal cortical tissues were harvested for DNA microarray, lipidomics, Luminex protein, and histological assays. DNA microarray analysis showed that sevoflurane exposure resulted in a broad identification of differentially expressed genes (DEGs) in the monkey brain. In general, these genes were associated with nervous system development, function, and neural cell viability. Notably, a number of DEGs were closely related to lipid metabolism. Lipidomic analysis demonstrated that critical lipid components, (eg, phosphatidylethanolamine, phosphatidylserine, and phosphatidylglycerol) were significantly downregulated by prolonged exposure of sevoflurane. Luminex protein analysis indicated abnormal levels of cytokines in sevoflurane-exposed brains. Consistently, Fluoro-Jade C staining revealed more degenerating neurons after sevoflurane exposure. These data demonstrate that a clinically relevant concentration of sevoflurane (2.5%) is capable of inducing and maintaining an effective surgical plane of anesthesia in the developing nonhuman primate and that a prolonged exposure of 9 h resulted in profound changes in gene expression, cytokine levels, lipid metabolism, and subsequently, neuronal damage. Generally, sevoflurane-induced neuronal damage was also associated with changes in lipid content, composition, or both; and specific lipid changes could provide insights into the molecular mechanism(s) underlying anesthetic-induced neurotoxicity and may be sensitive biomarkers for the early detection of anesthetic-induced neuronal damage.
Collapse
Affiliation(s)
- Fang Liu
- *Division of Neurotoxicology, National Center for Toxicological Research/Food and Drug Administration, Jefferson, AR 72079;
| | - Shuo W Rainosek
- Department of Anesthesiology, University of Arkansas for Medical Sciences, Little Rock, AR 72205
| | - Jessica L Frisch-Daiello
- Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute at Lake Nona, Orlando, FL 32827; and
| | - Tucker A Patterson
- *Division of Neurotoxicology, National Center for Toxicological Research/Food and Drug Administration, Jefferson, AR 72079
| | - Merle G Paule
- *Division of Neurotoxicology, National Center for Toxicological Research/Food and Drug Administration, Jefferson, AR 72079
| | - William Slikker
- Office of the Director, National Center for Toxicological Research/Food and Drug Administration, Jefferson, AR 72079
| | - Cheng Wang
- *Division of Neurotoxicology, National Center for Toxicological Research/Food and Drug Administration, Jefferson, AR 72079
| | - Xianlin Han
- Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute at Lake Nona, Orlando, FL 32827; and
| |
Collapse
|
60
|
The effects of acute and chronic administration of phosphatidylserine on cell proliferation and survival in the dentate gyrus of adult and middle-aged rats. Brain Res 2015; 1609:72-81. [DOI: 10.1016/j.brainres.2015.03.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 03/04/2015] [Accepted: 03/09/2015] [Indexed: 11/19/2022]
|
61
|
Khan MZ, He L. The role of polyunsaturated fatty acids and GPR40 receptor in brain. Neuropharmacology 2015; 113:639-651. [PMID: 26005184 DOI: 10.1016/j.neuropharm.2015.05.013] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 04/02/2015] [Accepted: 05/08/2015] [Indexed: 01/15/2023]
Abstract
Polyunsaturated fatty acids (PUFAs) are found in abundance in the nervous system. They perform significant functions for example boosting synaptogenesis, neurogenesis, inducing antinociception, stimulating gene expression and neuronal activity, preventing apoptosis and neuroinflammation. G-protein-coupled receptor 40 (GPR40), also called free fatty acid receptor 1 (FFA1), is ubiquitously expressed in various regions of the human brain including the olfactory bulb, midbrain, medulla oblongata, hippocampus, hypothalamus, cerebral cortex, cerebellum and in the spinal cord. GPR40, when binding with polyunsaturated fatty acids (PUFAs) has shown promising therapeutic potential. This review presents current knowledge regarding the pharmacological properties of GPR40 and addresses its functions in brain, with a focus on neurodevelopment & neurogenesis. Furthermore, the demonstration of GPR40 involvement in several neuropathological conditions such as apoptosis, inflammatory pain, Alzheimer's disease and Parkinson's disease. Although the results are encouraging, further research is needed to clarify their role in the treatment of inflammatory pain, Alzheimer's disease and Parkinson's disease. This article is part of the Special Issue entitled 'Lipid Sensing G Protein-Coupled Receptors in the CNS'.
Collapse
Affiliation(s)
- Muhammad Zahid Khan
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Ling He
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
62
|
Tostes MHFDS, Polonini HC, Mendes R, Brandão MAF, Gattaz WF, Raposo NRB. Fatty acid and phospholipase A2 plasma levels in children with autism. TRENDS IN PSYCHIATRY AND PSYCHOTHERAPY 2015; 35:76-80. [PMID: 25923188 DOI: 10.1590/s2237-60892013000100009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Accepted: 11/26/2012] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To evaluate fatty acid plasma levels, phospholipase A2 activity, and the developmental profiles of children with autism vs. control subjects. METHODS Twenty four children with autism underwent laboratory analysis for fatty acid quantification using gas chromatography and PLA2 activity determination by fluorometric assay. RESULTS No correlation was observed between the developmental quotient and fatty acid plasma levels. Phospholipase A2 activity was significantly higher among autistic children compared with controls. CONCLUSION The study did not show a correlation between fatty acid and phospholipase A2 plasma levels and the developmental profile of children with autism.
Collapse
Affiliation(s)
| | - Hudson Caetano Polonini
- Graduate Program in Health Sciences, Núcleo de Pesquisa e Inovação em Ciências da Saúde, UFJF
| | - Rosemeri Mendes
- Graduate Program in Health Sciences, Núcleo de Pesquisa e Inovação em Ciências da Saúde, UFJF
| | | | - Wagner Farid Gattaz
- Departamento e Instituto de Psiquiatria, Laboratório de Neurociências, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Nádia Rezende Barbosa Raposo
- Núcleo de Pesquisa e Inovação em Ciências da Saúde, Universidade Federal de Juiz de Fora, Juiz de Fora, MG, Brazil
| |
Collapse
|
63
|
Zhao G, Gao J, Liang S, Wang X, Sun C, Xia W, Hao Y, Li X, Cao Y, Wu L. Study of the serum levels of polyunsaturated fatty acids and the expression of related liver metabolic enzymes in a rat valproate-induced autism model. Int J Dev Neurosci 2015; 44:14-21. [PMID: 25916973 DOI: 10.1016/j.ijdevneu.2015.04.350] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 04/22/2015] [Accepted: 04/22/2015] [Indexed: 10/23/2022] Open
Abstract
To investigate whether the decreased level of serum polyunsaturated fatty acids (PUFAs) in patients with autism is associated with the expression of related liver metabolic enzymes, we selected rats that were exposed to valproic acid (VPA) on embryonic day 12.5 (E12.5) as a model of autism. We observed the serum levels of PUFAs and the expression of related liver metabolic enzymes, including Δ5-desaturase, Δ6-desaturase and elongase (Elovl2), in VPA-exposed and control rats on postnatal day 35 (PND35) and conducted sex dimorphic analysis. We found that the levels of serum PUFAs and related liver metabolic enzymes in the VPA rats were significantly reduced, in association with autism-like behavioral changes, the abnormal expression of apoptosis-related proteins and hippocampal neuronal injury, compared to the control rats and showed sex difference in VPA group. This finding indicated that rats exposed to VPA at the embryonic stage may exhibit reduced synthesis of serum PUFAs due to the down-regulation of liver metabolic enzymes, thereby inducing nervous system injury and behavioral changes, which is affected by sex in the meantime.
Collapse
Affiliation(s)
- Gang Zhao
- Department of Children's and Adolescent Health, Public Health College of Harbin Medical University, Harbin 150081, China
| | - Jingquan Gao
- Department of Nursing, Daqing Campus of Harbin Medical University, Daqing 163319, China
| | - Shuang Liang
- Department of Children's and Adolescent Health, Public Health College of Harbin Medical University, Harbin 150081, China
| | - Xuelai Wang
- Department of Children's and Adolescent Health, Public Health College of Harbin Medical University, Harbin 150081, China
| | - Caihong Sun
- Department of Children's and Adolescent Health, Public Health College of Harbin Medical University, Harbin 150081, China
| | - Wei Xia
- Department of Children's and Adolescent Health, Public Health College of Harbin Medical University, Harbin 150081, China
| | - Yanqiu Hao
- Department of Pediatric, the Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Xiang Li
- Department of Children's and Adolescent Health, Public Health College of Harbin Medical University, Harbin 150081, China
| | - Yonggang Cao
- Department of Pharmacology, Daqing Campus of Harbin Medical University, Daqing 163319, China.
| | - Lijie Wu
- Department of Children's and Adolescent Health, Public Health College of Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
64
|
Wellmann KA, George F, Brnouti F, Mooney SM. Docosahexaenoic acid partially ameliorates deficits in social behavior and ultrasonic vocalizations caused by prenatal ethanol exposure. Behav Brain Res 2015; 286:201-11. [PMID: 25746516 DOI: 10.1016/j.bbr.2015.02.048] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 02/23/2015] [Accepted: 02/24/2015] [Indexed: 01/17/2023]
Abstract
Prenatal ethanol exposure disrupts social behavior in humans and rodents. One system particularly important for social behavior is the somatosensory system. Prenatal ethanol exposure alters the structure and function of this area. Docosahexaenoic acid (DHA), an omega 3 polyunsaturated fatty acid, is necessary for normal brain development and brains from ethanol-exposed animals are DHA deficient. Thus, we determined whether postnatal DHA supplementation ameliorated behavioral deficits induced by prenatal ethanol exposure. Timed pregnant Long-Evans rats were assigned to one of three groups: ad libitum access to an ethanol-containing liquid diet, pair fed an isocaloric isonutritive non-alcohol liquid diet, or ad libitum access to chow and water. Pups were assigned to one of two postnatal treatment groups; gavaged intragastrically once per day between postnatal day (P)11 and P20 with DHA (10 mg/kg in artificial rat milk) or artificial rat milk. A third group was left untreated. Isolation-induced ultrasonic vocalizations (iUSVs) were recorded on P14. Social behavior and play-induced USVs were tested on P28 or P42. Somatosensory performance was tested with a gap crossing test around P33 or on P42. Anxiety was tested on elevated plus maze around P35. Animals exposed to ethanol prenatally vocalized less, play fought less, and crossed a significantly shorter gap than control-treated animals. Administration of DHA ameliorated these ethanol-induced deficits such that the ethanol-exposed animals given DHA were no longer significantly different to control-treated animals. Thus, DHA administration may have therapeutic value to reverse some of ethanol's damaging effects.
Collapse
Affiliation(s)
- Kristen A Wellmann
- Department of Pediatrics, University of Maryland, Baltimore, MD 21201, United States.
| | - Finney George
- Department of Pediatrics, University of Maryland, Baltimore, MD 21201, United States
| | - Fares Brnouti
- Department of Pediatrics, University of Maryland, Baltimore, MD 21201, United States
| | - Sandra M Mooney
- Department of Pediatrics, University of Maryland, Baltimore, MD 21201, United States.
| |
Collapse
|
65
|
Babchia N, DeAraujo A, Leclère L, Buteau B, Martine L, Grégoire S, Brétillon L. Docosahexaenoic acid modulates oxidative stress-induced apoptosis via PI3K/Akt m-TOR/p70S6K pathways in human RPE cells. EUR J LIPID SCI TECH 2015. [DOI: 10.1002/ejlt.201400155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Narjes Babchia
- CNRS; UMR 6265; Centre des Sciences du Goût et de l'Alimentation; Dijon France
- INRA; UMR 1324; Centre des Sciences du Goût et de l'Alimentation; Dijon France
- Université de Bourgogne; Centre des Sciences du Goût et de l'Alimentation; Dijon France
| | - Aline DeAraujo
- CNRS; UMR 6265; Centre des Sciences du Goût et de l'Alimentation; Dijon France
- INRA; UMR 1324; Centre des Sciences du Goût et de l'Alimentation; Dijon France
- Université de Bourgogne; Centre des Sciences du Goût et de l'Alimentation; Dijon France
| | - Laurent Leclère
- CNRS; UMR 6265; Centre des Sciences du Goût et de l'Alimentation; Dijon France
- INRA; UMR 1324; Centre des Sciences du Goût et de l'Alimentation; Dijon France
- Université de Bourgogne; Centre des Sciences du Goût et de l'Alimentation; Dijon France
| | - Bénédicte Buteau
- CNRS; UMR 6265; Centre des Sciences du Goût et de l'Alimentation; Dijon France
- INRA; UMR 1324; Centre des Sciences du Goût et de l'Alimentation; Dijon France
- Université de Bourgogne; Centre des Sciences du Goût et de l'Alimentation; Dijon France
| | - Lucy Martine
- CNRS; UMR 6265; Centre des Sciences du Goût et de l'Alimentation; Dijon France
- INRA; UMR 1324; Centre des Sciences du Goût et de l'Alimentation; Dijon France
- Université de Bourgogne; Centre des Sciences du Goût et de l'Alimentation; Dijon France
| | - Stéphane Grégoire
- CNRS; UMR 6265; Centre des Sciences du Goût et de l'Alimentation; Dijon France
- INRA; UMR 1324; Centre des Sciences du Goût et de l'Alimentation; Dijon France
- Université de Bourgogne; Centre des Sciences du Goût et de l'Alimentation; Dijon France
| | - Lionel Brétillon
- CNRS; UMR 6265; Centre des Sciences du Goût et de l'Alimentation; Dijon France
- INRA; UMR 1324; Centre des Sciences du Goût et de l'Alimentation; Dijon France
- Université de Bourgogne; Centre des Sciences du Goût et de l'Alimentation; Dijon France
| |
Collapse
|
66
|
Yamashita S, Kanno S, Nakagawa K, Kinoshita M, Miyazawa T. Extrinsic plasmalogens suppress neuronal apoptosis in mouse neuroblastoma Neuro-2A cells: importance of plasmalogen molecular species. RSC Adv 2015. [DOI: 10.1039/c5ra00632e] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Plasmalogen, especially those having 22:6, suppressed neuronal apoptosisviadeath receptor and mitochondrial pathways. These mechanisms of action of plasmalogen may be responsible for regulation of membrane functions and second messenger production.
Collapse
Affiliation(s)
- Shinji Yamashita
- Department of Food Science
- Obihiro University of Agriculture and Veterinary Medicine
- Obihiro
- Japan
- Food and Biodynamic Chemistry Laboratory
| | - Susumu Kanno
- Food and Biodynamic Chemistry Laboratory
- Graduate School of Agricultural Science
- Tohoku University
- Sendai
- Japan
| | - Kiyotaka Nakagawa
- Food and Biodynamic Chemistry Laboratory
- Graduate School of Agricultural Science
- Tohoku University
- Sendai
- Japan
| | - Mikio Kinoshita
- Department of Food Science
- Obihiro University of Agriculture and Veterinary Medicine
- Obihiro
- Japan
| | - Teruo Miyazawa
- Food and Biodynamic Chemistry Laboratory
- Graduate School of Agricultural Science
- Tohoku University
- Sendai
- Japan
| |
Collapse
|
67
|
Docosahexaenoic acid attenuates oxidative stress and protects human gingival fibroblasts against cytotoxicity induced by hydrogen peroxide and butyric acid. Arch Oral Biol 2015; 60:144-53. [DOI: 10.1016/j.archoralbio.2014.09.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 08/14/2014] [Accepted: 09/25/2014] [Indexed: 01/01/2023]
|
68
|
Polyunsaturated fatty acids and their metabolites in brain function and disease. Nat Rev Neurosci 2014; 15:771-85. [PMID: 25387473 DOI: 10.1038/nrn3820] [Citation(s) in RCA: 1029] [Impact Index Per Article: 93.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The brain is highly enriched with fatty acids. These include the polyunsaturated fatty acids (PUFAs) arachidonic acid and docosahexaenoic acid, which are largely esterified to the phospholipid cell membrane. Once PUFAs are released from the membrane, they can participate in signal transduction, either directly or after enzymatic conversion to a variety of bioactive derivatives ('mediators'). PUFAs and their mediators regulate several processes within the brain, such as neurotransmission, cell survival and neuroinflammation, and thereby mood and cognition. PUFA levels and the signalling pathways that they regulate are altered in various neurological disorders, including Alzheimer's disease and major depression. Diet and drugs targeting PUFAs may lead to novel therapeutic approaches for the prevention and treatment of brain disorders.
Collapse
|
69
|
Abstract
With more than 100 studies published over the past two decades, functional brain imaging research in gastroenterology has become an established field; one that has enabled improved insight into the supraspinal responses evoked by gastrointestinal stimulation both in health and disease. However, there remains considerable inter-study variation in the published results, largely owing to methodological differences in stimulation and recording techniques, heterogeneous patient selection, lack of control for psychological factors and so on. These issues with reproducibility, although not unique to studies of the gastrointestinal tract, can lead to unjustified inferences. To obtain consistent and more clinically relevant results, there is a need to optimize and standardize brain imaging studies across different centres. In addition, the use of complementary and more novel brain imaging modalities and analyses, which are now being used in other fields of research, might help unravel the factors at play in functional gastrointestinal disorders. This Review highlights the areas in which functional brain imaging has been useful and what it has revealed, the areas that are in need of improvement, and finally suggestions for future directions.
Collapse
|
70
|
Yokokura Y, Isobe Y, Matsueda S, Iwamoto R, Goto T, Yoshioka T, Urabe D, Inoue M, Arai H, Arita M. Identification of 14,20-dihydroxy-docosahexaenoic acid as a novel anti-inflammatory metabolite. ACTA ACUST UNITED AC 2014; 156:315-21. [DOI: 10.1093/jb/mvu044] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
71
|
Hughbanks-Wheaton DK, Birch DG, Fish GE, Spencer R, Pearson NS, Takacs A, Hoffman DR. Safety assessment of docosahexaenoic acid in X-linked retinitis pigmentosa: the 4-year DHAX trial. Invest Ophthalmol Vis Sci 2014; 55:4958-66. [PMID: 25015354 DOI: 10.1167/iovs.14-14437] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Docosahexaenoic acid (DHA) continues to be evaluated and recommended as treatment and prophylaxis for various diseases. We recently assessed efficacy of high-dose DHA supplementation to slow vision loss in patients with X-linked retinitis pigmentosa (XLRP) in a randomized clinical trial. Because DHA is a highly unsaturated fatty acid, it could serve as a target for free-radical induced oxidation, resulting in increased oxidative stress. Biosafety was monitored during the 4-year trial to determine whether DHA supplementation was associated with identifiable risks. METHODS Males (n = 78; 7-31 years) meeting entry criteria were enrolled. The modified intent-to-treat cohort (DHA = 33; placebo = 27) adhered to the protocol ≥ 1 year. Participants were randomized to an oral dose of 30 mg/kg/d DHA or placebo plus a daily multivitamin. Comprehensive metabolic analyses were assessed for group differences. Treatment-emergent adverse events including blood chemistry metabolites were recorded. RESULTS By year 4, supplementation elevated plasma and red blood cell-DHA 4.4- and 3.6-fold, respectively, compared with the placebo group (P < 0.00001). Over the trial duration, no significant differences between DHA and placebo groups were found for vitamin A, vitamin E, platelet aggregation, antioxidant activity, lipoprotein cholesterol, or oxidized LDL levels (all P > 0.14). Adverse events were transient and not considered severe (e.g., gastrointestinal [GI] irritability, blood chemistry alterations). One participant was unable to tolerate persistent GI discomfort. CONCLUSIONS Long-term, high-dose DHA supplementation to patients with XLRP was associated with limited safety risks in this 4-year trial. Nevertheless, GI symptoms should be monitored in all patients taking high dose DHA especially those with personal or family history of GI disturbances. (ClinicalTrials.gov number, NCT00100230.).
Collapse
Affiliation(s)
- Dianna K Hughbanks-Wheaton
- Retina Foundation of the Southwest, Dallas, Texas, United States Department of Ophthalmology, UT Southwestern Medical Center, Dallas, Texas, United States
| | - David G Birch
- Retina Foundation of the Southwest, Dallas, Texas, United States Department of Ophthalmology, UT Southwestern Medical Center, Dallas, Texas, United States
| | - Gary E Fish
- Texas Retina Associates, Dallas, Texas, United States
| | - Rand Spencer
- Texas Retina Associates, Dallas, Texas, United States
| | - N Shirlene Pearson
- Pearson Statistical Consulting & Expert Testimony, Richardson, Texas, United States
| | - Alison Takacs
- Retina Foundation of the Southwest, Dallas, Texas, United States
| | - Dennis R Hoffman
- Retina Foundation of the Southwest, Dallas, Texas, United States Department of Ophthalmology, UT Southwestern Medical Center, Dallas, Texas, United States
| |
Collapse
|
72
|
Kim HY, Huang BX, Spector AA. Phosphatidylserine in the brain: metabolism and function. Prog Lipid Res 2014; 56:1-18. [PMID: 24992464 DOI: 10.1016/j.plipres.2014.06.002] [Citation(s) in RCA: 238] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 06/18/2014] [Accepted: 06/21/2014] [Indexed: 01/08/2023]
Abstract
Phosphatidylserine (PS) is the major anionic phospholipid class particularly enriched in the inner leaflet of the plasma membrane in neural tissues. PS is synthesized from phosphatidylcholine or phosphatidylethanolamine by exchanging the base head group with serine, and this reaction is catalyzed by phosphatidylserine synthase 1 and phosphatidylserine synthase 2 located in the endoplasmic reticulum. Activation of Akt, Raf-1 and protein kinase C signaling, which supports neuronal survival and differentiation, requires interaction of these proteins with PS localized in the cytoplasmic leaflet of the plasma membrane. Furthermore, neurotransmitter release by exocytosis and a number of synaptic receptors and proteins are modulated by PS present in the neuronal membranes. Brain is highly enriched with docosahexaenoic acid (DHA), and brain PS has a high DHA content. By promoting PS synthesis, DHA can uniquely expand the PS pool in neuronal membranes and thereby influence PS-dependent signaling and protein function. Ethanol decreases DHA-promoted PS synthesis and accumulation in neurons, which may contribute to the deleterious effects of ethanol intake. Improvement of some memory functions has been observed in cognitively impaired subjects as a result of PS supplementation, but the mechanism is unclear.
Collapse
Affiliation(s)
- Hee-Yong Kim
- Laboratory of Molecular Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892-9410, United States.
| | - Bill X Huang
- Laboratory of Molecular Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892-9410, United States
| | - Arthur A Spector
- Laboratory of Molecular Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892-9410, United States
| |
Collapse
|
73
|
Pinsolle A, Roy P, Cansell M. Modulation of enzymatic PS synthesis by liposome membrane composition. Colloids Surf B Biointerfaces 2014; 115:157-63. [DOI: 10.1016/j.colsurfb.2013.11.036] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 10/29/2013] [Accepted: 11/18/2013] [Indexed: 11/30/2022]
|
74
|
Marin R, Casañas V, Pérez JA, Fabelo N, Fernandez CE, Diaz M. Oestrogens as modulators of neuronal signalosomes and brain lipid homeostasis related to protection against neurodegeneration. J Neuroendocrinol 2013; 25:1104-15. [PMID: 23795744 DOI: 10.1111/jne.12068] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 05/22/2013] [Accepted: 06/18/2013] [Indexed: 12/19/2022]
Abstract
Oestrogens trigger several pathways at the plasma membrane that exert beneficial actions against neurodegenerative diseases, such as Alzheimer's disease and Parkinson's disease. Part of these actions takes place in lipid rafts, which are membrane domains with a singular protein and lipid composition. These microdomains also represent a preferential site for signalling protein complexes, or signalosomes. A plausible hypothesis is that the dynamic interaction of signalosomes with different extracellular ligands may be at the basis of neuronal maintenance against different neuropathologies. Oestrogen receptors are localised in neuronal lipid rafts, taking part of macromolecular complexes together with a voltage-dependent anion channel (VDAC), and other molecules. Oestradiol binding to its receptor at this level enhances neuroprotection against amyloid-β degeneration through the activation of different signal transduction pathways, including VDAC gating modulation. Moreover, part of the stability and functionality of signalling platforms lays on the distribution of lipid hallmarks in these microstructures, which modulate membrane physicochemical properties, thus favouring molecular interactions. Interestingly, recent findings indicate a potential role of oestrogens in the preservation of neuronal membrane physiology related to lipid homeostasis. Thus, oestrogens and docosahexaenoic acid may act synergistically to stabilise brain lipid structure by regulating neuronal lipid biosynthetic pathways, suggesting that part of the neuroprotective effects elicited by oestrogens occur through mechanisms aimed at preserving lipid homeostasis. Overall, oestrogen mechanisms of neuroprotection may occur not only by its interaction with neuronal protein targets through nongenomic and genomic mechanisms, but also through its participation in membrane architecture stabilisation via 'lipostatic' mechanisms.
Collapse
Affiliation(s)
- R Marin
- Department of Physiology, Laboratory of Cellular Neurobiology, University of La Laguna, La Laguna, Tenerife, Spain
| | | | | | | | | | | |
Collapse
|
75
|
Murphy EJ. A lipid neurochemist's siren: docosahexaenoic acid and its elusive function in the central nervous system. J Neurochem 2013; 127:299-302. [PMID: 24117623 DOI: 10.1111/jnc.12439] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 08/26/2013] [Indexed: 11/28/2022]
Affiliation(s)
- Eric J Murphy
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, USA
| |
Collapse
|
76
|
Barceló-Coblijn G, Wold LE, Ren J, Murphy EJ. Prenatal ethanol exposure increases brain cholesterol content in adult rats. Lipids 2013; 48:1059-68. [PMID: 23996454 DOI: 10.1007/s11745-013-3821-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 07/17/2013] [Indexed: 11/29/2022]
Abstract
Fetal alcohol syndrome is the most severe expression of the fetal alcohol spectrum disorders (FASD). Although alterations in fetal and neonate brain fatty acid composition and cholesterol content are known to occur in animal models of FASD, the persistence of these alterations into adulthood is unknown. To address this question, we determined the effect of prenatal ethanol exposure on individual phospholipid class fatty acid composition, individual phospholipid class mass, and cholesterol mass in brains from 25-week-old rats that were exposed to ethanol during gestation beginning at gestational day 2. While total phospholipid mass was unaffected, phosphatidylinositol and cardiolipin mass was decreased 14 and 43 %, respectively. Exposure to prenatal ethanol modestly altered brain phospholipid fatty acid composition, and the most consistent change was a significant 1.1-fold increase in total polyunsaturated fatty acids (PUFA), in the n-3/n-6 ratio, and in the 22:6n-3 content in ethanolamine glycerophospholipids and in phosphatidylserine. In contrast, prenatal ethanol consumption significantly increased brain cholesterol mass 1.4-fold and the phospholipid to cholesterol ratio was significantly increased 1.3-fold. These results indicate that brain cholesterol mass was significantly increased in adult rats exposed prenatally to ethanol, but changes in phospholipid mass and phospholipid fatty acid composition were extremely limited. Importantly, suppression of postnatal ethanol consumption was not sufficient to reverse the large increase in cholesterol observed in the adult rats.
Collapse
Affiliation(s)
- Gwendolyn Barceló-Coblijn
- Department of Pharmacology, Physiology, and Therapeutics, School of Medicine and Health Sciences, University of North Dakota, 501 N. Columbia Rd, Room 3700, Grand Forks, ND, 58202-9037, USA
| | | | | | | |
Collapse
|
77
|
German OL, Monaco S, Agnolazza DL, Rotstein NP, Politi LE. Retinoid X receptor activation is essential for docosahexaenoic acid protection of retina photoreceptors. J Lipid Res 2013; 54:2236-2246. [PMID: 23723389 PMCID: PMC3708373 DOI: 10.1194/jlr.m039040] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 05/28/2013] [Indexed: 01/12/2023] Open
Abstract
We have established that docosahexaenoic acid (DHA), the major polyunsaturated fatty acid in the retina, promotes survival of rat retina photoreceptors during early development in vitro and upon oxidative stress by activating the ERK/MAPK signaling pathway. Here we have investigated whether DHA turns on this pathway through activation of retinoid X receptors (RXRs) or by inducing tyrosine kinase (Trk) receptor activation. We also evaluated whether DHA release from phospholipids was required for its protective effect. Addition of RXR antagonists (HX531, PA452) to rat retinal neuronal cultures inhibited DHA protection during early development in vitro and upon oxidative stress induced with Paraquat or H2O2. In contrast, the Trk inhibitor K252a did not affect DHA prevention of photoreceptor apoptosis. These results imply that activation of RXRs was required for DHA protection whereas Trk receptors were not involved in this protection. Pretreatment with 4-bromoenol lactone, a phospholipase A2 inhibitor, blocked DHA prevention of oxidative stress-induced apoptosis of photoreceptors. It is noteworthy that RXR agonists (HX630, PA024) also rescued photoreceptors from H2O2-induced apoptosis. These results provide the first evidence that activation of RXRs prevents photoreceptor apoptosis and suggest that DHA is first released from phospholipids and then activates RXRs to promote the survival of photoreceptors.
Collapse
Affiliation(s)
- Olga L German
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Universidad Nacional del Sur-CONICET, Buenos Aires, Argentina
| | - Sandra Monaco
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Universidad Nacional del Sur-CONICET, Buenos Aires, Argentina
| | - Daniela L Agnolazza
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Universidad Nacional del Sur-CONICET, Buenos Aires, Argentina
| | - Nora P Rotstein
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Universidad Nacional del Sur-CONICET, Buenos Aires, Argentina.
| | - Luis E Politi
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Universidad Nacional del Sur-CONICET, Buenos Aires, Argentina.
| |
Collapse
|
78
|
Kennedy DO, Jackson PA, Elliott JM, Scholey AB, Robertson BC, Greer J, Tiplady B, Buchanan T, Haskell CF. Cognitive and mood effects of 8 weeks' supplementation with 400 mg or 1000 mg of the omega-3 essential fatty acid docosahexaenoic acid (DHA) in healthy children aged 10–12 years. Nutr Neurosci 2013; 12:48-56. [DOI: 10.1179/147683009x388887] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
79
|
Hasadsri L, Wang BH, Lee JV, Erdman JW, Llano DA, Barbey AK, Wszalek T, Sharrock MF, Wang H(J. Omega-3 Fatty Acids as a Putative Treatment for Traumatic Brain Injury. J Neurotrauma 2013; 30:897-906. [DOI: 10.1089/neu.2012.2672] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Affiliation(s)
- Linda Hasadsri
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Bonnie H. Wang
- Department of Internal Medicine, University of Illinois College of Medicine at Urbana-Champaign, Urbana, Illinois
| | - James V. Lee
- Department of Internal Medicine, University of Illinois College of Medicine at Urbana-Champaign, Urbana, Illinois
| | - John W. Erdman
- Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, Urbana, Illinois
| | - Daniel A. Llano
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, Illinois
| | - Aron K. Barbey
- Department of Internal Medicine, University of Illinois College of Medicine at Urbana-Champaign, Urbana, Illinois
- Department of Psychology, University of Illinois Urbana-Champaign, Urbana, Illinois
- Department of Speech and Hearing Science, University of Illinois Urbana-Champaign, Urbana, Illinois
| | - Tracey Wszalek
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, Illinois
| | - Matthew F. Sharrock
- Department of Internal Medicine, University of Illinois College of Medicine at Urbana-Champaign, Urbana, Illinois
| | - Huan (John) Wang
- Department of Neurosurgery, University of Illinois College of Medicine at Urbana-Champaign, Urbana, Illinois
| |
Collapse
|
80
|
Rashid MA, Katakura M, Kharebava G, Kevala K, Kim HY. N-Docosahexaenoylethanolamine is a potent neurogenic factor for neural stem cell differentiation. J Neurochem 2013; 125:869-84. [PMID: 23570577 DOI: 10.1111/jnc.12255] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 03/29/2013] [Accepted: 03/30/2013] [Indexed: 01/13/2023]
Abstract
Docosahexaenoic acid (DHA) has been shown to promote neuronal differentiation of neural stem cells (NSCs) in vivo and in vitro. Previously, we found that N-docosahexaenoylethanolamine (synaptamide), an endogenous DHA metabolite with an endocannabinoid-like structure, promotes neurite growth, synaptogenesis, and synaptic function. In this study, we demonstrate that synaptamide potently induces neuronal differentiation of NSCs. Differentiating NSCs were capable of synthesizing synaptamide from DHA. Treatment of NSCs with synaptamide at low nanomolar concentrations significantly increased the number of MAP2 and Tuj-1-positive neurons with concomitant induction of protein kinase A (PKA)/cAMP response element binding protein (CREB) phosphorylation. Conversely, PKA inhibitors or PKA knockdown abolished the synaptamide-induced neuronal differentiation of NSCs. URB597, a fatty acid amide hydrolase (FAAH) inhibitor, elevated the level of DHA-derived synaptamide and further potentiated the DHA- or synaptamide-induced neuronal differentiation of NSCs. Similarly, NSCs obtained from FAAH KO mice exhibited greater capacity to induce neuronal differentiation in response to DHA or synaptamide compared to the wild type NSCs. Neither synaptamide nor DHA affected NSC differentiation into GFAP-positive glia cells. These results suggest that endogenously produced synaptamide is a potent mediator for neurogenic differentiation of NSCs acting through PKA/CREB activation.
Collapse
Affiliation(s)
- Mohammad A Rashid
- Laboratory of Molecular Signaling, DICBR, NIAAA, NIH, Bethesda, Maryland 20892-9410, USA
| | | | | | | | | |
Collapse
|
81
|
Nutritional modulation of cognitive function and mental health. J Nutr Biochem 2013; 24:725-43. [DOI: 10.1016/j.jnutbio.2013.01.002] [Citation(s) in RCA: 181] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 01/11/2013] [Accepted: 01/14/2013] [Indexed: 12/30/2022]
|
82
|
Experimental evidence of ω-3 polyunsaturated fatty acid modulation of inflammatory cytokines and bioactive lipid mediators: their potential role in inflammatory, neurodegenerative, and neoplastic diseases. BIOMED RESEARCH INTERNATIONAL 2013; 2013:743171. [PMID: 23691510 PMCID: PMC3652138 DOI: 10.1155/2013/743171] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 03/27/2013] [Accepted: 03/27/2013] [Indexed: 12/19/2022]
Abstract
A large body of evidence has emerged over the past years to show the critical role played by inflammation in the pathogenesis of several diseases including some cardiovascular, neoplastic, and neurodegenerative diseases, previously not considered inflammation-related. The anti-inflammatory action of ω-3 polyunsaturated fatty acids (PUFAs), as well as their potential healthy effects against the development and progression of the same diseases, has been widely studied by our and others' laboratories. As a result, a rethinking is taking place on the possible mechanisms underlying the beneficial effects of ω-3 PUFAs against these disorders, and, in particular, on the influence that they may exert on the molecular pathways involved in inflammatory process, including the production of inflammatory cytokines and lipid mediators active in the resolving phase of inflammation. In the present review we will summarize and discuss the current knowledge regarding the modulating effects of ω-3 PUFAs on the production of inflammatory cytokines and proresolving or protective lipid mediators in the context of inflammatory, metabolic, neurodegenerative, and neoplastic diseases.
Collapse
|
83
|
Das UN. Polyunsaturated fatty acids and their metabolites in the pathobiology of schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2013; 42:122-34. [PMID: 22735394 DOI: 10.1016/j.pnpbp.2012.06.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2011] [Revised: 05/27/2012] [Accepted: 06/13/2012] [Indexed: 01/01/2023]
Abstract
Schizophrenia can be considered as a low-grade systemic inflammatory disease with its origins in the perinatal period. It is likely that genetic, environmental, and nutritional factors interact to induce excess production of pro-inflammatory cytokines that, in turn, damage fetal neurons leading to the adult onset of schizophrenia. Polyunsaturated fatty acids (PUFAs) and their metabolites such as lipoxins, resolvins, protectins, maresins and nitrolipids not only have potent neuroprotective action but also are capable of inhibiting the production of pro-inflammatory cytokines. Decreased formation of PUFAs as a result of low activity of Δ(6) and Δ(5) desaturases can result in an increase in the production of pro-inflammatory cytokines due to the absence of negative control exerted by PUFAs and their anti-inflammatory metabolites that, in turn, may predispose to neuronal damage and development of schizophrenia in adult life. Furthermore, PUFAs are essential for brain growth and development. If this proposal is correct, this implies that perinatal and adult supplementation of PUFAs not only prevents but also helps in the treatment of schizophrenia. Furthermore, synthetic analogs of lipoxins, resolvins, and protectins may be of significant benefit in schizophrenia.
Collapse
Affiliation(s)
- Undurti N Das
- UND Life Sciences, 13800 Fairhill Road, Shaker Heights, OH 44120, USA.
| |
Collapse
|
84
|
Hamazaki K, Kim HY. Differential modification of the phospholipid profile by transient ischemia in rat hippocampal CA1 and CA3 regions. Prostaglandins Leukot Essent Fatty Acids 2013; 88:299-306. [PMID: 23395327 PMCID: PMC3622766 DOI: 10.1016/j.plefa.2013.01.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 01/04/2013] [Accepted: 01/05/2013] [Indexed: 01/11/2023]
Abstract
The hippocampal CA1 region is most susceptible to cerebral ischemia in both rodents and humans, whereas CA3 is remarkably resistant. Here, we investigated the possible role of membrane lipids in differential susceptibility in these regions. Transient ischemia was induced in rats via bilateral occlusion of common carotid arteries and membrane lipids were analyzed by mass spectrometry. While lipid profile differences between the intact CA1 and CA3 were rather minor, ischemia caused significant pyramidal cell death with concomittant reduction of phosphatidylserine, phosphatidylinositol, phosphatidylethanolamine, plasmalogen and sphingomyelin only in CA1. The phospholipid loss was evenly distributed in most molecular species. Ischemia also significantly increased cell death mediator ceramides only in CA1. Our data suggests that differential susceptibility to ischemia between CA1 and CA3 is not linked to their unique phospholipid profile. Also, selective activation of phospholipase A2, which primarily releases polyunsaturated fatty acids, might not be characteristic to cell death in CA1.
Collapse
Affiliation(s)
- Kei Hamazaki
- Laboratory of Molecular Signaling, Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health,5625 Fishers Lane, Room 3N-07, Bethesda, MD 20892-9410, United States
| | | |
Collapse
|
85
|
Lim SN, Huang W, Hall JC, Michael-Titus AT, Priestley JV. Improved outcome after spinal cord compression injury in mice treated with docosahexaenoic acid. Exp Neurol 2013; 239:13-27. [DOI: 10.1016/j.expneurol.2012.09.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 09/20/2012] [Accepted: 09/24/2012] [Indexed: 12/20/2022]
|
86
|
Lewis M, Ghassemi P, Hibbeln J. Therapeutic use of omega-3 fatty acids in severe head trauma. Am J Emerg Med 2013; 31:273.e5-8. [PMID: 22867826 PMCID: PMC3518659 DOI: 10.1016/j.ajem.2012.05.014] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 05/15/2012] [Accepted: 05/19/2012] [Indexed: 11/20/2022] Open
Abstract
Traumatic brain injury (TBI) has long been recognized as the leading cause of traumatic death and disability. Tremendous advances in surgical and intensive care unit management of the primary injury, including maintaining adequate oxygenation, controlling intracranial pressure, and ensuring proper cerebral perfusion pressure, have resulted in reduced mortality. However, the secondary injury phase of TBI is a prolonged pathogenic process characterized by neuroinflammation, excitatory amino acids, free radicals, and ion imbalance. There are no approved therapies to directly address these underlying processes. Here, we present a case that was intentionally treated with substantial amounts of omega-3 fatty acids (n-3FA) to provide the nutritional foundation for the brain to begin the healing process following severe TBI. Recent animal research supports the use of n-3FA, and clinical experience suggests that benefits may be possible from substantially and aggressively adding n-3FA to optimize the nutritional foundation of severe TBI patients and must be in place if the brain is to be given the opportunity to repair itself to the best possible extent. Administration early in the course of treatment, in the emergency department or sooner, has the potential to improve outcomes from this potentially devastating public health problem.
Collapse
Affiliation(s)
- Michael Lewis
- Brain Health Education and Research Foundation Arlington, VA
| | | | - Joseph Hibbeln
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, 3N-07, Rockville, MD 20852
| |
Collapse
|
87
|
The follicular microenviroment as a predictor of pregnancy: MALDI-TOF MS lipid profile in cumulus cells. J Assist Reprod Genet 2012; 29:1289-97. [PMID: 22968515 DOI: 10.1007/s10815-012-9859-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Accepted: 08/29/2012] [Indexed: 02/06/2023] Open
Abstract
PURPOSE This research proposed to study the changes in lipid composition in cumulus cells (CCs) from women who achieved pregnancy compared with women who did not, after in vitro fertilization treatment. This approach has the potential to provide novel information on the lipid metabolism of the CCs and as an additional method to predict pregnancy. METHOD Fifty-four samples from couples with tubal and male factor infertility and where the female partner was age 35 or younger were divided in two groups according to their level of hCG 14 days after embryo transfer as follows: (1) 23 samples in pregnant group and (2) 31 samples in non-pregnant group. Lipid extraction was performed by the Bligh-Dyer protocol, and lipid profiles were obtained by MALDI-TOF MS. Mass spectra data were processed with MassLynx, and statistical analysis was performed using MarkerLynx extended statistic. OPLS-DA model was built. RESULTS S-plot Analysis revealed three ions as potential markers in the pregnant group, and five ions in the non-pregnant group. These ions were identified in the human metabolome database (HMDB) as phosphatidylcholine in the pregnant group and as phosphatidylethanolamine, phosphatidylserine and phosphatidylinositol species in the non-pregnant group. These lipids might be involved in cell proliferation and differentiation, apoptosis and GAP junction regulation. CONCLUSION We conclude that MALDI-TOF MS can be used as an informative and fast analytical strategy to obtain and study the lipid profile of cumulus cells and can potentially be used as a supporting tool to predict pregnancy based on the metabolic state of the CCs.
Collapse
|
88
|
|
89
|
Bazan NG, Molina MF, Gordon WC. Docosahexaenoic acid signalolipidomics in nutrition: significance in aging, neuroinflammation, macular degeneration, Alzheimer's, and other neurodegenerative diseases. Annu Rev Nutr 2011; 31:321-51. [PMID: 21756134 DOI: 10.1146/annurev.nutr.012809.104635] [Citation(s) in RCA: 319] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Essential polyunsaturated fatty acids (PUFAs) are critical nutritional lipids that must be obtained from the diet to sustain homeostasis. Omega-3 and -6 PUFAs are key components of biomembranes and play important roles in cell integrity, development, maintenance, and function. The essential omega-3 fatty acid family member docosahexaenoic acid (DHA) is avidly retained and uniquely concentrated in the nervous system, particularly in photoreceptors and synaptic membranes. DHA plays a key role in vision, neuroprotection, successful aging, memory, and other functions. In addition, DHA displays anti-inflammatory and inflammatory resolving properties in contrast to the proinflammatory actions of several members of the omega-6 PUFAs family. This review discusses DHA signalolipidomics, comprising the cellular/tissue organization of DHA uptake, its distribution among cellular compartments, the organization and function of membrane domains rich in DHA-containing phospholipids, and the cellular and molecular events revealed by the uncovering of signaling pathways regulated by DHA and docosanoids, the DHA-derived bioactive lipids, which include neuroprotectin D1 (NPD1), a novel DHA-derived stereoselective mediator. NPD1 synthesis agonists include neurotrophins and oxidative stress; NPD1 elicits potent anti-inflammatory actions and prohomeostatic bioactivity, is anti-angiogenic, promotes corneal nerve regeneration, and induces cell survival. In the context of DHA signalolipidomics, this review highlights aging and the evolving studies on the significance of DHA in Alzheimer's disease, macular degeneration, Parkinson's disease, and other brain disorders. DHA signalolipidomics in the nervous system offers emerging targets for pharmaceutical intervention and clinical translation.
Collapse
Affiliation(s)
- Nicolas G Bazan
- Neuroscience Center of Excellence and Department of Ophthalmology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.
| | | | | |
Collapse
|
90
|
Kim HY, Spector AA, Xiong ZM. A synaptogenic amide N-docosahexaenoylethanolamide promotes hippocampal development. Prostaglandins Other Lipid Mediat 2011; 96:114-20. [PMID: 21810478 PMCID: PMC3215906 DOI: 10.1016/j.prostaglandins.2011.07.002] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 06/16/2011] [Accepted: 07/07/2011] [Indexed: 01/05/2023]
Abstract
Docosahexaenoic acid (DHA), the n-3 essential fatty acid that is highly enriched in the brain, increases neurite growth and synaptogenesis in cultured mouse fetal hippocampal neurons. These cellular effects may underlie the DHA-induced enhancement of hippocampus-dependent learning and memory functions. We found that N-docsahexaenoylethanolamide (DEA), an ethanolamide derivative of DHA, is a potent mediator for these actions. This is supported by the observation that DHA is converted to DEA by fetal mouse hippocampal neuron cultures and a hippocampal homogenate, and DEA is present endogenously in the mouse hippocampus. Furthermore, DEA stimulates neurite growth and synaptogenesis at substantially lower concentrations than DHA, and it enhances glutamatergic synaptic activities with concomitant increases in synapsin and glutamate receptor subunit expression in the hippocampal neurons. These findings suggest that DEA, an ethanolamide derivative of DHA, is a synaptogenic factor, and therefore we suggest utilizing the term 'synaptamide'. This brief review summarizes the neuronal production and actions of synaptamide and describes other N-docosahexaenoyl amides that are present in the brain.
Collapse
Affiliation(s)
- Hee-Yong Kim
- Laboratory of Molecular Signaling, National Institute of Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892-9410, USA.
| | | | | |
Collapse
|
91
|
Collins MA, Neafsey EJ. Neuroinflammatory pathways in binge alcohol-induced neuronal degeneration: oxidative stress cascade involving aquaporin, brain edema, and phospholipase A2 activation. Neurotox Res 2011; 21:70-8. [PMID: 21927955 DOI: 10.1007/s12640-011-9276-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Revised: 09/01/2011] [Accepted: 09/05/2011] [Indexed: 12/21/2022]
Abstract
Chronic binge alcohol exposure in adult rat models causes neuronal degeneration in the cortex and hippocampus that is not reduced by excitotoxic receptor antagonists, but is prevented by antioxidants. Neuroinflammatory (glial-neuronal) signaling pathways are believed to underlie the oxidative stress and brain damage. Based on our experimental results as well as increased knowledge about the pro-neuroinflammatory potential of glial water channels, we propose that induction of aquaporin-4 can be a critical initiating factor in alcohol's neurotoxic effects, through the instigation of cellular edema-based neuroinflammatory cascades involving increased phospholipase A2 activities, polyunsaturated fatty acid release/membrane depletion, decreased prosurvival signaling, and oxidative stress. A testable scheme for this pathway is presented that incorporates recent findings in the alcohol-brain literature indicating a role for neuroimmune activation (upregulation of NF-kappaB, proinflammatory cytokines, and toll-like receptors). We present the argument that such neuroimmune activation could be associated with or even dependent on increased aquaporin-4 and glial swelling as well.
Collapse
Affiliation(s)
- Michael A Collins
- Department of Molecular Pharmacology & Therapeutics, Stritch School of Medicine, Loyola University Chicago, 2160 South First Avenue, Maywood, IL 60153, USA.
| | | |
Collapse
|
92
|
Pascoe MC, Crewther SG, Carey LM, Crewther DP. What you eat is what you are – A role for polyunsaturated fatty acids in neuroinflammation induced depression? Clin Nutr 2011; 30:407-15. [DOI: 10.1016/j.clnu.2011.03.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Accepted: 03/27/2011] [Indexed: 01/17/2023]
|
93
|
Liu X, Shibata T, Hisaka S, Kawai Y, Osawa T. DHA Hydroperoxides as a Potential Inducer of Neuronal Cell Death: a Mitochondrial Dysfunction-Mediated Pathway. J Clin Biochem Nutr 2011; 43:26-33. [PMID: 18648656 PMCID: PMC2459249 DOI: 10.3164/jcbn.2008040] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2007] [Accepted: 01/29/2008] [Indexed: 01/24/2023] Open
Abstract
During the lipid peroxidation reaction, lipid hydroperoxides are formed as primary products. Several lines of evidence suggest that lipid hydroperoxides can trigger cell death in many cell types, including neurons. In a screening of lipid hydroperoxides which can induce toxicity in neuronal cells, we found docosahexaenoic acid hydroperoxides (DHA-OOH) induced much severe levels of reactive oxygen species generation and cell death in human neuroblastoma SH-SY5Y cells compared to the hydroperoxides of linoleic acid and arachidonic acid. Therefore, we focused on DHA-OOH, and demonstrated that DHA-OOH apparently induced an apoptosis in the neuronal cells through several apoptotic hallmarks including nuclei condensation, DNA fragmentation, poly (ADP-ribose) polymerase cleavage and increased activity of caspase-3. We also found the signaling changes in mitochondria-mediated apoptosis, such as cytochrome c release and increased expression of Bcl-2, as well as a dose-dependent attenuation of mitochondrial membrane potential in the DHA-OOH treated cells. These data indicated DHA hydroperoxide as a potential inducer of apoptosis in human neuroblastoma SH-SY5Y cells, which may be mediated by mitochondria dysfunction pathway.
Collapse
Affiliation(s)
- Xuebo Liu
- Laboratory of Food and Biodynamics, Graduate School of Bioagricultural Science, Nagoya University, Nagoya 464-8601, Japan
| | | | | | | | | |
Collapse
|
94
|
El-Ansary AK, Bacha AGB, Al-Ayahdi LY. Plasma fatty acids as diagnostic markers in autistic patients from Saudi Arabia. Lipids Health Dis 2011; 10:62. [PMID: 21510882 PMCID: PMC3107800 DOI: 10.1186/1476-511x-10-62] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2011] [Accepted: 04/21/2011] [Indexed: 11/10/2022] Open
Abstract
Backgrounds Autism is a family of developmental disorders of unknown origin. The disorder is characterized by behavioral, developmental, neuropathological and sensory abnormalities, and is usually diagnosed between the ages of 2 and 10 with peak prevalence rates observed in children aged 5-8 years. Recently, there has been heightened interest in the role of plasma free fatty acids (FA) in the pathology of neurological disorders. The aim of this study is to compare plasma fatty acid profiles of Saudi autistic patients with those of age-matching control subjects in an attempt to clarify the role of FA in the etiology of autism. Methods 26 autistic patients together with 26-age-matching controls were enrolled in the present study. Methyl esters of FA were extracted with hexane, and the fatty acid composition of the extract was analyzed on a gas chromatography. Results The obtained data proved that fatty acids are altered in the plasma of autistic patients, specifically showing an increase in most of the saturated fatty acids except for propionic acid, and a decrease in most of polyunsaturated fatty acids. The altered fatty acid profile was discussed in relation to oxidative stress, mitochondrial dysfunction and the high lead (Pb) concentration previously reported in Saudi autistic patients. Statistical analysis of the obtained data shows that most of the measured fatty acids were significantly different in autistic patients compared to age -matching controls. Conclusions Receiver Operating Characteristic (ROC) curve analysis shows satisfactory values of area under the curve (AUC) which could reflect the high degree of specificity and sensitivity of the altered fatty acids as biomarkers in autistic patients from Saudi Arabia.
Collapse
Affiliation(s)
- Afaf K El-Ansary
- Biochemistry Department, Science College, King Saud University, P,O Box 22452, Zip Code 11495, Riyadh, Saudi Arabia.
| | | | | |
Collapse
|
95
|
Huang BX, Akbar M, Kevala K, Kim HY. Phosphatidylserine is a critical modulator for Akt activation. J Cell Biol 2011; 192:979-92. [PMID: 21402788 PMCID: PMC3063130 DOI: 10.1083/jcb.201005100] [Citation(s) in RCA: 132] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Accepted: 02/17/2011] [Indexed: 12/25/2022] Open
Abstract
Akt activation relies on the binding of Akt to phosphatidylinositol-3,4,5-trisphosphate (PIP(3)) in the membrane. Here, we demonstrate that Akt activation requires not only PIP(3) but also membrane phosphatidylserine (PS). The extent of insulin-like growth factor-induced Akt activation and downstream signaling as well as cell survival under serum starvation conditions positively correlates with plasma membrane PS levels in living cells. PS promotes Akt-PIP(3) binding, participates in PIP(3)-induced Akt interdomain conformational changes for T308 phosphorylation, and causes an open conformation that allows for S473 phosphorylation by mTORC2. PS interacts with specific residues in the pleckstrin homology (PH) and regulatory (RD) domains of Akt. Disruption of PS-Akt interaction by mutation impairs Akt signaling and increases susceptibility to cell death. These data identify a critical function of PS for Akt activation and cell survival, particularly in conditions with limited PIP(3) availability. The novel molecular interaction mechanism for Akt activation suggests potential new targets for controlling Akt-dependent cell survival and proliferation.
Collapse
Affiliation(s)
- Bill X Huang
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
96
|
Cross-cultural issues of global significance. DEMENTIA 2010. [DOI: 10.1017/cbo9780511780615.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
97
|
Ma D, Zhang M, Mori Y, Yao C, Larsen CP, Yamashima T, Zhou L. Cellular localization of epidermal-type and brain-type fatty acid-binding proteins in adult hippocampus and their response to cerebral ischemia. Hippocampus 2010; 20:811-9. [PMID: 19623607 DOI: 10.1002/hipo.20682] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
This study aimed at an analysis of expression of epidermal-type and brain-type fatty acid-binding proteins (E-FABP and B-FABP, also called FABP5 and FABP7, respectively) in adult hippocampus and their potential value as neuroprotective factors after ischemic brain damage in monkey model. The immunostaining and Western blotting results show that FABP5 was mainly expressed in neurons, whereas FABP7 was primarily expressed in astrocytes and progenitors of the subgranular zone (SGZ). Interestingly, FABP5 expression in neurons increased in cornu Ammonis 1 (CA1) and remains stable within dentate gyrus (DG) after ischemia; FABP7 expression increased within both CA1 and SGZ. This indicates a potential role for FABP5 and FABP7 in intracellular fatty acid transport within different neural cells. The change in FABP5-7 expression within CA1 and DG of the adult postischemic hippocampus was compatible with previous findings of downregulation in CA1 neurons and upregulation in SGZ progenitor cells after ischemia. Altogether, the present data suggest that polyunsaturated fatty acids, such as docosahexaenoic acid, may act via FABP5 or 7 to regulate adult postischemic hippocampal neuronal antiapoptosis or neurogenesis in primates.
Collapse
Affiliation(s)
- Dexuan Ma
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
98
|
Chen P, Véricel E, Lagarde M, Guichardant M. Poxytrins, a class of oxygenated products from polyunsaturated fatty acids, potently inhibit blood platelet aggregation. FASEB J 2010; 25:382-8. [PMID: 20833872 DOI: 10.1096/fj.10-161836] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Docosahexaenoic acid (DHA), an important component of marine lipids, exhibits anti-inflammatory activity related to some of its oxygenated metabolites, such as neuroprotectin/protectin D1 [NPD1/PD1; 10(R),17(S)-dihydroxy-docosa-4Z,7Z, 11E,13E,15Z,19Z-hexaenoic acid] produced through the 15-lipoxygenase pathway. However, other metabolites from DHA can be produced through this pathway, and other polyunsaturated fatty acids (PUFAs) of nutritional value may be oxygenated as well. Their biological activities remain unknown. Isomers of protectin D1 were synthesized using soybean lipoxygenase and tested for their ability to inhibit human blood platelet aggregation. A geometric isomer called PDX, previously described with the 11E,13Z,15E geometry, instead of 11E,13E,15Z in PD1, inhibited platelet aggregation at submicromolar concentrations when induced by either collagen, arachidonic acid, or thromboxane. The inhibition occurred at the level of both the cyclooxygenase activity and thromboxane receptor site. Interestingly, all the metabolites tested exhibiting the E,Z,E-conjugated triene were active, whereas E,E,Z trienes (as in PD1) or all-trans (E,E,E) trienes were inactive. We conclude that PDX and other oxygenated products from PUFAs of nutritional interest, having the E,Z,E-conjugated triene motif and collectively named poxytrins (PUFA oxygenated trienes), might have antithrombotic potential.
Collapse
Affiliation(s)
- Ping Chen
- Université de Lyon, Unité Mixte de Recherche (UMR) 870, Institut National de la Santé et de la Recherche Médicale (INSERM)/Institut National des Sciences Appliquées–Lyon, and UMR 1235, Institut National de Recherche Agronomique, Villeurbanne, France
| | | | | | | |
Collapse
|
99
|
Babenko NA, Semenova YA. Effects of long-term fish oil-enriched diet on the sphingolipid metabolism in brain of old rats. Exp Gerontol 2010; 45:375-80. [DOI: 10.1016/j.exger.2010.02.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2009] [Revised: 02/22/2010] [Accepted: 02/23/2010] [Indexed: 01/25/2023]
|
100
|
Kim HY, Akbar M, Kim YS. Phosphatidylserine-dependent neuroprotective signaling promoted by docosahexaenoic acid. Prostaglandins Leukot Essent Fatty Acids 2010; 82:165-72. [PMID: 20207120 PMCID: PMC3383770 DOI: 10.1016/j.plefa.2010.02.025] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Enrichment of polyunsaturated fatty acids, particularly docosahexaenoic acid (DHA, 22:6n-3), in the brain is known to be critical for optimal brain development and function. Mechanisms for DHA's beneficial effects in the nervous system are not clearly understood at present. DHA is incorporated into the phospholipids in neuronal membranes, which in turn can influence not only the membrane chemical and physical properties but also the cell signaling involved in neuronal survival, proliferation and differentiation. Our studies have indicated that DHA supplementation promotes phosphatidylserine (PS) accumulation and inhibits neuronal cell death under challenged conditions, supporting a notion that DHA is an important neuroprotective agent. This article summarizes our findings on the DHA-mediated membrane-related signaling mechanisms that might explain some of the beneficial effects of DHA, particularly on neuronal survival.
Collapse
Affiliation(s)
- Hee-Yong Kim
- Laboratory of Molecular Signaling, NIAAA, NIH, Bethesda, MD 20892-9410, USA.
| | | | | |
Collapse
|