1
|
Frank GKW, Scolnick B. Therapeutic ketogenic diet as treatment for anorexia nervosa. Front Nutr 2024; 11:1392135. [PMID: 39296512 PMCID: PMC11409850 DOI: 10.3389/fnut.2024.1392135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 08/13/2024] [Indexed: 09/21/2024] Open
Abstract
Anorexia nervosa (AN) is a severe psychiatric disorder. However, we lack neurobiological models and interventions to explain and treat the core characteristics of food restriction, feeling fat, and body size overestimation. Research has made progress in understanding brain function involved in the pathophysiology of AN, but translating those results into biological therapies has been challenging. Studies have suggested that metabolic factors could contribute to developing and maintaining AN pathophysiology. Here, we describe a neurobiological model for why using a therapeutic ketogenic diet could address key alterations in brain function in AN and prevent the desire for weight loss and associated eating disorder-specific symptoms. This translational model is based on animal studies and human data and integrates behavioral traits, brain neural energy metabolism, and neurotransmitter function. Pilot data indicate that the intervention can dramatically reduce eating and body-related fears, although larger studies across illness stages still need to be conducted.
Collapse
Affiliation(s)
- Guido K W Frank
- Department of Psychiatry, San Diego School of Medicine, University of California, San Diego, San Diego, CA, United States
| | | |
Collapse
|
2
|
Alluri H, Peddaboina CS, Tharakan B. Evaluation of Tight Junction Integrity in Brain Endothelial Cells Using Confocal Microscopy. Methods Mol Biol 2024; 2711:257-262. [PMID: 37776464 DOI: 10.1007/978-1-0716-3429-5_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/02/2023]
Abstract
The blood-brain barrier (BBB) is a highly complex and dynamic microvascular barrier that protects the brain parenchyma from the entry of pathogens, toxins, and other macromolecules and is a critical structure that helps to maintain brain homeostasis. The BBB is formed mainly by brain capillary endothelial cells and perivascular astrocytes and pericytes. One of the primary properties of the BBB is a tight regulation of paracellular permeability due to the presence of tight junctional complexes (also, adherens and gap junctions) between the neighboring microvascular endothelial cells. Alterations in the assembly of the tight junctions impair BBB properties, particularly influenced barrier integrity and permeability. The tight junctions of the BBB are mainly composed of proteins including claudins, occludin, and zonula occludens-1 (ZO-1). Zonula occludens-1 binds to the actin cytoskeleton, and its localization provides valuable information on the status of BBB integrity and permeability. Immunofluorescence localization of ZO-1 and/or other tight junction proteins is a reliable indicator of barrier integrity and permeability in microvascular endothelial cells. In microvascular endothelial cells, f-actin stress fiber formation significantly influences the rate and size of the inter-endothelial cell gap that form as cells retract from their borders. Rhodamine phalloidin is a popular conjugate used as a fluorescent label for f-actin. Herein, we describe the procedures for ZO-1 immunofluorescence and f-actin labeling followed by confocal microscopic imaging to determine barrier integrity and tight junction organization in brain microvascular endothelial cells in vitro.
Collapse
Affiliation(s)
| | | | - Binu Tharakan
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
3
|
Guldemir HH, Buyukuslu N, Yigit P, Cakici C, Ozdemir EM. Effects of omega fatty acids on the short-term postprandial satiety related peptides in rats. INT J VITAM NUTR RES 2023; 93:401-409. [PMID: 34986656 DOI: 10.1024/0300-9831/a000743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We aimed to assess the effects of omega fatty acids on time depending on responses of satiety hormones. Sixty adult rats were randomly divided into 4 groups; linoleic acid (LA), α-linolenic acid (ALA), eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) groups. For each fatty acid, the dose of 400 mg/kg was applied by oral gavage. Blood samples were taken after the 15, 30, 60 and 120 minutes. Ghrelin, cholecystokinin (CCK), glucagon-like peptide-1 (GLP-1), peptide YY (PYY), leptin and insulin hormones were analyzed by ELISA. We observed the significant increases (p<0.05) of the levels of CCK between n-3 (ALA, at 60th min; EPA, at 30th and 60th min and DHA, at 60 min) and n-6 (LA) supplemented rats. The highest GLP-1 levels were in ALA (0.70 ng/mL) and DHA (0.67 ng/mL) supplemented groups at 60th and 120th min indicating n-3 fatty acids efficiency on satiety compared to LA. It seems that ALA at 60th min and EPA at 120th min could provide the highest satiety effect with the highest insulin response, while the efficiency of LA supplementation on insulin-induced satiety diminished. The only significant change in AUC values among all hormones was in the CCK of the ALA group (p=0.004). The level of leptin increased in DHA and EPA supplemented rats (p=0.140). Our results showed that dietary omega fatty acids influenced the releasing of hormones in different ways possibly depending on chain length or saturation degree. Comprehensive studies need to be addressed for each fatty acid on satiety-related peptide hormones.
Collapse
Affiliation(s)
- Hilal Hizli Guldemir
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Anadolu University, Eskisehir, Turkey
| | - Nihal Buyukuslu
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Istanbul Medipol University, Istanbul, Turkey
| | - Pakize Yigit
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Cagri Cakici
- Department of Medical Biochemistry, Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Ekrem Musa Ozdemir
- Medical Research Center (MEDITAM), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
| |
Collapse
|
4
|
de Ruiter Swain J, Michalopoulou E, Noch EK, Lukey MJ, Van Aelst L. Metabolic partitioning in the brain and its hijacking by glioblastoma. Genes Dev 2023; 37:681-702. [PMID: 37648371 PMCID: PMC10546978 DOI: 10.1101/gad.350693.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
The different cell types in the brain have highly specialized roles with unique metabolic requirements. Normal brain function requires the coordinated partitioning of metabolic pathways between these cells, such as in the neuron-astrocyte glutamate-glutamine cycle. An emerging theme in glioblastoma (GBM) biology is that malignant cells integrate into or "hijack" brain metabolism, co-opting neurons and glia for the supply of nutrients and recycling of waste products. Moreover, GBM cells communicate via signaling metabolites in the tumor microenvironment to promote tumor growth and induce immune suppression. Recent findings in this field point toward new therapeutic strategies to target the metabolic exchange processes that fuel tumorigenesis and suppress the anticancer immune response in GBM. Here, we provide an overview of the intercellular division of metabolic labor that occurs in both the normal brain and the GBM tumor microenvironment and then discuss the implications of these interactions for GBM therapy.
Collapse
Affiliation(s)
- Jed de Ruiter Swain
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
- Cold Spring Harbor Laboratory School of Biological Sciences, Cold Spring Harbor, New York 11724, USA
| | | | - Evan K Noch
- Department of Neurology, Division of Neuro-oncology, Weill Cornell Medicine, New York, New York 10021, USA
| | - Michael J Lukey
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA;
| | - Linda Van Aelst
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA;
| |
Collapse
|
5
|
Erythrocyte Plasma Membrane Lipid Composition Mirrors That of Neurons and Glial Cells in Murine Experimental In Vitro and In Vivo Inflammation. Cells 2023; 12:cells12040561. [PMID: 36831228 PMCID: PMC9953778 DOI: 10.3390/cells12040561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 01/30/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
Lipid membrane turnover and myelin repair play a central role in diseases and lesions of the central nervous system (CNS). The aim of the present study was to analyze lipid composition changes due to inflammatory conditions. We measured the fatty acid (FA) composition in erythrocytes (RBCs) and spinal cord tissue (gas chromatography) derived from mice affected by experimental allergic encephalomyelitis (EAE) in acute and remission phases; cholesterol membrane content (Filipin) and GM1 membrane assembly (CT-B) in EAE mouse RBCs, and in cultured neurons, oligodendroglial cells and macrophages exposed to inflammatory challenges. During the EAE acute phase, the RBC membrane showed a reduction in polyunsaturated FAs (PUFAs) and an increase in saturated FAs (SFAs) and the omega-6/omega-3 ratios, followed by a restoration to control levels in the remission phase in parallel with an increase in monounsaturated fatty acid residues. A decrease in PUFAs was also shown in the spinal cord. CT-B staining decreased and Filipin staining increased in RBCs during acute EAE, as well as in cultured macrophages, neurons and oligodendrocyte precursor cells exposed to inflammatory challenges. This regulation in lipid content suggests an increased cell membrane rigidity during the inflammatory phase of EAE and supports the investigation of peripheral cell membrane lipids as possible biomarkers for CNS lipid membrane concentration and assembly.
Collapse
|
6
|
Marrano N, Biondi G, Borrelli A, Rella M, Zambetta T, Di Gioia L, Caporusso M, Logroscino G, Perrini S, Giorgino F, Natalicchio A. Type 2 Diabetes and Alzheimer's Disease: The Emerging Role of Cellular Lipotoxicity. Biomolecules 2023; 13:183. [PMID: 36671568 PMCID: PMC9855893 DOI: 10.3390/biom13010183] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/06/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
Type 2 diabetes (T2D) and Alzheimer's diseases (AD) represent major health issues that have reached alarming levels in the last decades. Although growing evidence demonstrates that AD is a significant comorbidity of T2D, and there is a ~1.4-2-fold increase in the risk of developing AD among T2D patients, the involvement of possible common triggers in the pathogenesis of these two diseases remains largely unknown. Of note, recent mechanistic insights suggest that lipotoxicity could represent the missing ring in the pathogenetic mechanisms linking T2D to AD. Indeed, obesity, which represents the main cause of lipotoxicity, has been recognized as a major risk factor for both pathological conditions. Lipotoxicity can lead to inflammation, insulin resistance, oxidative stress, ceramide and amyloid accumulation, endoplasmic reticulum stress, ferroptosis, and autophagy, which are shared biological events in the pathogenesis of T2D and AD. In the current review, we try to provide a critical and comprehensive view of the common molecular pathways activated by lipotoxicity in T2D and AD, attempting to summarize how these mechanisms can drive future research and open the way to new therapeutic perspectives.
Collapse
Affiliation(s)
- Nicola Marrano
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Giuseppina Biondi
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Anna Borrelli
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Martina Rella
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Tommaso Zambetta
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Ludovico Di Gioia
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Mariangela Caporusso
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Giancarlo Logroscino
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124 Bari, Italy
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari Aldo Moro at Pia Fondazione Cardinale G. Panico, 73039 Lecce, Italy
| | - Sebastio Perrini
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Francesco Giorgino
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Annalisa Natalicchio
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| |
Collapse
|
7
|
Husain MA, Vachon A, Chouinard-Watkins R, Vandal M, Calon F, Plourde M. Investigating the plasma-liver-brain axis of omega-3 fatty acid metabolism in mouse knock-in for the human apolipoprotein E epsilon 4 allele. J Nutr Biochem 2023; 111:109181. [PMID: 36220526 DOI: 10.1016/j.jnutbio.2022.109181] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 06/30/2022] [Accepted: 09/13/2022] [Indexed: 11/06/2022]
Abstract
The metabolism of docosahexaenoic acid (DHA), an omega-3 fatty acid, is different in carriers of APOE4, the main genetic risk factor for late-onset Alzheimer's disease. The brain relies on the plasma DHA pool for its need, but the plasma-liver-brain axis in relation to cognition remains obscure. We hypothesized that this relationship is compromised in APOE4 mice considering the differences in fatty acid metabolism between APOE3 and APOE4 mice. Male and female APOE3 and APOE4 mice were fed either a diet enriched with DHA (0.7 g DHA/100 g diet) or a control diet for 8 months. There was a significant genotype × diet interaction for DHA concentration in the liver and adipose tissue. In the cortex, a genotype effect was found where APOE4 mice had a higher concentration of DHA than APOE3 mice fed the control diet. There was a significant genotype × diet interaction for the liver and hippocampal arachidonic acid (AA). APOE4 mice had 20-30% lower plasma DHA and AA concentrations than APOE3 mice, independent of diet. Plasma and liver DHA levels were significantly correlated in APOE3 and APOE4 mice. In APOE4 mice, there was a significant correlation between plasma, adipose tissues, cortex DHA and the Barnes maze and/or with a better recognition index. Moreover, higher AA levels in the liver and the hippocampus of APOE4 mice were correlated with lower cognitive performance. Our results suggest that there is a plasma-liver-brain axis of DHA that is modified in APOE4 mice. Moreover, our data support that APOE4 mice rely more on plasma DHA than APOE3 mice, especially in cognitive performance. Any disturbance in plasma DHA metabolism might have a greater impact on cognition in APOE4 carriers.
Collapse
Affiliation(s)
- Mohammed Amir Husain
- Centre de Recherche sur le Vieillissement, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada; Département de Médecine, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Annick Vachon
- Centre de Recherche sur le Vieillissement, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada
| | | | - Milène Vandal
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Frédéric Calon
- Institut de la nutrition et des aliments fonctionnels, Université Laval, Québec, Quebec, Canada; Faculté de pharmacie et center de recherche du CHU de Québec-Université Laval, Quebec, Canada
| | - Mélanie Plourde
- Centre de Recherche sur le Vieillissement, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada; Département de Médecine, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada; Institut de la nutrition et des aliments fonctionnels, Université Laval, Québec, Quebec, Canada.
| |
Collapse
|
8
|
Adrien V, Bosc N, Fumat H, Tessier C, Ferreri F, Mouchabac S, Tareste D, Nuss P. Higher stress response and altered quality of life in schizophrenia patients with low membrane levels of docosahexaenoic acid. Front Psychiatry 2023; 14:1089724. [PMID: 36816405 PMCID: PMC9937080 DOI: 10.3389/fpsyt.2023.1089724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 01/12/2023] [Indexed: 02/05/2023] Open
Abstract
Schizophrenia is a severe, chronic, and heterogeneous mental disorder that affects approximately 1% of the world population. Ongoing research aims at clustering schizophrenia heterogeneity into various "biotypes" to identify subgroups of individuals displaying homogeneous symptoms, etiopathogenesis, prognosis, and treatment response. The present study is in line with this approach and focuses on a biotype partly characterized by a specific membrane lipid composition. We have examined clinical and biological data of patients with stabilized schizophrenia, including the fatty acid content of their erythrocyte membranes, in particular the omega-3 docosahexaenoic acid (DHA). Two groups of patients of similar size were identified: the DHA- group (N = 19) with a lower proportion of membrane DHA as compared to the norm in the general population, and the DHAn group (N = 18) with a normal proportion of DHA. Compared to DHAn, DHA- patients had a higher number of hospitalizations and a lower quality of life in terms of perceived health and physical health. They also exhibited significant higher interleukin-6 and cortisol blood levels. These results emphasize the importance of measuring membrane lipid and immunoinflammatory biomarkers in stabilized patients to identify a specific subgroup and optimize non-pharmacological interventions. It could also guide future research aimed at proposing specific pharmacological treatments.
Collapse
Affiliation(s)
- Vladimir Adrien
- AP-HP, Sorbonne Université, Department of Psychiatry, Hôpital Saint-Antoine, Paris, France.,Infrastructure for Clinical Research in Neurosciences (iCRIN), Paris Brain Institute, Sorbonne Université, INSERM, CNRS, Paris, France.,Université Paris Cité, INSERM UMR-S 1266, Institut de Psychiatrie et Neurosciences de Paris, Paris, France
| | - Nicolas Bosc
- AP-HP, Sorbonne Université, Department of Psychiatry, Hôpital Saint-Antoine, Paris, France
| | - Hugo Fumat
- Université Paris Cité, INSERM UMR-S 1266, Institut de Psychiatrie et Neurosciences de Paris, Paris, France
| | - Cédric Tessier
- AP-HP, Sorbonne Université, Department of Psychiatry, Hôpital Saint-Antoine, Paris, France
| | - Florian Ferreri
- AP-HP, Sorbonne Université, Department of Psychiatry, Hôpital Saint-Antoine, Paris, France.,Infrastructure for Clinical Research in Neurosciences (iCRIN), Paris Brain Institute, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Stéphane Mouchabac
- AP-HP, Sorbonne Université, Department of Psychiatry, Hôpital Saint-Antoine, Paris, France.,Infrastructure for Clinical Research in Neurosciences (iCRIN), Paris Brain Institute, Sorbonne Université, INSERM, CNRS, Paris, France
| | - David Tareste
- Université Paris Cité, INSERM UMR-S 1266, Institut de Psychiatrie et Neurosciences de Paris, Paris, France
| | - Philippe Nuss
- AP-HP, Sorbonne Université, Department of Psychiatry, Hôpital Saint-Antoine, Paris, France.,Centre de Recherche Saint-Antoine, INSERM UMR S938, Sorbonne Université, Paris, France
| |
Collapse
|
9
|
FABP5 controls macrophage alternative activation and allergic asthma by selectively programming long-chain unsaturated fatty acid metabolism. Cell Rep 2022; 41:111668. [DOI: 10.1016/j.celrep.2022.111668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 09/13/2022] [Accepted: 10/25/2022] [Indexed: 11/17/2022] Open
|
10
|
Li M, Gao Y, Wang D, Hu X, Jiang J, Qing Y, Yang X, Cui G, Wang P, Zhang J, Sun L, Wan C. Impaired Membrane Lipid Homeostasis in Schizophrenia. Schizophr Bull 2022; 48:1125-1135. [PMID: 35751100 PMCID: PMC9434453 DOI: 10.1093/schbul/sbac011] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
BACKGROUND AND HYPOTHESIS Multiple lines of clinical, biochemical, and genetic evidence suggest that disturbances of membrane lipids and their metabolism are probably involved in the etiology of schizophrenia (SCZ). Lipids in the membrane are essential to neural development and brain function, however, their role in SCZ remains largely unexplored. STUDY DESIGN Here we investigated the lipidome of the erythrocyte membrane of 80 patients with SCZ and 40 healthy controls using ultra-performance liquid chromatography-mass spectrometry. Based on the membrane lipids profiling, we explored the potential mechanism of membrane phospholipids metabolism. STUDY RESULTS By comparing 812 quantified lipids, we found that in SCZ, membrane phosphatidylcholines and phosphatidylethanolamines, especially the plasmalogen, were significantly decreased. In addition, the total polyunsaturated fatty acids (PUFAs) in the membrane of SCZ were significantly reduced, resulting in a decrease in membrane fluidity. The accumulation of membrane oxidized lipids and the level of peripheral lipid peroxides increased, suggesting an elevated level of oxidative stress in SCZ. Further study of membrane-phospholipid-remodeling genes showed that activation of PLA2s and LPCATs expression in patients, supporting the imbalance of unsaturated and saturated fatty acyl remodeling in phospholipids of SCZ patients. CONCLUSIONS Our results suggest that the mechanism of impaired membrane lipid homeostasis is related to the activated phospholipid remodeling caused by excessive oxidative stress in SCZ. Disordered membrane lipids found in this study may reflect the membrane dysfunction in the central nervous system and impact neurotransmitter transmission in patients with SCZ, providing new evidence for the membrane lipids hypothesis of SCZ.
Collapse
Affiliation(s)
- Minghui Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Gao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Dandan Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaowen Hu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Jie Jiang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Qing
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Xuhan Yang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Gaoping Cui
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Pengkun Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Juan Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Liya Sun
- To whom correspondence should be addressed; Bio-X Institutes, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China, tel: +86-021-62822491, fax: +86-021-62932059, e-mail: (C.W.), (L.S.)
| | - Chunling Wan
- To whom correspondence should be addressed; Bio-X Institutes, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China, tel: +86-021-62822491, fax: +86-021-62932059, e-mail: (C.W.), (L.S.)
| |
Collapse
|
11
|
Terzo S, Amato A, Mulè F. From obesity to Alzheimer's disease through insulin resistance. J Diabetes Complications 2021; 35:108026. [PMID: 34454830 DOI: 10.1016/j.jdiacomp.2021.108026] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 07/29/2021] [Accepted: 08/14/2021] [Indexed: 12/27/2022]
Abstract
Alzheimer's disease is one of the most frequent forms of dementia. It is a progressive neurodegenerative disease, characterized by presence of amyloid plaques and neurofibrillary tangles in the brain. Obesity is regarded as abnormal fat accumulation with deleterious impact on human health. There is full scientific evidence that obesity and the metabolic comorbidities (e.g., insulin resistance, hyperglycaemia, and type 2 diabetes) are related to Alzheimer's disease and likely in the causative pathway. Numerous studies have identified several overlapping neurodegenerative mechanisms, including oxidative stress, mitochondrial dysfunction, and inflammation. In this review, we present how obesity and the associated lipotoxicity as well as chronic inflammation initiate a state of insulin resistance that in turn, may have a role in causing the characteristic cerebral alterations of AD. In particular, we focus on the molecular mechanisms linking the obesity-induced impairment in insulin signalling to the upregulation of Aβ aggregation, tau hyper-phosphorylation, inflammation, oxidative stress and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Simona Terzo
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, 90128, Italy.
| | - Antonella Amato
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, 90128, Italy.
| | - Flavia Mulè
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, 90128, Italy.
| |
Collapse
|
12
|
Desai A, Chen H, Kevala K, Kim HY. Higher n-3 Polyunsaturated Fatty Acid Diet Improves Long-Term Neuropathological and Functional Outcome after Repeated Mild Traumatic Brain Injury. J Neurotrauma 2021; 38:2622-2632. [PMID: 33913741 PMCID: PMC8403198 DOI: 10.1089/neu.2021.0096] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Repeated mild traumatic brain injury (TBI) can cause persistent neuropathological effects and is a major risk factor for chronic traumatic encephalopathy. PUFAs (n-3 polyunsaturated fatty acids) were shown to improve acute TBI outcomes in single-injury models in most cases. In this study, we demonstrate positive effects of dietary n-3 PUFA on long-term neuropathological and functional outcome in a clinically relevant model of repeated mild TBI using the Closed-Head Impact Model of Engineered Rotational Acceleration (CHIMERA). Adult mice, reared on n-3 PUFA adequate (higher n-3 PUFA) or deficient (lower n-3 PUFA) diets, were given a mild CHIMERA daily for 3 consecutive days. At 2 months after injury, visual function and spatial memory were evaluated. Glia cell activation was assessed by immunostaining using antibodies of ionized calcium-binding adaptor molecule 1 and glial fibrillary acidic protein, and axonal damage was examined using silver staining. Repeated CHIMERA (rCHIMERA)-induced gliosis was significantly suppressed in the optic tract, corpus callosum, and hippocampus of mice fed the n-3 PUFA adequate diet compared to the deficient diet group. Considerable axonal damage was detected in the optic tract after rCHIMERA, but the adequate diet group displayed less axonal damage compared to the deficient diet group. rCHIMERA induced a drastic reduction in N1 amplitude of the visual evoked potential in both diet groups and the a-wave amplitude of the electroretinogram in the deficient diet group. However, reduction of N1 and a-wave amplitude were less severe in the adequate diet group. The Morris water maze probe test indicated a significant decrease in the number of platform crossings in the deficient diet group compared to the adequate group. In summary, dietary n-3 PUFA can attenuate persistent glial cell activation and axonal damage and improve deficits in visual function and spatial memory after repeated mild TBI. These data support the neuroprotective potential of a higher n-3 PUFA diet in ameliorating the adverse outcome of repeated mild TBI.
Collapse
Affiliation(s)
- Abhishek Desai
- Laboratory of Molecular Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Huazhen Chen
- Laboratory of Molecular Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
- Center for Neuroscience and Regenerative Medicine, Henry M. Jackson Foundation, Bethesda, Maryland, USA
| | - Karl Kevala
- Laboratory of Molecular Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Hee-Yong Kim
- Laboratory of Molecular Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
- Center for Neuroscience and Regenerative Medicine, Henry M. Jackson Foundation, Bethesda, Maryland, USA
| |
Collapse
|
13
|
Pompura SL, Wagner A, Kitz A, LaPerche J, Yosef N, Dominguez-Villar M, Hafler DA. Oleic acid restores suppressive defects in tissue-resident FOXP3 Tregs from patients with multiple sclerosis. J Clin Invest 2021; 131:138519. [PMID: 33170805 DOI: 10.1172/jci138519] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 11/05/2020] [Indexed: 12/21/2022] Open
Abstract
FOXP3+ Tregs rely on fatty acid β-oxidation-driven (FAO-driven) oxidative phosphorylation (OXPHOS) for differentiation and function. Recent data demonstrate a role for Tregs in the maintenance of tissue homeostasis, with tissue-resident Tregs possessing tissue-specific transcriptomes. However, specific signals that establish tissue-resident Treg programs remain largely unknown. Tregs metabolically rely on FAO, and considering the lipid-rich environments of tissues, we hypothesized that environmental lipids drive Treg homeostasis. First, using human adipose tissue to model tissue residency, we identified oleic acid as the most prevalent free fatty acid. Mechanistically, oleic acid amplified Treg FAO-driven OXPHOS metabolism, creating a positive feedback mechanism that increased the expression of FOXP3 and phosphorylation of STAT5, which enhanced Treg-suppressive function. Comparing the transcriptomic program induced by oleic acid with proinflammatory arachidonic acid, we found that Tregs sorted from peripheral blood and adipose tissue of healthy donors transcriptomically resembled the Tregs treated in vitro with oleic acid, whereas Tregs from patients with multiple sclerosis (MS) more closely resembled an arachidonic acid transcriptomic profile. Finally, we found that oleic acid concentrations were reduced in patients with MS and that exposure of MS Tregs to oleic acid restored defects in their suppressive function. These data demonstrate the importance of fatty acids in regulating tissue inflammatory signals.
Collapse
Affiliation(s)
- Saige L Pompura
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Allon Wagner
- Department of Electrical Engineering and Computer Science, and the Center for Computational Biology, University of California Berkeley, Berkeley, California, USA
| | - Alexandra Kitz
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jacob LaPerche
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Nir Yosef
- Department of Electrical Engineering and Computer Science, and the Center for Computational Biology, University of California Berkeley, Berkeley, California, USA.,Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard University, Boston, Massachusetts, USA.,Chan-Zuckerberg Biohub, San Francisco, California, USA
| | - Margarita Dominguez-Villar
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA.,Faculty of Medicine, Imperial College London, London, United Kingdom
| | - David A Hafler
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA.,Broad Institute of MIT and Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
14
|
Mongan D, Healy C, Jones HJ, Zammit S, Cannon M, Cotter DR. Plasma polyunsaturated fatty acids and mental disorders in adolescence and early adulthood: cross-sectional and longitudinal associations in a general population cohort. Transl Psychiatry 2021; 11:321. [PMID: 34059620 PMCID: PMC8167090 DOI: 10.1038/s41398-021-01425-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 03/23/2021] [Accepted: 04/08/2021] [Indexed: 02/07/2023] Open
Abstract
Polyunsaturated fatty acids (PUFAs) may be pertinent to the development of mental disorders, for example via modulation of inflammation and synaptogenesis. We wished to examine cross-sectional and longitudinal associations between PUFAs and mental disorders in a large cohort of young people. Participants in the Avon Longitudinal Study of Parents and Children were interviewed and provided blood samples at two sampling periods when approximately 17 and 24 years old. Plasma PUFA measures (total omega-6 [n-6], total omega-3 [n-3], n-6:n-3 ratio and docosahexaenoic acid [DHA] percentage of total fatty acids) were assessed using nuclear magnetic resonance spectroscopy. Cross-sectional and longitudinal associations between standardised PUFA measures and three mental disorders (psychotic disorder, moderate/severe depressive disorder and generalised anxiety disorder [GAD]) were measured by logistic regression, adjusting for age, sex, body mass index and cigarette smoking. There was little evidence of cross-sectional associations between PUFA measures and mental disorders at age 17. At age 24, the n-6:n-3 ratio was positively associated with psychotic disorder, depressive disorder and GAD, while DHA was inversely associated with psychotic disorder. In longitudinal analyses, there was evidence of an inverse association between DHA at age 17 and incident psychotic disorder at age 24 (adjusted odds ratio 0.44, 95% confidence interval 0.22-0.87) with little such evidence for depressive disorder or GAD. There was little evidence for associations between change in PUFA measures from 17 to 24 years and incident mental disorders at 24 years. These findings provide support for associations between PUFAs and mental disorders in early adulthood, and in particular, for DHA in adolescence in relation to prevention of psychosis.
Collapse
Affiliation(s)
- David Mongan
- Department of Psychiatry, Royal College of Surgeons in Ireland, Dublin, Ireland.
| | - Colm Healy
- Department of Psychiatry, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Hannah J Jones
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- National Institute for Health Research Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust, University of Bristol, Bristol, UK
| | - Stan Zammit
- Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- National Institute for Health Research Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust, University of Bristol, Bristol, UK
- Division of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Mary Cannon
- Department of Psychiatry, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - David R Cotter
- Department of Psychiatry, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
15
|
Kung WM, Lin MS. Beneficial Impacts of Alpha-Eleostearic Acid from Wild Bitter Melon and Curcumin on Promotion of CDGSH Iron-Sulfur Domain 2: Therapeutic Roles in CNS Injuries and Diseases. Int J Mol Sci 2021; 22:ijms22073289. [PMID: 33804820 PMCID: PMC8037269 DOI: 10.3390/ijms22073289] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 03/20/2021] [Accepted: 03/21/2021] [Indexed: 02/05/2023] Open
Abstract
Neuroinflammation and abnormal mitochondrial function are related to the cause of aging, neurodegeneration, and neurotrauma. The activation of nuclear factor κB (NF-κB), exaggerating these two pathologies, underlies the pathogenesis for the aforementioned injuries and diseases in the central nervous system (CNS). CDGSH iron-sulfur domain 2 (CISD2) belongs to the human NEET protein family with the [2Fe-2S] cluster. CISD2 has been verified as an NFκB antagonist through the association with peroxisome proliferator-activated receptor-β (PPAR-β). This protective protein can be attenuated under circumstances of CNS injuries and diseases, thereby causing NFκB activation and exaggerating NFκB-provoked neuroinflammation and abnormal mitochondrial function. Consequently, CISD2-elevating plans of action provide pathways in the management of various disease categories. Various bioactive molecules derived from plants exert protective anti-oxidative and anti-inflammatory effects and serve as natural antioxidants, such as conjugated fatty acids and phenolic compounds. Herein, we have summarized pharmacological characters of the two phytochemicals, namely, alpha-eleostearic acid (α-ESA), an isomer of conjugated linolenic acids derived from wild bitter melon (Momordica charantia L. var. abbreviata Ser.), and curcumin, a polyphenol derived from rhizomes of Curcuma longa L. In this review, the unique function of the CISD2-elevating effect of α-ESA and curcumin are particularly emphasized, and these natural compounds are expected to serve as a potential therapeutic target for CNS injuries and diseases.
Collapse
Affiliation(s)
- Woon-Man Kung
- Department of Exercise and Health Promotion, College of Kinesiology and Health, Chinese Culture University, Taipei 11114, Taiwan;
| | - Muh-Shi Lin
- Division of Neurosurgery, Department of Surgery, Kuang Tien General Hospital, Taichung 43303, Taiwan
- Department of Biotechnology and Animal Science, College of Bioresources, National Ilan University, Yilan 26047, Taiwan
- Department of Biotechnology, College of Medical and Health Care, Hung Kuang University, Taichung 43302, Taiwan
- Department of Health Business Administration, College of Medical and Health Care, Hung Kuang University, Taichung 43302, Taiwan
- Correspondence: ; Tel.: +886-4-2665-1900
| |
Collapse
|
16
|
Stirton H, Meek BP, Edel AL, Solati Z, Surendran A, Aukema H, Modirrousta M, Ravandi A. Oxolipidomics profile in major depressive disorder: Comparing remitters and non-remitters to repetitive transcranial magnetic stimulation treatment. PLoS One 2021; 16:e0246592. [PMID: 33571313 PMCID: PMC7877633 DOI: 10.1371/journal.pone.0246592] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 01/19/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Repetitive Transcranial Magnetic Stimulation [rTMS] is increasingly being used to treat Major Depressive Disorder [MDD]. Given that not all patients respond to rTMS, it would be clinically useful to have reliable biomarkers that predict treatment response. Oxidized phosphatidylcholine [OxPC] and some oxylipins are important plasma biomarkers of oxidative stress and inflammation. Not only is depression associated with oxidative stress, but rTMS has been shown to have anti-oxidative effects. OBJECTIVES To investigate whether plasma oxolipidomics profiles could predict treatment response in patients with treatment resistant MDD. METHODS Fourty-eight patients undergoing rTMS treatment for MDD were recruited along with nine healthy control subjects. Plasma OxPCs and oxylipins were extracted and analyzed through high performance liquid chromatography coupled with mass spectrometry. Patients with a Hamilton Depression Rating Scale score [Ham-D] ≤7 post-treatment were defined as having entered remission. RESULTS Fifty-seven OxPC and 32 oxylipin species were identified in our subjects. MDD patients who entered remission following rTMS had significantly higher pre-rTMS levels of total and fragmented OxPCs compared to non-remitters and controls [one-way ANOVA, p<0.05]. However, no significant changes in OxPC levels were found as a result of rTMS, regardless of treatment response [p>0.05]. No differences in plasma oxylipins were found between remitters and non-remitters at baseline. CONCLUSION Certain categories of OxPCs may be useful predictive biomarkers for response to rTMS treatment in MDD. Given that elevated oxidized lipids may indicate higher levels of oxidative stress and inflammation in the brain, patients with this phenotype of depression may be more receptive to rTMS treatment.
Collapse
Affiliation(s)
- Hannah Stirton
- Cardiovascular Lipidomics Laboratory, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba, Canada
- Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Benjamin P. Meek
- Dept. of Psychiatry, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Andrea L. Edel
- Cardiovascular Lipidomics Laboratory, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba, Canada
| | - Zahra Solati
- Cardiovascular Lipidomics Laboratory, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba, Canada
- Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Arun Surendran
- Cardiovascular Lipidomics Laboratory, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba, Canada
- Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Harold Aukema
- Cardiovascular Lipidomics Laboratory, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba, Canada
| | - Mandana Modirrousta
- Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- Dept. of Psychiatry, University of Manitoba, Winnipeg, Manitoba, Canada
- * E-mail: (MM); (AR)
| | - Amir Ravandi
- Cardiovascular Lipidomics Laboratory, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba, Canada
- Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- * E-mail: (MM); (AR)
| |
Collapse
|
17
|
Huanglian Jiedu decoction remodels the periphery microenvironment to inhibit Alzheimer's disease progression based on the "brain-gut" axis through multiple integrated omics. ALZHEIMERS RESEARCH & THERAPY 2021; 13:44. [PMID: 33579351 PMCID: PMC7881564 DOI: 10.1186/s13195-021-00779-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 01/25/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND In recent years, excellent results have suggested an association between the "brain-gut" axis and Alzheimer's disease (AD) progression, yet the role of the "brain-gut" axis in AD pathogenesis still remains obscure. Herein, we provided a potential link between the central and peripheral neuroinflammatory disorders in AD progression. METHODS The Morris water maze (MWM) test, immunohistochemistry, ELISA, ProcartaPlex Multiplex immunoassay, multiple LC-MS/MS methods, and the V3-V4 regions of 16S rRNA genes were applied to explore potential biomarkers. RESULTS In Tg-APP/PS1 mice, gut dysbiosis and lipid metabolism were highly associated with AD-like neuroinflammation. The combination of inflammatory factors (IL-6 and INF-γ), phosphatidylcholines (PCs) and SCFA-producing bacteria were expected to be early diagnostic biomarkers for AD. Huanglian Jiedu decoction (HLJDD) suppressed gut dysbiosis and the associated Aβ accumulation, harnessed neuroinflammation and reversed cognitive impairment. CONCLUSION Together, our findings highlighted the roles of neuroinflammation induced by gut dysbiosis and lipid metabolism disorder in AD progression. This integrated metabolomics approach showed its potential to understand the complex mechanisms of HLJDD in the treatment of AD.
Collapse
|
18
|
Madison AA, Belury MA, Andridge R, Shrout MR, Renna ME, Malarkey WB, Bailey MT, Kiecolt-Glaser JK. Afternoon distraction: a high-saturated-fat meal and endotoxemia impact postmeal attention in a randomized crossover trial. Am J Clin Nutr 2020; 111:1150-1158. [PMID: 32393980 PMCID: PMC7266694 DOI: 10.1093/ajcn/nqaa085] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 04/06/2020] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Saturated-fat intake and endotoxemia can impair cognition. However, their acute impact on cognitive performance is unknown. OBJECTIVE This study assessed the impact of 2 high-fat meals and endotoxemia on attention. METHODS In this double-blind, randomized crossover trial, 51 women (n = 32 breast cancer survivors, n = 19 noncancer controls; mean ± SD age: 53 ± 8 y) completed the Continuous Performance Test (CPT) and had their blood drawn to assess endotoxemia markers LPS binding protein (LBP), soluble CD14 (sCD14), and the LBP to sCD14 ratio 1 h prior to eating either a high-saturated-fat meal or a high-oleic-sunflower-oil meal. Women again completed the CPT 5 h postmeal. At 1 to 4 wk later, women completed the same protocol but consumed the other meal. RESULTS In adjusted models, women had more difficulty distinguishing target stimuli from distractors after consuming the high-saturated-fat meal than they did after the oleic-sunflower-oil meal (B = 4.44, SE = 1.88, P = 0.02). Women with higher baseline LBP had less consistent response times (B = 0.002, SE = 0.0008, P = 0.04). Those with higher LBP and LBP:sCD14 were less able to sustain their attention throughout the entire CPT, as reflected by their progressively slower (B = 0.002, SE = 0.0006, P = 0.003; and B = 2.43, SE = 0.090, P = 0.008, respectively) and more erratic (B = 0.003, SE = 0.0008, P < 0.0001; and B = 3.29, SE = 1.17, P = 0.006, respectively) response times. Additionally, women with higher baseline LBP or sCD14 were less able to maintain or increase response speeds at higher interstimulus intervals (B = 0.002, SE = 0.0006, P = 0.02; and B = 0.006, SE = 0.003, P = 0.03, respectively), indicating greater difficulty adapting to changing task demands. Significant meal type by LBP and LBP:sCD14 interactions emerged (P < 0.05), such that high LBP and LBP:sCD14 erased between-meal cognitive differences, uniformly impairing performance. CONCLUSIONS These results suggest that higher LBP, sCD14, and LBP:sCD14 and saturated-fat intake individually and jointly influence attention. Endotoxemia may override the relative cognitive benefit of healthier oil choices.This trial is registered at clinicaltrials.gov as NCT04247763.
Collapse
Affiliation(s)
- Annelise A Madison
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, Columbus, OH, USA,Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Martha A Belury
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, Columbus, OH, USA,Department of Human Sciences, College of Education and Human Ecology, The Ohio State University, Columbus, OH, USA
| | - Rebecca Andridge
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, Columbus, OH, USA,College of Public Health, The Ohio State University, Columbus, OH, USA
| | - M Rosie Shrout
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Megan E Renna
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, Columbus, OH, USA
| | - William B Malarkey
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, Columbus, OH, USA,Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Michael T Bailey
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, Columbus, OH, USA,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA,Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Janice K Kiecolt-Glaser
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, Columbus, OH, USA,Department of Psychiatry and Behavioral Health, The Ohio State University College of Medicine, Columbus, OH, USA,Address correspondence to JKK-G (E-mail: )
| |
Collapse
|
19
|
White CJ, Lee J, Choi J, Chu T, Scafidi S, Wolfgang MJ. Determining the Bioenergetic Capacity for Fatty Acid Oxidation in the Mammalian Nervous System. Mol Cell Biol 2020; 40:e00037-20. [PMID: 32123009 PMCID: PMC7189099 DOI: 10.1128/mcb.00037-20] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 02/16/2020] [Indexed: 12/15/2022] Open
Abstract
The metabolic state of the brain can greatly impact neurologic function. Evidence of this includes the therapeutic benefit of a ketogenic diet in neurologic diseases, including epilepsy. However, brain lipid bioenergetics remain largely uncharacterized. The existence, capacity, and relevance of mitochondrial fatty acid β-oxidation (FAO) in the brain are highly controversial, with few genetic tools available to evaluate the question. We have provided evidence for the capacity of brain FAO using a pan-brain-specific conditional knockout (KO) mouse incapable of FAO due to the loss of carnitine palmitoyltransferase 2, the product of an obligate gene for FAO (CPT2B-/-). Loss of central nervous system (CNS) FAO did not result in gross neuroanatomical changes or systemic differences in metabolism. Loss of CPT2 in the brain did not result in robustly impaired behavior. We demonstrate by unbiased and targeted metabolomics that the mammalian brain oxidizes a substantial quantity of long-chain fatty acids in vitro and in vivo Loss of CNS FAO results in robust accumulation of long-chain acylcarnitines in the brain, suggesting that the mammalian brain mobilizes fatty acids for their oxidation, irrespective of diet or metabolic state. Together, these data demonstrate that the mammalian brain oxidizes fatty acids under normal circumstances with little influence from or on peripheral tissues.
Collapse
Affiliation(s)
- Cory J White
- Department of Biological Chemistry, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Jieun Lee
- Department of Biological Chemistry, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Joseph Choi
- Department of Biological Chemistry, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Tiffany Chu
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Susanna Scafidi
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Michael J Wolfgang
- Department of Biological Chemistry, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
20
|
O'Reilly ÉJ, Bjornevik K, Furtado JD, Kolonel LN, Le Marchand L, McCullough ML, Stevens VL, Shadyab AH, Snetselaar L, Manson JE, Ascherio A. Prediagnostic plasma polyunsaturated fatty acids and the risk of amyotrophic lateral sclerosis. Neurology 2020; 94:e811-e819. [PMID: 31796528 PMCID: PMC7136057 DOI: 10.1212/wnl.0000000000008676] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 09/06/2019] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE To examine the association between prediagnostic plasma polyunsaturated fatty acids levels (PUFA) and amyotrophic lateral sclerosis (ALS). METHODS We identified 275 individuals who developed ALS while enrolled in 5 US prospective cohorts, and randomly selected 2 controls, alive at the time of the case diagnosis, matched on cohort, birth year, sex, ethnicity, fasting status, and time of blood draw. We measured PUFA, expressed as percentages of total fatty acids, using gas liquid chromatography and used conditional logistic regression to estimate risk ratios (RR) and 95% confidence intervals (CI) for the association between PUFA and ALS. RESULTS There was no association between total, n-3, and n-6 PUFA, eicosapentaenoic acid, or docosapentaenoic acid levels and ALS. Higher plasma α-linolenic acid (ALA) in men was associated with lower risk of ALS in age- and matching factor-adjusted analyses (top vs bottom quartile: RR = 0.21 [95% CI 0.07, 0.58], p for trend = 0.004). In women, higher plasma arachidonic acid was associated with higher risk (top vs bottom quartile: RR = 1.65 [95% CI 0.99, 2.76], p for trend = 0.052). Multivariable adjustment, including correlated PUFA, did not change the findings for ALA and arachidonic acid. In men and women combined, higher plasma docosahexaenoic acid (DHA) was associated with higher risk of ALS (top vs bottom quartile: RR = 1.56 [95% CI 1.01, 2.41], p for trend = 0.054), but in multivariable models the association was only evident in men. CONCLUSIONS The majority of individual PUFAs were not associated with ALS. In men, ALA was inversely and DHA was positively related to risk of ALS, while in women arachidonic acid was positively related. These findings warrant confirmation in future studies.
Collapse
Affiliation(s)
- Éilis J O'Reilly
- From the Departments of Nutrition (É.J.O., K.B., J.D.F., A.A.) and Epidemiology (J.E.M., A.A.), Harvard T.H. Chan School of Public Health, Boston, MA; School of Public Health (É.J.O.), College of Medicine, University College Cork, Ireland; Epidemiology Program (L.N.K., L.L.M.), University of Hawaii Cancer Center, Honolulu; Behavioral and Epidemiology Research Group (M.L.M., V.L.S.), American Cancer Society, Atlanta, GA; Family Medicine and Public Health (A.H.S.), School of Medicine, University of California San Diego; Department of Epidemiology (L.S.), College of Public Health, University of Iowa, Iowa City; and Department of Medicine (J.E.M.) and Channing Division of Network Medicine (J.E.M., A.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA.
| | - Kjetil Bjornevik
- From the Departments of Nutrition (É.J.O., K.B., J.D.F., A.A.) and Epidemiology (J.E.M., A.A.), Harvard T.H. Chan School of Public Health, Boston, MA; School of Public Health (É.J.O.), College of Medicine, University College Cork, Ireland; Epidemiology Program (L.N.K., L.L.M.), University of Hawaii Cancer Center, Honolulu; Behavioral and Epidemiology Research Group (M.L.M., V.L.S.), American Cancer Society, Atlanta, GA; Family Medicine and Public Health (A.H.S.), School of Medicine, University of California San Diego; Department of Epidemiology (L.S.), College of Public Health, University of Iowa, Iowa City; and Department of Medicine (J.E.M.) and Channing Division of Network Medicine (J.E.M., A.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Jeremy D Furtado
- From the Departments of Nutrition (É.J.O., K.B., J.D.F., A.A.) and Epidemiology (J.E.M., A.A.), Harvard T.H. Chan School of Public Health, Boston, MA; School of Public Health (É.J.O.), College of Medicine, University College Cork, Ireland; Epidemiology Program (L.N.K., L.L.M.), University of Hawaii Cancer Center, Honolulu; Behavioral and Epidemiology Research Group (M.L.M., V.L.S.), American Cancer Society, Atlanta, GA; Family Medicine and Public Health (A.H.S.), School of Medicine, University of California San Diego; Department of Epidemiology (L.S.), College of Public Health, University of Iowa, Iowa City; and Department of Medicine (J.E.M.) and Channing Division of Network Medicine (J.E.M., A.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Laurence N Kolonel
- From the Departments of Nutrition (É.J.O., K.B., J.D.F., A.A.) and Epidemiology (J.E.M., A.A.), Harvard T.H. Chan School of Public Health, Boston, MA; School of Public Health (É.J.O.), College of Medicine, University College Cork, Ireland; Epidemiology Program (L.N.K., L.L.M.), University of Hawaii Cancer Center, Honolulu; Behavioral and Epidemiology Research Group (M.L.M., V.L.S.), American Cancer Society, Atlanta, GA; Family Medicine and Public Health (A.H.S.), School of Medicine, University of California San Diego; Department of Epidemiology (L.S.), College of Public Health, University of Iowa, Iowa City; and Department of Medicine (J.E.M.) and Channing Division of Network Medicine (J.E.M., A.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Loic Le Marchand
- From the Departments of Nutrition (É.J.O., K.B., J.D.F., A.A.) and Epidemiology (J.E.M., A.A.), Harvard T.H. Chan School of Public Health, Boston, MA; School of Public Health (É.J.O.), College of Medicine, University College Cork, Ireland; Epidemiology Program (L.N.K., L.L.M.), University of Hawaii Cancer Center, Honolulu; Behavioral and Epidemiology Research Group (M.L.M., V.L.S.), American Cancer Society, Atlanta, GA; Family Medicine and Public Health (A.H.S.), School of Medicine, University of California San Diego; Department of Epidemiology (L.S.), College of Public Health, University of Iowa, Iowa City; and Department of Medicine (J.E.M.) and Channing Division of Network Medicine (J.E.M., A.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Marjorie L McCullough
- From the Departments of Nutrition (É.J.O., K.B., J.D.F., A.A.) and Epidemiology (J.E.M., A.A.), Harvard T.H. Chan School of Public Health, Boston, MA; School of Public Health (É.J.O.), College of Medicine, University College Cork, Ireland; Epidemiology Program (L.N.K., L.L.M.), University of Hawaii Cancer Center, Honolulu; Behavioral and Epidemiology Research Group (M.L.M., V.L.S.), American Cancer Society, Atlanta, GA; Family Medicine and Public Health (A.H.S.), School of Medicine, University of California San Diego; Department of Epidemiology (L.S.), College of Public Health, University of Iowa, Iowa City; and Department of Medicine (J.E.M.) and Channing Division of Network Medicine (J.E.M., A.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Victoria L Stevens
- From the Departments of Nutrition (É.J.O., K.B., J.D.F., A.A.) and Epidemiology (J.E.M., A.A.), Harvard T.H. Chan School of Public Health, Boston, MA; School of Public Health (É.J.O.), College of Medicine, University College Cork, Ireland; Epidemiology Program (L.N.K., L.L.M.), University of Hawaii Cancer Center, Honolulu; Behavioral and Epidemiology Research Group (M.L.M., V.L.S.), American Cancer Society, Atlanta, GA; Family Medicine and Public Health (A.H.S.), School of Medicine, University of California San Diego; Department of Epidemiology (L.S.), College of Public Health, University of Iowa, Iowa City; and Department of Medicine (J.E.M.) and Channing Division of Network Medicine (J.E.M., A.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Aladdin H Shadyab
- From the Departments of Nutrition (É.J.O., K.B., J.D.F., A.A.) and Epidemiology (J.E.M., A.A.), Harvard T.H. Chan School of Public Health, Boston, MA; School of Public Health (É.J.O.), College of Medicine, University College Cork, Ireland; Epidemiology Program (L.N.K., L.L.M.), University of Hawaii Cancer Center, Honolulu; Behavioral and Epidemiology Research Group (M.L.M., V.L.S.), American Cancer Society, Atlanta, GA; Family Medicine and Public Health (A.H.S.), School of Medicine, University of California San Diego; Department of Epidemiology (L.S.), College of Public Health, University of Iowa, Iowa City; and Department of Medicine (J.E.M.) and Channing Division of Network Medicine (J.E.M., A.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Linda Snetselaar
- From the Departments of Nutrition (É.J.O., K.B., J.D.F., A.A.) and Epidemiology (J.E.M., A.A.), Harvard T.H. Chan School of Public Health, Boston, MA; School of Public Health (É.J.O.), College of Medicine, University College Cork, Ireland; Epidemiology Program (L.N.K., L.L.M.), University of Hawaii Cancer Center, Honolulu; Behavioral and Epidemiology Research Group (M.L.M., V.L.S.), American Cancer Society, Atlanta, GA; Family Medicine and Public Health (A.H.S.), School of Medicine, University of California San Diego; Department of Epidemiology (L.S.), College of Public Health, University of Iowa, Iowa City; and Department of Medicine (J.E.M.) and Channing Division of Network Medicine (J.E.M., A.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - JoAnn E Manson
- From the Departments of Nutrition (É.J.O., K.B., J.D.F., A.A.) and Epidemiology (J.E.M., A.A.), Harvard T.H. Chan School of Public Health, Boston, MA; School of Public Health (É.J.O.), College of Medicine, University College Cork, Ireland; Epidemiology Program (L.N.K., L.L.M.), University of Hawaii Cancer Center, Honolulu; Behavioral and Epidemiology Research Group (M.L.M., V.L.S.), American Cancer Society, Atlanta, GA; Family Medicine and Public Health (A.H.S.), School of Medicine, University of California San Diego; Department of Epidemiology (L.S.), College of Public Health, University of Iowa, Iowa City; and Department of Medicine (J.E.M.) and Channing Division of Network Medicine (J.E.M., A.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Alberto Ascherio
- From the Departments of Nutrition (É.J.O., K.B., J.D.F., A.A.) and Epidemiology (J.E.M., A.A.), Harvard T.H. Chan School of Public Health, Boston, MA; School of Public Health (É.J.O.), College of Medicine, University College Cork, Ireland; Epidemiology Program (L.N.K., L.L.M.), University of Hawaii Cancer Center, Honolulu; Behavioral and Epidemiology Research Group (M.L.M., V.L.S.), American Cancer Society, Atlanta, GA; Family Medicine and Public Health (A.H.S.), School of Medicine, University of California San Diego; Department of Epidemiology (L.S.), College of Public Health, University of Iowa, Iowa City; and Department of Medicine (J.E.M.) and Channing Division of Network Medicine (J.E.M., A.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
21
|
Loehfelm A, Elder MK, Boucsein A, Jones PP, Williams JM, Tups A. Docosahexaenoic acid prevents palmitate-induced insulin-dependent impairments of neuronal health. FASEB J 2020; 34:4635-4652. [PMID: 32030816 DOI: 10.1096/fj.201902517r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/19/2019] [Accepted: 01/20/2020] [Indexed: 02/06/2023]
Abstract
The importance of fatty acids (FAs) for healthy brain development and function has become more evident in the past decades. However, most studies focus on the hypothalamus as an important FA-sensing brain region involved in energy homeostasis. Less work has been done to evaluate the effects of FAs on brain regions such as the hippocampus or cortex, two important centres of learning, memory formation, and cognition. Furthermore, the mechanisms of how FAs modulate the neuronal development and function are incompletely understood. Therefore, this study examined the effects of the saturated FA palmitic acid (PA) and the polyunsaturated FA docosahexaenoic acid (DHA) on primary hippocampal and cortical cultures isolated from P0/P1 Sprague Dawley rat pups. Exposure to PA, but not DHA, resulted in severe morphological changes in primary neurons such as cell body swelling, axonal and dendritic blebbing, and a reduction in synaptic innervation, compromising healthy cell function and excitability. Pharmacological assessment revealed that the PA-mediated alterations were caused by overactivation of neuronal insulin signaling, demonstrated by insulin stimulation and phosphoinositide 3-kinase inhibition. Remarkably, co-exposure to DHA prevented all PA-induced morphological changes. This work provides new insights into how FAs can affect the cytoskeletal rearrangements and neuronal function via modulation of insulin signaling.
Collapse
Affiliation(s)
- Aline Loehfelm
- Department of Physiology, School of Medical Sciences, Centre for Neuroendocrinology and Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Megan K Elder
- Department of Anatomy, School of Medical Sciences, Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Alisa Boucsein
- Department of Physiology, School of Medical Sciences, Centre for Neuroendocrinology and Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Peter P Jones
- Department of Physiology and HeartOtago, School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | - Joanna M Williams
- Department of Anatomy, School of Medical Sciences, Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Alexander Tups
- Department of Physiology, School of Medical Sciences, Centre for Neuroendocrinology and Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| |
Collapse
|
22
|
Deng FL, Pan JX, Zheng P, Xia JJ, Yin BM, Liang WW, Li YF, Wu J, Xu F, Wu QY, Qu CH, Li W, Wang HY, Xie P. Metabonomics reveals peripheral and central short-chain fatty acid and amino acid dysfunction in a naturally occurring depressive model of macaques. Neuropsychiatr Dis Treat 2019; 15:1077-1088. [PMID: 31118641 PMCID: PMC6501704 DOI: 10.2147/ndt.s186071] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
PURPOSE Depression is a complex psychiatric disorder. Various depressive rodent models are usually constructed based on different pathogenesis hypotheses. MATERIALS AND METHODS Herein, using our previously established naturally occurring depressive (NOD) model in a non-human primate (cynomolgus monkey, Macaca fascularis), we performed metabolomics analysis of cerebrospinal fluid (CSF) from NOD female macaques (N=10) and age-and gender-matched healthy controls (HCs) (N=12). Multivariate statistical analysis was used to identify the differentially expressed metabolites between the two groups. Ingenuity Pathways Analysis and MetaboAnalyst were applied for predicted pathways and biological functions analysis. RESULTS Totally, 37 metabolites responsible for discriminating the two groups were identified. The NOD macaques were mainly characterized by perturbations of fatty acid biosynthesis, ABC transport system, and amino acid metabolism (eg, aspartate, glycine, serine, and threonine metabolism). Interestingly, we found that eight altered CSF metabolites belonging to short-chain fatty acids and amino acids were also observed in the serum of NOD macaques (N=13 per group). CONCLUSION Our findings suggest that peripheral and central short-chain fatty acids and amino acids are implicated in the onset of depression.
Collapse
Affiliation(s)
- Feng-Li Deng
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402460, People's Republic of China, .,Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China, .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China, .,School of Public Health and Management, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Jun-Xi Pan
- Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China, .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China, .,The First Affiliated Hospital of Kunming Medical University, Kunming 650032, People's Republic of China
| | - Peng Zheng
- Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China, .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China, .,Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Jin-Jun Xia
- Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China, .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China,
| | - Bang-Min Yin
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402460, People's Republic of China, .,Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China, .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China,
| | - Wei-Wei Liang
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402460, People's Republic of China, .,Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China, .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China,
| | - Yi-Fan Li
- Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China, .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China, .,Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Jing Wu
- Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China, .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China,
| | - Fan Xu
- Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China, .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China,
| | - Qing-Yuan Wu
- Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China, .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China, .,Department of Neurology, Three Gorges Central Hospital, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Chao-Hua Qu
- Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China, .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China,
| | - Wei Li
- Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China, .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China,
| | - Hai-Yang Wang
- Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China, .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China,
| | - Peng Xie
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402460, People's Republic of China, .,Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China, .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China,
| |
Collapse
|
23
|
Lee S, Youn K, Jun M. Major compounds of red ginseng oil attenuate Aβ 25-35-induced neuronal apoptosis and inflammation by modulating MAPK/NF-κB pathway. Food Funct 2018; 9:4122-4134. [PMID: 30014084 DOI: 10.1039/c8fo00795k] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
β-Amyloid (Aβ)-induced neuronal toxicity in Alzheimer's disease (AD) is associated with complex mechanisms. Thus, a multi-target approach might be suitable for AD treatment. Following our previous study on the neuroprotective effects of red ginseng oil extract, its major compounds, including linoleic acid (LA), β-sitosterol (BS), and stigmasterol (SS), were examined to elucidate the mechanism of anti-apoptosis and anti-inflammation in Aβ25-35-stimulated PC12 cells. The results showed that the three compounds mitigated Aβ25-35 toxicity by regulating oxidative stress, apoptotic responses, and pro-inflammatory mediators. LA and SS strongly regulated intrinsic apoptosis markers, such as mitochondrial membrane potential, intracellular Ca2+, Bax/Bcl-2 ratio, and caspases-9, -3, and -8. However, BS blocked only the intrinsic apoptotic pathway, particularly by suppressing Ca2+ accumulation. Furthermore, all three compounds downregulated iNOS and phospho-nuclear factor-κB, but only LA and SS inhibited the expression of cyclooxygenase-2 and phospho-IκB. In assays to evaluate MAPK expression for confirming upstream signal pathways, BS decreased the phosphorylation of p38 and ERK, but not JNK, while SS markedly decreased the phosphorylation of all three MAPKs, and LA clearly decreased the phosphorylation of ERK and JNK, but not p38. These results indicate that LA, BS, and SS act as neuroprotectives against Aβ25-35-induced injury by distinct molecular mechanisms, indicating their preventive and/or therapeutic potential to treat AD.
Collapse
Affiliation(s)
- Seonah Lee
- Department of Food Science and Nutrition, Dong-A University, Busan 604-714, Korea.
| | | | | |
Collapse
|
24
|
Schommer J, Marwarha G, Nagamoto-Combs K, Ghribi O. Palmitic Acid-Enriched Diet Increases α-Synuclein and Tyrosine Hydroxylase Expression Levels in the Mouse Brain. Front Neurosci 2018; 12:552. [PMID: 30127714 PMCID: PMC6087752 DOI: 10.3389/fnins.2018.00552] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 07/20/2018] [Indexed: 11/13/2022] Open
Abstract
Background: Accumulation of the α-synuclein (α-syn) protein and depletion of dopaminergic neurons in the substantia nigra are hallmarks of Parkinson's disease (PD). Currently, α-syn is under scrutiny as a potential pathogenic factor that may contribute to dopaminergic neuronal death in PD. However, there is a significant gap in our knowledge on what causes α-syn to accumulate and dopaminergic neurons to die. It is now strongly suggested that the nature of our dietary intake influences both epigenetic changes and disease-related genes and may thus potentially increase or reduce our risk of developing PD. Objective: In this study, we determined the extent to which a 3 month diet enriched in the saturated free fatty acid palmitate (PA) influences levels of α-syn and tyrosine hydroxylase, the rate limiting enzyme in dopamine synthesis in mice brains. Methods: We fed the m-Thy1-αSyn (m-Thy1) mouse model for PD and its matched control, the B6D2F1/J (B6D2) mouse a PA-enriched diet or a normal diet for 3 months. Levels of α-syn, tyrosine hydroxylase, and the biogenic amines dopamine and dopamine metabolites, serotonin and noradrenaline were determined. Results: We found that the PA-enriched diet induces an increase in α-syn and TH protein and mRNA expression levels in m-Thy1 transgenic mice. We also show that, while it didn't affect levels of biogenic amine content in the B6D2 mice, the PA-enriched diet significantly reduces dopamine metabolites and increases the level of serotonin in m-Thy1 mice. Conclusion: Altogether, our results demonstrate that a diet rich in the saturated fatty acid palmitate can modulate levels of α-syn, TH, dopamine, and serotonin which all are proteins and neurochemicals that play key roles in increasing or reducing the risk for many neurodegenerative diseases including PD.
Collapse
Affiliation(s)
- Jared Schommer
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, United States
| | - Gurdeep Marwarha
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, United States
| | - Kumi Nagamoto-Combs
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, United States
| | - Othman Ghribi
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, United States
| |
Collapse
|
25
|
Mocking RJT, Assies J, Ruhé HG, Schene AH. Focus on fatty acids in the neurometabolic pathophysiology of psychiatric disorders. J Inherit Metab Dis 2018. [PMID: 29524021 DOI: 10.1007/s10545-018-0158-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Continuous research into the pathophysiology of psychiatric disorders, such as major depressive disorder (MDD), posttraumatic stress disorder (PTSD), and schizophrenia, suggests an important role for metabolism. This narrative review will provide an up-to-date summary of how metabolism is thought to be involved in the pathophysiology of these psychiatric disorders. We will focus on (I) the important role of fatty acids in these metabolic alterations, (II) whether fatty acid alterations represent epiphenomena or risk factors, and (III) similarities and dissociations in fatty acid alterations between different psychiatric disorders. (Historical) epidemiological evidence links fatty acid intake to psychiatric disorder prevalence, corroborated by altered fatty acid concentrations measured in psychiatric patients. These fatty acid alterations are connected with other concomitant pathophysiological mechanisms, including biological stress (hypothalamic-pituitary-adrenal (HPA)-axis and oxidative stress), inflammation, and brain network structure and function. Metabolomics and lipidomics studies are underway to more deeply investigate this complex network of associated neurometabolic alterations. Supplementation of fatty acids as disease-modifying nutraceuticals has clinical potential, particularly add-on eicosapentaenoic acid (EPA) in depressed patients with markers of increased inflammation. However, by interpreting the observed fatty acid alterations as partly (mal)adaptive phenomena, we attempt to nuance translational expectations and provide new clinical applications for these novel neurometabolic insights, e.g., to predict treatment response or depression recurrence. In conclusion, placing fatty acids in context can contribute to further understanding and optimized treatment of psychiatric disorders, in order to diminish their overwhelming burden of disease.
Collapse
Affiliation(s)
- R J T Mocking
- Department of Psychiatry, Academic Medical Center, University of Amsterdam, Meibergdreef 5, Amsterdam, 1105 AZ, The Netherlands.
| | - J Assies
- Department of Psychiatry, Academic Medical Center, University of Amsterdam, Meibergdreef 5, Amsterdam, 1105 AZ, The Netherlands
| | - H G Ruhé
- Department of Psychiatry, Academic Medical Center, University of Amsterdam, Meibergdreef 5, Amsterdam, 1105 AZ, The Netherlands
- Warneford Hospital, Department of Psychiatry, University of Oxford, Oxford, UK
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, the Netherlands
| | - A H Schene
- Department of Psychiatry, Academic Medical Center, University of Amsterdam, Meibergdreef 5, Amsterdam, 1105 AZ, The Netherlands
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen, Nijmegen, the Netherlands
| |
Collapse
|
26
|
Ng YW, Say YH. Palmitic acid induces neurotoxicity and gliatoxicity in SH-SY5Y human neuroblastoma and T98G human glioblastoma cells. PeerJ 2018; 6:e4696. [PMID: 29713567 PMCID: PMC5924683 DOI: 10.7717/peerj.4696] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 04/12/2018] [Indexed: 12/21/2022] Open
Abstract
Background Obesity-related central nervous system (CNS) pathologies like neuroinflammation and reactive gliosis are associated with high-fat diet (HFD) related elevation of saturated fatty acids like palmitic acid (PA) in neurons and astrocytes of the brain. Methods Human neuroblastoma cells SH-SY5Y (as a neuronal model) and human glioblastoma cells T98G (as an astrocytic model), were treated with 100–500 µM PA, oleic acid (OA) or lauric acid (LA) for 24 h or 48 h, and their cell viability was assessed by 3-(4,5-dimetylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The effects of stable overexpression of γ-synuclein (γ-syn), a neuronal protein recently recognized as a novel regulator of lipid handling in adipocytes, and transient overexpression of Parkinson’s disease (PD) α-synuclein [α-syn; wild-type (wt) and its pathogenic mutants A53T, A30P and E46K] in SH-SY5Y and T98G cells, were also evaluated. The effects of co-treatment of PA with paraquat (PQ), a Parkinsonian pesticide, and leptin, a hormone involved in the brain-adipose axis, were also assessed. Cell death mode and cell cycle were analyzed by Annexin V/PI flow cytometry. Reactive oxygen species (ROS) level was determined using 2′,7′-dichlorofluorescien diacetate (DCFH-DA) assay and lipid peroxidation level was determined using thiobarbituric acid reactive substances (TBARS) assay. Results MTT assay revealed dose- and time-dependent PA cytotoxicity on SH-SY5Y and T98G cells, but not OA and LA. The cytotoxicity was significantly lower in SH-SY5Y-γ-syn cells, while transient overexpression of wt α-syn or its PD mutants (A30P and E46K, but not A53T) modestly (but still significantly) rescued the cytotoxicity of PA in SH-SY5Y and T98G cells. Co-treatment of increasing concentrations of PQ exacerbated PA’s neurotoxicity. Pre-treatment of leptin, an anti-apoptotic adipokine, did not successfully rescue SH-SY5Y cells from PA-induced cytotoxicity—suggesting a mechanism of PA-induced leptin resistance. Annexin V/PI flow cytometry analysis revealed PA-induced increase in percentages of cells in annexin V-positive/PI-negative quadrant (early apoptosis) and subG0-G1 fraction, accompanied by a decrease in G2-M phase cells. The PA-induced ROS production and lipid peroxidation was at greater extent in T98G as compared to that in SH-SY5Y. Discussion In conclusion, PA induces apoptosis by increasing oxidative stress in neurons and astrocytes. Taken together, the results suggest that HFD may cause neuronal and astrocytic damage, which indirectly proposes that CNS pathologies involving neuroinflammation and reactive gliosis could be prevented via the diet regimen.
Collapse
Affiliation(s)
- Yee-Wen Ng
- Department of Biomedical Science, Faculty of Science, Universiti Tunku Abdul Rahman (UTAR) Kampar Campus, Kampar, Perak, Malaysia
| | - Yee-How Say
- Department of Biomedical Science, Faculty of Science, Universiti Tunku Abdul Rahman (UTAR) Kampar Campus, Kampar, Perak, Malaysia
| |
Collapse
|
27
|
|
28
|
Youn K, Lee S, Jun M. Gamma-linolenic acid ameliorates Aβ-induced neuroinflammation through NF-κB and MAPK signalling pathways. J Funct Foods 2018. [DOI: 10.1016/j.jff.2017.12.065] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
29
|
Trovato Salinaro A, Pennisi M, Di Paola R, Scuto M, Crupi R, Cambria MT, Ontario ML, Tomasello M, Uva M, Maiolino L, Calabrese EJ, Cuzzocrea S, Calabrese V. Neuroinflammation and neurohormesis in the pathogenesis of Alzheimer's disease and Alzheimer-linked pathologies: modulation by nutritional mushrooms. IMMUNITY & AGEING 2018; 15:8. [PMID: 29456585 PMCID: PMC5813410 DOI: 10.1186/s12979-017-0108-1] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 12/28/2017] [Indexed: 02/08/2023]
Abstract
Human life develops and expands not only in time and space, but also in the retrograde permanent recollection and interweaving of memories. Therefore, individual human identity depends fully on a proper access to the autobiographical memory. Such access is hindered or lost under pathological conditions such as Alzheimer’s disease, including recently associated oxidant pathologies, such as ocular neural degeneration occurring in glaucoma or neurosensorial degeneration occurring in Menière’s disease. Oxidative stress and altered antioxidant systems have been suggested to play a role in the aetiology of major neurodegenerative disorders, and altered expression of genes sensing oxidative stress, as well as decreased cellular stress response mechanisms could synergistically contribute to the course of these oxidant disorders. Thus, the theory that low levels of stress can produce protective responses against the pathogenic processes is a frontier area of neurobiological research focal to understanding and developing therapeutic approaches to neurodegenerative disorders. Herein, we discuss cellular mechanisms underlying AD neuroinflammatory pathogenesis that are contributory to Alzheimer’s disease. We describe endogenous cellular defence mechanism modulation and neurohormesis as a potentially innovative approach to therapeutics for AD and other neurodegenerative conditions that are associated with mitochondrial dysfunction and neuroinflammation. Particularly, we consider the emerging role of the inflammasome as an important component of the neuroprotective network, as well as the importance of Coriolus and Hericium nutritional mushrooms in redox stress responsive mechanisms and neuroprotection.
Collapse
Affiliation(s)
- Angela Trovato Salinaro
- 1Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy
| | - Manuela Pennisi
- 1Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy.,Spinal Unit, Emergency Hospital "Cannizzaro", Catania, Italy
| | - Rosanna Di Paola
- 2Department of Chemical, Biological, Pharmaceutical and Environmental Sciences University of Messina, Messina, Italy
| | - Maria Scuto
- 1Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy
| | - Rosalia Crupi
- 2Department of Chemical, Biological, Pharmaceutical and Environmental Sciences University of Messina, Messina, Italy
| | - Maria Teresa Cambria
- 1Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy
| | - Maria Laura Ontario
- 1Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy
| | - Mario Tomasello
- 1Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy
| | - Maurizio Uva
- 3Department of Medical and Surgery Sciences and Advanced Technology, University of Catania, Catania, Italy
| | - Luigi Maiolino
- 3Department of Medical and Surgery Sciences and Advanced Technology, University of Catania, Catania, Italy
| | - Edward J Calabrese
- Environmental Health Sciences Division, School of Public Health, University of Massachusetts, Amherst, MA USA
| | - Salvatore Cuzzocrea
- 2Department of Chemical, Biological, Pharmaceutical and Environmental Sciences University of Messina, Messina, Italy
| | - Vittorio Calabrese
- 1Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy
| |
Collapse
|
30
|
Abdullah L, Evans JE, Emmerich T, Crynen G, Shackleton B, Keegan AP, Luis C, Tai L, LaDu MJ, Mullan M, Crawford F, Bachmeier C. APOE ε4 specific imbalance of arachidonic acid and docosahexaenoic acid in serum phospholipids identifies individuals with preclinical Mild Cognitive Impairment/Alzheimer's Disease. Aging (Albany NY) 2017; 9:964-985. [PMID: 28333036 PMCID: PMC5391242 DOI: 10.18632/aging.101203] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 03/11/2017] [Indexed: 01/09/2023]
Abstract
This study was designed to explore the influence of apolipoprotein E (APOE) on blood phospholipids (PL) in predicting preclinical Alzheimer's disease (AD). Lipidomic analyses were also performed on blood from an AD mouse model expressing human APOE isoforms (EFAD) and five AD mutations and from 195 cognitively normal participants, 23 of who converted to mild cognitive impairment (MCI)/AD within 3 years. APOE ε4-carriers converting to MCI/AD had high arachidonic acid (AA)/docosahexaenoic acid (DHA) ratios in PL compared to cognitively normal ε4 and non-ε4 carriers. Arachidonic acid and DHA containing PL species, ε4-status and Aβ42/Aβ40 ratios provided 91% accuracy in detecting MCI/AD. Fish oil/omega-3 fatty acid consumption was associated with lower AA/DHA ratios even among ε4 carriers. High plasma AA/DHA ratios were observed in E4FAD compared to EFAD mice with other isoforms. In particular, alterations in plasma AA and DHA containing PL species were also observed in the brains of E4FAD mice compared to E3FAD mice. Despite the small sample size and a short follow-up, these results suggest that blood PL could potentially serve as biomarkers of preclinical MCI/AD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Leon Tai
- University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Mary J LaDu
- University of Illinois at Chicago, Chicago, IL 60607, USA
| | | | | | | |
Collapse
|
31
|
Jernberg JN, Bowman CE, Wolfgang MJ, Scafidi S. Developmental regulation and localization of carnitine palmitoyltransferases (CPTs) in rat brain. J Neurochem 2017; 142:407-419. [PMID: 28512781 DOI: 10.1111/jnc.14072] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 04/24/2017] [Accepted: 05/06/2017] [Indexed: 11/28/2022]
Abstract
While the brain's high energy demands are largely met by glucose, brain is also equipped with the ability to oxidize fatty acids for energy and metabolism. The brain expresses the carnitine palmitoyltransferases (CPTs) that mediate carnitine-dependent entry of long-chain acyl-CoAs into the mitochondrial matrix for β-oxidation - CPT1a and CPT2 located on the outer and inner mitochondrial membranes, respectively. Their developmental profile, regional distribution and activity as well as cell type expression remain unknown. We determined that brain CPT1a RNA and total protein expression were unchanged throughout post-natal development (PND0, PND7, PND14, PND21 and PND50); however, CPT2 RNA peaked at PND 21 and remained unchanged through PND50 in all regions studied (cortex, hippocampus, midbrain, and cerebellum). Both long-chain acyl CoA dehydrogenase and medium acyl-CoA dehydrogenase showed a similar developmental profile to CPT2. Acylcarnitines, generated as a result of CPT1a activity, significantly increased with age and peaked at PND21 in all brain regions, concurrent with the increased expression of enzymes involved in mitochondrial β-oxidation. The CPT system is highly enriched in vivo in hippocampus and cerebellum, relative to cortex and midbrain, and is exclusively present in astrocytes and neural progenitor cells, while absent in neurons, microglia, and oligodendrocytes. Using radiolabeled oleate, we demonstrate regional differences in brain fatty acid oxidation that may be blocked by the irreversible CPT1a inhibitor etomoxir. This study contributes to the field of knowledge in brain cell-specific metabolic pathways, which are important for understanding normal brain development and aging, as well as pathophysiology of neurological diseases. Read the Editorial Comment for this article on page 347.
Collapse
Affiliation(s)
- Jennifer N Jernberg
- Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Caitlyn E Bowman
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael J Wolfgang
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Susanna Scafidi
- Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
32
|
Kumar S, Downie Ruiz Velasco A, Michlewski G. Oleic Acid Induces MiR-7 Processing through Remodeling of Pri-MiR-7/Protein Complex. J Mol Biol 2017; 429:1638-1649. [PMID: 28483648 PMCID: PMC5462424 DOI: 10.1016/j.jmb.2017.05.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 05/01/2017] [Accepted: 05/02/2017] [Indexed: 12/13/2022]
Abstract
MicroRNAs (miRs) play a vital role in governing cell function, with their levels tightly controlled at transcriptional and post-transcriptional levels. Different sets of RNA-binding proteins interact with primary miRs (pri-miRs) and precursor-miR transcripts (pre-miRs), controlling their biogenesis post-transcriptionally. The Hu antigen R (HuR)-mediated binding of Musashi homolog2 (MSI2) to the conserved terminal loop of pri-miR-7 regulates the levels of brain-enriched miR-7 formation in a tissue-specific manner. Here, we show that oleic acid (OA) inhibits the binding of proteins containing RNA recognition motifs (RRM) to the conserved terminal loop of pri-miR-7. Using electrophoretic mobility shift assays in HeLa cell extracts, we show that OA treatment disrupts pre-miR/protein complexes. Furthermore, OA rescues in vitro processing of pri-miR-7, which is otherwise blocked by HuR and MSI2 proteins. On the contrary, pri-miR-16 shows reduced processing in the presence of OA. This indicates that OA may inhibit the binding of other RRM-containing protein/s necessary for miR-16 processing. Finally, we demonstrate that OA induces mature miR-7 production in HeLa cells. Together, our results demonstrate that OA can regulate the processing of pri-miRs by remodeling their protein complexes. This provides a new tool to study RNA processing and a potential lead for small molecules that target the miR-7 biogenesis pathway. OA inhibits the binding of proteins containing RRM. OA rescues processing of pri-miR-7 in vitro. OA induces mature miR-7 production in HeLa cells.
Collapse
Affiliation(s)
- Santosh Kumar
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Michael Swann Building, Edinburgh, EH9 3BF, UK
| | - Angela Downie Ruiz Velasco
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Michael Swann Building, Edinburgh, EH9 3BF, UK
| | - Gracjan Michlewski
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Michael Swann Building, Edinburgh, EH9 3BF, UK.
| |
Collapse
|
33
|
Grabacka M, Pierzchalska M, Dean M, Reiss K. Regulation of Ketone Body Metabolism and the Role of PPARα. Int J Mol Sci 2016; 17:ijms17122093. [PMID: 27983603 PMCID: PMC5187893 DOI: 10.3390/ijms17122093] [Citation(s) in RCA: 211] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 12/06/2016] [Accepted: 12/07/2016] [Indexed: 12/28/2022] Open
Abstract
Ketogenesis and ketolysis are central metabolic processes activated during the response to fasting. Ketogenesis is regulated in multiple stages, and a nuclear receptor peroxisome proliferator activated receptor α (PPARα) is one of the key transcription factors taking part in this regulation. PPARα is an important element in the metabolic network, where it participates in signaling driven by the main nutrient sensors, such as AMP-activated protein kinase (AMPK), PPARγ coactivator 1α (PGC-1α), and mammalian (mechanistic) target of rapamycin (mTOR) and induces hormonal mediators, such as fibroblast growth factor 21 (FGF21). This work describes the regulation of ketogenesis and ketolysis in normal and malignant cells and briefly summarizes the positive effects of ketone bodies in various neuropathologic conditions.
Collapse
Affiliation(s)
- Maja Grabacka
- Department of Food Biotechnology, Faculty of Food Technology, University of Agriculture, ul. Balicka 122, 30-149 Kraków, Poland.
| | - Malgorzata Pierzchalska
- Department of Food Biotechnology, Faculty of Food Technology, University of Agriculture, ul. Balicka 122, 30-149 Kraków, Poland.
| | - Matthew Dean
- Neurological Cancer Research, Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, 1700 Tulane Ave, New Orleans, LA 70112, USA.
| | - Krzysztof Reiss
- Neurological Cancer Research, Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, 1700 Tulane Ave, New Orleans, LA 70112, USA.
| |
Collapse
|
34
|
Phan CW, David P, Naidu M, Wong KH, Sabaratnam V. Therapeutic potential of culinary-medicinal mushrooms for the management of neurodegenerative diseases: diversity, metabolite, and mechanism. Crit Rev Biotechnol 2016; 35:355-68. [PMID: 24654802 DOI: 10.3109/07388551.2014.887649] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Mushrooms have long been used not only as food but also for the treatment of various ailments. Although at its infancy, accumulated evidence suggested that culinary-medicinal mushrooms may play an important role in the prevention of many age-associated neurological dysfunctions, including Alzheimer's and Parkinson's diseases. Therefore, efforts have been devoted to a search for more mushroom species that may improve memory and cognition functions. Such mushrooms include Hericium erinaceus, Ganoderma lucidum, Sarcodon spp., Antrodia camphorata, Pleurotus giganteus, Lignosus rhinocerotis, Grifola frondosa, and many more. Here, we review over 20 different brain-improving culinary-medicinal mushrooms and at least 80 different bioactive secondary metabolites isolated from them. The mushrooms (either extracts from basidiocarps/mycelia or isolated compounds) reduced beta amyloid-induced neurotoxicity and had anti-acetylcholinesterase, neurite outgrowth stimulation, nerve growth factor (NGF) synthesis, neuroprotective, antioxidant, and anti-(neuro)inflammatory effects. The in vitro and in vivo studies on the molecular mechanisms responsible for the bioactive effects of mushrooms are also discussed. Mushrooms can be considered as useful therapeutic agents in the management and/or treatment of neurodegeneration diseases. However, this review focuses on in vitro evidence and clinical trials with humans are needed.
Collapse
Affiliation(s)
- Chia-Wei Phan
- a Mushroom Research Centre, Institute of Biological Sciences, Faculty of Science, University of Malaya , Kuala Lumpur , Malaysia
| | | | | | | | | |
Collapse
|
35
|
Lavandera JV, Saín J, Fariña AC, Bernal CA, González MA. N-3 fatty acids reduced trans fatty acids retention and increased docosahexaenoic acid levels in the brain. Nutr Neurosci 2016; 20:424-435. [PMID: 27098669 DOI: 10.1080/1028415x.2016.1173343] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION The levels of docosahexaenoic acid (DHA, 22:6n-3) and arachidonic acid (AA, 20:4n-6) are critical for the normal structure and function of the brain. Trans fatty acids (TFA) and the source of the dietary fatty acids (FA) interfere with long-chain polyunsaturated fatty acids (LC-PUFA) biosynthesis. OBJECTIVES The aim of this study was to investigate the effect of TFA supplementation in diets containing different proportions of n-9, n-6, and n-3 FA on the brain FA profile, including the retention of TFA, LC-PUFA levels, and n-6/n-3 PUFA ratios. These parameters were also investigated in the liver, considering that LC-PUFA are mainly bioconverted from their dietary precursors in this tissue and transported by serum to the brain. Also, stearoyl-CoA desaturase-1 (SCD1) and sterol regulatory element-binding protein-1c (SREBP-1c) gene expressions were evaluated. METHODS Male CF1 mice were fed (16 weeks) diets containing different oils (olive, corn, and rapeseed) with distinct proportions of n-9, n-6, and n-3 FA (55.2/17.2/0.7, 32.0/51.3/0.9, and 61.1/18.4/8.6), respectively, substituted or not with 0.75% of TFA. FA composition of the brain, liver, and serum was assessed by gas chromatography. RESULTS TFA were incorporated into, and therefore retained in the brain, liver, and serum. However, the magnitude of retention was dependent on the tissue and type of isomer. In the brain, total TFA retention was lower than 1% in all diets. DISCUSSION Dietary n-3 PUFA decreased TFA retention and increased DHA accretion in the brain. The results underscore the importance of the type of dietary FA on the retention of TFA in the brain and also on the changes of the FA profile.
Collapse
Affiliation(s)
- Jimena Verónica Lavandera
- a Cátedra de Bromatología y Nutrición, Facultad de Bioquímica y Ciencias Biológicas , Universidad Nacional del Litoral , Santa Fe , Argentina.,b Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) , Santa Fe , Argentina
| | - Juliana Saín
- a Cátedra de Bromatología y Nutrición, Facultad de Bioquímica y Ciencias Biológicas , Universidad Nacional del Litoral , Santa Fe , Argentina.,b Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) , Santa Fe , Argentina
| | - Ana Clara Fariña
- a Cátedra de Bromatología y Nutrición, Facultad de Bioquímica y Ciencias Biológicas , Universidad Nacional del Litoral , Santa Fe , Argentina
| | - Claudio Adrián Bernal
- a Cátedra de Bromatología y Nutrición, Facultad de Bioquímica y Ciencias Biológicas , Universidad Nacional del Litoral , Santa Fe , Argentina.,b Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) , Santa Fe , Argentina
| | - Marcela Aída González
- a Cátedra de Bromatología y Nutrición, Facultad de Bioquímica y Ciencias Biológicas , Universidad Nacional del Litoral , Santa Fe , Argentina
| |
Collapse
|
36
|
Heras-Sandoval D, Pedraza-Chaverri J, Pérez-Rojas JM. Role of docosahexaenoic acid in the modulation of glial cells in Alzheimer's disease. J Neuroinflammation 2016; 13:61. [PMID: 26965310 PMCID: PMC4787218 DOI: 10.1186/s12974-016-0525-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 03/03/2016] [Indexed: 01/25/2023] Open
Abstract
Docosahexaenoic acid (DHA) is an omega-3 (ω-3) long-chain polyunsaturated fatty acid (LCPUFA) relevant for brain function. It has largely been explored as a potential candidate to treat Alzheimer’s disease (AD). Clinical evidence favors a role for DHA in the improvement of cognition in very early stages of the AD. In response to stress or damage, DHA generates oxygenated derivatives called docosanoids that can activate the peroxisome proliferator-activated receptor γ (PPARγ). In conjunction with activated retinoid X receptors (RXR), PPARγ modulates inflammation, cell survival, and lipid metabolism. As an early event in AD, inflammation is associated with an excess of amyloid β peptide (Aβ) that contributes to neural insult. Glial cells are recognized to be actively involved during AD, and their dysfunction is associated with the early appearance of this pathology. These cells give support to neurons, remove amyloid β peptides from the brain, and modulate inflammation. Since DHA can modulate glial cell activity, the present work reviews the evidence about this modulation as well as the effect of docosanoids on neuroinflammation and in some AD models. The evidence supports PPARγ as a preferred target for gene modulation. The effective use of DHA and/or its derivatives in a subgroup of people at risk of developing AD is discussed.
Collapse
Affiliation(s)
- David Heras-Sandoval
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, 04510, México, DF, México.,Laboratorio de Farmacología, Subdirección de Investigación Básica, Instituto Nacional de Cancerología (INCan), Av. San Fernando #22, Tlalpan 14080, Apartado Postal 22026, México, DF, México
| | - José Pedraza-Chaverri
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, 04510, México, DF, México
| | - Jazmin M Pérez-Rojas
- Laboratorio de Farmacología, Subdirección de Investigación Básica, Instituto Nacional de Cancerología (INCan), Av. San Fernando #22, Tlalpan 14080, Apartado Postal 22026, México, DF, México.
| |
Collapse
|
37
|
Lei E, Vacy K, Boon WC. Fatty acids and their therapeutic potential in neurological disorders. Neurochem Int 2016; 95:75-84. [PMID: 26939763 DOI: 10.1016/j.neuint.2016.02.014] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 02/24/2016] [Accepted: 02/25/2016] [Indexed: 11/27/2022]
Abstract
There is little doubt that we are what we eat. Fatty acid supplementation and diets rich in fatty acids are being promoted as ways to a healthier brain. Short chain fatty acids are a product of intestinal microbiota metabolism of dietary fibre; and their derivatives are used as an anti-convulstant. They demonstrated therapeutic potential in neurodegenerative conditions as HDAC inhibitors; and while the mechanism is not well understood, have been shown to lower amyloid β in Alzheimer's Disease in preclinical studies. Medium chain fatty acids consumed as a mixture in dietary oils can induce ketogenesis without the need for a ketogentic diet. Hence, this has the potential to provide an alternative energy source to prevent neuronal cell death due to lack of glucose. Long chain fatty acids are commonly found in the diet as omega fatty acids. They act as an anti-oxidant protecting neuronal cell membranes from oxidative damage and as an anti-inflammatory mediator in the brain. We review which agents, from each fatty acid class, have the most therapeutic potential for neurological disorders (primarily Alzheimer's disease, Parkinson's disease, Autism Spectrum Disorder as well as possible applications to traumatic brain injury), by discussing what is known about their biological mechanisms from preclinical studies.
Collapse
Affiliation(s)
- Enie Lei
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Kristina Vacy
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Wah Chin Boon
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia; Dept of Anatomy and Developmental Biology, Monash University, Clayton, Victoria 3800, Australia.
| |
Collapse
|
38
|
Medema S, Mocking RJT, Koeter MWJ, Vaz FM, Meijer C, de Haan L, van Beveren NJM, Kahn R, de Haan L, van Os J, Wiersma D, Bruggeman R, Cahn W, Meijer C, Myin-Germeys I. Levels of Red Blood Cell Fatty Acids in Patients With Psychosis, Their Unaffected Siblings, and Healthy Controls. Schizophr Bull 2016; 42:358-68. [PMID: 26385764 PMCID: PMC4753602 DOI: 10.1093/schbul/sbv133] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Two recent meta-analyses showed decreased red blood cell (RBC) polyunsaturated fatty acids (FA) in schizophrenia and related disorders. However, both these meta-analyses report considerable heterogeneity, probably related to differences in patient samples between studies. Here, we investigated whether variations in RBC FA are associated with psychosis, and thus may be an intermediate phenotype of the disorder. METHODS For the present study, a total of 215 patients (87% outpatients), 187 siblings, and 98 controls were investigated for multiple FA analyses. Based on previous studies, we investigated docosahexaenoic acid (DHA), docosapentaenoic acid (DPA), arachidonic acid (AA), linoleic acid (LA), nervonic acid (NA), and eicasopentaenoic acid (EPA). On an exploratory basis, a large number of additional FA were investigated. Multilevel mixed models were used to compare the FA between the 3 groups. RESULTS Compared to controls, both patients and siblings showed significantly increased DHA, DPA, AA, and NA. LA was significantly higher in siblings compared to controls. EPA was not significantly different between the 3 groups. Also the exploratory FA were increased in patients and siblings. CONCLUSIONS We found increased RBC FA DHA, DPA, AA, and NA in patients and siblings compared to controls. The direction of change is similar in both patients and siblings, which may suggest a shared environment and/or an intermediate phenotype. Differences between patient samples reflecting stage of disorder, dietary patterns, medication use, and drug abuse are possible modifiers of FA, contributing to the heterogeneity in findings concerning FA in schizophrenia patients.
Collapse
Affiliation(s)
- Suzanne Medema
- Department of Psychiatry, Antes Center for Mental Health Care, Rotterdam, The Netherlands; Department of Psychiatry, Academic Medical Center, Amsterdam, The Netherlands
| | - Roel J T Mocking
- Department of Psychiatry, Academic Medical Center, Amsterdam, The Netherlands
| | - Maarten W J Koeter
- Department of Psychiatry, Academic Medical Center, Amsterdam, The Netherlands
| | - Frédéric M Vaz
- Laboratory of Genetic and Metabolic Diseases, Academic Medical Center, Amsterdam, The Netherlands
| | - Carin Meijer
- Department of Psychiatry, Academic Medical Center, Amsterdam, The Netherlands
| | - Lieuwe de Haan
- Department of Psychiatry, Academic Medical Center, Amsterdam, The Netherlands
| | - Nico J M van Beveren
- Department of Psychiatry, Antes Center for Mental Health Care, Rotterdam, The Netherlands; Department of Psychiatry, Erasmus MC, Rotterdam, The Netherlands; Department of Neuroscience, Erasmus MC, Rotterdam, The Netherlands;
| | - René Kahn
- Department of Psychiatry, University Medical Centre Utrecht, Rudolf Magnus Institute of Neuroscience, The Netherlands
| | - Lieuwe de Haan
- Department of Psychiatry, Academic Medical Center, Amsterdam, The Netherlands
| | - Jim van Os
- Department of Psychiatry and Psychology, Maastricht University Medical Center, The Netherlands
| | - Durk Wiersma
- Department of Psychiatry, University Medical Center Groningen, The Netherlands
| | - Richard Bruggeman
- Department of Psychiatry, University Medical Center Groningen, The Netherlands
| | - Wiepke Cahn
- Department of Psychiatry, University Medical Centre Utrecht, Rudolf Magnus Institute of Neuroscience, The Netherlands
| | - Carin Meijer
- Department of Psychiatry, Antes Center for Mental Health Care, Rotterdam, The Netherlands
| | - Inez Myin-Germeys
- Department of Psychiatry and Psychology, Maastricht University Medical Center, The Netherlands
| |
Collapse
|
39
|
|
40
|
Dornellas APS, Watanabe RLH, Pimentel GD, Boldarine VT, Nascimento CMO, Oyama LM, Ghebremeskel K, Wang Y, Bueno AA, Ribeiro EB. Deleterious effects of lard-enriched diet on tissues fatty acids composition and hypothalamic insulin actions. Prostaglandins Leukot Essent Fatty Acids 2015; 102-103:21-9. [PMID: 26525379 DOI: 10.1016/j.plefa.2015.10.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Revised: 08/28/2015] [Accepted: 10/01/2015] [Indexed: 01/14/2023]
Abstract
Altered tissue fatty acid (FA) composition may affect mechanisms involved in the control of energy homeostasis, including central insulin actions. In rats fed either standard chow or a lard-enriched chow (high in saturated/low in polyunsaturated FA, HS-LP) for eight weeks, we examined the FA composition of blood, hypothalamus, liver, and retroperitoneal, epididymal and mesenteric adipose tissues. Insulin-induced hypophagia and hypothalamic signaling were evaluated after intracerebroventricular insulin injection. HS-LP feeding increased saturated FA content in adipose tissues and serum while it decreased polyunsaturated FA content of adipose tissues, serum, and liver. Hypothalamic C20:5n-3 and C20:3n-6 contents increased while monounsaturated FA content decreased. HS-LP rats showed hyperglycemia, impaired insulin-induced hypophagia and hypothalamic insulin signaling. The results showed that, upon HS-LP feeding, peripheral tissues underwent potentially deleterious alterations in their FA composition, whist the hypothalamus was relatively preserved. However, hypothalamic insulin signaling and hypophagia were drastically impaired. These findings suggest that impairment of hypothalamic insulin actions by HS-LP feeding was not related to tissue FA composition.
Collapse
Affiliation(s)
- A P S Dornellas
- Department of Physiology, Division of Nutrition Physiology, Sao Paulo Federal University, Sao Paulo, Brazil
| | - R L H Watanabe
- Department of Physiology, Division of Nutrition Physiology, Sao Paulo Federal University, Sao Paulo, Brazil
| | - G D Pimentel
- Department of Physiology, Division of Nutrition Physiology, Sao Paulo Federal University, Sao Paulo, Brazil
| | - V T Boldarine
- Department of Physiology, Division of Nutrition Physiology, Sao Paulo Federal University, Sao Paulo, Brazil
| | - C M O Nascimento
- Department of Physiology, Division of Nutrition Physiology, Sao Paulo Federal University, Sao Paulo, Brazil
| | - L M Oyama
- Department of Physiology, Division of Nutrition Physiology, Sao Paulo Federal University, Sao Paulo, Brazil
| | - K Ghebremeskel
- Lipidomics and Nutrition Research Centre, Faculty of Life Sciences and Computing, London Metropolitan University, London, United Kingdom
| | - Y Wang
- Department of Medicine, Division of Infectious Diseases, Section of Paediatrics, Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | - A A Bueno
- Institute of Science and the Environment, University of Worcester, Worcester, United Kingdom
| | - E B Ribeiro
- Department of Physiology, Division of Nutrition Physiology, Sao Paulo Federal University, Sao Paulo, Brazil.
| |
Collapse
|
41
|
Assies J, Mocking RJT, Lok A, Koeter MWJ, Bockting CLH, Visser I, Pouwer F, Ruhé HG, Schene AH. Erythrocyte fatty acid profiles and plasma homocysteine, folate and vitamin B6 and B12 in recurrent depression: Implications for co-morbidity with cardiovascular disease. Psychiatry Res 2015; 229:992-8. [PMID: 26260568 DOI: 10.1016/j.psychres.2015.06.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 04/23/2015] [Accepted: 06/01/2015] [Indexed: 01/02/2023]
Abstract
Oxidative stress induced interactions between fatty acid (FA) and one-carbon metabolism may be involved in co-occurrence of major depressive disorder (MDD) and cardiovascular disease (CVD), which have been scarcely studied together. In 137 recurrent MDD-patients vs. 73 age- and sex-matched healthy controls, we simultaneously measured key components of one-carbon metabolism in plasma (homocysteine, folate, vitamins B6 and B12), and of FA-metabolism in red blood cell membranes [main polyunsaturated fatty acids (PUFAs) eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), and arachidonic acid (AA) and structural FA-indices (chain length, unsaturation, peroxidation)]. Results show significant positive associations of folate with EPA, DHA, and the peroxidation index, which were similar in patients and controls. After correction for confounders, these associations were lost except for EPA. Associations between B-vitamins and FA-parameters were non-significant, but also similar in patients and controls. Homocysteine and DHA were significantly less negatively associated in patients than in controls. In conclusion, these data indicate similarities but also differences in associations between parameters of one-carbon and FA-metabolism in recurrent MDD patients vs. controls, which may reflect differences in handling of oxidative stress. Further research should test the consequences of these differences, particularly the premature development of CVD in MDD.
Collapse
Affiliation(s)
- Johanna Assies
- Program for Mood Disorders, Department of Psychiatry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | - Roel J T Mocking
- Program for Mood Disorders, Department of Psychiatry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Anja Lok
- Program for Mood Disorders, Department of Psychiatry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Arq Psychotrauma Expert Group, Diemen, The Netherlands
| | - Maarten W J Koeter
- Program for Mood Disorders, Department of Psychiatry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Claudi L H Bockting
- Department of Clinical Psychology, University of Groningen, Groningen, The Netherlands; Department of Clinical and Health Psychology, University of Utrecht, Utrecht, The Netherlands
| | - Ieke Visser
- Program for Mood Disorders, Department of Psychiatry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - François Pouwer
- Department of Medical and Clinical Psychology, Center of Research on Psychology in Somatic Diseases (CoRPS), Tilburg University, The Netherlands
| | - Henricus G Ruhé
- Program for Mood Disorders, Department of Psychiatry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; University of Groningen, University Center for Psychiatry UMCG, Program for Mood and Anxiety Disorders, The Netherlands
| | - Aart H Schene
- Program for Mood Disorders, Department of Psychiatry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Psychiatry, Radboud University Medical Centre, Nijmegen, The Netherlands; Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
42
|
Alluri H, Wiggins-Dohlvik K, Davis ML, Huang JH, Tharakan B. Blood-brain barrier dysfunction following traumatic brain injury. Metab Brain Dis 2015; 30:1093-104. [PMID: 25624154 DOI: 10.1007/s11011-015-9651-7] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 01/13/2015] [Indexed: 01/24/2023]
Abstract
Traumatic brain injury is a serious cause of morbidity and mortality worldwide. After traumatic brain injury, the blood-brain barrier, the protective barrier between the brain and the intravascular compartment, becomes dysfunctional, leading to leakage of proteins, fluid, and transmigration of immune cells. As this leakage has profound clinical implications, including edema formation, elevated intracranial pressure and decreased perfusion pressure, much interest has been paid to better understanding the mechanisms responsible for these events. Various molecular pathways and numerous mediators have been found to be involved in the intricate process of regulating blood-brain barrier permeability following traumatic brain injury. This review provides an update to the existing knowledge about the various pathophysiological pathways and advancements in the field of blood-brain barrier dysfunction and hyperpermeability following traumatic brain injury, including the role of various tight junction proteins involved in blood-brain barrier integrity and regulation. We also address pitfalls of existing systems and propose strategies to improve the various debilitating functional deficits caused by this progressive epidemic.
Collapse
Affiliation(s)
- Himakarnika Alluri
- Department of Surgery, Baylor Scott & White Health & Texas A&M University Health Science Center, College of Medicine, 702 S.W. H.K. Dodgen Loop, Temple, TX, 76504, USA
| | | | | | | | | |
Collapse
|
43
|
Solomons NW, Bailey E, Soto Méndéz MJ, Campos R, Kraemer K, Salem N. Erythrocyte fatty acid status in a convenience sample of residents of the Guatemalan Pacific coastal plain. Prostaglandins Leukot Essent Fatty Acids 2015; 98:21-7. [PMID: 25979321 DOI: 10.1016/j.plefa.2015.03.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 03/24/2015] [Accepted: 03/26/2015] [Indexed: 12/16/2022]
Abstract
We report the fatty acid composition, and in particular, the n-3 and n-6 long-chain polyunsaturated fatty acids (LCPUFA), in erythrocytes from a convenience sample of 158 women and 135 schoolchildren residing in the southern Pacific Coast of Guatemala. Erythrocyte fatty acids were analyzed by gas-liquid chromatography with flame ionization detection and the profiles were expressed as a weight percent; the Omega-3 Index values were also determined. Schoolchildren had significantly higher mean ARA and total n-6 fatty acid levels than the women. Women had significantly higher EPA fatty acid levels than schoolchildren, but the reverse was true for DHA. For mean total n-3 fatty acid concentration, women and schoolchildren had similar values. The red cell weight percentages of selected fatty acids were also similar in women and schoolchildren. As compared with erythrocyte fatty acid data from developed countries, Guatemalan women and schoolchildren had consistently lower LCPUFA values. The traditional diet of Guatemalans living in the Pacific coastal region provided a worse erythrocyte fatty acid profile than that typically obtained from a Western diet. Additional fatty acid composition studies with associated dietary intake data in other inland locations may be useful for the interpretation of the nutritional status of Guatemalan children and adults.
Collapse
Affiliation(s)
- Noel W Solomons
- Center for Studies of Sensory Impairment, Aging and Metabolism (CeSSIAM), Guatemala City, Guatemala
| | - Eileen Bailey
- Nutritional Lipids, DSM Nutritional Products, Columbia, MD, USA
| | - María José Soto Méndéz
- Center for Studies of Sensory Impairment, Aging and Metabolism (CeSSIAM), Guatemala City, Guatemala
| | - Raquel Campos
- Center for Studies of Sensory Impairment, Aging and Metabolism (CeSSIAM), Guatemala City, Guatemala
| | - Klaus Kraemer
- Sight and Life, Basel, Switzerland; Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Norman Salem
- Nutritional Lipids, DSM Nutritional Products, Columbia, MD, USA.
| |
Collapse
|
44
|
Willebrords J, Pereira IVA, Maes M, Crespo Yanguas S, Colle I, Van Den Bossche B, Da Silva TC, de Oliveira CPMS, Andraus W, Alves VA, Cogliati B, Vinken M. Strategies, models and biomarkers in experimental non-alcoholic fatty liver disease research. Prog Lipid Res 2015; 59:106-25. [PMID: 26073454 DOI: 10.1016/j.plipres.2015.05.002] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 05/13/2015] [Accepted: 05/13/2015] [Indexed: 12/12/2022]
Abstract
Non-alcoholic fatty liver disease encompasses a spectrum of liver diseases, including simple steatosis, steatohepatitis, liver fibrosis and cirrhosis and hepatocellular carcinoma. Non-alcoholic fatty liver disease is currently the most dominant chronic liver disease in Western countries due to the fact that hepatic steatosis is associated with insulin resistance, type 2 diabetes mellitus, obesity, metabolic syndrome and drug-induced injury. A variety of chemicals, mainly drugs, and diets is known to cause hepatic steatosis in humans and rodents. Experimental non-alcoholic fatty liver disease models rely on the application of a diet or the administration of drugs to laboratory animals or the exposure of hepatic cell lines to these drugs. More recently, genetically modified rodents or zebrafish have been introduced as non-alcoholic fatty liver disease models. Considerable interest now lies in the discovery and development of novel non-invasive biomarkers of non-alcoholic fatty liver disease, with specific focus on hepatic steatosis. Experimental diagnostic biomarkers of non-alcoholic fatty liver disease, such as (epi)genetic parameters and '-omics'-based read-outs are still in their infancy, but show great promise. In this paper, the array of tools and models for the study of liver steatosis is discussed. Furthermore, the current state-of-art regarding experimental biomarkers such as epigenetic, genetic, transcriptomic, proteomic and metabonomic biomarkers will be reviewed.
Collapse
Affiliation(s)
- Joost Willebrords
- Department of In Vitro Toxicology and Dermato-Cosmetology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Isabel Veloso Alves Pereira
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, Av. Prof. Dr. Orlando Marques de Paiva, 87, São Paulo, Brazil.
| | - Michaël Maes
- Department of In Vitro Toxicology and Dermato-Cosmetology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Sara Crespo Yanguas
- Department of In Vitro Toxicology and Dermato-Cosmetology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Isabelle Colle
- Department of Hepatology and Gastroenterology, Algemeen Stedelijk Ziekenhuis Campus Aalst, Merestraat 80, 9300 Aalst, Belgium.
| | - Bert Van Den Bossche
- Department of Abdominal Surgery and Hepato-Pancreatico-Biliary Surgery, Algemeen Stedelijk Ziekenhuis Campus Aalst, Merestraat 80, 9300 Aalst, Belgium.
| | - Tereza Cristina Da Silva
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, Av. Prof. Dr. Orlando Marques de Paiva, 87, São Paulo, Brazil.
| | | | - Wellington Andraus
- Department of Gastroenterology, University of São Paulo School of Medicine, Av. Dr. Arnaldo, 455, São Paulo, Brazil.
| | - Venâncio Avancini Alves
- Laboratory of Medical Investigation, Department of Pathology, University of São Paulo School of Medicine, Av. Dr. Arnaldo, 455, São Paulo, Brazil.
| | - Bruno Cogliati
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, Av. Prof. Dr. Orlando Marques de Paiva, 87, São Paulo, Brazil.
| | - Mathieu Vinken
- Department of In Vitro Toxicology and Dermato-Cosmetology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| |
Collapse
|
45
|
Sartorius T, Häring HU. The Metabolic Role of Saturated and Monounsaturated Dietary Fatty Acids. MODULATION OF SLEEP BY OBESITY, DIABETES, AGE, AND DIET 2015:203-210. [DOI: 10.1016/b978-0-12-420168-2.00022-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
46
|
Barberger-Gateau P. Nutrition and brain aging: how can we move ahead? Eur J Clin Nutr 2014; 68:1245-9. [DOI: 10.1038/ejcn.2014.177] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 08/06/2014] [Indexed: 11/09/2022]
|
47
|
Guest J, Grant R, Garg M, Mori TA, Croft KD, Bilgin A. Cerebrospinal fluid levels of inflammation, oxidative stress and NAD+ are linked to differences in plasma carotenoid concentrations. J Neuroinflammation 2014; 11:117. [PMID: 24985027 PMCID: PMC4096526 DOI: 10.1186/1742-2094-11-117] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 05/27/2014] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND The consumption of foods rich in carotenoids that possess significant antioxidant and inflammatory modulating properties has been linked to reduced risk of neuropathology. The objective of this study was to evaluate the relationship between plasma carotenoid concentrations and plasma and cerebrospinal fluid (CSF) markers of inflammation, oxidative stress and nicotinamide adenine dinucleotide (NAD+) in an essentially healthy human cohort. METHODS Thirty-eight matched CSF and plasma samples were collected from consenting participants who required a spinal tap for the administration of anaesthetic. Plasma concentrations of carotenoids and both plasma and cerebrospinal fluid (CSF) levels of NAD(H) and markers of inflammation (IL-6, TNF-α) and oxidative stress (F2-isoprostanes, 8-OHdG and total antioxidant capacity) were quantified. RESULTS The average age of participants was 53 years (SD=20, interquartile range=38). Both α-carotene (P=0.01) and β-carotene (P<0.001) correlated positively with plasma total antioxidant capacity. A positive correlation was observed between α-carotene and CSF TNF-α levels (P=0.02). β-cryptoxanthin (P=0.04) and lycopene (P=0.02) inversely correlated with CSF and plasma IL-6 respectively. A positive correlation was also observed between lycopene and both plasma (P<0.001) and CSF (P<0.01) [NAD(H)]. Surprisingly no statistically significant associations were found between the most abundant carotenoids, lutein and zeaxanthin and either plasma or CSF markers of oxidative stress. CONCLUSION Together these findings suggest that consumption of carotenoids may modulate inflammation and enhance antioxidant defences within both the central nervous system (CNS) and systemic circulation. Increased levels of lycopene also appear to moderate decline in the essential pyridine nucleotide [NAD(H)] in both the plasma and the CSF.
Collapse
Affiliation(s)
- Jade Guest
- Australasian Research Institute, Sydney Adventist Hospital, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Wallace Wurth Building, office #203, Sydney, NSW 2052, Australia
| | - Ross Grant
- Australasian Research Institute, Sydney Adventist Hospital, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Wallace Wurth Building, office #203, Sydney, NSW 2052, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Manohar Garg
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia
| | - Trevor A Mori
- School of Medicine and Pharmacology, Royal Perth Hospital Unit, University of Western Australia, Perth, WA, Australia
| | - Kevin D Croft
- School of Medicine and Pharmacology, Royal Perth Hospital Unit, University of Western Australia, Perth, WA, Australia
| | - Ayse Bilgin
- Faculty of Science, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
48
|
Model CS, Gomes LM, Scaini G, Ferreira GK, Gonçalves CL, Rezin GT, Steckert AV, Valvassori SS, Varela RB, Quevedo J, Streck EL. Omega-3 fatty acids alter behavioral and oxidative stress parameters in animals subjected to fenproporex administration. Metab Brain Dis 2014; 29:185-92. [PMID: 24385143 DOI: 10.1007/s11011-013-9473-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 12/13/2013] [Indexed: 12/30/2022]
Abstract
Studies have consistently reported the participation of oxidative stress in bipolar disorder (BD). Evidences indicate that omega-3 (ω3) fatty acids play several important roles in brain development and functioning. Moreover, preclinical and clinical evidence suggests roles for ω3 fatty acids in BD. Considering these evidences, the present study aimed to investigate the effects of ω3 fatty acids on locomotor behavior and oxidative stress parameters (TBARS and protein carbonyl content) in brain of rats subjected to an animal model of mania induced by fenproporex. The fenproporex treatment increased locomotor behavior in saline-treated rats under reversion and prevention model, and ω3 fatty acids prevented fenproporex-related hyperactivity. Moreover, fenproporex increased protein carbonyls in the prefrontal cortex and cerebral cortex, and the administration of ω3 fatty acids reversed this effect. Lipid peroxidation products also are increased in prefrontal cortex, striatum, hippocampus and cerebral after fenproporex administration, but ω3 fatty acids reversed this damage only in the hippocampus. On the other hand, in the prevention model, fenproporex increased carbonyl content only in the cerebral cortex, and administration of ω3 fatty acids prevented this damage. Additionally, the administration of fenproporex resulted in a marked increased of TBARS in the prefrontal cortex, hippocampus, striatum and cerebral cortex, and prevent this damage in the prefrontal cortex, hippocampus and striatum. In conclusion, we are able to demonstrate that fenproporex-induced hyperlocomotion and damage through oxidative stress were prevented by ω3 fatty acids. Thus, the ω3 fatty acids may be important adjuvant therapy of bipolar disorder.
Collapse
Affiliation(s)
- Camila S Model
- Laboratório de Bioenergética, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Lopez S, Bermudez B, Montserrat-de la Paz S, Jaramillo S, Varela LM, Ortega-Gomez A, Abia R, Muriana FJG. Membrane composition and dynamics: a target of bioactive virgin olive oil constituents. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1838:1638-56. [PMID: 24440426 DOI: 10.1016/j.bbamem.2014.01.007] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 01/03/2014] [Accepted: 01/07/2014] [Indexed: 12/26/2022]
Abstract
The endogenous synthesis of lipids, which requires suitable dietary raw materials, is critical for the formation of membrane bilayers. In eukaryotic cells, phospholipids are the predominant membrane lipids and consist of hydrophobic acyl chains attached to a hydrophilic head group. The relative balance between saturated, monounsaturated, and polyunsaturated acyl chains is required for the organization and normal function of membranes. Virgin olive oil is the richest natural dietary source of the monounsaturated lipid oleic acid and is one of the key components of the healthy Mediterranean diet. Virgin olive oil also contains a unique constellation of many other lipophilic and amphipathic constituents whose health benefits are still being discovered. The focus of this review is the latest evidence regarding the impact of oleic acid and the minor constituents of virgin olive oil on the arrangement and behavior of lipid bilayers. We highlight the relevance of these interactions to the potential use of virgin olive oil in preserving the functional properties of membranes to maintain health and in modulating membrane functions that can be altered in several pathologies. This article is part of a Special Issue entitled: Membrane Structure and Function: Relevance in the Cell's Physiology, Pathology and Therapy.
Collapse
Affiliation(s)
- Sergio Lopez
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, CSIC, 41012 Seville, Spain
| | - Beatriz Bermudez
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, CSIC, 41012 Seville, Spain
| | | | - Sara Jaramillo
- Laboratory of Phytochemicals and Food Quality, Instituto de la Grasa, CSIC, 41014 Seville, Spain
| | - Lourdes M Varela
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, CSIC, 41012 Seville, Spain
| | - Almudena Ortega-Gomez
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, CSIC, 41012 Seville, Spain
| | - Rocio Abia
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, CSIC, 41012 Seville, Spain
| | - Francisco J G Muriana
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, CSIC, 41012 Seville, Spain.
| |
Collapse
|