1
|
Barrionuevo EM, Peralta E, Manzur De Nardi A, Monat J, Fallico MJ, Llanos MA, Gavernet L, Mustafá ER, Martin P, Talevi A. In Silico Screening Identification of Fatty Acids and Fatty Acid Derivatives with Antiseizure Activity: In Vitro and In Vivo Validation. Pharmaceutics 2024; 16:996. [PMID: 39204342 PMCID: PMC11357650 DOI: 10.3390/pharmaceutics16080996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/19/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024] Open
Abstract
High fat diets have been used as complementary treatments for seizure disorders for more than a century. Moreover, many fatty acids and derivatives, including the broad-spectrum antiseizure medication valproic acid, have been explored and used as pharmacological agents to treat epilepsy. In this work, we have explored the anticonvulsant potential of a large library of fatty acids and fatty acid derivatives, the LIPID MAPS Structure Database, using structure-based virtual screening to assess their ability to block the voltage-gated sodium channel 1.2 (NaV1.2), a validated target for antiseizure medications. Four of the resulting in silico hits were submitted for experimental confirmation using in vitro patch clamp experiments, and their protective role was evaluated in an acute mice seizure model, the Maximal Electroshock seizure model. These four compounds were found to protect mice against seizures. Two of them exhibited blocking effects on NaV1.2, CaV2.2, and CaV3.1.
Collapse
Affiliation(s)
- Emilia Mercedes Barrionuevo
- Laboratory of Bioactive Compound Research and Development (LIDeB), Faculty of Exact Sciences, National University of La Plata (UNLP), Blvd. 120 1489, La Plata 1900, Argentina
- Argentinean National Council of Scientific and Technical Research (CONICET), CCT La Plata, La Plata 1900, Argentina
| | - Estefanía Peralta
- Laboratory of Bioactive Compound Research and Development (LIDeB), Faculty of Exact Sciences, National University of La Plata (UNLP), Blvd. 120 1489, La Plata 1900, Argentina
- Argentinean National Council of Scientific and Technical Research (CONICET), CCT La Plata, La Plata 1900, Argentina
| | - Agustín Manzur De Nardi
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), Universidad Nacional de La Plata–CICPBA–CONICET, Boulevard 120 no. 1489, La Plata 1900, Argentina
| | - Juliana Monat
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), Universidad Nacional de La Plata–CICPBA–CONICET, Boulevard 120 no. 1489, La Plata 1900, Argentina
| | - Maximiliano José Fallico
- Laboratory of Bioactive Compound Research and Development (LIDeB), Faculty of Exact Sciences, National University of La Plata (UNLP), Blvd. 120 1489, La Plata 1900, Argentina
- Argentinean National Council of Scientific and Technical Research (CONICET), CCT La Plata, La Plata 1900, Argentina
| | - Manuel Augusto Llanos
- Laboratory of Bioactive Compound Research and Development (LIDeB), Faculty of Exact Sciences, National University of La Plata (UNLP), Blvd. 120 1489, La Plata 1900, Argentina
- Argentinean National Council of Scientific and Technical Research (CONICET), CCT La Plata, La Plata 1900, Argentina
| | - Luciana Gavernet
- Laboratory of Bioactive Compound Research and Development (LIDeB), Faculty of Exact Sciences, National University of La Plata (UNLP), Blvd. 120 1489, La Plata 1900, Argentina
- Argentinean National Council of Scientific and Technical Research (CONICET), CCT La Plata, La Plata 1900, Argentina
| | - Emilio Román Mustafá
- Electrophysiology Laboratory of the Multidisciplinary Institute of Cell Biology [Argentine Research Council (CONICET), Scientific Research Commission of the Province of Buenos Aires (CIC-PBA) and National University of La Plata (UNLP)], La Plata 1900, Argentina
| | - Pedro Martin
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), Universidad Nacional de La Plata–CICPBA–CONICET, Boulevard 120 no. 1489, La Plata 1900, Argentina
| | - Alan Talevi
- Laboratory of Bioactive Compound Research and Development (LIDeB), Faculty of Exact Sciences, National University of La Plata (UNLP), Blvd. 120 1489, La Plata 1900, Argentina
- Argentinean National Council of Scientific and Technical Research (CONICET), CCT La Plata, La Plata 1900, Argentina
| |
Collapse
|
2
|
Mitrović N, Adžić Bukvić M, Zarić Kontić M, Dragić M, Petrović S, Paunović M, Vučić V, Grković I. Flaxseed Oil Alleviates Trimethyltin-Induced Cell Injury and Inhibits the Pro-Inflammatory Activation of Astrocytes in the Hippocampus of Female Rats. Cells 2024; 13:1184. [PMID: 39056766 PMCID: PMC11274492 DOI: 10.3390/cells13141184] [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] [Received: 05/28/2024] [Revised: 06/28/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Exposure to the neurotoxin trimethyltin (TMT) selectively induces hippocampal neuronal injury and astrocyte activation accompanied with resultant neuroinflammation, which causes severe behavioral, cognitive, and memory impairment. A large body of evidence suggests that flaxseed oil (FSO), as one of the richest sources of essential omega-3 fatty acids, i.e., α-linolenic acids (ALA), displays neuroprotective properties. Here, we report the preventive effects of dietary FSO treatment in a rat model of TMT intoxication. The administration of FSO (1 mL/kg, orally) before and over the course of TMT intoxication (a single dose, 8 mg/kg, i.p.) reduced hippocampal cell death, prevented the activation of astrocytes, and inhibited their polarization toward a pro-inflammatory/neurotoxic phenotype. The underlying protective mechanism was delineated through the selective upregulation of BDNF and PI3K/Akt and the suppression of ERK activation in the hippocampus. Pretreatment with FSO reduced cell death and efficiently suppressed the expression of inflammatory molecules. These beneficial effects were accompanied by an increased intrahippocampal content of n-3 fatty acids. In vitro, ALA pretreatment prevented the TMT-induced polarization of cultured astrocytes towards the pro-inflammatory spectrum. Together, these findings support the beneficial neuroprotective properties of FSO/ALA against TMT-induced neurodegeneration and accompanied inflammation and hint at a promising preventive use of FSO in hippocampal degeneration and dysfunction.
Collapse
Affiliation(s)
- Nataša Mitrović
- Department of Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (M.Z.K.); (I.G.)
| | - Marija Adžić Bukvić
- Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (M.A.B.); (M.D.)
- Center for Laser Microscopy, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia
| | - Marina Zarić Kontić
- Department of Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (M.Z.K.); (I.G.)
| | - Milorad Dragić
- Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (M.A.B.); (M.D.)
| | - Snježana Petrović
- Group for Nutritional Biochemistry and Dietology, Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (S.P.); (M.P.); (V.V.)
| | - Marija Paunović
- Group for Nutritional Biochemistry and Dietology, Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (S.P.); (M.P.); (V.V.)
| | - Vesna Vučić
- Group for Nutritional Biochemistry and Dietology, Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (S.P.); (M.P.); (V.V.)
| | - Ivana Grković
- Department of Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (M.Z.K.); (I.G.)
| |
Collapse
|
3
|
Kim OY, Song J. Important roles of linoleic acid and α-linolenic acid in regulating cognitive impairment and neuropsychiatric issues in metabolic-related dementia. Life Sci 2024; 337:122356. [PMID: 38123015 DOI: 10.1016/j.lfs.2023.122356] [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] [Received: 09/23/2023] [Revised: 12/02/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
Metabolic syndrome (MetS), which is characterized by insulin resistance, high blood glucose, obesity, and dyslipidemia, is known to increase the risk of dementia accompanied by memory loss and depression. The direct pathways and specific mechanisms in the central nervous system (CNS) for addressing fatty acid imbalances in MetS have not yet been fully elucidated. Among polyunsaturated acids, linoleic acid (LA, n6-PUFA) and α-linolenic acid (ALA, n3-PUFA), which are two essential fatty acids that should be provided by food sources (e.g., vegetable oils and seeds), have been reported to regulate various cellular mechanisms including apoptosis, inflammatory responses, mitochondrial biogenesis, and insulin signaling. Furthermore, inadequate intake of LA and ALA is reported to be involved in neuropathology and neuropsychiatric diseases as well as imbalanced metabolic conditions. Herein, we review the roles of LA and ALA on metabolic-related dementia focusing on insulin resistance, dyslipidemia, synaptic plasticity, cognitive function, and neuropsychiatric issues. This review suggests that LA and ALA are important fatty acids for concurrent treatment of both MetS and neurological problems.
Collapse
Affiliation(s)
- Oh Yoen Kim
- Department of Food Science and Nutrition, Dong A University, Busan, Republic of Korea; Department of Health Sciences, Graduate School of Dong-A University, Busan, Republic of Korea.
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Seoul, Republic of Korea.
| |
Collapse
|
4
|
Hemida M, Rosendahl S, Jokinen TS, Moore R, Vuori KA, Anturaniemi J, Hielm-Björkman A. Assessing the association between supplemented puppyhood dietary fat sources and owner-reported epilepsy in adulthood, among Finnish companion dogs. Front Vet Sci 2023; 10:1227437. [PMID: 37781290 PMCID: PMC10540444 DOI: 10.3389/fvets.2023.1227437] [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: 05/23/2023] [Accepted: 08/25/2023] [Indexed: 10/03/2023] Open
Abstract
Introduction Epilepsy is a serious and common neurological condition in dogs, despite the wide number of antiepileptic drugs available, in approximately one third of the patients, epilepsy remains unsatisfactorily controlled. We aim to analyze whether feeding dietary fat sources during puppyhood was associated with canine epilepsy in adulthood. Methods A nested case-control study was compiled from the validated DogRisk food frequency questionnaire (DogRisk FFQ). DogRisk FFQ collected feeding, disease, and background data about the dog. The study sample consisted of 108 owner-reported epileptic cases and 397 non-epileptic controls. Each case was matched with up to four controls for the key confounding factors of sex, breed, and age. We analyzed associations between feeding as a puppy and owner-reported epilepsy as an adult dog using Cox regression. We tested 55 different food variables. Results We found that feeding fish fat from dietary sources at least once a week during puppyhood was inversely associated with epilepsy in later life in the unadjusted analysis [OR 0.46 (95% CI 0.25-0.83), p=0.01], while when adjusting for keeping conditions and dog characteristics the association was [OR 0.45 (95% CI 0.23-0.88), p=0.02]. When adjusted for keeping conditions, dog characteristics, and other feeding factors, the association was of similar magnitude but not significance [OR 0.56 (95% CI 0.27-1.15), p=0.12]. Discussion The study indicates possible protective associations of feeding the dog with dietary sources of fish fat against epilepsy, although the result could be confounded by other feeding factors. Findings are compatible with current knowledge regarding the role of omega-3 fatty acids and ketogenic diet, a low carbohydrate, high fat diet as supportive treatments of epilepsy. As our findings are based on observations, we suggest the possibility of causality but do not prove it. Dietary intervention studies should now be conducted to confirm our findings.
Collapse
Affiliation(s)
- Manal Hemida
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Beni-Suef University, Beni Suef, Egypt
| | - Sarah Rosendahl
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Tarja S. Jokinen
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Robin Moore
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Kristiina A. Vuori
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Johanna Anturaniemi
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Anna Hielm-Björkman
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
5
|
Ríos C, Aguirre-Aranda I, Avendaño-Estrada A, Ángel Ávila-Rodríguez M, Manjarrez-Marmolejo J, Franco-Pérez J, Islas-Cortez M, Ruiz-Diaz A, Méndez-Armenta M, Diaz-Ruiz A. Characterization of the anticonvulsant effect of dapsone on metabolic activity assessed by [ 18F]FDG -PET after kainic acid-induced status epilepticus in rats. Brain Res 2023; 1803:148227. [PMID: 36592802 DOI: 10.1016/j.brainres.2022.148227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 12/28/2022] [Accepted: 12/29/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND Development of effective drugs for epilepsy are needed, as nearly 30 % of epileptic patients, are resistant to current treatments. This study is aimed to characterize the anticonvulsant effect of dapsone (DDS), in the kainic acid (KA)-induced Status Epilepticus (SE) by recording the brain metabolic activity with an [18F]FDG-PET analysis. METHODS Wistar rats received KA (10 mg/kg, i.p., single dose) to produce sustained seizures. [18F]FDG-PET and electroencephalographic (EEG) studies were then performed. DDS or vehicle were administered 30 min before KA. [18F]FDG uptake and EEG were evaluated at baseline, 2 and 25 h after KA injection. Likewise, caspase-8, 3 hippocampal activities and Fluoro-Jade B neuronal degeneration and Hematoxylin-eosin staining were measured 25 h after KA. RESULTS PET data evaluated at 2 h showed hyper-uptake of [18F]FDG in the control group, which was decreased by DDS. At 25 h, hypo-uptake was observed in the control group and higher values due to DDS effect. EEG spectral power was increased 2 h after KA administration in the control group during the generalized tonic-clonic seizures, which was reversed by DDS, correlated with [18F]FDG-PET uptake changes. The values of caspases-8 activity decreased 48 and 43 % vs control group in the groups treated with DDS (12.5 y 25 mg/kg respectively), likewise; caspase-3 activity diminished by 57 and 53 %. Fewer degenerated neurons were observed due to DDS treatments. CONCLUSIONS This study pinpoints the anticonvulsant therapeutic potential of DDS. Given its safety and effectiveness, DDS may be a viable alternative for patients with drug-resistant epilepsy.
Collapse
Affiliation(s)
- Camilo Ríos
- Departamento de Neuroquímica Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez,14269 Ciudad de México, Mexico; Laboratorio de Neurofarmacología Molecular, Universidad Autónoma Metropolitana Xochimilco,04960 Ciudad de México, Mexico
| | - Iñigo Aguirre-Aranda
- Departamento de Neuroquímica Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez,14269 Ciudad de México, Mexico
| | - Arturo Avendaño-Estrada
- Unidad Radiofarmacia-Ciclotrón, División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México, 04510 Ciudad de México, Mexico
| | - Miguel Ángel Ávila-Rodríguez
- Unidad Radiofarmacia-Ciclotrón, División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México, 04510 Ciudad de México, Mexico
| | - Joaquín Manjarrez-Marmolejo
- Laboratorio de Fisiología de la Formación Reticular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, 14269 Ciudad de México. Mexico
| | - Javier Franco-Pérez
- Laboratorio de Fisiología de la Formación Reticular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, 14269 Ciudad de México. Mexico
| | - Marcela Islas-Cortez
- Doctorado en Ciencias Químico Biológicas, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Amairani Ruiz-Diaz
- Departamento de Neuroquímica Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez,14269 Ciudad de México, Mexico
| | - Marisela Méndez-Armenta
- Departamento de Neuroquímica Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez,14269 Ciudad de México, Mexico
| | - Araceli Diaz-Ruiz
- Departamento de Neuroquímica Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez,14269 Ciudad de México, Mexico.
| |
Collapse
|
6
|
Senn L, Costa AM, Avallone R, Socała K, Wlaź P, Biagini G. Is the peroxisome proliferator-activated receptor gamma a putative target for epilepsy treatment? Current evidence and future perspectives. Pharmacol Ther 2023; 241:108316. [PMID: 36436690 DOI: 10.1016/j.pharmthera.2022.108316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/20/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022]
Abstract
The peroxisome proliferator-activated receptor gamma (PPARγ), which belongs to the family of nuclear receptors, has been mainly studied as an important factor in metabolic disorders. However, in recent years the potential role of PPARγ in different neurological diseases has been increasingly investigated. Especially, in the search of therapeutic targets for patients with epilepsy the question of the involvement of PPARγ in seizure control has been raised. Epilepsy is a chronic neurological disorder causing a major impact on the psychological, social, and economic conditions of patients and their families, besides the problems of the disease itself. Considering that the world prevalence of epilepsy ranges between 0.5% - 1.0%, this condition is the fourth for importance among the other neurological disorders, following migraine, stroke, and dementia. Among others, temporal lobe epilepsy (TLE) is the most common form of epilepsy in adult patients. About 65% of individuals who receive antiseizure medications (ASMs) experience seizure independence. For those in whom seizures still recur, investigating PPARγ could lead to the development of novel ASMs. This review focuses on the most important findings from recent investigations about the potential intracellular PPARγ-dependent processes behind different compounds that exhibited anti-seizure effects. Additionally, recent clinical investigations are discussed along with the promising results found for PPARγ agonists and the ketogenic diet (KD) in various rodent models of epilepsy.
Collapse
Affiliation(s)
- Lara Senn
- Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; PhD School of Clinical and Experimental Medicine (CEM), University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Anna-Maria Costa
- Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Rossella Avallone
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Katarzyna Socała
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, PL 20-033 Lublin, Poland
| | - Piotr Wlaź
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, PL 20-033 Lublin, Poland
| | - Giuseppe Biagini
- Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy.
| |
Collapse
|
7
|
Kumar A, Kumari S, Singh D. Insights into the Cellular Interactions and Molecular Mechanisms of Ketogenic Diet for Comprehensive Management of Epilepsy. Curr Neuropharmacol 2022; 20:2034-2049. [PMID: 35450526 PMCID: PMC9886834 DOI: 10.2174/1570159x20666220420130109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 01/27/2022] [Accepted: 03/25/2022] [Indexed: 11/22/2022] Open
Abstract
A high-fat diet with appropriate protein and low carbohydrate content, widely known as the ketogenic diet (KD), is considered as an effective non-pharmacotherapeutic treatment option for certain types of epilepsies. Several preclinical and clinical studies have been carried out to elucidate its mechanism of antiepileptic action. Ketone bodies produced after KD's breakdown interact with cellular excito-inhibitory processes and inhibit abnormal neuronal firing. The generated ketone bodies decrease glutamate release by inhibiting the vesicular glutamate transporter 1 and alter the transmembrane potential by hyperpolarization. Apart from their effect on the well-known pathogenic mechanisms of epilepsy, some recent studies have shown the interaction of KD metabolites with novel neuronal targets, particularly adenosine receptors, adenosine triphosphate-sensitive potassium channel, mammalian target of rapamycin, histone deacetylase, hydroxycarboxylic acid receptors, and the NLR family pyrin domain containing 3 inflammasomes to suppress seizures. The role of KD in augmenting gut microbiota as a potential mechanism for epileptic seizure suppression has been established. Furthermore, some recent findings also support the beneficial effect of KD against epilepsy- associated comorbidities. Despite several advantages of the KD in epilepsy management, its use is also associated with a wide range of side effects. Hypoglycemia, excessive ketosis, acidosis, renal stones, cardiomyopathies, and other metabolic disturbances are the primary adverse effects observed with the use of KD. However, in some recent studies, modified KD has been tested with lesser side effects and better tolerability. The present review discusses the molecular mechanism of KD and its role in managing epilepsy and its associated comorbidities.
Collapse
Affiliation(s)
- Amit Kumar
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR- Institute of Himalayan Bioresource Technology, Palampur 176061, Himachal Pradesh, India; ,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Savita Kumari
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR- Institute of Himalayan Bioresource Technology, Palampur 176061, Himachal Pradesh, India; ,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Damanpreet Singh
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR- Institute of Himalayan Bioresource Technology, Palampur 176061, Himachal Pradesh, India; ,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India,Address correspondence to this author at the Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur-176061, Himachal Pradesh, India; Tel: +91-9417923132; E-mails: ;
| |
Collapse
|
8
|
Dave A, Pillai PP. Docosahexaenoic acid increased MeCP2 mediated mitochondrial respiratory complexes II and III enzyme activities in cortical astrocytes. J Biochem Mol Toxicol 2022; 36:e23002. [PMID: 35174922 DOI: 10.1002/jbt.23002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 12/16/2021] [Accepted: 01/19/2022] [Indexed: 11/08/2022]
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder caused by mutations in the methyl-CpG-binding protein 2 (MeCP2) in the neurons and glial cells of the central nervous system. Currently, therapeutics for RTT is aimed at restoring the loss-of-function by MeCP2 gene therapy, but that approach has multiple challenges. We have already reported impaired mitochondrial bioenergetics in MeCP2 deficient astrocytes. Docosahexaenoic acid (DHA), a polyunsaturated fatty acid, has been shown with health benefits, but its impact on mitochondrial functions in MeCP2 deficient astrocytes has never been paid much attention. The present study aimed to investigate the effects of DHA on mitochondrial respiratory chain regulation in MeCP2 knockdown astrocytes. We determined NADH dehydrogenase (ubiquinone) flavoprotein 2 (Ndufv2-complex-I), Ubiquinol cytochrome c reductase core protein (Uqcrc1-complex-III) genes expression, Ndufv2 protein expression, respiratory electron transport chain complex I, II, III, and IV enzyme activities, intracellular Ca+2 , reactive oxygen species (ROS) and mitochondrial membrane potential (MMP) in DHA pre-incubated MeCP2 knock-down rat primary cortical astrocytes. Our study demonstrates that 100 µM DHA increases MeCP2 gene and protein expression. Increases brain-derived neurotrophic factor (BDNF) and Uqcrc1 gene expression, Ndufv2 protein expression, but has no effect on glial fibrillary acidic protein (GFAP) gene expression. DHA treatment also increases mitochondrial respiratory Complexes II and III activities and reduces intracellular calcium levels. Taken together, the effects of DHA seem independent of MeCP2 deficiency in astrocytes. Hence, further studies are warranted to understand the complicated mechanisms of DHA and for its therapeutic significance in MeCP2-mediated mitochondrial dysfunction and in RTT disease.
Collapse
Affiliation(s)
- Arpita Dave
- Department of Zoology, Division of Neurobiology, The Maharaja Sayajirao University of Baroda, Gujarat, India
| | - Prakash P Pillai
- Department of Zoology, Division of Neurobiology, The Maharaja Sayajirao University of Baroda, Gujarat, India
| |
Collapse
|
9
|
Burda J, Burda R. Ischemic tolerance - blessing or curse. Physiol Res 2021; 70:661-670. [PMID: 34505532 DOI: 10.33549/physiolres.934644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Application of knowledge about ischemic tolerance to clinic requires the solid understanding of mechanism of creation of this phenomenon. This review summarizes research that has been carried out in many laboratories over a long period of time, but the main focus will be on own experimental research. The main emphasis is devoted to the possibility of preparing full tolerance in the donor's body and its transfer to the patient in the form of activated blood plasma. Such plasma could be administered as soon as the patient is transported to the hospital and would take effect immediately after administration to the patient's bloodstream. One chapter is also devoted to anticonditioning, i.e. the possibility of preventing the activation of tolerance. Anticonditioning could be used to treat oncologic patients. We expect that this method could increase effectiveness of cancer treatment. Cross-tolerance with a wide range of diverse stressors gives us the courage to assume that activated plasma can significantly help with a wide range of pathological events.
Collapse
Affiliation(s)
- J Burda
- Institute of Neurobiology of Biomedical Research Center, Slovak Academy of Sciences, Košice, Slovak Republic. Faculty of Medicine, Clinic of Trauma Surgery, P. J. Šafárik University, Košice, Slovak Republic.
| | | |
Collapse
|
10
|
Borsini A, Nicolaou A, Camacho-Muñoz D, Kendall AC, Di Benedetto MG, Giacobbe J, Su KP, Pariante CM. Omega-3 polyunsaturated fatty acids protect against inflammation through production of LOX and CYP450 lipid mediators: relevance for major depression and for human hippocampal neurogenesis. Mol Psychiatry 2021; 26:6773-6788. [PMID: 34131267 PMCID: PMC8760043 DOI: 10.1038/s41380-021-01160-8] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 04/29/2021] [Accepted: 05/05/2021] [Indexed: 02/04/2023]
Abstract
Eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) can exert antidepressant, anti-inflammatory and neuroprotective properties, but the exact molecular mechanism underlying their effects is still not fully understood. We conducted both in vitro and clinical investigations to test which EPA or DHA metabolites are involved in these anti-inflammatory, neuroprotective and antidepressant effects. In vitro, we used the human hippocampal progenitor cell line HPC0A07/03C, and pre-treated cells with either EPA or DHA, followed by interleukin 1beta (IL1β), IL6 and interferon-alpha (IFN-α). Both EPA and DHA prevented the reduction in neurogenesis and the increase in apoptosis induced by these cytokines; moreover, these effects were mediated by the lipoxygenase (LOX) and cytochrome P450 (CYP450) EPA/DHA metabolites, 5-hydroxyeicosapentaenoic acid (HEPE), 4-hydroxydocosahexaenoic acid (HDHA), 18-HEPE, 20-HDHA, 17(18)-epoxyeicosatetraenoic acid (EpETE) and 19(20)-epoxydocosapentaenoic acid (EpDPA), detected here for the first time in human hippocampal neurones using mass spectrometry lipidomics of the supernatant. In fact, like EPA/DHA, co-treatment with these metabolites prevented cytokines-induced reduction in neurogenesis and apoptosis. Moreover, co-treatment with 17(18)-EpETE and 19(20)-EpDPA and the soluble epoxide hydroxylase (sEH) inhibitor, TPPU (which prevents their conversion into dihydroxyeicosatetraenoic acid (DiHETE)/ dihydroxydocosapentaenoic acid (DiHDPA) metabolites) further enhanced their neurogenic and anti-apoptotic effects. Interestingly, these findings were replicated in a sample of n = 22 patients with a DSM-IV Major Depressive Disorder, randomly assigned to treatment with either EPA (3.0 g/day) or DHA (1.4 g/day) for 12 weeks, with exactly the same LOX and CYP450 lipid metabolites increased in the plasma of these patients following treatment with their precursor, EPA or DHA, and some evidence that higher levels of these metabolites were correlated with less severe depressive symptoms. Overall, our study provides the first evidence for the relevance of LOX- and CYP450-derived EPA/DHA bioactive lipid metabolites as neuroprotective molecular targets for human hippocampal neurogenesis and depression, and highlights the importance of sEH inhibitors as potential therapeutic strategy for patients suffering from depressive symptoms.
Collapse
Affiliation(s)
- Alessandra Borsini
- Stress, Psychiatry and Immunology Laboratory, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, King's College London, London, UK.
| | - Anna Nicolaou
- Laboratory for Lipidomics and Lipid Biology, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Dolores Camacho-Muñoz
- Laboratory for Lipidomics and Lipid Biology, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Alexandra C Kendall
- Laboratory for Lipidomics and Lipid Biology, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Maria Grazia Di Benedetto
- Stress, Psychiatry and Immunology Laboratory, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, King's College London, London, UK
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio, Fatebenefratelli, Brescia, Italy
| | - Juliette Giacobbe
- Stress, Psychiatry and Immunology Laboratory, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, King's College London, London, UK
| | - Kuan-Pin Su
- Stress, Psychiatry and Immunology Laboratory, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, King's College London, London, UK.
- College of Medicine, China Medical University, Taichung, Taiwan.
- Depression Center, An-Nan Hospital, China Medical University, Tainan, Taiwan.
| | - Carmine M Pariante
- Stress, Psychiatry and Immunology Laboratory, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, King's College London, London, UK
| |
Collapse
|
11
|
Characterization of metabolic activity induced by kainic acid in adult rat whole brain at the early stage: A 18FDG-PET study. Brain Res 2021; 1769:147621. [PMID: 34403661 DOI: 10.1016/j.brainres.2021.147621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/08/2021] [Accepted: 08/10/2021] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Brain metabolic processes are not fully characterized in the kainic acid (KA)-induced Status Epilepticus (KASE). Thus, we evaluated the usefulness of 18F-fluorodeoxyglucose positron emission tomography (FDG-PET) as an experimental strategy to evaluate in vivo, in a non-invasive way, the glucose consumption in several brain regions, in a semi-quantitative study to compare and to correlate with data from electroencephalography and histology studies. METHODS Sixteen male Wistar rats underwent FDG-PET scans at basal state and after KA injection. FDG-PET images were normalized to an MRI-based atlas and segmented to locate regions. Standardized uptake values (SUV) were obtained at several time points. EEGs and cell viability by histological analysis, were also evaluated. RESULTS FDG-PET data showed changes in regions such as: amygdala, hippocampus, accumbens, entorhinal cortex, motor cortex and hypothalamus. Remarkably, hippocampal hypermetabolism was found (mean SUV = 2.66 ± 0.057) 2 h after KA administration, while hypometabolism at 24 h (mean SUV = 1.83 ± 0.056) vs basal values (mean SUV = 2.19 ± 0.057). EEG showed increased spectral power values 2 h post-KA administration. Hippocampal viable-cell counting 24 h after KA was decreased, while Fluoro-Jade B-positive cells were increased, as compared to control rats, coinciding with the hypometabolism detected in the same region by semi-quantitative FDG-PET at 24 h after KASE. CONCLUSIONS PET is suitable to measure metabolic brain changes in the rat model of status epilepticus induced by KA (KASE) at the first 24 h, compared to that of EEG; PET data may also be sensitive to cell viability.
Collapse
|
12
|
Long-term dietary supplementation with plant-derived omega-3 fatty acid improves outcome in experimental ischemic stroke. Atherosclerosis 2021; 325:89-98. [PMID: 33915355 DOI: 10.1016/j.atherosclerosis.2021.04.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/26/2021] [Accepted: 04/13/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Early revascularization -the gold standard therapy for ischemic stroke- is often withheld in the elderly population due to high risk of complications. Thus, safe and effective preventive and therapeutic options are needed. The plant-derived omega-3-fatty-acid alpha-linolenic-acid (ALA) has emerged as a novel cardiovascular-protective agent. As of yet, little is known about its potential therapeutic effects on stroke. We hereby aimed to investigate the impact of a clinically relevant long-term dietary intervention with ALA on stroke outcome. METHODS Six month-old C57BL/6 wildtype males were either fed an ALA-rich (high ALA) or a control diet (low ALA) for 12 months. At 18 months, brain ischemia/reperfusion was induced by transient middle cerebral artery occlusion (tMCAO). Stroke size and neurological function were assessed. Functional blood-brain-barrier-(BBB) permeability and protein expression were assessed by immunohistochemistry. Baseline inflammatory markers were measured at 18 months. RESULTS High ALA-fed animals displayed decreased circulating TNF-α levels and Neutrophil-to-Lymphocyte Ratios at 18 months. Stroke size and neurological dysfunction were significantly reduced in high ALA-fed animals. Coherently to the reduced stroke size, functional BBB integrity and occludin endothelial expression were maintained by high ALA supplementation. Additionally, ALA reduced endothelial activation and thus recruitment and activation of macrophages and resident microglia. Finally, high ALA diet reduced the expression of BBB-degrading and neurotoxic MMP-3 and MMP-9. CONCLUSIONS We demonstrate the beneficial effects of a clinically relevant and feasible dietary intervention with a safe and readily available compound in the setting of stroke. The protective effects observed with ALA supplementation may relate to blunting of inflammation and might pave the way for novel stroke treatments.
Collapse
|
13
|
Borsini A, Stangl D, Jeffries AR, Pariante CM, Thuret S. The role of omega-3 fatty acids in preventing glucocorticoid-induced reduction in human hippocampal neurogenesis and increase in apoptosis. Transl Psychiatry 2020; 10:219. [PMID: 32636362 PMCID: PMC7341841 DOI: 10.1038/s41398-020-00908-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 06/15/2020] [Accepted: 06/23/2020] [Indexed: 12/21/2022] Open
Abstract
Glucocorticoids have been suggested to be involved in several neuropsychiatric disorders, including depression. One of the possible mechanisms through which glucocorticoids contribute to the development of the depressive symptomatology is via regulation of distinct neurogenic mechanisms in the brain. A preventive or protective approach for these patients might be the use of omega-3 polyunsaturated fatty acids (n-3 PUFAs), eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), which are known for they neuroprotective properties. We used the human hippocampal progenitor cell line HPC0A07/03C and pre-treated cells with either EPA or DHA, followed by treatment with the glucocorticoid cortisol either alone, or in co-treatment with the same n-3 PUFA during subsequent 3 days of proliferation and 7 days of differentiation. During proliferation, both EPA and DHA were able to prevent cortisol-induced reduction in proliferation and increase in apoptosis, when used in pre-treatment, and both pre- and co-treatment. During differentiation, EPA was able to prevent cortisol-induced reduction in neurogenesis and increase in apoptosis, when used in pre-treatment, and both pre- and co-treatment only during the proliferation stage; however, DHA required continuous treatment also during the differentiation stage to prevent cortisol-induced reduction in neurogenesis. Using transcriptomic analyses, we showed that both EPA and DHA regulated pathways involved in oxidative stress and immune response [e.g., nuclear factor (erythroid-derived 2)-like 2 (Nrf2), Signal transducer and activator of transcription 3 (STAT3), Interferon (IFN) and Interleukin (IL)-1 signaling], whereas DHA also regulated pathways involved in cell development and neuronal formation [e.g., cAMP-response element binding protein (CREB) signaling]. We provide the first evidence for treatment with both EPA and DHA to prevent cortisol-induced reduction in human hippocampal neurogenesis, and identify novel molecular mechanisms underlying these effects.
Collapse
Affiliation(s)
- Alessandra Borsini
- Section of Stress, Psychiatry and Immunology & Perinatal Psychiatry, King's College London, Institute of Psychiatry, Psychology & Neuroscience, Department of Psychological Medicine, London, UK.
- King's College London, Institute of Psychiatry, Psychology & Neuroscience, Department of Basic and Clinical Neuroscience, London, UK.
| | - Doris Stangl
- King's College London, Institute of Psychiatry, Psychology & Neuroscience, Department of Basic and Clinical Neuroscience, London, UK
| | | | - Carmine M Pariante
- Section of Stress, Psychiatry and Immunology & Perinatal Psychiatry, King's College London, Institute of Psychiatry, Psychology & Neuroscience, Department of Psychological Medicine, London, UK
| | - Sandrine Thuret
- King's College London, Institute of Psychiatry, Psychology & Neuroscience, Department of Basic and Clinical Neuroscience, London, UK.
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
14
|
Pietri M, Djillani A, Mazella J, Borsotto M, Heurteaux C. First evidence of protective effects on stroke recovery and post-stroke depression induced by sortilin-derived peptides. Neuropharmacology 2019; 158:107715. [PMID: 31325429 DOI: 10.1016/j.neuropharm.2019.107715] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/12/2019] [Accepted: 07/16/2019] [Indexed: 01/15/2023]
Abstract
Post-stroke depression (PSD) is the most common mood disorder following stroke with high relevance for outcome and survival of patients. The TREK-1 channel represents a crucial target in the pathogenesis of stroke and depression. Spadin and its short analog mini-spadin were reported to display potent antidepressant properties. We investigated the therapeutic effects of mini-spadin in a mouse model of focal ischemia and PSD. To activate TREK-1 and induce neuroprotection a single low dose of mini-spadin (0.03 μg/kg) was intraperitoneally injected 30 min after the onset of ischemia, once a day during 7 days post-ischemia. Then, to inhibit TREK-1 and induce antidepressant effect, the peptide was injected at higher concentration (3 μg/kg) once a day for 4 days/week until the sacrifice of animals. Electrophysiological studies showed that mini-spadin had a biphasic action on TREK-1. At low doses, the channel activity was increased whereas at higher doses it was inhibited. Mini-spadin prevented the loss of body weight and the delayed dopaminergic degeneration in substantia nigra and improved the motor and cognitive ischemia-induced deficits. Moreover, mini-spadin prevented PSD analyzed in the Forced Swim (FST) and Novelty Suppressed Feeding (NSF) tests. Finally, enhanced neurogenesis and synaptogenesis contributed to the beneficial effects of mini-spadin against stroke and PSD. This work reveals the first evidence that the modulation of TREK-1 channels in the early and chronic phases of stroke as well as the stimulation of brain plasticity by mini-spadin could play a key role in its brain protective effects against stroke and its deleterious consequences such as PSD.
Collapse
Affiliation(s)
- Mariel Pietri
- Université Côte D'Azur, CNRS, IPMC, UMR7275, 660 Route des Lucioles, Sophia Antipolis, 06560, Valbonne, France
| | - Alaeddine Djillani
- Université Côte D'Azur, CNRS, IPMC, UMR7275, 660 Route des Lucioles, Sophia Antipolis, 06560, Valbonne, France
| | - Jean Mazella
- Université Côte D'Azur, CNRS, IPMC, UMR7275, 660 Route des Lucioles, Sophia Antipolis, 06560, Valbonne, France
| | - Marc Borsotto
- Université Côte D'Azur, CNRS, IPMC, UMR7275, 660 Route des Lucioles, Sophia Antipolis, 06560, Valbonne, France
| | - Catherine Heurteaux
- Université Côte D'Azur, CNRS, IPMC, UMR7275, 660 Route des Lucioles, Sophia Antipolis, 06560, Valbonne, France.
| |
Collapse
|
15
|
Senbokuya N, Yoshioka H, Yagi T, Owada Y, Kinouchi H. Effects of FABP7 on functional recovery after spinal cord injury in adult mice. J Neurosurg Spine 2019; 31:291-297. [PMID: 31051461 DOI: 10.3171/2019.2.spine18844] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 02/13/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Elucidating the mechanisms of neuronal injury is crucial for the development of spinal cord injury (SCI) treatments. Brain-type fatty acid-binding protein 7 (FABP7) is expressed in the adult rodent brain, especially in astrocytes, and has been reported to play a role in astrocyte function in various types of brain damage; however, its role after SCI has not been well studied. In this study, the authors evaluated the expression change of FABP7 after SCI using a mouse spinal cord compression model and observed the effect of FABP7 gene knockout on neuronal damage and functional recovery after SCI. METHODS Female FABP7 knockout (KO) mice with a C57BL/6 background and their respective wild-type littermates were subjected to SCI with a vascular clip. The expression of FABP7, neuronal injury, and functional recovery after SCI were analyzed in both groups of mice. RESULTS Western blot analysis revealed upregulation of FABP7 in the wild-type mice, which reached its peak 14 days after SCI, with a significant difference in comparison to the control mice. Immunohistochemistry also showed upregulation of FABP7 at the same time points, mainly in proliferative astrocytes. The number of surviving ventral neurons in the FABP7-KO mice at 28 days after SCI was significantly lower than that observed in the wild-type mice. In addition, motor functional recovery in the FABP7-KO mice was significantly worse than that of the wild-type mice. CONCLUSIONS The findings of this study indicate that FABP7 could have a neuroprotective role that might be associated with modulation of astrocytes after SCI. FABP7 could potentially be a therapeutic target in the treatment of SCI.
Collapse
Affiliation(s)
- Nobuo Senbokuya
- 1Department of Neurosurgery, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi; and
| | - Hideyuki Yoshioka
- 1Department of Neurosurgery, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi; and
| | - Takashi Yagi
- 1Department of Neurosurgery, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi; and
| | - Yuji Owada
- 2Department of Organ Anatomy, Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
| | - Hiroyuki Kinouchi
- 1Department of Neurosurgery, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi; and
| |
Collapse
|
16
|
Gonzalo-Gobernado R, Ayuso MI, Sansone L, Bernal-Jiménez JJ, Ramos-Herrero VD, Sánchez-García E, Ramos TL, Abia R, Muriana FJG, Bermúdez B, Montaner J. Neuroprotective Effects of Diets Containing Olive Oil and DHA/EPA in a Mouse Model of Cerebral Ischemia. Nutrients 2019; 11:E1109. [PMID: 31109078 PMCID: PMC6566717 DOI: 10.3390/nu11051109] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 05/14/2019] [Indexed: 12/29/2022] Open
Abstract
Stroke is one of the leading causes of death worldwide and while there is increasing evidence that a Mediterranean diet might decrease the risk of a stroke, the effects of dietary fat composition on stroke outcomes have not been fully explored. We hypothesize that the brain damage provoked by a stroke would be different depending on the source of dietary fat. To test this, male C57BL/6J mice were fed for 4 weeks with a standard low-fat diet (LFD), a high-fat diet (HFD) rich in saturated fatty acids (HFD-SFA), an HFD containing monounsaturated fatty acids (MUFAs) from olive oil (HFD-OO), or an HFD containing MUFAs from olive oil plus polyunsaturated fatty acids (PUFAs) docosahexaenoic acid/eicosapentaenoic acid (DHA/EPA) (HFD-OO-ω3). These mice were then subjected to transient middle cerebral artery occlusion (tMCAo). Behavioural tests and histological analyses were performed 24 and/or 48 h after tMCAo in order to elucidate the impact of these diets with different fatty acid profiles on the ischemic lesion and on neurological functions. Mice fed with HFD-OO-ω3 displayed better histological outcomes after cerebral ischemia than mice that received an HFD-SFA or LFD. Furthermore, PUFA- and MUFA-enriched diets improved the motor function and neurological performance of ischemic mice relative to those fed with an LFD or HFD-SFA. These findings support the use of DHA/EPA-omega-3-fatty acid supplementation and olive oil as dietary source of MUFAs in order to reduce the damage and protect the brain when a stroke occurs.
Collapse
Affiliation(s)
- Rafael Gonzalo-Gobernado
- Neurovascular Research Group, Institute of Biomedicine of Seville, IBiS, Hospital Universitario Virgen del Rocío, Av. Manuel Siurot s/n, 41013 Seville; Spain.
| | - María Irene Ayuso
- Neurovascular Research Group, Institute of Biomedicine of Seville, IBiS, Hospital Universitario Virgen del Rocío, Av. Manuel Siurot s/n, 41013 Seville; Spain.
| | - Loredana Sansone
- Neurovascular Research Group, Institute of Biomedicine of Seville, IBiS, Hospital Universitario Virgen del Rocío, Av. Manuel Siurot s/n, 41013 Seville; Spain.
| | - Juan José Bernal-Jiménez
- Neurovascular Research Group, Institute of Biomedicine of Seville, IBiS, Hospital Universitario Virgen del Rocío, Av. Manuel Siurot s/n, 41013 Seville; Spain.
| | - Víctor Darío Ramos-Herrero
- Neurovascular Research Group, Institute of Biomedicine of Seville, IBiS, Hospital Universitario Virgen del Rocío, Av. Manuel Siurot s/n, 41013 Seville; Spain.
| | - Enrique Sánchez-García
- Neurovascular Research Group, Institute of Biomedicine of Seville, IBiS, Hospital Universitario Virgen del Rocío, Av. Manuel Siurot s/n, 41013 Seville; Spain.
| | - Teresa L Ramos
- Laboratory of Cell Therapy and New Therapeutic Targets in Onco-Hematology, Institute of Biomedicine of Seville, IBiS, Hospital Universitario Virgen del Rocío, Av. Manuel Siurot s/n, 41013 Seville, Spain.
| | - Rocío Abia
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, CSIC, Ctra. de Utrera Km. 1, 41013 Seville, Spain.
| | - Francisco J G Muriana
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, CSIC, Ctra. de Utrera Km. 1, 41013 Seville, Spain.
| | - Beatriz Bermúdez
- Department of Cellular Biology, School of Biology, University of Seville, Av. de la Reina Mercedes 6, 41012 Seville, Spain.
| | - Joan Montaner
- Neurovascular Research Group, Institute of Biomedicine of Seville, IBiS, Hospital Universitario Virgen del Rocío, Av. Manuel Siurot s/n, 41013 Seville; Spain.
- Department of Neurology, Hospital Universitario Virgen Macarena, Av. Doctor Fedriani 3, 41007 Seville, Spain.
| |
Collapse
|
17
|
Oxidative stress and stroke: a review of upstream and downstream antioxidant therapeutic options. ACTA ACUST UNITED AC 2019. [DOI: 10.1007/s00580-019-02940-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
18
|
Ríos C, Farfán-Briseño AC, Manjarrez-Marmolejo J, Franco-Pérez J, Méndez-Armenta M, Nava-Ruiz C, Caballero-Chacón S, Ruiz-Diaz A, Baron-Flores V, Díaz-Ruiz A. Efficacy of dapsone administered alone or in combination with diazepam to inhibit status epilepticus in rats. Brain Res 2019; 1708:181-187. [DOI: 10.1016/j.brainres.2018.12.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 11/13/2018] [Accepted: 12/12/2018] [Indexed: 12/15/2022]
|
19
|
Andrews RN, Dugan GO, Peiffer AM, Hawkins GA, Hanbury DB, Bourland JD, Hampson RE, Deadwyler SA, Cline JM. White Matter is the Predilection Site of Late-Delayed Radiation-Induced Brain Injury in Non-Human Primates. Radiat Res 2019; 191:217-231. [PMID: 30694733 PMCID: PMC6422025 DOI: 10.1667/rr15263.1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Fractionated whole-brain irradiation for the treatment of intracranial neoplasia causes progressive neurodegeneration and neuroinflammation. The long-term consequences of single-fraction high-dose irradiation to the brain are unknown. To assess the late effects of brain irradiation we compared transcriptomic gene expression profiles from nonhuman primates (NHP; rhesus macaques Macaca mulatta) receiving single-fraction total-body irradiation (TBI; n = 5, 6.75-8.05 Gy, 6-9 years prior to necropsy) to those receiving fractionated whole-brain irradiation (fWBI; n = 5, 40 Gy, 8 × 5 Gy fractions; 12 months prior to necropsy) and control comparators (n = 5). Gene expression profiles from the dorsolateral prefrontal cortex (DLPFC), hippocampus (HC) and deep white matter (WM; centrum semiovale) were compared. Stratified analyses by treatment and region revealed that radiation-induced transcriptomic alterations were most prominent in animals receiving fWBI, and primarily affected white matter in both TBI and fWBI groups. Unsupervised canonical and ontologic analysis revealed that TBI or fWBI animals demonstrated shared patterns of injury, including white matter neuroinflammation, increased expression of complement factors and T-cell activation. Both irradiated groups also showed evidence of impaired glutamatergic neurotransmission and signal transduction within white matter, but not within the dorsolateral prefrontal cortex or hippocampus. Signaling pathways and structural elements involved in extracellular matrix (ECM) deposition and remodeling were noted within the white matter of animals receiving fWBI, but not of those receiving TBI. These findings indicate that those animals receiving TBI are susceptible to neurological injury similar to that observed after fWBI, and these changes persist for years postirradiation. Transcriptomic profiling reaffirmed that macrophage/microglial-mediated neuroinflammation is present in radiation-induced brain injury (RIBI), and our data provide novel evidence that the complement system may contribute to the pathogenesis of RIBI. Finally, these data challenge the assumption that the hippocampus is the predilection site of injury in RIBI, and indicate that impaired glutamatergic neurotransmission may occur in white matter injury.
Collapse
Affiliation(s)
- Rachel N. Andrews
- Departments of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| | - Gregory O. Dugan
- Departments of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| | - Ann M. Peiffer
- Departments of Radiation Oncology, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
- Departments of Brain Tumor Center of Excellence, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| | - Gregory A. Hawkins
- Departments of Biochemistry, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
- Departments of Wake Forest Baptist Comprehensive Cancer Center, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| | - David B. Hanbury
- Department of Psychology, Averett University, Danville, Virginia 24541
| | - J. Daniel Bourland
- Departments of Radiation Oncology, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
- Departments of Brain Tumor Center of Excellence, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| | - Robert E. Hampson
- Departments of Physiology and Pharmacology, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| | - Samuel A. Deadwyler
- Departments of Physiology and Pharmacology, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| | - J. Mark Cline
- Departments of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
- Departments of Radiation Oncology, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| |
Collapse
|
20
|
Omega-3 Polyunsaturated Fatty Acid Deficiency and Progressive Neuropathology in Psychiatric Disorders: A Review of Translational Evidence and Candidate Mechanisms. Harv Rev Psychiatry 2019; 27:94-107. [PMID: 30633010 PMCID: PMC6411441 DOI: 10.1097/hrp.0000000000000199] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Meta-analytic evidence indicates that mood and psychotic disorders are associated with both omega-3 polyunsaturated fatty acid (omega-3 PUFA) deficits and progressive regional gray and white matter pathology. Although the association between omega-3 PUFA insufficiency and progressive neuropathological processes remains speculative, evidence from translational research suggests that omega-3 PUFA insufficiency may represent a plausible and modifiable risk factor not only for enduring neurodevelopmental abnormalities in brain structure and function, but also for increased vulnerability to neurodegenerative processes. Recent evidence from human neuroimaging studies suggests that lower omega-3 PUFA intake/status is associated with accelerated gray matter atrophy in healthy middle-aged and elderly adults, particularly in brain regions consistently implicated in mood and psychotic disorders, including the amygdala, anterior cingulate, hippocampus, prefrontal cortex, and temporal cortex. Human neuroimaging evidence also suggests that both low omega-3 PUFA intake/status and psychiatric disorders are associated with reductions in white matter microstructural integrity and increased rates of white matter hyperintensities. Preliminary evidence suggests that increasing omega-3 PUFA status is protective against gray matter atrophy and deficits in white matter microstructural integrity in patients with mood and psychotic disorders. Plausible mechanisms mediating this relationship include elevated pro-inflammatory signaling, increased synaptic regression, and reductions in cerebral perfusion. Together these associations encourage additional neuroimaging research to directly investigate whether increasing omega-3 PUFA status can mitigate neuropathological processes in patients with, or at high risk for, psychiatric disorders.
Collapse
|
21
|
Lamas JA, Fernández-Fernández D. Tandem pore TWIK-related potassium channels and neuroprotection. Neural Regen Res 2019; 14:1293-1308. [PMID: 30964046 PMCID: PMC6524494 DOI: 10.4103/1673-5374.253506] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
TWIK-related potassium channels (TREK) belong to a subfamily of the two-pore domain potassium channels family with three members, TREK1, TREK2 and TWIK-related arachidonic acid-activated potassium channels. The two-pore domain potassium channels is the last big family of channels being discovered, therefore it is not surprising that most of the information we know about TREK channels predominantly comes from the study of heterologously expressed channels. Notwithstanding, in this review we pay special attention to the limited amount of information available on native TREK-like channels and real neurons in relation to neuroprotection. Mainly we focus on the role of free fatty acids, lysophospholipids and other neuroprotective agents like riluzole in the modulation of TREK channels, emphasizing on how important this modulation may be for the development of new therapies against neuropathic pain, depression, schizophrenia, epilepsy, ischemia and cardiac complications.
Collapse
Affiliation(s)
- J Antonio Lamas
- Laboratory of Neuroscience, Biomedical Research Center (CINBIO), University of Vigo, Vigo, Galicia, Spain
| | - Diego Fernández-Fernández
- Laboratory of Neuroscience, Biomedical Research Center (CINBIO), University of Vigo, Vigo, Galicia, Spain
| |
Collapse
|
22
|
McNamara RK, Asch RH, Lindquist DM, Krikorian R. Role of polyunsaturated fatty acids in human brain structure and function across the lifespan: An update on neuroimaging findings. Prostaglandins Leukot Essent Fatty Acids 2018; 136:23-34. [PMID: 28529008 PMCID: PMC5680156 DOI: 10.1016/j.plefa.2017.05.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 04/14/2017] [Accepted: 05/08/2017] [Indexed: 01/07/2023]
Abstract
There is a substantial body of evidence from animal studies implicating polyunsaturated fatty acids (PUFA) in neuroinflammatory, neurotrophic, and neuroprotective processes in brain. However, direct evidence for a role of PUFA in human brain structure and function has been lacking. Over the last decade there has been a notable increase in neuroimaging studies that have investigated the impact of PUFA intake and/or blood levels (i.e., biostatus) on brain structure, function, and pathology in human subjects. The majority of these studies specifically evaluated associations between omega-3 PUFA intake and/or biostatus and neuroimaging outcomes using a variety of experimental designs and imaging techniques. This review provides an updated overview of these studies in an effort to identify patterns to guide and inform future research. While the weight of evidence provides general support for a beneficial effect of a habitual diet consisting of higher omega-3 PUFA intake on cortical structure and function in healthy human subjects, additional research is needed to replicate and extend these findings as well as identify response mediators and clarify mechanistic pathways. Controlled intervention trials are also needed to determine whether increasing n-3 PUFA biostatus can prevent or attenuate neuropathological brain changes observed in patients with or at risk for psychiatric disorders and dementia.
Collapse
Affiliation(s)
- Robert K McNamara
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH 45219, United States.
| | - Ruth H Asch
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH 45219, United States
| | - Diana M Lindquist
- Imaging Research Center, Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45267, United States
| | - Robert Krikorian
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH 45219, United States
| |
Collapse
|
23
|
Ferain A, Bonnineau C, Neefs I, Das K, Larondelle Y, Rees JF, Debier C, Lemaire B. Transcriptional effects of phospholipid fatty acid profile on rainbow trout liver cells exposed to methylmercury. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2018; 199:174-187. [PMID: 29649756 DOI: 10.1016/j.aquatox.2018.03.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/16/2018] [Accepted: 03/20/2018] [Indexed: 06/08/2023]
Abstract
Lipids, and their constitutive fatty acids, are key nutrients for fish health as they provide energy, maintain cell structure, are precursors of signalling molecules and act as nuclear receptor ligands. These specific roles may be of crucial importance in a context of exposure to pollutants. We recently showed that the fatty acid profile of rainbow trout liver cell phospholipids modulates sensitivity to an acute methylmercury challenge. In order to investigate mechanisms of effects, we herein tested whether specific polyunsaturated fatty acids (PUFAs) may protect cells from methylmercury through decreasing intracellular mercury accumulation and/or enhancing cellular defences (e.g. via modulation of gene expression patterns). We also investigated the inverse relationship and assessed the impact of methylmercury on cellular fatty acid metabolism. To do so, the fatty acid composition of rainbow trout liver cell phospholipids was first modified by incubating them in a medium enriched in a specific PUFA from either the n-3 family (alpha-linolenic acid, ALA; eicosapentaenoic acid, EPA) or the n-6 family (linoleic acid, LA; arachidonic acid, AA). Cells were then exposed to methylmercury (0.15 or 0.50 μM) for 24 h and sampled thereafter for assessing phospholipid fatty acid profile, intracellular total mercury burden, and expression pattern of genes involved in fatty acid metabolism, synthesis of PUFA-derived signalling molecules and stress response. We observed that cells incorporated the given PUFA and some biotransformation products in their phospholipids. Methylmercury had few impacts on this cellular phospholipid composition. None of the PUFA enrichments affected the cellular mercury burden, suggesting that the previously observed cytoprotection conferred by ALA and EPA was not linked to a global decrease in cellular accumulation of mercury. Fatty acid enrichments and methylmercury exposure both modulated gene expression patterns. Genes involved in the synthesis of PUFA-derived signalling molecules, in stress response and the orphan cytochrome P450 20A1 were identified as possible sites of interaction between fatty acids and methylmercury in rainbow trout liver cells.
Collapse
Affiliation(s)
- Aline Ferain
- Institute of Life Sciences, Université catholique de Louvain, Place Croix du Sud 2/L7.05.08, B-1348 Louvain-la-Neuve, Belgium.
| | - Chloé Bonnineau
- Institute of Life Sciences, Université catholique de Louvain, Place Croix du Sud 2/L7.05.08, B-1348 Louvain-la-Neuve, Belgium; Irstea, UR RiverLy, Centre de Lyon-Villeurbanne, 5, 69625 Villeurbanne, France
| | - Ineke Neefs
- Institute of Life Sciences, Université catholique de Louvain, Place Croix du Sud 2/L7.05.08, B-1348 Louvain-la-Neuve, Belgium
| | - Krishna Das
- Freshwater and Oceanic sciences Unit of reSearch (FOCUS), Laboratory of Oceanology, Université de Liège, Allée du 6 août B6C, B-4000 Liège, Belgium
| | - Yvan Larondelle
- Institute of Life Sciences, Université catholique de Louvain, Place Croix du Sud 2/L7.05.08, B-1348 Louvain-la-Neuve, Belgium
| | - Jean-François Rees
- Institute of Life Sciences, Université catholique de Louvain, Place Croix du Sud 2/L7.05.08, B-1348 Louvain-la-Neuve, Belgium
| | - Cathy Debier
- Institute of Life Sciences, Université catholique de Louvain, Place Croix du Sud 2/L7.05.08, B-1348 Louvain-la-Neuve, Belgium.
| | - Benjamin Lemaire
- Institute of Life Sciences, Université catholique de Louvain, Place Croix du Sud 2/L7.05.08, B-1348 Louvain-la-Neuve, Belgium
| |
Collapse
|
24
|
Lee AY, Lee MH, Lee S, Cho EJ. Neuroprotective Effect of Alpha-Linolenic Acid against Aβ-Mediated Inflammatory Responses in C6 Glial Cell. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:4853-4861. [PMID: 29668263 DOI: 10.1021/acs.jafc.8b00836] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Therapeutic approaches for neurodegeneration, such as Alzheimer's disease (AD), have been widely studied. One of the critical hallmarks of AD is accumulation of amyloid beta (Aβ). Aβ induces neurotoxicity and releases inflammatory mediators or cytokines through activation of glial cell, and these pathological features are observed in AD patient's brain. The purpose of this study is to investigate the protective effect of alpha-linolenic acid (ALA) on Aβ25-35-induced neurotoxicity in C6 glial cells. Exposure of C6 glial cells to 50 μM Aβ25-35 caused cell death, overproduction of nitric oxide (NO), and pro-inflammatory cytokines release [interleukin (IL)-6 and tumor necrosis factor-α], while treatment of ALA increased cell viability and markedly attenuated Aβ25-35-induced excessive production of NO and those inflammatory cytokines. Inhibitory effect of ALA on generation of NO and cytokines was mediated by down-regulation of inducible nitric oxide synthase and cyclooxygenase-2 protein and mRNA expressions. In addition, ALA treatment inhibited reactive oxygen species generation induced by Aβ25-35 through the enhancement of the nuclear factor-erythroid 2-related factor-2 (Nrf-2) protein levels and subsequent induction of heme-oxygenase-1 (HO-1) expression in C6 glial cells dose- and time-dependently. Furthermore, the levels of neprilysin and insulin-degrading enzyme protein expressions, which contribute to degradation of Aβ, were also increased by treatment of ALA compared to Aβ25-35-treated control group. In conclusion, effects of ALA on Aβ degradation were shown to be mediated through inhibition of inflammatory responses and activation of antioxidative system, Nrf-2/HO-1 signaling pathway, in C6 glial cells. Our findings suggest that ALA might have the potential for therapeutics of AD.
Collapse
Affiliation(s)
- Ah Young Lee
- Department of Food Science and Nutrition & Kimchi Research Institute , Pusan National University , Busan 46241 , Republic of Korea
| | - Myoung Hee Lee
- Department of Southern Area Crop Science , National Institute of Crop Science, Rural Development Administration , Gyeongnam 50424 , Republic of Korea
| | - Sanghyun Lee
- Department of Integrative Plant Science , Chung-Ang University , Gyeonggi 17546 , Republic of Korea
| | - Eun Ju Cho
- Department of Food Science and Nutrition & Kimchi Research Institute , Pusan National University , Busan 46241 , Republic of Korea
| |
Collapse
|
25
|
Geffard M, Mangas A, Bedat D, Coveñas R. GEMALS: A promising therapy for amyotrophic lateral sclerosis. Exp Ther Med 2018; 15:3203-3210. [PMID: 29545836 PMCID: PMC5841048 DOI: 10.3892/etm.2018.5868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 09/14/2017] [Indexed: 12/01/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease that currently has no cure. At present, the only approved treatment for ALS is Riluzole, a glutamate release blocker that improves life expectancy by 3–6 months. ALS-Endotherapia (GEMALS) is a novel therapeutic approach to treat ALS and the aim of the present study was to investigate the potential beneficial effects of this novel treatment. A total of 31 patients with ALS were assessed in the current study. Deceleration of the disease was observed in 83.87% (P<0.0001) of patients and mean life expectancy was increased by 38 months. Motor functions, including breathing, walking, salivation, speech, swallowing and writing, were also improved in patients treated with GEMALS. The results of the present study demonstrate that long-term treatment with GEMALS has a curative effect in patients with ALS. Furthermore, the overall effectiveness of GEMALS was assessed using the ALS Assessment Questionnaire. The score improvement was 76.2 and 100% for men and women, respectively (P<0.0001), compared with the worldwide reference score. The present study provides a promising basis for the use of GEMALS as a therapeutic treatment for patients with ALS; however, these results must be confirmed in a double-blinded and randomized clinical trial.
Collapse
Affiliation(s)
- Michel Geffard
- Research Department, Institute for Development of Research on Human Pathology and Therapeutics, 33400 Talence, France
| | - Arturo Mangas
- Research Department, Institute for Development of Research on Human Pathology and Therapeutics, 33400 Talence, France.,Gemacbio, Research Department, Lieu dit Berganton, 33127 Saint Jean d'Illac, France.,Institute of Neurosciences of Castilla y León, Laboratory of Neuroanatomy of The Peptidergic Systems, University of Salamanca, 37007 Salamanca, Spain
| | - Denis Bedat
- Research Department, Institute for Development of Research on Human Pathology and Therapeutics, 33400 Talence, France
| | - Rafael Coveñas
- Institute of Neurosciences of Castilla y León, Laboratory of Neuroanatomy of The Peptidergic Systems, University of Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
26
|
Zheng G, Zheng Q, Xu Q. Identification of key signaling pathways in cerebral small vessel disease using differential pathway network analysis. Exp Ther Med 2017; 14:4371-4376. [PMID: 29104648 DOI: 10.3892/etm.2017.5104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 05/11/2017] [Indexed: 11/06/2022] Open
Abstract
Cerebral small vessel disease (CSVD) primarily affects the perforating cerebral arterioles and capillaries, and results in injury to subcortical grey and white matter. Despite advances in determining the genetic basis of CSVD, the molecular mechanisms underlying the development and progression of CSVD remain unclear. The present study aimed to identify significant signaling pathways associated with CSVD based on differential pathway network analysis. Combining CSVD microarray data with human protein-protein interaction data and data from the Reactome pathway database, pathway interactions were constructed using the Spearman's correlation coefficient strategy. Pathway interactions with weight values of >0.95 were selected to construct the differential pathway network, which contained 715 differential pathway interactions covering 312 nodes and was visualized using Cytoscape software. A total of 15 hub pathways with a top 5% degree distribution in the differential pathway network were identified. The top 5 hub pathways were associated with the synthesis and metabolism of fatty acids. The results of the present study indicate that the synthesis and metabolism of fatty acids is associated with the occurrence and development of CSVD, and may thus provide insights to improve the early diagnosis and treatment of CSVD.
Collapse
Affiliation(s)
- Gang Zheng
- Department of Neurology, Yinzhou People's Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Qianlei Zheng
- Intensive Care Unit, Yinzhou People's Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Qianwei Xu
- Department of Rehabilitation, Yinzhou People's Hospital, Ningbo, Zhejiang 315040, P.R. China
| |
Collapse
|
27
|
Geffard M, Mangas A, Coveñas R. Follow-up of multiple sclerosis patients treated with Endotherapia (GEMSP). Biomed Rep 2017; 6:307-313. [PMID: 28451391 DOI: 10.3892/br.2017.857] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 01/23/2017] [Indexed: 11/06/2022] Open
Abstract
Endotherapia (GEMSP) is a novel therapeutic approach for multiple sclerosis (MS). The aim of the present study was to demonstrate the efficiency of Endotherapia in the follow-up of 193 patients with MS. The efficiency coefficient that was evaluated was the Expanded Disability Status Scale (EDSS) score, which is a functional scale of MS progression. The evaluated score of each patient during follow-up visits was compared with the theoretical score of the disease progression without GEMSP. The evolution of the EDSS score was evaluated according to the inclusion score. The quantitative global study of the EDSS score highlighted a statistically significant difference between the final average scores of the treatment with GEMSP (M) and worldwide reference (R) groups. The improvement of the M group compared with the R group was 24.5%. According to the final EDSS scores, the study highlighted a difference in favor of the M group with 62.0% for scores ≤3, 7.8% for scores between 3 and 6 and 19.6% for scores ≥6. According to the qualitative evolution of the EDSS scores, the improvements in favor of group M were 49.3% for scores ≤3, 79.1% for scores between 3 and 6 and 19.5% for scores ≥6. The qualitative study of the EDSS score showed a statistically significant success percentage; the success percentages were between 59.1 and 90.0%. In a larger population of MS patients, the data confirm the beneficial effects of GEMSP that were previously reported in pre-clinical and clinical studies. In addition, 78% of patients showed an improvement or deceleration of the disease.
Collapse
Affiliation(s)
- Michel Geffard
- Institut pour le Développement de la Recherche en Pathologie Humaine et Thérapeutique, 33400 Talence, France
| | - Arturo Mangas
- Gemacbio, Research Department, Lieu dit Berganton, 33127 Saint Jean d'Illac, France
| | - Rafael Coveñas
- Institute of Neurosciences of Castilla y León, Laboratory of Neuroanatomy of the Peptidergic Systems, University of Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
28
|
Tauskela JS, Bourourou M, Blondeau N. Tackling issues in the path toward clinical translation in brain conditioning: Potential offered by nutraceuticals. Brain Circ 2017; 3:78-86. [PMID: 30276308 PMCID: PMC6126266 DOI: 10.4103/bc.bc_8_17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 05/23/2017] [Accepted: 05/31/2017] [Indexed: 11/21/2022] Open
Abstract
Brief periods of ischemia have been shown in many experimental setups to provide tolerance against ischemia in multiple organs including the brain, when administered before (preconditioning) or even after (postconditioning) the normally lethal ischemia. In addition to these so-called ischemic conditionings, many pharmacological and natural agents (e.g., chemicals and nutraceuticals) can also act as potent pre- and post-conditioners. Deriving from the original concept of ischemic preconditioning, these various conditioning paradigms may be promising as clinical-stage therapies for prevention of ischemic-related injury, especially stroke. As no proven experimentally identified strategy has translated into clinical success, the experimental induction of neuroprotection using these various conditioning paradigms has raised several questions, even before considering translation to clinical studies in humans. The first aim of the review is to consider key questions on preclinical studies of pre- or post-conditioning modalities including those induced by chemical or nutraceuticals. Second, we make the argument that several key issues can be addressed by a novel concept, nutraceutical preconditioning. Specifically, α-linolenic acid (alpha-linolenic acid [ALA] an omega-3 polyunsaturated fatty acid), contained in plant-derived edible products, is essential in the daily diet, and a body of work has identified ALA as a pre- and post-conditioner of the brain. Nutritional intervention and functional food development are an emerging direction for preventing stroke damage, offering the potential to improving clinical outcomes through activation of the endogenous protective mechanisms known collectively as conditioning.
Collapse
Affiliation(s)
- Joseph S Tauskela
- Department of Translational Bioscience, Human Health Therapeutics, National Research Council Canada, Ottawa, Ontario, Canada K1A 0R6
| | - Miled Bourourou
- University of Côte d'Azur, Centre National de la Recherche Scientifique, IPMC, UMR7275 Sophia Antipolis, F-06560, France
| | - Nicolas Blondeau
- University of Côte d'Azur, Centre National de la Recherche Scientifique, IPMC, UMR7275 Sophia Antipolis, F-06560, France
| |
Collapse
|
29
|
Antollini SS, Barrantes FJ. Fatty Acid Regulation of Voltage- and Ligand-Gated Ion Channel Function. Front Physiol 2016; 7:573. [PMID: 27965583 PMCID: PMC5124694 DOI: 10.3389/fphys.2016.00573] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 11/09/2016] [Indexed: 12/25/2022] Open
Abstract
Free fatty acids (FFA) are essential components of the cell, where they play a key role in lipid and carbohydrate metabolism, and most particularly in cell membranes, where they are central actors in shaping the physicochemical properties of the lipid bilayer and the cellular adaptation to the environment. FFA are continuously being produced and degraded, and a feedback regulatory function has been attributed to their turnover. The massive increase observed under some pathological conditions, especially in brain, has been interpreted as a protective mechanism possibly operative on ion channels, which in some cases is of stimulatory nature and in other cases inhibitory. Here we discuss the correlation between the structure of FFA and their ability to modulate protein function, evaluating the influence of saturation/unsaturation, number of double bonds, and cis vs. trans isomerism. We further focus on the mechanisms of FFA modulation operating on voltage-gated and ligand-gated ion channel function, contrasting the still conflicting evidence on direct vs. indirect mechanisms of action.
Collapse
Affiliation(s)
- Silvia S Antollini
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (CONICET-UNS)Bahía Blanca, Argentina; Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del SurBahía Blanca, Argentina
| | | |
Collapse
|
30
|
Bourourou M, Heurteaux C, Blondeau N. Alpha-linolenic acid given as enteral or parenteral nutritional intervention against sensorimotor and cognitive deficits in a mouse model of ischemic stroke. Neuropharmacology 2016; 108:60-72. [PMID: 27133376 DOI: 10.1016/j.neuropharm.2016.04.040] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/26/2016] [Accepted: 04/27/2016] [Indexed: 02/03/2023]
Abstract
Stroke is a leading cause of disability and death worldwide. Numerous therapeutics applied acutely after stroke have failed to improve long-term clinical outcomes. An emerging direction is nutritional intervention with omega-3 polyunsaturated fatty acids acting as disease-modifying factors and targeting post-stroke disabilities. Our previous studies demonstrated that the omega-3 precursor, alpha-linolenic acid (ALA) administrated by injections or dietary supplementation reduces stroke damage by direct neuroprotection, and triggering brain artery vasodilatation and neuroplasticity. Successful translation of putative therapies will depend on demonstration of robust efficacy on common deficits resulting from stroke like loss of motor control and memory/learning. This study evaluated the value of ALA as adjunctive therapy for stroke recovery by comparing whether oral or intravenous supplementation of ALA best support recovery from ischemia. Motor and cognitive deficits were assessed using rotarod, pole and Morris water maze tests. ALA supplementation in diet was better than intravenous treatment in improving motor coordination, but this improvement was not due to a neuroprotective effect since infarct size was not reduced. Both types of ALA supplementation improved spatial learning and memory after stroke. This cognitive improvement correlated with higher survival of hippocampal neurons. These results support clinical investigation establishing therapeutic plans using ALA supplementation.
Collapse
Affiliation(s)
- Miled Bourourou
- Université de Nice Sophia Antipolis, IPMC, Sophia Antipolis, F-06560, France; CNRS, IPMC, Sophia Antipolis, F-06560, France
| | - Catherine Heurteaux
- Université de Nice Sophia Antipolis, IPMC, Sophia Antipolis, F-06560, France; CNRS, IPMC, Sophia Antipolis, F-06560, France
| | - Nicolas Blondeau
- Université de Nice Sophia Antipolis, IPMC, Sophia Antipolis, F-06560, France; CNRS, IPMC, Sophia Antipolis, F-06560, France.
| |
Collapse
|
31
|
Messamore E, McNamara RK. Detection and treatment of omega-3 fatty acid deficiency in psychiatric practice: Rationale and implementation. Lipids Health Dis 2016; 15:25. [PMID: 26860589 PMCID: PMC4748485 DOI: 10.1186/s12944-016-0196-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 02/04/2016] [Indexed: 11/10/2022] Open
Abstract
A body of translational evidence has implicated dietary deficiency in long-chain omega-3 (LCn-3) fatty acids, including eicosapenaenoic acid (EPA) and docosahexaenoic acid (DHA), in the pathophysiology and potentially etiology of different psychiatric disorders. Case–control studies have consistently observed low erythrocyte (red blood cell) EPA and/or DHA levels in patients with major depressive disorder, bipolar disorder, schizophrenia, and attention deficit hyperactivity disorder. Low erythrocyte EPA + DHA biostatus can be treated with fish oil-based formulations containing preformed EPA + DHA, and extant evidence suggests that fish oil supplementation is safe and well-tolerated and may have therapeutic benefits. These and other data provide a rationale for screening for and treating LCn-3 fatty acid deficiency in patients with psychiatric illness. To this end, we have implemented a pilot program that routinely measures blood fatty acid levels in psychiatric patients entering a residential inpatient clinic. To date over 130 blood samples, primarily from patients with treatment-refractory mood or anxiety disorders, have been collected and analyzed. Our initial results indicate that the majority (75 %) of patients exhibit whole blood EPA + DHA levels at ≤4 percent of total fatty acid composition, a rate that is significantly higher than general population norms (25 %). In a sub-set of cases, corrective treatment with fish oil-based products has resulted in improvements in psychiatric symptoms without notable side effects. In view of the urgent need for improvements in conventional treatment algorithms, these preliminary findings provide important support for expanding this approach in routine psychiatric practice.
Collapse
Affiliation(s)
- Erik Messamore
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, 260 Stetson Street, Rm. 3306, Cincinnati, OH, 45218-0516, USA.,Lindner Center of HOPE, Mason, OH, USA
| | - Robert K McNamara
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, 260 Stetson Street, Rm. 3306, Cincinnati, OH, 45218-0516, USA.
| |
Collapse
|
32
|
Weller J, Steinhäuser C, Seifert G. pH-Sensitive K+ Currents and Properties of K2P Channels in Murine Hippocampal Astrocytes. ION CHANNELS AS THERAPEUTIC TARGETS, PART A 2016; 103:263-94. [DOI: 10.1016/bs.apcsb.2015.10.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
33
|
Pusceddu MM, Nolan YM, Green HF, Robertson RC, Stanton C, Kelly P, Cryan JF, Dinan TG. The Omega-3 Polyunsaturated Fatty Acid Docosahexaenoic Acid (DHA) Reverses Corticosterone-Induced Changes in Cortical Neurons. Int J Neuropsychopharmacol 2015; 19:pyv130. [PMID: 26657646 PMCID: PMC4926793 DOI: 10.1093/ijnp/pyv130] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Accepted: 11/30/2015] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Chronic exposure to the glucocorticoid hormone corticosterone exerts cellular stress-induced toxic effects that have been associated with neurodegenerative and psychiatric disorders. Docosahexaenoic acid is a polyunsaturated fatty acid that has been shown to be of benefit in stress-related disorders, putatively through protective action in neurons. METHODS We investigated the protective effect of docosahexaenoic acid against glucocorticoid hormone corticosterone-induced cellular changes in cortical cell cultures containing both astrocytes and neurons. RESULTS We found that glucocorticoid hormone corticosterone (100, 150, 200 μM) at different time points (48 and 72 hours) induced a dose- and time-dependent reduction in cellular viability as assessed by methyl thiazolyl tetrazolium. Moreover, glucocorticoid hormone corticosterone (200 μM, 72 hours) decreased the percentage composition of neurons while increasing the percentage of astrocytes as assessed by βIII-tubulin and glial fibrillary acidic protein immunostaining, respectively. In contrast, docosahexaenoic acid treatment (6 μM) increased docosahexaenoic acid content and attenuated glucocorticoid hormone corticosterone (200 μM)-induced cell death (72 hours) in cortical cultures. This translates into a capacity for docosahexaenoic acid to prevent neuronal death as well as astrocyte overgrowth following chronic exposure to glucocorticoid hormone corticosterone. Furthermore, docosahexaenoic acid (6 μM) reversed glucocorticoid hormone corticosterone-induced neuronal apoptosis as assessed by terminal deoxynucleotidyl transferase-mediated nick-end labeling and attenuated glucocorticoid hormone corticosterone-induced reductions in brain derived neurotrophic factor mRNA expression in these cultures. Finally, docosahexaenoic acid inhibited glucocorticoid hormone corticosterone-induced downregulation of glucocorticoid receptor expression on βIII- tubulin-positive neurons. CONCLUSIONS This work supports the view that docosahexaenoic acid may be beneficial in ameliorating stress-related cellular changes in the brain and may be of value in psychiatric disorders.
Collapse
Affiliation(s)
- Matteo M Pusceddu
- Department of Psychiatry and Neurobehavioural Science (Dr Pusceddu and Prof. Dinan), APC Microbiome Institute (Drs Pusceddu and Stanton and Profs Cryan and Dinan), Department of Anatomy and Neuroscience (Drs Nolan and Green and Prof. Cryan), and School of Microbiology, University College Cork, Cork, Ireland (Mr Robertson); Moorepark Food Research Centre, Teagasc, Fermoy, Co. Cork, Ireland (Mr Robertson and Drs Stanton and Kelly)
| | - Yvonne M Nolan
- Department of Psychiatry and Neurobehavioural Science (Dr Pusceddu and Prof. Dinan), APC Microbiome Institute (Drs Pusceddu and Stanton and Profs Cryan and Dinan), Department of Anatomy and Neuroscience (Drs Nolan and Green and Prof. Cryan), and School of Microbiology, University College Cork, Cork, Ireland (Mr Robertson); Moorepark Food Research Centre, Teagasc, Fermoy, Co. Cork, Ireland (Mr Robertson and Drs Stanton and Kelly)
| | - Holly F Green
- Department of Psychiatry and Neurobehavioural Science (Dr Pusceddu and Prof. Dinan), APC Microbiome Institute (Drs Pusceddu and Stanton and Profs Cryan and Dinan), Department of Anatomy and Neuroscience (Drs Nolan and Green and Prof. Cryan), and School of Microbiology, University College Cork, Cork, Ireland (Mr Robertson); Moorepark Food Research Centre, Teagasc, Fermoy, Co. Cork, Ireland (Mr Robertson and Drs Stanton and Kelly)
| | - Ruairi C Robertson
- Department of Psychiatry and Neurobehavioural Science (Dr Pusceddu and Prof. Dinan), APC Microbiome Institute (Drs Pusceddu and Stanton and Profs Cryan and Dinan), Department of Anatomy and Neuroscience (Drs Nolan and Green and Prof. Cryan), and School of Microbiology, University College Cork, Cork, Ireland (Mr Robertson); Moorepark Food Research Centre, Teagasc, Fermoy, Co. Cork, Ireland (Mr Robertson and Drs Stanton and Kelly)
| | - Catherine Stanton
- Department of Psychiatry and Neurobehavioural Science (Dr Pusceddu and Prof. Dinan), APC Microbiome Institute (Drs Pusceddu and Stanton and Profs Cryan and Dinan), Department of Anatomy and Neuroscience (Drs Nolan and Green and Prof. Cryan), and School of Microbiology, University College Cork, Cork, Ireland (Mr Robertson); Moorepark Food Research Centre, Teagasc, Fermoy, Co. Cork, Ireland (Mr Robertson and Drs Stanton and Kelly)
| | - Philip Kelly
- Department of Psychiatry and Neurobehavioural Science (Dr Pusceddu and Prof. Dinan), APC Microbiome Institute (Drs Pusceddu and Stanton and Profs Cryan and Dinan), Department of Anatomy and Neuroscience (Drs Nolan and Green and Prof. Cryan), and School of Microbiology, University College Cork, Cork, Ireland (Mr Robertson); Moorepark Food Research Centre, Teagasc, Fermoy, Co. Cork, Ireland (Mr Robertson and Drs Stanton and Kelly)
| | - John F Cryan
- Department of Psychiatry and Neurobehavioural Science (Dr Pusceddu and Prof. Dinan), APC Microbiome Institute (Drs Pusceddu and Stanton and Profs Cryan and Dinan), Department of Anatomy and Neuroscience (Drs Nolan and Green and Prof. Cryan), and School of Microbiology, University College Cork, Cork, Ireland (Mr Robertson); Moorepark Food Research Centre, Teagasc, Fermoy, Co. Cork, Ireland (Mr Robertson and Drs Stanton and Kelly).
| | - Timothy G Dinan
- Department of Psychiatry and Neurobehavioural Science (Dr Pusceddu and Prof. Dinan), APC Microbiome Institute (Drs Pusceddu and Stanton and Profs Cryan and Dinan), Department of Anatomy and Neuroscience (Drs Nolan and Green and Prof. Cryan), and School of Microbiology, University College Cork, Cork, Ireland (Mr Robertson); Moorepark Food Research Centre, Teagasc, Fermoy, Co. Cork, Ireland (Mr Robertson and Drs Stanton and Kelly)
| |
Collapse
|
34
|
Furukawa A, Koriyama Y. A role of Heat Shock Protein 70 in Photoreceptor Cell Death: Potential as a Novel Therapeutic Target in Retinal Degeneration. CNS Neurosci Ther 2015; 22:7-14. [PMID: 26507240 DOI: 10.1111/cns.12471] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 09/23/2015] [Accepted: 09/25/2015] [Indexed: 01/17/2023] Open
Abstract
Retinal degenerative diseases (RDs) such as retinitis pigmentosa (RP) are a genetically heterogeneous group of disorders characterized by night blindness and peripheral vision loss, which caused by the dysfunction and death of photoreceptor cells. Although many causative gene mutations have been reported, the final common end stage is photoreceptor cell death. Unfortunately, no effective treatments or therapeutic agents have been discovered. Heat shock protein 70 (HSP70) is highly conserved and has antiapoptotic activities. A few reports have shown that HSP70 plays a role in RDs. Thus, we focused on the role of HSP70 in photoreceptor cell death. Using the N-methyl-N-nitrosourea (MNU)-induced photoreceptor cell death model in mice, we could examine two stages of the novel cell death mechanism; the early stage, including HSP70 cleavage through protein carbonylation by production of reactive oxygen species, lipid peroxidation and Ca(2+) influx/calpain activation, and the late stage of cathepsin and/or caspase activation. The upregulation of intact HSP70 expression by its inducer is likely to protect photoreceptor cells. In this review, we focus on the role of HSP70 and the novel cell death signaling process in RDs. We also describe candidate therapeutic agents for RDs.
Collapse
Affiliation(s)
- Ayako Furukawa
- Graduate School and Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan
| | - Yoshiki Koriyama
- Graduate School and Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan
| |
Collapse
|
35
|
Sabour H, Norouzi Javidan A, Latifi S, Shidfar F, Heshmat R, Emami Razavi SH, Vafa MR, Larijani B. Omega-3 fatty acids' effect on leptin and adiponectin concentrations in patients with spinal cord injury: A double-blinded randomized clinical trial. J Spinal Cord Med 2015; 38:599-606. [PMID: 25096818 PMCID: PMC4535802 DOI: 10.1179/2045772314y.0000000251] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
CONTEXT Omega-3 fatty acids have been recently proposed to induce neural improvement in patients with spinal cord injury (SCI) while affecting some hormones including leptin and adiponectin. OBJECTIVES We tried to evaluate the effect of omega-3 fatty acids on circulatory concentrations of leptin and adiponectin among these patients. DESIGN This study is a double-blinded randomized clinical trial with intervention duration of 14 months. SETTING A tertiary rehabilitation center. PARTICIPANTS Total of 104 patients with SCI who did not meet our exclusion criteria entered the study. Those with history of diabetes, cancer, endocrinology disease, acute infection, and use of special medications were excluded. Patients were divided randomly into the treatment and control group by using permuted balanced block randomization. INTERVENTION The treatment group received two MorDHA® capsules per day (each capsule contain 465 mg of docosahexaenoic acid (DHA) and 63 mg of eicosapentaenoic acid (EPA)) for 14 months while the control group received placebo capsules with similar color, shape, and taste. MAIN OUTCOMES MEASURES Leptin and adiponectin concentrations in plasma were measured at the beginning of trial and then after 6 and 14 months. RESULTS Fourteen months of treatment with DHA and EPA did not influence concentrations of leptin but adiponectin level was significantly decreased (P: 0.03). Weight was positively correlated with leptin level at stage 0 of trial (P: 0.008, r=0.41) while this association was attenuated through stages of trial after intervention. CONCLUSION Our data show that omega-3 fatty acids may not affect plasma concentrations of leptin but adiponectin level is decreased in patients with SCI. Moreover, this intervention influences the linear relationship between weight and leptin after 14 months administration of DHA and EPA.
Collapse
Affiliation(s)
- Hadis Sabour
- Brain and Spinal Injury Research Center (BASIR), Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Norouzi Javidan
- Brain and Spinal Injury Research Center (BASIR), Tehran University of Medical Sciences, Tehran, Iran
| | - Sahar Latifi
- Brain and Spinal Injury Research Center (BASIR), Tehran University of Medical Sciences, Tehran, Iran
| | - Farzad Shidfar
- Nutrition Department, Iran University of Medical Sciences, Tehran, Iran
| | - Ramin Heshmat
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed-Hassan Emami Razavi
- Brain and Spinal Injury Research Center (BASIR), Tehran University of Medical Sciences, Tehran, Iran
| | | | - Bagher Larijani
- Brain and Spinal Injury Research Center (BASIR), Tehran University of Medical Sciences, Tehran, Iran,Correspondence to: Bagher Larijani, Endocrinology and Metabolism Research Institute, 5th floor, Shariati Hospital, North Kargar Avenue, 1411413137, Tehran, Iran. ;
| |
Collapse
|
36
|
Blondeau N. The nutraceutical potential of omega-3 alpha-linolenic acid in reducing the consequences of stroke. Biochimie 2015; 120:49-55. [PMID: 26092420 DOI: 10.1016/j.biochi.2015.06.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 06/08/2015] [Indexed: 01/01/2023]
Abstract
Stroke is a worldwide major cause of mortality and morbidity. Preclinical studies have identified over 1000 molecules with brain-protective properties. More than 200 clinical trials have evaluated neuroprotective candidates for ischemic stroke yet, to date almost all failed, leading to a re-analysis of treatment strategies against stroke. An emerging view is to seek combinatory therapy, or discovering molecules able to stimulate multiple protective and regenerative mechanisms. A pertinent experimental approach to identify such candidates is the study of brain preconditioning, which refers to how the brain protects itself against ischemia and others stress-inducing stimuli. The recent discovery that nutrients like alpha-linolenic acid (ALA is an essential omega-3 polyunsaturated fatty acid required as part of our daily diet), may be an efficient brain preconditionner against stroke fosters the novel concept of brain preconditioning by nutraceuticals. This review stresses the underestimated role of nutrition in preventing and combating stroke. Although there is a consensus that increased consumption of salt, fatty foods and alcoholic beverages may promote pathologies like hypertension, obesity and alcoholism - all of which are well known risk factors of stroke - few risk factors are attributed to a deficiency in an essential nutrient in the diet. The ALA deficiency observed in the Western modern diets may itself constitute a risk factor. This review outlines how ALA supplementation by modification of the daily diet prevented mortality and cerebral damage in a rodent model of ischemic stroke. It also describes the pleiotropic ability of ALA to trigger responses that are multicellular, mechanistically diverse, resulting in neuronal protection, stimulation of neuroplasticity, and brain artery vasodilation. Overall, this review proposes a promising therapeutic opportunity by integrating a nutritional-based approach focusing on enriching the daily diet in ALA to prevent the devastating damage caused by stroke.
Collapse
Affiliation(s)
- Nicolas Blondeau
- Université de Nice Sophia Antipolis, IPMC, Sophia Antipolis, F-06560, France; CNRS, IPMC, Sophia Antipolis, F-06560, France.
| |
Collapse
|
37
|
Kuang Q, Purhonen P, Hebert H. Structure of potassium channels. Cell Mol Life Sci 2015; 72:3677-93. [PMID: 26070303 PMCID: PMC4565861 DOI: 10.1007/s00018-015-1948-5] [Citation(s) in RCA: 177] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 05/09/2015] [Accepted: 06/03/2015] [Indexed: 12/25/2022]
Abstract
Potassium channels ubiquitously exist in nearly all kingdoms of life and perform diverse but important functions. Since the first atomic structure of a prokaryotic potassium channel (KcsA, a channel from Streptomyces lividans) was determined, tremendous progress has been made in understanding the mechanism of potassium channels and channels conducting other ions. In this review, we discuss the structure of various kinds of potassium channels, including the potassium channel with the pore-forming domain only (KcsA), voltage-gated, inwardly rectifying, tandem pore domain, and ligand-gated ones. The general properties shared by all potassium channels are introduced first, followed by specific features in each class. Our purpose is to help readers to grasp the basic concepts, to be familiar with the property of the different domains, and to understand the structure and function of the potassium channels better.
Collapse
Affiliation(s)
- Qie Kuang
- Department of Biosciences and Nutrition, Karolinska Institutet, Novum, 14183, Huddinge, Sweden.
- School of Technology and Health, KTH Royal Institute of Technology, Novum, 14183, Huddinge, Sweden.
| | - Pasi Purhonen
- Department of Biosciences and Nutrition, Karolinska Institutet, Novum, 14183, Huddinge, Sweden
| | - Hans Hebert
- Department of Biosciences and Nutrition, Karolinska Institutet, Novum, 14183, Huddinge, Sweden
- School of Technology and Health, KTH Royal Institute of Technology, Novum, 14183, Huddinge, Sweden
| |
Collapse
|
38
|
Alpha-Linolenic Acid-Induced Increase in Neurogenesis is a Key Factor in the Improvement in the Passive Avoidance Task After Soman Exposure. Neuromolecular Med 2015; 17:251-69. [DOI: 10.1007/s12017-015-8353-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 04/11/2015] [Indexed: 02/01/2023]
|
39
|
Alyssum homolocarpum seeds: phytochemical analysis and effects of the seed oil on neural stem cell proliferation and differentiation. J Nat Med 2015; 69:387-96. [PMID: 25860174 DOI: 10.1007/s11418-015-0905-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 03/24/2015] [Indexed: 10/23/2022]
Abstract
Pharmacognostic evaluation of medicinal plants may assess their current applications and possibly results in finding new active components. In this study, ash and extractive values and high performance thin layer chromatography fingerprints of Alyssum homolocarpum (Brassicaceae) seed extracts were investigated to elucidate its composition. Differential scanning calorimetry and gas chromatography-mass spectrometry analysis were employed to determine the components of A. homolocarpum seed oil (AHO). Neurosphere assay, in vitro differentiation and immunofluorescence analysis were performed to evaluate the effects of oral administration of AHO (0.5 or 1 g/kg/day for 14 days) on proliferation and differentiation of neural stem cells (NSCs) in adult male BALB/c mice. Total, acid-insoluble and water-soluble ash values were determined as 45.83 ± 5.85, 6.67 ± 2.89 and 28.33 ± 2.89 mg/g, respectively. The extractive values were 4.90, 0.43 and 0.56 % (w/w) for n-hexane, dichloromethane and ethanolic extracts, respectively. Interestingly, AHO was mainly composed of α-linolenic acid (89.71 %), β-sitosterol (3.3 mg/g) and campesterol (0.86 mg/g). Administration of AHO at 1 g/kg/day significantly increased proliferation of NSCs, as evidenced by an increase in mean neurosphere-forming frequency per brain (872.7 ± 15.17) and neurosphere diameter (101 ± 2.48 µm) compared to the control group (424.3 ± 59.29 and 78.63 ± 1.7 µm, respectively; P < 0.05). AHO treatment did not affect in vitro differentiation of the harvested NSCs. Our data show that A. homolocarpum seed oil is a rich source of α-linolenic acid and β-sitosterol with potential therapeutic application to enhance NSC proliferation and recruitment in neurological diseases.
Collapse
|
40
|
Hadjighassem M, Kamalidehghan B, Shekarriz N, Baseerat A, Molavi N, Mehrpour M, Joghataei MT, Tondar M, Ahmadipour F, Meng GY. Oral consumption of α-linolenic acid increases serum BDNF levels in healthy adult humans. Nutr J 2015; 14:20. [PMID: 25889793 PMCID: PMC4353682 DOI: 10.1186/s12937-015-0012-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 02/16/2015] [Indexed: 02/04/2023] Open
Abstract
Background aims Dietary omega-6 and omega-3 fatty acids have remarkable impacts on the levels of DHA in the brain and retina. Low levels of DHA in plasma and blood hamper visual and neural development in children and cause dementia and cognitive decline in adults. The level of brain-derived neurotrophic factors (BDNF) changes with dietary omega-3 fatty acid intake. BDNF is known for its effects on promoting neurogenesis and neuronal survival. Methods In this study, we examined the effect of the oral consumption of α-Linolenic acid (ALA) on blood levels of BDNF and Malondialdehyde (MDA) in healthy adult humans. 30 healthy volunteers, 15 men and 15 women, were selected randomly. Each individual served as his or her own control. Before consuming the Flaxseed oil capsules, 5cc blood from each individual was sampled in order to measure the plasma levels of BDNF and MDA as baseline controls. During the experiment, each individual was given 3 oral capsules of flaxseed oil, containing 500mg of alpha linolenic acid, daily for one week. Then, plasma levels of BDNF and MDA were tested. Results The plasma levels of BDNF and MDA significantly (P < 0.05) increased in individuals who received the oral capsules of ALA. Plasma levels of BDNF increased more in the women in comparison with the men. Conclusion ALA treatment could be a feasible approach to reduce size of infarcts in stroke patients. Thus, ALA could be used in adjunction with routine stroke therapies to minimize brain lesions caused by stroke.
Collapse
Affiliation(s)
- Mahmoudreza Hadjighassem
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran. .,Brain and Spinal Cord Research Center, School of Advanced Medical Technologies, Tehran University of Medical Sciences, Tehran, Iran.
| | - Behnam Kamalidehghan
- Department of Pharmacy, Faculty of Medicine, University of Malaya (UM), Kuala Lumpur, Malaysia.
| | - Nima Shekarriz
- Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Argavan Baseerat
- Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Nima Molavi
- Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Masoud Mehrpour
- Department of Neurology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Taghi Joghataei
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran. .,Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Mahdi Tondar
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, USA.
| | - Fatemeh Ahmadipour
- Department of Pharmacy, Faculty of Medicine, University of Malaya (UM), Kuala Lumpur, Malaysia.
| | - Goh Yong Meng
- Department of Animal Science, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Malaysia.
| |
Collapse
|
41
|
Alpha-linolenic acid: an omega-3 fatty acid with neuroprotective properties-ready for use in the stroke clinic? BIOMED RESEARCH INTERNATIONAL 2015; 2015:519830. [PMID: 25789320 PMCID: PMC4350958 DOI: 10.1155/2015/519830] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 09/08/2014] [Indexed: 01/29/2023]
Abstract
Alpha-linolenic acid (ALA) is plant-based essential omega-3 polyunsaturated fatty acids that must be obtained through the diet. This could explain in part why the severe deficiency in omega-3 intake pointed by numerous epidemiologic studies may increase the brain's vulnerability representing an important risk factor in the development and/or deterioration of certain cardio- and neuropathologies. The roles of ALA in neurological disorders remain unclear, especially in stroke that is a leading cause of death. We and others have identified ALA as a potential nutraceutical to protect the brain from stroke, characterized by its pleiotropic effects in neuroprotection, vasodilation of brain arteries, and neuroplasticity. This review highlights how chronic administration of ALA protects against rodent models of hypoxic-ischemic injury and exerts an anti-depressant-like activity, effects that likely involve multiple mechanisms in brain, and may be applied in stroke prevention. One major effect may be through an increase in mature brain-derived neurotrophic factor (BDNF), a widely expressed protein in brain that plays critical roles in neuronal maintenance, and learning and memory. Understanding the precise roles of ALA in neurological disorders will provide the underpinnings for the development of new therapies for patients and families who could be devastated by these disorders.
Collapse
|
42
|
Martinc B, Grabnar I, Vovk T. Antioxidants as a preventive treatment for epileptic process: a review of the current status. Curr Neuropharmacol 2014; 12:527-50. [PMID: 25977679 PMCID: PMC4428026 DOI: 10.2174/1570159x12666140923205715] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Revised: 07/29/2014] [Accepted: 09/22/2014] [Indexed: 11/25/2022] Open
Abstract
Epilepsy is known as one of the most frequent neurological diseases, characterized by an enduring predisposition to generate epileptic seizures. Oxidative stress is believed to directly participate in pathways leading to neurodegeneration, which serves as the most important propagating factor, leading to the epileptic condition and cognitive decline. Moreover, there is also a growing body of evidence showing the disturbance of antioxidant system balance and consequently increased production of reactive species in patients with epilepsy. A meta-analysis, conducted in the present review confirms an association between epilepsy and increased lipid peroxidation. Furthermore, it was also shown that some of the antiepileptic drugs could potentially be responsible for additionally increased lipid peroxidation. Therefore, it is reasonable to propose that during the epileptic process neuroprotective treatment with antioxidants could lead to less sever structural damages, reduced epileptogenesis and milder cognitive deterioration. To evaluate this hypothesis studies investigating the neuroprotective therapeutic potential of various antioxidants in cells, animal seizure models and patients with epilepsy have been reviewed. Numerous beneficial effects of antioxidants on oxidative stress markers and in some cases also neuroprotective effects were observed in animal seizure models. However, despite these encouraging results, till now only a few antioxidants have been further applied to patients with epilepsy as an add-on therapy. Based on the several positive findings in animal models, a strong need for more carefully planned, randomized, double-blind, cross-over, placebo-controlled clinical trials for the evaluation of antioxidants efficacy in patients with epilepsy is warranted.
Collapse
Affiliation(s)
| | | | - Tomaž Vovk
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| |
Collapse
|
43
|
Göb E, Bittner S, Bobak N, Kraft P, Göbel K, Langhauser F, Homola GA, Brede M, Budde T, Meuth SG, Kleinschnitz C. The two-pore domain potassium channel KCNK5 deteriorates outcome in ischemic neurodegeneration. Pflugers Arch 2014; 467:973-87. [PMID: 25315980 DOI: 10.1007/s00424-014-1626-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 09/29/2014] [Accepted: 10/01/2014] [Indexed: 01/05/2023]
Abstract
Potassium channels can fulfill both beneficial and detrimental roles in neuronal damage during ischemic stroke. Earlier studies have characterized a neuroprotective role of the two-pore domain potassium channels KCNK2 (TREK1) and KCNK3 (TASK1). Protective neuronal hyperpolarization and prevention of intracellular Ca(2+) overload and glutamate excitotoxicity were suggested to be the underlying mechanisms. We here identify an unexpected role for the related KCNK5 channel in a mouse model of transient middle cerebral artery occlusion (tMCAO). KCNK5 is strongly upregulated on neurons upon cerebral ischemia, where it is most likely involved in the induction of neuronal apoptosis. Hypoxic conditions elevated neuronal expression levels of KCNK5 in acute brain slices and primary isolated neuronal cell cultures. In agreement, KCNK5 knockout mice had significantly reduced infarct volumes and improved neurologic function 24 h after 60 min of tMCAO and this protective effect was preserved at later stages of infarct development. KCNK5 deficiency resulted in a significantly reduced number of apoptotic neurons, a downregulation of pro-apoptotic and upregulation of anti-apoptotic factors. Results of adoptive transfer experiments of wild-type and Kcnk5 (-/-) immune cells into Rag1 (-/-) mice prior to tMCAO exclude a major role of KCNK5 in poststroke inflammatory reactions. In summary, KCNK5 expression is induced on neurons under ischemic conditions where it most likely exerts pro-apoptotic effects. Hence, pharmacological blockade of KCNK5 might have therapeutic potential in preventing ischemic neurodegeneration.
Collapse
Affiliation(s)
- Eva Göb
- Department of Neurology, University Clinics Würzburg, Würzburg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Liu Y, Sun Q, Chen X, Jing L, Wang W, Yu Z, Zhang G, Xie M. Linolenic acid provides multi-cellular protective effects after photothrombotic cerebral ischemia in rats. Neurochem Res 2014; 39:1797-808. [PMID: 25062759 DOI: 10.1007/s11064-014-1390-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 07/02/2014] [Accepted: 07/15/2014] [Indexed: 10/25/2022]
Abstract
Alpha-linolenic acid (LIN) has been shown to provide neuroprotective effects against cerebral ischemia. LIN is a potent activator of TREK-1 channel and LIN-induced neuroprotection disappears in Trek1-/- mice, suggesting that this channel is directly related to the LIN-induced resistance of brain against ischemia. However, the cellular mechanism underlying LIN induced neuroprotective effects after ischemia remains unclear. In this study, using a rat photochemical brain ischemia model, we investigated the effects of LIN on the protein abundance of astrocytic glutamate transporter and AQP4, microglia activation, cell apoptosis and behavioral recovery following ischemia. Administration of LIN rescued the protein abundance of astrocytic glutamate transporter GLT-1, decreased the protein abundance of AQP4 and brain edema, inhibited microglia activation, attenuated cell apoptosis and improved behavioral function recovery. Meanwhile, TREK-1 was widely distributed in the cortex and hippocampus, primarily localized in astrocytes and neurons. LIN could potentiate the TREK-1 mediated astrocytic passive conductance and hyperpolarize the membrane potential. Our results suggest that LIN provides multiple cellular neuroprotective effects in cerebral ischemia. TREK-1 may serve as a promising multi-mechanism therapeutic target for the treatment of stroke.
Collapse
Affiliation(s)
- Yang Liu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Traumatic brain injury (TBI) is the most important cause of disability in individuals under the age of 45 years and thus represents a significant social and economic burden. Evidence strongly suggests that oxidative stress is a cornerstone event leading to and propagating secondary injury mechanisms such as excitotoxicity, mitochondrial dysfunction, apoptosis, autophagy, brain edema, and inflammation. TBI has defied conventional approaches to diagnosis and therapy development because of its heterogeneity and complexity. Therefore, it is necessary to explore alternative approaches to therapy development for TBI. The aim of this review is to present a therapeutic approach for TBI, taking into account the evidence supporting the role for oxidative stress in the pathophysiological processes of secondary brain injury. The role of agents such as mitochondria-targeted antioxidants (melatonin and new mitochondria-targeted antioxidants), nicotinamide adenine dinucleotide phosphate (NADPH) inhibitors (antioxidant vitamins and apocynin), and other compounds having mainly antioxidant properties (hydrogen-rich saline, sulforaphane, U-83836E, omega-3, and polyphenols) is covered. The rationale for innovative antioxidant therapies based on current knowledge and particularly the most recent studies regarding this field is discussed. Particular considerations and translational potential of new TBI treatments are examined and a novel therapeutic proposal for TBI is presented.
Collapse
|
46
|
Abstract
A transient, ischemia-resistant phenotype known as "ischemic tolerance" can be established in brain in a rapid or delayed fashion by a preceding noninjurious "preconditioning" stimulus. Initial preclinical studies of this phenomenon relied primarily on brief periods of ischemia or hypoxia as preconditioning stimuli, but it was later realized that many other stressors, including pharmacologic ones, are also effective. This review highlights the surprisingly wide variety of drugs now known to promote ischemic tolerance, documented and to some extent mechanistically characterized in preclinical animal models of stroke. Although considerably more experimentation is needed to thoroughly validate the ability of any currently identified preconditioning agent to protect ischemic brain, the fact that some of these drugs are already clinically approved for other indications implies that the growing enthusiasm for translational success in the field of pharmacologic preconditioning may be well justified.
Collapse
|
47
|
McNamara RK, Jandacek R, Tso P, Weber W, Chu WJ, Strakowski SM, Adler CM, Delbello MP. Low docosahexaenoic acid status is associated with reduced indices in cortical integrity in the anterior cingulate of healthy male children: a 1H MRS Study. Nutr Neurosci 2013; 16:183-90. [PMID: 23582513 DOI: 10.1179/1476830512y.0000000045] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Docosahexaenoic acid (DHA, 22:6n-3) is the principal omega-3 fatty acid in mammalian brain gray matter, and emerging preclinical evidence suggests that DHA has neurotrophic and neuroprotective properties. This study investigated relationships among DHA status, neurocognitive performance, and cortical metabolism measured with proton magnetic resonance spectroscopy (1H MRS) in healthy developing male children (aged 8-10 years, n = 38). Subjects were segregated into low-DHA (n = 19) and high-DHA (n = 19) status groups by a median split of erythrocyte DHA levels. Group differences in 1H MRS indices of cortical metabolism, including choline (Cho), creatine (Cr), glutamine + glutamate + γ-aminobutyric acid (Glx), myo-inositol (mI), and n-acetyl aspartate (NAA), were determined in the right and left dorsolateral prefrontal cortex (R/L-DLPFC, BA9) and bilateral anterior cingulate cortex (ACC, BA32/33). Group differences in neurocognitive performance were evaluated with the Kaufman Brief Intelligence Test and identical-pairs version of the continuous performance task (CPT-IP). Subjects in the low-DHA group consumed fish less frequently (P = 0.02), had slower reaction times on the CPT-IP (P = 0.007), and exhibited lower mI (P = 0.007), NAA (P = 0.007), Cho (P = 0.009), and Cr (P = 0.01) concentrations in the ACC compared with the high-DHA group. There were no group differences in ACC Glx or any metabolite in the L-DLPFC and R-DLPFC. These data indicate that low-DHA status is associated with reduced indices of metabolic function in the ACC and slower reaction time during sustained attention in developing male children.
Collapse
Affiliation(s)
- Robert K McNamara
- Department of Psychiatry and Behavioral Neuroscience, Center for Imaging Research, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Omega-3 polyunsaturated fatty acid supplementation improves neurologic recovery and attenuates white matter injury after experimental traumatic brain injury. J Cereb Blood Flow Metab 2013; 33:1474-84. [PMID: 23801244 PMCID: PMC3764381 DOI: 10.1038/jcbfm.2013.108] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 05/30/2013] [Accepted: 06/04/2013] [Indexed: 12/26/2022]
Abstract
Dietary supplementation with omega-3 (ω-3) fatty acids is a safe, economical mean of preventive medicine that has shown protection against several neurologic disorders. The present study tested the hypothesis that this method is protective against controlled cortical impact (CCI). Indeed, mice fed with ω-3 polyunsaturated fatty acid (PUFA)-enriched diet for 2 months exhibited attenuated short and long-term behavioral deficits due to CCI. Although ω-3 PUFAs did not decrease cortical lesion volume, these fatty acids did protect against hippocampal neuronal loss after CCI and reduced pro-inflammatory response. Interestingly, ω-3 PUFAs prevented the loss of myelin basic protein (MPB), preserved the integrity of the myelin sheath, and maintained the nerve fiber conductivity in the CCI model. ω-3 PUFAs also directly protected oligodendrocyte cultures from excitotoxicity and blunted the microglial activation-induced death of oligodendrocytes in microglia/oligodendrocyte cocultures. In sum, ω-3 PUFAs elicit multifaceted protection against behavioral dysfunction, hippocampal neuronal loss, inflammation, and loss of myelination and impulse conductivity. The present report is the first demonstration that ω-3 PUFAs protect against white matter injury in vivo and in vitro. The protective impact of ω-3 PUFAs supports the clinical use of this dietary supplement as a prophylaxis against traumatic brain injury and other nervous system disorders.
Collapse
|
49
|
Kuratko CN, Barrett EC, Nelson EB, Norman S. The relationship of docosahexaenoic acid (DHA) with learning and behavior in healthy children: a review. Nutrients 2013; 5:2777-810. [PMID: 23877090 PMCID: PMC3738999 DOI: 10.3390/nu5072777] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 06/04/2013] [Accepted: 06/08/2013] [Indexed: 12/13/2022] Open
Abstract
Childhood is a period of brain growth and maturation. The long chain omega-3 fatty acid, docosahexaenoic acid (DHA), is a major lipid in the brain recognized as essential for normal brain function. In animals, low brain DHA results in impaired learning and behavior. In infants, DHA is important for optimal visual and cognitive development. The usual intake of DHA among toddlers and children is low and some studies show improvements in cognition and behavior as the result of supplementation with polyunsaturated fatty acids including DHA. The purpose of this review was to identify and evaluate current knowledge regarding the relationship of DHA with measures of learning and behavior in healthy school-age children. A systematic search of the literature identified 15 relevant publications for review. The search found studies which were diverse in purpose and design and without consistent conclusions regarding the treatment effect of DHA intake or biomarker status on specific cognitive tests. However, studies of brain activity reported benefits of DHA supplementation and over half of the studies reported a favorable role for DHA or long chain omega-3 fatty acids in at least one area of cognition or behavior. Studies also suggested an important role for DHA in school performance.
Collapse
Affiliation(s)
- Connye N. Kuratko
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-443-542-2552; Fax: +1-410-740-2985
| | - Erin Cernkovich Barrett
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-443-542-2552; Fax: +1-410-740-2985
| | - Edward B. Nelson
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-443-542-2552; Fax: +1-410-740-2985
| | - Salem Norman
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-443-542-2552; Fax: +1-410-740-2985
| |
Collapse
|
50
|
Antioxidant, anticonvulsive and neuroprotective effects of dapsone and phenobarbital against kainic acid-induced damage in rats. Neurochem Res 2013; 38:1819-27. [PMID: 23729301 DOI: 10.1007/s11064-013-1087-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 05/20/2013] [Accepted: 05/24/2013] [Indexed: 01/20/2023]
Abstract
Excitotoxicity due to glutamate receptors (GluRs) overactivation is a leading mechanism of oxidative damage and neuronal death in various diseases. We have shown that dapsone (DDS) was able to reduce both neurotoxicity and seizures associated to the administration of kainic acid (KA), an agonist acting on AMPA/KA receptors (GluK1-GluK5). Recently, it has been shown that phenobarbital (PB) is also able to reduce epileptic activity evoked by that receptor. In the present study, we tested the antioxidative, anticonvulsive and neuroprotective effects of DDS and PB administered alone or in combination upon KA toxicity to rats. Results showed that KA increased lipid peroxidation and diminished reduced glutathione (GSH), 24 h after KA administration and both drugs in combination or individually inhibited these events. Likewise, KA promotes mortality and this event was antagonized by effect of both treatments. Additionally, the behavioral evaluation showed that DDS and PB administered alone or in combination decreased the number of limbic seizures and reduced the percentage of animals showing tonic-clonic seizures versus the control group, which was administered only with KA. Finally, our study demonstrated that all of the treatments prevented the neuronal death of the pyramidal cell layer of hippocampal CA-3. In conclusion, the treatment with DDS and PB administrated alone or in combination exerted antioxidant, anticonvulsive and neuroprotective effects against the neurotoxicity induced by KA in rats, but their effects were not additive. Thus, it may be good options of treatment in diseases such as epilepsy and status epilepicus, administered separately.
Collapse
|