1
|
Chen MS, Hu CL, Jiang SK, Chong ZY, Chen JC. Modulation of secretory factors by lipofundin contributes to its anti‑neuroinflammatory effects. Exp Ther Med 2024; 27:169. [PMID: 38476917 PMCID: PMC10929000 DOI: 10.3892/etm.2024.12456] [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: 10/02/2023] [Accepted: 01/16/2024] [Indexed: 03/14/2024] Open
Abstract
As the global population ages, the prevalence of neuroinflammatory diseases such as Alzheimer's disease, Parkinson's disease and stroke continues to increase. Therefore, it is necessary to develop preventive and therapeutic methods against neuroinflammatory diseases. Lipofundin is a lipid emulsion commonly used in clinical anesthetic solvents and nutritional supplements. Lipid emulsions have been shown to possess anti-inflammatory properties. However, the potential beneficial effect of lipofundin against neuroinflammation requires elucidation. In the present study, two cell models were used to investigate the efficacy of lipofundin against neuroinflammation. In the first model, BV2 mouse microglial cells were treated with lipopolysaccharide (LPS) to induce nitric oxide (NO) production as a model of neuroinflammation. In the second model, HMC3 human microglial were activated by LPS, and changes in the secretion of factors associated with inflammation were analyzed using Luminex xMAP® technology. Griess assay results revealed that lipofundin significantly prevented and treated LPS-induced NO production. An anti-neuroinflammatory effect was also observed in HMC3 cells, where lipofundin exhibited excellent preventive and therapeutic properties by reducing the LPS-induced expression and secretion of interleukin-1β. Notably, lipofundin also promoted the secretion of certain growth factors, suggesting a potential neuroprotective effect. These results demonstrate that, in addition to its role as a solvent for drugs and nutritional support, lipofundin may also have beneficial effects in alleviating the progression of neuroinflammation. These findings may serve as an important reference for future translational medicine applications.
Collapse
Affiliation(s)
- Ming-Shan Chen
- Department of Anesthesiology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan, R.O.C
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung 41354, Taiwan, R.O.C
| | - Chia-Lin Hu
- Department of Anesthesiology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan, R.O.C
| | - Shin-Kuang Jiang
- Department of Neurology, China Medical University Hospital, Taichung 404332, Taiwan, R.O.C
| | - Zhi-Yong Chong
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi 600355, Taiwan, R.O.C
| | - Jui-Chieh Chen
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi 600355, Taiwan, R.O.C
| |
Collapse
|
2
|
Wen J, Satyanarayanan SK, Li A, Yan L, Zhao Z, Yuan Q, Su KP, Su H. Unraveling the impact of Omega-3 polyunsaturated fatty acids on blood-brain barrier (BBB) integrity and glymphatic function. Brain Behav Immun 2024; 115:335-355. [PMID: 37914102 DOI: 10.1016/j.bbi.2023.10.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/05/2023] [Accepted: 10/22/2023] [Indexed: 11/03/2023] Open
Abstract
Alzheimer's disease (AD) and other forms of dementia represent major public health challenges but effective therapeutic options are limited. Pathological brain aging is associated with microvascular changes and impaired clearance systems. The application of omega-3 polyunsaturated fatty acids (n-3 or omega-3 PUFAs) is one of the most promising nutritional interventions in neurodegenerative disorders from epidemiological data, clinical and pre-clinical studies. As essential components of neuronal membranes, n-3 PUFAs have shown neuroprotection and anti-inflammatory effects, as well as modulatory effects through microvascular pathophysiology, amyloid-beta (Aβ) clearance and glymphatic pathways. This review meticulously explores these underlying mechanisms that contribute to the beneficial effects of n-3 PUFAs against AD and dementia, synthesizing evidence from both animal and interventional studies.
Collapse
Affiliation(s)
- Jing Wen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau
| | - Senthil Kumaran Satyanarayanan
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong
| | - Ang Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau
| | - Lingli Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau
| | - Ziai Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau
| | - Qiuju Yuan
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong
| | - Kuan-Pin Su
- An-Nan Hospital, China Medical University, Tainan, Taiwan; Department of Psychiatry, China Medical University Hospital, Taichung, Taiwan; Mind-Body Interface Research Center (MBI-Lab), China Medical University Hospital, Taichung, Taiwan.
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau.
| |
Collapse
|
3
|
Zeng J, Bao T, Yang K, Zhu X, Wang S, Xiang W, Ge A, Zeng L, Ge J. The mechanism of microglia-mediated immune inflammation in ischemic stroke and the role of natural botanical components in regulating microglia: A review. Front Immunol 2022; 13:1047550. [PMID: 36818470 PMCID: PMC9933144 DOI: 10.3389/fimmu.2022.1047550] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 12/05/2022] [Indexed: 02/05/2023] Open
Abstract
Ischemic stroke (IS) is one of the most fatal diseases. Neuroimmunity, inflammation, and oxidative stress play important roles in various complex mechanisms of IS. In particular, the early proinflammatory response resulting from the overactivation of resident microglia and the infiltration of circulating monocytes and macrophages in the brain after cerebral ischemia leads to secondary brain injury. Microglia are innate immune cells in the brain that constantly monitor the brain microenvironment under normal conditions. Once ischemia occurs, microglia are activated to produce dual effects of neurotoxicity and neuroprotection, and the balance of the two effects determines the fate of damaged neurons. The activation of microglia is defined as the classical activation (M1 type) or alternative activation (M2 type). M1 type microglia secrete pro-inflammatory cytokines and neurotoxic mediators to exacerbate neuronal damage, while M2 type microglia promote a repairing anti-inflammatory response. Fine regulation of M1/M2 microglial activation to minimize damage and maximize protection has important therapeutic value. This review focuses on the interaction between M1/M2 microglia and other immune cells involved in the regulation of IS phenotypic characteristics, and the mechanism of natural plant components regulating microglia after IS, providing novel candidate drugs for regulating microglial balance and IS drug development.
Collapse
Affiliation(s)
- Jinsong Zeng
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Tingting Bao
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Kailin Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | | | - Shanshan Wang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Wang Xiang
- Department of Rheumatology, The First People's Hospital Changde City, Changde, Hunan, China
| | - Anqi Ge
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Liuting Zeng
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Jinwen Ge
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China.,Hunan Academy of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
4
|
Role of polyunsaturated fatty acids in ischemic stroke - A perspective of specialized pro-resolving mediators. Clin Nutr 2021; 40:2974-2987. [PMID: 33509668 DOI: 10.1016/j.clnu.2020.12.037] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 12/14/2020] [Accepted: 12/26/2020] [Indexed: 12/17/2022]
Abstract
Polyunsaturated fatty acids (PUFAs) have been proposed as beneficial for cardiovascular health. However, results from both epidemiological studies and clinical trials have been inconsistent, whereas most of the animal studies showed promising benefits of PUFAs in the prevention and treatment of ischemic stroke. In recent years, it has become clear that PUFAs are metabolized into various types of bioactive derivatives, including the specialized pro-resolving mediators (SPMs). SPMs exert multiple biofunctions, such as to limit excessive inflammatory responses, regulate lipid metabolism and immune cell functions, decrease production of pro-inflammatory factors, increase anti-inflammatory mediators, as well as to promote tissue repair and homeostasis. Inflammation has been recognised as a key contributor to the pathophysiology of acute ischemic stroke. Owing to their potent pro-resolving actions, SPMs are potential for development of novel anti-stroke therapy. In this review, we will summarize current knowledge of epidemiological studies, basic research and clinical trials concerning PUFAs in stroke prevention and treatment, with special attention to SPMs as the unsung heroes behind PUFAs.
Collapse
|
5
|
Manual Kollareth DJ, Deckelbaum RJ, Liu Z, Ramakrishnan R, Jouvene C, Serhan CN, Ten VS, Zirpoli H. Acute injection of a DHA triglyceride emulsion after hypoxic-ischemic brain injury in mice increases both DHA and EPA levels in blood and brain ✰. Prostaglandins Leukot Essent Fatty Acids 2020; 162:102176. [PMID: 33038830 PMCID: PMC7685398 DOI: 10.1016/j.plefa.2020.102176] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/21/2020] [Accepted: 09/09/2020] [Indexed: 12/31/2022]
Abstract
We recently reported that acute injection of docosahexaenoic acid (DHA) triglyceride emulsions (tri-DHA) conferred neuroprotection after hypoxic-ischemic (HI) injury in a neonatal mouse stroke model. We showed that exogenous DHA increased concentrations of DHA in brain mitochondria as well as DHA-derived specialized pro-resolving mediator (SPM) levels in the brain. The objective of the present study was to investigate the distribution of emulsion particles and changes in plasma lipid profiles after tri-DHA injection in naïve mice and in animals subjected to HI injury. We also examined whether tri-DHA injection would change DHA- and eicosapentaenoic acid (EPA)-derived SPM levels in the brain. To address this, neonatal (10-day-old) naïve and HI mice were injected with radiolabeled tri-DHA emulsion (0.375 g tri-DHA/kg bw), and blood clearance and tissue distribution were analyzed. Among all the organs assayed, the lowest uptake of emulsion particles was in the brain (<0.4% recovered dose) in both naïve and HI mice, while the liver had the highest uptake. Tri-DHA administration increased DHA concentrations in plasma lysophosphatidylcholine and non-esterified fatty acids. Additionally, treatment with tri-DHA after HI injury significantly elevated the levels of DHA-derived SPMs and monohydroxy-containing DHA-derived products in the brain. Further, tri-DHA administration increased resolvin E2 (RvE2, 5S,18R-dihydroxy-eicosa-6E,8Z,11Z,14Z,16E-pentaenoic acid) and monohydroxy-containing EPA-derived products in the brain. These results suggest that the transfer of DHA through plasma lipid pools plays an important role in DHA brain transport in neonatal mice subjected to HI injury. Furthermore, increases in EPA and EPA-derived SPMs following tri-DHA injection demonstrate interlinked metabolism of these two fatty acids. Hence, changes in both EPA and DHA profile patterns need to be considered when studying the protective effects of DHA after HI brain injury. Our results highlight the need for further investigation to differentiate the effects of DHA from EPA on neuroprotective pathways following HI damage. Such information could contribute to the development of specific DHA-EPA formulations to improve clinical endpoints and modulate potential biomarkers in ischemic brain injury.
Collapse
Affiliation(s)
| | - Richard J Deckelbaum
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY; Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY
| | - Zequn Liu
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY
| | - Rajasekhar Ramakrishnan
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY; Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY
| | - Charlotte Jouvene
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Harvard Institutes of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States of America
| | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Harvard Institutes of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States of America
| | - Vadim S Ten
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY
| | - Hylde Zirpoli
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY.
| |
Collapse
|
6
|
Lipid Emulsion Improves Functional Recovery in an Animal Model of Stroke. Int J Mol Sci 2020; 21:ijms21197373. [PMID: 33036206 PMCID: PMC7582956 DOI: 10.3390/ijms21197373] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/29/2020] [Accepted: 10/03/2020] [Indexed: 12/16/2022] Open
Abstract
Stroke is a life-threatening condition that leads to the death of many people around the world. Reperfusion injury after ischemic stroke is a recurrent problem associated with various surgical procedures that involve the removal of blockages in the brain arteries. Lipid emulsion was recently shown to attenuate ischemic reperfusion injury in the heart and to protect the brain from excitotoxicity. However, investigations on the protective mechanisms of lipid emulsion against ischemia in the brain are still lacking. This study aimed to determine the neuroprotective effects of lipid emulsion in an in vivo rat model of ischemic reperfusion injury through middle cerebral artery occlusion (MCAO). Under sodium pentobarbital anesthesia, rats were subjected to MCAO surgery and were administered with lipid emulsion through intra-arterial injection during reperfusion. The experimental animals were assessed for neurological deficit wherein the brains were extracted at 24 h after reperfusion for triphenyltetrazolium chloride staining, immunoblotting and qPCR. Neuroprotection was found to be dosage-dependent and the rats treated with 20% lipid emulsion had significantly decreased infarction volumes and lower Bederson scores. Phosphorylation of Akt and glycogen synthase kinase 3-β (GSK3-β) were increased in the 20% lipid-emulsion treated group. The Wnt-associated signals showed a marked increase with a concomitant decrease in signals of inflammatory markers in the group treated with 20% lipid emulsion. The protective effects of lipid emulsion and survival-related expression of genes such as Akt, GSK-3β, Wnt1 and β-catenin were reversed by the intra-peritoneal administration of XAV939 through the inhibition of the Wnt/β-catenin signaling pathway. These results suggest that lipid emulsion has neuroprotective effects against ischemic reperfusion injury in the brain through the modulation of the Wnt signaling pathway and may provide potential insights for the development of therapeutic targets.
Collapse
|
7
|
Rasouli B, Ghahari L, Safari M, Shahroozian E, Naeimi S. Combination therapy of the granulocyte colony stimulating factor and intravenous lipid emulsion protect the hippocampus after global ischemia in rat: focusing on CA1 region. Metab Brain Dis 2020; 35:991-997. [PMID: 32458336 DOI: 10.1007/s11011-020-00579-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 05/06/2020] [Indexed: 10/24/2022]
Abstract
Brain stroke is one of the causes of human death and disability worldwide. Global ischemia results in the accumulation of free radicals in the neurons. It leads to histologically brain damage. The CA1 region of the hippocampus is a sensitive area for free radicals. This study investigated the combined therapy of the Granulocyte colony stimulating factor (G-CSF) and the Intravenous lipid emulsion (ILE). These neuroprotective agents play a role in the regeneration of neurons. They improve the learning ability and memory in rats induced global ischemia. We divided 35 rats into five groups. The groups were sham group, ischemia group, G-CSF group, ILE group, and G-CSF plus ILE group. Ischemia was induced by occlusion of the bilateral common carotid about 10 min. The drugs applied on days 1, 3 and 7. The treated groups received subcutaneous injection of 20 μg/kg G-CSF and intravenous injection of 5 ml/kg ILE. After two weeks, the memory and learning ability of the rats was evaluated by the shuttle box. Hematoxylin and Eosin and Nissl and TUNEL stainings were used to determine the necrosis, normal and apoptotic cells. The combined therapy increased normal cells compared to the ischemia group. They decreased the number of necrotic and apoptosis cells in other groups. The combined group improved the passive avoidance test compared to the other groups. The combination therapy of G-CSF plus ILE is more effective than each alone.
Collapse
Affiliation(s)
- Babak Rasouli
- Department of Anatomy, Medical School, AJA University of Medical Sciences, Tehran, Iran
- Department of Basic Sciences, Faculty of Veterinary Medicine, Semnan University, Semnan, Iran
| | - Laya Ghahari
- Department of Anatomy, Medical School, AJA University of Medical Sciences, Tehran, Iran.
| | - Manouchehr Safari
- Research Center of Nervous System Stem Cells, Semnan University of Medical Science, Semnan, Iran
| | - Ebrahim Shahroozian
- Department of Basic Sciences, Faculty of Veterinary Medicine, Semnan University, Semnan, Iran
| | - Saeideh Naeimi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Semnan University, Semnan, Iran
| |
Collapse
|
8
|
Algae Oil Treatment Protects Retinal Ganglion Cells (RGCs) via ERK Signaling Pathway in Experimental Optic Nerve Ischemia. Mar Drugs 2020; 18:md18020083. [PMID: 32012745 PMCID: PMC7074556 DOI: 10.3390/md18020083] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/17/2022] Open
Abstract
Background: We investigated the therapeutic effects and related mechanisms of algae oil (ALG) to protect retinal ganglion cells (RGCs) in a rat model of anterior ischemic optic neuropathy (rAION). Methods: Rats were daily gavaged with ALG after rAION induction for seven days. The therapeutic effects of ALG on rAION were evaluated using flash visual evoked potentials (FVEPs), retrograde labeling of RGCs, TUNEL assay of the retina, and ED1 staining of optic nerves (ONs). The levels of inducible nitric oxide synthase (iNOS), IL-1β, TNF-α, Cl-caspase-3, ciliary neurotrophic factor (CNTF), and p-ERK were analyzed by using western blots. Results: Protection of visual function in FVEPs amplitude was noted, with a better preservation of the P1–N2 amplitude in the ALG-treated group (p = 0.032) than in the rAION group. The density of RGCs was 2.4-fold higher in the ALG-treated group compared to that in the rAION group (p < 0.0001). The number of ED1-positive cells in ONs was significantly reduced 4.1-fold in the ALG-treated group compared to those in the rAION group (p = 0.029). The number of apoptotic RGCs was 3.2-fold lower in number in the ALG-treated group (p = 0.001) than that in the rAION group. The ALG treatment inhibited ERK activation to reduce the levels of iNOS, IL-1β, TNF-α, and Cl-caspase-3 and to increase the level of CNTF in the rAION model. Conclusion: The treatment with ALG after rAION induction inhibits ERK activation to provide both anti-inflammatory and antiapoptotic effects in rAION.
Collapse
|
9
|
Mo Z, Tang C, Li H, Lei J, Zhu L, Kou L, Li H, Luo S, Li C, Chen W, Zhang L. Eicosapentaenoic acid prevents inflammation induced by acute cerebral infarction through inhibition of NLRP3 inflammasome activation. Life Sci 2019; 242:117133. [PMID: 31830477 DOI: 10.1016/j.lfs.2019.117133] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/27/2019] [Accepted: 11/29/2019] [Indexed: 01/20/2023]
Abstract
OBJECTIVE Acute cerebral infarction (ACI) is the most common type of acute cerebrovascular diseases resulting in high rate of death and disability. Numerous evidences show that inflammation is the leading cause of ischemic brain injury, thus anti-inflammatory therapy is an attractive candidate for ischemic brain damage. Eicosapentaenoic acid (EPA) exerts anti-inflammatory activity in lots of human inflammatory diseases, whereas its effect in ACI is left to elucidate. METHOD Nlpr3-/- mice, Gpr40-/-; Gpr120-/- mice and mice with right middle cerebral artery occlusion (MCAO) were used to detect NLR family pyrin domain containing 3 (NLRP3) inflammasome activation by Western Blot and the release of proinflammatory cytokines by ELISA. To estimate the acute ischemic condition in vitro, oxygen-glucose deprivation (OGD) was induced in BV2 microglia cells. Transfection of the shRNA targeting GPR40 and GPR120 mRNA into BV2 cells was also assessed. Apoptosis in ischemic cerebral tissues and BV2 cells was detected by terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick end labeling (TUNEL) assay and flow cytometry. RESULT Here we show that EPA suppresses ACI-induced inflammatory responses through blocking NLRP3 inflammasome activation. In addition, EPA inhibits NLRP3 inflammasome activation through G protein-coupled receptor 40 (GPR40) and GPR120. Importantly, EPA ameliorates ACI-induced apoptosis. CONCLUSION EPA exerts beneficial effect on ACI-induced inflammation through blocking NLRP3 inflammasome activation by GPR40 and GPR120. Our findings suggest the potential clinical use of EPA in ACI.
Collapse
Affiliation(s)
- Zhihuai Mo
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China
| | - Chaogang Tang
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China; Department of Neurology, Maoming People's Hospital, 525000, Guangdong, China
| | - Huiqing Li
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China
| | - Junjie Lei
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China
| | - Lingjuan Zhu
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China
| | - Li Kou
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China
| | - Hao Li
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China; Department of Neurology, Maoming People's Hospital, 525000, Guangdong, China
| | - Shijian Luo
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China
| | - Chunyi Li
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China
| | - Wenli Chen
- Department of Pharmacology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China.
| | - Lei Zhang
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China.
| |
Collapse
|
10
|
Ueno Y, Miyamoto N, Yamashiro K, Tanaka R, Hattori N. Omega-3 Polyunsaturated Fatty Acids and Stroke Burden. Int J Mol Sci 2019; 20:ijms20225549. [PMID: 31703271 PMCID: PMC6888676 DOI: 10.3390/ijms20225549] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 11/01/2019] [Accepted: 11/05/2019] [Indexed: 12/30/2022] Open
Abstract
Stroke is a major leading cause of death and disability worldwide. N-3 polyunsaturated fatty acids (PUFAs) including eicosapentaenoic acid and docosahexaenoic acid have potent anti-inflammatory effects, reduce platelet aggregation, and regress atherosclerotic plaques. Since the discovery that the Greenland Eskimo population, whose diet is high in marine n-3 PUFAs, have a lower incidence of coronary heart disease than Western populations, numerous epidemiological studies to explore the associations of dietary intakes of fish and n-3 PUFAs with cardiovascular diseases, and large-scale clinical trials to identify the benefits of treatment with n-3 PUFAs have been conducted. In most of these studies the incidence and mortality of stroke were also evaluated mainly as secondary endpoints. Thus, a systematic literature review regarding the association of dietary intake of n-3 PUFAs with stroke in the epidemiological studies and the treatment effects of n-3 PUFAs in the clinical trials was conducted. Moreover, recent experimental studies were also reviewed to explore the molecular mechanisms of the neuroprotective effects of n-3 PUFAs after stroke.
Collapse
Affiliation(s)
- Yuji Ueno
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo 113-8421, Japan; (N.M.); (K.Y.); (N.H.)
- Correspondence: ; Tel.: +81-3-3813-3111; Fax: +81-3-5800-0547
| | - Nobukazu Miyamoto
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo 113-8421, Japan; (N.M.); (K.Y.); (N.H.)
| | - Kazuo Yamashiro
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo 113-8421, Japan; (N.M.); (K.Y.); (N.H.)
| | - Ryota Tanaka
- Stroke Center and Division of Neurology, Department of Medicine, Jichi Medical University, Tochigi 329-0498, Japan;
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo 113-8421, Japan; (N.M.); (K.Y.); (N.H.)
| |
Collapse
|
11
|
He J, Huang Y, Du G, Wang Z, Xiang Y, Wang Q. Lasting spatial learning and memory deficits following chronic cerebral hypoperfusion are associated with hippocampal mitochondrial aging in rats. Neuroscience 2019; 415:215-229. [DOI: 10.1016/j.neuroscience.2019.04.044] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 04/18/2019] [Accepted: 04/23/2019] [Indexed: 12/16/2022]
|
12
|
Measuring Respiration in Isolated Murine Brain Mitochondria: Implications for Mechanistic Stroke Studies. Neuromolecular Med 2019; 21:493-504. [PMID: 31172441 DOI: 10.1007/s12017-019-08552-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 05/29/2019] [Indexed: 11/26/2022]
Abstract
Measuring mitochondrial respiration in brain tissue is very critical in understanding the physiology and pathology of the central nervous system. Particularly, measurement of respiration in isolated mitochondria provides the advantage over the whole cells or tissues as the changes in respiratory function are intrinsic to mitochondrial structures rather than the cellular signaling that regulates mitochondria. Moreover, a high-throughput technique for measuring mitochondrial respiration minimizes the experimental time and the sample-to-sample variation. Here, we provide a detailed protocol for measuring respiration in isolated brain non-synaptosomal mitochondria using Agilent Seahorse XFe24 Analyzer. We optimized the protocol for the amount of mitochondria and concentrations of ADP, oligomycin, and trifluoromethoxy carbonylcyanide phenylhydrazone (FCCP) for measuring respiratory parameters for complex I-mediated respiration. In addition, we measured complex II-mediated respiratory parameters. We observed that 10 µg of mitochondrial protein per well, ADP concentrations ranging between 2.5 and 10 mmol/L along with 5 µmol/L of oligomycin, and 5 µmol/L of FCCP are ideal for measuring the complex I-mediated respiration in isolated mouse brain mitochondria. Furthermore, we determined that 2.5 µg of mitochondrial protein per well is ideal for measuring complex II-mediated respiration. Notably, we provide a discussion of logical analysis of data and how the assay could be utilized to design mechanistic studies for experimental stroke. In conclusion, we provide detailed experimental design for measurement of various respiratory parameters in isolated brain mitochondria utilizing a novel high-throughput technique along with interpretation and analysis of data.
Collapse
|
13
|
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: 28] [Impact Index Per Article: 4.7] [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
|
14
|
Vinciguerra A, Cuomo O, Cepparulo P, Anzilotti S, Brancaccio P, Sirabella R, Guida N, Annunziato L, Pignataro G. Models and methods for conditioning the ischemic brain. J Neurosci Methods 2018; 310:63-74. [PMID: 30287283 DOI: 10.1016/j.jneumeth.2018.09.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 09/13/2018] [Accepted: 09/26/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND In the last decades the need to find new neuroprotective targets has addressed the researchers to investigate the endogenous molecular mechanisms that brain activates when exposed to a conditioning stimulus. Indeed, conditioning is an adaptive biological process activated by those interventions able to confer resistance to a deleterious brain event through the exposure to a sub-threshold insult. Specifically, preconditioning and postconditioning are realized when the conditioning stimulus is applied before or after, respectively, the harmul ischemia. AIMS AND RESULTS The present review will describe the most common methods to induce brain conditioning, with particular regards to surgical, physical exercise, temperature-induced and pharmacological approaches. It has been well recognized that when the subliminal stimulus is delivered after the ischemic insult, the achieved neuroprotection is comparable to that observed in models of ischemic preconditioning. In addition, subjecting the brain to both preconditioning as well as postconditioning did not cause greater protection than each treatment alone. CONCLUSIONS The last decades have provided fascinating insights into the mechanisms and potential application of strategies to induce brain conditioning. Since the identification of intrinsic cell-survival pathways should provide more direct opportunities for translational neuroprotection trials, an accurate examination of the different models of preconditioning and postconditioning is mandatory before starting any new project.
Collapse
Affiliation(s)
- Antonio Vinciguerra
- Division of Pharmacology, Department of Neuroscience, School of Medicine, "Federico II" University of Naples, Via Pansini, 5, 80131, Naples, Italy
| | - Ornella Cuomo
- Division of Pharmacology, Department of Neuroscience, School of Medicine, "Federico II" University of Naples, Via Pansini, 5, 80131, Naples, Italy
| | - Pasquale Cepparulo
- Division of Pharmacology, Department of Neuroscience, School of Medicine, "Federico II" University of Naples, Via Pansini, 5, 80131, Naples, Italy
| | | | - Paola Brancaccio
- Division of Pharmacology, Department of Neuroscience, School of Medicine, "Federico II" University of Naples, Via Pansini, 5, 80131, Naples, Italy
| | - Rossana Sirabella
- Division of Pharmacology, Department of Neuroscience, School of Medicine, "Federico II" University of Naples, Via Pansini, 5, 80131, Naples, Italy
| | | | | | - Giuseppe Pignataro
- Division of Pharmacology, Department of Neuroscience, School of Medicine, "Federico II" University of Naples, Via Pansini, 5, 80131, Naples, Italy.
| |
Collapse
|
15
|
Post-stroke DHA Treatment Protects Against Acute Ischemic Brain Injury by Skewing Macrophage Polarity Toward the M2 Phenotype. Transl Stroke Res 2018; 9:669-680. [PMID: 30203370 DOI: 10.1007/s12975-018-0662-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 08/30/2018] [Accepted: 09/03/2018] [Indexed: 12/12/2022]
Abstract
Systemic docosahexaenoic acid (DHA) has been explored as a clinically feasible protectant in stroke models. However, the mechanism for DHA-afforded neuroprotection remains elusive. Transient middle cerebral artery occlusion (tMCAO) was induced for 1 h. DHA (i.p., 10 mg/kg) was administered immediately after reperfusion and repeated daily for 3 days. Stroke outcomes, systemic inflammatory status, and microglia/macrophage phenotypic alterations were assessed 3 days after stroke. Macrophage depletion was induced by clodronate liposomes injection. Primary macrophage cultures were used to evaluate the direct effect of DHA on macrophages. We demonstrated that post-stroke DHA injection efficiently reduced brain infarct and ameliorated neurological deficits 3 days after tMCAO. Systemic DHA treatment significantly inhibited immune cell infiltration (macrophages, neutrophils, T lymphocytes, and B lymphocytes) and promoted macrophage polarization toward an anti-inflammatory M2 phenotype in the ischemic brain. Meanwhile, systemic DHA administration inhibited the otherwise elevated pro-inflammatory factors in blood and shifted circulating macrophage polarity toward M2 phenotype after ischemic stroke. The numbers of circulating immune cells in blood and spleen, however, were equivalent between DHA- and vehicle-treated groups. The protective effects of DHA were macrophage-dependent, as macrophage depletion abolished DHA-afforded neuroprotection. In vitro studies confirmed that DHA suppressed production of chemokines and pro-inflammatory cytokines from macrophages under inflammatory stimulation. These data indicate that post-stroke DHA treatment ameliorated acute ischemic brain injury in a macrophage-dependent manner and DHA enhanced macrophage phenotypic shift toward an anti-inflammatory phenotype to reduced central and peripheral inflammation after stroke.
Collapse
|
16
|
Mohammadi V, Khorvash F, Feizi A, Askari G. Does Alpha-lipoic Acid Supplementation Modulate Cardiovascular Risk Factors in Patients with Stroke? A Randomized, Double-blind Clinical Trial. Int J Prev Med 2018; 9:34. [PMID: 29721235 PMCID: PMC5907425 DOI: 10.4103/ijpvm.ijpvm_32_17] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 05/14/2017] [Indexed: 02/05/2023] Open
Abstract
Background: Stroke as a devastating condition is one of the major causes of death worldwide. It is accountable for long time disability with high personal and social cost in adults. There are several risk factors for stroke such as diabetes and hypertension. Alpha-lipoic acid (ALA) as an antioxidant can be a risk modifier in these patients. We designed this trial to scrutinize the possible effects of ALA consumption on some cardiovascular risk factors in patients experienced stroke. Methods: In this randomized, double-blind, placebo-controlled clinical trial, 67 patients experienced stroke were randomly allocated into two groups (taking a 600 mg ALA supplement or placebo daily for 12 weeks). Fasting blood sugar (FBS), fasting insulin and systolic (SBP), and diastolic blood pressure (DBP) were measured before and after intervention in this study. Statistical analyses were performed using SPSS version 16 (SPSS Inc., Chicago, IL, USA) software. Results: Primary features were similar in the intervention and placebo groups (P > 0.05). After the intervention period, SBP (P < 0.001), DBP (P < 0.001) and FBS (P < 0.001) reduced in ALA group compared with placebo group, significantly. No significant change was seen in insulin level (P = 0.82). Conclusions: Results of this trial indicated that 12 weeks supplementation with 600 mg ALA has beneficial effects on SBP, DBP, and FBS but has no effect on insulin level.
Collapse
Affiliation(s)
- Vida Mohammadi
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fariborz Khorvash
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Awat Feizi
- Department of Epidemiology and Biostatistics, Isfahan Endocrine and Metabolism Research Center, School of Public Health, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Gholamreza Askari
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
17
|
Fourrier C, Remus-Borel J, Greenhalgh AD, Guichardant M, Bernoud-Hubac N, Lagarde M, Joffre C, Layé S. Docosahexaenoic acid-containing choline phospholipid modulates LPS-induced neuroinflammation in vivo and in microglia in vitro. J Neuroinflammation 2017; 14:170. [PMID: 28838312 PMCID: PMC5571638 DOI: 10.1186/s12974-017-0939-x] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 08/09/2017] [Indexed: 12/27/2022] Open
Abstract
Background Neuroinflammatory processes are considered a double-edged sword, having both protective and detrimental effects in the brain. Microglia, the brain’s resident innate immune cells, are a key component of neuroinflammatory response. There is a growing interest in developing drugs to target microglia and control neuroinflammatory processes. In this regard, docosahexaenoic acid (DHA), the brain’s n-3 polyunsaturated fatty acid, is a promising molecule to regulate pro-inflammatory microglia and cytokine production. Several works reported that the bioavailability of DHA to the brain is higher when DHA is acylated to phospholipid. In this work, we analyzed the anti-inflammatory activity of DHA-phospholipid, either acetylated at the sn-1 position (AceDoPC, a stable form thought to have superior access to the brain) or acylated with palmitic acid at the sn-1 position (PC-DHA) using a lipopolysaccharide (LPS)-induced neuroinflammation model both in vitro and in vivo. Methods In vivo, adult C57Bl6/J mice were injected intravenously (i.v.) with either AceDoPC or PC-DHA 24 h prior to LPS (i.p.). For in vitro studies, immortalized murine microglia cells BV-2 were co-incubated with DHA forms and LPS. AceDoPC and PC-DHA effect on brain or BV-2 PUFA content was assessed by gas chromatography. LPS-induced pro-inflammatory cytokines interleukin IL-1β, IL-6, and tumor necrosis factor (TNF) α production were measured by quantitative PCR (qPCR) or multiplex. IL-6 receptors and associated signaling pathway STAT3 were assessed by FACS analysis and western-blot in vitro. Results In vivo, a single injection of AceDoPC or PC-DHA decreased LPS-induced IL-6 production in the hippocampus of mice. This effect could be linked to their direct effect on microglia, as revealed in vitro. In addition, AceDoPC or PC-DHA reduced IL-6 receptor while only AceDoPC decreased IL-6-induced STAT3 phosphorylation. Conclusions These results highlight the potency of administered DHA—acetylated to phospholipids—to rapidly regulate LPS-induced neuroinflammatory processes through their effect on microglia. In particular, both IL-6 production and signaling are targeted by AceDoPC in microglia.
Collapse
Affiliation(s)
- Célia Fourrier
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076, Bordeaux, France.,Bordeaux University, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076, Bordeaux, France
| | - Julie Remus-Borel
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076, Bordeaux, France.,Bordeaux University, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076, Bordeaux, France
| | - Andrew D Greenhalgh
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076, Bordeaux, France.,Bordeaux University, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076, Bordeaux, France
| | - Michel Guichardant
- CarMeN laboratory, INSERM UMR 1060, INRA UMR 1397, IMBL, INSA-Lyon, University of Lyon, Lyon, France
| | - Nathalie Bernoud-Hubac
- CarMeN laboratory, INSERM UMR 1060, INRA UMR 1397, IMBL, INSA-Lyon, University of Lyon, Lyon, France
| | - Michel Lagarde
- CarMeN laboratory, INSERM UMR 1060, INRA UMR 1397, IMBL, INSA-Lyon, University of Lyon, Lyon, France
| | - Corinne Joffre
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076, Bordeaux, France. .,Bordeaux University, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076, Bordeaux, France.
| | - Sophie Layé
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076, Bordeaux, France. .,Bordeaux University, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076, Bordeaux, France.
| |
Collapse
|
18
|
Mohammadi V, Khorvash F, Feizi A, Askari G. The effect of alpha-lipoic acid supplementation on anthropometric indices and food intake in patients who experienced stroke: A randomized, double-blind, placebo-controlled clinical trial. JOURNAL OF RESEARCH IN MEDICAL SCIENCES 2017; 22:98. [PMID: 28900454 PMCID: PMC5583617 DOI: 10.4103/jrms.jrms_1_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 04/26/2017] [Accepted: 05/12/2017] [Indexed: 01/05/2023]
Abstract
Background: Stroke as a devastating condition is a major cause of death worldwide. It is accountable for long-term disability with high personal and social cost in adults. Alpha-lipoic acid (ALA) is an eight-carbon, sulfur-containing compound with antioxidant properties which reduces body weight, changes other anthropometric indices, and regulates food intake by suppressing appetite and increasing metabolism This study was designed to evaluate the possible effects of ALA supplementation on anthropometric indices and dietary intake in patients with stroke. Materials and Methods: In this randomized, double-blind, placebo-controlled clinical trial, 67 patients with stroke were randomly allocated to two groups (taking a 600 mg ALA supplement or placebo daily for 12 weeks). Weight, waist circumference, energy, carbohydrate, protein, and fat intake were measured, and body mass index (BMI) was calculated before and after intervention. Dietary intake and statistical analyses were carried out using Nutritionist IV and SPSS (version 16; SPSS Inc., Chicago, IL, USA) software, respectively. Results: Primary features were similar in the intervention and placebo groups (P > 0.05). Waist circumference (P < 0.001), energy, carbohydrate, protein, and fat intake (P < 0.001) decreased significantly, after the intervention period, in ALA group compared with placebo. While no significant change was observed in weight (P = 0.26) and BMI (P = 0.56) in ALA supplementation group compared with placebo. Conclusion: Results of this trial indicated that 12-week supplementation with 600 mg ALA can decrease waist circumference and food intake (energy, carbohydrate, protein, and fat) in patients with stroke.
Collapse
Affiliation(s)
- Vida Mohammadi
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fariborz Khorvash
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Awat Feizi
- Department of Epidemiology and Biostatistics, School of Public Health, Isfahan Endocrine and Metabolism Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Gholamreza Askari
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
19
|
de Oliveira JN, Reis LO, Ferreira EDF, Godinho J, Bacarin CC, Soares LM, de Oliveira RMW, Milani H. Postischemic fish oil treatment confers task-dependent memory recovery. Physiol Behav 2017; 177:196-207. [DOI: 10.1016/j.physbeh.2017.05.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 04/27/2017] [Accepted: 05/05/2017] [Indexed: 12/20/2022]
|
20
|
Does Alpha-Lipoic Acid Comsumption Improve Lipid Profile In Patients With Stroke? A Randomized, Double Blind, Placebo-Controlled Clinical Trial. IRANIAN RED CRESCENT MEDICAL JOURNAL 2017. [DOI: 10.5812/ircmj.58765] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
21
|
Delhey LM, Nur Kilinc E, Yin L, Slattery JC, Tippett ML, Rose S, Bennuri SC, Kahler SG, Damle S, Legido A, Goldenthal MJ, Frye RE. The Effect of Mitochondrial Supplements on Mitochondrial Activity in Children with Autism Spectrum Disorder. J Clin Med 2017; 6:jcm6020018. [PMID: 28208802 PMCID: PMC5332922 DOI: 10.3390/jcm6020018] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 01/16/2017] [Accepted: 02/06/2017] [Indexed: 02/05/2023] Open
Abstract
Treatment for mitochondrial dysfunction is typically guided by expert opinion with a paucity of empirical evidence of the effect of treatment on mitochondrial activity. We examined citrate synthase and Complex I and IV activities using a validated buccal swab method in 127 children with autism spectrum disorder with and without mitochondrial disease, a portion of which were on common mitochondrial supplements. Mixed-model linear regression determined whether specific supplements altered the absolute mitochondrial activity as well as the relationship between the activities of mitochondrial components. Complex I activity was increased by fatty acid and folate supplementation, but folate only effected those with mitochondrial disease. Citrate synthase activity was increased by antioxidant supplementation but only for the mitochondrial disease subgroup. The relationship between Complex I and IV was modulated by folate while the relationship between Complex I and Citrate Synthase was modulated by both folate and B12. This study provides empirical support for common mitochondrial treatments and demonstrates that the relationship between activities of mitochondrial components might be a marker to follow in addition to absolute activities. Measurements of mitochondrial activity that can be practically repeated over time may be very useful to monitor the biochemical effects of treatments.
Collapse
Affiliation(s)
- Leanna M Delhey
- Arkansas Children's Research Institute, Little Rock, AR 72202, USA.
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA.
| | - Ekim Nur Kilinc
- Arkansas Children's Research Institute, Little Rock, AR 72202, USA.
| | - Li Yin
- Child and Adolescent Department, Mental Health Centre, West China Hospital of Sichuan University, Chengdu 610041, China.
| | - John C Slattery
- Arkansas Children's Research Institute, Little Rock, AR 72202, USA.
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA.
| | - Marie L Tippett
- Arkansas Children's Research Institute, Little Rock, AR 72202, USA.
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA.
| | - Shannon Rose
- Arkansas Children's Research Institute, Little Rock, AR 72202, USA.
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA.
| | - Sirish C Bennuri
- Arkansas Children's Research Institute, Little Rock, AR 72202, USA.
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA.
| | - Stephen G Kahler
- Arkansas Children's Research Institute, Little Rock, AR 72202, USA.
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA.
| | - Shirish Damle
- Department of Pediatrics, Drexel University College of Medicine, Neurology Section, St. Christopher's Hospital for Children, Philadelphia, PA 19134, USA.
| | - Agustin Legido
- Department of Pediatrics, Drexel University College of Medicine, Neurology Section, St. Christopher's Hospital for Children, Philadelphia, PA 19134, USA.
| | - Michael J Goldenthal
- Department of Pediatrics, Drexel University College of Medicine, Neurology Section, St. Christopher's Hospital for Children, Philadelphia, PA 19134, USA.
| | - Richard E Frye
- Arkansas Children's Research Institute, Little Rock, AR 72202, USA.
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA.
| |
Collapse
|