1
|
Gargiulo S, Albanese S, Megna R, Gramanzini M, Marsella G, Vecchiarelli L. Veterinary medical care in rodent models of stroke: Pitfalls and refinements to balance quality of science and animal welfare. Neuroscience 2025; 572:269-302. [PMID: 39894435 DOI: 10.1016/j.neuroscience.2025.01.044] [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: 09/26/2024] [Revised: 10/25/2024] [Accepted: 01/22/2025] [Indexed: 02/04/2025]
Abstract
Rodent models of cerebral ischemia provide a valuable contribution to a better understanding of stroke pathophysiology, to validate diagnostic methods, and to enable testing of new treatments for ischemia-reperfusion damage and comorbidities. However, ethical concerns have led to increased attention to the welfare aspects of such models. Supportive therapies are an essential part of the overall animal care and use program and should be tailored to the experimental model being studied, the regulatory requirements, and research objectives to achieve high-quality preclinical studies and ethical research practices. On the other hand, the use of veterinary medical treatments in preclinical models of stroke must balance the needs of animal care and potential sources of bias in experimental results. This report provides a systematic review of the scientific literature covering the relevant period from years 1988 to September 2024, with the aim to investigating veterinary medical interventions useful to minimize suffering in rodent models of stroke without producing experimental bias. The research findings, consolidated from 181 selected studies, published from 1991 to 2023, indicate the feasibility of implementing personalized protocols of anesthesia, analgesics, antibiotics, and other supportive therapies in rodent models of stroke, while avoiding scientific interferences. These data fill a gap in current knowledge and could be of interest for an interdisciplinary audience working with rodent models of stroke, stimulating further refinements to safeguard both animal welfare and the validity of experimental findings, and may promote the culture of ethical conduct in various research fields and disciplines.
Collapse
Affiliation(s)
- Sara Gargiulo
- Institute of Clinical Physiology, National Research Council, Via Fiorentina 1, 53100 Siena, Italy.
| | - Sandra Albanese
- Institute of Biostructures and Bioimaging, National Research Council, 80131 Naples, Italy.
| | - Rosario Megna
- Institute of Biostructures and Bioimaging, National Research Council, 80131 Naples, Italy.
| | - Matteo Gramanzini
- Institute of Chemical Sciences and Technologies "Giulio Natta", National Research Council, L.go F. Vito, 00168 Rome, Italy.
| | - Gerardo Marsella
- Animal Care Unit, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.
| | - Lidovina Vecchiarelli
- Animal Welfare at Animal and Plant Health Agency, Department for Environment Food and Rural Affairs, Midlands, UK.
| |
Collapse
|
2
|
Liu C, Guo J, Guan L, Li C, Hu X, Jin X, Xu B, Li J, Zhao H. Docosahexaenoic acid protects against ischemic stroke in diabetic mice by inhibiting inflammatory responses and apoptosis. Exp Neurol 2025; 385:115075. [PMID: 39608560 DOI: 10.1016/j.expneurol.2024.115075] [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: 09/19/2024] [Revised: 11/09/2024] [Accepted: 11/24/2024] [Indexed: 11/30/2024]
Abstract
This study was to explore whether docosahexaenoic acid (DHA) protects against ischemic stroke in diabetic mice and its mechanisms. DHA was administered to mice and its effects on stroke outcomes in type 1 diabetes mellitus were assessed 24 h and 3 days post-reperfusion using RNA sequencing, flow cytometry, multiplex immunoassays, and western-blotting analysis. In diabetic mice, DHA administration post-ischemic stroke significantly reduced cerebral infarct size, brain edema, and neurological impairments. Flow cytometric analysis demonstrated a notable decrease in the percentage of neutrophils in the ischemic brain, suggesting a mitigated inflammatory response. Western blotting assay revealed that pro-apoptotic protein Bax was reduced whereas anti-apoptotic protein Bcl-2 was increased, indicating the attenuation of apoptosis. Additionally, RNA sequencing of brain tissue highlighted significant transcriptomic changes, with downregulation of genes for several inflammatory pathways such as NF-kappa B signaling and upregulation of genes for neuroprotective pathways such as neuroactive ligand-receptor interaction. Similar transcriptomic changes in peripheral blood mononuclear cells indicated that DHA treatment resulted the systemic anti-inflammatory and neuroprotective response. DHA treatment mitigated cerebral ischemic injuries by dampening inflammatory responses and apoptosis in diabetic mice after ischemic stroke, highlighting its therapeutic potential for clinical management of stroke in diabetic patients.
Collapse
Affiliation(s)
- Cuiying Liu
- School of Nursing, Capital Medical University, Beijing, China; Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China.
| | - Jiayi Guo
- Department of Neurobiology, School of Basic Medical Science, Capital Medical University, Beijing, China.
| | - Longfei Guan
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Chenyang Li
- School of Nursing, Capital Medical University, Beijing, China.
| | - Xiaoyan Hu
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Xinchun Jin
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
| | - Baohui Xu
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, United States.
| | - Junfa Li
- Department of Neurobiology, School of Basic Medical Science, Capital Medical University, Beijing, China.
| | - Heng Zhao
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China.
| |
Collapse
|
3
|
Kerdiles O, Oye Mintsa Mi-mba MF, Coulombe K, Tremblay C, Émond V, Saint-Pierre M, Rouxel C, Berthiaume L, Julien P, Cicchetti F, Calon F. Additive neurorestorative effects of exercise and docosahexaenoic acid intake in a mouse model of Parkinson's disease. Neural Regen Res 2025; 20:574-586. [PMID: 38819068 PMCID: PMC11317935 DOI: 10.4103/nrr.nrr-d-23-00595] [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: 04/13/2023] [Revised: 11/23/2023] [Accepted: 01/30/2024] [Indexed: 06/01/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202502000-00033/figure1/v/2024-05-28T214302Z/r/image-tiff There is a need to develop interventions to slow or reverse the degeneration of dopamine neurons in Parkinson's disease after diagnosis. Given that preclinical and clinical studies suggest benefits of dietary n-3 polyunsaturated fatty acids, such as docosahexaenoic acid, and exercise in Parkinson's disease, we investigated whether both could synergistically interact to induce recovery of the dopaminergic pathway. First, mice received a unilateral stereotactic injection of 6-hydroxydopamine into the striatum to establish an animal model of nigrostriatal denervation. Four weeks after lesion, animals were fed a docosahexaenoic acid-enriched or a control diet for the next 8 weeks. During this period, the animals had access to a running wheel, which they could use or not. Docosahexaenoic acid treatment, voluntary exercise, or the combination of both had no effect on (i) distance traveled in the open field test, (ii) the percentage of contraversive rotations in the apomorphine-induction test or (iii) the number of tyrosine-hydroxylase-positive cells in the substantia nigra pars compacta. However, the docosahexaenoic acid diet increased the number of tyrosine-hydroxylase-positive terminals and induced a rise in dopamine concentrations in the lesioned striatum. Compared to docosahexaenoic acid treatment or exercise alone, the combination of docosahexaenoic acid and exercise (i) improved forelimb balance in the stepping test, (ii) decreased the striatal DOPAC/dopamine ratio and (iii) led to increased dopamine transporter levels in the lesioned striatum. The present results suggest that the combination of exercise and docosahexaenoic acid may act synergistically in the striatum of mice with a unilateral lesion of the dopaminergic system and provide support for clinical trials combining nutrition and physical exercise in the treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Olivier Kerdiles
- Faculté de pharmacie, Université Laval, 1050 Avenue de la Médecine, Quebec, QC, Canada
- Axe Neuroscience, Centre de recherche du CHU de Québec-Université Laval (Pavillon CHUL), 2705 Boulevard Laurier, Quebec, QC, Canada
- Institute of Nutrition and Functional Foods, Quebec, QC, Canada
- Optinutribrain International Associated Laboratory (NutriNeuro, France; INAF, Canada), Quebec, QC, Canada
| | - Méryl-Farelle Oye Mintsa Mi-mba
- Faculté de pharmacie, Université Laval, 1050 Avenue de la Médecine, Quebec, QC, Canada
- Axe Neuroscience, Centre de recherche du CHU de Québec-Université Laval (Pavillon CHUL), 2705 Boulevard Laurier, Quebec, QC, Canada
- Institute of Nutrition and Functional Foods, Quebec, QC, Canada
- Optinutribrain International Associated Laboratory (NutriNeuro, France; INAF, Canada), Quebec, QC, Canada
| | - Katherine Coulombe
- Axe Neuroscience, Centre de recherche du CHU de Québec-Université Laval (Pavillon CHUL), 2705 Boulevard Laurier, Quebec, QC, Canada
| | - Cyntia Tremblay
- Axe Neuroscience, Centre de recherche du CHU de Québec-Université Laval (Pavillon CHUL), 2705 Boulevard Laurier, Quebec, QC, Canada
- Optinutribrain International Associated Laboratory (NutriNeuro, France; INAF, Canada), Quebec, QC, Canada
| | - Vincent Émond
- Axe Neuroscience, Centre de recherche du CHU de Québec-Université Laval (Pavillon CHUL), 2705 Boulevard Laurier, Quebec, QC, Canada
- Optinutribrain International Associated Laboratory (NutriNeuro, France; INAF, Canada), Quebec, QC, Canada
| | - Martine Saint-Pierre
- Axe Neuroscience, Centre de recherche du CHU de Québec-Université Laval (Pavillon CHUL), 2705 Boulevard Laurier, Quebec, QC, Canada
| | - Clémence Rouxel
- Axe Neuroscience, Centre de recherche du CHU de Québec-Université Laval (Pavillon CHUL), 2705 Boulevard Laurier, Quebec, QC, Canada
- Institute of Nutrition and Functional Foods, Quebec, QC, Canada
| | - Line Berthiaume
- Axe Endocrinologie et Néphrologie, Centre de recherche du CHU de Québec-Université Laval, Quebec, QC, Canada
- Département de Médecine, Faculté de Médecine, Université Laval, Quebec, QC, Canada
| | - Pierre Julien
- Axe Endocrinologie et Néphrologie, Centre de recherche du CHU de Québec-Université Laval, Quebec, QC, Canada
- Département de Médecine, Faculté de Médecine, Université Laval, Quebec, QC, Canada
| | - Francesca Cicchetti
- Axe Neuroscience, Centre de recherche du CHU de Québec-Université Laval (Pavillon CHUL), 2705 Boulevard Laurier, Quebec, QC, Canada
- Département de Psychiatrie et Neurosciences, Faculté de Médecine, Quebec, QC, Canada
| | - Frédéric Calon
- Faculté de pharmacie, Université Laval, 1050 Avenue de la Médecine, Quebec, QC, Canada
- Axe Neuroscience, Centre de recherche du CHU de Québec-Université Laval (Pavillon CHUL), 2705 Boulevard Laurier, Quebec, QC, Canada
- Institute of Nutrition and Functional Foods, Quebec, QC, Canada
- Optinutribrain International Associated Laboratory (NutriNeuro, France; INAF, Canada), Quebec, QC, Canada
| |
Collapse
|
4
|
Oye Mintsa Mi-Mba MF, Lebbadi M, Alata W, Julien C, Emond V, Tremblay C, Fortin S, Barrow CJ, Bilodeau JF, Calon F. Differential impact of eicosapentaenoic acid and docosahexaenoic acid in an animal model of Alzheimer's disease. J Lipid Res 2024; 65:100682. [PMID: 39490923 DOI: 10.1016/j.jlr.2024.100682] [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: 06/24/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024] Open
Abstract
Dietary supplementation with n-3 polyunsaturated fatty acids improves cognitive performance in several animal models of Alzheimer's disease (AD), an effect often associated with reduced amyloid-beta and/or tau pathologies. However, it remains unclear to what extent eicosapentaenoic (EPA) provides additional benefits compared to docosahexaenoic acid (DHA). Here, male and female 3xTg-AD mice were fed for 3 months (13-16 months of age) the following diets: (1) control (no DHA/EPA), (2) DHA (1.1g/kg) and low EPA (0.4g/kg), or (3) DHA (0.9g/kg) with high EPA (9.2g/kg). The DHA and DHA + EPA diets respectively increased DHA by 19% and 8% in the frontal cortex of 3xTg-AD mice, compared to controls. Levels of EPA, which were below the detection limit after the control diet, reached 0.14% and 0.29% of total brain fatty acids after the DHA and DHA + EPA diet, respectively. DHA and DHA + EPA diets lowered brain arachidonic acid levels and the n-6:n-3 docosapentaenoic acid ratio. Brain uptake of free 14C-DHA measured through intracarotid brain perfusion, but not of 14C-EPA, was lower in 3xTg-AD than in NonTg mice. DHA and DHA + EPA diets in 3xTg-AD mice reduced cortical soluble phosphorylated tau (pS202) (-34% high-DHA, -34% DHA + EPA, P < 0.05) while increasing p21-activated kinase (+58% and +83%, P < 0.001; respectively). High EPA intake lowered insoluble phosphorylated tau (-31% vs. DHA, P < 0.05). No diet effect on amyloid-beta levels was observed. In conclusion, dietary intake of DHA and EPA leads to differential changes in brain PUFA while altering cerebral biomarkers consistent with beneficial effects against AD-like neuropathology.
Collapse
Affiliation(s)
- Méryl-Farelle Oye Mintsa Mi-Mba
- Faculty of Pharmacy, Laval University, Quebec, QC, Canada; Centre Hospitalier de l'Université Laval (CHUL) Research Center, Quebec, QC, Canada
| | - Meryem Lebbadi
- Faculty of Pharmacy, Laval University, Quebec, QC, Canada; Centre Hospitalier de l'Université Laval (CHUL) Research Center, Quebec, QC, Canada
| | - Waël Alata
- Faculty of Pharmacy, Laval University, Quebec, QC, Canada; Centre Hospitalier de l'Université Laval (CHUL) Research Center, Quebec, QC, Canada
| | - Carl Julien
- Faculty of Pharmacy, Laval University, Quebec, QC, Canada; Centre Hospitalier de l'Université Laval (CHUL) Research Center, Quebec, QC, Canada
| | - Vincent Emond
- Centre Hospitalier de l'Université Laval (CHUL) Research Center, Quebec, QC, Canada
| | - Cyntia Tremblay
- Centre Hospitalier de l'Université Laval (CHUL) Research Center, Quebec, QC, Canada
| | - Samuel Fortin
- Centre de recherche sur les biotechnologies marines, Rimouski, QC, Canada
| | - Colin J Barrow
- Centre for Sustainable Bioproducts, Deakin University Geelong, Victoria, Australia
| | - Jean-François Bilodeau
- Centre Hospitalier de l'Université Laval (CHUL) Research Center, Quebec, QC, Canada; Department of medicine, Faculty of Medecine, Laval University, Quebec, QC, Canada
| | - Frédéric Calon
- Faculty of Pharmacy, Laval University, Quebec, QC, Canada; Centre Hospitalier de l'Université Laval (CHUL) Research Center, Quebec, QC, Canada.
| |
Collapse
|
5
|
Jin Y, Pu T, Zhang T, Sun Q, Han Y, Han S, Wang G, Yang S, Zhang Y. DHA plays a protective role in cerebral ischemia-reperfusion injury by affecting macrophage/microglia type polarization. Brain Res 2024; 1846:149278. [PMID: 39413982 DOI: 10.1016/j.brainres.2024.149278] [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: 07/12/2024] [Revised: 09/24/2024] [Accepted: 10/11/2024] [Indexed: 10/18/2024]
Abstract
A close correlation exists between the macrophage/microglia(MΦ/MG) polarization states and the development of cerebral ischemia and reperfusion (I/R). Therefore it is of great significance to research on how to modulate the MΦ/MG states for improved patient outcomes. In particular, regulatory mechanisms involved in this process remain to be identified. Hereby, we aim to shed light on how docosahexaenoic acid (DHA) actively modulates the switch between M1 and M2 macrophage states by restraining the NACHT-LRR-PYD-containing protein three inflammasome (NALP3). We found that NALP3-positive cells were detected in clinical human cerebral infarction tissue samples and the mouse tMCAO model. In mice after DHA treatment, the number of NALP3-positive cells was significantly reduced, significantly decreasing infarct volume and improving the postoperative physical status of mice. NALP3-positive cells were found to be MΦ/MG after co-staining with CD11b. By extracting peritoneal macrophages, it was verified that DHA inhibited the activation of NALP3 and regulated the transformation of M1 and M2 cells, thereby reducing I/R injury.
Collapse
Affiliation(s)
- Yimin Jin
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, No. 23 Youzheng Street, Nangang District, Heilongjiang 150001, PR China
| | - Tiantian Pu
- Department of Neurobiology, Harbin Medical University, No. 194 Xuefu Road, Harbin, Heilongjiang 150081, PR China
| | - Tongshuai Zhang
- Department of Neurobiology, Harbin Medical University, No. 194 Xuefu Road, Harbin, Heilongjiang 150081, PR China
| | - Qixu Sun
- Digestive System Department, Yantai Penglai People's Hospital, No. 89 Xianhou Road, Yantai, Shandong 265600, PR China
| | - Yang Han
- Department of Neurobiology, Harbin Medical University, No. 194 Xuefu Road, Harbin, Heilongjiang 150081, PR China; Department of Anesthesiology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, No. 10 Huadong Road, Nanning, Guangxi 530011, PR China
| | - Siyu Han
- Department of Neurobiology, Harbin Medical University, No. 194 Xuefu Road, Harbin, Heilongjiang 150081, PR China
| | - Guangyou Wang
- Department of Neurobiology, Harbin Medical University, No. 194 Xuefu Road, Harbin, Heilongjiang 150081, PR China.
| | - Shanshan Yang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, No. 23 Youzheng Street, Nangang District, Heilongjiang 150001, PR China.
| | - Yao Zhang
- Department of Neurobiology, Harbin Medical University, No. 194 Xuefu Road, Harbin, Heilongjiang 150081, PR China.
| |
Collapse
|
6
|
Poblete RA, Pena J, Kuo G, Tarzi F, Nguyen PL, Cen SY, Yaceczko S, Louie SG, Lewis MR, Martin M, Amar AP, Sanossian N, Sung G, Lyden PD. Immunonutrition with Omega-3 Fatty Acid Supplementation in Severe TBI: Retrospective Analysis of Patient Characteristics and Outcomes. Neurotrauma Rep 2024; 5:574-583. [PMID: 39036427 PMCID: PMC11257118 DOI: 10.1089/neur.2024.0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024] Open
Abstract
Early evidence-based medical interventions to improve patient outcomes after traumatic brain injury (TBI) are lacking. In patients admitted to the ICU after TBI, optimization of nutrition is an emerging field of interest. Specialized enteral nutrition (EN) formulas that include immunonutrition containing omega-3 polyunsaturated fatty acids (n-3 PUFAs) have been developed and are used for their proposed anti-inflammatory and proimmune properties; however, their use has not been rigorously studied in human TBI populations. A single-center, retrospective, descriptive observational study was conducted at the LAC + USC Medical Center. Patients with severe TBI (sTBI, Glasgow Coma Scale score ≤ 8) who remained in the ICU for ≥2 weeks and received EN were identified between 2017 and 2022 using the institutional trauma registry. Those who received immunonutrition formulas containing n-3 PUFAs were compared with those who received standard, polymeric EN with regard to baseline characteristics, clinical markers of inflammation and immune function, and short-term clinical outcomes. A total of 151 patients with sTBI were analyzed. Those who received immunonutrition with n-3 PUFA supplementation were more likely to be male, younger, Hispanic/Latinx, and have polytrauma needing non-central nervous system surgery. No differences in clinical markers of inflammation or infection rate were found. In multivariate regression analysis, immunonutrition was associated with reduced hospital length of stay (LOS). ICU LOS was also reduced in the subgroup of patients with polytrauma and TBI. This study identifies important differences in patient characteristics and outcomes associated with the EN formula prescribed. Study results can directly inform a prospective pragmatic study of immunonutrition with n-3 PUFA supplementation aimed to confirm the biomechanistic and clinical benefits of the intervention.
Collapse
Affiliation(s)
- Roy A. Poblete
- Department of Neurology, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Jesus Pena
- Department of Neurology, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Grace Kuo
- Department of Neurology, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Fawaz Tarzi
- Department of Neurology, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Peggy L. Nguyen
- Department of Neurology, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Steven Y. Cen
- Department of Neurology, University of Southern California Keck School of Medicine, Los Angeles, California, USA
- Department of Radiology, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Shelby Yaceczko
- UCLA Health, University of California, Los Angeles, California, USA
| | - Stan G. Louie
- Department of Clinical Pharmacy, University of Southern California School of Pharmacy, Los Angeles, California, USA
| | - Meghan R. Lewis
- Department of Surgery, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Matthew Martin
- Department of Surgery, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Arun P. Amar
- Department of Neurological Surgery, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Nerses Sanossian
- Department of Neurology, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Gene Sung
- Department of Neurology, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Patrick D. Lyden
- Department of Neurology, University of Southern California Keck School of Medicine, Los Angeles, California, USA
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
7
|
Jensen M, Liu S, Stepula E, Martella D, Birjandi AA, Farrell‐Dillon K, Chan KLA, Parsons M, Chiappini C, Chapple SJ, Mann GE, Vercauteren T, Abbate V, Bergholt MS. Opto-Lipidomics of Tissues. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2302962. [PMID: 38145965 PMCID: PMC11005704 DOI: 10.1002/advs.202302962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 11/30/2023] [Indexed: 12/27/2023]
Abstract
Lipid metabolism and signaling play pivotal functions in biology and disease development. Despite this, currently available optical techniques are limited in their ability to directly visualize the lipidome in tissues. In this study, opto-lipidomics, a new approach to optical molecular tissue imaging is introduced. The capability of vibrational Raman spectroscopy is expanded to identify individual lipids in complex tissue matrices through correlation with desorption electrospray ionization (DESI) - mass spectrometry (MS) imaging in an integrated instrument. A computational pipeline of inter-modality analysis is established to infer lipidomic information from optical vibrational spectra. Opto-lipidomic imaging of transient cerebral ischemia-reperfusion injury in a murine model of ischemic stroke demonstrates the visualization and identification of lipids in disease with high molecular specificity using Raman scattered light. Furthermore, opto-lipidomics in a handheld fiber-optic Raman probe is deployed and demonstrates real-time classification of bulk brain tissues based on specific lipid abundances. Opto-lipidomics opens a host of new opportunities to study lipid biomarkers for diagnostics, prognostics, and novel therapeutic targets.
Collapse
Affiliation(s)
- Magnus Jensen
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUK
| | - Shiyue Liu
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUK
- Institute of Pharmaceutical ScienceKing's College LondonLondonSE1 9NHUK
| | - Elzbieta Stepula
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUK
| | - Davide Martella
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUK
| | - Anahid A. Birjandi
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUK
| | - Keith Farrell‐Dillon
- King's British Heart Foundation Centre of Research ExcellenceSchool of Cardiovascular and Metabolic Medicine & SciencesFaculty of Life Sciences & MedicineKing's College London150 Stamford StreetLondonSE1 9NHUK
| | | | - Maddy Parsons
- Randall Centre for Cell and Molecular BiophysicsKing's College LondonLondonSE1 1ULUK
| | - Ciro Chiappini
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUK
| | - Sarah J. Chapple
- Institute of Pharmaceutical ScienceKing's College LondonLondonSE1 9NHUK
- King's British Heart Foundation Centre of Research ExcellenceSchool of Cardiovascular and Metabolic Medicine & SciencesFaculty of Life Sciences & MedicineKing's College London150 Stamford StreetLondonSE1 9NHUK
| | - Giovanni E. Mann
- Institute of Pharmaceutical ScienceKing's College LondonLondonSE1 9NHUK
- King's British Heart Foundation Centre of Research ExcellenceSchool of Cardiovascular and Metabolic Medicine & SciencesFaculty of Life Sciences & MedicineKing's College London150 Stamford StreetLondonSE1 9NHUK
| | - Tom Vercauteren
- School of Biomedical Engineering and Imaging SciencesKing's College LondonLondonWC2R 2LSUK
| | - Vincenzo Abbate
- Department of AnalyticalEnvironmental and Forensic SciencesKing's College London150 Stamford StreetLondonSE1 9NHUK
| | - Mads S. Bergholt
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUK
| |
Collapse
|
8
|
Rahimian R, Guruswamy R, Boutej H, Cordeau P, Weng YC, Kriz J. Targeting SRSF3 restores immune mRNA translation in microglia/macrophages following cerebral ischemia. Mol Ther 2024; 32:783-799. [PMID: 38196192 PMCID: PMC10928149 DOI: 10.1016/j.ymthe.2024.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 10/20/2023] [Accepted: 01/05/2024] [Indexed: 01/11/2024] Open
Abstract
We recently described a novel ribosome-based regulatory mechanism/checkpoint that controls innate immune gene translation and microglial activation in non-sterile inflammation orchestrated by RNA binding protein SRSF3. Here we describe a role of SRSF3 in the regulation of microglia/macrophage activation phenotypes after experimental stroke. Using a model-system for analysis of the dynamic translational state of microglial ribosomes we show that 24 h after stroke highly upregulated immune mRNAs are not translated resulting in a marked dissociation of mRNA and protein networks in activated microglia/macrophages. Next, microglial activation after stroke was characterized by a robust increase in pSRSF3/SRSF3 expression levels. Targeted knockdown of SRSF3 using intranasal delivery of siRNA 24 h after stroke caused a marked knockdown of endogenous protein. Further analyses revealed that treatment with SRSF3-siRNA alleviated translational arrest of selected genes and induced a transient but significant increase in innate immune signaling and IBA1+ immunoreactivity peaking 5 days after initial injury. Importantly, delayed SRSF3-mediated increase in immune signaling markedly reduced the size of ischemic lesion measured 7 days after stroke. Together, our findings suggest that targeting SRSF3 and immune mRNA translation may open new avenues for molecular/therapeutic reprogramming of innate immune response after ischemic injury.
Collapse
Affiliation(s)
- Reza Rahimian
- CERVO Brain Research Centre and Department of Psychiatry and Neuroscience, Université Laval, Québec, QC G1J 2G3, Canada
| | - Revathy Guruswamy
- CERVO Brain Research Centre and Department of Psychiatry and Neuroscience, Université Laval, Québec, QC G1J 2G3, Canada
| | - Hejer Boutej
- CERVO Brain Research Centre and Department of Psychiatry and Neuroscience, Université Laval, Québec, QC G1J 2G3, Canada
| | - Pierre Cordeau
- CERVO Brain Research Centre and Department of Psychiatry and Neuroscience, Université Laval, Québec, QC G1J 2G3, Canada
| | - Yuan Cheng Weng
- CERVO Brain Research Centre and Department of Psychiatry and Neuroscience, Université Laval, Québec, QC G1J 2G3, Canada
| | - Jasna Kriz
- CERVO Brain Research Centre and Department of Psychiatry and Neuroscience, Université Laval, Québec, QC G1J 2G3, Canada; Faculty of Medicine, Université Laval, Québec, QC G1J 2G3, Canada.
| |
Collapse
|
9
|
Di Martino E, Rayasam A, Vexler ZS. Brain Maturation as a Fundamental Factor in Immune-Neurovascular Interactions in Stroke. Transl Stroke Res 2024; 15:69-86. [PMID: 36705821 PMCID: PMC10796425 DOI: 10.1007/s12975-022-01111-7] [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: 07/13/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 01/28/2023]
Abstract
Injuries in the developing brain cause significant long-term neurological deficits. Emerging clinical and preclinical data have demonstrated that the pathophysiology of neonatal and childhood stroke share similar mechanisms that regulate brain damage, but also have distinct molecular signatures and cellular pathways. The focus of this review is on two different diseases-neonatal and childhood stroke-with emphasis on similarities and distinctions identified thus far in rodent models of these diseases. This includes the susceptibility of distinct cell types to brain injury with particular emphasis on the role of resident and peripheral immune populations in modulating stroke outcome. Furthermore, we discuss some of the most recent and relevant findings in relation to the immune-neurovascular crosstalk and how the influence of inflammatory mediators is dependent on specific brain maturation stages. Finally, we comment on the current state of treatments geared toward inducing neuroprotection and promoting brain repair after injury and highlight that future prophylactic and therapeutic strategies for stroke should be age-specific and consider gender differences in order to achieve optimal translational success.
Collapse
Affiliation(s)
- Elena Di Martino
- Department of Neurology, University California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA
| | - Aditya Rayasam
- Department of Neurology, University California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA
| | - Zinaida S Vexler
- Department of Neurology, University California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA.
| |
Collapse
|
10
|
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
|
11
|
Naeem S, Ali L, Jaffar N, Khan SS, Shafiq Y, Suri S, Tahir A. Shark fish oil prevents scopolamine-induced memory impairment in an experimental model. Metab Brain Dis 2024; 39:15-27. [PMID: 38008885 DOI: 10.1007/s11011-023-01320-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 11/02/2023] [Indexed: 11/28/2023]
Abstract
Fish oil has been known for its antioxidant, cardioprotective, anti-inflammatory, and neuroprotective characteristics due to the presence of polyunsaturated fatty acids (PUFAs) that are essential for optimal brain function and mental health. The present study investigated the effect of Carcharhinus Bleekeri (Shark Fish) oil on learning and memory functions in scopolamine-induced amnesia in rats. Locomotor and memory-enhancing activity in scopolamine-induced amnesic rats was investigated by assessing the open field and passive avoidance paradigm. Forty male Albino mice were divided into 4 equal groups (n = 10) as bellow: 1 - control (received 0.9% saline), 2 - SCOP (received scopolamine 2 mg/kg for 21 days), 3 - SCOP + SFO (received scopolamine and fish oil 5 mg/kg/ day for 21 days), 4 - SCOP + Donepezil groups (received 3 mg/kg/day for 21 days). SFO produced significant (P < 0.01) locomotor and memory-enhancing activities in open-field and passive avoidance paradigm models. Additionally, SFO restored the Acetylcholine (ACh) concentration in the hippocampus (p < 0.05) and remarkably prevented the degradation of monoamines. Histology of brain tissue showed marked cellular distortion in the scopolamine-treated group, while the SFO treatment restored distortion in the brain's hippocampus region. These results suggest that the SFO significantly ameliorates scopolamine-induced spatial memory impairment by attenuating the ACh and monoamine concentrations in the rat's hippocampus.
Collapse
Affiliation(s)
- Sadaf Naeem
- Institute of Pharmaceutical Sciences, Jinnah Sindh Medical University, Karachi, Pakistan.
- Department of Pharmacology, Faculty of Pharmacy, Hamdard University, Karachi, Pakistan.
| | - Liaquat Ali
- Department of Pharmacology, Faculty of Pharmacy, Hamdard University, Karachi, Pakistan
| | - Nazish Jaffar
- Department of Pathology, Sindh Medical College, Jinnah Sindh Medical University, Karachi, Pakistan
| | - Saira Saeed Khan
- Department of Pharmacology, Faculty of Pharmacy, University of Karachi, Karachi, Pakistan
| | - Yousra Shafiq
- Institute of Pharmaceutical Sciences, Jinnah Sindh Medical University, Karachi, Pakistan
| | - Sadia Suri
- Department of Pharmacology, Faculty of Pharmacy, Ziauddin University, Karachi, Pakistan
| | - Anosh Tahir
- Dow Institute of Medical Technology, Dow University of Health Sciences, Karachi, Pakistan
| |
Collapse
|
12
|
Carpinter BA, Renhe DC, Bellei JCB, Vieira CD, Rodolphi CM, Ferreira MVR, de Freitas CS, Neto AFDS, Coelho EAF, Mietto BDS, Gomes FLR, Rocha VN, Scopel KKG. DHA-rich fish oil plays a protective role against experimental cerebral malaria by controlling inflammatory and mechanical events from infection. J Nutr Biochem 2024; 123:109492. [PMID: 37866427 DOI: 10.1016/j.jnutbio.2023.109492] [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: 02/04/2023] [Revised: 05/17/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023]
Abstract
Every year, thousands of children, particularly those under 5 years old, die because of cerebral malaria (CM). Following conventional treatment, approximately 25% of surviving individuals have lifelong severe neurocognitive sequelae. Therefore, improved conventional therapies or effective alternative therapies that prevent the severe infection are crucial. Omega-3 (Ω-3) polyunsaturated fatty acids (PUFAs) are known to have antioxidative and anti-inflammatory effects and protect against diverse neurological disorders, including Alzheimer's and Parkinson's diseases. However, little is known regarding the effects of Ω-3 PUFAs against parasitic infections. In this study, C57BL/6 mice received supplemental treatment of a fish oil rich in the Ω-3 PUFA, docosahexaenoic acid (DHA), which was started 15 days prior to infection with Plasmodium berghei ANKA and was maintained until the end of the study. Animals treated with the highest doses of DHA, 3.0 and 6.0 g/kg body weight, had 60 and 80% chance of survival, respectively, while all nontreated mice died by the 7th day postinfection due to CM. Furthermore, the parasite load during the critical period for CM development (5th to 11th day postinfection) was controlled in treated mice. However, after this period all animals developed high levels of parasitemia until the 20th day of infection. DHA treatment also effectively reduced blood-brain barrier (BBB) damage and brain edema and completely prevented brain hemorrhage and vascular occlusion. A strong anti-inflammatory profile was observed in the brains of DHA-treated mice, as well as, an increased number of neutrophil and reduced number of CD8+ T leukocytes in the spleen. Thus, this is the first study to demonstrate that the prophylactic use of DHA-rich fish oil exerts protective effects against experimental CM, reducing the mechanical and immunological events caused by the P. berghei ANKA infection.
Collapse
Affiliation(s)
- Bárbara Albuquerque Carpinter
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Daniela Chaves Renhe
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Jéssica Correa Bezerra Bellei
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Carolina David Vieira
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Cinthia Magalhães Rodolphi
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | | | - Camila Simões de Freitas
- Post-graduation Program in Health Sciences, Infectology and Tropical Medicine, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Adolfo Firmino da Silva Neto
- Department of Biology, Research Centre of Cellular Biology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Eduardo Antônio Ferraz Coelho
- Post-graduation Program in Health Sciences, Infectology and Tropical Medicine, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Bruno de Siqueira Mietto
- Department of Biology, Research Centre of Cellular Biology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | | | - Vinicius Novaes Rocha
- Department of Veterinary Medicine, Research Centre of Pathology and Veterinary Histology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Kézia Katiani Gorza Scopel
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil.
| |
Collapse
|
13
|
Poblete RA, Pena JE, Kuo G, Tarzi F, Nguyen PL, Cen SY, Yaceczko S, Louie SG, Lewis MR, Martin M, Amar AP, Sanossian N, Sung G, Lyden PD. Immunonutrition with Omega-3 Fatty Acid Supplementation in Severe TBI: Retrospective Analysis of Patient Characteristics and Outcomes. RESEARCH SQUARE 2023:rs.3.rs-3548036. [PMID: 37986931 PMCID: PMC10659558 DOI: 10.21203/rs.3.rs-3548036/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Background Early evidence-based medical interventions to improve patient outcomes after traumatic brain injury (TBI) are lacking. In patients admitted to the ICU after TBI, optimization of nutrition is an emerging field of interest. Specialized enteral nutrition (EN) formulas that include immunonutrition containing omega-3 polyunsaturated fatty acids (n-3 PUFAs) have been developed and are used for their proposed anti-inflammatory and pro-immune properties; however, their use has not been rigorously studied in human TBI populations. Methods A single-center, retrospective, descriptive observational study was conducted at LAC + USC Medical Center. Patients with severe TBI (sTBI, Glasgow Coma Scale score ≤ 8) who remained in the ICU for ≥ 2 weeks and received EN were identified between 2017 and 2022 using the institutional trauma registry. Those who received immunonutrition formulas containing n-3 PUFAs were compared to those who received standard, polymeric EN in regard to baseline characteristics, clinical markers of inflammation and immune function, and short-term clinical outcomes. Results A total of 151 patients with sTBI were analyzed. Those who received immunonutrition with n-3 PUFA supplementation were more likely to be male, younger, Hispanic/Latinx, and have polytrauma needing non-central nervous system surgery. No differences in clinical markers of inflammation or infection rate were found. In multivariate regression analysis, immunonutrition was associated with reduced hospital length of stay (LOS). ICU LOS was also reduced in the subgroup of patients with polytrauma and TBI. Conclusion This study identifies important differences in patient characteristics and outcomes associated with the EN formula prescribed. Study results can directly inform a prospective pragmatic study of immunonutrition with n-3 PUFA supplementation aimed to confirm the biomechanistic and clinical benefits of the intervention.
Collapse
Affiliation(s)
- Roy A Poblete
- University of Southern California Keck School of Medicine
| | - Jesus E Pena
- University of Southern California Keck School of Medicine
| | - Grace Kuo
- University of Southern California Keck School of Medicine
| | - Fawaz Tarzi
- University of Southern California Keck School of Medicine
| | - Peggy L Nguyen
- University of Southern California Keck School of Medicine
| | - Steven Y Cen
- University of Southern California Keck School of Medicine
| | - Shelby Yaceczko
- University of California Los Angeles Health System: UCLA Health
| | - Stan G Louie
- University of Southern California School of Pharmacy
| | - Meghan R Lewis
- University of Southern California Keck School of Medicine
| | - Matthew Martin
- University of Southern California Keck School of Medicine
| | - Arun P Amar
- University of Southern California Keck School of Medicine
| | | | - Gene Sung
- University of Southern California Keck School of Medicine
| | | |
Collapse
|
14
|
Jiang Z, Zeng Z, He H, Li M, Lan Y, Hui J, Bie P, Chen Y, Liu H, Fan H, Xia H. Lycium barbarum glycopeptide alleviates neuroinflammation in spinal cord injury via modulating docosahexaenoic acid to inhibiting MAPKs/NF-kB and pyroptosis pathways. J Transl Med 2023; 21:770. [PMID: 37907930 PMCID: PMC10617163 DOI: 10.1186/s12967-023-04648-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 10/21/2023] [Indexed: 11/02/2023] Open
Abstract
BACKGROUND Lycium barbarum polysaccharide (LBP) is an active ingredient extracted from Lycium barbarum that inhibits neuroinflammation, and Lycium barbarum glycopeptide (LbGp) is a glycoprotein with immunological activity that was purified and isolated from LBP. Previous studies have shown that LbGp can regulate the immune microenvironment, but its specific mechanism of action remains unclear. AIMS In this study, we aimed to explore the mechanism of action of LbGp in the treatment of spinal cord injury through metabolomics and molecular experiments. METHODS SD male rats were randomly assigned to three experimental groups, and after establishing the spinal cord hemisection model, LbGp was administered orally. Spinal cord tissue was sampled on the seventh day after surgery for molecular and metabolomic experiments. In vitro, LbGp was administered to mimic the inflammatory microenvironment by activating microglia, and its mechanism of action in suppressing neuroinflammation was further elaborated using metabolomics and molecular biology techniques such as western blotting and q-PCR. RESULTS In vivo and in vitro experiments found that LbGp can improve the inflammatory microenvironment by inhibiting the NF-kB and pyroptosis pathways. Furthermore, LbGp induced the secretion of docosahexaenoic acid (DHA) by microglia, and DHA inhibited neuroinflammation through the MAPK/NF-κB and pyroptosis pathways. CONCLUSIONS In summary, we hypothesize that LbGp improves the inflammatory microenvironment by regulating the secretion of DHA by microglia and thereby inhibiting the MAPK/NF-κB and pyroptosis pathways and promoting nerve repair and motor function recovery. This study provides a new direction for the treatment of spinal cord injury and elucidates the potential mechanism of action of LbGp.
Collapse
Affiliation(s)
- Zhanfeng Jiang
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, People's Republic of China
- Ningxia Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, China
| | - Zhong Zeng
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, People's Republic of China
- Ningxia Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, China
| | - He He
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, People's Republic of China
- Ningxia Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, China
| | - Mei Li
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, People's Republic of China
- Ningxia Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, China
| | - Yuanxiang Lan
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, People's Republic of China
- Ningxia Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, China
| | - Jianwen Hui
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, People's Republic of China
- Ningxia Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, China
| | - Pengfei Bie
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, People's Republic of China
- Ningxia Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, China
| | - Yanjun Chen
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, People's Republic of China
- Ningxia Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, China
| | - Hao Liu
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, People's Republic of China
| | - Heng Fan
- Ningxia Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, China.
| | - Hechun Xia
- Ningxia Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, China.
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, People's Republic of China.
| |
Collapse
|
15
|
Poblete RA, Yaceczko S, Aliakbar R, Saini P, Hazany S, Breit H, Louie SG, Lyden PD, Partikian A. Optimization of Nutrition after Brain Injury: Mechanistic and Therapeutic Considerations. Biomedicines 2023; 11:2551. [PMID: 37760993 PMCID: PMC10526443 DOI: 10.3390/biomedicines11092551] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Emerging science continues to establish the detrimental effects of malnutrition in acute neurological diseases such as traumatic brain injury, stroke, status epilepticus and anoxic brain injury. The primary pathological pathways responsible for secondary brain injury include neuroinflammation, catabolism, immune suppression and metabolic failure, and these are exacerbated by malnutrition. Given this, there is growing interest in novel nutritional interventions to promote neurological recovery after acute brain injury. In this review, we will describe how malnutrition impacts the biomolecular mechanisms of secondary brain injury in acute neurological disorders, and how nutritional status can be optimized in both pediatric and adult populations. We will further highlight emerging therapeutic approaches, including specialized diets that aim to resolve neuroinflammation, immunodeficiency and metabolic crisis, by providing pre-clinical and clinical evidence that their use promotes neurologic recovery. Using nutrition as a targeted treatment is appealing for several reasons that will be discussed. Given the high mortality and both short- and long-term morbidity associated with acute brain injuries, novel translational and clinical approaches are needed.
Collapse
Affiliation(s)
- Roy A. Poblete
- Department of Neurology, Keck School of Medicine, The University of Southern California, 1540 Alcazar Street, Suite 215, Los Angeles, CA 90033, USA; (R.A.); (P.S.); (H.B.)
| | - Shelby Yaceczko
- UCLA Health, University of California, 100 Medical Plaza, Suite 345, Los Angeles, CA 90024, USA;
| | - Raya Aliakbar
- Department of Neurology, Keck School of Medicine, The University of Southern California, 1540 Alcazar Street, Suite 215, Los Angeles, CA 90033, USA; (R.A.); (P.S.); (H.B.)
| | - Pravesh Saini
- Department of Neurology, Keck School of Medicine, The University of Southern California, 1540 Alcazar Street, Suite 215, Los Angeles, CA 90033, USA; (R.A.); (P.S.); (H.B.)
| | - Saman Hazany
- Department of Radiology, Keck School of Medicine, The University of Southern California, 1500 San Pablo Street, Los Angeles, CA 90033, USA;
| | - Hannah Breit
- Department of Neurology, Keck School of Medicine, The University of Southern California, 1540 Alcazar Street, Suite 215, Los Angeles, CA 90033, USA; (R.A.); (P.S.); (H.B.)
| | - Stan G. Louie
- Department of Clinical Pharmacy, School of Pharmacy, The University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089, USA;
| | - Patrick D. Lyden
- Department of Neurology, Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, The University of Southern California, 1540 Alcazar Street, Suite 215, Los Angeles, CA 90033, USA;
| | - Arthur Partikian
- Department of Neurology, Department of Pediatrics, Keck School of Medicine, The University of Southern California, 2010 Zonal Avenue, Building B, 3P61, Los Angeles, CA 90033, USA;
| |
Collapse
|
16
|
Srakočić S, Gorup D, Kutlić D, Petrović A, Tarabykin V, Gajović S. Reactivation of corticogenesis-related transcriptional factors BCL11B and SATB2 after ischemic lesion of the adult mouse brain. Sci Rep 2023; 13:8539. [PMID: 37237015 DOI: 10.1038/s41598-023-35515-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 05/19/2023] [Indexed: 05/28/2023] Open
Abstract
The aim of this study was to characterize expression of corticogenesis-related transcription factors BCL11B and SATB2 after brain ischemic lesion in the adult mice, and to analyze their correlation to the subsequent brain recovery. Ischemic brain lesion was induced by transient middle cerebral artery occlusion followed by reperfusion, and the animals with ischemic lesion were compared to the sham controls. Progression of the brain damage and subsequent recovery was longitudinally monitored structurally, by magnetic resonance imaging, and functionally, by neurological deficit assessment. Seven days after the ischemic injury the brains were isolated and analyzed by immunohistochemistry. The results showed higher expression in the brain of both, BCL11B and SATB2 in the animals with ischemic lesion compared to the sham controls. The co-expression of both markers, BCL11B and SATB2, increased in the ischemic brains, as well as the co-expression of BCL11B with the beneficial transcriptional factor ATF3 but not its co-expression with detrimental HDAC2. BCL11B was mainly implicated in the ipsilateral and SATB2 in the contralateral brain hemisphere, and their level in these regions correlated with the functional recovery rate. The results indicate that the reactivation of corticogenesis-related transcription factors BCL11B and SATB2 is beneficial after brain ischemic lesion.
Collapse
Affiliation(s)
- Sanja Srakočić
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, 10000, Zagreb, Croatia
| | - Dunja Gorup
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, 10000, Zagreb, Croatia
- Universität Zürich, Universitätspital Zürich, Zürich, Switzerland
| | - Dominik Kutlić
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, 10000, Zagreb, Croatia
| | - Ante Petrović
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, 10000, Zagreb, Croatia
| | - Victor Tarabykin
- Institute of Cell Biology and Neurobiology, Charité-Universitätsmedizin, Berlin, Germany
- Institute of Neuroscience, University of Nizhny Novgorod, Pr. Gagarina 24, Nizhny Novgorod, Russia
| | - Srećko Gajović
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, 10000, Zagreb, Croatia.
| |
Collapse
|
17
|
Unsaturated Fatty Acids and Their Immunomodulatory Properties. BIOLOGY 2023; 12:biology12020279. [PMID: 36829556 PMCID: PMC9953405 DOI: 10.3390/biology12020279] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/07/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023]
Abstract
Oils are an essential part of the human diet and are primarily derived from plant (or sometimes fish) sources. Several of them exhibit anti-inflammatory properties. Specific diets, such as Mediterranean diet, that are high in ω-3 polyunsaturated fatty acids (PUFAs) and ω-9 monounsaturated fatty acids (MUFAs) have even been shown to exert an overall positive impact on human health. One of the most widely used supplements in the developed world is fish oil, which contains high amounts of PUFAs docosahexaenoic and eicosapentaenoic acid. This review is focused on the natural sources of various polyunsaturated and monounsaturated fatty acids in the human diet, and their role as precursor molecules in immune signaling pathways. Consideration is also given to their role in CNS immunity. Recent findings from clinical trials utilizing various fatty acids or diets high in specific fatty acids are reviewed, along with the mechanisms through which fatty acids exert their anti-inflammatory properties. An overall understanding of diversity of polyunsaturated fatty acids and their role in several molecular signaling pathways is useful in formulating diets that reduce inflammation and increase longevity.
Collapse
|
18
|
Hao J, Feng Y, Xu X, Li L, Yang K, Dai G, Gao W, Zhang M, Fan Y, Yin T, Wang J, Yang B, Jiao L, Zhang L. Plasma Lipid Mediators Associate With Clinical Outcome After Successful Endovascular Thrombectomy in Patients With Acute Ischemic Stroke. Front Immunol 2022; 13:917974. [PMID: 35865524 PMCID: PMC9295711 DOI: 10.3389/fimmu.2022.917974] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundNeuroinflammatory response contributes to early neurological deterioration (END) and unfavorable long-term functional outcome in patients with acute ischemic stroke (AIS) who recanalized successfully by endovascular thrombectomy (EVT), but there are no reliable biomarkers for their accurate prediction. Here, we sought to determine the temporal plasma profiles of the bioactive lipid mediators lipoxin A4 (LXA4), resolvin D1 (RvD1), and leukotriene B4 (LTB4) for their associations with clinical outcome.MethodsWe quantified levels of LXA4, RvD1, and LTB4 in blood samples retrospectively and longitudinally collected from consecutive AIS patients who underwent complete angiographic recanalization by EVT at admission (pre-EVT) and 24 hrs post-EVT. The primary outcome was unfavorable long-term functional outcome, defined as a 90-day modified Rankin Scale score of 3-6. Secondary outcome was END, defined as an increase in National Institutes of Health Stroke Scale (NIHSS) score ≥4 points at 24 hrs post-EVT.ResultsEighty-one consecutive AIS patients and 20 healthy subjects were recruited for this study. Plasma levels of LXA4, RvD1, and LTB4 were significantly increased in post-EVT samples from AIS patients, as compared to those of healthy controls. END occurred in 17 (20.99%) patients, and 38 (46.91%) had unfavorable 90-day functional outcome. Multiple logistic regression analyses demonstrated that post-EVT levels of LXA4 (adjusted odd ratio [OR] 0.992, 95% confidence interval [CI] 0.987-0.998), ΔLXA4 (adjusted OR 0.995, 95% CI 0.991-0.999), LTB4 (adjusted OR 1.003, 95% CI 1.001-1.005), ΔLTB4 (adjusted OR 1.004, 95% CI 1.002-1.006), and post-EVT LXA4/LTB4 (adjusted OR 0.023, 95% CI 0.001-0.433) and RvD1/LTB4 (adjusted OR 0.196, 95% CI 0.057-0.682) ratios independently predicted END, and post-EVT LXA4 levels (adjusted OR 0.995, 95% CI 0.992-0.999), ΔLXA4 levels (adjusted OR 0.996, 95% CI 0.993-0.999), and post-EVT LXA4/LTB4 ratio (adjusted OR 0.285, 95% CI 0.096-0.845) independently predicted unfavorable 90-day functional outcome. These were validated using receiver operating characteristic curve analyses.ConclusionsPlasma lipid mediators measured 24 hrs post-EVT were independent predictors for early and long-term outcomes. Further studies are needed to determine their causal-effect relationship, and whether the imbalance between anti-inflammatory/pro-resolving and pro-inflammatory lipid mediators could be a potential adjunct therapeutic target.
Collapse
Affiliation(s)
- Jiheng Hao
- Department of Neurosurgery, Liaocheng People’s hospital, Liaocheng, China
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yao Feng
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Xin Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
- *Correspondence: Xin Xu, ; Liqun Jiao, ; Liyong Zhang,
| | - Long Li
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Kun Yang
- Department of Evidence-based Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Gaolei Dai
- Department of Intervention, Liaocheng People’s hospital, Liaocheng, China
| | - Weiwei Gao
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
| | - Meng Zhang
- Department of Neurosurgery, Liaocheng People’s hospital, Liaocheng, China
| | - Yaming Fan
- Department of Neurosurgery, Liaocheng People’s hospital, Liaocheng, China
| | - Tengkun Yin
- Department of Neurosurgery, Liaocheng People’s hospital, Liaocheng, China
| | - Jiyue Wang
- Department of Neurosurgery, Liaocheng People’s hospital, Liaocheng, China
| | - Bin Yang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Liqun Jiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
- Department of Interventional Neuroradiology, Xuanwu Hospital, Capital Medical Universit, Beijing, China
- *Correspondence: Xin Xu, ; Liqun Jiao, ; Liyong Zhang,
| | - Liyong Zhang
- Department of Neurosurgery, Liaocheng People’s hospital, Liaocheng, China
- *Correspondence: Xin Xu, ; Liqun Jiao, ; Liyong Zhang,
| |
Collapse
|
19
|
Chappus-McCendie H, Poulin MA, Chouinard-Watkins R, Vandal M, Calon F, Lauzon MA, Plourde M. A diet rich in docosahexaenoic acid enhances reactive astrogliosis and ramified microglia morphology in apolipoprotein E epsilon 4-targeted replacement mice. AGING BRAIN 2022; 2:100046. [PMID: 36908881 PMCID: PMC9997137 DOI: 10.1016/j.nbas.2022.100046] [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: 05/02/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 10/16/2022] Open
Abstract
Docosahexaenoic acid (DHA) consumption reduces spatial memory impairment in mice carrying the human apolipoprotein E ε4 (APOE4) allele. The current study evaluated whether astrocyte and microglia morphology contribute to the mechanism of this result. APOE3 and APOE4 mice were fed either a DHA-enriched diet or a control diet from 4 to 12 months of age. Coronal brain sections were immunostained for GFAP, Iba1, and NeuN. Astrocytes from APOE4 mice exhibited signs of reactive astrogliosis compared to APOE3 mice. Consumption of DHA exacerbated reactive astrocyte morphology in APOE4 carriers. Microglia from APOE4-control mice exhibited characteristics of amoeboid morphology and other characteristics of ramified morphology (more processes, greater process complexity, and greater distance between neighboring microglia). DHA enhanced ramified microglia morphology in APOE4 mice. In addition, APOE4 mice fed the DHA diet had lower hippocampal concentrations of interleukin-7, lipopolysaccharide-induced CXC chemokine and monocyte chemoattractant protein 1, and higher concentration of interferon-gamma compared to APOE4-control mice. Our results indicate that a diet rich in DHA enhances reactive astrogliosis and ramified microglia morphology in APOE4 mice.
Collapse
Affiliation(s)
- Hillary Chappus-McCendie
- Centre de Recherche sur le Vieillissement, CIUSSS de l'Estrie-CHUS, Sherbrooke, QC, Canada.,Département de pharmacologie-physiologie, Université de Sherbrooke, Sherbrooke, QC, Canada.,Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Marc-Antoine Poulin
- Département de génie chimique et de génie biotechnologique, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Raphaël Chouinard-Watkins
- Centre de Recherche sur le Vieillissement, CIUSSS de l'Estrie-CHUS, Sherbrooke, QC, Canada.,Département de pharmacologie-physiologie, Université de Sherbrooke, Sherbrooke, QC, Canada.,Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de la nutrition et des aliments fonctionnels, Université Laval, Québec, QC, Canada
| | - Milène Vandal
- Institut de la nutrition et des aliments fonctionnels, Université Laval, Québec, QC, Canada.,Faculté de pharmacie et centre de recherche du CHU de Québec-Université Laval, QC, Canada
| | - Frédéric Calon
- Institut de la nutrition et des aliments fonctionnels, Université Laval, Québec, QC, Canada.,Faculté de pharmacie et centre de recherche du CHU de Québec-Université Laval, QC, Canada
| | - Marc-Antoine Lauzon
- Centre de Recherche sur le Vieillissement, CIUSSS de l'Estrie-CHUS, Sherbrooke, QC, Canada.,Département de génie chimique et de génie biotechnologique, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Mélanie Plourde
- Centre de Recherche sur le Vieillissement, CIUSSS de l'Estrie-CHUS, Sherbrooke, QC, Canada.,Département de pharmacologie-physiologie, Université de Sherbrooke, Sherbrooke, QC, Canada.,Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de la nutrition et des aliments fonctionnels, Université Laval, Québec, QC, Canada.,Département de Médecine, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
20
|
The effect of omega-3 polyunsaturated fatty acids on stroke treatment and prevention: a systematic review and meta-analysis. NUTR HOSP 2022; 39:924-935. [DOI: 10.20960/nh.04148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
21
|
Lamontagne-Proulx J, Coulombe K, Dahhani F, Côté M, Guyaz C, Tremblay C, Di Marzo V, Flamand N, Calon F, Soulet D. Effect of Docosahexaenoic Acid (DHA) at the Enteric Level in a Synucleinopathy Mouse Model. Nutrients 2021; 13:nu13124218. [PMID: 34959768 PMCID: PMC8703327 DOI: 10.3390/nu13124218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/18/2021] [Accepted: 11/22/2021] [Indexed: 12/15/2022] Open
Abstract
The aggregation of alpha-synuclein protein (αSyn) is a hallmark of Parkinson’s disease (PD). Considerable evidence suggests that PD involves an early aggregation of αSyn in the enteric nervous system (ENS), spreading to the brain. While it has previously been reported that omega-3 polyunsaturated fatty acids (ω-3 PUFA) acts as neuroprotective agents in the brain in murine models of PD, their effect in the ENS remains undefined. Here, we studied the effect of dietary supplementation with docosahexaenoic acid (DHA, an ω-3 PUFA), on the ENS, with a particular focus on enteric dopaminergic (DAergic) neurons. Thy1-αSyn mice, which overexpress human αSyn, were fed ad libitum with a control diet, a low ω-3 PUFA diet or a diet supplemented with microencapsulated DHA and then compared with wild-type littermates. Our data indicate that Thy1-αSyn mice showed a lower density of enteric dopaminergic neurons compared with non-transgenic animals. This decrease was prevented by dietary DHA. Although we found that DHA reduced microgliosis in the striatum, we did not observe any evidence of peripheral inflammation. However, we showed that dietary intake of DHA promoted a build-up of ω-3 PUFA-derived endocannabinoid (eCB)-like mediators in plasma and an increase in glucagon-like peptide-1 (GLP-1) and the redox regulator, Nrf2 in the ENS. Taken together, our results suggest that DHA exerts neuroprotection of enteric DAergic neurons in the Thy1-αSyn mice, possibly through alterations in eCB-like mediators, GLP-1 and Nrf2.
Collapse
Affiliation(s)
- Jérôme Lamontagne-Proulx
- Centre de Recherche du CHU de Québec, Québec, QC G1V 4G2, Canada; (J.L.-P.); (K.C.); (M.C.); (C.T.); (F.C.)
- Faculté de Pharmacie, Université Laval, Québec, QC G1V 0A6, Canada;
| | - Katherine Coulombe
- Centre de Recherche du CHU de Québec, Québec, QC G1V 4G2, Canada; (J.L.-P.); (K.C.); (M.C.); (C.T.); (F.C.)
| | - Fadil Dahhani
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Québec, QC G1V 4G5, Canada; (F.D.); (V.D.); (N.F.)
- Canada Excellence Research in the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Québec, QC G1V 4G5, Canada
| | - Mélissa Côté
- Centre de Recherche du CHU de Québec, Québec, QC G1V 4G2, Canada; (J.L.-P.); (K.C.); (M.C.); (C.T.); (F.C.)
| | - Cédric Guyaz
- Faculté de Pharmacie, Université Laval, Québec, QC G1V 0A6, Canada;
| | - Cyntia Tremblay
- Centre de Recherche du CHU de Québec, Québec, QC G1V 4G2, Canada; (J.L.-P.); (K.C.); (M.C.); (C.T.); (F.C.)
- Faculté de Pharmacie, Université Laval, Québec, QC G1V 0A6, Canada;
| | - Vincenzo Di Marzo
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Québec, QC G1V 4G5, Canada; (F.D.); (V.D.); (N.F.)
- Canada Excellence Research in the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Québec, QC G1V 4G5, Canada
- Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels (INAF) et Centre NUTRISS, Université Laval, Québec, QC G1V 0A6, Canada
| | - Nicolas Flamand
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Québec, QC G1V 4G5, Canada; (F.D.); (V.D.); (N.F.)
- Canada Excellence Research in the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Québec, QC G1V 4G5, Canada
- Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Frédéric Calon
- Centre de Recherche du CHU de Québec, Québec, QC G1V 4G2, Canada; (J.L.-P.); (K.C.); (M.C.); (C.T.); (F.C.)
- Faculté de Pharmacie, Université Laval, Québec, QC G1V 0A6, Canada;
- Laboratoire International Associé OptiNutriBrain, (NutriNeuro France-INAF Canada), Québec, QC G1V 0A6, Canada
| | - Denis Soulet
- Centre de Recherche du CHU de Québec, Québec, QC G1V 4G2, Canada; (J.L.-P.); (K.C.); (M.C.); (C.T.); (F.C.)
- Faculté de Pharmacie, Université Laval, Québec, QC G1V 0A6, Canada;
- Institut sur la Nutrition et les Aliments Fonctionnels (INAF) et Centre NUTRISS, Université Laval, Québec, QC G1V 0A6, Canada
- Correspondence: ; Tel.: +1-418-654-2296
| |
Collapse
|
22
|
Koçancı FG. Role of Fatty Acid Chemical Structures on Underlying Mechanisms of Neurodegenerative Diseases and Gut Microbiota. EUR J LIPID SCI TECH 2021. [DOI: 10.1002/ejlt.202000341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Fatma Gonca Koçancı
- Vocational High School of Health Services Department of Medical Laboratory Techniques Alanya Alaaddin Keykubat University Alanya/Antalya 07425 Turkey
| |
Collapse
|
23
|
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
|
24
|
Rist PM, Buring JE, Cook NR, Manson JE, Rexrode KM. Effect of vitamin D and/or omega-3 fatty acid supplementation on stroke outcomes: A randomized trial. Eur J Neurol 2020; 28:809-815. [PMID: 33131164 DOI: 10.1111/ene.14623] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 10/24/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND PURPOSE Among stroke patients, low serum 25-hydroxyvitamin D predicts poor outcomes. In mice, higher omega-3 (n-3) fatty acid intake diminishes brain damage after stroke. In this study, we tested whether vitamin D or n-3 fatty acids supplementation prior to stroke reduces the risk of functional limitations and physical disability after stroke. METHODS We used data from VITAL (the VITamin D and OmegA-3 TriaL) which randomized middle-aged and older men and women without cardiovascular disease to vitamin D3 (2000 IU/day) and/or marine n-3 fatty acids (1 g/day) and followed them for incident stroke events. Individuals experiencing a non-fatal stroke were mailed questionnaires assessing functional limitations (the physical performance scale adapted from Nagi) and physical disability (the modified Katz Activities of Daily Living and Rosow-Breslau Functional Health scales). We used logistic regression to analyze associations between randomized treatment and limitations on each scale. RESULTS A total of 290 individuals experienced their first stroke during the trial, of whom 197 stroke survivors completed the stroke outcomes questionnaire a median of 1.4 years after diagnosis. We observed no associations between randomized treatment to vitamin D and functional limitations (odds ratio [OR] 1.01, 95% confidence interval [CI] 0.52, 1.97) or physical disability (Rosow-Breslau scale: OR 0.92, 95% CI 0.50, 1.67; Katz scale: OR 1.03, 95% CI 0.31, 3.42). Those randomized to n-3 fatty acids had a non-significantly lower risk of functional limitations (OR 0.55, 95% CI 0.28, 1.09) and physical disability (Rosow-Breslau scale: OR 0.56, 95% CI 0.31, 1.02; Katz sclae: OR 0.32, 95% CI 0.50, 1.67). CONCLUSION Vitamin D or omega-3 fatty acid supplementation prior to stroke did not result in significantly improved post-stroke outcomes.
Collapse
Affiliation(s)
- Pamela M Rist
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Julie E Buring
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Nancy R Cook
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - JoAnn E Manson
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Kathryn M Rexrode
- Division of Women's Health, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
25
|
Zirpoli H, Chang CL, Carpentier YA, Michael-Titus AT, Ten VS, Deckelbaum RJ. Novel Approaches for Omega-3 Fatty Acid Therapeutics: Chronic Versus Acute Administration to Protect Heart, Brain, and Spinal Cord. Annu Rev Nutr 2020; 40:161-187. [PMID: 32966188 DOI: 10.1146/annurev-nutr-082018-124539] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This article reviews novel approaches for omega-3 fatty acid (FA) therapeutics and the linked molecular mechanisms in cardiovascular and central nervous system (CNS) diseases. In vitro and in vivo research studies indicate that omega-3 FAs affect synergic mechanisms that include modulation of cell membrane fluidity, regulation of intracellular signaling pathways, and production of bioactive mediators. We compare how chronic and acute treatments with omega-3 FAs differentially trigger pathways of protection in heart, brain, and spinal cord injuries. We also summarize recent omega-3 FA randomized clinical trials and meta-analyses and discuss possible reasons for controversial results, with suggestions on improving the study design for future clinical trials. Acute treatment with omega-3 FAs offers a novel approach for preserving cardiac and neurological functions, and the combinations of acute treatment with chronic administration of omega-3 FAs might represent an additional therapeutic strategy for ameliorating adverse cardiovascular and CNS outcomes.
Collapse
Affiliation(s)
- Hylde Zirpoli
- Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA;
| | - Chuchun L Chang
- Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA;
| | - Yvon A Carpentier
- Clinical Nutrition Unit, Université Libre de Bruxelles, 1050 Brussels, Belgium.,Nutrition Lipid Developments, SPRL, 1050 Brussels, Belgium
| | - Adina T Michael-Titus
- Center for Neuroscience, Surgery, and Trauma, Queen Mary University of London, London E1 4NS, United Kingdom
| | - Vadim S Ten
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Richard J Deckelbaum
- Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA; .,Department of Pediatrics, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
26
|
Reyes LD, Haight T, Desai A, Chen H, Bosomtwi A, Korotcov A, Dardzinski B, Kim HY, Pierpaoli C. Investigation of the effect of dietary intake of omega-3 polyunsaturated fatty acids on trauma-induced white matter injury with quantitative diffusion MRI in mice. J Neurosci Res 2020; 98:2232-2244. [PMID: 32840025 PMCID: PMC7589213 DOI: 10.1002/jnr.24705] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 07/09/2020] [Accepted: 07/11/2020] [Indexed: 01/26/2023]
Abstract
Previous studies suggest that long-term supplementation and dietary intake of omega-3 polyunsaturated fatty acids (PUFAs) may have neuroprotective effects following brain injury. The objective of this study was to investigate potential neuroprotective effects of omega-3 PUFAs on white matter following closed-head trauma. The closed-head injury model of engineered rotational acceleration (CHIMERA) produces a reproducible injury in the optic tract and brachium of the superior colliculus in mice. Damage is detectable using diffusion tensor imaging (DTI) metrics, particularly fractional anisotropy (FA), with sensitivity comparable to histology. We acquired in vivo (n = 38) and ex vivo (n = 41) DTI data in mice divided into sham and CHIMERA groups with two dietary groups: one deficient in omega-3 PUFAs and one adequate in omega-3 PUFAs. We examined injury effects (reduction in FA) and neuroprotection (FA reduction modulated by diet) in the optic tract and brachium. We verified that diet did not affect FA in sham animals. In injured animals, we found significantly reduced FA in the optic tract and brachium (~10% reduction, p < 0.001), and Bayes factor analysis showed strong evidence to reject the null hypothesis. However, Bayes factor analysis showed substantial evidence to accept the null hypothesis of no diet-related FA differences in injured animals in the in vivo and ex vivo samples. Our results indicate no neuroprotective effect from adequate dietary omega-3 PUFA intake on white matter damage following traumatic brain injury. Since damage from CHIMERA mainly affects white matter, our results do not necessarily contradict previous findings showing omega-3 PUFA-mediated neuroprotection in gray matter.
Collapse
Affiliation(s)
- Laura D Reyes
- Quantitative Medical Imaging Section, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Thaddeus Haight
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA.,Center for Neuroscience and Regenerative Medicine, Uniformed Services University, Bethesda, MD, USA
| | - Abhishek Desai
- Laboratory of Molecular Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Huazhen Chen
- Laboratory of Molecular Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Asamoah Bosomtwi
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA.,Georgia Cancer Center, Augusta University, Augusta, GA, USA.,Department of Radiology and Radiological Sciences, Uniformed Services University, Bethesda, MD, USA
| | - Alexandru Korotcov
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA.,Department of Radiology and Radiological Sciences, Uniformed Services University, Bethesda, MD, USA
| | - Bernard Dardzinski
- Department of Radiology and Radiological Sciences, Uniformed Services University, Bethesda, MD, USA
| | - Hee-Yong Kim
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University, Bethesda, MD, USA
| | - Carlo Pierpaoli
- Quantitative Medical Imaging Section, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
27
|
Zhang W, Zhu L, An C, Wang R, Yang L, Yu W, Li P, Gao Y. The blood brain barrier in cerebral ischemic injury – Disruption and repair. BRAIN HEMORRHAGES 2020. [DOI: 10.1016/j.hest.2019.12.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
28
|
Rahimian R, Lalancette-Hébert M, Weng YC, Sato S, Kriz J. Glucosamine-mediated immunomodulation after stroke is sexually dimorphic. Brain Behav Immun Health 2020; 3:100041. [PMID: 34589834 PMCID: PMC8474641 DOI: 10.1016/j.bbih.2020.100041] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/22/2020] [Accepted: 01/24/2020] [Indexed: 12/30/2022] Open
Abstract
Growing evidence suggests that galectin-3 (Gal-3) is instrumental in orchestrating innate immune response and microglia activation following different brain pathologies. However, its role remains controversial. We recently showed that a readily available natural product glucosamine may act as a strong modulator of Gal-3. Glucosamine is a naturally occurring sugar and a precursor in the synthesis of glycosylated proteins. It is often used as a supplement to treat symptoms of various inflammatory conditions. Our recent work suggests that by increasing the synthesis and availability of Gal-3 ligands and/or by regulating its expression levels, glucosamine may significantly modulate Gal-3 signaling. Because evidence suggests that Gal-3 might be differentially regulated after ischemic injury in the brains of female mice, here we examined and compared the immunomodulatory potential of glucosamine in male and female stroke. The mice were subjected to transient middle cerebral artery occlusion (MCAO), followed by different reperfusion periods. The short-term 5 days treatment with glucosamine (150 mg/kg i.p.) was initiated 2 hrs after stroke. To visualize the effects of glucosamine treatment on post-stroke inflammation, we took advantage of a transgenic mouse model bearing the dual reporter system luciferase/GFP under transcriptional control of a murine TLR2 promoter (TLR2-luc-GFP) allowing in vivo bioluminescence imaging of innate immune response and microglial activation. We report that after stroke, both, male and female mice strongly up-regulate the TLR2 bioluminescence signals from activated microglia, however, the observed in vivo immunomodulatory effects of glucosamine after stroke were sex-dependent. Analysis of cytokine profiles at protein level, in glucosamine-treated male mice 72hsr after stroke, revealed down regulation of pro-inflammatory cytokines, an increase in levels of anti-inflammatory cytokines including IL-4, IL13 and colony stimulating factors MCFC and GM-CSF and a significant decrease in the size of ischemic lesion in male mice. Conversely, in female mice glucosamine markedly increases the pro-inflammatory signaling and exacerbates ischemic injury. Analysis of the downstream signaling target of glucosamine/Gal-3 revealed that glucosamine administration restored PPAR-γ activity in male but not in female mice 3 days following MCAO. Together, our results suggest that glucosamine acts as a fine tuner of post-ischemic inflammation in a sex dependent-manner and may have therapeutic potential after stroke in males. Based on our results propose that targeting immune system after stroke may require adapted sex-specific therapeutic approaches. Immunomodulatory effects of glucosamine are sex dependent. Glucosamine differentially modulates galectin-3/IL4R signaling in male and female mice. Glucosamine restores PPAR-gamma transcriptional activity in male mice and protects against stroke in male mice. Glucosamine increases inflammatory signaling and exacerbates ischemic injury in female mice.
Collapse
Affiliation(s)
- Reza Rahimian
- CERVO Brain Research Centre and Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, QC, G1J 2G3, Canada
| | - Melanie Lalancette-Hébert
- CERVO Brain Research Centre and Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, QC, G1J 2G3, Canada
| | - Yuan Cheng Weng
- CERVO Brain Research Centre and Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, QC, G1J 2G3, Canada
| | - Sachiko Sato
- Glycobiology Laboratory, Research Centre for Infectious Disease, Université Laval, QC, G1V 4G2, Canada
| | - Jasna Kriz
- CERVO Brain Research Centre and Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, QC, G1J 2G3, Canada
| |
Collapse
|
29
|
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: 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: 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
|
30
|
Pinçon A, De Montgolfier O, Akkoyunlu N, Daneault C, Pouliot P, Villeneuve L, Lesage F, Levy BI, Thorin-Trescases N, Thorin É, Ruiz M. Non-Alcoholic Fatty Liver Disease, and the Underlying Altered Fatty Acid Metabolism, Reveals Brain Hypoperfusion and Contributes to the Cognitive Decline in APP/PS1 Mice. Metabolites 2019; 9:metabo9050104. [PMID: 31130652 PMCID: PMC6572466 DOI: 10.3390/metabo9050104] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/16/2019] [Accepted: 05/21/2019] [Indexed: 12/22/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), the leading cause of chronic liver disease, is associated with cognitive decline in middle-aged adults, but the mechanisms underlying this association are not clear. We hypothesized that NAFLD would unveil the appearance of brain hypoperfusion in association with altered plasma and brain lipid metabolism. To test our hypothesis, amyloid precursor protein/presenilin-1 (APP/PS1) transgenic mice were fed a standard diet or a high-fat, cholesterol and cholate diet, inducing NAFLD without obesity and hyperglycemia. The diet-induced NAFLD disturbed monounsaturated and polyunsaturated fatty acid (MUFAs, PUFAs) metabolism in the plasma, liver, and brain, and particularly reduced n-3 PUFAs levels. These alterations in lipid homeostasis were associated in the brain with an increased expression of Tnfα, Cox2, p21, and Nox2, reminiscent of brain inflammation, senescence, and oxidative stress. In addition, compared to wild-type (WT) mice, while brain perfusion was similar in APP/PS1 mice fed with a chow diet, NAFLD in APP/PS1 mice reveals cerebral hypoperfusion and furthered cognitive decline. NAFLD reduced plasma β40- and β42-amyloid levels and altered hepatic but not brain expression of genes involved in β-amyloid peptide production and clearance. Altogether, our results suggest that in a mouse model of Alzheimer disease (AD) diet-induced NAFLD contributes to the development and progression of brain abnormalities through unbalanced brain MUFAs and PUFAs metabolism and cerebral hypoperfusion, irrespective of brain amyloid pathology that may ultimately contribute to the pathogenesis of AD.
Collapse
Affiliation(s)
- Anthony Pinçon
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada.
- Research Center, Montreal Heart Institute, University of Montreal, Montreal, QC H1T 1C8, Canada.
| | - Olivia De Montgolfier
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada.
- Research Center, Montreal Heart Institute, University of Montreal, Montreal, QC H1T 1C8, Canada.
| | - Nilay Akkoyunlu
- Research Center, Montreal Heart Institute, University of Montreal, Montreal, QC H1T 1C8, Canada.
| | - Caroline Daneault
- Research Center, Montreal Heart Institute, University of Montreal, Montreal, QC H1T 1C8, Canada.
| | - Philippe Pouliot
- Research Center, Montreal Heart Institute, University of Montreal, Montreal, QC H1T 1C8, Canada.
- Department of Electrical Engineering, Ecole Polytechnique de Montréal, Montreal, QC H3T 1J4, Canada.
| | - Louis Villeneuve
- Research Center, Montreal Heart Institute, University of Montreal, Montreal, QC H1T 1C8, Canada.
| | - Frédéric Lesage
- Research Center, Montreal Heart Institute, University of Montreal, Montreal, QC H1T 1C8, Canada.
- Department of Electrical Engineering, Ecole Polytechnique de Montréal, Montreal, QC H3T 1J4, Canada.
| | - Bernard I Levy
- Institut des Vaisseaux et du Sang, Hôpital Lariboisière, 75010 Paris, France.
| | | | - Éric Thorin
- Research Center, Montreal Heart Institute, University of Montreal, Montreal, QC H1T 1C8, Canada.
- Department of Surgery, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada.
| | - Matthieu Ruiz
- Research Center, Montreal Heart Institute, University of Montreal, Montreal, QC H1T 1C8, Canada.
- Department of Medecine, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada.
| |
Collapse
|
31
|
Delayed Galectin-3-Mediated Reprogramming of Microglia After Stroke is Protective. Mol Neurobiol 2019; 56:6371-6385. [PMID: 30798442 DOI: 10.1007/s12035-019-1527-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 02/13/2019] [Indexed: 01/06/2023]
Abstract
Galectin-3 (Gal-3), a β-galactoside-binding lectin, has recently emerged as a molecule with immunoregulatory functions. We investigated the effects of Gal-3 on microglia morphology, migration, and secretory profile under physiological conditions and in the context of ischemic injury. We show that in the control conditions, exposure to recombinant Gal-3 increases microglial ramification and motility in vitro and in vivo via an IL-4-dependent mechanism. Importantly, after stroke, Gal-3 exerted marked immune-modulatory properties. Delivery of Gal-3 at 24 h after middle cerebral artery occlusion (MCAO) was associated with an increase in Ym1-positive microglia and decrease in iNOS. Analysis of cytokine profiles at the protein level revealed downregulation of pro-inflammatory cytokines and a marked upregulation of the anti-inflammatory cytokine, IL-4, 24 h after i.c.v. injection of Gal-3. Importantly, the observed shift in cytokines in microglia was associated with a significant decrease in the infarct size. Taken together, our results suggest that when delivered well after ischemic injury, Gal-3 might fine tune innate immunity and induce a therapeutic shift in microglia polarization.
Collapse
|
32
|
Orchard TS, Gaudier-Diaz MM, Phuwamongkolwiwat-Chu P, Andridge R, Lustberg MB, Bomser J, Cole RM, Belury MA, DeVries AC. Low Sucrose, Omega-3 Enriched Diet Has Region-Specific Effects on Neuroinflammation and Synaptic Function Markers in a Mouse Model of Doxorubicin-Based Chemotherapy. Nutrients 2018; 10:E2004. [PMID: 30567351 PMCID: PMC6316589 DOI: 10.3390/nu10122004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 11/21/2018] [Accepted: 12/13/2018] [Indexed: 12/21/2022] Open
Abstract
Chemotherapeutic agents such as doxorubicin may negatively affect long-term brain functioning in cancer survivors; neuroinflammation may play a causal role. Dietary approaches that reduce inflammation, such as lowering sucrose and increasing eicosapentaenoic acid plus docosahexaenoic acid (EPA + DHA), may attenuate chemotherapy-induced neuroinflammation and synaptic damage, thereby improving quality of life. Ovariectomized, C57BL/6 mice were assigned to a chemotherapy (9 mg/kg doxorubicin + 90 mg/kg cyclophosphamide) or vehicle two-injection regimen, with injections two and four weeks after starting diets. In Study 1, mice received low sucrose diets with EPA + DHA or No EPA + DHA for four to six weeks; tissues were collected four, seven, or 14 days after the second injection. Compared to vehicle, chemotherapy increased pro-inflammatory cytokine IL-1β at day seven in the cortex and hippocampus, and reduced gene expression of synaptic marker Shank 3 at all timepoints in cortex, while EPA + DHA increased expression of Shank 3. In Study 2, high or low sucrose/EPA + DHA or No EPA + DHA diets were fed for five weeks; tissues were collected ten days after the second injection. Among chemotherapy-treated mice, brain DHA was higher with low sucrose feeding. Furthermore, low sucrose increased gene expression of Shank 1, while EPA + DHA increased expression of Shank 3 and reduced protein concentrations of pro-inflammatory markers IL-5, IL-6 and KC/GRO in the cortex, but not the hippocampus. Low sucrose, EPA + DHA diets may attenuate neuroinflammation and synaptic damage induced by doxorubicin-based chemotherapy in specific brain regions.
Collapse
Affiliation(s)
- Tonya S Orchard
- Department of Human Sciences, Human Nutrition Program, The Ohio State University, Columbus, OH 43210, USA.
| | - Monica M Gaudier-Diaz
- Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, NC 27707, USA.
| | | | - Rebecca Andridge
- Division of Biostatistics, The Ohio State University, Columbus, OH 43210, USA.
| | - Maryam B Lustberg
- Division of Medical Oncology, The Ohio State University, Columbus, OH 43210, USA.
| | - Joshua Bomser
- Department of Human Sciences, Human Nutrition Program, The Ohio State University, Columbus, OH 43210, USA.
| | - Rachel M Cole
- Department of Human Sciences, Human Nutrition Program, The Ohio State University, Columbus, OH 43210, USA.
| | - Martha A Belury
- Department of Human Sciences, Human Nutrition Program, The Ohio State University, Columbus, OH 43210, USA.
| | - A Courtney DeVries
- Department of Neuroscience, West Virginia University, Morgantown, WV 26506, USA.
| |
Collapse
|
33
|
Thammisetty SS, Pedragosa J, Weng YC, Calon F, Planas A, Kriz J. Age-related deregulation of TDP-43 after stroke enhances NF-κB-mediated inflammation and neuronal damage. J Neuroinflammation 2018; 15:312. [PMID: 30413172 PMCID: PMC6230239 DOI: 10.1186/s12974-018-1350-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 10/29/2018] [Indexed: 12/13/2022] Open
Abstract
Background TDP-43 has been identified as a disease-associated protein in several chronic neurodegenerative disorders and increasing evidence suggests its potentially pathogenic role following brain injuries. Normally expressed in nucleus, under pathological conditions TDP-43 forms cytoplasmic ubiquitinated inclusions in which it is abnormally phosphorylated and cleaved to generate a 35 and a 25 kDa C-terminal fragments. In the present study, we investigated age-related expression patterns of TDP-43 in neurons and glia and its role as modulator of inflammation following ischemic injury. Methods Wild-type and TDP-43 transgenic mice of different age groups were subjected to transient middle cerebral artery occlusion. The role of TDP-43 in modulation of inflammation was assessed using immunofluorescence, Western blot analysis, and in vivo bioluminescence imaging. Finally, post-mortem stroke human brain sections were analyzed for TDP-43 protein by immunohistochemistry. Results We report here an age-related increase and formation of ubiquitinated TDP-43 cytoplasmic inclusions after stroke. The observed deregulation in TDP-43 expression patterns was associated with an increase in microglial activation and innate immune signaling as revealed by in vivo bioluminescence imaging and immunofluorescence analysis. The presence of ubiquitinated TDP-43 aggregates and its cleaved TDP-35 and TDP-25 fragments was markedly increased in older, 12-month-old mice leading to larger infarctions and a significant increase in in neuronal death. Importantly, unlike the hallmark neuropathological features associated with chronic neurodegenerative disorders, the TDP-43-positive cytoplasmic inclusions detected after stroke were not phosphorylated. Next, we showed that an increase and/or overexpression of the cytoplasmic TDP-43 drives the pathogenic NF-κB response and further increases levels of pro-inflammatory markers and ischemic injury after stroke in age-dependent manner. Finally, analyses of the post-mortem stroke brain tissues revealed the presence of the cytoplasmic TDP-43 immunoreactive structures after human stroke. Conclusion Together, our findings suggest that the level of cytoplasmic TDP-43 increases with aging and may act as an age-related mediator of inflammation and neuronal injury after stroke. Thus, targeting cytoplasmic TDP-43 may have a therapeutic potential after stroke. Electronic supplementary material The online version of this article (10.1186/s12974-018-1350-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sai Sampath Thammisetty
- CERVO Brain Research Centre, Université Laval, 2601 Chemin de la Canardière, Québec, QC, G1J 2G3, Canada.,Faculty of Pharmacy, Université Laval, Québec, QC, G1J2G3, Canada
| | | | - Yuan Cheng Weng
- CERVO Brain Research Centre, Université Laval, 2601 Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Frédéric Calon
- Research Centre of the CHUQ, Université Laval, Québec, QC, G1J2G3, Canada.,Faculty of Pharmacy, Université Laval, Québec, QC, G1J2G3, Canada
| | | | - Jasna Kriz
- CERVO Brain Research Centre, Université Laval, 2601 Chemin de la Canardière, Québec, QC, G1J 2G3, Canada. .,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, 2601 Chemin de la Canardière, Québec, QC, G1J2G3, Canada.
| |
Collapse
|
34
|
Li M, Liu J, Bi Y, Chen J, Zhao L. Potential Medications or Compounds Acting on Toll-like Receptors in Cerebral Ischemia. Curr Neuropharmacol 2018; 16:160-175. [PMID: 28571545 PMCID: PMC5883378 DOI: 10.2174/1570159x15666170601125139] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 05/24/2017] [Accepted: 05/31/2017] [Indexed: 01/22/2023] Open
Abstract
Background: Toll-like receptors play an integral role in the process of inflammatory response after ischemic in-jury. The therapeutic potential acting on TLRs is worth of evaluations. The aim of this review was to introduce readers some potential medications or compounds which could alleviate the ischemic damage via TLRs. Methods: Research articles online on TLRs were reviewed. Categorizations were listed according to the follows, methods acting on TLRs directly, modulations of MyD88 or TRIF signaling pathway, and the ischemic tolerance induced by the pre-conditioning or postconditioning with TLR ligands or minor cerebral ischemia via acting on TLRs. Results: There are only a few studies concerning on direct effects. Anti-TLR4 or anti-TLR2 therapies may serve as promis-ing strategies in acute events. Approaches targeting on inhibiting NF-κB signaling pathway and enhancing interferon regu-latory factor dependent signaling have attracted great interests. Not only drugs but compounds extracted from traditional Chinese medicine have been used to identify their neuroprotective effects against cerebral ischemia. In addition, many re-searchers have reported the positive therapeutic effects of preconditioning with agonists of TLR2, 3, 4, 7 and 9. Several trails have also explored the potential of postconditioning, which provide a new idea in ischemic treatments. Considering all the evidence above, many drugs and new compounds may have great potential to reduce ischemic insults. Conclusion: This review will focus on promising therapies which exerting neuroprotective effects against ischemic injury by acting on TLRs.
Collapse
Affiliation(s)
- Man Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jing Liu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Department of Neurology, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430033, China
| | - Ying Bi
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jixiang Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lei Zhao
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
35
|
Boutej H, Rahimian R, Thammisetty SS, Béland LC, Lalancette-Hébert M, Kriz J. Diverging mRNA and Protein Networks in Activated Microglia Reveal SRSF3 Suppresses Translation of Highly Upregulated Innate Immune Transcripts. Cell Rep 2018; 21:3220-3233. [PMID: 29241548 DOI: 10.1016/j.celrep.2017.11.058] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/26/2017] [Accepted: 11/16/2017] [Indexed: 01/15/2023] Open
Abstract
Uncontrolled microglial activation may lead to the development of inflammation-induced brain damage. Here, we uncover a ribosome-based mechanism/checkpoint involved in control of the innate immune response and microglial activation. Using an in vivo model system for analysis of the dynamic translational state of microglial ribosomes, with mRNAs as input and newly synthesized peptides as an output, we find a marked dissociation of microglia mRNA and protein networks following innate immune challenge. Highly upregulated and ribosome-associated mRNAs were not translated, resulting in two distinct microglial molecular signatures, a highly specialized pro-inflammatory mRNA signature and an immunomodulatory/homeostatic protein signature. We find that this is due to specific translational suppression of highly expressed mRNAs through a 3' UTR-mediated mechanism involving the RNA-binding protein SRSF3. This discovery suggests avenues for therapeutic modulation of innate immune response in resident microglia.
Collapse
Affiliation(s)
- Hejer Boutej
- CERVO Brain Research Centre and Department of Psychiatry and Neuroscience, Faculty of Medicine, Laval University, Québec, QC G1J2G3, Canada
| | - Reza Rahimian
- CERVO Brain Research Centre and Department of Psychiatry and Neuroscience, Faculty of Medicine, Laval University, Québec, QC G1J2G3, Canada
| | - Sai Sampath Thammisetty
- CERVO Brain Research Centre and Department of Psychiatry and Neuroscience, Faculty of Medicine, Laval University, Québec, QC G1J2G3, Canada
| | - Louis-Charles Béland
- CERVO Brain Research Centre and Department of Psychiatry and Neuroscience, Faculty of Medicine, Laval University, Québec, QC G1J2G3, Canada
| | - Mélanie Lalancette-Hébert
- CERVO Brain Research Centre and Department of Psychiatry and Neuroscience, Faculty of Medicine, Laval University, Québec, QC G1J2G3, Canada
| | - Jasna Kriz
- CERVO Brain Research Centre and Department of Psychiatry and Neuroscience, Faculty of Medicine, Laval University, Québec, QC G1J2G3, Canada.
| |
Collapse
|
36
|
Luo C, Ren H, Yao X, Shi Z, Liang F, Kang JX, Wan JB, Pei Z, Su KP, Su H. Enriched Brain Omega-3 Polyunsaturated Fatty Acids Confer Neuroprotection against Microinfarction. EBioMedicine 2018; 32:50-61. [PMID: 29880270 PMCID: PMC6021265 DOI: 10.1016/j.ebiom.2018.05.028] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 05/10/2018] [Accepted: 05/23/2018] [Indexed: 01/03/2023] Open
Abstract
Cerebral microinfarcts have significant effects on the development of geriatric neurological disorders, including vascular dementia and Alzheimer's disease. However, little is known about the pathophysiological mechanisms involved in the evolution of microinfarcts and potential treatment and prevention against these microvascular ischemic lesions. In the present study, the "single cortical microinfarct model" generated via occluding a penetrating arteriole by femtosecond laser ablation and the "multiple diffuse microinfarcts model" induced by unilateral injection of cholesterol crystals through the internal carotid artery were established to investigate the pathophysiological mechanisms underlying the evolution of microinfarcts and the effects of omega-3 polyunsaturated fatty acids (ω-3 PUFAs) on alleviating microinfarct burdens and functional deficits. The occlusion of a single penetrating arteriole led to a distinct cortical microinfarct, which manifested as neuronal loss and occupation of activated glial cells in the ischemic core. Using Fat-1 transgenic mice and fish oil supplements, we demonstrated that both endogenously-generated and exogenously-delivered ω-3 PUFAs significantly inhibited the activation of receptor-interacting serine/threonine protein kinases 1 (RIPK1) and its downstream apoptosis-associated proteins, mitigated cell apoptosis, and anatomically reduced the microinfarct size. The protective effects of ω-3 PUFAs against microinfarcts were further verified in a multiple diffuse microinfarcts model, where ω-3 PUFAs significantly attenuated cell apoptosis as revealed by TUNEL staining, alleviated the diffuse microinfarct burdens and remarkably improved the functional deficits as evidenced by reduced spontaneous anxiety, increased preference for the novel object, and improved hippocampal-based learning and short-term memory. Together, these findings demonstrate that enriched brain ω-3 PUFAs are effective for reducing microinfarct burdens and improving the function deficits, which support the clinical research and application of ω-3 PUFAs in the treatment or prophylaxis in vascular dementia.
Collapse
Affiliation(s)
- Chuanming Luo
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China; Department of Neurology, Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 510080, China
| | - Huixia Ren
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Xiaoli Yao
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, First Affiliated Hospital Sun Yat-Sen University, Guangzhou 510080, China
| | - Zhe Shi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Fengyin Liang
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, First Affiliated Hospital Sun Yat-Sen University, Guangzhou 510080, China
| | - Jing X Kang
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jian-Bo Wan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Zhong Pei
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, First Affiliated Hospital Sun Yat-Sen University, Guangzhou 510080, China
| | - Kuan-Pin Su
- Department of Psychiatry & Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, College of Medicine, China Medical University, Taichung, Taiwan
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
| |
Collapse
|
37
|
Shinde AB, Baboota RK, Denis S, Loizides-Mangold U, Peeters A, Espeel M, Malheiro AR, Riezman H, Vinckier S, Vaz FM, Brites P, Ferdinandusse S, Van Veldhoven PP, Baes M. Mitochondrial disruption in peroxisome deficient cells is hepatocyte selective but is not mediated by common hepatic peroxisomal metabolites. Mitochondrion 2018; 39:51-59. [DOI: 10.1016/j.mito.2017.08.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 08/25/2017] [Indexed: 01/06/2023]
|
38
|
Layé S, Nadjar A, Joffre C, Bazinet RP. Anti-Inflammatory Effects of Omega-3 Fatty Acids in the Brain: Physiological Mechanisms and Relevance to Pharmacology. Pharmacol Rev 2018; 70:12-38. [PMID: 29217656 DOI: 10.1124/pr.117.014092] [Citation(s) in RCA: 253] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 09/05/2017] [Indexed: 12/17/2022] Open
Abstract
Classically, polyunsaturated fatty acids (PUFA) were largely thought to be relatively inert structural components of brain, largely important for the formation of cellular membranes. Over the past 10 years, a host of bioactive lipid mediators that are enzymatically derived from arachidonic acid, the main n-6 PUFA, and docosahexaenoic acid, the main n-3 PUFA in the brain, known to regulate peripheral immune function, have been detected in the brain and shown to regulate microglia activation. Recent advances have focused on how PUFA regulate the molecular signaling of microglia, especially in the context of neuroinflammation and behavior. Several active drugs regulate brain lipid signaling and provide proof of concept for targeting the brain. Because brain lipid metabolism relies on a complex integration of diet, peripheral metabolism, including the liver and blood, which supply the brain with PUFAs that can be altered by genetics, sex, and aging, there are many pathways that can be disrupted, leading to altered brain lipid homeostasis. Brain lipid signaling pathways are altered in neurologic disorders and may be viable targets for the development of novel therapeutics. In this study, we discuss in particular how n-3 PUFAs and their metabolites regulate microglia phenotype and function to exert their anti-inflammatory and proresolving activities in the brain.
Collapse
Affiliation(s)
- Sophie Layé
- Institut National pour la Recherche Agronomique and Bordeaux University, Nutrition et Neurobiologie Intégrée, UMR 1286, Bordeaux, France (S.L., A.N., C.J.); and Department of Nutritional Sciences, University of Toronto, Ontario, Canada (R.P.B.)
| | - Agnès Nadjar
- Institut National pour la Recherche Agronomique and Bordeaux University, Nutrition et Neurobiologie Intégrée, UMR 1286, Bordeaux, France (S.L., A.N., C.J.); and Department of Nutritional Sciences, University of Toronto, Ontario, Canada (R.P.B.)
| | - Corinne Joffre
- Institut National pour la Recherche Agronomique and Bordeaux University, Nutrition et Neurobiologie Intégrée, UMR 1286, Bordeaux, France (S.L., A.N., C.J.); and Department of Nutritional Sciences, University of Toronto, Ontario, Canada (R.P.B.)
| | - Richard P Bazinet
- Institut National pour la Recherche Agronomique and Bordeaux University, Nutrition et Neurobiologie Intégrée, UMR 1286, Bordeaux, France (S.L., A.N., C.J.); and Department of Nutritional Sciences, University of Toronto, Ontario, Canada (R.P.B.)
| |
Collapse
|
39
|
Impact of DHA intake in a mouse model of synucleinopathy. Exp Neurol 2017; 301:39-49. [PMID: 29229294 DOI: 10.1016/j.expneurol.2017.12.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 11/26/2017] [Accepted: 12/06/2017] [Indexed: 11/22/2022]
Abstract
Polyunsaturated fatty acids omega-3 (n-3 PUFA), such as docosahexaenoic acid (DHA), have been shown to prevent, and partially reverse, neurotoxin-induced nigrostriatal denervation in animal models of Parkinson's disease (PD). However, the accumulation of α-synuclein (αSyn) in cerebral tissues is equally important to the pathophysiology. To determine whether DHA intake improves various aspects related to synucleinopathy, ninety male mice overexpressing human αSyn under the Thy-1 promoter (Thy1-αSyn) were fed one of three diets (specially formulated control, low n-3 PUFA or high DHA) and compared to non-transgenic C57/BL6 littermate mice exposed to a control diet. Thy1-αSyn mice displayed impaired motor skills, lower dopaminergic neuronal counts within the substantia nigra (-13%) in parallel to decreased levels of the striatal dopamine transporter (DAT) (-24%), as well as reduced NeuN (-41%) and synaptic proteins PSD-95 (-51%), synaptophysin (-80%) and vesicular acetylcholine transporter (VChAT) (-40%) in the cerebral cortex compared to C57/BL6 mice. However, no significant difference in dopamine concentrations was observed by HPLC analysis between Thy1-αSyn and non-transgenic C57/BL6 littermates under the control diet. The most striking finding was a favorable effect of DHA on the survival/longevity of Thy1-αSyn mice (+51% survival rate at 12months of age). However, dietary DHA supplementation did not have a significant effect on other parameters examined in this study, despite increased striatal dopamine concentrations. While human αSyn monomers and oligomers were detected in the cortex of Thy1-αSyn mice, the effects of the diets were limited to a small increase of 42kDa oligomers in insoluble protein fractions upon n-3 PUFA deprivation. Overall, our data indicate that a diet rich in n-3 PUFA has a beneficial effect on the longevity of a murine model of α-synucleinopathy without a major impact on the dopamine system and motor impairments, nor αSyn levels.
Collapse
|
40
|
Kerdiles O, Layé S, Calon F. Omega-3 polyunsaturated fatty acids and brain health: Preclinical evidence for the prevention of neurodegenerative diseases. Trends Food Sci Technol 2017. [DOI: 10.1016/j.tifs.2017.09.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
41
|
Lalancette-Hébert M, Faustino J, Thammisetty SS, Chip S, Vexler ZS, Kriz J. Live imaging of the innate immune response in neonates reveals differential TLR2 dependent activation patterns in sterile inflammation and infection. Brain Behav Immun 2017; 65:312-327. [PMID: 28579520 PMCID: PMC6151183 DOI: 10.1016/j.bbi.2017.05.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 05/31/2017] [Accepted: 05/31/2017] [Indexed: 01/07/2023] Open
Abstract
Activation of microglial cells in response to brain injury and/or immune stimuli is associated with a marked induction of Toll-like receptors (TLRs). While in adult brain, the contribution of individual TLRs, including TLR2, in pathophysiological cascades has been well established, their role and spatial and temporal induction patterns in immature brain are far less understood. To examine whether infectious stimuli and sterile inflammatory stimuli trigger distinct TLR2-mediated innate immune responses, we used three models in postnatal day 9 (P9) mice, a model of infection induced by systemic endotoxin injection and two models of sterile inflammation, intra-cortical IL-1β injection and transient middle cerebral artery occlusion (tMCAO). We took advantage of a transgenic mouse model bearing the dual reporter system luciferase/GFP under transcriptional control of a murine TLR2 promoter (TLR2-luc-GFP) to visualize the TLR2 response in the living neonatal brain and then determined neuroinflammation, microglial activation and leukocyte infiltration. We show that in physiological postnatal brain development the in vivo TLR2-luc signal undergoes a marked ∼30-fold decline and temporal-spatial changes during the second and third postnatal weeks. We then show that while endotoxin robustly induces the in vivo TLR2-luc signal in the living brain and increases levels of several inflammatory cytokines and chemokines, the in vivo TLR2-luc signal is reduced after both IL-1β and tMCAO and the inflammatory response is muted. Immunofluorescence revealed that microglial cells are the predominant source of TLR2 production during postnatal brain development and in all three neonatal models studied. Flow cytometry revealed developmental changes in CD11b+/CD45+ and CD11b+/Ly6C+ cell populations, involvement of cells of the monocyte lineage, but lack of Ly6G+ neutrophils or CD3+ cells in acutely injured neonatal brains. Cumulatively, our results suggest distinct TLR2 induction patterns following PAMP and DAMP - mediated inflammation in immature brain.
Collapse
Affiliation(s)
- Melanie Lalancette-Hébert
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Laval University, Research Center of the IUSMQ, 2601, de la Canardière, Québec, QC G1J 2G3, Canada
| | - Joel Faustino
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158-0663, USA
| | - Sai Sampath Thammisetty
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Laval University, Research Center of the IUSMQ, 2601, de la Canardière, Québec, QC G1J 2G3, Canada
| | - Sophorn Chip
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158-0663, USA
| | - Zinaida S Vexler
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158-0663, USA.
| | - Jasna Kriz
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Laval University, Research Center of the IUSMQ, 2601, de la Canardière, Québec, QC G1J 2G3, Canada.
| |
Collapse
|
42
|
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
|
43
|
Ren H, Yang Z, Luo C, Zeng H, Li P, Kang JX, Wan JB, He C, Su H. Enriched Endogenous Omega-3 Fatty Acids in Mice Ameliorate Parenchymal Cell Death After Traumatic Brain Injury. Mol Neurobiol 2017; 54:3317-3326. [PMID: 27167127 DOI: 10.1007/s12035-016-9931-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 05/04/2016] [Indexed: 12/18/2022]
Abstract
Currently no effective therapies are available for the treatment of traumatic brain injury (TBI). Early intervention that specifically provides neuroprotection is of most importance which profoundly influences the outcome of TBI. In the present study, we adopted a closed-skull mild TBI model to investigate potential roles of omega-3 polyunsaturated fatty acids (ω-3 PUFAs) in protecting against TBI. Using two-photon laser scanning microscopy (2PLSM), parenchymal cell death and reactive oxidative species (ROS) expression were directly observed and recorded after TBI through a thinned skull bone window. Fat-1 mice with high endogenous ω-3 PUFAs significantly inhibited ROS expression and attenuated parenchymal cell death after compression injury during the early injury phase. Elevated generation of glutathione (GSH) and neuroprotectin D1 (NPD1) in the parenchyma of fat-1 mice could be the contributor to the beneficial role of ω-3 PUFAs in TBI. The results of the study suggest that ω-3 PUFAs is an effective neuroprotectant as an early pharmacological intervention for TBI and the information derived from this study may help guide dietary advice for those who are susceptible to repetitive mild TBI.
Collapse
Affiliation(s)
- Huixia Ren
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Zhen Yang
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China
| | - Chuanming Luo
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Haitao Zeng
- Center for Reproductive Medicine, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Peng Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Jing X Kang
- Laboratory for Lipid Medicine and Technology, the Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jian-Bo Wan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Chengwei He
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
| |
Collapse
|
44
|
Amplification of mGlu 5-Endocannabinoid Signaling Rescues Behavioral and Synaptic Deficits in a Mouse Model of Adolescent and Adult Dietary Polyunsaturated Fatty Acid Imbalance. J Neurosci 2017. [PMID: 28630250 DOI: 10.1523/jneurosci.3516-16.2017] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Energy-dense, yet nutritionally poor food is a high-risk factor for mental health disorders. This is of particular concern during adolescence, a period often associated with increased consumption of low nutritional content food and higher prevalence of mental health disorders. Indeed, there is an urgent need to understand the mechanisms linking unhealthy diet and mental disorders. Deficiency in n-3 polyunsaturated fatty acids (PUFAs) is a hallmark of poor nutrition and mood disorders. Here, we developed a mouse model of n-3 PUFA deficiency lasting from adolescence into adulthood. Starting nutritional deficits in dietary n-3 PUFAs during adolescence decreased n-3 PUFAs in both medial prefrontal cortex (mPFC) and nucleus accumbens, increased anxiety-like behavior, and decreased cognitive function in adulthood. Importantly, we discovered that endocannabinoid/mGlu5-mediated LTD in the mPFC and accumbens was abolished in adult n-3-deficient mice. Additionally, mPFC NMDAR-dependent LTP was also lacking in the n-3-deficient group. Pharmacological enhancement of the mGlu5/eCB signaling complex, by positive allosteric modulation of mGlu5 or inhibition of endocannabinoid 2-arachidonylglycerol degradation, fully restored synaptic plasticity and normalized emotional and cognitive behaviors in malnourished adult mice. Our data support a model where nutrition is a key environmental factor influencing the working synaptic range into adulthood, long after the end of the perinatal period. These findings have important implications for the identification of nutritional risk factors for disease and design of new treatments for the behavioral deficits associated with nutritional n-3 PUFA deficiency.SIGNIFICANCE STATEMENT In a mouse model mimicking n-3 PUFA dietary deficiency during adolescence and adulthood, we found strong increases in anxiety and anhedonia which lead to decreases in specific cognitive functions in adulthood. We found that endocannabinoid/mGlu5-mediated LTD and NMDAR-dependent LTP were lacking in adult n-3-deficient mice. Acute positive allosteric modulation of mGlu5 or inhibition of endocannabinoid degradation normalized behaviors and synaptic functions in n-3 PUFA-deficient adult mice. These findings have important implications for the identification of nutritional risk for disease and the design of new treatments for the behavioral deficits associated with nutritional n-3 PUFAs' imbalance.
Collapse
|
45
|
Wang Y, Plastina P, Vincken JP, Jansen R, Balvers M, ten Klooster JP, Gruppen H, Witkamp R, Meijerink J. N-Docosahexaenoyl Dopamine, an Endocannabinoid-like Conjugate of Dopamine and the n-3 Fatty Acid Docosahexaenoic Acid, Attenuates Lipopolysaccharide-Induced Activation of Microglia and Macrophages via COX-2. ACS Chem Neurosci 2017; 8:548-557. [PMID: 28292183 DOI: 10.1021/acschemneuro.6b00298] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Several studies indicate that the n-3 long-chain polyunsaturated fatty acid docosahexaenoic acid (DHA) contributes to an attenuated inflammatory status in the development of neurodegenerative disorders, such as Alzheimer's and Parkinson's disease. To explain these effects, different mechanisms are being proposed, including those involving endocannabinoids and related signaling molecules. Many of these compounds belong to the fatty acid amides, conjugates of fatty acids with biogenic amines. Conjugates of DHA with ethanolamine or serotonin have previously been shown to possess anti-inflammatory and potentially neuroprotective properties. Here, we synthesized another amine conjugate of DHA, N-docosahexaenoyl dopamine (DHDA), and tested its immune-modulatory properties in both RAW 264.7 macrophages and BV-2 microglial cells. N-Docosahexaenoyl dopamine significantly suppressed the production of nitric oxide (NO), the cytokine interleukin-6 (IL-6), and the chemokines macrophage-inflammatory protein-3α (CCL20) and monocyte chemoattractant protein-1 (MCP-1), whereas its parent compounds, dopamine and DHA, were ineffective. Further exploration of potential effects of DHDA on key inflammatory mediators revealed that cyclooxygenase-2 (COX-2) mRNA level and production of prostaglandin E2 (PGE2) were concentration-dependently inhibited in macrophages. In activated BV-2 cells, PGE2 production was also reduced, without changes in COX-2 mRNA levels. In addition, DHDA did not affect NF-kB activity in a reporter cell line. Finally, the immune-modulatory activities of DHDA were compared with those of N-arachidonoyl dopamine (NADA) and similar potencies were found in both cell types. Taken together, our data suggest that DHDA, a potentially endogenous endocannabinoid, may be an additional member of the group of immune-modulating n-3 fatty acid-derived lipid mediators.
Collapse
Affiliation(s)
| | - Pierluigi Plastina
- Department
of Chemistry and Chemical Technologies, University of Calabria, 87036 Cosenza, Italy
| | | | | | | | - Jean Paul ten Klooster
- Research Centre Technology & Innovation, Innovative Testing in Life Sciences and Chemistry, University of Applied Sciences, 3584 CH Utrecht, The Netherlands
| | | | | | | |
Collapse
|
46
|
Neuroprotective diets for stroke. Neurochem Int 2017; 107:4-10. [PMID: 28161467 DOI: 10.1016/j.neuint.2017.01.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 01/20/2017] [Accepted: 01/24/2017] [Indexed: 11/22/2022]
Abstract
Stroke is one of the main causes of death and disability in the elderly. In the last few years, there has been increasing evidence that suggests the influence of the diet on the decrease of stroke risk. Probably, because of the presence of bioactive components with beneficial effects such as antioxidant or anti-inflammatory properties. This article reviews several dietary bioactive compounds from studies in models of cerebral ischemia that have obtained promising results decreasing cerebral damage. We propose that many of these compounds present in diet could be good candidates to test new neuroprotection approaches focused on reducing the damage and protecting the brain before stroke occurs.
Collapse
|
47
|
Orchard TS, Gaudier-Diaz MM, Weinhold KR, Courtney DeVries A. Clearing the fog: a review of the effects of dietary omega-3 fatty acids and added sugars on chemotherapy-induced cognitive deficits. Breast Cancer Res Treat 2016; 161:391-398. [PMID: 27933449 DOI: 10.1007/s10549-016-4073-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 11/29/2016] [Indexed: 02/07/2023]
Abstract
Cancer treatments such as chemotherapy have been an important part of extending survival in women diagnosed with breast cancer. However, chemotherapy can cause potentially toxic side effects in the brain that impair memory, verbal fluency, and processing speed in up to 30% of women treated. Women report that post-chemotherapy cognitive deficits negatively impact quality of life and may last up to ten years after treatment. Mechanisms underlying these cognitive impairments are not fully understood, but emerging evidence suggests that chemotherapy induces structural changes in the brain, produces neuroinflammation, and reduces adult hippocampal neurogenesis. Dietary approaches that modify inflammation and neurogenesis are promising strategies for reducing chemotherapy-induced cognitive deficits in breast cancer survivors. In this review, we describe the cognitive and neuronal side effects associated with commonly used chemotherapy treatments for breast cancer, and we focus on the often opposing actions of omega-3 fatty acids and added sugars on cognitive function, neuroinflammation, and adult hippocampal neurogenesis. Omega-3 fatty acids administered concurrently with doxorubicin chemotherapy have been shown to prevent depressive-like behaviors and reduce neuroinflammation, oxidative stress, and neural apoptosis in rodent models. In contrast, diets high in added sugars may interact with n-3 FAs to diminish their anti-inflammatory activity or act independently to increase neuroinflammation, reduce adult hippocampal neurogenesis, and promote cognitive deficits. We propose that a diet rich in long-chain, marine-derived omega-3 fatty acids and low in added sugars may be an ideal pattern for preventing or alleviating neuroinflammation and oxidative stress, thereby protecting neurons from the toxic effects of chemotherapy. Research testing this hypothesis could lead to the identification of modifiable dietary choices to reduce the long-term impact of chemotherapy on the cognitive functions that are important to quality of life in breast cancer survivors.
Collapse
Affiliation(s)
- Tonya S Orchard
- Department of Human Sciences, The Ohio State University, 325 Campbell Hall, 1787 Neil Avenue, Columbus, OH, 43210, USA.
| | - Monica M Gaudier-Diaz
- Department of Neuroscience, The Ohio State University Wexner Medical Center, 614 Biomedical Research Tower, 460 West 12th Avenue, Columbus, OH, 43210, USA
| | - Kellie R Weinhold
- Department of Human Sciences, The Ohio State University, 325 Campbell Hall, 1787 Neil Avenue, Columbus, OH, 43210, USA
| | - A Courtney DeVries
- Department of Neuroscience, The Ohio State University Wexner Medical Center, 614 Biomedical Research Tower, 460 West 12th Avenue, Columbus, OH, 43210, USA
| |
Collapse
|
48
|
DHA-supplemented diet increases the survival of rats following asphyxia-induced cardiac arrest and cardiopulmonary bypass resuscitation. Sci Rep 2016; 6:36545. [PMID: 27811958 PMCID: PMC5109906 DOI: 10.1038/srep36545] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 10/17/2016] [Indexed: 12/27/2022] Open
Abstract
Accumulating evidence illustrates the beneficial effects of dietary docosahexaenoic acid (DHA) on cardiovascular diseases. However, its effects on cardiac arrest (CA) remain controversial in epidemiological studies and have not been reported in controlled animal studies. Here, we examined whether dietary DHA can improve survival, the most important endpoint in CA. Male Sprague-Dawley rats were randomized into two groups and received either a control diet or a DHA-supplemented diet for 7–8 weeks. Rats were then subjected to 20 min asphyxia-induced cardiac arrest followed by 30 min cardiopulmonary bypass resuscitation. Rat survival was monitored for additional 3.5 h following resuscitation. In the control group, 1 of 9 rats survived for 4 h, whereas 6 of 9 rats survived in the DHA-treated group. Surviving rats in the DHA-treated group displayed moderately improved hemodynamics compared to rats in the control group 1 h after the start of resuscitation. Rats in the control group showed no sign of brain function whereas rats in the DHA-treated group had recurrent seizures and spontaneous respiration, suggesting dietary DHA also protects the brain. Overall, our study shows that dietary DHA significantly improves rat survival following 20 min of severe CA.
Collapse
|
49
|
Pu H, Jiang X, Hu X, Xia J, Hong D, Zhang W, Gao Y, Chen J, Shi Y. Delayed Docosahexaenoic Acid Treatment Combined with Dietary Supplementation of Omega-3 Fatty Acids Promotes Long-Term Neurovascular Restoration After Ischemic Stroke. Transl Stroke Res 2016; 7:521-534. [PMID: 27566736 DOI: 10.1007/s12975-016-0498-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 08/16/2016] [Accepted: 08/18/2016] [Indexed: 12/14/2022]
Abstract
Prophylactic dietary intake of omega-3 polyunsaturated fatty acids (n-3 PUFAs) has been shown to remarkably ameliorate ischemic brain injury. However, the therapeutic efficacy of n-3 PUFA administration post-stroke, especially its impact on neurovascular remodeling and long-term neurological recovery, has not been fully characterized thus far. In this study, we investigated the effect of n-3 PUFA supplementation, as well as in combination with docosahexaenoic acid (DHA) injections, on long-term stroke outcomes. Mice were subjected to transient middle cerebral artery occlusion (MCAO) before randomly assigned to four groups to receive the following: (1) low dose of n-3 PUFAs as the vehicle control, (2) intraperitoneal DHA injections, (3) n-3 PUFA dietary supplement, or (4) combined treatment of (2) and (3). Neurological deficits and brain atrophy, neurogenesis, angiogenesis, and glial scar formation were assessed up to 28 days after MCAO. Results revealed that groups 2 and 3 showed only marginal reduction in post-stroke tissue loss and attenuation of cognitive deficits. Interestingly, group 4 exhibited significantly reduced tissue atrophy and improved cognitive functions compared to groups 2 and 3 with just a single treatment. Mechanistically, the combined treatment promoted post-stroke neurogenesis and angiogenesis, as well as reduced glial scar formation, all of which significantly correlated with the improved spatial memory in the Morris water maze. These results demonstrate an effective therapeutic regimen to enhance neurovascular restoration and long-term cognitive recovery in the mouse model of MCAO. Combined post-stroke DHA treatment and n-3 PUFA dietary supplementation thus may be a potential clinically translatable therapy for stroke or related brain disorders.
Collapse
Affiliation(s)
- Hongjian Pu
- Geriatric Research, Educational, and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA.,Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Xiaoyan Jiang
- State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China.,Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Xiaoming Hu
- Geriatric Research, Educational, and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA.,State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China.,Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Jinchao Xia
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Dandan Hong
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Wenting Zhang
- State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China.,Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Jun Chen
- Geriatric Research, Educational, and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA. .,State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China. .,Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| | - Yejie Shi
- Geriatric Research, Educational, and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA. .,Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
50
|
Coulombe K, Saint-Pierre M, Cisbani G, St-Amour I, Gibrat C, Giguère-Rancourt A, Calon F, Cicchetti F. Partial neurorescue effects of DHA following a 6-OHDA lesion of the mouse dopaminergic system. J Nutr Biochem 2016; 30:133-42. [DOI: 10.1016/j.jnutbio.2015.11.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 11/10/2015] [Accepted: 11/20/2015] [Indexed: 01/12/2023]
|