1
|
Liu D, Yang S, Yu S. Interactions Between Ferroptosis and Oxidative Stress in Ischemic Stroke. Antioxidants (Basel) 2024; 13:1329. [PMID: 39594471 PMCID: PMC11591163 DOI: 10.3390/antiox13111329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/10/2024] [Accepted: 10/16/2024] [Indexed: 11/28/2024] Open
Abstract
Ischemic stroke is a devastating condition that occurs due to the interruption of blood flow to the brain, resulting in a range of cellular and molecular changes. In recent years, there has been growing interest in the role of ferroptosis, a newly identified form of regulated cell death, in ischemic stroke. Ferroptosis is driven by the accumulation of lipid peroxides and is characterized by the loss of membrane integrity. Additionally, oxidative stress, which refers to an imbalance between prooxidants and antioxidants, is a hallmark of ischemic stroke and significantly contributes to the pathogenesis of the disease. In this review, we explore the interactions between ferroptosis and oxidative stress in ischemic stroke. We examine the underlying mechanisms through which oxidative stress induces ferroptosis and how ferroptosis, in turn, exacerbates oxidative stress. Furthermore, we discuss potential therapeutic strategies that target both ferroptosis and oxidative stress in the treatment of ischemic stroke. Overall, this review highlights the complex interplay between ferroptosis and oxidative stress in ischemic stroke and underscores the need for further research to identify novel therapeutic targets for this condition.
Collapse
Affiliation(s)
| | - Sha Yang
- College of Acupuncture and Massage, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China;
| | - Shuguang Yu
- College of Acupuncture and Massage, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China;
| |
Collapse
|
2
|
Zhaliazka K, Kurouski D. Nanoscale Structural Characterization of Amyloid β 1-42 Oligomers and Fibrils Grown in the Presence of Fatty Acids. ACS Chem Neurosci 2024; 15:3344-3353. [PMID: 39222387 DOI: 10.1021/acschemneuro.4c00275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Mono- and polyunsaturated fatty acids (FAs) are broadly used as food supplements. However, their effect on the aggregation of amyloidogenic proteins remains unclear. In this study, we investigated the effect of a large number of mono- and polyunsaturated, as well as fully saturated FAs on the aggregation of amyloid β1-42 (Aβ1-42) peptide. A progressive aggregation of this peptide is the expected molecular cause of Alzheimer's disease (AD), one of the most common neurodegenerative pathologies in the world. We found that arachidonic and stearic acids delayed the aggregation of Aβ1-42. Using Nano-Infrared spectroscopy, we found that FAs caused very little if any changes in the secondary structure of Aβ1-42 oligomers and fibrils formed at different stages of protein aggregation. However, the analyzed mono- and polyunsaturated, as well as fully saturated FAs uniquely altered the toxicity of Aβ1-42 fibrils. We found a direct relationship between the degree of FAs unsaturation and toxicity of Aβ1-42 fibrils formed in their presence. Specifically, with an increase in the degree of unsaturation, the toxicity Aβ1-42/FA fibrils increased. These results indicate that fully saturated or monounsaturated FAs could be used to decrease the toxicity of amyloid aggregates and, consequently, decelerate the development of AD.
Collapse
Affiliation(s)
- Kiryl Zhaliazka
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - Dmitry Kurouski
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843, United States
| |
Collapse
|
3
|
Ciesielski TH, Tosto G, Durodoye RO, Rajabali F, Akinyemi RO, Byrd GS, Bush WS, Kunkle BW, Reitz C, Vance JM, Pericak-Vance MA, Haines JL, Williams SM. Country Level Incidence of Alzheimer Disease and Related Dementias is Associated with Increased Omega6 PUFA Consumption. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.07.24311637. [PMID: 39148832 PMCID: PMC11326357 DOI: 10.1101/2024.08.07.24311637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
INTRODUCTION Clinical and genetic studies have implicated lipid dysfunction in Alzheimer Disease (AD) pathogenesis. However, lipid consumption at the individual-level does not vary greatly within most cohorts, and multiple lipids are rarely measured in any one study. METHODS Mean country-level lipid intakes were compared to Age-Standardized Alzheimer-Disease-Incidence-Rates(ASAIR) in 183 countries across all inhabited continents. Penalized spline regression and multivariable-adjusted linear regression, including a lag between intake and incidence, were used to assess the relationships between five lipid intakes and ASAIR. Validation was conducted using longitudinal within-country changes between 1990 and 2019. RESULTS Omega6 Polyunsaturated-Fatty-Acid(PUFA) intake exhibited a positive linear relationship with ASAIR(multivariable-adjusted model: β=2.44; 95%CI: 1.70, 3.19; p=1.38×10-9). ASAIR also increased with saturated-fat, trans-fat, and dietary-cholesterol up to a threshold. The association between Omega6-PUFA and ASAIR was confirmed using longitudinal intake changes. DISCUSSION Decreasing Omega6-PUFA consumption on the country-level may have substantial benefits in reducing the country-level burden of AD.
Collapse
Affiliation(s)
- Timothy H Ciesielski
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Ave. Cleveland, OH 44106
| | - Giuseppe Tosto
- Taub Institute for Research on Alzheimer Disease and the Aging Brain, Department of Neurology, Columbia University College of Physicians and Surgeons, 630 West 168th Street New York, NY 10032
| | - Razaq O Durodoye
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Ave. Cleveland, OH 44106
| | - Farid Rajabali
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine 1501 NW 10th Ave. Biomedical Research Building, Miami, FL 33136
| | - Rufus O Akinyemi
- Neuroscience and Ageing Research Unit, Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, P.M.B 5017 G.P.O Ibadan,Oyo State, Nigeria
| | - Goldie S Byrd
- Maya Angelou Center for Health Equity, Wake Forest University School of Medicine, 525 Vine Street Suite #150, 1st Floor, Winston-Salem, NC 27101
| | - William S Bush
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Ave. Cleveland, OH 44106
- Cleveland Institute for Computational Biology, Case Western Reserve University, 2-530 Wolstein Research Building, 2103 Cornell Road, Cleveland, OH
| | - Brian W Kunkle
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine 1501 NW 10th Ave. Biomedical Research Building, Miami, FL 33136
| | - Christiane Reitz
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Ave. Cleveland, OH 44106
- Taub Institute for Research on Alzheimer Disease and the Aging Brain, Department of Neurology, Columbia University College of Physicians and Surgeons, 630 West 168th Street New York, NY 10032
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine 1501 NW 10th Ave. Biomedical Research Building, Miami, FL 33136
- Neuroscience and Ageing Research Unit, Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, P.M.B 5017 G.P.O Ibadan,Oyo State, Nigeria
- Maya Angelou Center for Health Equity, Wake Forest University School of Medicine, 525 Vine Street Suite #150, 1st Floor, Winston-Salem, NC 27101
- Cleveland Institute for Computational Biology, Case Western Reserve University, 2-530 Wolstein Research Building, 2103 Cornell Road, Cleveland, OH
| | - Jeffery M Vance
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine 1501 NW 10th Ave. Biomedical Research Building, Miami, FL 33136
| | - Margaret A Pericak-Vance
- Neuroscience and Ageing Research Unit, Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, P.M.B 5017 G.P.O Ibadan,Oyo State, Nigeria
| | - Jonathan L Haines
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Ave. Cleveland, OH 44106
- Cleveland Institute for Computational Biology, Case Western Reserve University, 2-530 Wolstein Research Building, 2103 Cornell Road, Cleveland, OH
| | - Scott M Williams
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Ave. Cleveland, OH 44106
- Cleveland Institute for Computational Biology, Case Western Reserve University, 2-530 Wolstein Research Building, 2103 Cornell Road, Cleveland, OH
| |
Collapse
|
4
|
Meng X, Song Q, Liu Z, Liu X, Wang Y, Liu J. Neurotoxic β-amyloid oligomers cause mitochondrial dysfunction-the trigger for PANoptosis in neurons. Front Aging Neurosci 2024; 16:1400544. [PMID: 38808033 PMCID: PMC11130508 DOI: 10.3389/fnagi.2024.1400544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 04/29/2024] [Indexed: 05/30/2024] Open
Abstract
As the global population ages, the incidence of elderly patients with dementia, represented by Alzheimer's disease (AD), will continue to increase. Previous studies have suggested that β-amyloid protein (Aβ) deposition is a key factor leading to AD. However, the clinical efficacy of treating AD with anti-Aβ protein antibodies is not satisfactory, suggesting that Aβ amyloidosis may be a pathological change rather than a key factor leading to AD. Identification of the causes of AD and development of corresponding prevention and treatment strategies is an important goal of current research. Following the discovery of soluble oligomeric forms of Aβ (AβO) in 1998, scientists began to focus on the neurotoxicity of AβOs. As an endogenous neurotoxin, the active growth of AβOs can lead to neuronal death, which is believed to occur before plaque formation, suggesting that AβOs are the key factors leading to AD. PANoptosis, a newly proposed concept of cell death that includes known modes of pyroptosis, apoptosis, and necroptosis, is a form of cell death regulated by the PANoptosome complex. Neuronal survival depends on proper mitochondrial function. Under conditions of AβO interference, mitochondrial dysfunction occurs, releasing lethal contents as potential upstream effectors of the PANoptosome. Considering the critical role of neurons in cognitive function and the development of AD as well as the regulatory role of mitochondrial function in neuronal survival, investigation of the potential mechanisms leading to neuronal PANoptosis is crucial. This review describes the disruption of neuronal mitochondrial function by AβOs and elucidates how AβOs may activate neuronal PANoptosis by causing mitochondrial dysfunction during the development of AD, providing guidance for the development of targeted neuronal treatment strategies.
Collapse
Affiliation(s)
| | | | | | | | | | - Jinyu Liu
- Department of Toxicology, School of Public Health, Jilin University, Changchun, China
| |
Collapse
|
5
|
Sharma C, Mazumder A. A Comprehensive Review on Potential Molecular Drug Targets for the Management of Alzheimer's Disease. Cent Nerv Syst Agents Med Chem 2024; 24:45-56. [PMID: 38305393 DOI: 10.2174/0118715249263300231116062740] [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: 06/20/2023] [Revised: 08/25/2023] [Accepted: 10/04/2023] [Indexed: 02/03/2024]
Abstract
Alzheimer's disease (AD) is an onset and incurable neurodegenerative disorder that has been linked to various genetic, environmental, and lifestyle factors. Recent research has revealed several potential targets for drug development, such as the prevention of Aβ production and removal, prevention of tau hyperphosphorylation, and keeping neurons alive. Drugs that target numerous ADrelated variables have been developed, and early results are encouraging. This review provides a concise map of the different receptor signaling pathways associated with Alzheimer's Disease, as well as insight into drug design based on these pathways. It discusses the molecular mechanisms of AD pathogenesis, such as oxidative stress, aging, Aβ turnover, thiol groups, and mitochondrial activities, and their role in the disease. It also reviews the potential drug targets, in vivo active agents, and docking studies done in AD and provides prospects for future drug development. This review intends to provide more clarity on the molecular processes that occur in Alzheimer's patient's brains, which can be of use in diagnosing and preventing the condition.
Collapse
Affiliation(s)
- Chanchal Sharma
- Noida Institute of Engineering and Technology (Pharmacy Institute), 19 Knowledge Park-II, Institutional Area, Greater Noida-201306, Uttar Pradesh, India
| | - Avijit Mazumder
- Noida Institute of Engineering and Technology (Pharmacy Institute), 19 Knowledge Park-II, Institutional Area, Greater Noida-201306, Uttar Pradesh, India
| |
Collapse
|
6
|
Chen F, Kang R, Liu J, Tang D. The ACSL4 Network Regulates Cell Death and Autophagy in Diseases. BIOLOGY 2023; 12:864. [PMID: 37372148 DOI: 10.3390/biology12060864] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/05/2023] [Accepted: 06/11/2023] [Indexed: 06/29/2023]
Abstract
Lipid metabolism, cell death, and autophagy are interconnected processes in cells. Dysregulation of lipid metabolism can lead to cell death, such as via ferroptosis and apoptosis, while lipids also play a crucial role in the regulation of autophagosome formation. An increased autophagic response not only promotes cell survival but also causes cell death depending on the context, especially when selectively degrading antioxidant proteins or organelles that promote ferroptosis. ACSL4 is an enzyme that catalyzes the formation of long-chain acyl-CoA molecules, which are important intermediates in the biosynthesis of various types of lipids. ACSL4 is found in many tissues and is particularly abundant in the brain, liver, and adipose tissue. Dysregulation of ACSL4 is linked to a variety of diseases, including cancer, neurodegenerative disorders, cardiovascular disease, acute kidney injury, and metabolic disorders (such as obesity and non-alcoholic fatty liver disease). In this review, we introduce the structure, function, and regulation of ACSL4; discuss its role in apoptosis, ferroptosis, and autophagy; summarize its pathological function; and explore the potential implications of targeting ACSL4 in the treatment of various diseases.
Collapse
Affiliation(s)
- Fangquan Chen
- DAMP Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 511436, China
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jiao Liu
- DAMP Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 511436, China
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
7
|
Akefe IO, Osborne SL, Matthews B, Wallis TP, Meunier FA. Lipids and Secretory Vesicle Exocytosis. ADVANCES IN NEUROBIOLOGY 2023; 33:357-397. [PMID: 37615874 DOI: 10.1007/978-3-031-34229-5_14] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
In recent years, the number of studies implicating lipids in the regulation of synaptic vesicle exocytosis has risen considerably. It has become increasingly clear that lipids such as phosphoinositides, lysophospholipids, cholesterol, arachidonic acid and myristic acid play critical regulatory roles in the processes leading up to exocytosis. Lipids may affect membrane fusion reactions by altering the physical properties of the membrane, recruiting key regulatory proteins, concentrating proteins into exocytic "hotspots" or by modulating protein functions allosterically. Discrete changes in phosphoinositides concentration are involved in multiple trafficking events including exocytosis and endocytosis. Lipid-modifying enzymes such as the DDHD2 isoform of phospholipase A1 were recently shown to contribute to memory acquisition via dynamic modifications of the brain lipid landscape. Considering the increasing reports on neurodegenerative disorders associated with aberrant intracellular trafficking, an improved understanding of the control of lipid pathways is physiologically and clinically significant and will afford unique insights into mechanisms and therapeutic methods for neurodegenerative diseases. Consequently, this chapter will discuss the different classes of lipids, phospholipase enzymes, the evidence linking them to synaptic neurotransmitter release and how they act to regulate key steps in the multi-step process leading to neuronal communication and memory acquisition.
Collapse
Affiliation(s)
- Isaac O Akefe
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - Shona L Osborne
- ARC Training Centre for Innovation in Biomedical Imaging Technology (CIBIT), The University of Queensland, St Lucia, QLD, Australia
| | - Benjamin Matthews
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - Tristan P Wallis
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - Frédéric A Meunier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia.
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia.
| |
Collapse
|
8
|
Bogie JF, Guns J, Vanherle S. Lipid metabolism in neurodegenerative diseases. CELLULAR LIPID IN HEALTH AND DISEASE 2023:389-419. [DOI: 10.1016/b978-0-323-95582-9.00008-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
9
|
Pinchaud K, Hafeez Z, Auger S, Chatel JM, Chadi S, Langella P, Paoli J, Dary-Mourot A, Maguin-Gaté K, Olivier JL. Impact of Dietary Arachidonic Acid on Gut Microbiota Composition and Gut-Brain Axis in Male BALB/C Mice. Nutrients 2022; 14:nu14245338. [PMID: 36558497 PMCID: PMC9786182 DOI: 10.3390/nu14245338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Although arachidonic acid (ARA) is the precursor of the majority of eicosanoids, its influence as a food component on health is not well known. Therefore, we investigated its impact on the gut microbiota and gut-brain axis. Groups of male BALB/c mice were fed either a standard diet containing 5% lipids (Std-ARA) or 15%-lipid diets without ARA (HL-ARA) or with 1% ARA (HL + ARA) for 9 weeks. Fatty acid profiles of all three diets were the same. The HL-ARA diet favored the growth of Bifidobacterium pseudolongum contrary to the HL + ARA diet that favored the pro-inflammatory Escherichia-Shigella genus in fecal microbiota. Dietary ARA intake induced 4- and 15-fold colic overexpression of the pro-inflammatory markers IL-1β and CD40, respectively, without affecting those of TNFα and adiponectin. In the brain, dietary ARA intake led to moderate overexpression of GFAP in the hippocampus and cortex. Both the hyperlipidic diets reduced IL-6 and IL-12 in the brain. For the first time, it was shown that dietary ARA altered the gut microbiota, led to low-grade colic inflammation, and induced astrogliosis in the brain. Further work is necessary to determine the involved mechanisms.
Collapse
Affiliation(s)
- Katleen Pinchaud
- Calbinotox (UR7488), Université de Lorraine, 54000 Nancy, France
| | - Zeeshan Hafeez
- Calbinotox (UR7488), Université de Lorraine, 54000 Nancy, France
| | - Sandrine Auger
- INRAE, Université Paris-Saclay, AgroParisTech, UMR 1319 Micalis Institute, 78352 Jouy-en-Josas, France
| | - Jean-Marc Chatel
- INRAE, Université Paris-Saclay, AgroParisTech, UMR 1319 Micalis Institute, 78352 Jouy-en-Josas, France
| | - Sead Chadi
- INRAE, Université Paris-Saclay, AgroParisTech, UMR 1319 Micalis Institute, 78352 Jouy-en-Josas, France
| | - Philippe Langella
- INRAE, Université Paris-Saclay, AgroParisTech, UMR 1319 Micalis Institute, 78352 Jouy-en-Josas, France
| | - Justine Paoli
- Calbinotox (UR7488), Université de Lorraine, 54000 Nancy, France
| | | | - Katy Maguin-Gaté
- Calbinotox (UR7488), Université de Lorraine, 54000 Nancy, France
| | - Jean Luc Olivier
- Calbinotox (UR7488), Université de Lorraine, 54000 Nancy, France
- CHRU de Nancy, Pôle des Laboratoires, Service de Biochimie-Biologie Moléculaire-Nutrition, 54000 Nancy, France
- Correspondence:
| |
Collapse
|
10
|
Wu Z, Sun J, Liao Z, Qiao J, Chen C, Ling C, Wang H. An update on the therapeutic implications of long-chain acyl-coenzyme A synthetases in nervous system diseases. Front Neurosci 2022; 16:1030512. [PMID: 36507355 PMCID: PMC9731139 DOI: 10.3389/fnins.2022.1030512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/07/2022] [Indexed: 11/25/2022] Open
Abstract
Long-chain acyl-coenzyme A synthetases (ACSLs) are a family of CoA synthetases that activate fatty acid (FA) with chain lengths of 12-20 carbon atoms by forming the acyl-AMP derivative in an isozyme-specific manner. This family mainly includes five members (ACSL1, ACSL3, ACSL4, ACSL5, and ACSL6), which are thought to have specific and different functions in FA metabolism and oxidative stress of mammals. Accumulating evidence shows that the dysfunction of ACSLs is likely to affect cell proliferation and lead to metabolic diseases in multiple organs and systems through different signaling pathways and molecular mechanisms. Hence, a central theme of this review is to emphasize the therapeutic implications of ACSLs in nervous system disorders.
Collapse
Affiliation(s)
- Zhimin Wu
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jun Sun
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhi Liao
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jia Qiao
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Chuan Chen
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Cong Ling
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hui Wang
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China,*Correspondence: Hui Wang,
| |
Collapse
|
11
|
Fadó R, Molins A, Rojas R, Casals N. Feeding the Brain: Effect of Nutrients on Cognition, Synaptic Function, and AMPA Receptors. Nutrients 2022; 14:nu14194137. [PMID: 36235789 PMCID: PMC9572450 DOI: 10.3390/nu14194137] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 11/05/2022] Open
Abstract
In recent decades, traditional eating habits have been replaced by a more globalized diet, rich in saturated fatty acids and simple sugars. Extensive evidence shows that these dietary factors contribute to cognitive health impairment as well as increase the incidence of metabolic diseases such as obesity and diabetes. However, how these nutrients modulate synaptic function and neuroplasticity is poorly understood. We review the Western, ketogenic, and paleolithic diets for their effects on cognition and correlations with synaptic changes, focusing mainly (but not exclusively) on animal model studies aimed at tracing molecular alterations that may contribute to impaired human cognition. We observe that memory and learning deficits mediated by high-fat/high-sugar diets, even over short exposure times, are associated with reduced arborization, widened synaptic cleft, narrowed post-synaptic zone, and decreased activity-dependent synaptic plasticity in the hippocampus, and also observe that these alterations correlate with deregulation of the AMPA-type glutamate ionotropic receptors (AMPARs) that are crucial to neuroplasticity. Furthermore, we explored which diet-mediated mechanisms modulate synaptic AMPARs and whether certain supplements or nutritional interventions could reverse deleterious effects, contributing to improved learning and memory in older people and patients with Alzheimer’s disease.
Collapse
Affiliation(s)
- Rut Fadó
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, E-08195 Sant Cugat del Vallès, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
- Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, E-08193 Cerdanyola del Vallès, Spain
- Correspondence: ; Tel.: +34-93-504-20-00
| | - Anna Molins
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, E-08195 Sant Cugat del Vallès, Spain
| | - Rocío Rojas
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, E-08195 Sant Cugat del Vallès, Spain
| | - Núria Casals
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, E-08195 Sant Cugat del Vallès, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| |
Collapse
|
12
|
Ma H, Dong Y, Chu Y, Guo Y, Li L. The mechanisms of ferroptosis and its role in alzheimer’s disease. Front Mol Biosci 2022; 9:965064. [PMID: 36090039 PMCID: PMC9459389 DOI: 10.3389/fmolb.2022.965064] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 07/25/2022] [Indexed: 12/06/2022] Open
Abstract
Alzheimer’s disease (AD) accounts for two-thirds of all dementia cases, affecting 50 million people worldwide. Only four of the more than 100 AD drugs developed thus far have successfully improved AD symptoms. Furthermore, these improvements are only temporary, as no treatment can stop or reverse AD progression. A growing number of recent studies have demonstrated that iron-dependent programmed cell death, known as ferroptosis, contributes to AD-mediated nerve cell death. The ferroptosis pathways within nerve cells include iron homeostasis regulation, cystine/glutamate (Glu) reverse transporter (system xc−), glutathione (GSH)/glutathione peroxidase 4 (GPX4), and lipid peroxidation. In the regulation pathway of AD iron homeostasis, abnormal iron uptake, excretion and storage in nerve cells lead to increased intracellular free iron and Fenton reactions. Furthermore, decreased Glu transporter expression leads to Glu accumulation outside nerve cells, resulting in the inhibition of the system xc− pathway. GSH depletion causes abnormalities in GPX4, leading to excessive accumulation of lipid peroxides. Alterations in these specific pathways and amino acid metabolism eventually lead to ferroptosis. This review explores the connection between AD and the ferroptosis signaling pathways and amino acid metabolism, potentially informing future AD diagnosis and treatment methodologies.
Collapse
Affiliation(s)
- Hongyue Ma
- Department of Neurology, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, China
| | - Yan Dong
- Department of Neurology, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, China
| | - Yanhui Chu
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, China
| | - Yanqin Guo
- Department of Neurology, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, China
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, China
- *Correspondence: Yanqin Guo, ; Luxin Li,
| | - Luxin Li
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, China
- *Correspondence: Yanqin Guo, ; Luxin Li,
| |
Collapse
|
13
|
Dietary fatty acids affect learning and memory ability via regulating inflammatory factors in obese mice. J Nutr Biochem 2022; 103:108959. [PMID: 35158028 DOI: 10.1016/j.jnutbio.2022.108959] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 01/10/2022] [Accepted: 01/19/2022] [Indexed: 12/17/2022]
Abstract
OBJECTIVE High-fat diets are linked to obesity, contributing to the alterations in inflammatory signaling pathways, which is associated with cognitive function. We aim to investigate the mechanisms by which various different types of dietary fatty acids affecting cognitive function in obese mice through the gut/brain axis-inflammatory signaling pathway. METHODS AND RESULTS Eight-week-old male C57BL/6 mice were fed with basal diet (control group), lard high-fat diet (containing long-chain saturated fatty acid (LCSFA group)), coconut oil high-fat diet (containing medium-chain saturated fatty acid (MCSFA group)), linseed oil high-fat diet (containing n-3 polyunsaturated fatty acid (n-3 PUFA group)), soybean oil high-fat diet (containing n-6 polyunsaturated fatty acid (n-6 PUFA group)), olive oil high-fat diet (containing monounsaturated fatty acid (MUFA group)) and 8% hydrogenated soybean oil high-fat diet (containing trans fatty acid (TFA group)) respectively for 16 weeks. Our results revealed that the mean escape latency was significantly prolonged in LCSFA group, and the latency to cross the platform location of n-6 PUFA and TFA groups were increased significantly. The differences of inflammatory markers and toll-like receptor-myeloid differentiation factor-88-nuclear factor kappa-B (TLR-MyD88-NF-κB) inflammatory signaling pathway expressions among all groups reached statistical significances. CONCLUSION Compared to basal diet, high-fat diets enriched in LCSFA, MCSFA, n-6 PUFA, MUFA, and TFA might exert detrimental effects on cognitive function in obese mice via regulating the inflammatory markers and inflammatory signaling pathway in brain and intestine. High-fat diet enriched in n-3 PUFA might exhibit different effect on modulating inflammatory responses in different tissues and might benefit to cognitive function.
Collapse
|
14
|
Shao Y, Ouyang Y, Li T, Liu X, Xu X, Li S, Xu G, Le W. Alteration of Metabolic Profile and Potential Biomarkers in the Plasma of Alzheimer's Disease. Aging Dis 2020; 11:1459-1470. [PMID: 33269100 PMCID: PMC7673846 DOI: 10.14336/ad.2020.0217] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 02/17/2020] [Indexed: 12/23/2022] Open
Abstract
The expending of elderly population worldwide has resulted in a dramatic rise in the incidence of chronic diseases such as Alzheimer's disease (AD). Inadequate understanding of the mechanisms underlying AD has hampered the development of efficient tools for definitive diagnosis and curative interventions. Previous studies have attempted to discover reliable biomarkers of AD, but these biomarkers can only be measured through invasive (neuropathological markers in cerebrospinal fluid) or expensive (positron emission tomography scanning or magnetic resonance imaging) techniques. Metabolomics is a high-throughput technology that can detect and catalog large numbers of small metabolites and may be a useful tool for characterization of AD and identification of biomarkers. In this study, we used ultra-performance liquid chromatography-mass spectrometry based untargeted metabolomics to measure the concentrations of plasma metabolites in a cohort of subjects with AD (n=44) and cognitively normal controls (Ctrl, n=94). The AD group showed marked reductions in levels of polyunsaturated fatty acids, acyl-carnitines, degradation products of tryptophan, and elevated levels of bile acids compared to the Ctrl group. We then validated the results using an independent cohort that included subjects with AD (n=30), mild cognitive impairment (MCI, n=13), healthy controls (n=43), and non-AD neurological disease controls (NDC, n=31). We identified five metabolites comprising cholic acid, chenodeoxycholic acid, allocholic acid, indolelactic acid, and tryptophan that were able to distinguish patients with AD from both Ctrl and NDC with satisfactory sensitivity and specificity. The concentrations of these metabolites were significantly correlated with disease severity. Our results also suggested that altered bile acid profiles in AD and MCI might indicate early risk for the development of AD. These findings may allow for development of new approaches for diagnosis of AD and may provide novel insights into AD pathogenesis.
Collapse
Affiliation(s)
- Yaping Shao
- Center for Clinical Research on Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China.
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China.
| | - Yang Ouyang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China.
- University of Chinese Academy of Sciences, Beijing, China
| | - Tianbai Li
- Center for Clinical Research on Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China.
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China.
| | - Xinyao Liu
- Center for Clinical Research on Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China.
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China.
| | - Xiaojiao Xu
- Center for Clinical Research on Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China.
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China.
| | - Song Li
- Center for Clinical Research on Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China.
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China.
| | - Guowang Xu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China.
| | - Weidong Le
- Center for Clinical Research on Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China.
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China.
| |
Collapse
|
15
|
O'Connor M, Shentu YP, Wang G, Hu WT, Xu ZD, Wang XC, Liu R, Man HY. Acetylation of AMPA Receptors Regulates Receptor Trafficking and Rescues Memory Deficits in Alzheimer's Disease. iScience 2020; 23:101465. [PMID: 32861999 PMCID: PMC7476873 DOI: 10.1016/j.isci.2020.101465] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 07/21/2020] [Accepted: 08/13/2020] [Indexed: 12/26/2022] Open
Abstract
In Alzheimer's disease (AD), decreases in the amount and synaptic localization of AMPA receptors (AMPARs) result in weakened synaptic activity and dysfunction in synaptic plasticity, leading to impairments in cognitive functions. We have previously found that AMPARs are subject to lysine acetylation, resulting in higher AMPAR stability and protein accumulation. Here we report that AMPAR acetylation was significantly reduced in AD and neurons with Aβ incubation. We identified p300 as the acetyltransferase responsible for AMPAR acetylation and found that enhancing GluA1 acetylation ameliorated Aβ-induced reductions in total and cell-surface AMPARs. Importantly, expression of acetylation mimetic GluA1 (GluA1-4KQ) in APP/PS1 mice rescued impairments in synaptic plasticity and memory. These findings indicate that Aβ-induced reduction in AMPAR acetylation and stability contributes to synaptopathy and memory deficiency in AD, suggesting that AMPAR acetylation may be an effective molecular target for AD therapeutics.
Collapse
Affiliation(s)
- Margaret O'Connor
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - Yang-Ping Shentu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Pathology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guan Wang
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - Wen-Ting Hu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhen-Dong Xu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiao-Chuan Wang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Rong Liu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Heng-Ye Man
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, 72 East Concord St., L-603, Boston, MA 02118, USA
- Center for Systems Neuroscience, Boston University, 610 Commonwealth Avenue, Boston, MA, USA
| |
Collapse
|
16
|
Hosseini M, Poljak A, Braidy N, Crawford J, Sachdev P. Blood fatty acids in Alzheimer's disease and mild cognitive impairment: A meta-analysis and systematic review. Ageing Res Rev 2020; 60:101043. [PMID: 32194194 DOI: 10.1016/j.arr.2020.101043] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 12/23/2019] [Accepted: 03/05/2020] [Indexed: 12/31/2022]
Abstract
Plasma fatty acids have been reported to be dysregulated in mild cognitive impairment (MCI) and Alzheimer's disease (AD), though outcomes are not always consistent, and subject numbers often small. Our aim was to use a meta-analysis and systematic review approach to identify if plasma fatty acid dysregulation would be observed in case control studies of AD and MCI. Six databases were searched for studies reporting quantified levels of fatty acids in MCI and/or AD individuals, relative to cognitively normal controls. Docosahexaenoic (DHA) and vaccenic acids were significantly lower and higher respectively in MCI relative to controls. Total fatty acids were 27.2% lower in AD relative to controls, and this was reflected almost uniformly in all specific fatty acids in AD. Changes to plasma/serum fatty acids were identified in both MCI and AD relative to age and gender matched controls. Differences were greatest in AD, in both total number of fatty acids significantly altered, and the degree of change. Docosahexaenoic acid was lower in both MCI and AD, suggesting that it may be a driver of pathology.
Collapse
Affiliation(s)
- Mahboobeh Hosseini
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia.
| | - Anne Poljak
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia; Mark Wainwright Analytical Centre, Bioanalytical Mass Spectrometry Facility, University of New South Wales, Sydney, Australia; School of Medical Sciences, University of New South Wales, Sydney, Australia.
| | - Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia.
| | - John Crawford
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia.
| | - Perminder Sachdev
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia; Neuropsychiatric Institute, Euroa Centre, Prince of Wales Hospital, Sydney, Australia.
| |
Collapse
|
17
|
Fatty acid metabolism in the progression and resolution of CNS disorders. Adv Drug Deliv Rev 2020; 159:198-213. [PMID: 31987838 DOI: 10.1016/j.addr.2020.01.004] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/13/2020] [Accepted: 01/23/2020] [Indexed: 12/15/2022]
Abstract
Recent advances in lipidomics and metabolomics have unveiled the complexity of fatty acid metabolism and the fatty acid lipidome in health and disease. A growing body of evidence indicates that imbalances in the metabolism and level of fatty acids drive the initiation and progression of central nervous system (CNS) disorders such as multiple sclerosis, Alzheimer's disease, and Parkinson's disease. Here, we provide an in-depth overview on the impact of the β-oxidation, synthesis, desaturation, elongation, and peroxidation of fatty acids on the pathophysiology of these and other neurological disorders. Furthermore, we discuss the impact of individual fatty acids species, acquired through the diet or endogenously synthesized in mammals, on neuroinflammation, neurodegeneration, and CNS repair. The findings discussed in this review highlight the therapeutic potential of modulators of fatty acid metabolism and the fatty acid lipidome in CNS disorders, and underscore the diagnostic value of lipidome signatures in these diseases.
Collapse
|
18
|
The effects of omega-3 fatty acid deficiency during development on oxidative fatty acid degradation during maturity in a mouse model of Alzheimer's disease. Neurobiol Aging 2019; 79:66-74. [PMID: 31029017 DOI: 10.1016/j.neurobiolaging.2019.03.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 02/25/2019] [Accepted: 03/05/2019] [Indexed: 12/13/2022]
Abstract
Metabolic conditions during brain development may have long-term consequences on brain metabolism, thereby influencing the risk of neurodegenerative disease in later life. To ascertain the long-term consequences of omega-3 (ω3) fatty acid deficiency during brain development on oxidative fatty acid degradation in the brain and the development of Alzheimer-like pathology, wild-type (WT) female mice were fed diets that were either replete or deficient in ω3 fatty acids for 5 weeks. These females were then mated with hemizygous 5xFAD male transgenic (TG) mouse models of Alzheimer's disease, and the progeny were continued on diets that were either ω3-replete or ω3-deficient. When the progeny were 6 months of age, they received radiolabeled arachidonic acid (ARA) by intracerebroventricular injection. Five days after these injections, the brains were harvested and oxidative degradation of the radiolabeled ARA was characterized. Among the progeny of female mice on an ω3-replete diet, TG progeny had lower PSD-95 expression and higher oxidative ARA degradation than WT progeny. Progeny on an ω3-deficient diet, however, had no significant differences in PSD-95 expression between TG and WT mice, or in the extent of ARA degradation. In TG mice, an ω3-deficient diet reduced oxidative ARA degradation to a greater extent than in WT mice. The reductions in oxidative ARA degradation occurred even if the progeny of female mice on an ω3-deficient diet resumed an ω3-replete diet immediately on weaning. These results demonstrate that dietary ω3 fatty acid deficiency during development can cause long-term changes in the expression of a synaptic marker and long-term reductions in the rate of ARA degradation in the WT brain, which are not completely alleviated by an ω3-replete diet after weaning. The elimination of differences between TG and WT mice by an ω3-deficient diet suggests that mechanisms regulating PSD-95 expression and the oxidative degradation of ARA are related and that the timing of dietary ω3 intake during development may influence Alzheimer's disease-related pathological changes later in life.
Collapse
|
19
|
Maternal Dietary Docosahexaenoic Acid Alters Lipid Peroxidation Products and (n-3)/(n-6) Fatty Acid Balance in Offspring Mice. Metabolites 2019; 9:metabo9030040. [PMID: 30832208 PMCID: PMC6468482 DOI: 10.3390/metabo9030040] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 02/24/2019] [Accepted: 02/26/2019] [Indexed: 12/11/2022] Open
Abstract
The abundance of docosahexaenoic acid (DHA) in the mammalian brain has generated substantial interest in the search for its roles in regulating brain functions. Our recent study with a gene/stress mouse model provided evidence to support the ability for the maternal supplement of DHA to alleviate autism-associated behavior in the offspring. DHA and arachidonic acid (ARA) are substrates of enzymatic and non-enzymatic reactions, and lipid peroxidation results in the production of 4-hydroxyhexenal (4-HHE) and 4-hydroxynonenal (4-HNE), respectively. In this study, we examine whether a maternal DHA-supplemented diet alters fatty acids (FAs), as well as lipid peroxidation products in the pup brain, heart and plasma by a targeted metabolite approach. Pups in the maternal DHA-supplemented diet group showed an increase in DHA and a concomitant decrease in ARA in all brain regions examined. However, significant increases in 4-HHE, and not 4-HNE, were found mainly in the cerebral cortex and hippocampus. Analysis of heart and plasma showed large increases in DHA and 4-HHE, but a significant decrease in 4-HNE levels only in plasma. Taken together, the DHA-supplemented maternal diet alters the (n-3)/(n-6) FA ratio, and increases 4-HHE levels in pup brain, heart and plasma. These effects may contribute to the beneficial effects of DHA on neurodevelopment, as well as functional changes in other body organs.
Collapse
|
20
|
Liang J, Wu Y, Yuan H, Yang Y, Xiong Q, Liang C, Li Z, Li C, Zhang G, Lai X, Hu Y, Hou S. Dendrobium officinale polysaccharides attenuate learning and memory disabilities via anti-oxidant and anti-inflammatory actions. Int J Biol Macromol 2018; 126:414-426. [PMID: 30593810 DOI: 10.1016/j.ijbiomac.2018.12.230] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 12/14/2018] [Accepted: 12/22/2018] [Indexed: 12/29/2022]
Abstract
The aim of this study was to explore the therapeutic effect and underling mechanism of Dendrobium officinale polysaccharides (DOPS) on two well-established animal models of learning and memory disabilities. Model of estrogen deficiency caused learning and memory disability can be induced by ovariectomy in mice, and mice were injected subcutaneously with d-galactose, which can also cause cognitive decline. H&E staining and Nissl staining were employed to confirm the protective effect of DOPS on hippocampal neuron. Morris water maze test, biochemical analysis, immunohistochemistry and immunofluorescence assay were used to study the effect and underlying mechanism of DOPS on two different learning and memory impairment models. Administration of DOPS significantly improved learning and memory disability in both models. Further studies showed that DOPS could attenuate oxidative stress and reduce neuro-inflammation via up-regulating expressions of Nrf2/HO-1 pathway and inhibiting activation of astrocytes and microglia in ovariectomy- and d-galactose-induced cognitive decline. These findings suggest that DOPS have an appreciable therapeutic effect on learning and memory disabilities and its mechanism may be related to activate Nrf2/HO-1 pathway to reduce oxidative stress and neuro-inflammation.
Collapse
Affiliation(s)
- Jian Liang
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, and Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, China
| | - Yanfang Wu
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, and Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, China
| | - Han Yuan
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, and Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, China
| | - Yiqi Yang
- The second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, China
| | - Qingping Xiong
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, and Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, China
| | - Chuyan Liang
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510080, Guangdong, China
| | - Zhimeng Li
- The Fifth People's Hospital of Tangshan, Tangshan 063004, Hebei, PR China
| | - Cantao Li
- School of pharmaceutical sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, China
| | - Guifang Zhang
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, and Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, China
| | - Xiaoping Lai
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, and Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, China.
| | - Youdong Hu
- Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an 223002, Jiangsu, PR China.
| | - Shaozhen Hou
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, and Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, China; School of pharmaceutical sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, China.
| |
Collapse
|