1
|
Singh MK, Ryu TH, Nguyen MN, Yu K. Inhibition of high-fat diet-induced miRNA ameliorates tau toxicity in Drosophila. Biochem Biophys Res Commun 2024; 733:150446. [PMID: 39067249 DOI: 10.1016/j.bbrc.2024.150446] [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: 07/02/2024] [Revised: 07/16/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
Alzheimer's disease (AD), caused by amyloid beta (Aβ) plaques and Tau tangles, is a neurodegenerative disease characterized by progressive memory impairment and cognitive dysfunction. High-fat diet (HFD), which induces type 2 diabetes, exacerbates Aβ plaque deposition in the brain. To investigate the function of HFD in Tau-mediated AD, we fed an HFD to the Drosophila Tau model and found that HFD aggravates Tau-induced neurological phenotypes. Since microRNAs (miRNAs) are biomarkers for diabetes and AD, we evaluated the expression levels of common miRNAs of HFD and AD in HFD-fed Tau model fly brains. Among the common miRNAs, the expression levels of Let-7 and miR-34 were increased. We found that the inhibition of these miRNAs alleviates Tau-mediated AD phenotypes. Our research provides valuable insights into how HFD accelerates tau toxicity. Additionally, our work highlights the therapeutic potential of targeting Let-7 and miR-34 to develop innovative treatment approaches for AD.
Collapse
Affiliation(s)
- Manish Kumar Singh
- Metabolism and Neurophysiology Research Group, Disease Target Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Tae Hoon Ryu
- Metabolism and Neurophysiology Research Group, Disease Target Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Minh Nguyet Nguyen
- Metabolism and Neurophysiology Research Group, Disease Target Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea; Department of Bioscience, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Kweon Yu
- Metabolism and Neurophysiology Research Group, Disease Target Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea; Department of Bioscience, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea.
| |
Collapse
|
2
|
Allerton TD, Stampley JE, Li Z, Yu X, Quiariate H, Doiron JE, White G, Wigger Z, Gartia MR, Lefer DJ, Soto P, Irving BA. Nitric oxide donors rescue metabolic and mitochondrial dysfunction in obese Alzheimer's model. Sci Rep 2024; 14:26118. [PMID: 39478095 PMCID: PMC11525932 DOI: 10.1038/s41598-024-75870-8] [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: 03/12/2024] [Accepted: 10/08/2024] [Indexed: 11/02/2024] Open
Abstract
Reduced nitric oxide (NO) bioavailability is a pathological link between obesity and Alzheimer's disease (AD). Obesity-associated metabolic and mitochondrial bioenergetic dysfunction are key drivers of AD pathology. The hypothalamus is a critical brain region during the development of obesity and dysfunction is an area implicated in the development of AD. NO is an essential mediator of blood flow and mitochondrial bioenergetic function, but the role of NO in obesity-AD is not entirely clear. We investigated diet-induced obesity in female APPswe/PS1dE9 (APP) mouse model of AD, which we treated with two different NO donors (sodium nitrite or L-citrulline). After 26 weeks of a high-fat diet, female APP mice had higher adiposity, insulin resistance, and mitochondrial dysfunction (hypothalamus) than non-transgenic littermate (wild type) controls. Treatment with either sodium nitrite or L-citrulline did not reduce adiposity but improved whole-body energy expenditure, substrate oxidation, and insulin sensitivity. Notably, both NO donors restored hypothalamic mitochondrial respiration in APP mice. Our findings suggest that NO is an essential mediator of whole-body metabolism and hypothalamic mitochondrial function, which are severely impacted by the dual insults of obesity and AD pathology.
Collapse
Affiliation(s)
- Timothy D Allerton
- Vascular Metabolism Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA.
| | - James E Stampley
- Department of Kinesiology, Louisiana State University, Baton Rouge, LA, USA
| | - Zhen Li
- Department of Cardiac Surgery, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Xiaoman Yu
- Department of Cardiac Surgery, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Heather Quiariate
- Vascular Metabolism Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
- Department of Kinesiology, Louisiana State University, Baton Rouge, LA, USA
| | - Jake E Doiron
- Vascular Metabolism Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Ginger White
- Department of Psychology, Louisiana State University, Baton Rouge, LA, USA
| | - Zach Wigger
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Manas Ranjan Gartia
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA, USA
| | - David J Lefer
- Department of Cardiac Surgery, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Paul Soto
- Department of Psychology, Louisiana State University, Baton Rouge, LA, USA
| | - Brian A Irving
- Department of Kinesiology, Louisiana State University, Baton Rouge, LA, USA
| |
Collapse
|
3
|
Babalola JA, Stracke A, Loeffler T, Schilcher I, Sideromenos S, Flunkert S, Neddens J, Lignell A, Prokesch M, Pazenboeck U, Strobl H, Tadic J, Leitinger G, Lass A, Hutter-Paier B, Hoefler G. Effect of astaxanthin in type-2 diabetes -induced APPxhQC transgenic and NTG mice. Mol Metab 2024; 85:101959. [PMID: 38763496 PMCID: PMC11153249 DOI: 10.1016/j.molmet.2024.101959] [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: 02/19/2024] [Revised: 05/09/2024] [Accepted: 05/15/2024] [Indexed: 05/21/2024] Open
Abstract
OBJECTIVES Aggregation and misfolding of amyloid beta (Aβ) and tau proteins, suggested to arise from post-translational modification processes, are thought to be the main cause of Alzheimer's disease (AD). Additionally, a plethora of evidence exists that links metabolic dysfunctions such as obesity, type 2 diabetes (T2D), and dyslipidemia to the pathogenesis of AD. We thus investigated the combinatory effect of T2D and human glutaminyl cyclase activity (pyroglutamylation), on the pathology of AD and whether astaxanthin (ASX) treatment ameliorates accompanying pathophysiological manifestations. METHODS Male transgenic AD mice, APPxhQC, expressing human APP751 with the Swedish and the London mutation and human glutaminyl cyclase (hQC) enzyme and their non-transgenic (NTG) littermates were used. Both APPxhQC and NTG mice were allocated to 3 groups, control, T2D-control, and T2D-ASX. Mice were fed control or high fat diet ± ASX for 13 weeks starting at an age of 11-12 months. High fat diet fed mice were further treated with streptozocin for T2D induction. Effects of genotype, T2D induction, and ASX treatment were evaluated by analysing glycemic readouts, lipid concentration, Aβ deposition, hippocampus-dependent cognitive function and nutrient sensing using immunosorbent assay, ELISA-based assays, western blotting, immunofluorescence staining, and behavioral testing via Morris water maze (MWM), respectively. RESULTS APPxhQC mice presented a higher glucose sensitivity compared to NTG mice. T2D-induced brain dysfunction was more severe in NTG compared to the APPxhQC mice. T2D induction impaired memory functions while increasing hepatic LC3B, ABCA1, and p65 levels in NTG mice. T2D induction resulted in a progressive shift of Aβ from the soluble to insoluble form in APPxhQC mice. ASX treatment reversed T2D-induced memory dysfunction in NTG mice and in parallel increased hepatic pAKT while decreasing p65 and increasing cerebral p-S6rp and p65 levels. ASX treatment reduced soluble Aβ38 and Aβ40 and insoluble Aβ40 levels in T2D-induced APPxhQC mice. CONCLUSIONS We demonstrate that T2D induction in APPxhQC mice poses additional risk for AD pathology as seen by increased Aβ deposition. Although ASX treatment reduced Aβ expression in T2D-induced APPxhQC mice and rescued T2D-induced memory impairment in NTG mice, ASX treatment alone may not be effective in cases of T2D comorbidity and AD.
Collapse
Affiliation(s)
| | - Anika Stracke
- Division of Immunology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Austria
| | | | | | - Spyridon Sideromenos
- QPS Austria GmbH, Grambach, Austria; Medical University of Vienna, Vienna, Austria
| | | | | | | | | | - Ute Pazenboeck
- Division of Immunology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Austria
| | - Herbert Strobl
- Division of Immunology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Austria
| | - Jelena Tadic
- Institute of Molecular Biosciences, University of Graz, Austria
| | - Gerd Leitinger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | - Achim Lass
- Institute of Molecular Biosciences, University of Graz, Austria
| | | | - Gerald Hoefler
- Diagnostic and Research Institute of Pathology Medical University of Graz, Graz, Austria.
| |
Collapse
|
4
|
do Nascimento FV, de Freitas BS, Dos Passos MP, Kleverston L, de Souza Dos Santos C, Kist LW, Bogo MR, Bromberg E, Schröder N. A high fat diet potentiates neonatal iron overload-induced memory impairments in rats. Eur J Nutr 2024; 63:1163-1175. [PMID: 38358514 DOI: 10.1007/s00394-024-03333-x] [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: 06/06/2023] [Accepted: 01/23/2024] [Indexed: 02/16/2024]
Abstract
PURPOSE The present study aimed at evaluating possible synergistic effects between two risk factors for cognitive decline and neurodegenerative disorders, i.e. iron overload and exposure to a hypercaloric/hyperlipidic diet, on cognition, insulin resistance, and hippocampal GLUT1, GLUT3, Insr mRNA expression, and AKT phosporylation. METHODS Male Wistar rats were treated with iron (30 mg/kg carbonyl iron) or vehicle (5% sorbitol in water) from 12 to 14th post-natal days. Iron-treated rats received a standard laboratory diet or a high fat diet from weaning to adulthood (9 months of age). Recognition and emotional memory, peripheral blood glucose and insulin levels were evaluated. Glucose transporters (GLUT 1 and GLUT3) and insulin signaling were analyzed in the hippocampus of rats. RESULTS Both iron overload and exposure to a high fat diet induced memory deficits. Remarkably, the association of iron with the high fat diet induced more severe cognitive deficits. Iron overload in the neonatal period induced higher insulin levels associated with significantly higher HOMA-IR, an index of insulin resistance. Long-term exposure to a high fat diet resulted in higher fasting glucose levels. Iron treatment induced changes in Insr and GLUT1 expression in the hippocampus. At the level of intracellular signaling, both iron treatment and the high fat diet decreased AKT phosphorylation. CONCLUSION The combination of iron overload with exposure to a high fat diet only led to synergistic deleterious effect on emotional memory, while the effects induced by iron and by the high fat diet on AKT phosphorylation were comparable. These findings indicate that there is, at least to some extent, an additive effect of iron combined with the diet. Further studies investigating the mechanisms associated to deleterious effects on cognition and susceptibility for the development of age-associated neurodegenerative disorders are warranted.
Collapse
Affiliation(s)
- Filipe Valvassori do Nascimento
- Neurobiology and Developmental Biology Laboratory, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Betânia Souza de Freitas
- Neurobiology and Developmental Biology Laboratory, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Maiara Priscila Dos Passos
- Neurobiology and Developmental Biology Laboratory, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Luiza Kleverston
- Neurobiology and Developmental Biology Laboratory, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Cristophod de Souza Dos Santos
- Neurobiology and Developmental Biology Laboratory, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Luiza Wilges Kist
- Laboratory of Genomics and Molecular Biology, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
- Excitotoxicity and Neuroprotection (INCT-EN), National Institute of Science and Technology for Brain Diseases, Porto Alegre, Brazil
| | - Maurício Reis Bogo
- Laboratory of Genomics and Molecular Biology, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
- Excitotoxicity and Neuroprotection (INCT-EN), National Institute of Science and Technology for Brain Diseases, Porto Alegre, Brazil
| | - Elke Bromberg
- Neurobiology and Developmental Biology Laboratory, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
- National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brasília, Brazil
| | - Nadja Schröder
- National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brasília, Brazil.
- Department of Physiology, Institute for Basic Health Sciences, Federal University of Rio Grande do Sul, Rua Ramiro Barcelos, 2600, Porto Alegre, RS, 90035-003, Brazil.
| |
Collapse
|
5
|
Lai AY, Almanza DLV, Ribeiro JA, Hill ME, Mandrozos M, Koletar MM, Stefanovic B, McLaurin J. Obesity Facilitates Sex-Specific Improvement In Cognition And Neuronal Function In A Rat Model Of Alzheimer's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.11.575200. [PMID: 38328066 PMCID: PMC10849478 DOI: 10.1101/2024.01.11.575200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Obesity reduces or increases the risk of developing Alzheimer's disease (AD) depending on whether it is assessed in mid-life or late-life. There is currently no consensus on the relationship between obesity and AD or the mechanism or their interaction. Here, we aim to differentiate the cause-and-effect relationship between obesity and AD in a controlled rat model of AD. We induced obesity in 9-month-old TgF344-AD rats, that is pathology-load wise similar to early symptomatic phase of human AD. To more accurately model human obesity, we fed both TgF344-AD and non-transgenic littermates a varied high-carbohydrate-high-fat diet consisting of human food for 3 months. Obesity increased overall glucose metabolism and slowed cognitive decline in TgF344-AD rats, specifically executive function, without affecting non-transgenic rats. Pathological analyses of prefrontal cortex and hippocampus showed that obesity in TgF344-AD rats produced varied effects, with increased density of myelin and oligodendrocytes, lowered density and activation of microglia that we propose contributes to the cognitive improvement. However, obesity also decreased neuronal density, and promoted deposition of amyloid-beta plaques and tau inclusions. After 6 months on the high-carbohydrate-high-fat diet, detrimental effects on density of neurons, amyloid-beta plaques, and tau inclusions persisted while the beneficial effects on myelin, microglia, and cognitive functions remained albeit with a lower effect size. By examining the effect of sex, we found that both beneficial and detrimental effects of obesity were stronger in female TgF344-AD rats indicating that obesity during early symptomatic phase of AD is protective in females.
Collapse
|
6
|
Valentin-Escalera J, Leclerc M, Calon F. High-Fat Diets in Animal Models of Alzheimer's Disease: How Can Eating Too Much Fat Increase Alzheimer's Disease Risk? J Alzheimers Dis 2024; 97:977-1005. [PMID: 38217592 PMCID: PMC10836579 DOI: 10.3233/jad-230118] [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] [Accepted: 11/15/2023] [Indexed: 01/15/2024]
Abstract
High dietary intake of saturated fatty acids is a suspected risk factor for neurodegenerative diseases, including Alzheimer's disease (AD). To decipher the causal link behind these associations, high-fat diets (HFD) have been repeatedly investigated in animal models. Preclinical studies allow full control over dietary composition, avoiding ethical concerns in clinical trials. The goal of the present article is to provide a narrative review of reports on HFD in animal models of AD. Eligibility criteria included mouse models of AD fed a HFD defined as > 35% of fat/weight and western diets containing > 1% cholesterol or > 15% sugar. MEDLINE and Embase databases were searched from 1946 to August 2022, and 32 preclinical studies were included in the review. HFD-induced obesity and metabolic disturbances such as insulin resistance and glucose intolerance have been replicated in most studies, but with methodological variability. Most studies have found an aggravating effect of HFD on brain Aβ pathology, whereas tau pathology has been much less studied, and results are more equivocal. While most reports show HFD-induced impairment on cognitive behavior, confounding factors may blur their interpretation. In summary, despite conflicting results, exposing rodents to diets highly enriched in saturated fat induces not only metabolic defects, but also cognitive impairment often accompanied by aggravated neuropathological markers, most notably Aβ burden. Although there are important variations between methods, particularly the lack of diet characterization, these studies collectively suggest that excessive intake of saturated fat should be avoided in order to lower the incidence of AD.
Collapse
Affiliation(s)
- Josue Valentin-Escalera
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du centre Hospitalier de l'Université Laval (CHUL), Québec, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, Québec, Canada
- OptiNutriBrain - Laboratoire International Associé (NutriNeuro France-INAF Canada)
| | - Manon Leclerc
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du centre Hospitalier de l'Université Laval (CHUL), Québec, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, Québec, Canada
- OptiNutriBrain - Laboratoire International Associé (NutriNeuro France-INAF Canada)
| | - Frédéric Calon
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du centre Hospitalier de l'Université Laval (CHUL), Québec, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, Québec, Canada
- OptiNutriBrain - Laboratoire International Associé (NutriNeuro France-INAF Canada)
| |
Collapse
|
7
|
Abi-Ghanem C, Salinero AE, Smith RM, Kelly RD, Belanger KM, Richard RN, Paul AS, Herzog AA, Thrasher CA, Rybka KA, Riccio D, Gannon OJ, Kordit D, Kyaw NR, Mansour FM, Groom E, Brooks HL, Robison LS, Pumiglia K, Zuloaga DG, Zuloaga KL. Effects of Menopause and High Fat Diet on Metabolic Outcomes in a Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2024; 101:1177-1194. [PMID: 39302361 DOI: 10.3233/jad-231332] [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: 09/22/2024]
Abstract
Background About two-thirds of those with Alzheimer's disease (AD) are women, most of whom are post-menopausal. Menopause accelerates dementia risk by increasing the risk for metabolic, cardiovascular, and cerebrovascular diseases. Mid-life metabolic disease (obesity, diabetes/prediabetes) is a well-known risk factor for dementia. A high fat diet can lead to poor metabolic health in both humans and rodents. Objective Our goal was to determine the effects of a high fat diet on metabolic outcomes in the AppNL-F knock-in mouse model of AD and assess the effects of menopause. Methods First, 3-month-old AppNL-F and WT female mice were placed on either a control or a high fat diet until 10 months of age then assessed for metabolic outcomes. Next, we did a more extensive assessment in AppNL-F mice that were administered VCD (4-vinylcyclohexene diepoxide) or vehicle (oil) and placed on a control or high fat diet for 7 months. VCD was used to model menopause by causing accelerated ovarian failure. Results Compared to WT controls, AD female mice had worse glucose intolerance. Menopause led to metabolic impairment (weight gain and glucose intolerance) and further exacerbated obesity in response to a high fat diet. There were interactions between diet and menopause on some metabolic health serum biomarkers and the expression of hypothalamic markers related to energy balance. Conclusions This work highlights the need to model endocrine aging in animal models of dementia and will contribute to further understanding the interaction between menopause and metabolic health in the context of AD.
Collapse
Affiliation(s)
- Charly Abi-Ghanem
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Abigail E Salinero
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Rachel M Smith
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Richard D Kelly
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Kasey M Belanger
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Riane N Richard
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Aaron S Paul
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Ava A Herzog
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Christina A Thrasher
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Krystyna A Rybka
- Department of Psychology and Center for Neuroscience Research, State University of New York at Albany, Albany, NY, USA
| | - David Riccio
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Olivia J Gannon
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - David Kordit
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Nyi-Rein Kyaw
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Febronia M Mansour
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Emily Groom
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Heddwen L Brooks
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Lisa S Robison
- Department of Psychology and Neuroscience, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Kevin Pumiglia
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, USA
| | - Damian G Zuloaga
- Department of Psychology and Center for Neuroscience Research, State University of New York at Albany, Albany, NY, USA
| | - Kristen L Zuloaga
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| |
Collapse
|
8
|
Neto A, Fernandes A, Barateiro A. The complex relationship between obesity and neurodegenerative diseases: an updated review. Front Cell Neurosci 2023; 17:1294420. [PMID: 38026693 PMCID: PMC10665538 DOI: 10.3389/fncel.2023.1294420] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Obesity is a global epidemic, affecting roughly 30% of the world's population and predicted to rise. This disease results from genetic, behavioral, societal, and environmental factors, leading to excessive fat accumulation, due to insufficient energy expenditure. The adipose tissue, once seen as a simple storage depot, is now recognized as a complex organ with various functions, including hormone regulation and modulation of metabolism, inflammation, and homeostasis. Obesity is associated with a low-grade inflammatory state and has been linked to neurodegenerative diseases like multiple sclerosis (MS), Alzheimer's (AD), and Parkinson's (PD). Mechanistically, reduced adipose expandability leads to hypertrophic adipocytes, triggering inflammation, insulin and leptin resistance, blood-brain barrier disruption, altered brain metabolism, neuronal inflammation, brain atrophy, and cognitive decline. Obesity impacts neurodegenerative disorders through shared underlying mechanisms, underscoring its potential as a modifiable risk factor for these diseases. Nevertheless, further research is needed to fully grasp the intricate connections between obesity and neurodegeneration. Collaborative efforts in this field hold promise for innovative strategies to address this complex relationship and develop effective prevention and treatment methods, which also includes specific diets and physical activities, ultimately improving quality of life and health.
Collapse
Affiliation(s)
- Alexandre Neto
- Central Nervous System, Blood and Peripheral Inflammation, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Adelaide Fernandes
- Central Nervous System, Blood and Peripheral Inflammation, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Andreia Barateiro
- Central Nervous System, Blood and Peripheral Inflammation, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
9
|
Lippi SLP, Barkey RE, Rodriguez MN. High-fat diet negatively affects brain markers, cognitive behaviors, and noncognitive behaviors in the rTg4510 tau mouse model. Physiol Behav 2023; 271:114316. [PMID: 37543107 DOI: 10.1016/j.physbeh.2023.114316] [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: 05/22/2023] [Revised: 07/11/2023] [Accepted: 08/01/2023] [Indexed: 08/07/2023]
Abstract
Alzheimer's disease (AD) drastically impacts cognitive and noncognitive behaviors in both humans and animal models. Two hallmark proteins in AD, amyloid-β plaques and tau neurofibrillary tangles, accumulate in regions of the brain critical for learning and memory, including the hippocampus. Poor dietary choices have been shown to exacerbate cognitive deficits seen in AD. In this study, we assessed the effects of a high-fat diet (HFD - 60 kcal% fat) on cognitive & noncognitive behaviors as well as on brain markers in the rTg4510 tau mouse model. While all mice learned the Morris Water Maze (MWM) task, it was noted that on the last day of acquisition female tau mice had a significantly higher latency to find the platform than male tau mice (p < 0.01). Mice given the HFD spent significantly less time in the target quadrant than those given a control diet (CD) (p < 0.05). Tau mice showed impaired burrowing (p < 0.05) and nesting behaviors (p < 0.001) compared to WT mice and HFD administration worsened burrowing in tau mice. Tau mice exhibited greater levels of glial fibrillary acidic protein (GFAP) (p < 0.05) and significantly less hippocampal cell density than WT mice (p < 0.001). We observed trends of HFD mice having greater levels of GFAP and greater average tangle size than CD mice. These results highlight the importance of dietary choices, especially in older populations more susceptible to AD and its effects.
Collapse
Affiliation(s)
- Stephen L P Lippi
- University of Texas at San Antonio, Dept. Psychology, San Antonio, TX 78249, United States.
| | - Rachel E Barkey
- Pennsylvania State University College of Medicine, Dept. Neural and Behavioral Sciences, 700 HMC Crescent Road, Hershey, PA 17033, United States
| | - Mya N Rodriguez
- MD Anderson UTHealth Houston Graduate School of Biomedical Sciences, 6767 Bertner Ave, Houston, TX 77030, United States
| |
Collapse
|
10
|
Goikolea J, Latorre-Leal M, Tsagkogianni C, Pikkupeura S, Gulyas B, Cedazo-Minguez A, Loera-Valencia R, Björkhem I, Rodriguez Rodriguez P, Maioli S. Different effects of CYP27A1 and CYP7B1 on cognitive function: Two mouse models in comparison. J Steroid Biochem Mol Biol 2023; 234:106387. [PMID: 37648096 DOI: 10.1016/j.jsbmb.2023.106387] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/27/2023] [Indexed: 09/01/2023]
Abstract
The oxysterol 27-hydroxycholesterol (27OHC) is produced by the enzyme sterol 27-hydroxylase (Cyp27A1) and is mainly catabolized to 7α-Hydroxy-3-oxo-4-cholestenoic acid (7-HOCA) by the enzyme cytochrome P-450 oxysterol 7α-hydroxylase (Cyp7B1). 27OHC is mostly produced in the liver and can reach the brain by crossing the blood-brain barrier. A large body of evidence shows that CYP27A1 overexpression and high levels of 27OHC have a detrimental effect on the brain, causing cognitive and synaptic dysfunction together with a decrease in glucose uptake in mice. In this work, we analyzed two mouse models with high levels of 27OHC: Cyp7B1 knock-out mice and CYP27A1 overexpressing mice. Despite the accumulation of 27OHC in both models, Cyp7B1 knock-out mice maintained intact learning and memory capacities, neuronal morphology, and brain glucose uptake over time. Neurons treated with the Cyp7B1 metabolite 7-HOCA did not show changes in synaptic genes and 27OHC-treated Cyp7B1 knock-out neurons could not counteract 27OHC detrimental effects. This suggests that 7-HOCA and Cyp7B1 deletion in neurons do not mediate the neuroprotective effects observed in Cyp7B1 knock-out animals. RNA-seq of neuronal nuclei sorted from Cyp7B1 knock-out brains revealed upregulation of genes likely to confer neuroprotection to these animals. Differently from Cyp7B1 knock-out mice, transcriptomic data from CYP27A1 overexpressing neurons showed significant downregulation of genes associated with synaptic function and several metabolic processes. Our results suggest that the differences observed in the two models may be mediated by the higher levels of Cyp7B1 substrates such as 25-hydroxycholesterol and 3β-Adiol in the knock-out mice and that CYP27A1 overexpressing mice may be a more suitable model for studying 27-OHC-specific signaling. We believe that future studies on Cyp7B1 and Cyp27A1 will contribute to a better understanding of the pathogenic mechanisms of neurodegenerative diseases like Alzheimer's disease and may lead to potential new therapeutic approaches.
Collapse
Affiliation(s)
- Julen Goikolea
- Karolinska Institutet, Department of Neurobiology Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Stockholm, Sweden
| | - Maria Latorre-Leal
- Karolinska Institutet, Department of Neurobiology Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Stockholm, Sweden
| | - Christina Tsagkogianni
- Karolinska Institutet, Department of Neurobiology Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Stockholm, Sweden
| | - Sonja Pikkupeura
- Karolinska Institutet, Department of Neurobiology Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Stockholm, Sweden
| | - Balazs Gulyas
- Karolinska Institutet, Department of Clinical Neuroscience, Stockholm, Sweden
| | - Angel Cedazo-Minguez
- Karolinska Institutet, Department of Neurobiology Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Stockholm, Sweden
| | - Raul Loera-Valencia
- Karolinska Institutet, Department of Neurobiology Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Stockholm, Sweden; Tecnologico de Monterrey, School of Medicine and Health Sciences, Chihuahua, Mexico
| | - Ingemar Björkhem
- Karolinska Institutet, Department of Laboratory Medicine, Huddinge, Sweden
| | - Patricia Rodriguez Rodriguez
- Karolinska Institutet, Department of Neurobiology Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Stockholm, Sweden
| | - Silvia Maioli
- Karolinska Institutet, Department of Neurobiology Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Stockholm, Sweden.
| |
Collapse
|
11
|
Mengr A, Strnadová V, Strnad Š, Vrkoslav V, Pelantová H, Kuzma M, Comptdaer T, Železná B, Kuneš J, Galas MC, Pačesová A, Maletínská L. Feeding High-Fat Diet Accelerates Development of Peripheral and Central Insulin Resistance and Inflammation and Worsens AD-like Pathology in APP/PS1 Mice. Nutrients 2023; 15:3690. [PMID: 37686722 PMCID: PMC10490051 DOI: 10.3390/nu15173690] [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: 07/13/2023] [Revised: 08/15/2023] [Accepted: 08/16/2023] [Indexed: 09/10/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive brain disorder characterized by extracellular amyloid-β (Aβ) plaques, intracellular neurofibrillary tangles formed by hyperphosphorylated Tau protein and neuroinflammation. Previous research has shown that obesity and type 2 diabetes mellitus, underlined by insulin resistance (IR), are risk factors for neurodegenerative disorders. In this study, obesity-induced peripheral and central IR and inflammation were studied in relation to AD-like pathology in the brains and periphery of APP/PS1 mice, a model of Aβ pathology, fed a high-fat diet (HFD). APP/PS1 mice and their wild-type controls fed either a standard diet or HFD were characterized at the ages of 3, 6 and 10 months by metabolic parameters related to obesity via mass spectroscopy, nuclear magnetic resonance, immunoblotting and immunohistochemistry to quantify how obesity affected AD pathology. The HFD induced substantial peripheral IR leading to central IR. APP/PS1-fed HFD mice had more pronounced IR, glucose intolerance and liver steatosis than their WT controls. The HFD worsened Aβ pathology in the hippocampi of APP/PS1 mice and significantly supported both peripheral and central inflammation. This study reveals a deleterious effect of obesity-related mild peripheral inflammation and prediabetes on the development of Aβ and Tau pathology and neuroinflammation in APP/PS1 mice.
Collapse
Affiliation(s)
- Anna Mengr
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, Prague 6, 166 10 Prague, Czech Republic; (A.M.); (V.S.); (Š.S.); (V.V.); (B.Ž.); (J.K.)
| | - Veronika Strnadová
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, Prague 6, 166 10 Prague, Czech Republic; (A.M.); (V.S.); (Š.S.); (V.V.); (B.Ž.); (J.K.)
| | - Štěpán Strnad
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, Prague 6, 166 10 Prague, Czech Republic; (A.M.); (V.S.); (Š.S.); (V.V.); (B.Ž.); (J.K.)
| | - Vladimír Vrkoslav
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, Prague 6, 166 10 Prague, Czech Republic; (A.M.); (V.S.); (Š.S.); (V.V.); (B.Ž.); (J.K.)
| | - Helena Pelantová
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, Prague 4, 142 20 Prague, Czech Republic; (H.P.); (M.K.)
| | - Marek Kuzma
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, Prague 4, 142 20 Prague, Czech Republic; (H.P.); (M.K.)
| | - Thomas Comptdaer
- University of Lille, Inserm, CHU Lille, CNRS, LilNCog-Lille Neuroscience & Cognition, F-59000 Lille, France; (T.C.); (M.-C.G.)
| | - Blanka Železná
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, Prague 6, 166 10 Prague, Czech Republic; (A.M.); (V.S.); (Š.S.); (V.V.); (B.Ž.); (J.K.)
| | - Jaroslav Kuneš
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, Prague 6, 166 10 Prague, Czech Republic; (A.M.); (V.S.); (Š.S.); (V.V.); (B.Ž.); (J.K.)
- Institute of Physiology of the Czech Academy of Sciences, Vídeňská 1083, Prague 4, 142 20 Prague, Czech Republic
| | - Marie-Christine Galas
- University of Lille, Inserm, CHU Lille, CNRS, LilNCog-Lille Neuroscience & Cognition, F-59000 Lille, France; (T.C.); (M.-C.G.)
| | - Andrea Pačesová
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, Prague 6, 166 10 Prague, Czech Republic; (A.M.); (V.S.); (Š.S.); (V.V.); (B.Ž.); (J.K.)
| | - Lenka Maletínská
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, Prague 6, 166 10 Prague, Czech Republic; (A.M.); (V.S.); (Š.S.); (V.V.); (B.Ž.); (J.K.)
| |
Collapse
|
12
|
Doroszkiewicz J, Mroczko J, Rutkowski P, Mroczko B. Molecular Aspects of a Diet as a New Pathway in the Prevention and Treatment of Alzheimer's Disease. Int J Mol Sci 2023; 24:10751. [PMID: 37445928 PMCID: PMC10341644 DOI: 10.3390/ijms241310751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/19/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Alzheimer's disease is the most common cause of dementia in the world. Lack of an established pathology makes it difficult to develop suitable approaches and treatment for the disease. Besides known hallmarks, including amyloid β peptides cumulating in plaques and hyperphosphorylated tau forming NFTs, inflammation also plays an important role, with known connections to the diet. In AD, adhering to reasonable nutrition according to age-related principles is recommended. The diet should be high in neuroprotective foods, such as polyunsaturated fatty acids, antioxidants, and B vitamins. In addition, foods capable of rising BDNF should be considered because of the known profitable results of this molecule in AD. Adhering to beneficial diets might result in improvements in memory, cognition, and biomarkers and might even reduce the risk of developing AD. In this review, we discuss the effects of various diets, foods, and nutrients on brain health and possible connections to Alzheimer's disease.
Collapse
Affiliation(s)
- Julia Doroszkiewicz
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-089 Bialystok, Poland; (J.M.); (B.M.)
| | - Jan Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-089 Bialystok, Poland; (J.M.); (B.M.)
| | | | - Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-089 Bialystok, Poland; (J.M.); (B.M.)
- Department of Biochemical Diagnostics, Medical University of Białystok, 15-089 Bialystok, Poland
| |
Collapse
|
13
|
So SW, Fleming KM, Nixon JP, Butterick TA. Early Life Obesity Increases Neuroinflammation, Amyloid Beta Deposition, and Cognitive Decline in a Mouse Model of Alzheimer's Disease. Nutrients 2023; 15:nu15112494. [PMID: 37299457 DOI: 10.3390/nu15112494] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/23/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
Obesity, a known risk factor of Alzheimer's disease (AD), increases the activation of microglia, leading to a proinflammatory phenotype. Our previous work shows that a high fat diet (HFD) can cause neuroinflammation and cognitive decline in mice. We hypothesized that proinflammatory activation of brain microglia in obesity exacerbates AD pathology and increases the accumulation of amyloid beta (Aβ) plaques. Presently, we tested cognitive function in 8-month-old male and female APP/PS1 mice fed a HFD, starting at 1.5 months of age. Locomotor activity, anxiety-like behavior, behavioral despair, and spatial memory were all assessed through behavioral tests. Microgliosis and Aβ deposition were measured in multiple brain regions through immunohistochemical analysis. Our results show that a HFD decreases locomotor activity, while increasing anxiety-like behavior and behavioral despair independent of genotype. A HFD led to increased memory deficits in both sexes, with HFD-fed APP/PS1 mice performing the worst out of all groups. Immunohistochemical analysis showed increased microgliosis in mice fed a HFD. This was accompanied by an increase in Aβ deposition in the HFD-fed APP/PS1 mice. Together, our results support that HFD-induced obesity exacerbates neuroinflammation and Aβ deposition in a young adult AD mouse model, leading to increased memory deficits and cognitive decline in both sexes.
Collapse
Affiliation(s)
- Simon W So
- Minneapolis Veterans Affairs Health Care System, Minneapolis, MN 55417, USA
- Department of Neuroscience, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA
| | - Kendra M Fleming
- Minneapolis Veterans Affairs Health Care System, Minneapolis, MN 55417, USA
- Department of Neuroscience, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA
| | - Joshua P Nixon
- Minneapolis Veterans Affairs Health Care System, Minneapolis, MN 55417, USA
- Department of Food Science and Nutrition, University of Minnesota Twin Cities, St. Paul, MN 55108, USA
- Department of Surgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Tammy A Butterick
- Minneapolis Veterans Affairs Health Care System, Minneapolis, MN 55417, USA
- Department of Neuroscience, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA
- Department of Food Science and Nutrition, University of Minnesota Twin Cities, St. Paul, MN 55108, USA
| |
Collapse
|
14
|
Lim HK, Bae S, Han K, Kang BM, Jeong Y, Kim SG, Suh M. Seizure-induced neutrophil adhesion in brain capillaries leads to a decrease in postictal cerebral blood flow. iScience 2023; 26:106655. [PMID: 37168551 PMCID: PMC10164910 DOI: 10.1016/j.isci.2023.106655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 03/23/2023] [Accepted: 04/06/2023] [Indexed: 05/13/2023] Open
Abstract
Cerebral hypoperfusion has been proposed as a potential cause of postictal neurological dysfunction in epilepsy, but its underlying mechanism is still unclear. We show that a 30% reduction in postictal cerebral blood flow (CBF) has two contributing factors: the early hypoperfusion up to ∼30 min post-seizure was mainly induced by arteriolar constriction, while the hypoperfusion that persisted for over an hour was due to increased capillary stalling induced by neutrophil adhesion to brain capillaries, decreased red blood cell (RBC) flow accompanied by constriction of capillaries and venules, and elevated intercellular adhesion molecule-1 (ICAM-1) expression. Administration of antibodies against the neutrophil marker Ly6G and against LFA-1, which mediates adhesive interactions with ICAM-1, prevented neutrophil adhesion and recovered the prolonged CBF reductions to control levels. Our findings provide evidence that seizure-induced neutrophil adhesion to cerebral microvessels via ICAM-1 leads to prolonged postictal hypoperfusion, which may underlie neurological dysfunction in epilepsy.
Collapse
Affiliation(s)
- Hyun-Kyoung Lim
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, South Korea
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon 16419, South Korea
| | - Sungjun Bae
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, South Korea
- IMNEWRUN Inc, N Center Bldg. A 5F, Sungkyunkwan University, Suwon 16419, South Korea
| | - Kayoung Han
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, South Korea
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon 16419, South Korea
| | - Bok-Man Kang
- IMNEWRUN Inc, N Center Bldg. A 5F, Sungkyunkwan University, Suwon 16419, South Korea
| | - Yoonyi Jeong
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon 16419, South Korea
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, South Korea
- Department of Intelligent Precision Healthcare Convergence (IPHC), Sungkyunkwan University, Suwon 16419, South Korea
| | - Seong-Gi Kim
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon 16419, South Korea
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, South Korea
- Department of Intelligent Precision Healthcare Convergence (IPHC), Sungkyunkwan University, Suwon 16419, South Korea
| | - Minah Suh
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, South Korea
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon 16419, South Korea
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, South Korea
- IMNEWRUN Inc, N Center Bldg. A 5F, Sungkyunkwan University, Suwon 16419, South Korea
- Department of Intelligent Precision Healthcare Convergence (IPHC), Sungkyunkwan University, Suwon 16419, South Korea
- Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Suwon 16419, South Korea
| |
Collapse
|
15
|
Yavari M, Ramalingam L, Harris BN, Kahathuduwa CN, Chavira A, Biltz C, Mounce L, Maldonado KA, Scoggin S, Zu Y, Kalupahana NS, Yosofvand M, Moussa H, Moustaid-Moussa N. Eicosapentaenoic Acid Protects against Metabolic Impairments in the APPswe/PS1dE9 Alzheimer's Disease Mouse Model. J Nutr 2023; 153:1038-1051. [PMID: 36781072 PMCID: PMC10273166 DOI: 10.1016/j.tjnut.2023.01.030] [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: 09/03/2022] [Revised: 01/18/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is an age-related neurodegenerative disease characterized by amyloid-β (Aβ) plaques. Systemic inflammation and obesity may exacerbate AD pathogenesis. We previously reported anti-inflammatory and anti-obesity effects of EPA in mice. OBJECTIVES We aimed to determine whether EPA reduces obesity-associated metabolic dysfunctions and Aβ accumulation in AD amyloidogenic mice. METHODS Two-mo-old APPswe/PS1dE9 transgenic (TG) mice and non-TG littermates were randomly assigned to low fat (LF; 10% kcal fat), high fat (HF; 45% kcal fat), or EPA (36 g/kg)-supplemented HF diets. Body composition, glucose tolerance, and energy expenditure were measured, and serum and brain metabolic markers were tested 38 wk postintervention. Outcomes were statistically analyzed via 3-factor ANOVA, modeling genotype, sex, and diet interactions. RESULTS HF-fed males gained more weight than females (Δ = 61 mg; P < 0.001). Compared with LF, HF increased body weights of wild-type (WT) males (Δ = 31 mg; P < 0.001). EPA reduced HF-induced weight gain in WT males (Δ = 24 mg; P = 0.054) but not in females. HF mice showed decreased glucose clearance and respiratory energy compared with LF-fed groups (Δ = -1.31 g/dL; P < 0.001), with no significant effects of EPA. However, EPA conferred metabolic improvements by decreasing serum leptin and insulin (Δ = -2.51 g/mL and Δ = -0.694 ng/mL, respectively compared with HF, P ≤ 0.05) and increasing adiponectin (Δ = 21.6 ng/mL; P < 0.001). As we expected, TG mice expressed higher serum and brain Aβ than WT mice (Δ = 0.131 ng/mL; P < 0.001 and Δ = 0.56%; P < 0.01, respectively), and EPA reduced serum Aβ1-40 in TG males compared with HF (Δ = 0.053 ng/mL; P ≤ 0.05). CONCLUSIONS To our knowledge, this is the first report that EPA reduces serum Aβ1-40 in obese AD male mice, warranting further investigations into tissue-specific mechanisms of EPA in AD.
Collapse
Affiliation(s)
- Mahsa Yavari
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA; Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX, USA
| | - Latha Ramalingam
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| | - Breanna N Harris
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX, USA; Department of Biological Sciences, Texas Tech University, Lubbock, TX, USA
| | - Chanaka Nadeeshan Kahathuduwa
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX, USA; Department of Laboratory Science and Primary Care, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Angela Chavira
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| | - Caroline Biltz
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| | - Logan Mounce
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, USA
| | | | - Shane Scoggin
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| | - Yujiao Zu
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA; Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX, USA
| | - Nishan Sudheera Kalupahana
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA; Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX, USA; Department of Physiology, University of Peradeniya, Sri Lanka
| | - Mohammad Yosofvand
- Department of Mechanical Engineering, Texas Tech University, Lubbock, TX, USA
| | - Hanna Moussa
- Department of Mechanical Engineering, Texas Tech University, Lubbock, TX, USA
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA; Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX, USA.
| |
Collapse
|
16
|
Lee Y, Park HR, Lee JY, Kim J, Yang S, Lee C, Kim K, Kim HS, Chang SC, Lee J. Low-dose curcumin enhances hippocampal neurogenesis and memory retention in young mice. Arch Pharm Res 2023; 46:423-437. [PMID: 36947339 DOI: 10.1007/s12272-023-01440-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/02/2023] [Indexed: 03/23/2023]
Abstract
Adult neurogenesis generates new functional neurons from adult neural stem cells in various regions, including the subventricular zone (SVZ) of the lateral ventricles and subgranular zone (SGZ) of hippocampal dentate gyrus (DG). Available evidence shows hippocampal neurogenesis can be negatively or positively regulated by dietary components. In a previous study, we reported that curcumin (diferuloylmethane; a polyphenolic found in curry spice) stimulates the proliferation of embryonic neural stem cells (NSCs) by activating adaptive cellular stress responses. Here, we investigated whether subchronic administration of curcumin (once daily at 0.4, 2, or 10 mg/kg for 14 days) promotes hippocampal neurogenesis and neurocognitive function in young (5-week-old) mice. Oral administration of low-dose curcumin (0.4 mg/kg) increased the proliferation and survival of newly generated cells in hippocampus, but surprisingly, high-dose curcumin (10 mg/kg) did not effectively upregulate the proliferation or survival of newborn cells. Furthermore, hippocampal BDNF levels and phosphorylated CREB activity were elevated in only low-dose curcumin-treated mice. Passive avoidance testing revealed that low-dose curcumin increased cross-over latency times, indicating enhanced memory retention, and an in vitro study showed that low-concentration curcumin increased the proliferative activity of neural progenitor cells (NPCs) by upregulating NF1X levels. Collectively, our findings suggest that low-dose curcumin has neurogenic effects and that it may prevent age and neurodegenerative disease-related cognitive deficits.
Collapse
Affiliation(s)
- Yujeong Lee
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea
- Cognitive Science Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Hee Ra Park
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea
- Department of KM Science Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseong-daero, Yuseong-gu, Daejeon, 34054, Republic of Korea
| | - Joo Yeon Lee
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea
| | - Jaehoon Kim
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea
| | - Seonguk Yang
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea
| | - Chany Lee
- Cognitive Science Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Kipom Kim
- Research Strategy Office, Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Hyung Sik Kim
- Division of Toxicology, School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Seung-Cheol Chang
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan, 46241, Republic of Korea
| | - Jaewon Lee
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea.
| |
Collapse
|
17
|
Ruiz-Uribe NE, Bracko O, Swallow M, Omurzakov A, Dash S, Uchida H, Xiang D, Haft-Javaherian M, Falkenhain K, Lamont ME, Ali M, Njiru BN, Chang HY, Tan AY, Xiang JZ, Iadecola C, Park L, Sanchez T, Nishimura N, Schaffer CB. Vascular oxidative stress causes neutrophil arrest in brain capillaries, leading to decreased cerebral blood flow and contributing to memory impairment in a mouse model of Alzheimer’s disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.15.528710. [PMID: 36824768 PMCID: PMC9949082 DOI: 10.1101/2023.02.15.528710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
INTRODUCTION In this study, we explore the role of oxidative stress produced by NOX2-containing NADPH oxidase as a molecular mechanism causing capillary stalling and cerebral blood flow deficits in the APP/PS1 mouse model of AD. METHODS We inhibited NOX2 in APP/PS1 mice by administering a 10 mg/kg dose of the peptide inhibitor gp91-ds-tat i.p., for two weeks. We used in vivo two-photon imaging to measure capillary stalling, penetrating arteriole flow, and vascular inflammation. We also characterized short-term memory function and gene expression changes in cerebral microvessels. RESULTS We found that after NOX2 inhibition capillary stalling, as well as parenchymal and vascular inflammation, were significantly reduced. In addition, we found a significant increase in penetrating arteriole flow, followed by an improvement in short-term memory, and downregulation of inflammatory gene expression pathways. DISCUSSION Oxidative stress is a major mechanism leading to microvascular dysfunction in AD, and represents an important therapeutic target.
Collapse
|
18
|
Ibeh S, Bakkar NMZ, Ahmad F, Nwaiwu J, Barsa C, Mekhjian S, Reslan MA, Eid AH, Harati H, Nabha S, Mechref Y, El-Yazbi AF, Kobeissy F. High fat diet exacerbates long-term metabolic, neuropathological, and behavioral derangements in an experimental mouse model of traumatic brain injury. Life Sci 2023; 314:121316. [PMID: 36565814 DOI: 10.1016/j.lfs.2022.121316] [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: 10/20/2022] [Revised: 12/13/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
AIMS Traumatic brain injury (TBI) constitutes a serious public health concern. Although TBI targets the brain, it can exert several systemic effects which can worsen the complications observed in TBI subjects. Currently, there is no FDA-approved therapy available for its treatment. Thus, there has been an increasing need to understand other factors that could modulate TBI outcomes. Among the factors involved are diet and lifestyle. High-fat diets (HFD), rich in saturated fat, have been associated with adverse effects on brain health. MAIN METHODS To study this phenomenon, an experimental mouse model of open head injury, induced by the controlled cortical impact was used along with high-fat feeding to evaluate the impact of HFD on brain injury outcomes. Mice were fed HFD for a period of two months where several neurological, behavioral, and molecular outcomes were assessed to investigate the impact on chronic consequences of the injury 30 days post-TBI. KEY FINDINGS Two months of HFD feeding, together with TBI, led to a notable metabolic, neurological, and behavioral impairment. HFD was associated with increased blood glucose and fat-to-lean ratio. Spatial learning and memory, as well as motor coordination, were all significantly impaired. Notably, HFD aggravated neuroinflammation, oxidative stress, and neurodegeneration. Also, cell proliferation post-TBI was repressed by HFD, which was accompanied by an increased lesion volume. SIGNIFICANCE Our research indicated that chronic HFD feeding can worsen functional outcomes, predispose to neurodegeneration, and decrease brain recovery post-TBI. This sheds light on the clinical impact of HFD on TBI pathophysiology and rehabilitation as well.
Collapse
Affiliation(s)
- Stanley Ibeh
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Nour-Mounira Z Bakkar
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Fatima Ahmad
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Neuroscience Research Center, Lebanese University, Beirut, Lebanon
| | - Judith Nwaiwu
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Deparment of Chemistry, Texas Tech University, Lubbock, TX, USA
| | - Chloe Barsa
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Sarine Mekhjian
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Mohammad Amine Reslan
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Hayat Harati
- Neuroscience Research Center, Lebanese University, Beirut, Lebanon
| | - Sanaa Nabha
- Neuroscience Research Center, Lebanese University, Beirut, Lebanon
| | - Yehia Mechref
- Deparment of Chemistry, Texas Tech University, Lubbock, TX, USA
| | - Ahmed F El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Deparment of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt; Faculty of Pharmacy, Alamein International University, Al-Alamein, Egypt.
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Morehouse School of Medicine, Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), 720 Westview Dr. SW, Atlanta, GA 30310, USA.
| |
Collapse
|
19
|
Chandra S, Sisodia SS, Vassar RJ. The gut microbiome in Alzheimer's disease: what we know and what remains to be explored. Mol Neurodegener 2023; 18:9. [PMID: 36721148 PMCID: PMC9889249 DOI: 10.1186/s13024-023-00595-7] [Citation(s) in RCA: 60] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/06/2023] [Indexed: 02/02/2023] Open
Abstract
Alzheimer's disease (AD), the most common cause of dementia, results in a sustained decline in cognition. There are currently few effective disease modifying therapies for AD, but insights into the mechanisms that mediate the onset and progression of disease may lead to new, effective therapeutic strategies. Amyloid beta oligomers and plaques, tau aggregates, and neuroinflammation play a critical role in neurodegeneration and impact clinical AD progression. The upstream modulators of these pathological features have not been fully clarified, but recent evidence indicates that the gut microbiome (GMB) may have an influence on these features and therefore may influence AD progression in human patients. In this review, we summarize studies that have identified alterations in the GMB that correlate with pathophysiology in AD patients and AD mouse models. Additionally, we discuss findings with GMB manipulations in AD models and potential GMB-targeted therapeutics for AD. Lastly, we discuss diet, sleep, and exercise as potential modifiers of the relationship between the GMB and AD and conclude with future directions and recommendations for further studies of this topic.
Collapse
Affiliation(s)
- Sidhanth Chandra
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
- Medical Scientist Training Program, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Sangram S. Sisodia
- Department of Neurobiology, University of Chicago, Chicago, IL 60637 USA
| | - Robert J. Vassar
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| |
Collapse
|
20
|
Novoa C, Salazar P, Cisternas P, Gherardelli C, Vera-Salazar R, Zolezzi JM, Inestrosa NC. Inflammation context in Alzheimer's disease, a relationship intricate to define. Biol Res 2022; 55:39. [PMID: 36550479 PMCID: PMC9784299 DOI: 10.1186/s40659-022-00404-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 11/15/2022] [Indexed: 12/24/2022] Open
Abstract
Alzheimer's disease (AD), the most common form of dementia, is characterized by the accumulation of amyloid β (Aβ) and hyperphosphorylated tau protein aggregates. Importantly, Aβ and tau species are able to activate astrocytes and microglia, which release several proinflammatory cytokines, such as tumor necrosis factor α (TNF-α) and interleukin 1β (IL-1β), together with reactive oxygen (ROS) and nitrogen species (RNS), triggering neuroinflammation. However, this inflammatory response has a dual function: it can play a protective role by increasing Aβ degradation and clearance, but it can also contribute to Aβ and tau overproduction and induce neurodegeneration and synaptic loss. Due to the significant role of inflammation in the pathogenesis of AD, several inflammatory mediators have been proposed as AD markers, such as TNF-α, IL-1β, Iba-1, GFAP, NF-κB, TLR2, and MHCII. Importantly, the use of anti-inflammatory drugs such as NSAIDs has emerged as a potential treatment against AD. Moreover, diseases related to systemic or local inflammation, including infections, cerebrovascular accidents, and obesity, have been proposed as risk factors for the development of AD. In the following review, we focus on key inflammatory processes associated with AD pathogenesis.
Collapse
Affiliation(s)
- Catalina Novoa
- Centro de Envejecimiento y Regeneración (CARE-UC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda Bernardo O'Higgins 340, P.O. Box 114-D, Santiago, Chile
| | - Paulina Salazar
- Centro de Envejecimiento y Regeneración (CARE-UC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda Bernardo O'Higgins 340, P.O. Box 114-D, Santiago, Chile
| | - Pedro Cisternas
- Instituto de Ciencias de la Salud, Universidad de O'Higgins, Rancagua, Chile
| | - Camila Gherardelli
- Centro de Envejecimiento y Regeneración (CARE-UC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda Bernardo O'Higgins 340, P.O. Box 114-D, Santiago, Chile
| | - Roberto Vera-Salazar
- Facultad de Ciencias Médicas, Escuela de Kinesiología, Universidad de Santiago de Chile, Santiago, Chile
| | - Juan M Zolezzi
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE-UC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda Bernardo O'Higgins 340, P.O. Box 114-D, Santiago, Chile.
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile.
| |
Collapse
|
21
|
Amelianchik A, Sweetland-Martin L, Norris EH. The effect of dietary fat consumption on Alzheimer's disease pathogenesis in mouse models. Transl Psychiatry 2022; 12:293. [PMID: 35869065 PMCID: PMC9307654 DOI: 10.1038/s41398-022-02067-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 11/25/2022] Open
Abstract
Alzheimer's disease (AD) is a fatal cognitive disorder with proteinaceous brain deposits, neuroinflammation, cerebrovascular dysfunction, and extensive neuronal loss over time. AD is a multifactorial disease, and lifestyle factors, including diet, are likely associated with the development of AD pathology. Since obesity and diabetes are recognized as risk factors for AD, it might be predicted that a high-fat diet (HFD) would worsen AD pathology. However, modeling HFD-induced obesity in AD animal models has yielded inconclusive results. Some studies report a deleterious effect of HFD on Aβ accumulation, neuroinflammation, and cognitive function, while others report that HFD worsens memory without affecting AD brain pathology. Moreover, several studies report no major effect of HFD on AD-related phenotypes in mice, while other studies show that HFD might, in fact, be protective. The lack of a clear association between dietary fat consumption and AD-related pathology and cognitive function in AD mouse models might be explained by experimental variations, including AD mouse model, sex and age of the animals, composition of the HFD, and timeline of HFD consumption. In this review, we summarize recent studies that aimed at elucidating the effect of HFD-induced obesity on AD-related pathology in mice and provide an overview of the factors that may have contributed to the results reported in these studies. Based on the heterogeneity of these animal model studies and given that the human population itself is quite disparate, it is likely that people will benefit most from individualized nutritional plans based on their medical history and clinical profiles.
Collapse
Affiliation(s)
- Anna Amelianchik
- Patricia and John Rosenwald Laboratory of Neurobiology & Genetics, The Rockefeller University, 1230 York Avenue, New York, NY, USA
| | - Lauren Sweetland-Martin
- Patricia and John Rosenwald Laboratory of Neurobiology & Genetics, The Rockefeller University, 1230 York Avenue, New York, NY, USA
| | - Erin H Norris
- Patricia and John Rosenwald Laboratory of Neurobiology & Genetics, The Rockefeller University, 1230 York Avenue, New York, NY, USA.
| |
Collapse
|
22
|
Bah TM, Allen EM, Garcia-Jaramillo M, Perez R, Zarnegarnia Y, Davis CM, Bloom MB, Magana AA, Choi J, Bobe G, Pike MM, Raber J, Maier CS, Alkayed NJ. GPR39 Deficiency Impairs Memory and Alters Oxylipins and Inflammatory Cytokines Without Affecting Cerebral Blood Flow in a High-Fat Diet Mouse Model of Cognitive Impairment. Front Cell Neurosci 2022; 16:893030. [PMID: 35875352 PMCID: PMC9298837 DOI: 10.3389/fncel.2022.893030] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/20/2022] [Indexed: 12/30/2022] Open
Abstract
Vascular cognitive impairment (VCI) is the second most common cause of dementia. There is no treatment for VCI, in part due to a lack of understanding of the underlying mechanisms. The G-protein coupled receptor 39 (GPR39) is regulated by arachidonic acid (AA)-derived oxylipins that have been implicated in VCI. Furthermore, GPR39 is increased in microglia of post mortem human brains with VCI. Carriers of homozygous GPR39 SNPs have a higher burden of white matter hyperintensity, an MRI marker of VCI. We tested the hypothesis that GPR39 plays a protective role against high-fat diet (HFD)-induced cognitive impairment, in part mediated via oxylipins actions on cerebral blood flow (CBF) and neuroinflammation. Homozygous (KO) and heterozygous (Het) GPR39 knockout mice and wild-type (WT) littermates with and without HFD for 8 months were tested for cognitive performance using the novel object recognition (NOR) and the Morris water maze (MWM) tests, followed by CBF measurements using MRI. Brain tissue and plasma oxylipins were quantified with high-performance liquid chromatography coupled to mass spectrometry. Cytokines and chemokines were measured using a multiplex assay. KO mice, regardless of diet, swam further away from platform location in the MWM compared to WT and Het mice. In the NOR test, there were no effects of genotype or diet. Brain and plasma AA-derived oxylipins formed by 11- and 15-lipoxygenase (LOX), cyclooxygenase (COX) and non-enzymatically were increased by HFD and GPR39 deletion. Interleukin-10 (IL-10) was lower in KO mice on HFD than standard diet (STD), whereas IL-4, interferon γ-induced protein-10 (IP-10) and monocyte chemotactic protein-3 (MCP-3) were altered by diet in both WT and KO, but were not affected by genotype. Resting CBF was reduced in WT and KO mice on HFD, with no change in vasoreactivity. The deletion of GPR39 did not change CBF compared to WT mice on either STD or HFD. We conclude that GPR39 plays a role in spatial memory retention and protects against HFD-induced cognitive impairment in part by modulating inflammation and AA-derived oxylipins. The results indicate that GPR39 and oxylipin pathways play a role and may serve as therapeutic targets in VCI.
Collapse
Affiliation(s)
- Thierno M. Bah
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Elyse M. Allen
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Manuel Garcia-Jaramillo
- Department of Chemistry, Oregon State University, Corvallis, OR, United States
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, United States
| | - Ruby Perez
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Yalda Zarnegarnia
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Catherine M. Davis
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Madeline B. Bloom
- Department of Chemistry, Oregon State University, Corvallis, OR, United States
| | - Armando A. Magana
- Department of Chemistry, Oregon State University, Corvallis, OR, United States
| | - Jaewoo Choi
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
| | - Gerd Bobe
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
| | - Martin M. Pike
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR, United States
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
- Departments of Neurology, Radiation Medicine, and Psychiatry, Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, United States
- College of Pharmacy, Oregon State University, Corvallis, OR, United States
| | - Claudia S. Maier
- Department of Chemistry, Oregon State University, Corvallis, OR, United States
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
| | - Nabil J. Alkayed
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, United States
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
23
|
Saiyasit N, Butlig EAR, Chaney SD, Traylor MK, Hawley NA, Randall RB, Bobinger HV, Frizell CA, Trimm F, Crook ED, Lin M, Hill BD, Keller JL, Nelson AR. Neurovascular Dysfunction in Diverse Communities With Health Disparities-Contributions to Dementia and Alzheimer's Disease. Front Neurosci 2022; 16:915405. [PMID: 35844216 PMCID: PMC9279126 DOI: 10.3389/fnins.2022.915405] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/31/2022] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease and related dementias (ADRD) are an expanding worldwide crisis. In the absence of scientific breakthroughs, the global prevalence of ADRD will continue to increase as more people are living longer. Racial or ethnic minority groups have an increased risk and incidence of ADRD and have often been neglected by the scientific research community. There is mounting evidence that vascular insults in the brain can initiate a series of biological events leading to neurodegeneration, cognitive impairment, and ADRD. We are a group of researchers interested in developing and expanding ADRD research, with an emphasis on vascular contributions to dementia, to serve our local diverse community. Toward this goal, the primary objective of this review was to investigate and better understand health disparities in Alabama and the contributions of the social determinants of health to those disparities, particularly in the context of vascular dysfunction in ADRD. Here, we explain the neurovascular dysfunction associated with Alzheimer's disease (AD) as well as the intrinsic and extrinsic risk factors contributing to dysfunction of the neurovascular unit (NVU). Next, we ascertain ethnoregional health disparities of individuals living in Alabama, as well as relevant vascular risk factors linked to AD. We also discuss current pharmaceutical and non-pharmaceutical treatment options for neurovascular dysfunction, mild cognitive impairment (MCI) and AD, including relevant studies and ongoing clinical trials. Overall, individuals in Alabama are adversely affected by social and structural determinants of health leading to health disparities, driven by rurality, ethnic minority status, and lower socioeconomic status (SES). In general, these communities have limited access to healthcare and healthy food and other amenities resulting in decreased opportunities for early diagnosis of and pharmaceutical treatments for ADRD. Although this review is focused on the current state of health disparities of ADRD patients in Alabama, future studies must include diversity of race, ethnicity, and region to best be able to treat all individuals affected by ADRD.
Collapse
Affiliation(s)
- Napatsorn Saiyasit
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Evan-Angelo R. Butlig
- Department of Neurology, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA, United States
| | - Samantha D. Chaney
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Miranda K. Traylor
- Department of Health, Kinesiology, and Sport, University of South Alabama, Mobile, AL, United States
| | - Nanako A. Hawley
- Department of Psychology, University of South Alabama, Mobile, AL, United States
| | - Ryleigh B. Randall
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Hanna V. Bobinger
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Carl A. Frizell
- Department of Physician Assistant Studies, University of South Alabama, Mobile, AL, United States
| | - Franklin Trimm
- College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Errol D. Crook
- Department of Internal Medicine, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Mike Lin
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Benjamin D. Hill
- Department of Psychology, University of South Alabama, Mobile, AL, United States
| | - Joshua L. Keller
- Department of Health, Kinesiology, and Sport, University of South Alabama, Mobile, AL, United States
| | - Amy R. Nelson
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| |
Collapse
|
24
|
Oblak AL, Kotredes KP, Pandey RS, Reagan AM, Ingraham C, Perkins B, Lloyd C, Baker D, Lin PB, Soni DM, Tsai AP, Persohn SA, Bedwell AA, Eldridge K, Speedy R, Meyer JA, Peters JS, Figueiredo LL, Sasner M, Territo PR, Sukoff Rizzo SJ, Carter GW, Lamb BT, Howell GR. Plcg2M28L Interacts With High Fat/High Sugar Diet to Accelerate Alzheimer's Disease-Relevant Phenotypes in Mice. Front Aging Neurosci 2022; 14:886575. [PMID: 35813947 PMCID: PMC9263289 DOI: 10.3389/fnagi.2022.886575] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/09/2022] [Indexed: 11/30/2022] Open
Abstract
Obesity is recognized as a significant risk factor for Alzheimer's disease (AD). Studies have supported the notion that obesity accelerates AD-related pathophysiology in mouse models of AD. The majority of studies, to date, have focused on the use of early-onset AD models. Here, we evaluate the impact of genetic risk factors on late-onset AD (LOAD) in mice fed with a high fat/high sugar diet (HFD). We focused on three mouse models created through the IU/JAX/PITT MODEL-AD Center. These included a combined risk model with APOE4 and a variant in triggering receptor expressed on myeloid cells 2 (Trem2R47H ). We have termed this model, LOAD1. Additional variants including the M28L variant in phospholipase C Gamma 2 (Plcg2M28L ) and the 677C > T variant in methylenetetrahydrofolate reductase (Mthfr 677C > T ) were engineered by CRISPR onto LOAD1 to generate LOAD1.Plcg2M28L and LOAD1.Mthfr 677C > T . At 2 months of age, animals were placed on an HFD that induces obesity or a control diet (CD), until 12 months of age. Throughout the study, blood was collected to assess the levels of cholesterol and glucose. Positron emission tomography/computed tomography (PET/CT) was completed prior to sacrifice to image for glucose utilization and brain perfusion. After the completion of the study, blood and brains were collected for analysis. As expected, animals fed a HFD, showed a significant increase in body weight compared to those fed a CD. Glucose increased as a function of HFD in females only with cholesterol increasing in both sexes. Interestingly, LOAD1.Plcg2M28L demonstrated an increase in microglia density and alterations in regional brain glucose and perfusion on HFD. These changes were not observed in LOAD1 or LOAD1.Mthfr 677C > T animals fed with HFD. Furthermore, LOAD1.Plcg2M28L but not LOAD1.Mthfr 677C > T or LOAD1 animals showed transcriptomics correlations with human AD modules. Our results show that HFD affects the brain in a genotype-specific manner. Further insight into this process may have significant implications for the development of lifestyle interventions for the treatment of AD.
Collapse
Affiliation(s)
- Adrian L. Oblak
- Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Radiology & Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | | | - Ravi S. Pandey
- The Jackson Laboratory, Bar Harbor, ME, United States
- Jackson Laboratory for Genomic Medicine, Farmington, CT, United States
| | | | - Cynthia Ingraham
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Bridget Perkins
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Christopher Lloyd
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Deborah Baker
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Peter B. Lin
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Disha M. Soni
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Andy P. Tsai
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Scott A. Persohn
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Amanda A. Bedwell
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Kierra Eldridge
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Rachael Speedy
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Jill A. Meyer
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Johnathan S. Peters
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Lucas L. Figueiredo
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | | | - Paul R. Territo
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
- Department of Medicine, Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Stacey J. Sukoff Rizzo
- Department of Medicine, Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | | | - Bruce T. Lamb
- Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Radiology & Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | | |
Collapse
|
25
|
Gannon OJ, Robison LS, Salinero AE, Abi-Ghanem C, Mansour FM, Kelly RD, Tyagi A, Brawley RR, Ogg JD, Zuloaga KL. High-fat diet exacerbates cognitive decline in mouse models of Alzheimer's disease and mixed dementia in a sex-dependent manner. J Neuroinflammation 2022; 19:110. [PMID: 35568928 PMCID: PMC9107741 DOI: 10.1186/s12974-022-02466-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 04/21/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Approximately 70% of Alzheimer's disease (AD) patients have co-morbid vascular contributions to cognitive impairment and dementia (VCID); this highly prevalent overlap of dementia subtypes is known as mixed dementia (MxD). AD is more prevalent in women, while VCID is slightly more prevalent in men. Sex differences in risk factors may contribute to sex differences in dementia subtypes. Unlike metabolically healthy women, diabetic women are more likely to develop VCID than diabetic men. Prediabetes is 3× more prevalent than diabetes and is linked to earlier onset of dementia in women, but not men. How prediabetes influences underlying pathology and cognitive outcomes across different dementia subtypes is unknown. To fill this gap in knowledge, we investigated the impact of diet-induced prediabetes and biological sex on cognitive function and neuropathology in mouse models of AD and MxD. METHODS Male and female 3xTg-AD mice received a sham (AD model) or unilateral common carotid artery occlusion surgery to induce chronic cerebral hypoperfusion (MxD model). Mice were fed a control or high fat (HF; 60% fat) diet from 3 to 7 months of age. In both sexes, HF diet elicited a prediabetic phenotype (impaired glucose tolerance) and weight gain. RESULTS In females, but not males, metabolic consequences of a HF diet were more severe in AD or MxD mice compared to WT. In both sexes, HF-fed AD or MxD mice displayed deficits in spatial memory in the Morris water maze (MWM). In females, but not males, HF-fed AD and MxD mice also displayed impaired spatial learning in the MWM. In females, but not males, AD or MxD caused deficits in activities of daily living, regardless of diet. Astrogliosis was more severe in AD and MxD females compared to males. Further, AD/MxD females had more amyloid beta plaques and hippocampal levels of insoluble amyloid beta 40 and 42 than AD/MxD males. In females, but not males, more severe glucose intolerance (prediabetes) was correlated with increased hippocampal microgliosis. CONCLUSIONS High-fat diet had a wider array of metabolic, cognitive, and neuropathological consequences in AD and MxD females compared to males. These findings shed light on potential underlying mechanisms by which prediabetes may lead to earlier dementia onset in women.
Collapse
Affiliation(s)
- Olivia J. Gannon
- grid.413558.e0000 0001 0427 8745Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY 12208 USA
| | - Lisa S. Robison
- grid.413558.e0000 0001 0427 8745Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY 12208 USA ,grid.261241.20000 0001 2168 8324Department of Psychology & Neuroscience, Nova Southeastern University, 3301 College Avenue, Fort Lauderdale, FL 33314 USA ,grid.264307.40000 0000 9688 1551Department of Psychology, Stetson University, 421 N Woodland Blvd, DeLand, FL 32723 USA
| | - Abigail E. Salinero
- grid.413558.e0000 0001 0427 8745Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY 12208 USA
| | - Charly Abi-Ghanem
- grid.413558.e0000 0001 0427 8745Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY 12208 USA
| | - Febronia M. Mansour
- grid.413558.e0000 0001 0427 8745Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY 12208 USA
| | - Richard D. Kelly
- grid.413558.e0000 0001 0427 8745Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY 12208 USA
| | - Alvira Tyagi
- grid.413558.e0000 0001 0427 8745Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY 12208 USA
| | - Rebekah R. Brawley
- grid.264307.40000 0000 9688 1551Department of Psychology, Stetson University, 421 N Woodland Blvd, DeLand, FL 32723 USA
| | - Jordan D. Ogg
- grid.264307.40000 0000 9688 1551Department of Psychology, Stetson University, 421 N Woodland Blvd, DeLand, FL 32723 USA
| | - Kristen L. Zuloaga
- grid.413558.e0000 0001 0427 8745Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY 12208 USA
| |
Collapse
|
26
|
Zou X, Yuan Y, Liao Y, Jiang C, Zhao F, Ding D, Gu Y, Chen L, Chu Y, Hsu Y, Liebig PA, Xu B, Mao Y. Moyamoya disease: A human model for chronic hypoperfusion and intervention in Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12285. [PMID: 35415209 PMCID: PMC8985488 DOI: 10.1002/trc2.12285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 02/10/2022] [Accepted: 03/03/2022] [Indexed: 11/07/2022]
Abstract
Introduction Chronic cerebral hypoperfusion has been considered the etiology for sporadic Alzheimer's disease (AD). However, no valid clinical evidence exists due to the similar risk factors between cerebrovascular disease and AD. Methods We used moyamoya disease (MMD) as a model of chronic hypoperfusion and cognitive impairment, without other etiology interference. Results Based on the previous reports and preliminary findings, we hypothesized that chronic cerebral hypoperfusion could be an independent upstream crucial variable, resulting in AD, and induce pathological hallmarks such as amyloid beta peptide and hyperphosphorylated tau accumulation. Discussion Timely intervention with revascularisation would help reverse the brain damage with AD hallmarks and lead to cognitive improvement.
Collapse
Affiliation(s)
- Xiang Zou
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
| | - Yifan Yuan
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
| | - Yujun Liao
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
| | - Conglin Jiang
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
| | - Fan Zhao
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
| | - Ding Ding
- Huashan HospitalInstitute of NeurologyFudan UniversityShanghaiChina
- National Clinical Research Center for Aging and MedicineHuashan HospitalFudan UniversityShanghaiChina
| | - Yuxiang Gu
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
| | - Liang Chen
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
- Tianqiao and Chrissy Chen International Institute for Brain DiseasesShanghaiChina
| | - Ying‐Hua Chu
- MR CollaborationSiemens Healthineers Ltd.ShanghaiChina
| | - Yi‐Cheng Hsu
- MR CollaborationSiemens Healthineers Ltd.ShanghaiChina
| | | | - Bin Xu
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
| | - Ying Mao
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
- Huashan HospitalInstitute of NeurologyFudan UniversityShanghaiChina
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceSchool of Basic Medical Sciences and Institutes of Brain ScienceFudan UniversityShanghaiChina
| |
Collapse
|
27
|
Ali M, Falkenhain K, Njiru BN, Murtaza-Ali M, Ruiz-Uribe NE, Haft-Javaherian M, Catchers S, Nishimura N, Schaffer CB, Bracko O. VEGF signalling causes stalls in brain capillaries and reduces cerebral blood flow in Alzheimer's mice. Brain 2022; 145:1449-1463. [PMID: 35048960 PMCID: PMC9150081 DOI: 10.1093/brain/awab387] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/09/2021] [Accepted: 09/22/2021] [Indexed: 11/16/2022] Open
Abstract
Increased incidence of stalled capillary blood flow caused by adhesion of
leucocytes to the brain microvascular endothelium leads to a 17%
reduction of cerebral blood flow and exacerbates short-term memory loss in
multiple mouse models of Alzheimer’s disease. Here, we report that vascular endothelial growth factor (VEGF) signalling at the
luminal side of the brain microvasculature plays an integral role in the
capillary stalling phenomenon of the APP/PS1 mouse model. Administration of the anti-mouse VEGF-A164 antibody, an isoform that inhibits
blood–brain barrier hyperpermeability, reduced the number of stalled
capillaries within an hour of injection, leading to an immediate increase in
average capillary blood flow but not capillary diameter. VEGF-A inhibition also
reduced the overall endothelial nitric oxide synthase protein concentrations,
increased occludin levels and decreased the penetration of circulating Evans
Blue dye across the blood–brain barrier into the brain parenchyma,
suggesting increased blood–brain barrier integrity. Capillaries prone to
neutrophil adhesion after anti-VEGF-A treatment also had lower occludin
concentrations than flowing capillaries. Taken together, our findings demonstrate that VEGF-A signalling in APP/PS1 mice
contributes to aberrant endothelial nitric oxide synthase /occludin-associated
blood–brain barrier permeability, increases the incidence of capillary
stalls, and leads to reductions in cerebral blood flow. Reducing leucocyte
adhesion by inhibiting luminal VEGF signalling may provide a novel and
well-tolerated strategy for improving brain microvascular blood flow in
Alzheimer’s disease patients.
Collapse
Affiliation(s)
- Muhammad Ali
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA, 148532
| | - Kaja Falkenhain
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA, 148532
| | - Brendah N Njiru
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA, 148532
| | - Muhammad Murtaza-Ali
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA, 148532
| | - Nancy E Ruiz-Uribe
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA, 148532
| | | | | | - Nozomi Nishimura
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA, 148532
| | - Chris B Schaffer
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA, 148532
| | - Oliver Bracko
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA, 148532
| |
Collapse
|
28
|
Chen P, Chen F, Lei J, Wang G, Zhou B. The Gut Microbiota Metabolite Urolithin B Improves Cognitive Deficits by Inhibiting Cyt C-Mediated Apoptosis and Promoting the Survival of Neurons Through the PI3K Pathway in Aging Mice. Front Pharmacol 2021; 12:768097. [PMID: 34867396 PMCID: PMC8634731 DOI: 10.3389/fphar.2021.768097] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/22/2021] [Indexed: 12/15/2022] Open
Abstract
Background: Despite considerable advances in pharmacotherapy, more effective therapeutic interventions for aging-related neurodegenerative disorders (NDs), such as Alzheimer’s disease (AD), remain limited. Urolithin B (UB), one of the major subcategories of urolithins (microbiota metabolites) found in various tissues after ellagitannin consumption, has been shown to possess antioxidant, anti-inflammatory, and antiapoptotic effects. However, the neuroprotective effect of UB on brain aging in mice and its potential mechanisms were still unknown. Methods: In the current research, we first assessed the ameliorative effects of UB on oxidative injury and apoptosis induced by H2O2 in neuro-2a cells. Then a subcutaneous injection of D-galactose in mice for 8 weeks was used to establish the aging model to evaluate the protective effects of UB. The capacity of memory and learning, alterations of hippocampus histology and corresponding molecular mechanisms were all evaluated. Results: The D-gal-induced accelerated aging model in vivo demonstrated that UB could significantly ameliorate deficits in learning and memory by inhibiting the accumulation of advanced glycation end products (AGEs) and elevating the expression and activity of Cu, Zn-SOD and CAT. Furthermore, UB downregulated the c-Jun N-terminal kinase (JNK) signaling pathway and prevented cytochrome c release from isolated mitochondria, thereby inhibiting neuronal apoptosis during the aging process. More importantly, UB stimulation of aging mice activated ERK and phosphoinositide 3-kinase (PI3K), leading to neuronal survival along with Akt and p44/42 mitogen-activated protein kinase (MAPK) phosphorylation and activation. Conclusion: In summary, UB effectively alleviated cognitive deficits and ameliorated brain aging-related conditions and could be considered a healthcare product to prevent aging-associated NDs such as AD.
Collapse
Affiliation(s)
- Peng Chen
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan, China
| | - Fuchao Chen
- Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, China.,Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Shiyan, China
| | - Jiexin Lei
- Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Gaohua Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Benhong Zhou
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan, China.,Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
29
|
Solovyev R, Kalinin AA, Gabruseva T. 3D convolutional neural networks for stalled brain capillary detection. Comput Biol Med 2021; 141:105089. [PMID: 34920160 DOI: 10.1016/j.compbiomed.2021.105089] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 11/26/2021] [Accepted: 11/26/2021] [Indexed: 01/08/2023]
Abstract
Adequate blood supply is critical for normal brain function. Brain vasculature dysfunctions, including stalled blood flow in cerebral capillaries, are associated with cognitive decline and pathogenesis in Alzheimer's disease. Recent advances in imaging technology enabled generation of high-quality 3D images that can be used to visualize stalled blood vessels. However, localization of stalled vessels in 3D images is often required as the first step for downstream analysis. When performed manually, this process is tedious, time-consuming, and error-prone. Here, we describe a deep learning-based approach for automatic detection of stalled capillaries in brain images based on 3D convolutional neural networks. Our approach includes custom 3D data augmentations and a weights transfer method that re-uses weights from 2D models pre-trained on natural images for initialization of 3D networks. We used an ensemble of several 3D models to produce the winning submission to the "Clog Loss: Advance Alzheimer's Research with Stall Catchers" machine learning competition that challenged the participants with classifying blood vessels in 3D image stacks as stalled or flowing. In this setting, our approach outperformed other methods and demonstrated state-of-the-art results, achieving 85% Matthews correlation coefficient, 85% sensitivity, and 99.3% specificity. The source code for our solution is publicly available.
Collapse
Affiliation(s)
- Roman Solovyev
- Institute for Design Problems in Microelectronics of Russian Academy of Sciences, 3, Sovetskaya Street, Moscow, 124 365, Russian Federation.
| | - Alexandr A Kalinin
- Shenzhen Research Institute of Big Data, Shenzhen, 518 172, Guangdong, China; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48 109, USA
| | | |
Collapse
|
30
|
Frausto DM, Forsyth CB, Keshavarzian A, Voigt RM. Dietary Regulation of Gut-Brain Axis in Alzheimer's Disease: Importance of Microbiota Metabolites. Front Neurosci 2021; 15:736814. [PMID: 34867153 PMCID: PMC8639879 DOI: 10.3389/fnins.2021.736814] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/18/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that impacts 45 million people worldwide and is ranked as the 6th top cause of death among all adults by the Centers for Disease Control and Prevention. While genetics is an important risk factor for the development of AD, environment and lifestyle are also contributing risk factors. One such environmental factor is diet, which has emerged as a key influencer of AD development/progression as well as cognition. Diets containing large quantities of saturated/trans-fats, refined carbohydrates, limited intake of fiber, and alcohol are associated with cognitive dysfunction while conversely diets low in saturated/trans-fats (i.e., bad fats), high mono/polyunsaturated fats (i.e., good fats), high in fiber and polyphenols are associated with better cognitive function and memory in both humans and animal models. Mechanistically, this could be the direct consequence of dietary components (lipids, vitamins, polyphenols) on the brain, but other mechanisms are also likely to be important. Diet is considered to be the single greatest factor influencing the intestinal microbiome. Diet robustly influences the types and function of micro-organisms (called microbiota) that reside in the gastrointestinal tract. Availability of different types of nutrients (from the diet) will favor or disfavor the abundance and function of certain groups of microbiota. Microbiota are highly metabolically active and produce many metabolites and other factors that can affect the brain including cognition and the development and clinical progression of AD. This review summarizes data to support a model in which microbiota metabolites influence brain function and AD.
Collapse
Affiliation(s)
- Dulce M. Frausto
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Christopher B. Forsyth
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
| | - Ali Keshavarzian
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
- Department of Physiology, Rush University Medical Center, Chicago, IL, United States
| | - Robin M. Voigt
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
| |
Collapse
|
31
|
Zhou R, Chen LL, Yang H, Li L, Liu J, Chen L, Hong WJ, Wang CG, Ma JJ, Huang J, Zhou XF, Liu D, Zhou HD. Effect of High Cholesterol Regulation of LRP1 and RAGE on Aβ Transport Across the Blood-Brain Barrier in Alzheimer's Disease. Curr Alzheimer Res 2021; 18:428-442. [PMID: 34488598 DOI: 10.2174/1567205018666210906092940] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 04/01/2021] [Accepted: 06/09/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND High cholesterol aggravates the risk development of Alzheimer's disease (AD). AD is closely related to the transport impairment of Amyloid-β (Aβ) in the blood-brain barrier. It is unclear whether high cholesterol affects the risk of cognitive impairment in AD by affecting Aβ transport. The purpose of the study is to investigate whether high cholesterol regulates Aβ transport through low-density Lipoprotein Receptor-Related Protein 1 (LRP1) and Receptor for Advanced Glycation End products (RAGE) in the risk development of AD. METHODS We established high cholesterol AD mice model. The learning and memory functions were evaluated by Morris Water Maze (MWM). Cerebral microvascular endothelial cells were isolated, cultured, and observed. The expression levels of LRP1 and RAGE of endothelial cells and their effect on Aβ transport in vivo were observed. The expression level of LRP1 and RAGE was detected in cultured microvessels after using Wnt inhibitor DKK-1 and β-catenin inhibitor XAV-939. RESULTS Hypercholesterolemia exacerbated spatial learning and memory impairment. Hypercholesterolemia increased serum Aβ40 level, while serum Aβ42 level did not change significantly. Hypercholesterolemia decreased LRP1 expression and increased RAGE expression in cerebral microvascular endothelial cells. Hypercholesterolemia increased brain apoptosis in AD mice. In in vitro experiment, high cholesterol decreased LRP1 expression and increased RAGE expression, increased Aβ40 expression in cerebral microvascular endothelial cells. High cholesterol regulated the expressions of LRP1 and RAGE and transcriptional activity of LRP1 and RAGE promoters by the Wnt/β-catenin signaling pathway. CONCLUSION High cholesterol decreased LRP1 expression and increased RAGE expression in cerebral microvascular endothelial cells, which led to Aβ transport disorder in the blood-brain barrier. Increased Aβ deposition in the brain aggravated apoptosis in the brain, resulting to cognitive impairment of AD mice.
Collapse
Affiliation(s)
- Rui Zhou
- Department of Orthopedics, The Orthopedic Surgery Center of Chinese PLA, Southwest Hospital, Army Medical University, Chongqing 400042, China
| | - Li-Li Chen
- Department of Neurology, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Hai Yang
- Department of Neurology, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Ling Li
- Department of Neurology, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Juan Liu
- Department of Neurology, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Le Chen
- Postgraduate School, Bengbu Medical College, Anhui 233004, China
| | - Wen-Juan Hong
- Postgraduate School, Bengbu Medical College, Anhui 233004, China
| | - Cong-Guo Wang
- Postgraduate School, Bengbu Medical College, Anhui 233004, China
| | - Jing-Jing Ma
- Postgraduate School, Bengbu Medical College, Anhui 233004, China
| | - Jie Huang
- Postgraduate School, Bengbu Medical College, Anhui 233004, China
| | - Xin-Fu Zhou
- School of Pharmacy and Medical Sciences and Sansom Institute, University of South Australia, Adelaide, SA, Australia
| | - Dong Liu
- Laboratory of Field Surgery Institute, Army Medical University, Chongqing 400042, China
| | - Hua-Dong Zhou
- Department of Neurology, Daping Hospital, Army Medical University, Chongqing 400042, China
| |
Collapse
|
32
|
Reveglia P, Paolillo C, Ferretti G, De Carlo A, Angiolillo A, Nasso R, Caputo M, Matrone C, Di Costanzo A, Corso G. Challenges in LC-MS-based metabolomics for Alzheimer's disease early detection: targeted approaches versus untargeted approaches. Metabolomics 2021; 17:78. [PMID: 34453619 PMCID: PMC8403122 DOI: 10.1007/s11306-021-01828-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 08/06/2021] [Indexed: 01/22/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is one of the most common causes of dementia in old people. Neuronal deficits such as loss of memory, language and problem-solving are severely compromised in affected patients. The molecular features of AD are Aβ deposits in plaques or in oligomeric structures and neurofibrillary tau tangles in brain. However, the challenge is that Aβ is only one piece of the puzzle, and recent findings continue to support the hypothesis that their presence is not sufficient to predict decline along the AD outcome. In this regard, metabolomic-based techniques are acquiring a growing interest for either the early diagnosis of diseases or the therapy monitoring. Mass spectrometry is one the most common analytical platforms used for detection, quantification, and characterization of metabolic biomarkers. In the past years, both targeted and untargeted strategies have been applied to identify possible interesting compounds. AIM OF REVIEW The overall goal of this review is to guide the reader through the most recent studies in which LC-MS-based metabolomics has been proposed as a powerful tool for the identification of new diagnostic biomarkers in AD. To this aim, herein studies spanning the period 2009-2020 have been reported. Advantages and disadvantages of targeted vs untargeted metabolomic approaches have been outlined and critically discussed.
Collapse
Affiliation(s)
- Pierluigi Reveglia
- Department of Clinical and Experimental Medicine, University of Foggia, 71122, Foggia, Italy
| | - Carmela Paolillo
- Department of Clinical and Experimental Medicine, University of Foggia, 71122, Foggia, Italy
| | - Gabriella Ferretti
- Department of Neuroscience, School of Medicine, University of Naples Federico II, 80131, Napoli, Italy
| | - Armando De Carlo
- Department of Clinical and Experimental Medicine, University of Foggia, 71122, Foggia, Italy
- Policlinico Riuniti University Hospital, 71122, Foggia, Italy
| | - Antonella Angiolillo
- Department of Medicine and Health Sciences, Center for Research and Training in Aging Medicine, University of Molise, 86100, Campobasso, Italy
| | - Rosarita Nasso
- Department of Neuroscience, School of Medicine, University of Naples Federico II, 80131, Napoli, Italy
| | - Mafalda Caputo
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131, Napoli, Italy
| | - Carmela Matrone
- Department of Neuroscience, School of Medicine, University of Naples Federico II, 80131, Napoli, Italy
| | - Alfonso Di Costanzo
- Department of Medicine and Health Sciences, Center for Research and Training in Aging Medicine, University of Molise, 86100, Campobasso, Italy
| | - Gaetano Corso
- Department of Clinical and Experimental Medicine, University of Foggia, 71122, Foggia, Italy.
- Policlinico Riuniti University Hospital, 71122, Foggia, Italy.
| |
Collapse
|
33
|
Mazzei G, Ikegami R, Abolhassani N, Haruyama N, Sakumi K, Saito T, Saido TC, Nakabeppu Y. A high-fat diet exacerbates the Alzheimer's disease pathology in the hippocampus of the App NL-F/NL-F knock-in mouse model. Aging Cell 2021; 20:e13429. [PMID: 34245097 PMCID: PMC8373331 DOI: 10.1111/acel.13429] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 06/01/2021] [Accepted: 06/15/2021] [Indexed: 12/14/2022] Open
Abstract
Insulin resistance and diabetes mellitus are major risk factors for Alzheimer's disease (AD), and studies with transgenic mouse models of AD have provided supportive evidence with some controversies. To overcome potential artifacts derived from transgenes, we used a knock‐in mouse model, AppNL−F/NL−F, which accumulates Aβ plaques from 6 months of age and shows mild cognitive impairment at 18 months of age, without the overproduction of APP. In the present study, 6‐month‐old male AppNL−F/NL−F and wild‐type mice were fed a regular or high‐fat diet (HFD) for 12 months. HFD treatment caused obesity and impaired glucose tolerance (i.e., T2DM conditions) in both wild‐type and AppNL−F/NL−F mice, but only the latter animals exhibited an impaired cognitive function accompanied by marked increases in both Aβ deposition and microgliosis as well as insulin resistance in the hippocampus. Furthermore, HFD‐fed AppNL−F/NL−F mice exhibited a significant decrease in volume of the granule cell layer in the dentate gyrus and an increased accumulation of 8‐oxoguanine, an oxidized guanine base, in the nuclei of granule cells. Gene expression profiling by microarrays revealed that the populations of the cell types in hippocampus were not significantly different between the two mouse lines, regardless of the diet. In addition, HFD treatment decreased the expression of the Aβ binding protein transthyretin (TTR) in AppNL−F/NL−F mice, suggesting that the depletion of TTR underlies the increased Aβ deposition in the hippocampus of HFD‐fed AppNL−F/NL−F mice.
Collapse
Affiliation(s)
- Guianfranco Mazzei
- Division of Neurofunctional Genomics Department of Immunobiology and Neuroscience Medical Institute of Bioregulation Kyushu University Fukuoka Japan
| | - Ryohei Ikegami
- Division of Neurofunctional Genomics Department of Immunobiology and Neuroscience Medical Institute of Bioregulation Kyushu University Fukuoka Japan
| | - Nona Abolhassani
- Division of Neurofunctional Genomics Department of Immunobiology and Neuroscience Medical Institute of Bioregulation Kyushu University Fukuoka Japan
| | - Naoki Haruyama
- Division of Neurofunctional Genomics Department of Immunobiology and Neuroscience Medical Institute of Bioregulation Kyushu University Fukuoka Japan
| | - Kunihiko Sakumi
- Division of Neurofunctional Genomics Department of Immunobiology and Neuroscience Medical Institute of Bioregulation Kyushu University Fukuoka Japan
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience RIKEN Center for Brain Science Saitama Japan
- Department of Neurocognitive Science Institute of Brain Science Nagoya City University Graduate School of Medical Sciences Nagoya Japan
| | - Takaomi C. Saido
- Laboratory for Proteolytic Neuroscience RIKEN Center for Brain Science Saitama Japan
| | - Yusaku Nakabeppu
- Division of Neurofunctional Genomics Department of Immunobiology and Neuroscience Medical Institute of Bioregulation Kyushu University Fukuoka Japan
| |
Collapse
|
34
|
Sharma S. High fat diet and its effects on cognitive health: alterations of neuronal and vascular components of brain. Physiol Behav 2021; 240:113528. [PMID: 34260890 DOI: 10.1016/j.physbeh.2021.113528] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 07/03/2021] [Accepted: 07/06/2021] [Indexed: 01/01/2023]
Abstract
It has been well recognized that intake of diets rich in saturated fats could result in development of metabolic disorders such as type 2 diabetes mellitus, obesity and cardiovascular diseases. Recent studies have suggested that intake of high fat diet (HFD) is also associated with cognitive dysfunction. Various preclinical studies have demonstrated the impact of short and long term HFD feeding on the biochemical and behavioural alterations. This review summarizes studies and the protocols used to assess the impacts of HFD feeding on cognitive performance in rodents. Further, it discuss the key mechanisms that are altered by HFD feeding, such as, insulin resistance, oxidative stress, neuro-inflammation, transcriptional dysregulation and loss of synaptic plasticity. Along with these, HFD feeding also alters the vascular components of brain such as loss of BBB integrity and reduced cerebral blood flow. It is highly possible that these factors are responsible for the development of cognitive deficits as a result of HFD feeding.
Collapse
Affiliation(s)
- Sorabh Sharma
- Division of Medical Sciences, University of Victoria, PO Box 1700 STN CSC, Victoria, BC, V8W2Y2, Canada.
| |
Collapse
|
35
|
Sferra R, Pompili S, Cappariello A, Gaudio E, Latella G, Vetuschi A. Prolonged Chronic Consumption of a High Fat with Sucrose Diet Alters the Morphology of the Small Intestine. Int J Mol Sci 2021; 22:ijms22147280. [PMID: 34298894 PMCID: PMC8303301 DOI: 10.3390/ijms22147280] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 02/07/2023] Open
Abstract
(1) The high-fat diet (HFD) of western countries has dramatic effect on the health of several organs, including the digestive tract, leading to the accumulation of fats that can also trigger a chronic inflammatory process, such as that which occurs in non-alcohol steatohepatitis. The effects of a HFD on the small intestine, the organ involved in the absorption of this class of nutrients, are still poorly investigated. (2) To address this aspect, we administered a combined HFD with sucrose (HFD w/Suc, fat: 58% Kcal) regimen (18 months) to mice and investigated the morphological and molecular changes that occurred in the wall of proximal tract of the small intestine compared to the intestine of mice fed with a standard diet (SD) (fat: 18% Kcal). (3) We found an accumulation of lipid droplets in the mucosa of HFD w/Suc-fed mice that led to a disarrangement of mucosa architecture. Furthermore, we assessed the expression of several key players involved in lipid metabolism and inflammation, such as perilipin, leptin, leptin receptor, PI3K, p-mTOR, p-Akt, and TNF-α. All these molecules were increased in HFD mice compared to the SD group. We also evaluated anti-inflammatory molecules like adiponectin, adiponectin receptor, and PPAR-γ, and observed their significant reduction in the HFD w/Suc group compared to the control. Our data are in line with the knowledge that improper eating habits present a primary harmful assault on the bowel and the entire body's health. (4) These results represent a promising starting point for future studies, helping to better understand the complex and not fully elucidated spectrum of intestinal alterations induced by the overconsumption of fat.
Collapse
Affiliation(s)
- Roberta Sferra
- Department of Biotechnological and Applied Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.P.); (A.C.); (A.V.)
- Correspondence: ; Tel.: +39-0862-433504
| | - Simona Pompili
- Department of Biotechnological and Applied Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.P.); (A.C.); (A.V.)
| | - Alfredo Cappariello
- Department of Biotechnological and Applied Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.P.); (A.C.); (A.V.)
| | - Eugenio Gaudio
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences, Sapienza University of Rome, 00161 Rome, Italy;
| | - Giovanni Latella
- Department of Life, Health and Environmental Sciences, Division of Gastroenterology, Hepatology, and Nutrition, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Antonella Vetuschi
- Department of Biotechnological and Applied Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.P.); (A.C.); (A.V.)
| |
Collapse
|
36
|
Bracko O, Cruz Hernández JC, Park L, Nishimura N, Schaffer CB. Causes and consequences of baseline cerebral blood flow reductions in Alzheimer's disease. J Cereb Blood Flow Metab 2021; 41:1501-1516. [PMID: 33444096 PMCID: PMC8221770 DOI: 10.1177/0271678x20982383] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/27/2020] [Accepted: 11/16/2020] [Indexed: 12/23/2022]
Abstract
Reductions of baseline cerebral blood flow (CBF) of ∼10-20% are a common symptom of Alzheimer's disease (AD) that appear early in disease progression and correlate with the severity of cognitive impairment. These CBF deficits are replicated in mouse models of AD and recent work shows that increasing baseline CBF can rapidly improve the performance of AD mice on short term memory tasks. Despite the potential role these data suggest for CBF reductions in causing cognitive symptoms and contributing to brain pathology in AD, there remains a poor understanding of the molecular and cellular mechanisms causing them. This review compiles data on CBF reductions and on the correlation of AD-related CBF deficits with disease comorbidities (e.g. cardiovascular and genetic risk factors) and outcomes (e.g. cognitive performance and brain pathology) from studies in both patients and mouse models, and discusses several potential mechanisms proposed to contribute to CBF reductions, based primarily on work in AD mouse models. Future research aimed at improving our understanding of the importance of and interplay between different mechanisms for CBF reduction, as well as at determining the role these mechanisms play in AD patients could guide the development of future therapies that target CBF reductions in AD.
Collapse
Affiliation(s)
- Oliver Bracko
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Jean C Cruz Hernández
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Laibaik Park
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, USA
| | - Nozomi Nishimura
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Chris B Schaffer
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| |
Collapse
|
37
|
Amelianchik A, Merkel J, Palanisamy P, Kaneki S, Hyatt E, Norris EH. The protective effect of early dietary fat consumption on Alzheimer's disease-related pathology and cognitive function in mice. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 7:e12173. [PMID: 34084889 PMCID: PMC8144936 DOI: 10.1002/trc2.12173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 03/24/2021] [Accepted: 04/05/2021] [Indexed: 11/12/2022]
Abstract
INTRODUCTION It has been suggested that obesity may influence Alzheimer's disease (AD) pathogenesis, yet the numerous publications on this topic have inconsistent results and conclusions. METHODS Our study examined the effect of varying the timing of high-fat diet (HFD) consumption on AD-related pathology and cognition in transgenic Tg6799 AD mice. RESULTS HFD feeding starting at or before 3 months of age, prior to severe AD pathology, had protective effects in AD mice: reduced extracellular amyloid beta (Aβ) deposition, decreased fibrinogen extravasation into the brain parenchyma, and improved cognitive function. However, delaying HFD consumption until 6 months of age, when AD pathology is ubiquitous, reduced these protective effects in AD mice. DISCUSSION Overall, we demonstrate that the timeline of HFD consumption may play an important role in how dietary fats affect AD pathogenesis and cognitive function.
Collapse
Affiliation(s)
- Anna Amelianchik
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkUSA
| | - Jonathan Merkel
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkUSA
- Paul Flechsig Institute of Brain ResearchLeipzig UniversityLeipzigGermany
| | - Premkumar Palanisamy
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkUSA
| | - Shigeru Kaneki
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkUSA
| | - Emily Hyatt
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkUSA
| | - Erin H. Norris
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkUSA
| |
Collapse
|
38
|
Vinuesa A, Pomilio C, Gregosa A, Bentivegna M, Presa J, Bellotto M, Saravia F, Beauquis J. Inflammation and Insulin Resistance as Risk Factors and Potential Therapeutic Targets for Alzheimer's Disease. Front Neurosci 2021; 15:653651. [PMID: 33967682 PMCID: PMC8102834 DOI: 10.3389/fnins.2021.653651] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/31/2021] [Indexed: 12/21/2022] Open
Abstract
Overnutrition and modern diets containing high proportions of saturated fat are among the major factors contributing to a low-grade state of inflammation, hyperglycemia and dyslipidemia. In the last decades, the global rise of type 2 diabetes and obesity prevalence has elicited a great interest in understanding how changes in metabolic function lead to an increased risk for premature brain aging and the development of neurodegenerative disorders such as Alzheimer's disease (AD). Cognitive impairment and decreased neurogenic capacity could be a consequence of metabolic disturbances. In these scenarios, the interplay between inflammation and insulin resistance could represent a potential therapeutic target to prevent or ameliorate neurodegeneration and cognitive impairment. The present review aims to provide an update on the impact of metabolic stress pathways on AD with a focus on inflammation and insulin resistance as risk factors and therapeutic targets.
Collapse
Affiliation(s)
- Angeles Vinuesa
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carlos Pomilio
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Amal Gregosa
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Melisa Bentivegna
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Jessica Presa
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Melina Bellotto
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Flavia Saravia
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Juan Beauquis
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
39
|
Cho S, Lee H, Seo J. Impact of Genetic Risk Factors for Alzheimer's Disease on Brain Glucose Metabolism. Mol Neurobiol 2021; 58:2608-2619. [PMID: 33479841 DOI: 10.1007/s12035-021-02297-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/13/2021] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disease that affects more than 30 million people worldwide. Despite growing knowledge of AD pathophysiology, a complete understanding of the pathogenic mechanisms underpinning AD is lacking, and there is currently no cure for AD. Extant literature suggests that AD is a polygenic and multifactorial disease underscored by complex and dynamic pathogenic mechanisms. Despite extensive research and clinical trials, there has been a dearth of novel drugs for AD treatment on the market since memantine in 2003. This lack of therapeutic success has directed the entire research community to approach the disease from a different angle. In this review, we discuss growing evidence for the close link between altered glucose metabolism and AD pathogenesis by exploring how genetic risk factors for AD are associated with dysfunctional glucose metabolism. We also discuss modification of genes responsible for metabolic pathways implicated in AD pathology.
Collapse
Affiliation(s)
- Sukhee Cho
- Department of Brain and Cognitive Sciences, DGIST, Daegu, 42988, South Korea
| | - Hyein Lee
- Department of Brain and Cognitive Sciences, DGIST, Daegu, 42988, South Korea
| | - Jinsoo Seo
- Department of Brain and Cognitive Sciences, DGIST, Daegu, 42988, South Korea.
| |
Collapse
|
40
|
de Bem AF, Krolow R, Farias HR, de Rezende VL, Gelain DP, Moreira JCF, Duarte JMDN, de Oliveira J. Animal Models of Metabolic Disorders in the Study of Neurodegenerative Diseases: An Overview. Front Neurosci 2021; 14:604150. [PMID: 33536868 PMCID: PMC7848140 DOI: 10.3389/fnins.2020.604150] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/24/2020] [Indexed: 12/21/2022] Open
Abstract
The incidence of metabolic disorders, as well as of neurodegenerative diseases—mainly the sporadic forms of Alzheimer’s and Parkinson’s disease—are increasing worldwide. Notably, obesity, diabetes, and hypercholesterolemia have been indicated as early risk factors for sporadic forms of Alzheimer’s and Parkinson’s disease. These conditions share a range of molecular and cellular features, including protein aggregation, oxidative stress, neuroinflammation, and blood-brain barrier dysfunction, all of which contribute to neuronal death and cognitive impairment. Rodent models of obesity, diabetes, and hypercholesterolemia exhibit all the hallmarks of these degenerative diseases, and represent an interesting approach to the study of the phenotypic features and pathogenic mechanisms of neurodegenerative disorders. We review the main pathological aspects of Alzheimer’s and Parkinson’s disease as summarized in rodent models of obesity, diabetes, and hypercholesterolemia.
Collapse
Affiliation(s)
- Andreza Fabro de Bem
- Department of Physiological Sciences, Institute of Biology, University of Brasilia, Brazilia, Brazil
| | - Rachel Krolow
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Hémelin Resende Farias
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Victória Linden de Rezende
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Daniel Pens Gelain
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - José Cláudio Fonseca Moreira
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - João Miguel das Neves Duarte
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Jade de Oliveira
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
41
|
Natunen T, Martiskainen H, Marttinen M, Gabbouj S, Koivisto H, Kemppainen S, Kaipainen S, Takalo M, Svobodová H, Leppänen L, Kemiläinen B, Ryhänen S, Kuulasmaa T, Rahunen E, Juutinen S, Mäkinen P, Miettinen P, Rauramaa T, Pihlajamäki J, Haapasalo A, Leinonen V, Tanila H, Hiltunen M. Diabetic phenotype in mouse and humans reduces the number of microglia around β-amyloid plaques. Mol Neurodegener 2020; 15:66. [PMID: 33168021 PMCID: PMC7653710 DOI: 10.1186/s13024-020-00415-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 10/26/2020] [Indexed: 02/08/2023] Open
Abstract
Background Alzheimer’s disease (AD) is the most common neurodegenerative disease and type 2 diabetes (T2D) plays an important role in conferring the risk for AD. Although AD and T2D share common features, the common molecular mechanisms underlying these two diseases remain elusive. Methods Mice with different AD- and/or tauopathy-linked genetic backgrounds (APPswe/PS1dE9, Tau P301L and APPswe/PS1dE9/Tau P301L) were fed for 6 months with standard diet or typical Western diet (TWD). After behavioral and metabolic assessments of the mice, the effects of TWD on global gene expression as well as dystrophic neurite and microglia pathology were elucidated. Consequently, mechanistic aspects related to autophagy, cell survival, phagocytic uptake as well as Trem2/Dap12 signaling pathway, were assessed in microglia upon modulation of PI3K-Akt signaling. To evaluate whether the mouse model-derived results translate to human patients, the effects of diabetic phenotype on microglial pathology were assessed in cortical biopsies of idiopathic normal pressure hydrocephalus (iNPH) patients encompassing β-amyloid pathology. Results TWD led to obesity and diabetic phenotype in all mice regardless of the genetic background. TWD also exacerbated memory and learning impairment in APPswe/PS1dE9 and Tau P301L mice. Gene co-expression network analysis revealed impaired microglial responses to AD-related pathologies in APPswe/PS1dE9 and APPswe/PS1dE9/Tau P301L mice upon TWD, pointing specifically towards aberrant microglial functionality due to altered downstream signaling of Trem2 and PI3K-Akt. Accordingly, fewer microglia, which did not show morphological changes, and increased number of dystrophic neurites around β-amyloid plaques were discovered in the hippocampus of TWD mice. Mechanistic studies in mouse microglia revealed that interference of PI3K-Akt signaling significantly decreased phagocytic uptake and proinflammatory response. Moreover, increased activity of Syk-kinase upon ligand-induced activation of Trem2/Dap12 signaling was detected. Finally, characterization of microglial pathology in cortical biopsies of iNPH patients revealed a significant decrease in the number of microglia per β-amyloid plaque in obese individuals with concomitant T2D as compared to both normal weight and obese individuals without T2D. Conclusions Collectively, these results suggest that diabetic phenotype in mice and humans mechanistically associates with abnormally reduced microglial responses to β-amyloid pathology and further suggest that AD and T2D share overlapping pathomechanisms, likely involving altered immune function in the brain. Supplementary Information The online version contains supplementary material available at 10.1186/s13024-020-00415-2.
Collapse
Affiliation(s)
- Teemu Natunen
- Institute of Biomedicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Henna Martiskainen
- Institute of Biomedicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Mikael Marttinen
- Institute of Biomedicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Sami Gabbouj
- Institute of Biomedicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Hennariikka Koivisto
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Susanna Kemppainen
- Institute of Biomedicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Satu Kaipainen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Mari Takalo
- Institute of Biomedicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Helena Svobodová
- Department of Simulation and Virtual Medical Education, Faculty of Medicine, Comenius University, Bratislava, Slovak Republic
| | - Luukas Leppänen
- Institute of Biomedicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Benjam Kemiläinen
- Department of Neurosurgery, Kuopio University Hospital, and Institute of Clinical Medicine, Unit of Neurosurgery, University of Eastern Finland, Kuopio, Finland
| | - Simo Ryhänen
- Institute of Biomedicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Teemu Kuulasmaa
- Institute of Biomedicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Eija Rahunen
- Institute of Biomedicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Sisko Juutinen
- Institute of Biomedicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Petra Mäkinen
- Institute of Biomedicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Pasi Miettinen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Tuomas Rauramaa
- Department of Pathology, Kuopio University Hospital, and Institute of Clinical Medicine, Unit of Pathology, University of Eastern Finland, Kuopio, Finland
| | - Jussi Pihlajamäki
- Department of Clinical Nutrition, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Annakaisa Haapasalo
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Ville Leinonen
- Department of Neurosurgery, Kuopio University Hospital, and Institute of Clinical Medicine, Unit of Neurosurgery, University of Eastern Finland, Kuopio, Finland
| | - Heikki Tanila
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Mikko Hiltunen
- Institute of Biomedicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.
| |
Collapse
|
42
|
Robison LS, Gannon OJ, Thomas MA, Salinero AE, Abi-Ghanem C, Poitelon Y, Belin S, Zuloaga KL. Role of sex and high-fat diet in metabolic and hypothalamic disturbances in the 3xTg-AD mouse model of Alzheimer's disease. J Neuroinflammation 2020; 17:285. [PMID: 32993686 PMCID: PMC7526387 DOI: 10.1186/s12974-020-01956-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 09/18/2020] [Indexed: 12/14/2022] Open
Abstract
Background Hypothalamic dysfunction occurs early in the clinical course of Alzheimer’s disease (AD), likely contributing to disturbances in feeding behavior and metabolic function that are often observed years prior to the onset of cognitive symptoms. Late-life weight loss and low BMI are associated with increased risk of dementia and faster progression of disease. However, high-fat diet and metabolic disease (e.g., obesity, type 2 diabetes), particularly in mid-life, are associated with increased risk of AD, as well as exacerbated AD pathology and behavioral deficits in animal models. In the current study, we explored possible relationships between hypothalamic function, diet/metabolic status, and AD. Considering the sex bias in AD, with women representing two-thirds of AD patients, we sought to determine whether these relationships vary by sex. Methods WT and 3xTg-AD male and female mice were fed a control (10% fat) or high-fat (HF 60% fat) diet from ~ 3–7 months of age, then tested for metabolic and hypothalamic disturbances. Results On control diet, male 3xTg-AD mice displayed decreased body weight, reduced fat mass, hypoleptinemia, and mild systemic inflammation, as well as increased expression of gliosis- and inflammation-related genes in the hypothalamus (Iba1, GFAP, TNF-α, IL-1β). In contrast, female 3xTg-AD mice on control diet displayed metabolic disturbances opposite that of 3xTg-AD males (increased body and fat mass, impaired glucose tolerance). HF diet resulted in expected metabolic alterations across groups (increased body and fat mass; glucose intolerance; increased plasma insulin and leptin, decreased ghrelin; nonalcoholic fatty liver disease-related pathology). HF diet resulted in the greatest weight gain, adiposity, and glucose intolerance in 3xTg-AD females, which were associated with markedly increased hypothalamic expression of GFAP and IL-1β, as well as GFAP labeling in several hypothalamic nuclei that regulate energy balance. In contrast, HF diet increased diabetes markers and systemic inflammation preferentially in AD males but did not exacerbate hypothalamic inflammation in this group. Conclusions These findings provide further evidence for the roles of hypothalamic and metabolic dysfunction in AD, which in the 3xTg-AD mouse model appears to be dependent on both sex and diet.
Collapse
Affiliation(s)
- Lisa S Robison
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Olivia J Gannon
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Melissa A Thomas
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Abigail E Salinero
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Charly Abi-Ghanem
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Yannick Poitelon
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Sophie Belin
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Kristen L Zuloaga
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA.
| |
Collapse
|