1
|
Zeng J, Cheong LYT, Lo CH. Therapeutic targeting of obesity-induced neuroinflammation and neurodegeneration. Front Endocrinol (Lausanne) 2025; 15:1456948. [PMID: 39897964 PMCID: PMC11781992 DOI: 10.3389/fendo.2024.1456948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 12/30/2024] [Indexed: 02/04/2025] Open
Abstract
Obesity is a major modifiable risk factor leading to neuroinflammation and neurodegeneration. Excessive fat storage in obesity promotes the progressive infiltration of immune cells into adipose tissue, resulting in the release of pro-inflammatory factors such as cytokines and adipokines. These inflammatory mediators circulate through the bloodstream, propagating inflammation both in the periphery and in the central nervous system. Gut dysbiosis, which results in a leaky intestinal barrier, exacerbates inflammation and plays a significant role in linking obesity to the pathogenesis of neuroinflammation and neurodegeneration through the gut-brain/gut-brain-liver axis. Inflammatory states within the brain can lead to insulin resistance, mitochondrial dysfunction, autolysosomal dysfunction, and increased oxidative stress. These disruptions impair normal neuronal function and subsequently lead to cognitive decline and motor deficits, similar to the pathologies observed in major neurodegenerative diseases, including Alzheimer's disease, multiple sclerosis, and Parkinson's disease. Understanding the underlying disease mechanisms is crucial for developing therapeutic strategies to address defects in these inflammatory and metabolic pathways. In this review, we summarize and provide insights into different therapeutic strategies, including methods to alter gut dysbiosis, lifestyle changes, dietary supplementation, as well as pharmacological agents derived from natural sources, that target obesity-induced neuroinflammation and neurodegeneration.
Collapse
Affiliation(s)
- Jialiu Zeng
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, United States
- Interdisciplinary Neuroscience Program, Syracuse University, Syracuse, NY, United States
| | - Lenny Yi Tong Cheong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Chih Hung Lo
- Interdisciplinary Neuroscience Program, Syracuse University, Syracuse, NY, United States
- Department of Biology, Syracuse University, Syracuse, NY, United States
| |
Collapse
|
2
|
Reitsema VA, Schreuder L, Gerrits E, Eggen BJL, Goris M, Laman JD, de Rooij SE, Wesseling EM, Bouma HR, Henning RH. Calorie restriction increases the sensitivity of progeroid Ercc1 Δ/- mice to acute (neuro)inflammation. GeroScience 2024:10.1007/s11357-024-01347-1. [PMID: 39287878 DOI: 10.1007/s11357-024-01347-1] [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: 06/07/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024] Open
Abstract
Hospitalized elderly patients frequently suffer from delirium, especially in the context of sepsis-associated encephalopathy. Current treatments of delirium are merely symptomatic. Calorie restriction (CR) is both a promising strategy to protect against sepsis and has beneficial effects on aging-induced neurodegeneration. In this study, we investigated whether six weeks of 30% CR had protective effects on lipopolysaccharide (LPS) induced (neuro)inflammation in wild-type (WT) and progeroid mice deficient in the DNA excision-repair gene Ercc1 (Ercc1Δ/-). While CR did not affect the LPS-induced inflammatory response in WT mice, CR exaggerated the peripheral inflammatory response in Ercc1Δ/- mice, as evidenced by an increase of pro-inflammatory serum cytokines (TNF-α, IL-1β, and IFN-γ) and kidney injury marker Ngal. Neuroinflammatory effects were assessed by RNA-sequencing of isolated microglia. Similarly, CR did not affect microglia gene expression in WT mice, but increased neuroinflammation-associated gene expression in Ercc1Δ/- mice. In conclusion, CR increases the peripheral and brain inflammatory response of Ercc1Δ/- mice to a systemic inflammatory stimulus.
Collapse
Affiliation(s)
- V A Reitsema
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - L Schreuder
- Department of Internal Medicine, University Center for Geriatric Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - E Gerrits
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - B J L Eggen
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - M Goris
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - J D Laman
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - S E de Rooij
- Department of Internal Medicine, University Center for Geriatric Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - E M Wesseling
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - H R Bouma
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
- Department of Acute Care, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - R H Henning
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
3
|
Strohm AO, Majewska AK. Physical exercise regulates microglia in health and disease. Front Neurosci 2024; 18:1420322. [PMID: 38911597 PMCID: PMC11192042 DOI: 10.3389/fnins.2024.1420322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 05/20/2024] [Indexed: 06/25/2024] Open
Abstract
There is a well-established link between physical activity and brain health. As such, the effectiveness of physical exercise as a therapeutic strategy has been explored in a variety of neurological contexts. To determine the extent to which physical exercise could be most beneficial under different circumstances, studies are needed to uncover the underlying mechanisms behind the benefits of physical activity. Interest has grown in understanding how physical activity can regulate microglia, the resident immune cells of the central nervous system. Microglia are key mediators of neuroinflammatory processes and play a role in maintaining brain homeostasis in healthy and pathological settings. Here, we explore the evidence suggesting that physical activity has the potential to regulate microglia activity in various animal models. We emphasize key areas where future research could contribute to uncovering the therapeutic benefits of engaging in physical exercise.
Collapse
Affiliation(s)
- Alexandra O. Strohm
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Ania K. Majewska
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, United States
- Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, United States
- Center for Visual Science, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
4
|
Salvi J, Andreoletti P, Audinat E, Balland E, Ben Fradj S, Cherkaoui-Malki M, Heurtaux T, Liénard F, Nédélec E, Rovère C, Savary S, Véjux A, Trompier D, Benani A. Microgliosis: a double-edged sword in the control of food intake. FEBS J 2024; 291:615-631. [PMID: 35880408 DOI: 10.1111/febs.16583] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 06/30/2022] [Accepted: 07/25/2022] [Indexed: 02/16/2024]
Abstract
Maintaining energy balance is essential for survival and health. This physiological function is controlled by the brain, which adapts food intake to energy needs. Indeed, the brain constantly receives a multitude of biological signals that are derived from digested foods or that originate from the gastrointestinal tract, energy stores (liver and adipose tissues) and other metabolically active organs (muscles). These signals, which include circulating nutrients, hormones and neuronal inputs from the periphery, collectively provide information on the overall energy status of the body. In the brain, several neuronal populations can specifically detect these signals. Nutrient-sensing neurons are found in discrete brain areas and are highly enriched in the hypothalamus. In turn, specialized brain circuits coordinate homeostatic responses acting mainly on appetite, peripheral metabolism, activity and arousal. Accumulating evidence shows that hypothalamic microglial cells located at the vicinity of these circuits can influence the brain control of energy balance. However, microglial cells could have opposite effects on energy balance, that is homeostatic or detrimental, and the conditions for this shift are not totally understood yet. One hypothesis relies on the extent of microglial activation, and nutritional lipids can considerably change it.
Collapse
Affiliation(s)
- Juliette Salvi
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Pierre Andreoletti
- Laboratoire Bio-PeroxIL, Université Bourgogne Franche-Comté, Dijon, France
| | - Etienne Audinat
- IGF, Université de Montpellier, CNRS, Inserm, Montpellier, France
| | - Eglantine Balland
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Notting Hill, Australia
| | - Selma Ben Fradj
- IPMC, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Valbonne, France
| | | | - Tony Heurtaux
- Luxembourg Center of Neuropathology (LCNP), Dudelange, Luxembourg
- Department of Life Sciences and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Fabienne Liénard
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Emmanuelle Nédélec
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Carole Rovère
- IPMC, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Valbonne, France
| | - Stéphane Savary
- Laboratoire Bio-PeroxIL, Université Bourgogne Franche-Comté, Dijon, France
| | - Anne Véjux
- Laboratoire Bio-PeroxIL, Université Bourgogne Franche-Comté, Dijon, France
| | - Doriane Trompier
- Laboratoire Bio-PeroxIL, Université Bourgogne Franche-Comté, Dijon, France
| | - Alexandre Benani
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| |
Collapse
|
5
|
Raju RP, Cai L, Tyagi A, Pugazhenthi S. Interactions of Cellular Energetic Gene Clusters in the Alzheimer's Mouse Brain. Mol Neurobiol 2024; 61:476-486. [PMID: 37632678 PMCID: PMC10843700 DOI: 10.1007/s12035-023-03551-0] [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: 04/11/2023] [Accepted: 07/30/2023] [Indexed: 08/28/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in the aging population. The pathological characteristics include extracellular senile plaques and intracellular neurofibrillary tangles. In addition, mitochondrial dysfunction, oxidative stress, and neuroinflammation contribute to AD pathogenesis. In this study, we sought to determine the crosstalk between different pathways in the brain of 5XFAD mice, a mouse model for amyloid pathology, by RNA-seq analysis. We observed significant changes in the expression of genes (1288 genes; adj p value < 0.05; log2-fold > 1 and < 1) related to pathways including oxidation-reduction, oxidative phosphorylation, innate immune response, ribosomal protein synthesis, and ubiquitin proteosome system. The most striking feature was the downregulation of genes related to oxidation-reduction process with changes in the expression of a large number of mitochondrial genes. We also observed an upregulation of several immune response genes. Gene interaction network of oxidation-reduction related genes further confirmed a tight cluster of mitochondrial genes. Furthermore, gene interaction analysis of all the 1288 genes showed at least three distinct interaction clusters, with the predominant one relating to cellular energetics. In summary, we identified 1288 genes distinctly different in the 5XFAD brain compared to the WT brain and found cellular energetics to be the most distinct gene cluster in the AD mouse brain.
Collapse
Affiliation(s)
| | - Lun Cai
- Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Alpna Tyagi
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
- Department of Medicine, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA
| | - Subbiah Pugazhenthi
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA.
- Department of Medicine, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
6
|
Wang L, Wang R, Yu X, Shi Y, Li S, Yuan Y. Effects of Calorie Restriction and Fasting on Macrophage: Potential Impact on Disease Outcomes? Mol Nutr Food Res 2023; 67:e2300380. [PMID: 37771201 DOI: 10.1002/mnfr.202300380] [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: 06/08/2023] [Revised: 08/29/2023] [Indexed: 09/30/2023]
Abstract
Energy restriction, including calorie restriction and fasting, has garnered significant attention for its potential therapeutic effects on a range of chronic diseases (such as diabetes, obesity, and cancer) and aging. Since macrophages are critical players in many diseases, their response to energy restriction may impact disease outcomes. However, the diverse metabolic patterns and functions of macrophages can lead to variability in the effects of energy restriction on macrophages across different tissues and disease states. This review outlines the effects of energy restriction on macrophages in several diseases, offering valuable guidance for future studies and insights into the clinical applications of calorie restriction and fasting.
Collapse
Affiliation(s)
- Lei Wang
- Department of Pharmacy, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 280 Mohe Road, Shanghai, 201999, China
| | - Rong Wang
- Department of Pharmacy, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 280 Mohe Road, Shanghai, 201999, China
| | - Xiaoyan Yu
- Department of Pharmacy, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 280 Mohe Road, Shanghai, 201999, China
| | - Yuhuan Shi
- Department of Pharmacy, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 280 Mohe Road, Shanghai, 201999, China
| | - Shengnan Li
- Department of Pharmacy, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 280 Mohe Road, Shanghai, 201999, China
| | - Yongfang Yuan
- Department of Pharmacy, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 280 Mohe Road, Shanghai, 201999, China
| |
Collapse
|
7
|
Chadarevian JP, Lombroso SI, Peet GC, Hasselmann J, Tu C, Marzan DE, Capocchi J, Purnell FS, Nemec KM, Lahian A, Escobar A, England W, Chaluvadi S, O'Brien CA, Yaqoob F, Aisenberg WH, Porras-Paniagua M, Bennett ML, Davtyan H, Spitale RC, Blurton-Jones M, Bennett FC. Engineering an inhibitor-resistant human CSF1R variant for microglia replacement. J Exp Med 2023; 220:e20220857. [PMID: 36584406 DOI: 10.1084/jem.20220857] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 11/11/2022] [Accepted: 12/13/2022] [Indexed: 12/31/2022] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) can replace endogenous microglia with circulation-derived macrophages but has high mortality. To mitigate the risks of HSCT and expand the potential for microglia replacement, we engineered an inhibitor-resistant CSF1R that enables robust microglia replacement. A glycine to alanine substitution at position 795 of human CSF1R (G795A) confers resistance to multiple CSF1R inhibitors, including PLX3397 and PLX5622. Biochemical and cell-based assays show no discernable gain or loss of function. G795A- but not wildtype-CSF1R expressing macrophages efficiently engraft the brain of PLX3397-treated mice and persist after cessation of inhibitor treatment. To gauge translational potential, we CRISPR engineered human-induced pluripotent stem cell-derived microglia (iMG) to express G795A. Xenotransplantation studies demonstrate that G795A-iMG exhibit nearly identical gene expression to wildtype iMG, respond to inflammatory stimuli, and progressively expand in the presence of PLX3397, replacing endogenous microglia to fully occupy the brain. In sum, we engineered a human CSF1R variant that enables nontoxic, cell type, and tissue-specific replacement of microglia.
Collapse
Affiliation(s)
- Jean Paul Chadarevian
- Department of Neurobiology & Behavior, University of California, Irvine , Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine , Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine , Irvine, CA, USA
| | - Sonia I Lombroso
- Department of Psychiatry, Perelman School of Medicine , University of Pennsylvania, Philadelphia, PA, USA
- Pharmacology Graduate Group, Biomedical Graduate Studies Program, University of Pennsylvania , Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania , Philadelphia, PA, USA
| | - Graham C Peet
- Department of Psychiatry, Perelman School of Medicine , University of Pennsylvania, Philadelphia, PA, USA
- Neuroscience Graduate Program and Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus , Aurora, CO, USA
| | - Jonathan Hasselmann
- Department of Neurobiology & Behavior, University of California, Irvine , Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine , Irvine, CA, USA
| | - Christina Tu
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine , Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine , Irvine, CA, USA
| | - Dave E Marzan
- Department of Biology, The College of New Jersey , Ewing, NJ, USA
| | - Joia Capocchi
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine , Irvine, CA, USA
| | - Freddy S Purnell
- Department of Psychiatry, Perelman School of Medicine , University of Pennsylvania, Philadelphia, PA, USA
| | - Kelsey M Nemec
- Department of Psychiatry, Perelman School of Medicine , University of Pennsylvania, Philadelphia, PA, USA
- Department of Neuroscience, Perelman School of Medicine , Philadelphia, PA, USA
| | - Alina Lahian
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine , Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine , Irvine, CA, USA
| | - Adrian Escobar
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine , Irvine, CA, USA
| | - Whitney England
- Department of Pharmaceutical Sciences, University of California, Irvine , Irvine, CA, USA
| | - Sai Chaluvadi
- Department of Psychiatry, Perelman School of Medicine , University of Pennsylvania, Philadelphia, PA, USA
- Department of Neuroscience, Perelman School of Medicine , Philadelphia, PA, USA
| | - Carleigh A O'Brien
- Department of Psychiatry, Perelman School of Medicine , University of Pennsylvania, Philadelphia, PA, USA
| | - Fazeela Yaqoob
- Department of Psychiatry, Perelman School of Medicine , University of Pennsylvania, Philadelphia, PA, USA
| | - William H Aisenberg
- Department of Psychiatry, Perelman School of Medicine , University of Pennsylvania, Philadelphia, PA, USA
| | | | - Mariko L Bennett
- Department of Neuroscience, Perelman School of Medicine , Philadelphia, PA, USA
- Division of Neurology, Children's Hospital of Philadelphia , Philadelphia, PA, USA
| | - Hayk Davtyan
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine , Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine , Irvine, CA, USA
| | - Robert C Spitale
- Department of Pharmaceutical Sciences, University of California, Irvine , Irvine, CA, USA
| | - Mathew Blurton-Jones
- Department of Neurobiology & Behavior, University of California, Irvine , Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine , Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine , Irvine, CA, USA
| | - F Chris Bennett
- Department of Psychiatry, Perelman School of Medicine , University of Pennsylvania, Philadelphia, PA, USA
- Division of Neurology, Children's Hospital of Philadelphia , Philadelphia, PA, USA
| |
Collapse
|
8
|
Oza MJ, Gaikwad AB, Kulkarni YA. Effect of diet and nutrition on neuroinflammation: An overview. DIET AND NUTRITION IN NEUROLOGICAL DISORDERS 2023:597-611. [DOI: 10.1016/b978-0-323-89834-8.00030-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
9
|
Tyagi A, Musa M, Labeikovsky W, Pugazhenthi S. Sirt3 deficiency induced down regulation of insulin degrading enzyme in comorbid Alzheimer's disease with metabolic syndrome. Sci Rep 2022; 12:19808. [PMID: 36396721 PMCID: PMC9672095 DOI: 10.1038/s41598-022-23652-5] [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: 07/29/2022] [Accepted: 11/03/2022] [Indexed: 11/18/2022] Open
Abstract
SIRT3 deacetylates mitochondrial proteins, thereby enhancing their function. We have previously demonstrated that Sirt3 gene deletion leads to brain mitochondrial dysfunction and neuroinflammation. We also reported that silencing of Sirt3 gene in APP/PS1 mice results in exacerbation of insulin resistance, neuroinflammation and β amyloid plaque deposition. To further understand how metabolic syndrome and amyloid pathology interact, we performed RNA-seq analysis of the brain samples of APP/PS1/Sirt3-/- mice. Gene expression patterns were modulated in metabolic and inflammatory pathways by Sirt3 gene deletion, amyloid pathology, and the combination. Following Sirt3 gene deletion, a key finding was the decreased expression of insulin-degrading enzyme (IDE), an enzyme that regulates the levels of insulin and Aβ peptides. Western diet feeding of Sirt3-/- and APP/PS1 mice resulted in decrease of IDE protein, parallel to Sirt3 downregulation. Conversely, activation of SIRT3 by nicotinamide riboside in vivo and in vitro resulted in IDE upregulation. SIRT3 activation in vivo also increased the levels of neprilysin, another Aβ degrading enzyme and decreased the levels of BACE1 which generates Aβ peptide suggesting SIRT3's role in amyloid plaque reduction. Our findings provide a plausible mechanism linking metabolic syndrome and amyloid pathology. SIRT3 may be a potential therapeutic target to treat AD.
Collapse
Affiliation(s)
- Alpna Tyagi
- grid.422100.50000 0000 9751 469XRocky Mountain Regional VA Medical Center, Aurora, CO USA ,grid.430503.10000 0001 0703 675XDepartment of Medicine, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045 USA
| | - Musa Musa
- grid.422100.50000 0000 9751 469XRocky Mountain Regional VA Medical Center, Aurora, CO USA
| | - Wladimir Labeikovsky
- grid.430503.10000 0001 0703 675XDepartment of Education and Research, Strauss Health Sciences Library, University of Colorado Anschutz Medical Campus, Aurora, CO 80045 USA
| | - Subbiah Pugazhenthi
- grid.422100.50000 0000 9751 469XRocky Mountain Regional VA Medical Center, Aurora, CO USA ,grid.430503.10000 0001 0703 675XDepartment of Medicine, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045 USA
| |
Collapse
|
10
|
Wang Z, Cui Y, Wen L, Yu H, Feng J, Yuan W, He X. Dietary Restriction against Parkinson's Disease: What We Know So Far. Nutrients 2022; 14:nu14194108. [PMID: 36235760 PMCID: PMC9571011 DOI: 10.3390/nu14194108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/24/2022] [Accepted: 09/28/2022] [Indexed: 11/16/2022] Open
Abstract
Dietary restriction (DR) is defined as a moderate reduction in food intake while avoiding malnutrition. The beneficial effects of DR are being increasingly acknowledged in aging and in a series of age-related neurodegenerative disorders, for example, Parkinson's disease (PD). To date, the pathogenesis of PD remains elusive and there is no cure for it in spite of intensive research over decades. In this review, we summarize the current knowledge on the efficacy of DR on PD, focusing on the underlying mechanisms involving general metabolism, neuroendocrinolgy, neuroinflammation, gut microbiome, and so on. We anticipate that this review will provide future perspectives for PD prevention and treatment.
Collapse
Affiliation(s)
- Zhonglei Wang
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yueran Cui
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Lulu Wen
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Haiyang Yu
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Juan Feng
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Wei Yuan
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang 110001, China
- Correspondence: (W.Y.); (X.H.); Tel.: +86-024-8328-3360 (W.Y.); +86-024-96615-28111 (X.H.)
| | - Xin He
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang 110004, China
- Correspondence: (W.Y.); (X.H.); Tel.: +86-024-8328-3360 (W.Y.); +86-024-96615-28111 (X.H.)
| |
Collapse
|
11
|
Robbins JP, Solito E. Does Neuroinflammation Underlie the Cognitive Changes Observed With Dietary Interventions? Front Neurosci 2022; 16:854050. [PMID: 35620671 PMCID: PMC9127342 DOI: 10.3389/fnins.2022.854050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/24/2022] [Indexed: 11/13/2022] Open
Abstract
Dietary interventions, such as calorie restriction and ketogenic diet, have been extensively studied in ageing research, including in cognitive decline. Epidemiological studies indicate beneficial effects of certain dietary regimes on mental health, including mood disorders and dementia. However, randomised-controlled trials (the gold-standard of evidence-based medicine) on calorie restriction diets and the ketogenic diet have yet to show clinically convincing effects in neuropsychiatric disorders. This review will examine the quality of studies and evidence base for the ketogenic and calorie restriction diets in common neuropsychiatric conditions, collating findings from preclinical experiments, case reports or small clinical studies, and randomised controlled clinical trials. The major cellular mechanisms that mediate the effects of these dietary interventions on brain health include neuroinflammation, neuroprotection, and neuromodulation. We will discuss the studies that have investigated the roles of these pathways and their interactions. Popularity of the ketogenic and calorie restriction diets has grown both in the public domain and in psychiatry research, allowing for informed review of the efficacy, the limitations, and the side effects of these diets in specific patient populations. In this review we will summarise the clinical evidence for these diets in neuropsychiatry and make suggestions to improve clinical translation of future research studies.
Collapse
Affiliation(s)
- Jacqueline P. Robbins
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Egle Solito
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
12
|
Redefining microglia states: Lessons and limits of human and mouse models to study microglia states in neurodegenerative diseases. Semin Immunol 2022; 60:101651. [PMID: 36155944 DOI: 10.1016/j.smim.2022.101651] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 09/03/2022] [Indexed: 01/15/2023]
Abstract
Microglia are resident macrophages of the brain parenchyma and play an essential role in various aspects of brain development, plasticity, and homeostasis. With recent advances in single-cell RNA-sequencing, heterogeneous microglia transcriptional states have been identified in both animal models of neurodegenerative disorders and patients. However, the functional roles of these microglia states remain unclear; specifically, the question of whether individual states or combinations of states are protective or detrimental (or both) in the context of disease progression. To attempt to answer this, the field has largely relied on studies employing mouse models, human in vitro and chimeric models, and human post-mortem tissue, all of which have their caveats, but used in combination can enable new biological insight and validation of candidate disease pathways and mechanisms. In this review, we summarize our current understanding of disease-associated microglia states and phenotypes in neurodegenerative disorders, discuss important considerations when comparing mouse and human microglia states and functions, and identify areas of microglia biology where species differences might limit our understanding of microglia state.
Collapse
|
13
|
Long-term intake of total energy and fat in relation to subjective cognitive decline. Eur J Epidemiol 2022; 37:133-146. [PMID: 34748116 PMCID: PMC8960333 DOI: 10.1007/s10654-021-00814-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 10/11/2021] [Indexed: 02/03/2023]
Abstract
Diet is one of the modifiable risk factors for cognitive decline. However, human studies on total energy intake and cognitive function have remained limited and studies on fat intake and cognitive decline have been inconclusive. We aimed to examine prospectively the associations between long-term intakes of total energy and fat with subsequent subjective cognitive decline (SCD). A total of 49,493 women from the Nurses' Health Study and 27,842 men from the Health Professionals Follow-up Study were followed for over 20 years. Average dietary intake was calculated based on repeated food frequency questionnaires (SFFQs), and Poisson regression was used to evaluate associations. Higher total energy intake was significantly associated with greater odds of SCD in both cohorts. Comparing the highest with lowest quintiles of total energy intake, the pooled multivariable-adjusted ORs (95% CIs) for a 3-unit increment in SCD, corresponding to poor versus normal SCD, was 2.77 (2.53, 2.94). Each 500 kcal/day greater intake of total energy was associated with 48% higher odds of SCD. Intakes of both total fat and total carbohydrate appeared to contribute to the positive association between total energy intake and SCD although for the same percent of energy, the association was stronger for total fat. In conclusion, higher intakes of total energy, total fat, and total carbohydrate were adversely associated with SCD. Whether these associations are causal is unclear and deserves further investigation.
Collapse
|
14
|
Mayorga-Weber G, Rivera FJ, Castro MA. Neuron-glia (mis)interactions in brain energy metabolism during aging. J Neurosci Res 2022; 100:835-854. [PMID: 35085408 DOI: 10.1002/jnr.25015] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/08/2021] [Accepted: 12/06/2021] [Indexed: 02/06/2023]
Abstract
Life expectancy in humans is increasing, resulting in a growing aging population, that is accompanied by an increased disposition to develop cognitive deterioration. Hypometabolism is one of the multiple factors related to inefficient brain function during aging. This review emphasizes the metabolic interactions between glial cells (astrocytes, oligodendrocytes, and microglia) and neurons, particularly, during aging. Glial cells provide support and protection to neurons allowing adequate synaptic activity. We address metabolic coupling from the expression of transporters, availability of substrates, metabolic pathways, and mitochondrial activity. In aging, the main metabolic exchange machinery is altered with inefficient levels of nutrients and detrimental mitochondrial activity that results in high reactive oxygen species levels and reduced ATP production, generating a highly inflammatory environment that favors deregulated cell death. Here, we provide an overview of the glial-to-neuron mechanisms, from the molecular components to the cell types, emphasizing aging as the crucial risk factor for developing neurodegenerative/neuroinflammatory diseases.
Collapse
Affiliation(s)
- Gonzalo Mayorga-Weber
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Francisco J Rivera
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile.,Laboratory of Stem Cells and Neuroregeneration, Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile.,Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Maite A Castro
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile.,Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile.,Janelia Research Campus, HHMI, Ashburn, VA, USA
| |
Collapse
|
15
|
de Fraga LS, Tassinari ID, Jantsch J, Guedes RP, Bambini-Junior V. 'A picture is worth a thousand words': The use of microscopy for imaging neuroinflammation. Clin Exp Immunol 2021; 206:325-345. [PMID: 34596237 DOI: 10.1111/cei.13669] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 01/08/2023] Open
Abstract
Since the first studies of the nervous system by the Nobel laureates Camillo Golgi and Santiago Ramon y Cajal using simple dyes and conventional light microscopes, microscopy has come a long way to the most recent techniques that make it possible to perform images in live cells and animals in health and disease. Many pathological conditions of the central nervous system have already been linked to inflammatory responses. In this scenario, several available markers and techniques can help imaging and unveil the neuroinflammatory process. Moreover, microscopy imaging techniques have become even more necessary to validate the large quantity of data generated in the era of 'omics'. This review aims to highlight how to assess neuroinflammation by using microscopy as a tool to provide specific details about the cell's architecture during neuroinflammatory conditions. First, we describe specific markers that have been used in light microscopy studies and that are widely applied to unravel and describe neuroinflammatory mechanisms in distinct conditions. Then, we discuss some important methodologies that facilitate the imaging of these markers, such as immunohistochemistry and immunofluorescence techniques. Emphasis will be given to studies using two-photon microscopy, an approach that revolutionized the real-time assessment of neuroinflammatory processes. Finally, some studies integrating omics with microscopy will be presented. The fusion of these techniques is developing, but the high amount of data generated from these applications will certainly improve comprehension of the molecular mechanisms involved in neuroinflammation.
Collapse
Affiliation(s)
- Luciano Stürmer de Fraga
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Isadora D'Ávila Tassinari
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Jeferson Jantsch
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Renata Padilha Guedes
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Victorio Bambini-Junior
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire (UCLan), Preston, UK
| |
Collapse
|
16
|
Fontana L, Ghezzi L, Cross AH, Piccio L. Effects of dietary restriction on neuroinflammation in neurodegenerative diseases. J Exp Med 2021; 218:211666. [PMID: 33416892 PMCID: PMC7802371 DOI: 10.1084/jem.20190086] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/29/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022] Open
Abstract
Recent and accumulating work in experimental animal models and humans shows that diet has a much more pervasive and prominent role than previously thought in modulating neuroinflammatory and neurodegenerative mechanisms leading to some of the most common chronic central nervous system (CNS) diseases. Chronic or intermittent food restriction has profound effects in shaping brain and peripheral metabolism, immunity, and gut microbiome biology. Interactions among calorie intake, meal frequency, diet quality, and the gut microbiome modulate specific metabolic and molecular pathways that regulate cellular, tissue, and organ homeostasis as well as inflammation during normal brain aging and CNS neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and multiple sclerosis, among others. This review discusses these findings and their potential application to the prevention and treatment of CNS neuroinflammatory diseases and the promotion of healthy brain aging.
Collapse
Affiliation(s)
- Luigi Fontana
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, Australia.,Department of Clinical and Experimental Sciences, Brescia University, Brescia, Italy
| | - Laura Ghezzi
- Department of Neurology, Washington University in St. Louis, St. Louis, MO.,University of Milan, Milan, Italy
| | - Anne H Cross
- Department of Neurology, Washington University in St. Louis, St. Louis, MO
| | - Laura Piccio
- Department of Neurology, Washington University in St. Louis, St. Louis, MO.,Brain and Mind Centre, School of Medical Sciences, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
17
|
Augusto-Oliveira M, Verkhratsky A. Lifestyle-dependent microglial plasticity: training the brain guardians. Biol Direct 2021; 16:12. [PMID: 34353376 PMCID: PMC8340437 DOI: 10.1186/s13062-021-00297-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 07/29/2021] [Indexed: 02/07/2023] Open
Abstract
Lifestyle is one of the most powerful instruments shaping mankind; the lifestyle includes many aspects of interactions with the environment, from nourishment and education to physical activity and quality of sleep. All these factors taken in complex affect neuroplasticity and define brain performance and cognitive longevity. In particular, physical exercise, exposure to enriched environment and dieting act through complex modifications of microglial cells, which change their phenotype and modulate their functional activity thus translating lifestyle events into remodelling of brain homoeostasis and reshaping neural networks ultimately enhancing neuroprotection and cognitive longevity.
Collapse
Affiliation(s)
- Marcus Augusto-Oliveira
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belém, 66075-110, Brazil.
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK. .,Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, 01102, Vilnius, Lithuania. .,Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011, Bilbao, Spain. .,Department of Neurosciences, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain.
| |
Collapse
|
18
|
Snijders GJLJ, Sneeboer MAM, Fernández-Andreu A, Udine E, Boks MP, Ormel PR, van Berlekom AB, van Mierlo HC, Bӧttcher C, Priller J, Raj T, Hol EM, Kahn RS, de Witte LD. Distinct non-inflammatory signature of microglia in post-mortem brain tissue of patients with major depressive disorder. Mol Psychiatry 2021; 26:3336-3349. [PMID: 33028963 DOI: 10.1038/s41380-020-00896-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 08/22/2020] [Accepted: 09/17/2020] [Indexed: 02/06/2023]
Abstract
Findings from epidemiological studies, biomarker measurements and animal experiments suggest a role for aberrant immune processes in the pathogenesis of major depressive disorder (MDD). Microglia, the resident immune cells of the brain, are likely to play a key role in these processes. Previous post-mortem studies reported conflicting findings regarding microglial activation and an in-depth profiling of those cells in MDD is lacking. The aim of this study was therefore to characterize the phenotype and function of microglia in MDD. We isolated microglia from post-mortem brain tissue of patients with MDD (n = 13-19) and control donors (n = 12-25). Using flow cytometry and quantitative Polymerase Chain Reaction (qPCR), we measured protein and mRNA levels of a panel of microglial markers across four different brain regions (medial frontal gyrus, superior temporal gyrus, thalamus, and subventricular zone). In MDD cases, we found a significant upregulation of CX3CR1 and TMEM119 mRNA expression and a downregulation of CD163 mRNA expression and CD14 protein expression across the four brain regions. Expression levels of microglial activation markers, such as HLA-DRA, IL6, and IL1β, as well as the inflammatory responses to lipopolysaccharide and dexamethasone were unchanged. Our findings suggest that microglia enhance homeostatic functions in MDD but are not immune activated.
Collapse
Affiliation(s)
- Gijsje J L J Snijders
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands.
| | - Marjolein A M Sneeboer
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands.,Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands
| | - Alba Fernández-Andreu
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands
| | - Evan Udine
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Marco P Boks
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands
| | - Paul R Ormel
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands.,Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands
| | - Amber Berdenis van Berlekom
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands.,Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands
| | - Hans C van Mierlo
- Department of Psychiatry, St. Antonius Hospital, Nieuwegein, Koekoekslaan 1, 3430, EM, Nieuwegein, The Netherlands
| | - Chotima Bӧttcher
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Josef Priller
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany.,DZNE and BIH, 10117, Berlin, Germany.,University of Edinburgh and UK DRI, Edinburgh, EH16 4SB, UK
| | - Towfique Raj
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Elly M Hol
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands.,Neuroimmunology, Netherlands Institute for Neuroscience, an institute of the royal academy of arts and sciences, 1105, BA, Amsterdam, The Netherlands
| | - René S Kahn
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands.,Mental Illness Research Education Clinical, Centers of Excellence, VA, Mental Health, Veterans, Bronx, NY, USA
| | - Lot D de Witte
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands.,Mental Illness Research Education Clinical, Centers of Excellence, VA, Mental Health, Veterans, Bronx, NY, USA
| |
Collapse
|
19
|
Alexaki VI. The Impact of Obesity on Microglial Function: Immune, Metabolic and Endocrine Perspectives. Cells 2021; 10:cells10071584. [PMID: 34201844 PMCID: PMC8307603 DOI: 10.3390/cells10071584] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/18/2021] [Accepted: 06/22/2021] [Indexed: 02/06/2023] Open
Abstract
Increased life expectancy in combination with modern life style and high prevalence of obesity are important risk factors for development of neurodegenerative diseases. Neuroinflammation is a feature of neurodegenerative diseases, and microglia, the innate immune cells of the brain, are central players in it. The present review discusses the effects of obesity, chronic peripheral inflammation and obesity-associated metabolic and endocrine perturbations, including insulin resistance, dyslipidemia and increased glucocorticoid levels, on microglial function.
Collapse
Affiliation(s)
- Vasileia Ismini Alexaki
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| |
Collapse
|
20
|
No-added-oil mediterranean diet: A novel aging deceleration diet? AGING AND HEALTH RESEARCH 2021. [DOI: 10.1016/j.ahr.2021.100017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
21
|
Duggan MR, Parikh V. Microglia and modifiable life factors: Potential contributions to cognitive resilience in aging. Behav Brain Res 2021; 405:113207. [PMID: 33640394 PMCID: PMC8005490 DOI: 10.1016/j.bbr.2021.113207] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/27/2021] [Accepted: 02/20/2021] [Indexed: 02/08/2023]
Abstract
Given the increasing prevalence of age-related cognitive decline, it is relevant to consider the factors and mechanisms that might facilitate an individual's resiliency to such deficits. Growing evidence suggests a preeminent role of microglia, the prime mediator of innate immunity within the central nervous system. Human and animal investigations suggest aberrant microglial functioning and neuroinflammation are not only characteristic of the aged brain, but also might contribute to age-related dementia and Alzheimer's Disease. Conversely, accumulating data suggest that modifiable lifestyle factors (MLFs), such as healthy diet, exercise and cognitive engagement, can reliably afford cognitive benefits by potentially suppressing inflammation in the aging brain. The present review highlights recent advances in our understanding of the role for microglia in maintaining brain homeostasis and cognitive functioning in aging. Moreover, we propose an integrated, mechanistic model that postulates an individual's resiliency to cognitive decline afforded by MLFs might be mediated by the mitigation of aberrant microglia activation in aging, and subsequent suppression of neuroinflammation.
Collapse
Affiliation(s)
- Michael R Duggan
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, 19122, United States
| | - Vinay Parikh
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, 19122, United States.
| |
Collapse
|
22
|
Vitamin D Deficiency Induces Chronic Pain and Microglial Phenotypic Changes in Mice. Int J Mol Sci 2021; 22:ijms22073604. [PMID: 33808491 PMCID: PMC8036382 DOI: 10.3390/ijms22073604] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/22/2021] [Accepted: 03/26/2021] [Indexed: 02/07/2023] Open
Abstract
The bioactive form of vitamin D, 1,25-dihydroxyvitamin D (1,25D3), exerts immunomodulatory actions resulting in neuroprotective effects potentially useful against neurodegenerative and autoimmune diseases. In fact, vitamin D deficiency status has been correlated with painful manifestations associated with different pathological conditions. In this study, we have investigated the effects of vitamin D deficiency on microglia cells, as they represent the main immune cells responsible for early defense at central nervous system (CNS), including chronic pain states. For this purpose, we have employed a model of low vitamin D intake during gestation to evaluate possible changes in primary microglia cells obtained from postnatal day(P)2-3 pups. Afterwards, pain measurement and microglia morphological analysis in the spinal cord level and in brain regions involved in the integration of pain perception were performed in the parents subjected to vitamin D restriction. In cultured microglia, we detected a reactive-activated and proliferative-phenotype associated with intracellular reactive oxygen species (ROS) generation. Oxidative stress was closely correlated with the extent of DNA damage and increased β-galactosidase (B-gal) activity. Interestingly, the incubation with 25D3 or 1,25D3 or palmitoylethanolamide, an endogenous ligand of peroxisome proliferator-activated-receptor-alpha (PPAR-α), reduced most of these effects. Morphological analysis of ex-vivo microglia obtained from vitamin-D-deficient adult mice revealed an increased number of activated microglia in the spinal cord, while in the brain microglia appeared in a dystrophic phenotype. Remarkably, activated (spinal) or dystrophic (brain) microglia were detected in a prominent manner in females. Our data indicate that vitamin D deficiency produces profound modifications in microglia, suggesting a possible role of these cells in the sensorial dysfunctions associated with hypovitaminosis D.
Collapse
|
23
|
Mapping of Microglial Brain Region, Sex and Age Heterogeneity in Obesity. Int J Mol Sci 2021; 22:ijms22063141. [PMID: 33808700 PMCID: PMC8003547 DOI: 10.3390/ijms22063141] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 12/27/2022] Open
Abstract
The prevalence of obesity has increased rapidly in recent years and has put a huge burden on healthcare worldwide. Obesity is associated with an increased risk for many comorbidities, such as cardiovascular diseases, type 2 diabetes and hypertension. The hypothalamus is a key brain region involved in the regulation of food intake and energy expenditure. Research on experimental animals has shown neuronal loss, as well as microglial activation in the hypothalamus, due to dietary-induced obesity. Microglia, the resident immune cells in the brain, are responsible for maintaining the brain homeostasis and, thus, providing an optimal environment for neuronal function. Interestingly, in obesity, microglial cells not only get activated in the hypothalamus but in other brain regions as well. Obesity is also highly associated with changes in hippocampal function, which could ultimately result in cognitive decline and dementia. Moreover, changes have also been reported in the striatum and cortex. Microglial heterogeneity is still poorly understood, not only in the context of brain region but, also, age and sex. This review will provide an overview of the currently available data on the phenotypic differences of microglial innate immunity in obesity, dependent on brain region, sex and age.
Collapse
|
24
|
Torres W, Nava M, Galbán N, Gómez Y, Morillo V, Rojas M, Cano C, Chacín M, D Marco L, Herazo Y, Velasco M, Bermúdez V, Rojas-Quintero J. Anti-Aging Effect of Metformin: A Molecular and Therapeutical Perspective. Curr Pharm Des 2021; 26:4496-4508. [PMID: 32674728 DOI: 10.2174/1381612826666200716161610] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 06/09/2020] [Indexed: 12/25/2022]
Abstract
Aging is a time-dependent inevitable process, in which cellular homeostasis is affected, which has an impact on tissue function. This represents a risk factor for the development of numerous non-transmissible diseases. In consequence, the scientific community continues to search for therapeutic measures capable of improving quality of life and delaying cellular aging. At the center of this research is metformin, a widely used drug in Type 2 Diabetes Mellitus treatment that has a reduced adverse effects profile. Furthermore, there is evidence that this drug has beneficial health effects that go beyond its anti-hyperglycemic properties. Among these effects, its geronto-protection capability stands out. There is growing evidence that points out to an increased life expectancy as well as the quality of life in model organisms treated with metformin. Therefore, there is an abundance of research centered on elucidating the mechanism through which metformin has its anti-aging effects. Among these, the AMPK, mTORC1, SIRT1, FOXO, NF.kB, and DICER1 pathways can be mentioned. Furthermore, studies have highlighted the possibility of a role for the gut microbiome in these processes. The next step is the design of clinical essays that have as a goal evaluating the efficacy and safety of metformin as an anti-aging drug in humans to create a paradigm in the medical horizon. The question being if metformin is, in fact, the new antiaging therapy in humans?
Collapse
Affiliation(s)
- Wheeler Torres
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Manuel Nava
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Nestor Galbán
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Yosselin Gómez
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Valery Morillo
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Milagros Rojas
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Clímaco Cano
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Maricarmen Chacín
- Universidad Simón Bolívar, Facultad de Ciencias de la Salud, Barranquilla, Colombia
| | - Luis D Marco
- Hospital Clínico Universitario, INCLIVA, Nephrology Department, Valencia, España
| | - Yaneth Herazo
- Universidad Simón Bolívar, Facultad de Ciencias de la Salud, Barranquilla, Colombia
| | - Manuel Velasco
- Clinical Pharmacologic Unit, Vargas School of Medicine, Universidad Central de Venezuela, Caracas,
Venezuela
| | - Valmore Bermúdez
- Universidad Simón Bolívar, Facultad de Ciencias de la Salud, Barranquilla, Colombia
| | - Joselyn Rojas-Quintero
- Pulmonary and Critical Care Medicine Department, Brigham and Women’s Hospital, Harvard Medical School,
Boston, MA 02115, USA
| |
Collapse
|
25
|
Fu X, Zeng H, Zhao J, Zhou G, Zhou H, Zhuang J, Xu C, Li J, Peng Y, Cao Y, Li Y, Chen H, Wang L, Yan F, Chen G. Inhibition of Dectin-1 Ameliorates Neuroinflammation by Regulating Microglia/Macrophage Phenotype After Intracerebral Hemorrhage in Mice. Transl Stroke Res 2021; 12:1018-1034. [PMID: 33539006 DOI: 10.1007/s12975-021-00889-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/03/2020] [Accepted: 01/10/2021] [Indexed: 01/28/2023]
Abstract
Polarization of microglia/macrophages toward the pro-inflammatory phenotype is an important contributor to neuroinflammation after intracerebral hemorrhage (ICH). Dectin-1 is a pattern recognition receptor that has been reported to play a key role in regulating neuroinflammation in ischemic stroke and spinal cord injury. However, the role and mechanism of action of Dectin-1 after ICH remains unclear. In this study, we investigated the effect of Dectin-1 on modulating the microglia/macrophage phenotype and neuroinflammation and the possible underlying mechanism after ICH. We found that Dectin-1 expression increased after ICH, and was mainly localized in microglia/macrophages. Neutrophil infiltration and microglia/macrophage polarization toward the pro-inflammatory phenotype increased after ICH. However, treatment with a Dectin-1 inhibitor reversed these phenomena and induced a shift the anti-inflammatory phenotype in microglia/macrophages; this resulted in alleviation of neurological dysfunction and facilitated hematoma clearance after ICH. We also found that Dectin-1 crosstalks with the downstream pro-inflammatory pathway, Card9/NF-κB, by activating spleen tyrosine kinase (Syk) both in vivo and in vitro. In conclusion, our data suggest that Dectin-1 is involved in the microglia/macrophage polarization and functional recovery after ICH, and that this mechanism, at least in part, may contribute to the involvement of the Syk/Card9/NF-kB pathway.
Collapse
Affiliation(s)
- Xiongjie Fu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Hanhai Zeng
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Jikuang Zhao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China.,Department of Neurosurgery, Ningbo First Hospital, Ningbo Hospital, Zhejiang University School of Medicine, Ningbo, China
| | - Guoyang Zhou
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Hang Zhou
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Jianfeng Zhuang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Chaoran Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Jianru Li
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Yucong Peng
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Yang Cao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Yin Li
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Huaijun Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Lin Wang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Feng Yan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China.
| | - Gao Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China.
| |
Collapse
|
26
|
Changing Functional Signatures of Microglia along the Axis of Brain Aging. Int J Mol Sci 2021; 22:ijms22031091. [PMID: 33499206 PMCID: PMC7865559 DOI: 10.3390/ijms22031091] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 12/19/2022] Open
Abstract
Microglia, the innate immune cells of the brain, are commonly perceived as resident macrophages of the central nervous system (CNS). This definition, however, requires further specification, as under healthy homeostatic conditions, neither morphological nor functional properties of microglia mirror those of classical macrophages. Indeed, microglia adapt exceptionally well to their microenvironment, becoming a legitimate member of the cellular brain architecture. The ramified or surveillant microglia in the young adult brain are characterized by specific morphology (small cell body and long, thin motile processes) and physiology (a unique pattern of Ca2+ signaling, responsiveness to various neurotransmitters and hormones, in addition to classic “immune” stimuli). Their numerous physiological functions far exceed and complement their immune capabilities. As the brain ages, the respective changes in the microglial microenvironment impact the functional properties of microglia, triggering further rounds of adaptation. In this review, we discuss the recent data showing how functional properties of microglia adapt to age-related changes in brain parenchyma in a sex-specific manner, with a specific focus on early changes occurring at middle age as well as some strategies counteracting the aging of microglia.
Collapse
|
27
|
Yang Y, Zhang L. The effects of caloric restriction and its mimetics in Alzheimer's disease through autophagy pathways. Food Funct 2020; 11:1211-1224. [PMID: 32068753 DOI: 10.1039/c9fo02611h] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that commonly occurs among older individuals. Increasing evidence suggests that a low-caloric diet might be a promising adjuvant therapeutic strategy for slowing or preventing the pathogenesis and progression of AD through the induction of autophagy. Several intracellular pathways have been implicated in caloric restriction (CR)-induced autophagy. In this review, we summarized the efficacy of CR as well as its mimetics (resveratrol, spermidine, aspirin, rapamycin, metformin, and curcumin) in improving cognitive function of rodent models of AD. On the basis of recent in vitro and animal studies, the beneficial effects of CR- or caloric restriction mimetics-induced autophagy in alleviating amyloid burden and tau pathology of AD were also discussed.
Collapse
Affiliation(s)
- Yi Yang
- Department of Pharmacology, Hangzhou Key Laboratory of Medical Neurobiology, School of Medicine, Hangzhou Normal University, Hangzhou 310036, China.
| | - Lihui Zhang
- Department of Pharmacology, Hangzhou Key Laboratory of Medical Neurobiology, School of Medicine, Hangzhou Normal University, Hangzhou 310036, China.
| |
Collapse
|
28
|
Dridi H, Liu X, Yuan Q, Reiken S, Yehia M, Sittenfeld L, Apostolou P, Buron J, Sicard P, Matecki S, Thireau J, Menuet C, Lacampagne A, Marks AR. Role of defective calcium regulation in cardiorespiratory dysfunction in Huntington's disease. JCI Insight 2020; 5:140614. [PMID: 32897880 PMCID: PMC7566717 DOI: 10.1172/jci.insight.140614] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 09/02/2020] [Indexed: 12/19/2022] Open
Abstract
Huntington’s disease (HD) is a progressive, autosomal dominant neurodegenerative disorder affecting striatal neurons beginning in young adults with loss of muscle coordination and cognitive decline. Less appreciated is the fact that patients with HD also exhibit cardiac and respiratory dysfunction, including pulmonary insufficiency and cardiac arrhythmias. The underlying mechanism for these symptoms is poorly understood. In the present study we provide insight into the cause of cardiorespiratory dysfunction in HD and identify a potentially novel therapeutic target. We now show that intracellular calcium (Ca2+) leak via posttranslationally modified ryanodine receptor/intracellular calcium release (RyR) channels plays an important role in HD pathology. RyR channels were oxidized, PKA phosphorylated, and leaky in brain, heart, and diaphragm both in patients with HD and in a murine model of HD (Q175). HD mice (Q175) with endoplasmic reticulum Ca2+ leak exhibited cognitive dysfunction, decreased parasympathetic tone associated with cardiac arrhythmias, and reduced diaphragmatic contractile function resulting in impaired respiratory function. Defects in cognitive, motor, and respiratory functions were ameliorated by treatment with a novel Rycal small-molecule drug (S107) that fixes leaky RyR. Thus, leaky RyRs likely play a role in neuronal, cardiac, and diaphragmatic pathophysiology in HD, and RyRs are a potential novel therapeutic target. This study explores the role of ryanodine receptor calcium channels in the brain, the heart, and the diaphragm and central versus peripheral pathophysiological mechanisms in Huntington’s disease.
Collapse
Affiliation(s)
- Haikel Dridi
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Xiaoping Liu
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Qi Yuan
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Steve Reiken
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Mohamad Yehia
- PHYMEDEXP, University of Montpellier, CNRS, INSERM, CHRU Montpellier, Montpellier, France
| | - Leah Sittenfeld
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Panagiota Apostolou
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Julie Buron
- Institut de Neurobiologie de la Méditerranée, INMED UMR1249, INSERM, Aix-Marseille Université, Marseille, France
| | - Pierre Sicard
- PHYMEDEXP, University of Montpellier, CNRS, INSERM, CHRU Montpellier, Montpellier, France
| | - Stefan Matecki
- PHYMEDEXP, University of Montpellier, CNRS, INSERM, CHRU Montpellier, Montpellier, France
| | - Jérome Thireau
- PHYMEDEXP, University of Montpellier, CNRS, INSERM, CHRU Montpellier, Montpellier, France.,LIA MusCaRyR, CNRS, Montpellier, France
| | - Clement Menuet
- Institut de Neurobiologie de la Méditerranée, INMED UMR1249, INSERM, Aix-Marseille Université, Marseille, France
| | - Alain Lacampagne
- PHYMEDEXP, University of Montpellier, CNRS, INSERM, CHRU Montpellier, Montpellier, France.,LIA MusCaRyR, CNRS, Montpellier, France
| | - Andrew R Marks
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| |
Collapse
|
29
|
Diaz A, Muñoz‐Arenas G, Caporal‐Hernandez K, Vázquez‐Roque R, Lopez‐Lopez G, Kozina A, Espinosa B, Flores G, Treviño S, Guevara J. Gallic acid improves recognition memory and decreases oxidative‐inflammatory damage in the rat hippocampus with metabolic syndrome. Synapse 2020; 75:e22186. [PMID: 32780904 DOI: 10.1002/syn.22186] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/28/2020] [Accepted: 08/07/2020] [Indexed: 12/20/2022]
Affiliation(s)
- Alfonso Diaz
- Facultad de Ciencias Quimicas Benemerita Universidad Autonoma de Puebla Puebla Mexico
| | | | | | - Rubén Vázquez‐Roque
- Laboratorio de Neuropsiquiatria Instituto de Fisiologia Benemerita Universidad Autonoma de Puebla Puebla Mexico
| | - Gustavo Lopez‐Lopez
- Facultad de Ciencias Quimicas Benemerita Universidad Autonoma de Puebla Puebla Mexico
| | - Anna Kozina
- Instituto de Química Universidad Nacional Autonoma de México Ciudad de Mexico Mexico
| | - Blanca Espinosa
- Departamento de Bioquimica Instituto Nacional de Enfermedades Respiratorias, ICV Ciudad de Mexico Mexico
| | - Gonzalo Flores
- Laboratorio de Neuropsiquiatria Instituto de Fisiologia Benemerita Universidad Autonoma de Puebla Puebla Mexico
| | - Samuel Treviño
- Facultad de Ciencias Quimicas Benemerita Universidad Autonoma de Puebla Puebla Mexico
| | - Jorge Guevara
- Departamento de Bioquimica Facultad de Medicina Universidad Nacional Autonoma de México Ciudad de Mexico Mexico
| |
Collapse
|
30
|
Rudzki L, Maes M. The Microbiota-Gut-Immune-Glia (MGIG) Axis in Major Depression. Mol Neurobiol 2020; 57:4269-4295. [DOI: 10.1007/s12035-020-01961-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/28/2020] [Indexed: 02/08/2023]
|
31
|
Wang X, Yang J, Lu T, Zhan Z, Wei W, Lyu X, Jiang Y, Xue X. The effect of swimming exercise and diet on the hypothalamic inflammation of ApoE-/- mice based on SIRT1-NF-κB-GnRH expression. Aging (Albany NY) 2020; 12:11085-11099. [PMID: 32518216 PMCID: PMC7346084 DOI: 10.18632/aging.103323] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 04/28/2020] [Indexed: 04/13/2023]
Abstract
A high-fat diet and sedentary lifestyle could accelerate aging and hypothalamic inflammation. In order to explore the regulatory mechanisms of lifestyle in the hypothalamus, swimming exercise and diet control were applied in the high-fat diet ApoE-/- mice in our study. 20-week-old ApoE-/- mice fed with 12-week high-fat diet were treated by high-fat diet, diet control and swimming exercise. The results showed that hypothalamic inflammation, glial cells activation and cognition decline were induced by high-fat diet. Compared with the diet control, hypothalamic inflammation, glial cells activation and learning and memory impairment were effectively alleviated by swimming exercise plus diet control, which was related to the increasing expression of SIRT1, inhibiting the expression of NF-κB and raising secretion of GnRH in the hypothalamus. These findings supported the hypothesis that hypothalamic inflammation was susceptible to exercise and diet, which was strongly associated with SIRT1-NF-κB-GnRH expression in the hypothalamus.
Collapse
Affiliation(s)
- Xialei Wang
- The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou 350003, China
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350112, China
| | - Jingda Yang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350112, China
| | - Taotao Lu
- The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou 350003, China
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350112, China
| | - Zengtu Zhan
- The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou 350003, China
| | - Wei Wei
- The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou 350003, China
| | - Xinru Lyu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350112, China
| | - Yijing Jiang
- The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou 350003, China
| | - Xiehua Xue
- The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou 350003, China
| |
Collapse
|
32
|
Ye XC, Hao Q, Ma WJ, Zhao QC, Wang WW, Yin HH, Zhang T, Wang M, Zan K, Yang XX, Zhang ZH, Shi HJ, Zu J, Raza HK, Zhang XL, Geng DQ, Hu JX, Cui GY. Dectin-1/Syk signaling triggers neuroinflammation after ischemic stroke in mice. J Neuroinflammation 2020; 17:17. [PMID: 31926564 PMCID: PMC6954534 DOI: 10.1186/s12974-019-1693-z] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 12/26/2019] [Indexed: 12/13/2022] Open
Abstract
Background Dendritic cell-associated C-type lectin-1 (Dectin-1) receptor has been reported to be involved in neuroinflammation in Alzheimer’s disease and traumatic brain injury. The present study was designed to investigate the role of Dectin-1 and its downstream target spleen tyrosine kinase (Syk) in early brain injury after ischemic stroke using a focal cortex ischemic stroke model. Methods Adult male C57BL/6 J mice were subjected to a cerebral focal ischemia model of ischemic stroke. The neurological score, adhesive removal test, and foot-fault test were evaluated on days 1, 3, 5, and 7 after ischemic stroke. Dectin-1, Syk, phosphorylated (p)-Syk, tumor necrosis factor-α (TNF-α), and inducible nitric oxide synthase (iNOS) expression was analyzed via western blotting in ischemic brain tissue after ischemic stroke and in BV2 microglial cells subjected to oxygen-glucose deprivation/reoxygenation (OGD/R) injury in vitro. The brain infarct volume and Iba1-positive cells were evaluated using Nissl’s and immunofluorescence staining, respectively. The Dectin-1 antagonist laminarin (LAM) and a selective inhibitor of Syk phosphorylation (piceatannol; PIC) were used for the intervention. Results Dectin-1, Syk, and p-Syk expression was significantly enhanced on days 3, 5, and 7 and peaked on day 3 after ischemic stroke. The Dectin-1 antagonist LAM or Syk inhibitor PIC decreased the number of Iba1-positive cells and TNF-α and iNOS expression, decreased the brain infarct volume, and improved neurological functions on day 3 after ischemic stroke. In addition, the in vitro data revealed that Dectin-1, Syk, and p-Syk expression was increased following the 3-h OGD and 0, 3, and 6 h of reperfusion in BV2 microglial cells. LAM and PIC also decreased TNF-α and iNOS expression 3 h after OGD/R induction. Conclusion Dectin-1/Syk signaling plays a crucial role in inflammatory activation after ischemic stroke, and further investigation of Dectin-1/Syk signaling in stroke is warranted.
Collapse
Affiliation(s)
- Xin-Chun Ye
- Institute of Stroke Center and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, People's Republic of China.
| | - Qi Hao
- Department of Neurology, Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Xuzhou, People's Republic of China
| | - Wei-Jing Ma
- Institute of Stroke Center and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Qiu-Chen Zhao
- Department of Neurology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, People's Republic of China
| | - Wei-Wei Wang
- Department of Rehabilitation Medicine, Linyi Cancer Hospital, Shandong, People's Republic of China
| | - Han-Han Yin
- Institute of Stroke Center and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Tao Zhang
- Institute of Stroke Center and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Miao Wang
- Institute of Stroke Center and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Kun Zan
- Institute of Stroke Center and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Xin-Xin Yang
- Institute of Stroke Center and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Zuo-Hui Zhang
- Institute of Stroke Center and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Hong-Juan Shi
- Institute of Stroke Center and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Jie Zu
- Institute of Stroke Center and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Hafiz Khuram Raza
- Institute of Stroke Center and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Xue-Ling Zhang
- Department of Neurology, Suqian People's Hospital of Nanjing Drum tower Hospital Group, Suqian, Jiangsu, People's Republic of China
| | - De-Qin Geng
- Institute of Stroke Center and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Jin-Xia Hu
- Institute of Stroke Center and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, People's Republic of China.
| | - Gui-Yun Cui
- Institute of Stroke Center and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, People's Republic of China.
| |
Collapse
|
33
|
Yang H, Graham LC, Reagan AM, Grabowska WA, Schott WH, Howell GR. Transcriptome profiling of brain myeloid cells revealed activation of Itgal, Trem1, and Spp1 in western diet-induced obesity. J Neuroinflammation 2019; 16:169. [PMID: 31426806 PMCID: PMC6700800 DOI: 10.1186/s12974-019-1527-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 06/20/2019] [Indexed: 12/18/2022] Open
Abstract
Background Environmental factors are critical in the development of age-related cognitive decline and dementia. A western diet (WD) can cause nutrient deficiency and inflammation that could impact cognition directly. It is increasingly recognized that innate immune responses by brain myeloid cells, such as resident microglia, and infiltrating peripheral monocytes/macrophages may represent an essential link between a WD, cognitive decline, and dementia. Our previous data demonstrated that chronic consumption of a WD induced inflammation through brain myeloid cells in aging mice and a mouse model of Alzheimer’s disease (AD). However, the subtypes of myeloid cells that contribute to the WD-induced inflammation remain unclear. Methods C57BL/6J (B6), myeloid cell reporter mice (B6.Ccr2RFP/+Cx3cr1GFP/+), and Ccr2-deficient mice (B6.Ccr2RFP/RFP) were fed a WD or a control chow diet (CD) from 2 to 6 or 12 months of age. CD11b+CD45lo and CD11b+CD45hi cells from WD- and CD-fed B6 or Ccr2-deficient mice were characterized using flow cytometry, RNA-sequencing, and immunofluorescence. Results Ccr2::RFP expressing myeloid cells were significantly increased in brains of WD- compared to CD-fed mice, but were not elevated in Ccr2-deficient WD-fed mice. The percent of CD11b+CD45hi cells was significantly increased in WD- compared to CD-fed mice. Comparison of RNA-sequencing data with immune cell data in ImmGen supports that CD11b+CD45hi cells from WD-fed mice are enriched for peripheral monocytes and neutrophils. Ingenuity pathway analysis predicted these cells elicit proinflammatory responses that may be damaging to the brain. Using stringent criteria for gene expression levels between CD11b+CD45hi and CD11b+CD45lo cells, we identified approximately 70 genes that we predict are uniquely expressed in infiltrating cells, including Itgal, Trem1, and Spp1 (osteopontin, OPN). Finally, we show a significantly greater number of OPN+IBA1– cells in WD- compared to CD-fed mice that we propose are activated neutrophils based on ImmGen data. OPN+IBA1– cells are not significantly increased in Ccr2-deficient WD-fed mice. Conclusions These data further support the model that peripheral myeloid cells enter the brain in response to diet-induced obesity. Elucidating their contribution to age-related cognitive decline and age-related neurodegenerative diseases should offer new avenues for therapeutic intervention in Alzheimer’s disease and related dementias, where diet/obesity are major risk factors. Electronic supplementary material The online version of this article (10.1186/s12974-019-1527-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Leah C Graham
- The Jackson Laboratory, Bar Harbor, ME, USA.,Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | | | | | | | - Gareth R Howell
- The Jackson Laboratory, Bar Harbor, ME, USA.,Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA.,Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, USA
| |
Collapse
|
34
|
Maulik M, Mitra S, Sweeney M, Lu B, Taylor BE, Bult-Ito A. Complex interaction of dietary fat and Alaskan bog blueberry supplementation influences manganese mediated neurotoxicity and behavioral impairments. J Funct Foods 2019; 53:306-317. [PMID: 31558914 DOI: 10.1016/j.jff.2018.12.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Dietary fat modulates neuronal health and contributes to age-related nervous system disorders. However, the complex interaction between dietary fat and supplementation and its consequences on neurotoxic pathophysiology has been sparsely explored. The indigenous Alaskan bog blueberry (BB), Vaccinum uliginosum, is known to have anti-inflammatory properties, mostly attributed to its rich polyphenolic content. Here, we evaluate the interplay between dietary fat and BB supplementation on sub-chronic manganese (Mn) exposure that inflicts neurotoxicity and behavioral impairments. In both low-fat and normal-fat diets, BB supplementation attenuated the behavioral and the molecular hallmarks of Mn-induced neurotoxicity. On the contrary, a high-fat diet was found to exacerbate these Mn-induced pathological features. Furthermore, BB supplementation failed to recover the behavioral deficits in mice subjected to a high fat diet in Mn-treated mice. Overall, our results demonstrate the importance of including a dietary regimen comprised of polyphenolic rich supplements with low-fat content in combating age-related neurodegenerative disorders.
Collapse
Affiliation(s)
- Malabika Maulik
- Department of Chemistry and Biochemistry, University of Alaska Fairbanks, Fairbanks, AK, USA.,Biomedical Learning and Student Training (BLaST) Program, University of Alaska Fairbanks, Fairbanks, AK, USA
| | - Swarup Mitra
- IDeA Network of Biomedical Research Excellence (INBRE), University of Alaska Fairbanks, Fairbanks, AK, USA.,Department of Pharmacology and Toxicology, The Research Institution on Addiction, Program in Neuroscience, The State University of New York at Buffalo, Buffalo, NY, USA
| | - McKenzie Sweeney
- Biomedical Learning and Student Training (BLaST) Program, University of Alaska Fairbanks, Fairbanks, AK, USA.,Department of Biology and Wildlife, University of Alaska Fairbanks, Fairbanks, AK, USA
| | - Brianna Lu
- Biomedical Learning and Student Training (BLaST) Program, University of Alaska Fairbanks, Fairbanks, AK, USA.,Department of Biology and Wildlife, University of Alaska Fairbanks, Fairbanks, AK, USA
| | - Barbara E Taylor
- Department of Biological Sciences, College of Natural Science and Mathematics, California State University Long Beach, Long Beach, CA, USA
| | - Abel Bult-Ito
- Biomedical Learning and Student Training (BLaST) Program, University of Alaska Fairbanks, Fairbanks, AK, USA.,Department of Biology and Wildlife, University of Alaska Fairbanks, Fairbanks, AK, USA
| |
Collapse
|
35
|
van Beek AA, Van den Bossche J, Mastroberardino PG, de Winther MPJ, Leenen PJM. Metabolic Alterations in Aging Macrophages: Ingredients for Inflammaging? Trends Immunol 2019; 40:113-127. [PMID: 30626541 DOI: 10.1016/j.it.2018.12.007] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 12/12/2022]
Abstract
Aging is a complex process with an impact on essentially all organs. Declined cellular repair causes increased damage at genomic and proteomic levels upon aging. This can lead to systemic changes in metabolism and pro-inflammatory cytokine production, resulting in low-grade inflammation, or 'inflammaging'. Tissue macrophages, gatekeepers of parenchymal homeostasis and integrity, are prime inflammatory cytokine producers, as well as initiators and regulators of inflammation. In this opinion piece, we summarize intrinsic alterations in macrophage phenotype and function with age. We propose that alternatively activated macrophages (M2-like), which are yet pro-inflammatory, can accumulate in tissues and promote inflammaging. Age-related increases in endoplasmic reticulum stress and mitochondrial dysfunction might be cell-intrinsic forces driving this unusual phenotype.
Collapse
Affiliation(s)
- Adriaan A van Beek
- Top Institute Food and Nutrition, Nieuwe Kanaal 9A, 6709 PA Wageningen, The Netherlands; Cell Biology and Immunology Group, Wageningen University, De Elst 1, 6709 PG Wageningen, The Netherlands; Department of Immunology, Erasmus University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Jan Van den Bossche
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam, Netherlands; Amsterdam UMC, University of Amsterdam, Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam, The Netherlands
| | - Pier G Mastroberardino
- Department of Genetics, Erasmus University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Menno P J de Winther
- Amsterdam UMC, University of Amsterdam, Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam, The Netherlands; Institute for Cardiovascular Prevention (IPEK), Munich, Germany
| | - Pieter J M Leenen
- Department of Immunology, Erasmus University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands.
| |
Collapse
|
36
|
Pires PW, McClain JL, Hayoz SF, Dorrance AM. Mineralocorticoid receptor antagonism prevents obesity-induced cerebral artery remodeling and reduces white matter injury in rats. Microcirculation 2018; 25:e12460. [PMID: 29758591 DOI: 10.1111/micc.12460] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 05/06/2018] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Midlife obesity is a risk factor for dementia development. Obesity has also been linked to hyperaldosteronism, and this can be modeled in rats by high fat (HF) feeding from weaning. Aldosterone, or activation of the mineralocorticoid receptor (MR) causes cerebrovascular injury in lean hypertensive rats. We hypothesized that rats fed a HF diet would show inward middle cerebral artery (MCA) remodeling that could be prevented by MR antagonism. We further proposed that the cerebral artery remodeling would be associated with white mater injury. METHODS Three-week-old male Sprague-Dawley rats were fed a HF diet ± the MR antagonist canrenoic acid (Canr) for 17 weeks. Control rats received normal chow (control NC). MCA structure was assessed by pressure myography. RESULTS The MCAs from HF fed rats had smaller lumens and thicker walls when compared to arteries from control NC rats; Canr prevented the MCA remodeling associated with HF feeding. HF feeding increased the mRNA expression of markers of cell proliferation and vascular inflammation in cerebral arteries and Canr treatment prevented this. White mater injury was increased in the rats fed the HF diet and this was reduced by Canr treatment. The expression of doublecortin, a marker of new and immature neurons was reduced in HF fed rats, and MR antagonism normalized this. CONCLUSIONS These data suggest that HF feeding leads to MR dependent remodeling of the MCA and this is associated with markers of dementia development.
Collapse
Affiliation(s)
- Paulo W Pires
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA.,Department of Pharmacology, Center for Cardiovascular Research, University of Nevada School of Medicine, Reno, NV, USA
| | - Jonathon L McClain
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Sebastian F Hayoz
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA.,Department of Biomedical Sciences, East Tennessee State University, Johnson City, TN, USA
| | - Anne M Dorrance
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| |
Collapse
|