1
|
Zheng F, Dong T, Chen Y, Wang L, Peng G. Border-associated macrophages: From physiology to therapeutic targets in Alzheimer's disease. Exp Neurol 2024; 383:115021. [PMID: 39461707 DOI: 10.1016/j.expneurol.2024.115021] [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: 06/27/2024] [Revised: 10/15/2024] [Accepted: 10/20/2024] [Indexed: 10/29/2024]
Abstract
Border-associated macrophages (BAMs) constitute a highly heterogeneous group of central nervous system-resident macrophages at the brain boundaries. Despite their significance, BAMs have mainly been overlooked compared to microglia, resulting in a limited understanding of their functions. However, recent advancements in single-cell immunophenotyping and transcriptomic analyses of BAMs have revealed a previously unrecognized complexity in these cells, in addition to their critical roles under non-pathological conditions and diseases like Alzheimer's disease (AD), Parkinson's disease, glioma, and ischemic stroke. In this review, we discuss the origins, self-renewal capabilities, and extensive heterogeneity of BAMs, and clarify their important physiological functions such as immune monitoring, waste removal and vascular permeability regulation. We also summarize experimental evidence linking BAMs to the progression of AD. Finally, we review therapeutic strategies targeting brain innate immune cells mainly focusing on strategies aimed at modulating BAMs to treat AD and evaluate their potential in clinical applications.
Collapse
Affiliation(s)
- Fangxue Zheng
- Department of Neurology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Taiwei Dong
- Hangzhou Normal University School of Basic Medical Sciences, Hangzhou, China
| | - Yi Chen
- Department of Neurology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Lang Wang
- Department of Neurology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, Hangzhou, China.
| | - Guoping Peng
- Department of Neurology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
2
|
Muzio L, Perego J. CNS Resident Innate Immune Cells: Guardians of CNS Homeostasis. Int J Mol Sci 2024; 25:4865. [PMID: 38732082 PMCID: PMC11084235 DOI: 10.3390/ijms25094865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Although the CNS has been considered for a long time an immune-privileged organ, it is now well known that both the parenchyma and non-parenchymal tissue (meninges, perivascular space, and choroid plexus) are richly populated in resident immune cells. The advent of more powerful tools for multiplex immunophenotyping, such as single-cell RNA sequencing technique and upscale multiparametric flow and mass spectrometry, helped in discriminating between resident and infiltrating cells and, above all, the different spectrum of phenotypes distinguishing border-associated macrophages. Here, we focus our attention on resident innate immune players and their primary role in both CNS homeostasis and pathological neuroinflammation and neurodegeneration, two key interconnected aspects of the immunopathology of multiple sclerosis.
Collapse
Affiliation(s)
- Luca Muzio
- Neuroimmunology Lab, IRCCS San Raffaele Scientific Institute, Institute of Experimental Neurology, 20133 Milan, Italy;
| | | |
Collapse
|
3
|
Shang Y, Wang X, Su S, Ji F, Shao D, Duan C, Chen T, Liang C, Zhang D, Lu H. Identifying of immune-associated genes for assessing the obesity-associated risk to the offspring in maternal obesity: A bioinformatics and machine learning. CNS Neurosci Ther 2024; 30:e14700. [PMID: 38544384 PMCID: PMC10973700 DOI: 10.1111/cns.14700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 05/14/2024] Open
Abstract
BACKGROUND Perinatal exposure to maternal obesity predisposes offspring to develop obesity later in life. Immune dysregulation in the hypothalamus, the brain center governing energy homeostasis, is pivotal in obesity development. This study aimed to identify key candidate genes associated with the risk of offspring obesity in maternal obesity. METHODS We obtained obesity-related datasets from the Gene Expression Omnibus (GEO) database. GSE135830 comprises gene expression data from the hypothalamus of mouse offspring in a maternal obesity model induced by a high-fat diet model (maternal high-fat diet (mHFD) group and maternal chow (mChow) group), while GSE127056 consists of hypothalamus microarray data from young adult mice with obesity (high-fat diet (HFD) and Chow groups). We identified differentially expressed genes (DEGs) and module genes using Limma and weighted gene co-expression network analysis (WGCNA), conducted functional enrichment analysis, and employed a machine learning algorithm (least absolute shrinkage and selection operator (LASSO) regression) to pinpoint candidate hub genes for diagnosing obesity-associated risk in offspring of maternal obesity. We constructed a nomogram receiver operating characteristic (ROC) curve to evaluate the diagnostic value. Additionally, we analyzed immune cell infiltration to investigate immune cell dysregulation in maternal obesity. Furthermore, we verified the expression of the candidate hub genes both in vivo and in vitro. RESULTS The GSE135830 dataset revealed 2868 DEGs between the mHFD offspring and the mChow group and 2627 WGCNA module genes related to maternal obesity. The overlap of DEGs and module genes in the offspring with maternal obesity in GSE135830 primarily enriched in neurodevelopment and immune regulation. In the GSE127056 dataset, 133 DEGs were identified in the hypothalamus of HFD-induced adult obese individuals. A total of 13 genes intersected between the GSE127056 adult obesity DEGs and the GSE135830 maternal obesity module genes that were primarily enriched in neurodevelopment and the immune response. Following machine learning, two candidate hub genes were chosen for nomogram construction. Diagnostic value evaluation by ROC analysis determined Sytl4 and Kncn2 as hub genes for maternal obesity in the offspring. A gene regulatory network with transcription factor-miRNA interactions was established. Dysregulated immune cells were observed in the hypothalamus of offspring with maternal obesity. Expression of Sytl4 and Kncn2 was validated in a mouse model of hypothalamic inflammation and a palmitic acid-stimulated microglial inflammation model. CONCLUSION Two candidate hub genes (Sytl4 and Kcnc2) were identified and a nomogram was developed to predict obesity risk in offspring with maternal obesity. These findings offer potential diagnostic candidate genes for identifying obesity-associated risks in the offspring of obese mothers.
Collapse
Affiliation(s)
- Yanxing Shang
- Medical Research Center, Affiliated Hospital 2Nantong UniversityNantongChina
- Jiangsu Provincial Medical Key Discipline (Laboratory) Cultivation Unit, Medical Research CenterNantong First People's HospitalNantongChina
- Nantong Clinical Medical College of Kangda College of Nanjing Medical UniversityNantongChina
- Nantong Municipal Key Laboratory of Metabolic Immunology and Disease MicroenvironmentNantong First People's HospitalNantongChina
| | - Xueqin Wang
- Department of Endocrinology, Affiliated Hospital 2Nantong UniversityNantongChina
| | - Sixuan Su
- Medical Research Center, Affiliated Hospital 2Nantong UniversityNantongChina
- Jiangsu Provincial Medical Key Discipline (Laboratory) Cultivation Unit, Medical Research CenterNantong First People's HospitalNantongChina
- Nantong Clinical Medical College of Kangda College of Nanjing Medical UniversityNantongChina
- Nantong Municipal Key Laboratory of Metabolic Immunology and Disease MicroenvironmentNantong First People's HospitalNantongChina
- Department of Pathogen Biology, Medical CollegeNantong UniversityNantongChina
| | - Feng Ji
- Medical Research Center, Affiliated Hospital 2Nantong UniversityNantongChina
- Jiangsu Provincial Medical Key Discipline (Laboratory) Cultivation Unit, Medical Research CenterNantong First People's HospitalNantongChina
- Nantong Clinical Medical College of Kangda College of Nanjing Medical UniversityNantongChina
- Nantong Municipal Key Laboratory of Metabolic Immunology and Disease MicroenvironmentNantong First People's HospitalNantongChina
| | - Donghai Shao
- Medical Research Center, Affiliated Hospital 2Nantong UniversityNantongChina
- Jiangsu Provincial Medical Key Discipline (Laboratory) Cultivation Unit, Medical Research CenterNantong First People's HospitalNantongChina
- Nantong Clinical Medical College of Kangda College of Nanjing Medical UniversityNantongChina
- Nantong Municipal Key Laboratory of Metabolic Immunology and Disease MicroenvironmentNantong First People's HospitalNantongChina
| | - Chengwei Duan
- Medical Research Center, Affiliated Hospital 2Nantong UniversityNantongChina
- Jiangsu Provincial Medical Key Discipline (Laboratory) Cultivation Unit, Medical Research CenterNantong First People's HospitalNantongChina
- Nantong Clinical Medical College of Kangda College of Nanjing Medical UniversityNantongChina
- Nantong Municipal Key Laboratory of Metabolic Immunology and Disease MicroenvironmentNantong First People's HospitalNantongChina
| | - Tianpeng Chen
- Medical Research Center, Affiliated Hospital 2Nantong UniversityNantongChina
- Jiangsu Provincial Medical Key Discipline (Laboratory) Cultivation Unit, Medical Research CenterNantong First People's HospitalNantongChina
- Nantong Clinical Medical College of Kangda College of Nanjing Medical UniversityNantongChina
- Nantong Municipal Key Laboratory of Metabolic Immunology and Disease MicroenvironmentNantong First People's HospitalNantongChina
| | - Caixia Liang
- Medical Research Center, Affiliated Hospital 2Nantong UniversityNantongChina
- Jiangsu Provincial Medical Key Discipline (Laboratory) Cultivation Unit, Medical Research CenterNantong First People's HospitalNantongChina
- Nantong Clinical Medical College of Kangda College of Nanjing Medical UniversityNantongChina
- Nantong Municipal Key Laboratory of Metabolic Immunology and Disease MicroenvironmentNantong First People's HospitalNantongChina
| | - Dongmei Zhang
- Medical Research Center, Affiliated Hospital 2Nantong UniversityNantongChina
- Jiangsu Provincial Medical Key Discipline (Laboratory) Cultivation Unit, Medical Research CenterNantong First People's HospitalNantongChina
- Nantong Clinical Medical College of Kangda College of Nanjing Medical UniversityNantongChina
- Nantong Municipal Key Laboratory of Metabolic Immunology and Disease MicroenvironmentNantong First People's HospitalNantongChina
- Department of Pathogen Biology, Medical CollegeNantong UniversityNantongChina
| | - Hongjian Lu
- Medical Research Center, Affiliated Hospital 2Nantong UniversityNantongChina
- Jiangsu Provincial Medical Key Discipline (Laboratory) Cultivation Unit, Medical Research CenterNantong First People's HospitalNantongChina
- Department of Rehabilitation Medicine, Affiliated Hospital 2Nantong UniversityNantongChina
| |
Collapse
|
4
|
He L, Duan X, Li S, Zhang R, Dai X, Lu M. Unveiling the role of astrocytes in postoperative cognitive dysfunction. Ageing Res Rev 2024; 95:102223. [PMID: 38325753 DOI: 10.1016/j.arr.2024.102223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 02/02/2024] [Accepted: 02/02/2024] [Indexed: 02/09/2024]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder, characterized by progressive cognitive decline and the accumulation of amyloid-beta plaques, tau tangles, and neuroinflammation in the brain. Postoperative cognitive dysfunction (POCD) is a prevalent and debilitating condition characterized by cognitive decline following neuroinflammation and oxidative stress induced by procedures. POCD and AD are two conditions that share similarities in the underlying mechanisms and pathophysiology. Compared to normal aging individuals, individuals with POCD are at a higher risk for developing AD. Emerging evidence suggests that astrocytes, the most abundant glial cells in the central nervous system, play a critical role in the pathogenesis of these conditions. Comprehensive functions of astrocyte in AD has been extensively explored, but very little is known about POCD may experience late-onset AD pathogenesis. Herein, in this context, we mainly explore the multifaceted roles of astrocytes in the context of POCD, highlighting their involvement in neuroinflammation, neurotransmitter regulation, synaptic plasticity and neurotrophic support, and discuss how POCD may augment the onset of AD. Additionally, we discuss potential therapeutic strategies targeting astrocytes to mitigate or prevent POCD, which hold promise for improving the quality of life for patients undergoing surgeries and against AD in the future.
Collapse
Affiliation(s)
- Liang He
- Department of Anesthesiology, Yan'an Hospital of Kunming City, Kunming 650051, China.
| | - Xiyuan Duan
- Department of Anesthesiology, Yan'an Hospital of Kunming City, Kunming 650051, China
| | - Shikuo Li
- Department of Anesthesiology, Yan'an Hospital of Kunming City, Kunming 650051, China
| | - Ruqiang Zhang
- Department of Anesthesiology, Yan'an Hospital of Kunming City, Kunming 650051, China
| | - Xulei Dai
- Department of Clinical Laboratory Science, Xingtai Medical College, Xingtai 050054, China
| | - Meilin Lu
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China.
| |
Collapse
|
5
|
Samoilova YG, Matveeva MV, Spirina LV, Podchinenova DV, Oleinik OA, Galyukova DE. Neuroinflammation in Obese Children. Bull Exp Biol Med 2024; 176:386-389. [PMID: 38340199 DOI: 10.1007/s10517-024-06029-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Indexed: 02/12/2024]
Abstract
Obesity is associated with chronic persistent inflammation due to a pool of tissue macrophages that can penetrate the blood-brain barrier and cause neuroinflammation. The analysis of the association of CD14+CD163+ monocytes in the peripheral blood with cognitive functions in 56 obese children (mean age 11.95 (9.45; 14.45) years) was carried out. The control group consisted of 10 children (mean age 10.4 (9.3; 13.8) years). Standard deviation of the body mass index (SDS BMI) and height (SDS height) were calculated using WHO AnthroPlus software (for children of 6-19 years). Body composition was assessed using bioimpedance measurement. Mononuclear cells were isolated from whole blood by centrifugation on a Ficoll-Urografin density gradient (ρ=1.077 g/ml). The content of CD14+CD163+ monocytes in the peripheral blood was assessed by flow cytometry. To analyze cognitive functions, the intelligence coefficient (IQ) was calculated and a Russian adaptation of the Rey test was performed. We found an increase in the number of M2-polarized CD14+CD163+ monocytes in the peripheral blood with an increase in the obesity degree and in the presence of cognitive decline, as well as a negative correlation of the level of M2-polarized monocytes and IQ, taking into account the excess of visceral fat. The revealed data on the relationship of M2-polarized CD14+CD163+ peripheral blood monocytes with obesity in children and the development of neuropsychological deficiency confirm the role of peripheral visceral obesity and neuroinflammation.
Collapse
Affiliation(s)
- Yu G Samoilova
- Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia
| | - M V Matveeva
- Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia.
| | - L V Spirina
- Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia
| | - D V Podchinenova
- Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia
| | - O A Oleinik
- Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia
| | - D E Galyukova
- Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia
| |
Collapse
|
6
|
Santisteban MM, Schaeffer S, Anfray A, Faraco G, Brea D, Wang G, Sobanko MJ, Sciortino R, Racchumi G, Waisman A, Park L, Anrather J, Iadecola C. Meningeal interleukin-17-producing T cells mediate cognitive impairment in a mouse model of salt-sensitive hypertension. Nat Neurosci 2024; 27:63-77. [PMID: 38049579 PMCID: PMC10999222 DOI: 10.1038/s41593-023-01497-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 10/16/2023] [Indexed: 12/06/2023]
Abstract
Hypertension (HTN), a disease afflicting over one billion individuals worldwide, is a leading cause of cognitive impairment, the mechanisms of which remain poorly understood. In the present study, in a mouse model of HTN, we find that the neurovascular and cognitive dysfunction depends on interleukin (IL)-17, a cytokine elevated in individuals with HTN. However, neither circulating IL-17 nor brain angiotensin signaling can account for the dysfunction. Rather, IL-17 produced by T cells in the dura mater is the mediator released in the cerebrospinal fluid and activating IL-17 receptors on border-associated macrophages (BAMs). Accordingly, depleting BAMs, deleting IL-17 receptor A in brain macrophages or suppressing meningeal T cells rescues cognitive function without attenuating blood pressure elevation, circulating IL-17 or brain angiotensin signaling. Our data unveil a critical role of meningeal T cells and macrophage IL-17 signaling in the neurovascular and cognitive dysfunction in a mouse model of HTN.
Collapse
Affiliation(s)
- Monica M Santisteban
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Samantha Schaeffer
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Antoine Anfray
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Giuseppe Faraco
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - David Brea
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Department of Neuroscience and Experimental Therapeutics, Instituto de Investigaciones Biomédicas de Barcelona, Barcelona, Spain
| | - Gang Wang
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Melissa J Sobanko
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Rose Sciortino
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Gianfranco Racchumi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center, Mainz, Germany
| | - Laibaik Park
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Josef Anrather
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
7
|
Nakajima S, Demers G, Machuca-Parra AI, Pour ZD, Bairamian D, Bouyakdan K, Fisette A, Kabahizi A, Robb J, Rodaros D, Laurent C, Ferreira G, Arbour N, Alquier T, Fulton S. Central activation of the fatty acid sensor GPR120 suppresses microglia reactivity and alleviates sickness- and anxiety-like behaviors. J Neuroinflammation 2023; 20:302. [PMID: 38111048 PMCID: PMC10729532 DOI: 10.1186/s12974-023-02978-5] [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: 06/30/2023] [Accepted: 11/29/2023] [Indexed: 12/20/2023] Open
Abstract
G protein-coupled receptor 120 (GPR120, Ffar4) is a sensor for long-chain fatty acids including omega-3 polyunsaturated fatty acids (n-3 PUFAs) known for beneficial effects on inflammation, metabolism, and mood. GPR120 mediates the anti-inflammatory and insulin-sensitizing effects of n-3 PUFAs in peripheral tissues. The aim of this study was to determine the impact of GPR120 stimulation on microglial reactivity, neuroinflammation and sickness- and anxiety-like behaviors by acute proinflammatory insults. We found GPR120 mRNA to be enriched in both murine and human microglia, and in situ hybridization revealed GPR120 expression in microglia of the nucleus accumbens (NAc) in mice. In a manner similar to or exceeding n-3 PUFAs, GPR120 agonism (Compound A, CpdA) strongly attenuated lipopolysaccharide (LPS)-induced proinflammatory marker expression in primary mouse microglia, including tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β), and inhibited nuclear factor-ĸB translocation to the nucleus. Central administration of CpdA to adult mice blunted LPS-induced hypolocomotion and anxiety-like behavior and reduced TNF-α, IL-1β and IBA-1 (microglia marker) mRNA in the NAc, a brain region modulating anxiety and motivation and implicated in neuroinflammation-induced mood deficits. GPR120 agonist pre-treatment attenuated NAc microglia reactivity and alleviated sickness-like behaviors elicited by central injection TNF-α and IL-1β. These findings suggest that microglial GPR120 contributes to neuroimmune regulation and behavioral changes in response to acute infection and elevated brain cytokines. GPR120 may participate in the protective action of n-3 PUFAs at the neural and behavioral level and offers potential as treatment target for neuroinflammatory conditions.
Collapse
Affiliation(s)
- Shingo Nakajima
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, H3T1J4, Canada
| | - Geneviève Demers
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, H3T1J4, Canada
- Department of Nutrition, Université de Montréal, Montréal, QC, H3T1J4, Canada
| | - Arturo Israel Machuca-Parra
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, H3T1J4, Canada
| | - Zahra Dashtehei Pour
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, H3T1J4, Canada
- Department of Nutrition, Université de Montréal, Montréal, QC, H3T1J4, Canada
| | - Diane Bairamian
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, H3T1J4, Canada
- Department of Nutrition, Université de Montréal, Montréal, QC, H3T1J4, Canada
| | - Khalil Bouyakdan
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, H3T1J4, Canada
| | - Alexandre Fisette
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, H3T1J4, Canada
- Research Group in Cellular Signaling, Department of Medical Biology, Université du Québec à Trois-Rivières, Trois-Rivières, Canada
| | - Anita Kabahizi
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, H3T1J4, Canada
- Department of Nutrition, Université de Montréal, Montréal, QC, H3T1J4, Canada
| | - Josephine Robb
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, H3T1J4, Canada
| | - Demetra Rodaros
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, H3T1J4, Canada
| | - Cyril Laurent
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, H3T1J4, Canada
- Department of Neuroscience, Université de Montréal, Montréal, QC, H3T1J4, Canada
| | - Guillaume Ferreira
- Nutrition and Integrative Neurobiology Unit, UMR 1286, INRA-Université de Bordeaux, Bordeaux, France
| | - Nathalie Arbour
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, H3T1J4, Canada
- Department of Neuroscience, Université de Montréal, Montréal, QC, H3T1J4, Canada
| | - Thierry Alquier
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, H3T1J4, Canada
- Department of Medicine, Université de Montréal, Montréal, QC, H3T1J4, Canada
| | - Stephanie Fulton
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, H3T1J4, Canada.
- Department of Nutrition, Université de Montréal, Montréal, QC, H3T1J4, Canada.
| |
Collapse
|
8
|
Liu J, Aylor KW, Liu Z. Liraglutide and Exercise Synergistically Attenuate Vascular Inflammation and Enhance Metabolic Insulin Action in Early Diet-Induced Obesity. Diabetes 2023; 72:918-931. [PMID: 37074396 PMCID: PMC10281235 DOI: 10.2337/db22-0745] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 04/12/2023] [Indexed: 04/20/2023]
Abstract
Inflammation-induced vascular insulin resistance is an early event in diet-induced obesity and contributes to metabolic insulin resistance. To examine whether exercise and glucagon-like peptide 1 (GLP-1) receptor agonism, alone or in combination, modulate vascular and metabolic insulin actions during obesity development, we performed a euglycemic insulin clamp in adult male rats after 2 weeks of high-fat diet feeding with either access to a running wheel (exercise), liraglutide, or both. Rats exhibited increased visceral adiposity and blunted microvascular and metabolic insulin responses. Exercise and liraglutide alone each improved muscle insulin sensitivity, but their combination fully restored insulin-mediated glucose disposal rates. The combined exercise and liraglutide intervention enhanced insulin-mediated muscle microvascular perfusion, reduced perivascular macrophage accumulation and superoxide production in the muscle, attenuated blood vessel inflammation, and improved endothelial function, along with increasing endothelial nucleus translocation of NRF2 and increasing endothelial AMPK phosphorylation. We conclude that exercise and liraglutide synergistically enhance the metabolic actions of insulin and reduce vascular oxidative stress and inflammation in the early stage of obesity development. Our data suggest that early combination use of exercise and GLP-1 receptor agonism might be an effective strategy in preventing vascular and metabolic insulin resistance and associated complications during the development of obesity. ARTICLE HIGHLIGHTS Inflammation-induced vascular insulin resistance occurs early in diet-induced obesity and contributes to metabolic insulin resistance. We examined whether exercise and GLP-1 receptor agonism, alone or in combination, modulate vascular and metabolic insulin actions during obesity development. We found that exercise and liraglutide synergistically enhanced the metabolic actions of insulin and reduced perimicrovascular macrophage accumulation, vascular oxidative stress, and inflammation in the early stage of obesity development. Our data suggest that early combination use of exercise and a GLP-1 receptor agonist might be an effective strategy in preventing vascular and metabolic insulin resistance and associated complications during the development of obesity.
Collapse
Affiliation(s)
- Jia Liu
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, VA
| | - Kevin W. Aylor
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, VA
| | - Zhenqi Liu
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, VA
| |
Collapse
|
9
|
Chen S, Li J, Meng S, He T, Shi Z, Wang C, Wang Y, Cao H, Huang Y, Zhang Y, Gong Y, Gao Y. Microglia and macrophages in the neuro-glia-vascular unit: From identity to functions. Neurobiol Dis 2023; 179:106066. [PMID: 36889483 DOI: 10.1016/j.nbd.2023.106066] [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: 12/25/2022] [Revised: 02/27/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Although both are myeloid cells located surrounding cerebral vasculature, vessel-associated microglia (VAM) and perivascular macrophages (PVMs) can be distinguished by their distinct morphologies, signatures and microscopic location. As key component of neuro-glia-vascular unit (NGVU), they play prominent roles in neurovasculature development and pathological process of various central nervous system (CNS) diseases, including phagocytosis, angiogenesis, vessel damage/protection and blood flow regulation, therefore serving as potential targets for therapeutics of a broad array of CNS diseases. Herein, we will provide a comprehensive overview of heterogeneity of VAM/PVMs, highlight limitations of current understanding in this field, and discuss possible directions of future investigations.
Collapse
Affiliation(s)
- Shuning Chen
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Jiaying Li
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Shan Meng
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Tingyu He
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Ziyu Shi
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Chenran Wang
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yana Wang
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Hui Cao
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yichen Huang
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yue Zhang
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Ye Gong
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China.
| | - Yanqin Gao
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China.
| |
Collapse
|
10
|
The Concept of Healthy Behaviours in Obesity May Have Unintended Consequences. Nutrients 2022; 15:nu15010012. [PMID: 36615669 PMCID: PMC9824122 DOI: 10.3390/nu15010012] [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: 11/22/2022] [Revised: 12/13/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Obesity has become a global epidemic, representing a major health crisis, with a significant impact both in human and financial terms. Obesity was originally seen as a condition, not a disease, which was considered self-inflicted. Thus, it was understandable that a simplistic approach, such as eat less and move more was proposed to manage obesity. Over the last 25 years, the perception of obesity has been gradually changing and the awareness has risen that it is a disease in its own right and not just a precipitating factor for type 2 diabetes, non-alcoholic fatty liver disease (NAFLD), etc. Creation of a comprehensive algorithm for the management of obesity needs to be informed by an in-depth understanding of the issues impacting the provision of treatment. Promotion of healthy behaviours is essential to help the population become healthier, but these are not obesity treatment strategies. Twenty percent of patients with obesity may respond to approaches based on healthy behaviour, but the 80% who do not respond should not be stigmatised but rather their treatment should be escalated. The unintended consequences of promoting healthy behaviours to patients with obesity can be mitigated by understanding that obesity is likely to be a subset of complex diseases, that require chronic disease management. Once the biology of the disease has been addressed, then healthy behaviours may play an invaluable role in optimising self-care within a chronic disease management strategy.
Collapse
|
11
|
Xie L, Zheng L, Chen W, Zhai X, Guo Y, Zhang Y, Li Y, Yu W, Lai Z, Zhu Z, Li P. Trends in perivascular macrophages research from 1997 to 2021: A bibliometric analysis. CNS Neurosci Ther 2022; 29:816-830. [PMID: 36514189 PMCID: PMC9928555 DOI: 10.1111/cns.14034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 11/02/2022] [Accepted: 11/09/2022] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION Perivascular macrophages (PVMs) play pivotal roles in maintaining the physiological function of the brain. Dysfunction of PVMs is emerging as an important mechanism in various disease conditions in the brain. METHODS In this work, we analyzed recent research advances in PVMs, especially in the brain, from the Web of Science (WoS) core database using bibliometric analysis based on the search terms "perivascular macrophages" and "perivascular macrophage" on October 27, 2021. Visualization and collaboration analysis were performed by Citespace (5.8 R3 mac). RESULTS We found 2384 articles published between 1997 and 2021 in the field of PVMs, which were selected for analysis. PVMs were involved in several physio-pathological fields, in which Neurosciences and Neurology, Neuroscience, Immunology, Pathology, and Cardiovascular System and Cardiology were most reported. The research focuses on PVMs mainly in the central nervous system (CNS), inflammation, macrophage or T-cell, and disease, and highlights the related basic research regarding its activation, oxidative stress, angiotensin II, and insulin resistance. Tumor-associated macrophage, obesity, myeloid cell, and inflammation were relatively recent highlight keywords that attracted increasing attention in recent years. Harvard Univ, Vrije Univ Amsterdam, occupied important positions in the research field of PVMs. Meanwhile, PVM research in China (Peking Univ, Sun Yat Sen Univ, Shanghai Jiao Tong Univ, and Shandong Univ) is on the rise. Cluster co-citation analysis revealed that the mechanisms of CNS PVMs and related brain diseases are major specialties associated with PVMs, while PVMs in perivascular adipose tissue and vascular diseases or obesity are another big category of PVMs hotspots. CONCLUSION In conclusion, the research on PVMs continues to deepen, and the hotspots are constantly changing. Future studies of PVMs could have multiple disciplines intersecting.
Collapse
Affiliation(s)
- Lv Xie
- Department of AnesthesiologyClinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Li Zheng
- Department of AnesthesiologyClinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Weijie Chen
- Department of AnesthesiologyClinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiaozhu Zhai
- Department of AnesthesiologyClinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yunlu Guo
- Department of AnesthesiologyClinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yueman Zhang
- Department of AnesthesiologyClinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yan Li
- Department of AnesthesiologyClinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Weifeng Yu
- Department of AnesthesiologyClinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zhongmeng Lai
- Department of AnesthesiologyFujian Medical University Union HospitalFuzhouFujianChina
| | - Ziyu Zhu
- Department of AnesthesiologyClinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Peiying Li
- Department of AnesthesiologyClinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
12
|
Russo C, Valle MS, Russo A, Malaguarnera L. The Interplay between Ghrelin and Microglia in Neuroinflammation: Implications for Obesity and Neurodegenerative Diseases. Int J Mol Sci 2022; 23:ijms232113432. [PMID: 36362220 PMCID: PMC9654207 DOI: 10.3390/ijms232113432] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/29/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022] Open
Abstract
Numerous studies have shown that microglia are capable of producing a wide range of chemokines to promote inflammatory processes within the central nervous system (CNS). These cells share many phenotypical and functional characteristics with macrophages, suggesting that microglia participate in innate immune responses in the brain. Neuroinflammation induces neurometabolic alterations and increases in energy consumption. Microglia may constitute an important therapeutic target in neuroinflammation. Recent research has attempted to clarify the role of Ghre signaling in microglia on the regulation of energy balance, obesity, neuroinflammation and the occurrence of neurodegenerative diseases. These studies strongly suggest that Ghre modulates microglia activity and thus affects the pathophysiology of neurodegenerative diseases. This review aims to summarize what is known from the current literature on the way in which Ghre modulates microglial activity during neuroinflammation and their impact on neurometabolic alterations in neurodegenerative diseases. Understanding the role of Ghre in microglial activation/inhibition regulation could provide promising strategies for downregulating neuroinflammation and consequently for diminishing negative neurological outcomes.
Collapse
Affiliation(s)
- Cristina Russo
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| | - Maria Stella Valle
- Laboratory of Neuro-Biomechanics, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Antonella Russo
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Lucia Malaguarnera
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
- Correspondence:
| |
Collapse
|
13
|
Trends in Gliosis in Obesity, and the Role of Antioxidants as a Therapeutic Alternative. Antioxidants (Basel) 2022; 11:antiox11101972. [PMID: 36290695 PMCID: PMC9598641 DOI: 10.3390/antiox11101972] [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: 08/24/2022] [Revised: 09/23/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022] Open
Abstract
Obesity remains a global health problem. Chronic low-grade inflammation in this pathology has been related to comorbidities such as cognitive alterations that, in the long term, can lead to neurodegenerative diseases. Neuroinflammation or gliosis in patients with obesity and type 2 diabetes mellitus has been related to the effect of adipokines, high lipid levels and glucose, which increase the production of free radicals. Cerebral gliosis can be a risk factor for developing neurodegenerative diseases, and antioxidants could be an alternative for the prevention and treatment of neural comorbidities in obese patients. AIM Identify the immunological and oxidative stress mechanisms that produce gliosis in patients with obesity and propose antioxidants as an alternative to reducing neuroinflammation. METHOD Advanced searches were performed in scientific databases: PubMed, ProQuest, EBSCO, and the Science Citation index for research on the physiopathology of gliosis in obese patients and for the possible role of antioxidants in its management. CONCLUSION Patients with obesity can develop neuroinflammation, conditioned by various adipokines, excess lipids and glucose, which results in an increase in free radicals that must be neutralized with antioxidants to reduce gliosis and the risk of long-term neurodegeneration.
Collapse
|
14
|
Clayton RW, Lovell-Badge R, Galichet C. The Properties and Functions of Glial Cell Types of the Hypothalamic Median Eminence. Front Endocrinol (Lausanne) 2022; 13:953995. [PMID: 35966104 PMCID: PMC9363565 DOI: 10.3389/fendo.2022.953995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 06/17/2022] [Indexed: 02/06/2023] Open
Abstract
The median eminence (ME) is part of the neuroendocrine system (NES) that functions as a crucial interface between the hypothalamus and pituitary gland. The ME contains many non-neuronal cell types, including oligodendrocytes, oligodendrocyte precursor cells (OPCs), tanycytes, astrocytes, pericytes, microglia and other immune cells, which may be involved in the regulation of NES function. For example, in mice, ablation of tanycytes (a special class of ependymal glia with stem cell-like functions) results in weight gain, feeding, insulin insensitivity and increased visceral adipose, consistent with the demonstrated ability of these cells to sense and transport both glucose and leptin, and to differentiate into neurons that control feeding and metabolism in the hypothalamus. To give a further example, OPCs in the ME of mice have been shown to rapidly respond to dietary signals, in turn controlling composition of the extracellular matrix in the ME, derived from oligodendrocyte-lineage cells, which may contribute to the previously described role of these cells in actively maintaining leptin-receptor-expressing dendrites in the ME. In this review, we explore and discuss recent advances such as these, that have developed our understanding of how the various cell types of the ME contribute to its function in the NES as the interface between the hypothalamus and pituitary gland. We also highlight avenues of future research which promise to uncover additional functions of the ME and the glia, stem and progenitor cells it contains.
Collapse
|