1
|
Dali R, Langlet F. Tanycytes in the nexus of hypothalamic inflammation, appetite control, and obesity. Physiol Behav 2025; 296:114917. [PMID: 40222438 DOI: 10.1016/j.physbeh.2025.114917] [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: 12/30/2024] [Revised: 03/16/2025] [Accepted: 04/10/2025] [Indexed: 04/15/2025]
Abstract
Hypothalamic inflammation has been identified as a critical factor driving the development of obesity and associated metabolic disorders. This inflammation-related disruption of energy balance relies on alterations in metabolic cues sensing and hypothalamic cellular functions, together leading to overeating and weight gain. Within the hypothalamic cellular networks controlling energy balance, recent studies have highlighted the significance of glial dysfunction in these processes, suggesting that these cells could provide new avenues for weight loss therapies. Glia rapidly activates following the consumption of a high-fat diet, even after a very short exposure, and contributes to the disruption of the entire system through inflammatory crosstalk. This review explores recent progress in understanding the molecular interactions between glial cells and neurons in hypothalamic inflammation related to obesity, diabetes, and associated complications. Notably, it highlights specialized ependymal cells called tanycytes, whose role is still underestimated in hypothalamic inflammation, and examines the potential for targeting this cell type as a treatment strategy for metabolic disorders.
Collapse
Affiliation(s)
- Rafik Dali
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland; Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Fanny Langlet
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland; Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
2
|
He Q, Bennett AN, Zhang C, Zhang JY, Tong S, Chan KHK. Nutritional interventions for preventing cognitive decline in patients with mild cognitive impairment and Alzheimer's disease: A comprehensive network meta-analysis and Mendelian Randomization study. Clin Nutr ESPEN 2025; 67:555-566. [PMID: 40147763 DOI: 10.1016/j.clnesp.2025.03.040] [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: 01/24/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND As the population ages rapidly, cognitive impairment, especially in conditions like Alzheimer's disease (AD) and mild cognitive impairment (MCI), has become a crucial public health issue. Nutritional interventions have garnered attention as a promising non-pharmacological strategy for maintaining cognitive function and decelerating its decline. OBJECTIVE This study aimed to evaluate the effectiveness of various nutritional interventions in preventing cognitive impairment and elucidate intricate biological pathways linking nutritional interventions to cognitive function through a comprehensive approach involving systematic review, network meta-analysis (NMA), and Mendelian randomization (MR) analysis. METHODS We utilized pair-wise comparisons and NMA to evaluate the efficacy of different nutritional interventions on cognitive function in patients with decreased cognitive abilities. A systematic search in three biomedical databases was performed for double-blinded, randomized controlled trials (RCTs) or head-to-head comparisons up to December 31, 2024. The NMA has been registered at the International Prospective Register of Systematic Reviews (PROSPERO; CRD42022331173). Moreover, to clarify the biological mechanisms linking nutritional interventions to cognitive impairment, we conducted two-sample MR analyses to assess the potential causal relationships between 9 genetically predicted nutrient levels derived from extensive genome-wide association studies (GWASs) and 12 biomarkers linked to brain aging. RESULTS This study encompassed 52 trials with 8452 participants, 9 GWASs examining genetically predicted nutrient levels with a total of 603,996 participants, and 12 GWASs investigating brain aging biomarkers with a total of 2,405,530 participants. The NMA demonstrated that the multi-ingredient intervention outperformed other interventions significantly (standardized mean difference [SMD] = 2.03; 95 % credible interval [95 % CrI] = 0.97-3.09, P = 0.0002). In the MR analysis, the findings indicated that the multi-ingredient intervention was linked to reduced C-reactive protein (CRP) levels (odds ratios [OR] = 0.96, 95 % confidence interval [95 % CI] = 0.93-0.99, P = 0.014), suggesting that the multi-ingredient intervention may mitigate cognitive impairment by reducing inflammation. CONCLUSIONS Our NMA amalgamated evidence underscoring multi-ingredient interventions as the most efficacious strategy for attenuating cognitive decline in individuals with MCI and AD. Furthermore, the MR analysis unveiled the mechanisms underpinning the protective effects of multi-ingredient interventions, potentially offering benefits even in the early stages of neurodegeneration by mitigating oxidative stress and inflammation.
Collapse
Affiliation(s)
- Qian He
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Adam N Bennett
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Chuyun Zhang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Jia Yue Zhang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Shuyao Tong
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Kei Hang Katie Chan
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong Special Administrative Region of China; Department of Electrical Engineering, City University of Hong Kong, Hong Kong Special Administrative Region of China; Department of Epidemiology, Centre for Global Cardiometabolic Health, Brown University, RI, USA.
| |
Collapse
|
3
|
Tsuruta K, Sato Y, Nango H, Sakata Y, Ishikawa H, Tsuboi M, Miyagishi H, Kosuge Y. Pentadecyl®, an odd-chain-rich triglyceride mixture derived from Aurantiochytrium oil, attenuates lipopolysaccharide-induced inflammatory cytokine production in BV-2 microglial cells. Int Immunopharmacol 2025; 158:114810. [PMID: 40349404 DOI: 10.1016/j.intimp.2025.114810] [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: 02/25/2025] [Revised: 04/26/2025] [Accepted: 05/04/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND AND AIM Microglia are the primary immune cells of the central nervous system that play pivotal roles in health and disease. Abnormally activated microglia secrete proinflammatory factors and play essential roles in neurodegenerative disease progression. This study investigated the potential effects of Pentadecyl, rich in odd-numbered fatty acids, such as pentadecanoic acid, isolated from Aurantiochytrium limacinum, on the lipopolysaccharide (LPS)-induced immune response of BV-2 microglial cells. EXPERIMENTAL PROCEDURE Cell viability was detected using MTT and LIVE/DEAD assays. mRNA and protein levels of inflammatory cytokines and signaling factors were assessed using real-time PCR and western blotting, respectively. RESULTS AND CONCLUSION Pentadecyl did not affect MTT-reducing activity or the number of dead cells stained with ethidium homodimer-1. Pentadecyl selectively mitigated the LPS-induced overproduction of pro-inflammatory cytokines, including interleukin (IL)-6 and IL-1β, at the transcriptional and protein levels, whereas tumor necrosis factor-alpha (TNF-α) expression remained unchanged. Western blot analysis showed that Pentadecyl downregulated the LPS-induced increase in the phosphorylation of signal transducer and activator of transcription 3 (STAT3) but did not affect the phosphorylation of p65, a component of nuclear factor-kappa B, or p38 and c-Jun N-terminal kinase, both of which are mitogen-activated protein kinases. Similar to Pentadecyl, Stattic, a representative STAT3 inhibitor, preferentially suppressed the LPS-induced upregulation of IL-6 and IL-1β mRNA expression, whereas its inhibitory effect on TNF-α expression was relatively modest. These results indicate that Pentadecyl suppresses LPS-induced pro-inflammatory cytokine production without affecting cell survival by regulating the STAT3 signaling pathway in BV-2 cells.
Collapse
Affiliation(s)
- Komugi Tsuruta
- Laboratory of Pharmacology, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi-Shi, Chiba 274-8555, Japan..
| | - Yusei Sato
- Laboratory of Pharmacology, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi-Shi, Chiba 274-8555, Japan..
| | - Hiroshi Nango
- Laboratory of Pharmacology, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi-Shi, Chiba 274-8555, Japan..
| | - Yasuko Sakata
- Research and Development Division of Sea Act, Sea Act Co., Ltd., 2-17-8 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan..
| | - Hideaki Ishikawa
- Research and Development Division of Sea Act, Sea Act Co., Ltd., 2-17-8 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan..
| | - Makoto Tsuboi
- Research and Development Division of Sea Act, Sea Act Co., Ltd., 2-17-8 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan..
| | - Hiroko Miyagishi
- Laboratory of Pharmacology, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi-Shi, Chiba 274-8555, Japan..
| | - Yasuhiro Kosuge
- Laboratory of Pharmacology, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi-Shi, Chiba 274-8555, Japan..
| |
Collapse
|
4
|
Yang L, Geng Y, Qian Y, Zong N, Xia S, Yang H, Bao X, Chen J, Xu Y. Loss of Plxdc2 exacerbates microglia-mediated neuroinflammation and ischemic brain injury. Exp Neurol 2025; 391:115302. [PMID: 40345569 DOI: 10.1016/j.expneurol.2025.115302] [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: 03/29/2025] [Revised: 04/30/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
Microglia are the supervisors maintaining intracerebral homeostasis, which function importantly in determining the outcome of ischemic stroke. Plxdc2 is a single-transmembrane protein and mainly studied in the development of central nervous system and cancers, whereas its role in the function of microglia remains elusive. In this study, based on our previous scRNA-seq of ischemic brain and transcriptomic analysis of microglia isolated from the ischemic brain, we found that Plxdc2 was abundantly expressed in microglia and remarkably downregulated after stroke. Further, with adeno-associated virus (AAV) overexpressing or lentivirus interfering Plxdc2 in microglia in vivo, Plxdc2 was proved to protect against ischemic brain injury. Plxdc2 helps maintain microglial homeostatic state both in vitro and in vivo, and downregulation of Plxdc2 exacerbated microglial inflammatory response. In addition, we found that Plxdc2 participated in regulating the activation of NF-κB p65 signaling, and also modulated microglial lipid metabolism. Moreover, Plxdc2 was found to facilitate the activation of PPARγ, which might account for its impact on NF-κB p65 signaling and lipid metabolism in microglia. Overall, our results illustrated a vital role of Plxdc2 in modulating post-stroke microglial activation, which holds potential to be a novel target for immunomodulation in ischemic stroke.
Collapse
Affiliation(s)
- Lixuan Yang
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210000, China
| | - Yang Geng
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210000, China
| | - Yi Qian
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210000, China
| | - Ningning Zong
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210000, China
| | - Shengnan Xia
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210000, China
| | - Haiyan Yang
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210000, China
| | - Xinyu Bao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210000, China
| | - Jian Chen
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210000, China.
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210000, China; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China; Nanjing Neurology Clinical Medical Center, Nanjing, China.
| |
Collapse
|
5
|
Li HD, Zheng JY, Tan KW, Su JX, Chen W, Pang RK, Wu GL, Qiu YH, Li XX, Cai YF, Zhang SJ. Salvianolic acid B (SalB) improves high-fat diet (HFD)-caused cognitive impairment in mice by modulating the Trem2/Dap12 pathway in vivo and in vitro. Int Immunopharmacol 2025; 153:114461. [PMID: 40101423 DOI: 10.1016/j.intimp.2025.114461] [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: 12/13/2024] [Revised: 02/23/2025] [Accepted: 03/08/2025] [Indexed: 03/20/2025]
Abstract
Salvianolic acid B (SalB), which extracted from Salvia miltiorrhiza Bunge (Labiatae), is a traditional Chinese medicine. SalB is widely used in nervous system diseases. This study evaluated the protective effect of SalB on high-fat diet (HFD)-induced cognitive impairment and its mechanisms in vivo and in vitro. The behavior tests demonstrated that SalB alleviated motor skills and learning capacity in HFD mice. Animal experiments have confirmed that SalB reduced the mRNA expression of inflammatory markers and the Trem2/Dap12 pathway in HIP. Furthermore, SalB inhibited the microglia Trem2/Dap12 pathway in HIP. In vivo, palmitic acid (PA) was used to intervene in BV2 cells to construct an inflammatory. SalB reduced the mRNA expression of inflammatory markers and inhibited the Trem2/Dap12 pathway in BV2 cells. In conclusion, SalB treatment may serve as a possible therapy for cognitive impairment induced by HFD.
Collapse
Affiliation(s)
- Hong-Dan Li
- Department of Neurology, Nanning Hospital of Traditional Chinese Medicine, Nanning 530000, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou 510000, China
| | - Jia-Yi Zheng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou 510000, China
| | - Kai-Wen Tan
- Department of Neurology, Nanning Hospital of Traditional Chinese Medicine, Nanning 530000, China
| | - Jin-Xun Su
- Department of Neurology, Nanning Hospital of Traditional Chinese Medicine, Nanning 530000, China
| | - Wei Chen
- Department of Neurology, Nanning Hospital of Traditional Chinese Medicine, Nanning 530000, China
| | - Rui-Kang Pang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou 510000, China
| | - Guang-Liang Wu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou 510000, China
| | - Yu-Hui Qiu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou 510000, China
| | - Xiao-Xiao Li
- Research Center for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hong Kong 999077, China; State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518000, China.
| | - Ye-Feng Cai
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou 510000, China.
| | - Shi-Jie Zhang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou 510000, China.
| |
Collapse
|
6
|
Crain E, Minaya DM, de La Serre CB. Microbiota-induced inflammation mediates the impacts of a Western diet on hippocampal-dependent memory. Nutr Res 2025; 138:89-106. [PMID: 40339190 DOI: 10.1016/j.nutres.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 04/01/2025] [Accepted: 04/01/2025] [Indexed: 05/10/2025]
Abstract
Obesity is associated with impaired hippocampal-dependent memory, but the mechanisms driving this pathology are not fully understood. Western diets (WD) contribute to obesity, and previous reviews have described a role for WD in impaired hippocampal-dependent memory. However, there is need for a more detailed description of the pathways by which WD may impair memory. The short vs long-term effect of specific dietary components on brain structure and functions as well as the precise mechanism and molecular pathways involved are still not fully understood. This review focuses on the mechanisms and effects of gut microbiota-driven neuroinflammation. WD leads to changes and imbalance in bacterial taxa abundances that are deleterious to the host health (gut dysbiosis) and studies in rodent models show these changes are sufficient to impair hippocampal-dependent memory. Here, we discuss a variety of proposed mechanisms linking microbiota composition to hippocampal function, with a focus on neuroinflammation. Gut microbiota impacts gastrointestinal barrier function, leading to increased circulating proinflammatory bacterial products, increased blood-brain barrier permeability, and neuroinflammation.
Collapse
Affiliation(s)
- Eden Crain
- Department of Nutritional Sciences, University of Georgia, Athens, GA, USA
| | - Dulce M Minaya
- Department of Nutritional Sciences, University of Georgia, Athens, GA, USA
| | - Claire B de La Serre
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
7
|
Ogawa S, Hori H, Niwa M, Itoh M, Lin M, Yoshida F, Ino K, Kawanishi H, Narita M, Nakano W, Imai R, Matsui M, Kamo T, Kunugi H, Hattori K, Kim Y. Serum lipid and plasma fatty acid profiles in PTSD patients and healthy individuals: Associations with symptoms, cognitive function, and inflammatory markers. Prog Neuropsychopharmacol Biol Psychiatry 2025; 138:111298. [PMID: 39988258 DOI: 10.1016/j.pnpbp.2025.111298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 01/22/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Increasing evidence suggests that posttraumatic stress disorder (PTSD), a serious mental health condition, is associated with physical health problems. Lipid-related molecules are crucial for central nervous system functions associated with PTSD symptoms; however, case-control studies exploring the relationship between PTSD and lipid-related molecules are scarce. We examined 68 civilian PTSD patients and 97 healthy controls, evaluating PTSD symptoms, childhood maltreatment history, suicidality, and cognitive functions. Cholesterol, triglycerides, and inflammation-related marker levels were analyzed in serum, while fatty acid levels were measured in plasma. Compared to controls, patients exhibited significantly lower high-density lipoprotein cholesterol and n-6 linoleic acid levels, alongside higher saturated palmitic acid levels and the triene-to-tetraene (T/T) ratio. PTSD symptoms, particularly hyperarousal, were significantly positively correlated with n-6 γ-linolenic, n-6 dihomo-γ-linolenic, and n-9 mead acid levels, and the T/T ratio. Cognitive functions were significantly positively correlated with n-3 docosahexaenoic acid and total n-3 fatty acid levels, and negatively correlated with saturated lauric, palmitic, and total saturated fatty acid levels. Suicidality was significantly positively correlated with dihomo-γ-linolenic acid, mead acid levels, and the T/T ratio, and negatively correlated with polyunsaturated fatty acid (PUFA) levels. Inflammation-related marker levels were significantly correlated with higher palmitic, n-9 oleic, and total n-9 fatty acid levels, and lower linoleic acid and PUFA levels. Latent profile analysis (LPA) revealed distinct subgroups associated with unique fatty acid profiles. These lipid-related alterations may improve the understanding of PTSD pathophysiology. Distinct fatty acid profiles identified by LPA may help subtype PTSD patients and guide nutrition-based personalized treatment strategies.
Collapse
Affiliation(s)
- Shintaro Ogawa
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan; Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.
| | - Hiroaki Hori
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan; Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.
| | - Madoka Niwa
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Mariko Itoh
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan; Center for Environmental and Health Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Mingming Lin
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan; Laboratory for Imagination and Executive Functions, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Fuyuko Yoshida
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan; Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Keiko Ino
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan; Department of Psychiatry and Cognitive-Behavioral Medicine, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Hitomi Kawanishi
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan; Department of Advanced Neuroimaging, Integrative Brain Imaging Center, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan; Department of Behavioral Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Megumi Narita
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Wakako Nakano
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Risa Imai
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan; Department of Psychiatry and Cognitive-Behavioral Medicine, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan; Risa Irinaka Mental Clinic, Nagoya, Aichi, Japan
| | - Mie Matsui
- Department of Clinical Cognitive Neuroscience, Institute of Liberal Arts and Science, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Toshiko Kamo
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan; Wakamatsu-cho Mental and Skin Clinic, Shinjuku, Tokyo, Japan
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan; Department of Psychiatry, Teikyo University School of Medicine, Itabashi, Tokyo, Japan
| | - Kotaro Hattori
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan; Department of Bioresources, Medical Genome Center, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Yoshiharu Kim
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| |
Collapse
|
8
|
Prakash P, Randolph CE, Walker KA, Chopra G. Lipids: Emerging Players of Microglial Biology. Glia 2025; 73:657-677. [PMID: 39688320 PMCID: PMC11784843 DOI: 10.1002/glia.24654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024]
Abstract
Lipids are small molecule immunomodulators that play critical roles in maintaining cellular health and function. Microglia, the resident immune cells of the central nervous system, regulate lipid metabolism both in the extracellular environment and within intracellular compartments through various mechanisms. For instance, glycerophospholipids and fatty acids interact with protein receptors on the microglial surface, such as the Triggering Receptor Expressed on Myeloid Cells 2, influencing cellular functions like phagocytosis and migration. Moreover, cholesterol is essential not only for microglial survival but, along with other lipids such as fatty acids, is crucial for the formation, function, and accumulation of lipid droplets, which modulate microglial activity in inflammatory diseases. Other lipids, including acylcarnitines and ceramides, participate in various signaling pathways within microglia. Despite the complexity of the microglial lipidome, only a few studies have investigated the effects of specific lipid classes on microglial biology. In this review, we focus on major lipid classes and their roles in modulating microglial function. We also discuss novel analytical techniques for characterizing the microglial lipidome and highlight gaps in current knowledge, suggesting new directions for future research on microglial lipid biology.
Collapse
Affiliation(s)
- Priya Prakash
- Department of ChemistryPurdue UniversityWest LafayetteIndianaUSA
- Neuroscience Institute, NYU Grossman School of MedicineNew YorkNew YorkUSA
| | | | | | - Gaurav Chopra
- Department of ChemistryPurdue UniversityWest LafayetteIndianaUSA
- Purdue Institute for Integrative Neuroscience, Purdue UniversityWest LafayetteIndianaUSA
- Purdue Institute for Drug Discovery, Purdue UniversityWest LafayetteIndianaUSA
- Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue UniversityWest LafayetteIndianaUSA
- Regenstrief Center for Healthcare Engineering, Purdue UniversityWest LafayetteIndianaUSA
| |
Collapse
|
9
|
Song M, Bai Y, Song F. High-fat diet and neuroinflammation: The role of mitochondria. Pharmacol Res 2025; 212:107615. [PMID: 39842474 DOI: 10.1016/j.phrs.2025.107615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 12/28/2024] [Accepted: 01/17/2025] [Indexed: 01/24/2025]
Abstract
In recent years, increasing evidence has supported that high-fat diet (HFD) can induce the chronic, low-grade neuroinflammation in the brain, which is closely associated with the impairment of cognitive function. As the key organelles responsible for energy metabolism in the cell, mitochondria are believed to involved in the pathogenesis of a variety of neurological disorders. This review summarizes the current progress in the field of the relationship between HFD exposure and neurodegenerative diseases, and outline the major routines of HFD induced neuroinflammation and its pathological significance in the pathogenesis of neurodegenerative diseases. Furthermore, the article highlights the pivotal role of mitochondrial dysfunction in driving the neuroinflammation in the setting of HFD. Danger-associated molecular patterns (DAMPs) from damaged mitochondria can activate innate immune signaling pathways, while mitochondrial dysfunction itself can lead to metabolic remodeling of inflammatory cells, thus inducing neuroinflammation. More importantly, mitochondrial damage, neuroinflammation, and insulin resistance caused by HFD form a mutually reinforcing vicious cycle, ultimately leading to the death of neurons and promoting the progression of neurodegenerative diseases. Thus, in-depth elucidation of the role and underlying mechanisms of mitochondrial dysfunction in HFD-induced metabolic disorders may not only expand our understanding of the mechanistic linkages between HFD and etiology of neurodegenerative diseases, but also help develop the specific strategies for the prevention and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Mingxue Song
- Department of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, China.
| | - Yao Bai
- NHC Key Laboratory of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing 100021, China.
| | - Fuyong Song
- Department of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, China.
| |
Collapse
|
10
|
Hou J, Lv Z, Wang Y, Chen D. The gut microbiota regulates diabetic retinopathy in adult rats. Front Microbiol 2025; 16:1479792. [PMID: 39949626 PMCID: PMC11822567 DOI: 10.3389/fmicb.2025.1479792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 01/06/2025] [Indexed: 02/16/2025] Open
Abstract
Introduction Diabetic retinopathy (DR) is the most common complication of diabetes. Neuronal apoptosis, activated microglia, and microvascular changes are early features of DR. The gut microbiota is critical for the maturation and activation of microglia in the brain, and DR patients exhibit gut dysbiosis. However, the effect of the gut microbiota on retinal microglia under normal or diabetic conditions is still unclear. Methods Type 2 diabetes (T2D) was established in male adult Brown Norway (BN) rats, and they were treated with gavage of broad-spectrum antibiotic (ABX) suspension. Retinal fundus fluorescein angiography was performed to observe the dynamic growth process and leakage of blood vessels. Retro-orbital injection of FITC-Dextran was performed to observe the changes in blood-retinal barriers. After treatment with ABX and diabetes lasting for more than 6 months, 16S RNA sequencing of stool samples was performed to determine changes in the gut microbiome and mass spectrometry was used to analyze metabolome changes. IBA1, IB4, and Brn3 staining were performed on adult rats' retinal wholemount or sections to observe the changes in microglia, blood vessels and the number of ganglion cells. Results Long-term (6 months) T2D caused gut dysbiosis with increased average taxa numbers. We showed that broad-spectrum antibiotics (ABXs) gavage can reduce the average number of gut microbiota taxa and retinal microglia in adult male BN rats with or without T2D. Interestingly, adult male BN rats with T2D for more than 6 months showed a loss of retinal ganglion cells (RGCs) without significant changes in retinal microglia or retinal vascular vessels. However, ABX gavage reduced retinal microglia and alleviated RGC damage in these T2D rats. Conclusion Our data suggests that ABX gavage-induced gut dysbiosis can reduce retinal microglia in adult rats and alleviate RGC loss in long-term T2D rats. Targeting the gut microbiota may be a future therapeutic strategy for DR management.
Collapse
Affiliation(s)
- Jueyu Hou
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
- Research Laboratory of Ophthalmology and Vision Sciences, Eye Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Zhongping Lv
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
- Research Laboratory of Ophthalmology and Vision Sciences, Eye Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Yujiao Wang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
- Research Laboratory of Ophthalmology and Vision Sciences, Eye Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Danian Chen
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
- Research Laboratory of Ophthalmology and Vision Sciences, Eye Research Institute, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Colombo G, Monsorno K, Paolicelli RC. Metabolic control of microglia in health and disease. HANDBOOK OF CLINICAL NEUROLOGY 2025; 209:143-159. [PMID: 40122622 DOI: 10.1016/b978-0-443-19104-6.00009-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Metabolic states within cells are tightly linked to functional outcomes and finely regulated by nutrient availability. A growing body of the literature supports the idea that various metabolites can influence cellular functions, such as cell differentiation, migration, and proliferation in different contexts, with ample evidence coming from the immune system. Additionally, certain functional programs can trigger significant metabolic changes within cells, which are crucial not only to meet high energy demands, but also to produce intermediate metabolites necessary to support specific tasks. Microglia, the resident innate immune cells of the central nervous system, are constantly active, surveying the brain parenchyma and providing support to neighboring cells in the brain. They exhibit high metabolic flexibility, capable of quickly undergoing metabolic reprogramming based on nutrient availability and functional requirements. In this chapter, we will discuss the major metabolic pathways within cells and provide examples of how relevant enzymes and metabolites can impact microglial function in physiologic and pathologic contexts.
Collapse
Affiliation(s)
- Gloria Colombo
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Katia Monsorno
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Rosa C Paolicelli
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
12
|
Ontiveros-Ángel P, Vega-Torres JD, Simon TB, Williams V, Inostroza-Nives Y, Alvarado-Crespo N, Gonzalez YV, Pompolius M, Katzka W, Lou J, Sharafeddin F, De la Peña I, Dong T, Gupta A, Viet CT, Febo M, Obenaus A, Nair A, Figueroa JD. Early-life obesogenic environment integrates immunometabolic and epigenetic signatures governing neuroinflammation. Brain Behav Immun Health 2024; 42:100879. [PMID: 39430879 PMCID: PMC11490928 DOI: 10.1016/j.bbih.2024.100879] [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: 09/24/2024] [Accepted: 09/29/2024] [Indexed: 10/22/2024] Open
Abstract
Childhood overweight/obesity is associated with stress-related psychopathology, yet the pathways connecting childhood obesity to stress susceptibility are poorly understood. We employed a systems biology approach with 62 adolescent Lewis rats fed a Western-like high-saturated fat diet (WD, 41% kcal from fat) or a control diet (CD, 13% kcal from fat). A subset of rats underwent a 31-day model of predator exposures and social instability (PSS). Effects were assessed using behavioral tests, DTI (diffusion tensor imaging), NODDI (neurite orientation dispersion and density imaging), 16S rRNA gene sequencing for gut microbiome profiling, hippocampal microglia analysis, and targeted gene methylation. Parallel experiments on human microglia cells (HMC3) examined how palmitic acid influences cortisol-related inflammatory responses. Rats exposed to WD and PSS exhibited deficits in sociability, increased fear/anxiety-like behaviors, food consumption, and body weight. WD/PSS altered hippocampal microstructure (subiculum, CA1, dentate gyrus), and microbiome analysis showed a reduced abundance of members of the phylum Firmicutes. WD/PSS synergistically promoted neuroinflammatory changes in hippocampal microglia, linked with microbiome shifts and altered Fkbp5 expression/methylation. In HMC3, palmitate disrupted cortisol responses, affecting morphology, phagocytic markers, and cytokine release, partially mediated by FKBP5. This study identifies gene-environment interactions that influence microglia biology and may contribute to the connection between childhood obesity and stress-related psychopathology later in life.
Collapse
Affiliation(s)
- Perla Ontiveros-Ángel
- Center for Health Disparities and Molecular Medicine and Department of Basic Sciences, Physiology Division, Department of Basic Sciences, Loma Linda University Health School of Medicine, Loma Linda, CA, USA
| | - Julio David Vega-Torres
- Center for Health Disparities and Molecular Medicine and Department of Basic Sciences, Physiology Division, Department of Basic Sciences, Loma Linda University Health School of Medicine, Loma Linda, CA, USA
| | - Timothy B. Simon
- Center for Health Disparities and Molecular Medicine and Department of Basic Sciences, Physiology Division, Department of Basic Sciences, Loma Linda University Health School of Medicine, Loma Linda, CA, USA
| | - Vivianna Williams
- Center for Health Disparities and Molecular Medicine and Department of Basic Sciences, Physiology Division, Department of Basic Sciences, Loma Linda University Health School of Medicine, Loma Linda, CA, USA
| | - Yaritza Inostroza-Nives
- Department of Biochemistry and Pharmacology, San Juan Bautista School of Medicine, Caguas, Puerto Rico, USA
| | - Nashareth Alvarado-Crespo
- Department of Biochemistry and Pharmacology, San Juan Bautista School of Medicine, Caguas, Puerto Rico, USA
| | - Yarimar Vega Gonzalez
- Department of Biochemistry and Pharmacology, San Juan Bautista School of Medicine, Caguas, Puerto Rico, USA
| | - Marjory Pompolius
- Translational Research Imaging Laboratory, Department of Psychiatry, Department of Neuroscience, College of Medicine, University of Florida Health, Gainesville, FL, USA
| | - William Katzka
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, UCLA Microbiome Center, University of California, Los Angeles, CA, USA
| | - John Lou
- Loma Linda University Health School of Behavioral Health, Loma Linda, CA, USA
| | - Fransua Sharafeddin
- Center for Health Disparities and Molecular Medicine and Department of Basic Sciences, Physiology Division, Department of Basic Sciences, Loma Linda University Health School of Medicine, Loma Linda, CA, USA
| | - Ike De la Peña
- Department of Pharmaceutical and Administrative Sciences, Loma Linda University Health School of Pharmacy, Loma Linda, CA, USA
| | - Tien Dong
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, UCLA Microbiome Center, University of California, Los Angeles, CA, USA
| | - Arpana Gupta
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, UCLA Microbiome Center, University of California, Los Angeles, CA, USA
| | - Chi T. Viet
- Department of Oral & Maxillofacial Surgery, Loma Linda University Health School of Dentistry, Loma Linda, CA, USA
| | - Marcelo Febo
- Translational Research Imaging Laboratory, Department of Psychiatry, Department of Neuroscience, College of Medicine, University of Florida Health, Gainesville, FL, USA
| | - Andre Obenaus
- Department of Pediatrics, University of California Irvine, Irvine, CA, USA
| | - Aarti Nair
- Department of Psychology, Loma Linda University, Loma Linda, CA, USA
| | - Johnny D. Figueroa
- Center for Health Disparities and Molecular Medicine and Department of Basic Sciences, Physiology Division, Department of Basic Sciences, Loma Linda University Health School of Medicine, Loma Linda, CA, USA
| |
Collapse
|
13
|
Perdaens O, van Pesch V. Should We Consider Neurodegeneration by Itself or in a Triangulation with Neuroinflammation and Demyelination? The Example of Multiple Sclerosis and Beyond. Int J Mol Sci 2024; 25:12637. [PMID: 39684351 PMCID: PMC11641818 DOI: 10.3390/ijms252312637] [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: 10/31/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Neurodegeneration is preeminent in many neurological diseases, and still a major burden we fail to manage in patient's care. Its pathogenesis is complicated, intricate, and far from being completely understood. Taking multiple sclerosis as an example, we propose that neurodegeneration is neither a cause nor a consequence by itself. Mitochondrial dysfunction, leading to energy deficiency and ion imbalance, plays a key role in neurodegeneration, and is partly caused by the oxidative stress generated by microglia and astrocytes. Nodal and paranodal disruption, with or without myelin alteration, is further involved. Myelin loss exposes the axons directly to the inflammatory and oxidative environment. Moreover, oligodendrocytes provide a singular metabolic and trophic support to axons, but do not emerge unscathed from the pathological events, by primary myelin defects and cell apoptosis or secondary to neuroinflammation or axonal damage. Hereby, trophic failure might be an overlooked contributor to neurodegeneration. Thus, a complex interplay between neuroinflammation, demyelination, and neurodegeneration, wherein each is primarily and secondarily involved, might offer a more comprehensive understanding of the pathogenesis and help establishing novel therapeutic strategies for many neurological diseases and beyond.
Collapse
Affiliation(s)
- Océane Perdaens
- Neurochemistry Group, Institute of NeuroScience, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium;
| | - Vincent van Pesch
- Neurochemistry Group, Institute of NeuroScience, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium;
- Department of Neurology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| |
Collapse
|
14
|
Porcedda C, Manca C, Carta G, Piras F, Banni S, Sogos V, Murru E. Anti-neuroinflammatory effects of conjugated linoleic acid isomers, c9,t11 and t10,c12, on activated BV-2 microglial cells. Front Cell Neurosci 2024; 18:1442786. [PMID: 39398647 PMCID: PMC11466893 DOI: 10.3389/fncel.2024.1442786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 09/09/2024] [Indexed: 10/15/2024] Open
Abstract
Conjugated linoleic acid (CLA) isomers exhibit anti-inflammatory properties within the central nervous system (CNS). This study investigated the effects of CLA isomers c9,t11 and t10,c12 on fatty acid (FA) and N-acylethanolamine (NAE) profiles and their association with pro-inflammatory molecule expression in BV-2 microglia cell line, the CNS's resident immune cells responsible for maintaining neuronal activity and immune homeostasis. BV-2 cells were treated with 25 μM of c9,t11-CLA, t10,c12-CLA, or oleic acid (OA) for 24 h, followed by lipopolysaccharide (LPS) stimulation. After treatment, the cell's FA and NAE profiles and pro-inflammatory molecule expression were analyzed. Our results demonstrated that CLA isomers mitigate LPS-induced morphological changes in BV-2 cells and reduce gene expression and protein levels of inflammatory markers. This effect was linked to an upregulation of acyl-CoA oxidase 1, a key enzyme in the anti-inflammatory peroxisomal beta-oxidation pathway that efficiently metabolizes CLA isomers. Notably, t10,c12-CLA significantly suppressed stearoyl-CoA desaturase 1, impacting monounsaturated fatty acid synthesis. The NAEs profile was remarkably altered by CLA isomers, with a significant release of the anti-neuroinflammatory mediator docosahexaenoic acid (DHA)-derived N-acylethanolamine (DHAEA). In conclusion, our findings suggest that the anti-neuroinflammatory effects of CLA isomers are due to their unique influences on FA metabolism and the modulation of bioactive FA-derived NAEs, highlighting a potential strategy for nutritional intervention in conditions characterized by neuroinflammation.
Collapse
Affiliation(s)
| | | | | | | | | | - Valeria Sogos
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | | |
Collapse
|
15
|
Emmons HA, Fordahl SC. Moderate-intensity aerobic exercise enhanced dopamine signaling in diet-induced obese female mice without preventing body weight gain. Neuroscience 2024; 555:1-10. [PMID: 39032807 PMCID: PMC11344652 DOI: 10.1016/j.neuroscience.2024.07.020] [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: 02/28/2024] [Revised: 07/08/2024] [Accepted: 07/11/2024] [Indexed: 07/23/2024]
Abstract
Obesity continues to rise in prevalence and financial burden despite strong evidence linking it to an increased risk of developing several chronic diseases. Dopamine response and receptor density are shown to decrease under conditions of obesity. However, it is unclear if this could be a potential mechanism for treatment without drugs that have a potential for abuse. Therefore, the aim of this study was to investigate whether moderate-intensity exercise could reduce body weight gain and the associated decreases in dopamine signaling observed with high-fat diet-induced adiposity. We hypothesized that exercise would attenuate body weight gain and diet-induced inflammation in high-fat (HF)-fed mice, resulting in dopamine signaling (release and reuptake rate) comparable to sedentary, low-fat (LF)-fed counterparts. This hypothesis was tested using a mouse model of diet-induced obesity (DIO) and fast-scan cyclic voltammetry to measure evoked dopamine release and reuptake rates. Although the exercise protocol employed in this study was not sufficient to prevent significant body weight gain, there was an enhancement of dopamine signaling observed in female mice fed a HF diet that underwent treadmill running. Additionally, aerobic treadmill exercise enhanced the sensitivity to amphetamine (AMPH) in this same group of exercised, HF-fed females. The estrous cycle might influence the ability of exercise to enhance dopamine signaling in females, an effect not observed in male groups. Further research into females by estrous cycle phase, in addition to determining the optimal intensity and duration of aerobic exercise, are logical next steps.
Collapse
Affiliation(s)
| | - Steve C Fordahl
- UNC Greensboro, Department of Nutrition, Greensboro NC, USA.
| |
Collapse
|
16
|
Momen YS, Mishra J, Kumar N. Brain-Gut and Microbiota-Gut-Brain Communication in Type-2 Diabetes Linked Alzheimer's Disease. Nutrients 2024; 16:2558. [PMID: 39125436 PMCID: PMC11313915 DOI: 10.3390/nu16152558] [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: 05/27/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 08/12/2024] Open
Abstract
The gastrointestinal (GI) tract, home to the largest microbial population in the human body, plays a crucial role in overall health through various mechanisms. Recent advancements in research have revealed the potential implications of gut-brain and vice-versa communication mediated by gut-microbiota and their microbial products in various diseases including type-2 diabetes and Alzheimer's disease (AD). AD is the most common type of dementia where most of cases are sporadic with no clearly identified cause. However, multiple factors are implicated in the progression of sporadic AD which can be classified as non-modifiable (e.g., genetic) and modifiable (e.g. Type-2 diabetes, diet etc.). Present review focusses on key players particularly the modifiable factors such as Type-2 diabetes (T2D) and diet and their implications in microbiota-gut-brain (MGB) and brain-gut (BG) communication and cognitive functions of healthy brain and their dysfunction in Alzheimer's Disease. Special emphasis has been given on elucidation of the mechanistic aspects of the impact of diet on gut-microbiota and the implications of some of the gut-microbial products in T2D and AD pathology. For example, mechanistically, HFD induces gut dysbiosis with driven metabolites that in turn cause loss of integrity of intestinal barrier with concomitant colonic and systemic chronic low-grade inflammation, associated with obesity and T2D. HFD-induced obesity and T2D parallel neuroinflammation, deposition of Amyloid β (Aβ), and ultimately cognitive impairment. The review also provides a new perspective of the impact of diet on brain-gut and microbiota-gut-brain communication in terms of transcription factors as a commonly spoken language that may facilitates the interaction between gut and brain of obese diabetic patients who are at a higher risk of developing cognitive impairment and AD. Other commonality such as tyrosine kinase expression and functions maintaining intestinal integrity on one hand and the phagocytic clarence by migratory microglial functions in brain are also discussed. Lastly, the characterization of the key players future research that might shed lights on novel potential pharmacological target to impede AD progression are also discussed.
Collapse
Affiliation(s)
| | | | - Narendra Kumar
- Department of Pharmaceutical Sciences, ILR College of Pharmacy, Texas A&M Health Science Center, Kingsville, TX 78363, USA
| |
Collapse
|
17
|
Karnik R, Vohra A, Khatri M, Dalvi N, Vyas HS, Shah H, Gohil S, Kanojiya S, Devkar R. Diet/photoperiod mediated changes in cerebellar clock genes causes locomotor shifts and imperative changes in BDNF-TrkB pathway. Neurosci Lett 2024; 835:137843. [PMID: 38821201 DOI: 10.1016/j.neulet.2024.137843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
Neuropsychological studies report anxiety and depression like symptoms in patients suffering from lifestyle disorder but its impact on locomotor function lacks clarity. Our study investigates locomotor deficits resulting due to perturbations in cerebellum of high fat diet (HFD), chronodisruption (CD) or a combination (HCD) model of lifestyle disorder. Significant downregulation in levels of cerebellar clock genes (Bmal-1, Clock, Per 1 and Per 2) and Bdnf-Trkb pathway genes (Bdnf, TrkB and Syn1 levels) were recorded. Further, locomotor deficits were observed in all the three experimental groups as evidenced by actimeter test, pole test and wire hanging test. Nuclear pyknosis of Purkinje cells, their derangement and inflammation were the hallmark of cerebellar tissue of all the three experimental groups. Taken together, this study generates important links between cerebellar clock oscillations, locomotor function and Bdnf-TrkB signaling.
Collapse
Affiliation(s)
- Rhydham Karnik
- Division of Chronobiology and Metabolic Endocrinology, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, India; Dr. Vikram Sarabhai Institute of Cell & Molecular Biology, Faculty of Science, The Maharaja Sayajirao University of Baroda, India
| | - Aliasgar Vohra
- Division of Chronobiology and Metabolic Endocrinology, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, India; Department of Neurology, Washington University in St. Louis, Saint Louis, MO, 63110, USA
| | - Mahamadtezib Khatri
- Division of Chronobiology and Metabolic Endocrinology, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, India
| | - Nilay Dalvi
- Division of Chronobiology and Metabolic Endocrinology, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, India
| | - Hitarthi S Vyas
- Division of Chronobiology and Metabolic Endocrinology, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, India; Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Helly Shah
- Division of Chronobiology and Metabolic Endocrinology, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, India; Dr. Vikram Sarabhai Institute of Cell & Molecular Biology, Faculty of Science, The Maharaja Sayajirao University of Baroda, India
| | - Sujitsinh Gohil
- Division of Chronobiology and Metabolic Endocrinology, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, India
| | - Smit Kanojiya
- Division of Chronobiology and Metabolic Endocrinology, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, India; Dr. Vikram Sarabhai Institute of Cell & Molecular Biology, Faculty of Science, The Maharaja Sayajirao University of Baroda, India
| | - Ranjitsinh Devkar
- Division of Chronobiology and Metabolic Endocrinology, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, India; Dr. Vikram Sarabhai Institute of Cell & Molecular Biology, Faculty of Science, The Maharaja Sayajirao University of Baroda, India.
| |
Collapse
|
18
|
Le Thuc O, García-Cáceres C. Obesity-induced inflammation: connecting the periphery to the brain. Nat Metab 2024; 6:1237-1252. [PMID: 38997442 DOI: 10.1038/s42255-024-01079-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 06/11/2024] [Indexed: 07/14/2024]
Abstract
Obesity is often associated with a chronic, low-grade inflammatory state affecting the entire body. This sustained inflammatory state disrupts the coordinated communication between the periphery and the brain, which has a crucial role in maintaining homeostasis through humoural, nutrient-mediated, immune and nervous signalling pathways. The inflammatory changes induced by obesity specifically affect communication interfaces, including the blood-brain barrier, glymphatic system and meninges. Consequently, brain areas near the third ventricle, including the hypothalamus and other cognition-relevant regions, become susceptible to impairments, resulting in energy homeostasis dysregulation and an elevated risk of cognitive impairments such as Alzheimer's disease and dementia. This Review explores the intricate communication between the brain and the periphery, highlighting the effect of obesity-induced inflammation on brain function.
Collapse
Affiliation(s)
- Ophélia Le Thuc
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Cristina García-Cáceres
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
19
|
Chinnathambi S, Desale SE. The crosstalk between extracellular matrix proteins and Tau. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 141:447-466. [PMID: 38960482 DOI: 10.1016/bs.apcsb.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
Alzheimer's disease is progressive neurodegenerative disease characterize by the presence of extracellular accumulation of amyloid-β plaques and intracellular deposits of neurofibrillary tangles of Tau. Apart from axonal depositions pathological aggregated Tau protein is known to secrete into extracellular spaces and propagate through seeding mechanism. Microglia, the immune cells of the brain display modest ability to internalize the extracellular Tau and degrade it through endolysosomal pathway. However, the excessive burden of pathoproteins weakens the phagocytic ability of microglia. Extracellular supplementation of omega-3 fatty acids (n-3) may regulate the phagocytosis of microglia as they mediate the anti-inflammatory polarization of microglia through membrane lipid compositions changes. The internalization of extracellular Tau in the microglia is regulated by cortical membrane-associated actin remodeling driven by interplay of actin-binding proteins. On the other hand, Tau display capability bind and interact with various actin-binding protein owing to the presence of proline-rich domain in the structure and regulate their activation. In this study, we hypothesize that internalization of Tau in the presence of omega-3 fatty acids would propagate the Tau-mediated activation of actin-binding proteins as well as extracellular matrix and in turn modulate cortical actin remodeling for phagocytosis.
Collapse
Affiliation(s)
- Subashchandrabose Chinnathambi
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Institute of National Importance, Bangalore, Karnataka, India
| | - Smita Eknath Desale
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Institute of National Importance, Bangalore, Karnataka, India
| |
Collapse
|
20
|
Poxleitner M, Hoffmann SHL, Berezhnoy G, Ionescu TM, Gonzalez-Menendez I, Maier FC, Seyfried D, Ehrlichmann W, Quintanilla-Martinez L, Schmid AM, Reischl G, Trautwein C, Maurer A, Pichler BJ, Herfert K, Beziere N. Western diet increases brain metabolism and adaptive immune responses in a mouse model of amyloidosis. J Neuroinflammation 2024; 21:129. [PMID: 38745337 PMCID: PMC11092112 DOI: 10.1186/s12974-024-03080-0] [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: 01/18/2024] [Accepted: 03/29/2024] [Indexed: 05/16/2024] Open
Abstract
Diet-induced increase in body weight is a growing health concern worldwide. Often accompanied by a low-grade metabolic inflammation that changes systemic functions, diet-induced alterations may contribute to neurodegenerative disorder progression as well. This study aims to non-invasively investigate diet-induced metabolic and inflammatory effects in the brain of an APPPS1 mouse model of Alzheimer's disease. [18F]FDG, [18F]FTHA, and [18F]GE-180 were used for in vivo PET imaging in wild-type and APPPS1 mice. Ex vivo flow cytometry and histology in brains complemented the in vivo findings. 1H- magnetic resonance spectroscopy in the liver, plasma metabolomics and flow cytometry of the white adipose tissue were used to confirm metaflammatory condition in the periphery. We found disrupted glucose and fatty acid metabolism after Western diet consumption, with only small regional changes in glial-dependent neuroinflammation in the brains of APPPS1 mice. Further ex vivo investigations revealed cytotoxic T cell involvement in the brains of Western diet-fed mice and a disrupted plasma metabolome. 1H-magentic resonance spectroscopy and immunological results revealed diet-dependent inflammatory-like misbalance in livers and fatty tissue. Our multimodal imaging study highlights the role of the brain-liver-fat axis and the adaptive immune system in the disruption of brain homeostasis in amyloid models of Alzheimer's disease.
Collapse
Affiliation(s)
- Marilena Poxleitner
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Sabrina H L Hoffmann
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Georgy Berezhnoy
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Tudor M Ionescu
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Irene Gonzalez-Menendez
- Department of Pathology and Neuropathology, University Hospital Tübingen, Eberhard Karls University, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Florian C Maier
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Dominik Seyfried
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Walter Ehrlichmann
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Leticia Quintanilla-Martinez
- Department of Pathology and Neuropathology, University Hospital Tübingen, Eberhard Karls University, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Andreas M Schmid
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Gerald Reischl
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Christoph Trautwein
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Andreas Maurer
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Bernd J Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Kristina Herfert
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany.
| | - Nicolas Beziere
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany.
- Cluster of Excellence CMFI (EXC 2124) "Controlling Microbes to Fight Infections", Eberhard Karls University, Tübingen, Germany.
| |
Collapse
|
21
|
Nicholas DA, Mbongue JC, Garcia-Pérez D, Sorensen D, Ferguson Bennit H, De Leon M, Langridge WHR. Exploring the Interplay between Fatty Acids, Inflammation, and Type 2 Diabetes. IMMUNO 2024; 4:91-107. [PMID: 39606781 PMCID: PMC11600342 DOI: 10.3390/immuno4010006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024] Open
Abstract
Around 285 million people worldwide currently have type 2 diabetes and it is projected that this number will be surpassed by 2030. Therefore, it is of the utmost importance to enhance our comprehension of the disease's development. The regulation of diet, obesity, and inflammation in type 2 diabetes is believed to play a crucial role in enhancing insulin sensitivity and reducing the risk of onset diabetes. Obesity leads to an increase in visceral adipose tissue, which is a prominent site of inflammation in type 2 diabetes. Dyslipidemia, on the other hand, plays a significant role in attracting activated immune cells such as macrophages, dendritic cells, T cells, NK cells, and B cells to visceral adipose tissue. These immune cells are a primary source of pro-inflammatory cytokines that are believed to promote insulin resistance. This review delves into the influence of elevated dietary free saturated fatty acids and examines the cellular and molecular factors associated with insulin resistance in the initiation of inflammation induced by obesity. Furthermore, it explores novel concepts related to diet-induced inflammation and its relationship with type 2 diabetes.
Collapse
Affiliation(s)
- Dequina A. Nicholas
- School of Biological Sciences, University of California Irvine, Irvine, CA 92697, USA
| | - Jacques C. Mbongue
- Department of Biological Sciences, School of Arts and Sciences, Oakwood University, Huntsville, AL 35896, USA
| | - Darysbel Garcia-Pérez
- Center for Health Disparities and Molecular Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 11085, USA
- Division of Molecular Genetics and Microbiology, School of Medicine Alumni Hall, Loma Linda University, Rm 102, 11021 Campus Street, Loma Linda, CA 92350, USA
| | - Dane Sorensen
- Center for Perinatal Biology, Division of Physiology, Loma Linda School of Medicine, Rm A572, 11234 Anderson Street, Loma Linda, CA 92350, USA
| | - Heather Ferguson Bennit
- Center for Health Disparities and Molecular Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 11085, USA
| | - Marino De Leon
- Center for Health Disparities and Molecular Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 11085, USA
| | - William H. R. Langridge
- Center for Health Disparities and Molecular Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 11085, USA
| |
Collapse
|
22
|
Sanchez C, Colson C, Gautier N, Noser P, Salvi J, Villet M, Fleuriot L, Peltier C, Schlich P, Brau F, Sharif A, Altintas A, Amri EZ, Nahon JL, Blondeau N, Benani A, Barrès R, Rovère C. Dietary fatty acid composition drives neuroinflammation and impaired behavior in obesity. Brain Behav Immun 2024; 117:330-346. [PMID: 38309640 DOI: 10.1016/j.bbi.2024.01.216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 01/17/2024] [Accepted: 01/20/2024] [Indexed: 02/05/2024] Open
Abstract
Nutrient composition in obesogenic diets may influence the severity of disorders associated with obesity such as insulin-resistance and chronic inflammation. Here we hypothesized that obesogenic diets rich in fat and varying in fatty acid composition, particularly in omega 6 (ω6) to omega 3 (ω3) ratio, have various effects on energy metabolism, neuroinflammation and behavior. Mice were fed either a control diet or a high fat diet (HFD) containing either low (LO), medium (ME) or high (HI) ω6/ω3 ratio. Mice from the HFD-LO group consumed less calories and exhibited less body weight gain compared to other HFD groups. Both HFD-ME and HFD-HI impaired glucose metabolism while HFD-LO partly prevented insulin intolerance and was associated with normal leptin levels despite higher subcutaneous and perigonadal adiposity. Only HFD-HI increased anxiety and impaired spatial memory, together with increased inflammation in the hypothalamus and hippocampus. Our results show that impaired glucose metabolism and neuroinflammation are uncoupled, and support that diets with a high ω6/ω3 ratio are associated with neuroinflammation and the behavioral deterioration coupled with the consumption of diets rich in fat.
Collapse
Affiliation(s)
- Clara Sanchez
- Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, France
| | - Cécilia Colson
- Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, France; Université Côte d'Azur, Institut de Biologie de Valrose, CNRS, INSERM, France
| | - Nadine Gautier
- Université Côte d'Azur, Institut de Biologie de Valrose, CNRS, INSERM, France
| | - Pascal Noser
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Denmark
| | - Juliette Salvi
- Université Bourgogne Franche-Comté, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAe, France
| | - Maxime Villet
- Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, France
| | - Lucile Fleuriot
- Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, France
| | - Caroline Peltier
- Université Bourgogne Franche-Comté, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAe, France
| | - Pascal Schlich
- Université Bourgogne Franche-Comté, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAe, France
| | - Frédéric Brau
- Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, France
| | - Ariane Sharif
- Université de Lille, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neurosciences & Cognition, UMR-S 1172, Lille France
| | - Ali Altintas
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Denmark
| | - Ez-Zoubir Amri
- Université Côte d'Azur, Institut de Biologie de Valrose, CNRS, INSERM, France
| | - Jean-Louis Nahon
- Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, France
| | - Nicolas Blondeau
- Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, France
| | - Alexandre Benani
- Université Bourgogne Franche-Comté, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAe, France
| | - Romain Barrès
- Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, France; Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Denmark
| | - Carole Rovère
- Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, France.
| |
Collapse
|
23
|
Otunla AA, Shanmugarajah K, Davies AH, Shalhoub J. Lipotoxicity and immunometabolism in ischemic acute kidney injury: current perspectives and future directions. Front Pharmacol 2024; 15:1355674. [PMID: 38464721 PMCID: PMC10924325 DOI: 10.3389/fphar.2024.1355674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/12/2024] [Indexed: 03/12/2024] Open
Abstract
Dysregulated lipid metabolism is implicated in the pathophysiology of a range of kidney diseases. The specific mechanisms through which lipotoxicity contributes to acute kidney injury (AKI) remain poorly understood. Herein we review the cardinal features of lipotoxic injury in ischemic kidney injury; lipid accumulation and mitochondrial lipotoxicity. We then explore a new mechanism of lipotoxicity, what we define as "immunometabolic" lipotoxicity, and discuss the potential therapeutic implications of targeting this lipotoxicity using lipid lowering medications.
Collapse
Affiliation(s)
- Afolarin A. Otunla
- Department of Surgical Biotechnology, University College London, London, United Kingdom
| | | | - Alun H. Davies
- UK and Imperial Vascular Unit, Section of Vascular Surgery, Department of Surgery and Cancer, Imperial College London, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Joseph Shalhoub
- UK and Imperial Vascular Unit, Section of Vascular Surgery, Department of Surgery and Cancer, Imperial College London, Imperial College Healthcare NHS Trust, London, United Kingdom
| |
Collapse
|
24
|
Liu J, Zaidi A, Pike CJ. Microglia/macrophage-specific deletion of TLR-4 protects against neural effects of diet-induced obesity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.13.580189. [PMID: 38405877 PMCID: PMC10888944 DOI: 10.1101/2024.02.13.580189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Obesity is associated with numerous adverse neural effects, including reduced neurogenesis, cognitive impairment, and increased risks for developing Alzheimer's disease (AD) and vascular dementia. Obesity is also characterized by chronic, low-grade inflammation that is implicated in mediating negative consequences body-wide. Toll-like receptor 4 (TLR4) signaling from peripheral macrophages is implicated as an essential regulator of the systemic inflammatory effects of obesity. In the brain, obesity drives chronic neuroinflammation that involves microglial activation, however the contributions of microglia-derived TLR4 signaling to the consequences of obesity are poorly understood. To investigate this issue, we first generated mice that carry an inducible, microglia/macrophage-specific deletion of TLR4 that yields long-term TLR4 knockout only in brain indicating microglial specificity. Next, we analyzed the effects of microglial TLR4 deletion on systemic and neural effects of a 16-week of exposure to control versus obesogenic high-fat diets. In male mice, TLR4 deletion generally yielded limited effects on diet-induced systemic metabolic dysfunction but significantly reduced neuroinflammation and impairments in neurogenesis and cognitive performance. In female mice maintained on obesogenic diet, TLR4 deletion partially protected against weight gain, adiposity, and metabolic impairments. Compared to males, females showed milder diet-induced neural consequences, against which TLR4 deletion was protective. Collectively, these findings demonstrate a central role of microglial TLR4 signaling in mediating the neural effects of obesogenic diet and highlight sexual dimorphic responses to both diet and TLR4.
Collapse
|
25
|
Muscat SM, Butler MJ, Bettes MN, DeMarsh JW, Scaria EA, Deems NP, Barrientos RM. Post-operative cognitive dysfunction is exacerbated by high-fat diet via TLR4 and prevented by dietary DHA supplementation. Brain Behav Immun 2024; 116:385-401. [PMID: 38145855 PMCID: PMC10872288 DOI: 10.1016/j.bbi.2023.12.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/04/2023] [Accepted: 12/20/2023] [Indexed: 12/27/2023] Open
Abstract
Post-operative cognitive dysfunction (POCD) is an abrupt decline in neurocognitive function arising shortly after surgery and persisting for weeks to months, increasing the risk of dementia diagnosis. Advanced age, obesity, and comorbidities linked to high-fat diet (HFD) consumption such as diabetes and hypertension have been identified as risk factors for POCD, although underlying mechanisms remain unclear. We have previously shown that surgery alone, or 3-days of HFD can each evoke sufficient neuroinflammation to cause memory deficits in aged, but not young rats. The aim of the present study was to determine if HFD consumption before surgery would potentiate and prolong the subsequent neuroinflammatory response and memory deficits, and if so, to determine the extent to which these effects depend on activation of the innate immune receptor TLR4, which both insults are known to stimulate. Young-adult (3mo) & aged (24mo) male F344xBN F1 rats were fed standard chow or HFD for 3-days immediately before sham surgery or laparotomy. In aged rats, the combination of HFD and surgery caused persistent deficits in contextual memory and cued-fear memory, though it was determined that HFD alone was sufficient to cause the long-lasting cued-fear memory deficits. In young adult rats, HFD + surgery caused only cued-fear memory deficits. Elevated proinflammatory gene expression in the hippocampus of both young and aged rats that received HFD + surgery persisted for at least 3-weeks after surgery. In a separate experiment, rats were administered the TLR4-specific antagonist, LPS-RS, immediately before HFD onset, which ameliorated the HFD + surgery-associated neuroinflammation and memory deficits. Similarly, dietary DHA supplementation for 4 weeks prior to HFD onset blunted the neuroinflammatory response to surgery and prevented development of persistent memory deficits. These results suggest that HFD 1) increases risk of persistent POCD-associated memory impairments following surgery in male rats in 2) a TLR4-dependent manner, which 3) can be targeted by DHA supplementation to mitigate development of persistent POCD.
Collapse
Affiliation(s)
- Stephanie M Muscat
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Michael J Butler
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Menaz N Bettes
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - James W DeMarsh
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Emmanuel A Scaria
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Nicholas P Deems
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Ruth M Barrientos
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA; Department of Psychiatry & Behavioral Health, The Ohio State University, Columbus, OH, USA; Chronic Brain Injury Program, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
26
|
Yu Q, Yang Y, Xu T, Cai Y, Yang Z, Yuan F. Palmitoleic acid protects microglia from palmitate-induced neurotoxicity in vitro. PLoS One 2024; 19:e0297031. [PMID: 38241239 PMCID: PMC10798504 DOI: 10.1371/journal.pone.0297031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 12/26/2023] [Indexed: 01/21/2024] Open
Abstract
Although palmitoleic acid (POA) is a lipokine with beneficial effects on obesity and is produced as a byproduct from the manufacture of prescription omega-3 fatty acids, its role in nervous system inflammation is still unknown. This study aims to examine the mechanisms and protective effects of POA against palmitic acid (PA)-induced microglial death. PA-induced microglial death was used as a model for POA intervention. Various inhibitors were employed to suppress potential routes of PA entry into the cell. Immunofluorescence staining and Western blotting were conducted to elucidate the protective pathways involved. The results suggest POA has the potential to eliminate PA-induced lactate dehydrogenase (LDH) release, which decreases the overall number of propidium iodide (PI)-positive cells compared with control. Moreover, POA has the potential to significantly increase lipid droplets (LDs) in the cytoplasm, without causing any lysosomal damage. POA inhibited both canonical and non-canonical gasdermin D (GSDMD)-mediated pyroptosis and gasdermin E (GSDME)-mediated pyroptosis, which PA typically induces. Additionally, POA inhibited the endoplasmic reticulum (ER) stress and apoptosis-related proteins induced by PA. Based on the findings, POA can exert a protective effect on microglial death induced by PA via pathways related to pyroptosis, apoptosis, ER stress, and LDs.
Collapse
Affiliation(s)
- Qingting Yu
- Department of Pharmacy, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | - Yanzhuo Yang
- Department of Pharmacy, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | - Ting Xu
- Department of Pharmacy, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | | | - Zuisu Yang
- Department of Pharmacy, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | - Falei Yuan
- Department of Pharmacy, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| |
Collapse
|
27
|
Delpech JC, Valdearcos M, Nadjar A. Stress and Microglia: A Double-edged Relationship. ADVANCES IN NEUROBIOLOGY 2024; 37:333-342. [PMID: 39207700 DOI: 10.1007/978-3-031-55529-9_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglia are highly dynamic cells and acquire different activation states to modulate their multiple functions, which are tightly regulated by the central nervous system microenvironment in which they reside. In response to stress, that is to the appearance of non-physiological signals in their vicinity, microglia will adapt their function in order to promote a return to brain homeostasis. However, when these stress signals are chronically present, microglial response may not be adapted and lead to the establishment of a pathological state. The aim of this book chapter is to examine the substantial literature around the ability of acute and chronic stressors to affect microglial structure and function, with a special focus on psychosocial and nutritional stresses. We also discuss the molecular mechanisms known to date that explain the link between exposure to stressors and microglial activation.
Collapse
Affiliation(s)
| | - Martin Valdearcos
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Agnès Nadjar
- Neurocentre Magendie, U1215, INSERM-Université de Bordeaux, Bordeaux, France.
- Institut Universitaire de France (IUF), Paris, France.
| |
Collapse
|
28
|
Yao Z, Zhu Z, Chen X, Li X. Ameliorative Effect of Raspberry Ketone on Hypothalamic Inflammation in High Fat Diet-Induced Obese Mice. J Nutr Sci Vitaminol (Tokyo) 2024; 70:496-502. [PMID: 39756970 DOI: 10.3177/jnsv.70.496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
This study aimed to investigate the regulatory effects of raspberry ketone on hypothalamic inflammation and its mechanism. Mouse microglia cells (BV2 cells) were cultured in vitro with palmitic acid (100 μM) to induce inflammation model and then incubated with raspberry ketone (5, 20, 50 μM) alone or raspberry ketone (50 μM) and the specific inhibitor of uncoupling protein 2 (UCP2), genipin (10 μM), to test the role of UCP2 in raspberry ketone regulatory of inflammation. Meanwhile, C57BL/6J mice were fed a high-fat diet containing raspberry ketone (0.2%, wt/wt) for 16 wk or 7 d to observe the effects of raspberry ketone on the body weights and hypothalamic inflammation of mice. The expression levels of inflammatory factors, including interleukin-6 (IL-6), interleukin-1beta (IL-1β) and tumour necrosis factor alpha (TNF-α), were detected using RT-qPCR, Elisa, and Western blotting, respectively. At the cellular level, raspberry ketone reduced the content of inflammatory factors in BV2 cells and in the cell culture medium. Genipin inhibited the anti-inflammatory effect of raspberry ketone on BV2 cells. At the animal level, after 16 wk of feeding, raspberry ketone-containing diets significantly reduced the body weight of mice, but had no significant effect on the mRNA expression level of hypothalamic inflammatory factors. On the other hand, 7 d of raspberry ketone gavage significantly reduced mRNA and protein expression of hypothalamic inflammatory factors. The results of this study suggest that raspberry ketone could regulate high-fat diet-induced obesity in mice, and the specific mechanism may be to inhibit hypothalamic inflammation in mice by regulating UCP2 gene expression.
Collapse
Affiliation(s)
- Zhao Yao
- College of Health Industry, Sichuan Tourism University
| | - Zhenhua Zhu
- Key Laboratory of Sichuan Cuisine Artificial Intelligence, Sichuan Tourism University
- Cuisine Science Key Laboratory of Sichuan Province, Sichuan Tourism University
| | - Xingyou Chen
- Key Laboratory of Sichuan Cuisine Artificial Intelligence, Sichuan Tourism University
| | - Xiaoping Li
- Key Laboratory of Sichuan Cuisine Artificial Intelligence, Sichuan Tourism University
- Cuisine Science Key Laboratory of Sichuan Province, Sichuan Tourism University
- College of Culinary, Sichuan Tourism University
- State Key Laboratory of Food Science and Resource, Nanchang University
| |
Collapse
|
29
|
Garcia-Segura ME, Pluchino S, Peruzzotti-Jametti L. Metabolic Control of Microglia. ADVANCES IN NEUROBIOLOGY 2024; 37:607-622. [PMID: 39207716 DOI: 10.1007/978-3-031-55529-9_34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglia, immune sentinels of the central nervous system (CNS), play a critical role in maintaining its health and integrity. This chapter delves into the concept of immunometabolism, exploring how microglial metabolism shapes their diverse immune functions. It examines the impact of cell metabolism on microglia during various CNS states, including homeostasis, development, aging, and inflammation. Particularly in CNS inflammation, the chapter discusses how metabolic rewiring in microglia can initiate, resolve, or perpetuate inflammatory responses. The potential of targeting microglial metabolism as a therapeutic strategy for chronic CNS disorders with prominent innate immune cell activation is also explored.
Collapse
Affiliation(s)
- Monica Emili Garcia-Segura
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Stefano Pluchino
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Luca Peruzzotti-Jametti
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK.
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| |
Collapse
|
30
|
Gong L, Liang J, Xie L, Zhang Z, Mei Z, Zhang W. Metabolic Reprogramming in Gliocyte Post-cerebral Ischemia/ Reperfusion: From Pathophysiology to Therapeutic Potential. Curr Neuropharmacol 2024; 22:1672-1696. [PMID: 38362904 PMCID: PMC11284719 DOI: 10.2174/1570159x22666240131121032] [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: 08/21/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 02/17/2024] Open
Abstract
Ischemic stroke is a leading cause of disability and death worldwide. However, the clinical efficacy of recanalization therapy as a preferred option is significantly hindered by reperfusion injury. The transformation between different phenotypes of gliocytes is closely associated with cerebral ischemia/ reperfusion injury (CI/RI). Moreover, gliocyte polarization induces metabolic reprogramming, which refers to the shift in gliocyte phenotype and the overall transformation of the metabolic network to compensate for energy demand and building block requirements during CI/RI caused by hypoxia, energy deficiency, and oxidative stress. Within microglia, the pro-inflammatory phenotype exhibits upregulated glycolysis, pentose phosphate pathway, fatty acid synthesis, and glutamine synthesis, whereas the anti-inflammatory phenotype demonstrates enhanced mitochondrial oxidative phosphorylation and fatty acid oxidation. Reactive astrocytes display increased glycolysis but impaired glycogenolysis and reduced glutamate uptake after CI/RI. There is mounting evidence suggesting that manipulation of energy metabolism homeostasis can induce microglial cells and astrocytes to switch from neurotoxic to neuroprotective phenotypes. A comprehensive understanding of underlying mechanisms and manipulation strategies targeting metabolic pathways could potentially enable gliocytes to be reprogrammed toward beneficial functions while opening new therapeutic avenues for CI/RI treatment. This review provides an overview of current insights into metabolic reprogramming mechanisms in microglia and astrocytes within the pathophysiological context of CI/RI, along with potential pharmacological targets. Herein, we emphasize the potential of metabolic reprogramming of gliocytes as a therapeutic target for CI/RI and aim to offer a novel perspective in the treatment of CI/RI.
Collapse
Affiliation(s)
- Lipeng Gong
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Junjie Liang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Letian Xie
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Zhanwei Zhang
- Department of Neurosurgery, First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410007, China
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, College of Medicine and Health Sciences, China Three Gorges University, Yichang, Hubei 443002, China
| | - Wenli Zhang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| |
Collapse
|
31
|
Seliga AK, Zabłocki K, Bandorowicz-Pikuła J. Palmitate Stimulates Expression of the von Willebrand Factor and Modulates Toll-like Receptors Level and Activity in Human Umbilical Vein Endothelial Cells (HUVECs). Int J Mol Sci 2023; 25:254. [PMID: 38203423 PMCID: PMC10779284 DOI: 10.3390/ijms25010254] [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: 11/16/2023] [Revised: 12/12/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
An increased concentration of palmitate in circulation is one of the most harmful factors in obesity. The von Willebrand factor (vWF), a protein involved in haemostasis, is produced and secreted by the vascular endothelium. An increased level of vWF in obese patients is associated with thrombosis and cardiovascular disease. The aim of this study was to investigate a palmitate effect on vWF in endothelial cells and understand the mechanisms of palmitate-activated signalling. Human umbilical vein endothelial cells (HUVECs) incubated in the presence of palmitate, exhibited an increased VWF gene expression, vWF protein maturation, and stimulated vWF secretion. Cardamonin, a Nuclear Factor kappa B (NF-κB) inhibitor, abolished the palmitate effect on VWF expression. The inhibition of Toll-like receptor (TLR) 2 with C29 resulted in the TLR4 overactivation in palmitate-treated cells. Palmitate, in the presence of TLR4 inhibitor TAK-242, leads to a higher expression of TLR6, CD36, and TIRAP. The silencing of TLR4 resulted in an increase in TLR2 level and vice versa. The obtained results indicate a potential mechanism of obesity-induced thrombotic complication caused by fatty acid activation of NF-κB signalling and vWF upregulation and help to identify various compensatory mechanisms related to TLR4 signal transduction.
Collapse
Affiliation(s)
| | | | - Joanna Bandorowicz-Pikuła
- Laboratory of Cellular Metabolism, Nencki Institute of Experimental Biology PAS, 3 Pasteur Str., 02-093 Warsaw, Poland; (A.K.S.); (K.Z.)
| |
Collapse
|
32
|
Neto A, Fernandes A, Barateiro A. The complex relationship between obesity and neurodegenerative diseases: an updated review. Front Cell Neurosci 2023; 17:1294420. [PMID: 38026693 PMCID: PMC10665538 DOI: 10.3389/fncel.2023.1294420] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Obesity is a global epidemic, affecting roughly 30% of the world's population and predicted to rise. This disease results from genetic, behavioral, societal, and environmental factors, leading to excessive fat accumulation, due to insufficient energy expenditure. The adipose tissue, once seen as a simple storage depot, is now recognized as a complex organ with various functions, including hormone regulation and modulation of metabolism, inflammation, and homeostasis. Obesity is associated with a low-grade inflammatory state and has been linked to neurodegenerative diseases like multiple sclerosis (MS), Alzheimer's (AD), and Parkinson's (PD). Mechanistically, reduced adipose expandability leads to hypertrophic adipocytes, triggering inflammation, insulin and leptin resistance, blood-brain barrier disruption, altered brain metabolism, neuronal inflammation, brain atrophy, and cognitive decline. Obesity impacts neurodegenerative disorders through shared underlying mechanisms, underscoring its potential as a modifiable risk factor for these diseases. Nevertheless, further research is needed to fully grasp the intricate connections between obesity and neurodegeneration. Collaborative efforts in this field hold promise for innovative strategies to address this complex relationship and develop effective prevention and treatment methods, which also includes specific diets and physical activities, ultimately improving quality of life and health.
Collapse
Affiliation(s)
- Alexandre Neto
- Central Nervous System, Blood and Peripheral Inflammation, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Adelaide Fernandes
- Central Nervous System, Blood and Peripheral Inflammation, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Andreia Barateiro
- Central Nervous System, Blood and Peripheral Inflammation, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
33
|
Lepiarz-Raba I, Gbadamosi I, Florea R, Paolicelli RC, Jawaid A. Metabolic regulation of microglial phagocytosis: Implications for Alzheimer's disease therapeutics. Transl Neurodegener 2023; 12:48. [PMID: 37908010 PMCID: PMC10617244 DOI: 10.1186/s40035-023-00382-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 10/23/2023] [Indexed: 11/02/2023] Open
Abstract
Microglia, the resident immune cells of the brain, are increasingly implicated in the regulation of brain health and disease. Microglia perform multiple functions in the central nervous system, including surveillance, phagocytosis and release of a variety of soluble factors. Importantly, a majority of their functions are closely related to changes in their metabolism. This natural inter-dependency between core microglial properties and metabolism offers a unique opportunity to modulate microglial activities via nutritional or metabolic interventions. In this review, we examine the existing scientific literature to synthesize the hypothesis that microglial phagocytosis of amyloid beta (Aβ) aggregates in Alzheimer's disease (AD) can be selectively enhanced via metabolic interventions. We first review the basics of microglial metabolism and the effects of common metabolites, such as glucose, lipids, ketone bodies, glutamine, pyruvate and lactate, on microglial inflammatory and phagocytic properties. Next, we examine the evidence for dysregulation of microglial metabolism in AD. This is followed by a review of in vivo studies on metabolic manipulation of microglial functions to ascertain their therapeutic potential in AD. Finally, we discuss the effects of metabolic factors on microglial phagocytosis of healthy synapses, a pathological process that also contributes to the progression of AD. We conclude by enlisting the current challenges that need to be addressed before strategies to harness microglial phagocytosis to clear pathological protein deposits in AD and other neurodegenerative disorders can be widely adopted.
Collapse
Affiliation(s)
- Izabela Lepiarz-Raba
- Laboratory for Translational Research in Neuropsychiatric Disorders (TREND), BRAINCITY: Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Warsaw, Poland.
| | - Ismail Gbadamosi
- Laboratory for Translational Research in Neuropsychiatric Disorders (TREND), BRAINCITY: Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Roberta Florea
- Swiss Federal Institute of Technology (ETH), Zurich, Switzerland
| | | | - Ali Jawaid
- Laboratory for Translational Research in Neuropsychiatric Disorders (TREND), BRAINCITY: Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Warsaw, Poland.
| |
Collapse
|
34
|
Miao J, Chen L, Pan X, Li L, Zhao B, Lan J. Microglial Metabolic Reprogramming: Emerging Insights and Therapeutic Strategies in Neurodegenerative Diseases. Cell Mol Neurobiol 2023; 43:3191-3210. [PMID: 37341833 PMCID: PMC11410021 DOI: 10.1007/s10571-023-01376-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 06/14/2023] [Indexed: 06/22/2023]
Abstract
Microglia, the resident immune cells of the central nervous system, play a critical role in maintaining brain homeostasis. However, in neurodegenerative conditions, microglial cells undergo metabolic reprogramming in response to pathological stimuli, including Aβ plaques, Tau tangles, and α-synuclein aggregates. This metabolic shift is characterized by a transition from oxidative phosphorylation (OXPHOS) to glycolysis, increased glucose uptake, enhanced production of lactate, lipids, and succinate, and upregulation of glycolytic enzymes. These metabolic adaptations result in altered microglial functions, such as amplified inflammatory responses and diminished phagocytic capacity, which exacerbate neurodegeneration. This review highlights recent advances in understanding the molecular mechanisms underlying microglial metabolic reprogramming in neurodegenerative diseases and discusses potential therapeutic strategies targeting microglial metabolism to mitigate neuroinflammation and promote brain health. Microglial Metabolic Reprogramming in Neurodegenerative Diseases This graphical abstract illustrates the metabolic shift in microglial cells in response to pathological stimuli and highlights potential therapeutic strategies targeting microglial metabolism for improved brain health.
Collapse
Affiliation(s)
- Jifei Miao
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Shenzhen, China
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Lihua Chen
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Xiaojin Pan
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Liqing Li
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Beibei Zhao
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Shenzhen, China.
| | - Jiao Lan
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Shenzhen, China.
| |
Collapse
|
35
|
Emmons H, Wallace C, Fordahl S. Interleukin-6 and tumor necrosis factor-α attenuate dopamine release in mice fed a high-fat diet, but not medium or low-fat diets. Nutr Neurosci 2023; 26:864-874. [PMID: 35900193 PMCID: PMC9883593 DOI: 10.1080/1028415x.2022.2103613] [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] [Indexed: 01/31/2023]
Abstract
Chronic low-grade inflammation is associated with a state of diet-induced obesity that impacts systemic tissues and can cross the blood-brain barrier to act directly on the brain. The extent to which pro-inflammatory cytokines released in these conditions affect dopamine presynaptic neurotransmission has not been previously investigated. The purpose of this study was to examine how dopamine terminals are affected by pro-inflammatory cytokines, and to determine if dietary fat consumption potentiates cytokine effects on dopamine release and reuptake rate in the nucleus accumbens (NAc). Male and female C57BL/6J mice were fed high, medium, or low-fat diets (60%, 30%, or 10% total kcals from fat, respectively) for six weeks. Fast scan cyclic voltammetry (FSCV) was used to measure dopamine release and reuptake rate in the NAc core from ex vivo coronal brain slices. Electrically evoked dopamine release and the maximal rate of dopamine reuptake (Vmax) were significantly lower in mice fed the 30% and 60% high-fat diets compared to the 10% low-fat group (p < 0.05). IL-6 5 or 10 nM or TNFα 30 or 300 nM was added to artificial cerebrospinal fluid (aCSF) bathed over brain slices during FSCV. No effect on dopamine release or Vmax was observed with lower concentrations. However, 10 nM IL-6 and 300 nM TNFα significantly reduced dopamine release in the 60% fat group (p < 0.05). No effect of added cytokine was observed on Vmax. Overall, these data provide evidence that dietary fat increases neural responsiveness to cytokines, which may help inform comorbidities between diet-induced obesity and depression or other mood disorders.
Collapse
Affiliation(s)
- H.A. Emmons
- UNC Greensboro, Department of Nutrition, Greensboro NC
| | - C.W. Wallace
- UNC Greensboro, Department of Nutrition, Greensboro NC
- Wake Forest School of Medicine, Physiology and Pharmacology, Winston-Salem NC
| | - S.C. Fordahl
- UNC Greensboro, Department of Nutrition, Greensboro NC
| |
Collapse
|
36
|
Butler MJ, Mackey-Alfonso SE, Massa N, Baskin KK, Barrientos RM. Dietary fatty acids differentially impact phagocytosis, inflammatory gene expression, and mitochondrial respiration in microglial and neuronal cell models. Front Cell Neurosci 2023; 17:1227241. [PMID: 37636589 PMCID: PMC10448530 DOI: 10.3389/fncel.2023.1227241] [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: 05/31/2023] [Accepted: 07/27/2023] [Indexed: 08/29/2023] Open
Abstract
The consumption of diets high in saturated fatty acids and/or refined carbohydrates are associated with neuroinflammation, cognitive dysfunction, and neurodegenerative disease. In contrast, diets high in polyunsaturated fatty acids are associated with anti-inflammatory and neuroprotective effects. We have previously shown that high fat diet (HFD) consumption increases saturated fatty acids and decreases polyunsaturated fatty acids in the hippocampus. We have further shown that HFD elicits exaggerated neuroinflammation and reduced synaptic elements, and results in robust memory deficits in aged rats. Here, we examined the impact of palmitate, an abundant dietary saturated fat, on a variety of cellular responses in BV2 microglia and HippoE-14 neurons, and the extent to which the omega-3 fatty acid, docosahexaenoic acid (DHA), would buffer against these responses. Our data demonstrate that DHA pretreatment prevents or partially attenuates palmitate-induced alterations in proinflammatory, endoplasmic reticulum stress, and mitochondrial damage-associated gene expression in both cell types. Furthermore, we show that synaptoneurosomes isolated from aged, HFD-fed mice are engulfed by BV2 microglia at a faster rate than synaptoneurosomes isolated from aged, chow-fed mice, suggesting HFD alters signaling at synapses to hasten their engulfment by microglia. Consistent with this notion, we found modest increases in complement proteins and a decrease in CD47 protein expression on synaptoneurosomes isolated from the hippocampus of aged, HFD-fed mice. Interestingly, palmitate reduced BV2 microglial phagocytosis, but only of synaptoneurosomes isolated from chow-fed mice, an effect that was prevented by DHA pretreatment. Lastly, we measured the impact of palmitate and DHA on mitochondrial function in both microglial and neuronal cell models using the Seahorse XFe96 Analyzer. These data indicate that DHA pretreatment does not mitigate palmitate-induced reductions in mitochondrial respiration in BV2 microglia and HippoE-14 neurons, suggesting DHA may be acting downstream of mitochondrial function to exert its protective effects. Together, this study provides evidence that DHA can ameliorate the negative impact of palmitate on a variety of cellular functions in microglia- and neuron-like cells.
Collapse
Affiliation(s)
- Michael J. Butler
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, United States
| | - Sabrina E. Mackey-Alfonso
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
- Medical Scientist Training Program, The Ohio State University, Columbus, OH, United States
| | - Nashali Massa
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
| | - Kedryn K. Baskin
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Ruth M. Barrientos
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, United States
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States
- Chronic Brain Injury Program, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
37
|
Patlola SR, Donohoe G, McKernan DP. Counting the Toll of Inflammation on Schizophrenia-A Potential Role for Toll-like Receptors. Biomolecules 2023; 13:1188. [PMID: 37627253 PMCID: PMC10452856 DOI: 10.3390/biom13081188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/25/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
Toll-like receptors (TLRs) are a family of pattern recognition receptors (PRRs) that are ubiquitously expressed in the human body. They protect the brain and central nervous system from self and foreign antigens/pathogens. The immune response elicited by these receptors culminates in the release of cytokines, chemokines, and interferons causing an inflammatory response, which can be both beneficial and harmful to neurodevelopment. In addition, the detrimental effects of TLR activation have been implicated in multiple neurodegenerative diseases such as Alzheimer's, multiple sclerosis, etc. Many studies also support the theory that cytokine imbalance may be involved in schizophrenia, and a vast amount of literature showcases the deleterious effects of this imbalance on cognitive performance in the human population. In this review, we examine the current literature on TLRs, their potential role in the pathogenesis of schizophrenia, factors affecting TLR activity that contribute towards the risk of schizophrenia, and lastly, the role of TLRs and their impact on cognitive performance in schizophrenia.
Collapse
Affiliation(s)
- Saahithh Redddi Patlola
- Department of Pharmacology & Therapeutics, School of Medicine, University of Galway, H91 TK33 Galway, Ireland;
| | - Gary Donohoe
- School of Psychology, University of Galway, H91 TK33 Galway, Ireland;
| | - Declan P. McKernan
- Department of Pharmacology & Therapeutics, School of Medicine, University of Galway, H91 TK33 Galway, Ireland;
| |
Collapse
|
38
|
Salsinha AS, Socodato R, Rodrigues A, Vale-Silva R, Relvas JB, Pintado M, Rodríguez-Alcalá LM. Potential of omega-3 and conjugated fatty acids to control microglia inflammatory imbalance elicited by obesogenic nutrients. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159331. [PMID: 37172801 DOI: 10.1016/j.bbalip.2023.159331] [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: 01/05/2023] [Revised: 04/05/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023]
Abstract
High-fat diet-induced obesity detrimentally affects brain function by inducing chronic low-grade inflammation. This neuroinflammation is, at least in part, likely to be mediated by microglia, which are the main immune cell population in the brain. Microglia express a wide range of lipid-sensitive receptors and their activity can be modulated by fatty acids that cross the blood-brain barrier. Here, by combining live cell imaging and FRET technology we assessed how different fatty acids modulate microglia activity. We demonstrate that the combined action of fructose and palmitic acid induce Ikβα degradation and nuclear translocation of the p65 subunit nuclear factor kB (NF-κB) in HCM3 human microglia. Such obesogenic nutrients also lead to reactive oxygen species production and LynSrc activation (critical regulators of microglia inflammation). Importantly, short-time exposure to omega-3 (EPA and DHA), CLA and CLNA are sufficient to abolish NF-κB pathway activation, suggesting a potential neuroprotective role. Omega-3 and CLA also show an antioxidant potential by inhibiting reactive oxygen species production, and the activation of LynSrc in microglia. Furthermore, using chemical agonists (TUG-891) and antagonists (AH7614) of GPR120/FFA4, we demonstrated that omega-3, CLA and CLNA inhibition of the NF-κB pathway is mediated by this receptor, while omega-3 and CLA antioxidant potential occurs through different signaling mechanisms.
Collapse
Affiliation(s)
- A S Salsinha
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina, Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho, 1327, 4169-005 Porto, Portugal; Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, 4200-135 Porto, Portugal; Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
| | - R Socodato
- Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, 4200-135 Porto, Portugal; Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
| | - A Rodrigues
- Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, 4200-135 Porto, Portugal; Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
| | - R Vale-Silva
- Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, 4200-135 Porto, Portugal; Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal.; Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, 4050-313 Porto, Portugal
| | - J B Relvas
- Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, 4200-135 Porto, Portugal; Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
| | - M Pintado
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina, Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho, 1327, 4169-005 Porto, Portugal.
| | - L M Rodríguez-Alcalá
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina, Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho, 1327, 4169-005 Porto, Portugal.
| |
Collapse
|
39
|
Padovani-Claudio DA, Ramos CJ, Capozzi ME, Penn JS. Elucidating glial responses to products of diabetes-associated systemic dyshomeostasis. Prog Retin Eye Res 2023; 94:101151. [PMID: 37028118 PMCID: PMC10683564 DOI: 10.1016/j.preteyeres.2022.101151] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 04/08/2023]
Abstract
Diabetic retinopathy (DR) is a leading cause of blindness in working age adults. DR has non-proliferative stages, characterized in part by retinal neuroinflammation and ischemia, and proliferative stages, characterized by retinal angiogenesis. Several systemic factors, including poor glycemic control, hypertension, and hyperlipidemia, increase the risk of DR progression to vision-threatening stages. Identification of cellular or molecular targets in early DR events could allow more prompt interventions pre-empting DR progression to vision-threatening stages. Glia mediate homeostasis and repair. They contribute to immune surveillance and defense, cytokine and growth factor production and secretion, ion and neurotransmitter balance, neuroprotection, and, potentially, regeneration. Therefore, it is likely that glia orchestrate events throughout the development and progression of retinopathy. Understanding glial responses to products of diabetes-associated systemic dyshomeostasis may reveal novel insights into the pathophysiology of DR and guide the development of novel therapies for this potentially blinding condition. In this article, first, we review normal glial functions and their putative roles in the development of DR. We then describe glial transcriptome alterations in response to systemic circulating factors that are upregulated in patients with diabetes and diabetes-related comorbidities; namely glucose in hyperglycemia, angiotensin II in hypertension, and the free fatty acid palmitic acid in hyperlipidemia. Finally, we discuss potential benefits and challenges associated with studying glia as targets of DR therapeutic interventions. In vitro stimulation of glia with glucose, angiotensin II and palmitic acid suggests that: 1) astrocytes may be more responsive than other glia to these products of systemic dyshomeostasis; 2) the effects of hyperglycemia on glia are likely to be largely osmotic; 3) fatty acid accumulation may compound DR pathophysiology by promoting predominantly proinflammatory and proangiogenic transcriptional alterations of macro and microglia; and 4) cell-targeted therapies may offer safer and more effective avenues for DR treatment as they may circumvent the complication of pleiotropism in retinal cell responses. Although several molecules previously implicated in DR pathophysiology are validated in this review, some less explored molecules emerge as potential therapeutic targets. Whereas much is known regarding glial cell activation, future studies characterizing the role of glia in DR and how their activation is regulated and sustained (independently or as part of retinal cell networks) may help elucidate mechanisms of DR pathogenesis and identify novel drug targets for this blinding disease.
Collapse
Affiliation(s)
- Dolly Ann Padovani-Claudio
- Department of Ophthalmology and Visual Sciences, Vanderbilt University School of Medicine, B3321A Medical Center North, 1161 21st Avenue South, Nashville, TN, 37232-0011, USA.
| | - Carla J Ramos
- Department of Ophthalmology and Visual Sciences, Vanderbilt University School of Medicine, AA1324 Medical Center North, 1161 21st Avenue South, Nashville, TN, 37232-0011, USA.
| | - Megan E Capozzi
- Duke Molecular Physiology Institute, Duke University School of Medicine, 300 North Duke Street, Durham, NC, 27701, USA.
| | - John S Penn
- Department of Ophthalmology and Visual Sciences, Vanderbilt University School of Medicine, B3307 Medical Center North, 1161 21st Avenue South, Nashville, TN, 37232-0011, USA.
| |
Collapse
|
40
|
Hung HC, Tsai SF, Chou HW, Tsai MJ, Hsu PL, Kuo YM. Dietary fatty acids differentially affect secretion of pro-inflammatory cytokines in human THP-1 monocytes. Sci Rep 2023; 13:5511. [PMID: 37016048 PMCID: PMC10073224 DOI: 10.1038/s41598-023-32710-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 03/31/2023] [Indexed: 04/06/2023] Open
Abstract
Monocytes are a major population of circulating immune cells that play a crucial role in producing pro-inflammatory cytokines in the body. The actions of monocytes are known to be influenced by the combinations and concentrations of certain fatty acids (FAs) in blood and dietary fats. However, systemic comparisons of the effects of FAs on cytokine secretion by monocytes have not be performed. In this study, we compared how six saturated FAs (SFAs), two monounsaturated FAs (MUFAs), and seven polyunsaturated FAs (PUFAs) modulate human THP-1 monocyte secretion of TNF, IL-1β, and IL-6 in the absence or presence of lipopolysaccharide. SFAs generally stimulated resting THP-1 cells to secrete pro-inflammatory cytokines, with stearic acid being the most potent species. In contrast, MUFAs and PUFAs inhibited lipopolysaccharide-induced secretion of pro-inflammatory cytokines. Interestingly, the inhibitory potentials of MUFAs and PUFAs followed U-shaped (TNF and IL-1β) or inverted U-shaped (IL-6) dose-response curves. Among the MUFAs and PUFAs that were analyzed, docosahexaenoic acid (C22:6 n-3) exhibited the largest number of double bonds and was found to be the most potent anti-inflammatory compound. Together, our findings reveal that the chemical compositions and concentrations of dietary FAs are key factors in the intricate regulation of monocyte-mediated inflammation.
Collapse
Affiliation(s)
- Hao-Chang Hung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, 81362, Taiwan
| | - Sheng-Feng Tsai
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, 1 Ta Hsueh Road, Tainan, 70101, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Hsuan-Wen Chou
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
| | - Ming-Jun Tsai
- School of Medicine, College of Medicine, China Medical University, Taichung, 40402, Taiwan
- Department of Neurology, China Medical University Hospital, Taichung, 40447, Taiwan
- Department of Neurology, An-Nan-Hospital, China Medical University, Tainan, 709204, Taiwan
| | - Pei-Ling Hsu
- Department of Anatomy, School of Medicine, College of Medicine, Kaohsiung Medical University, 100 Shih-Chuan 1St Road, Kaohsiung, 80708, Taiwan.
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan.
| | - Yu-Min Kuo
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, 1 Ta Hsueh Road, Tainan, 70101, Taiwan.
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.
| |
Collapse
|
41
|
Muscat SM, Butler MJ, Mackey-Alfonso SE, Barrientos RM. Young adult and aged female rats are vulnerable to amygdala-dependent, but not hippocampus-dependent, memory impairment following short-term high-fat diet. Brain Res Bull 2023; 195:145-156. [PMID: 36870621 PMCID: PMC10257807 DOI: 10.1016/j.brainresbull.2023.03.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/18/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023]
Abstract
Global populations are increasingly consuming diets high in saturated fats and refined carbohydrates, and such diets have been well-associated with heightened inflammation and neurological dysfunction. Notably, older individuals are particularly vulnerable to the impact of unhealthy diet on cognition, even after a single meal, and pre-clinical rodent studies have demonstrated that short-term consumption of high-fat diet (HFD) induces marked increases in neuroinflammation and cognitive impairment. Unfortunately though, to date, most studies on the topic of nutrition and cognition, especially in aging, have been performed only in male rodents. This is especially concerning given that older females are more vulnerable to develop certain memory deficits and/or severe memory-related pathologies than males. Thus, the aim of the present study was to determine the extent to which short-term HFD consumption impacts memory function and neuroinflammation in female rats. Young adult (3 months) and aged (20-22 months) female rats were fed HFD for 3 days. Using contextual fear conditioning, we found that HFD had no effect on long-term contextual memory (hippocampus-dependent) at either age, but impaired long-term auditory-cued memory (amygdala-dependent) regardless of age. Gene expression of Il-1β was markedly dysregulated in the amygdala, but not hippocampus, of both young and aged rats after 3 days of HFD. Interestingly, modulation of IL-1 signaling via central administration of the IL-1 receptor antagonist (which we have previously demonstrated to be protective in males) had no impact on memory function following the HFD in females. Investigation of the memory-associated gene Pacap and its receptor Pac1r revealed differential effects of HFD on their expression in the hippocampus and amygdala. Specifically, HFD induced increased expression of Pacap and Pac1r in the hippocampus, whereas decreased Pacap was observed in the amygdala. Collectively, these data suggest that both young adult and aged female rats are vulnerable to amygdala-dependent (but not hippocampus-dependent) memory impairments following short-term HFD consumption, and identify potential mechanisms related to IL-1β and PACAP signaling in these differential effects. Notably, these findings are strikingly different than those previously reported in male rats using the same diet regimen and behavioral paradigms, and highlight the importance of examining potential sex differences in the context of neuroimmune-associated cognitive dysfunction.
Collapse
Affiliation(s)
- Stephanie M Muscat
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Michael J Butler
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Sabrina E Mackey-Alfonso
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Medical Scientist Training Program, The Ohio State University, Columbus, OH, USA
| | - Ruth M Barrientos
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA; Chronic Brain Injury Program, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
42
|
Vargas-Soria M, García-Alloza M, Corraliza-Gómez M. Effects of diabetes on microglial physiology: a systematic review of in vitro, preclinical and clinical studies. J Neuroinflammation 2023; 20:57. [PMID: 36869375 PMCID: PMC9983227 DOI: 10.1186/s12974-023-02740-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 02/16/2023] [Indexed: 03/05/2023] Open
Abstract
Diabetes mellitus is a heterogeneous chronic metabolic disorder characterized by the presence of hyperglycemia, commonly preceded by a prediabetic state. The excess of blood glucose can damage multiple organs, including the brain. In fact, cognitive decline and dementia are increasingly being recognized as important comorbidities of diabetes. Despite the largely consistent link between diabetes and dementia, the underlying causes of neurodegeneration in diabetic patients remain to be elucidated. A common factor for almost all neurological disorders is neuroinflammation, a complex inflammatory process in the central nervous system for the most part orchestrated by microglial cells, the main representatives of the immune system in the brain. In this context, our research question aimed to understand how diabetes affects brain and/or retinal microglia physiology. We conducted a systematic search in PubMed and Web of Science to identify research items addressing the effects of diabetes on microglial phenotypic modulation, including critical neuroinflammatory mediators and their pathways. The literature search yielded 1327 records, including 18 patents. Based on the title and abstracts, 830 papers were screened from which 250 primary research papers met the eligibility criteria (original research articles with patients or with a strict diabetes model without comorbidities, that included direct data about microglia in the brain or retina), and 17 additional research papers were included through forward and backward citations, resulting in a total of 267 primary research articles included in the scoping systematic review. We reviewed all primary publications investigating the effects of diabetes and/or its main pathophysiological traits on microglia, including in vitro studies, preclinical models of diabetes and clinical studies on diabetic patients. Although a strict classification of microglia remains elusive given their capacity to adapt to the environment and their morphological, ultrastructural and molecular dynamism, diabetes modulates microglial phenotypic states, triggering specific responses that include upregulation of activity markers (such as Iba1, CD11b, CD68, MHC-II and F4/80), morphological shift to amoeboid shape, secretion of a wide variety of cytokines and chemokines, metabolic reprogramming and generalized increase of oxidative stress. Pathways commonly activated by diabetes-related conditions include NF-κB, NLRP3 inflammasome, fractalkine/CX3CR1, MAPKs, AGEs/RAGE and Akt/mTOR. Altogether, the detailed portrait of complex interactions between diabetes and microglia physiology presented here can be regarded as an important starting point for future research focused on the microglia-metabolism interface.
Collapse
Affiliation(s)
- María Vargas-Soria
- Division of Physiology, School of Medicine, Universidad de Cadiz, Cadiz, Spain.,Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), Cadiz, Spain
| | - Mónica García-Alloza
- Division of Physiology, School of Medicine, Universidad de Cadiz, Cadiz, Spain.,Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), Cadiz, Spain
| | - Miriam Corraliza-Gómez
- Division of Physiology, School of Medicine, Universidad de Cadiz, Cadiz, Spain. .,Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), Cadiz, Spain.
| |
Collapse
|
43
|
Neto J, Jantsch J, Rodrigues F, Squizani S, Eller S, Oliveira TF, Silveira AK, Moreira JCF, Giovenardi M, Porawski M, Guedes RP. Impact of cafeteria diet and n3 supplementation on the intestinal microbiota, fatty acids levels, neuroinflammatory markers and social memory in male rats. Physiol Behav 2023; 260:114068. [PMID: 36567032 DOI: 10.1016/j.physbeh.2022.114068] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/08/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To assess the effects of omega-3 (n3) supplementation on intestinal microbiota, fatty acids profile, neuroinflammation, and social memory of cafeteria diet (CAF)-fed rats. METHODS Male Wistar rats were fed with CAF for 20 weeks. Omega-3 (500 mg/kg/day) was supplemented between the 16th and 20th week. Colon morphology, intestinal microbiota composition, short-chain fatty acids (SCFA) and lipopolysaccharide (LPS) in the plasma, fatty acids profile, TLR-4 and claudin-5 expressions in the brain, and social memory were investigated. RESULTS CAF reduced colon length, crypts' depth, and microbiota diversity, while n3 increased the Firmicutes/Bacteroidetes ratio. CAF increased SCFA plasma levels, but n3 reduced butyrate and isobutyrate in obese rats. LPS was increased in CAF-fed rats, and n3 decreased its levels. In the cerebral cortex, n3 increased caprylic, palmitic, stearic, tricosanoic, lignoceric, myristoleic, and linoleic acids. CAF increased palmitic acid and TLR-4 expression in the cerebral cortex while decreasing claudin-5 in the hippocampus. In the social memory test, CAF-fed animals showed greater social interaction with no effect of n3. CONCLUSIONS The lack of n3 effect in some of the evaluated parameters may be due to the severity of the obesity caused by CAF. However, n3 reduced LPS levels, suggesting its ability to reverse endotoxemia.
Collapse
Affiliation(s)
- João Neto
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Brazil
| | - Jeferson Jantsch
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Brazil
| | - Fernanda Rodrigues
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Brazil
| | - Samia Squizani
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Brazil
| | - Sarah Eller
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Brazil
| | - Tiago Franco Oliveira
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Brazil
| | | | - José Cláudio Fonseca Moreira
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Brazil; Departamento de Bioquímica da Universidade Federal do Rio Grande do Sul (UFRGS), Brazil
| | - Marcia Giovenardi
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Brazil; Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Brazil
| | - Marilene Porawski
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Brazil; Programa de Pós-Graduação em Medicina: Hepatologia, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Brazil
| | - Renata Padilha Guedes
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Brazil; Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Brazil.
| |
Collapse
|
44
|
Unsaturated Fatty Acids and Their Immunomodulatory Properties. BIOLOGY 2023; 12:biology12020279. [PMID: 36829556 PMCID: PMC9953405 DOI: 10.3390/biology12020279] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/07/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023]
Abstract
Oils are an essential part of the human diet and are primarily derived from plant (or sometimes fish) sources. Several of them exhibit anti-inflammatory properties. Specific diets, such as Mediterranean diet, that are high in ω-3 polyunsaturated fatty acids (PUFAs) and ω-9 monounsaturated fatty acids (MUFAs) have even been shown to exert an overall positive impact on human health. One of the most widely used supplements in the developed world is fish oil, which contains high amounts of PUFAs docosahexaenoic and eicosapentaenoic acid. This review is focused on the natural sources of various polyunsaturated and monounsaturated fatty acids in the human diet, and their role as precursor molecules in immune signaling pathways. Consideration is also given to their role in CNS immunity. Recent findings from clinical trials utilizing various fatty acids or diets high in specific fatty acids are reviewed, along with the mechanisms through which fatty acids exert their anti-inflammatory properties. An overall understanding of diversity of polyunsaturated fatty acids and their role in several molecular signaling pathways is useful in formulating diets that reduce inflammation and increase longevity.
Collapse
|
45
|
Ganji R, Paulo JA, Xi Y, Kline I, Zhu J, Clemen CS, Weihl CC, Purdy JG, Gygi SP, Raman M. The p97-UBXD8 complex regulates ER-Mitochondria contact sites by altering membrane lipid saturation and composition. Nat Commun 2023; 14:638. [PMID: 36746962 PMCID: PMC9902492 DOI: 10.1038/s41467-023-36298-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/25/2023] [Indexed: 02/08/2023] Open
Abstract
The intimate association between the endoplasmic reticulum (ER) and mitochondrial membranes at ER-Mitochondria contact sites (ERMCS) is a platform for critical cellular processes, particularly lipid synthesis. How contacts are remodeled and the impact of altered contacts on lipid metabolism remains poorly understood. We show that the p97 AAA-ATPase and its adaptor ubiquitin-X domain adaptor 8 (UBXD8) regulate ERMCS. The p97-UBXD8 complex localizes to contacts and its loss increases contacts in a manner that is dependent on p97 catalytic activity. Quantitative proteomics and lipidomics of ERMCS demonstrates alterations in proteins regulating lipid metabolism and a significant change in membrane lipid saturation upon UBXD8 deletion. Loss of p97-UBXD8 increased membrane lipid saturation via SREBP1 and the lipid desaturase SCD1. Aberrant contacts can be rescued by unsaturated fatty acids or overexpression of SCD1. We find that the SREBP1-SCD1 pathway is negatively impacted in the brains of mice with p97 mutations that cause neurodegeneration. We propose that contacts are exquisitely sensitive to alterations to membrane lipid composition and saturation.
Collapse
Affiliation(s)
- Rakesh Ganji
- Department of Developmental Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Yuecheng Xi
- Department of Immunobiology, BIO5 Institute, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Ian Kline
- Department of Immunobiology, BIO5 Institute, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Jiang Zhu
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
- Ilumina Inc., San Diego, CA, USA
| | - Christoph S Clemen
- Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
- Center for Physiology and Pathophysiology, Institute of Vegetative Physiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Conrad C Weihl
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - John G Purdy
- Department of Immunobiology, BIO5 Institute, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Steve P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Malavika Raman
- Department of Developmental Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
46
|
Marrano N, Biondi G, Borrelli A, Rella M, Zambetta T, Di Gioia L, Caporusso M, Logroscino G, Perrini S, Giorgino F, Natalicchio A. Type 2 Diabetes and Alzheimer's Disease: The Emerging Role of Cellular Lipotoxicity. Biomolecules 2023; 13:183. [PMID: 36671568 PMCID: PMC9855893 DOI: 10.3390/biom13010183] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/06/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
Type 2 diabetes (T2D) and Alzheimer's diseases (AD) represent major health issues that have reached alarming levels in the last decades. Although growing evidence demonstrates that AD is a significant comorbidity of T2D, and there is a ~1.4-2-fold increase in the risk of developing AD among T2D patients, the involvement of possible common triggers in the pathogenesis of these two diseases remains largely unknown. Of note, recent mechanistic insights suggest that lipotoxicity could represent the missing ring in the pathogenetic mechanisms linking T2D to AD. Indeed, obesity, which represents the main cause of lipotoxicity, has been recognized as a major risk factor for both pathological conditions. Lipotoxicity can lead to inflammation, insulin resistance, oxidative stress, ceramide and amyloid accumulation, endoplasmic reticulum stress, ferroptosis, and autophagy, which are shared biological events in the pathogenesis of T2D and AD. In the current review, we try to provide a critical and comprehensive view of the common molecular pathways activated by lipotoxicity in T2D and AD, attempting to summarize how these mechanisms can drive future research and open the way to new therapeutic perspectives.
Collapse
Affiliation(s)
- Nicola Marrano
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Giuseppina Biondi
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Anna Borrelli
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Martina Rella
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Tommaso Zambetta
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Ludovico Di Gioia
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Mariangela Caporusso
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Giancarlo Logroscino
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124 Bari, Italy
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari Aldo Moro at Pia Fondazione Cardinale G. Panico, 73039 Lecce, Italy
| | - Sebastio Perrini
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Francesco Giorgino
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Annalisa Natalicchio
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy
| |
Collapse
|
47
|
Marcos JL, Olivares-Barraza R, Ceballo K, Wastavino M, Ortiz V, Riquelme J, Martínez-Pinto J, Muñoz P, Cruz G, Sotomayor-Zárate R. Obesogenic Diet-Induced Neuroinflammation: A Pathological Link between Hedonic and Homeostatic Control of Food Intake. Int J Mol Sci 2023; 24:ijms24021468. [PMID: 36674982 PMCID: PMC9866213 DOI: 10.3390/ijms24021468] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/22/2022] [Accepted: 12/26/2022] [Indexed: 01/13/2023] Open
Abstract
Obesity-induced neuroinflammation is a chronic aseptic central nervous system inflammation that presents systemic characteristics associated with increased pro-inflammatory cytokines such as interleukin 1 beta (IL-1β) and interleukin 18 (IL-18) and the presence of microglia and reactive astrogliosis as well as the activation of the NLRP3 inflammasome. The obesity pandemic is associated with lifestyle changes, including an excessive intake of obesogenic foods and decreased physical activity. Brain areas such as the lateral hypothalamus (LH), lateral septum (LS), ventral tegmental area (VTA), and nucleus accumbens (NAcc) have been implicated in the homeostatic and hedonic control of feeding in experimental models of diet-induced obesity. In this context, a chronic lipid intake triggers neuroinflammation in several brain regions such as the hypothalamus, hippocampus, and amygdala. This review aims to present the background defining the significant impact of neuroinflammation and how this, when induced by an obesogenic diet, can affect feeding control, triggering metabolic and neurological alterations.
Collapse
Affiliation(s)
- José Luis Marcos
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Escuela de Ciencias Agrícolas y Veterinarias, Universidad Viña del Mar, Viña del Mar 2572007, Chile
- Programa de Doctorado en Ciencias e Ingeniería para la Salud, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Rossy Olivares-Barraza
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Programa de Doctorado en Ciencias Mención Neurociencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Karina Ceballo
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Programa de Doctorado en Ciencias Mención Neurociencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Melisa Wastavino
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Víctor Ortiz
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Julio Riquelme
- Escuela de Medicina y Centro de Neurología Traslacional (CENTRAS), Facultad de Medicina, Universidad de Valparaíso, Viña del Mar 2540064, Chile
| | - Jonathan Martínez-Pinto
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Pablo Muñoz
- Escuela de Medicina y Centro de Neurología Traslacional (CENTRAS), Facultad de Medicina, Universidad de Valparaíso, Viña del Mar 2540064, Chile
| | - Gonzalo Cruz
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Ramón Sotomayor-Zárate
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Correspondence: ; Tel.: +56-32-2508050
| |
Collapse
|
48
|
Salsinha AS, Socodato R, Relvas JB, Pintado M. The pro- and antiinflammatory activity of fatty acids. BIOACTIVE LIPIDS 2023:51-75. [DOI: 10.1016/b978-0-12-824043-4.00002-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
49
|
Salsinha AS, Rodríguez-Alcalá LM, Pimentel LL, Pintado M. Role of bioactive lipids in obesity. BIOACTIVE LIPIDS 2023:133-167. [DOI: 10.1016/b978-0-12-824043-4.00012-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
50
|
Bogie JF, Guns J, Vanherle S. Lipid metabolism in neurodegenerative diseases. CELLULAR LIPID IN HEALTH AND DISEASE 2023:389-419. [DOI: 10.1016/b978-0-323-95582-9.00008-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|