1
|
Wu J, Xu H, Wang S, Weng H, Luo Z, Ou G, Chen Y, Xu L, So KF, Deng L, Zhang L, Chen X. Regular exercise ameliorates high-fat diet-induced depressive-like behaviors by activating hippocampal neuronal autophagy and enhancing synaptic plasticity. Cell Death Dis 2024; 15:737. [PMID: 39389946 PMCID: PMC11467387 DOI: 10.1038/s41419-024-07132-4] [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: 01/15/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024]
Abstract
Exercise enhances synaptic plasticity and alleviates depression symptoms, but the mechanism through which exercise improves high-fat diet-induced depression remains unclear. In this study, 6-week-old male C57BL/6J mice were administered a high-fat diet (HFD, 60% kcal from fat) to a HFD model for 8 weeks. The RUN group also received 1 h of daily treadmill exercise in combination with the HFD. Depressive-like behaviors were evaluated by behavioral assessments for all groups. The key mediator of the effect of exercise on high-fat diet-induced depressive-like behaviors was detected by RNA-seq. The morphology and function of the neurons were evaluated via Nissl staining, Golgi staining, electron microscopy and electrophysiological experiments. The results showed that exercise attenuated high-fat diet-induced depressive-like behavior and reversed hippocampal gene expression changes. RNA-seq revealed Wnt5a, which was a key mediator of the effect of exercise on high-fat diet-induced depressive-like behaviors. Further work revealed that exercise significantly activated neuronal autophagy in the hippocampal CA1 region via the Wnt5a/CamkII signaling pathway, which enhanced synaptic plasticity to alleviate HFD-induced depressive-like behavior. However, the Wnt5a inhibitor Box5 suppressed the ameliorative effects of exercise. Therefore, this work highlights the critical role of Wnt5a, which is necessary for exercise to improve high-fat diet-induced depression.
Collapse
Affiliation(s)
- Jialin Wu
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Huachong Xu
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China.
- Key Laboratory of Central CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China.
| | - Shiqi Wang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Huandi Weng
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
- Key Laboratory of Central CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Zhihua Luo
- Key Laboratory of Central CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Guosen Ou
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Yaokang Chen
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Lu Xu
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Kwok-Fai So
- Key Laboratory of Central CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Li Deng
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China.
| | - Li Zhang
- Key Laboratory of Central CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China.
| | - Xiaoyin Chen
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China.
| |
Collapse
|
2
|
Meng X, Li D, Kan R, Xiang Y, Pan L, Guo Y, Yu P, Luo P, Zou H, Huang L, Zhu Y, Mao B, He Y, Xie L, Xu J, Liu X, Li W, Chen Y, Zhu S, Yang Y, Yu X. Inhibition of ANGPTL8 protects against diabetes-associated cognitive dysfunction by reducing synaptic loss via the PirB signaling pathway. J Neuroinflammation 2024; 21:192. [PMID: 39095838 PMCID: PMC11297729 DOI: 10.1186/s12974-024-03183-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2D) is associated with an increased risk of cognitive dysfunction. Angiopoietin-like protein 8 (ANGPTL8) is an important regulator in T2D, but the role of ANGPTL8 in diabetes-associated cognitive dysfunction remains unknown. Here, we explored the role of ANGPTL8 in diabetes-associated cognitive dysfunction through its interaction with paired immunoglobulin-like receptor B (PirB) in the central nervous system. METHODS The levels of ANGPTL8 in type 2 diabetic patients with cognitive dysfunction and control individuals were measured. Mouse models of diabetes-associated cognitive dysfunction were constructed to investigate the role of ANGPTL8 in cognitive function. The cognitive function of the mice was assessed by the Barnes Maze test and the novel object recognition test, and levels of ANGPTL8, synaptic and axonal markers, and pro-inflammatory cytokines were measured. Primary neurons and microglia were treated with recombinant ANGPTL8 protein (rA8), and subsequent changes were examined. In addition, the changes induced by ANGPTL8 were validated after blocking PirB and its downstream pathways. Finally, mice with central nervous system-specific knockout of Angptl8 and PirB-/- mice were generated, and relevant in vivo experiments were performed. RESULTS Here, we demonstrated that in the diabetic brain, ANGPTL8 was secreted by neurons into the hippocampus, resulting in neuroinflammation and impairment of synaptic plasticity. Moreover, neuron-specific Angptl8 knockout prevented diabetes-associated cognitive dysfunction and neuroinflammation. Mechanistically, ANGPTL8 acted in parallel to neurons and microglia via its receptor PirB, manifesting as downregulation of synaptic and axonal markers in neurons and upregulation of proinflammatory cytokine expression in microglia. In vivo, PirB-/- mice exhibited resistance to ANGPTL8-induced neuroinflammation and synaptic damage. CONCLUSION Taken together, our findings reveal the role of ANGPTL8 in the pathogenesis of diabetes-associated cognitive dysfunction and identify the ANGPTL8-PirB signaling pathway as a potential target for the management of this condition.
Collapse
Affiliation(s)
- Xiaoyu Meng
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China
| | - Danpei Li
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China
| | - Ranran Kan
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China
| | - Yuxi Xiang
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China
| | - Limeng Pan
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China
| | - Yaming Guo
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China
| | - Peng Yu
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China
| | - Peiqiong Luo
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China
| | - Huajie Zou
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China
| | - Li Huang
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China
| | - Yurong Zhu
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China
| | - Beibei Mao
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China
| | - Yi He
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China
| | - Lei Xie
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China
| | - Jialu Xu
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China
| | - Xiaoyan Liu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenjun Li
- Computer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Chen
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China
| | - Suiqiang Zhu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Yang
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China.
| | - Xuefeng Yu
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China.
| |
Collapse
|
3
|
Shetty S, Duesman SJ, Patel S, Huynh P, Toh P, Shroff S, Das A, Chowhan D, Keller B, Alvarez J, Fisher-Foye R, Sebra R, Beaumont K, McAlpine CS, Rajbhandari P, Rajbhandari AK. Sex-specific role of high-fat diet and stress on behavior, energy metabolism, and the ventromedial hypothalamus. Biol Sex Differ 2024; 15:55. [PMID: 39010139 PMCID: PMC11247790 DOI: 10.1186/s13293-024-00628-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 06/11/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Scientific evidence highlights the influence of biological sex on the relationship between stress and metabolic dysfunctions. However, there is limited understanding of how diet and stress concurrently contribute to metabolic dysregulation in both males and females. Our study aimed to investigate the combined effects of high-fat diet (HFD) induced obesity and repeated stress on fear-related behaviors, metabolic, immune, and hypothalamic outcomes in male and female mice. METHODS To investigate this, we used a highly reliable rodent behavioral model that faithfully recapitulates key aspects of post-traumatic stress disorder (PTSD)-like fear. We subjected mice to footshock stressor followed by a weekly singular footshock stressor or no stressor for 14 weeks while on either an HFD or chow diet. At weeks 10 and 14 we conducted glucose tolerance and insulin sensitivity measurements. Additionally, we placed the mice in metabolic chambers to perform indirect calorimetric measurements. Finally, we collected brain and peripheral tissues for cellular analysis. RESULTS We observed that HFD-induced obesity disrupted fear memory extinction, increased glucose intolerance, and affected energy expenditure specifically in male mice. Conversely, female mice on HFD exhibited reduced respiratory exchange ratio (RER), and a significant defect in glucose tolerance only when subjected to repeated stress. Furthermore, the combination of repeated stress and HFD led to sex-specific alterations in proinflammatory markers and hematopoietic stem cells across various peripheral metabolic tissues. Single-nuclei RNA sequencing (snRNAseq) analysis of the ventromedial hypothalamus (VMH) revealed microglial activation in female mice on HFD, while male mice on HFD exhibited astrocytic activation under repeated stress. CONCLUSIONS Overall, our findings provide insights into complex interplay between repeated stress, high-fat diet regimen, and their cumulative effects on health, including their potential contribution to the development of PTSD-like stress and metabolic dysfunctions, emphasizing the need for further research to fully understand these interconnected pathways and their implications for health.
Collapse
Affiliation(s)
- Sanutha Shetty
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Samuel J Duesman
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sanil Patel
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Pacific Huynh
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Pamela Toh
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sanjana Shroff
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anika Das
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Excellence in Youth Education, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Disha Chowhan
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benjamin Keller
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Johana Alvarez
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Rachel Fisher-Foye
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Robert Sebra
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kristin Beaumont
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cameron S McAlpine
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Prashant Rajbhandari
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Disease Mechanism and Therapeutics Program, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Abha K Rajbhandari
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
4
|
Costa A, Lucarini E. Treating chronic stress and chronic pain by manipulating gut microbiota with diet: can we kill two birds with one stone? Nutr Neurosci 2024:1-24. [PMID: 38889540 DOI: 10.1080/1028415x.2024.2365021] [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: 06/20/2024]
Abstract
Background: Chronic stress and chronic pain are closely linked by the capacity to exacerbate each other, sharing common roots in the brain and in the gut. The strict intersection between these two neurological diseases makes important to have a therapeutic strategy aimed at preventing both to maintain mental health in patients. Diet is an modifiable lifestyle factor associated with gut-brain axis diseases and there is growing interest in its use as adjuvant to main therapies. Several evidence attest the impact of specific diets or nutrients on chronic stress-related disorders and pain with a good degree of certainty. A daily adequate intake of foods containing micronutrients such as amino acids, minerals and vitamins, as well as the reduction in the consumption of processed food products can have a positive impact on microbiota and gut health. Many nutrients are endowed of prebiotic, anti-inflammatory, immunomodulatory and neuroprotective potential which make them useful tools helping the management of chronic stress and pain in patients. Dietary regimes, as intermittent fasting or caloric restriction, are promising, although further studies are needed to optimize protocols according to patient's medical history, age and sex. Moreover, by supporting gut microbiota health with diet is possible to attenuate comorbidities such as obesity, gastrointestinal dysfunction and mood disorders, thus reducing healthcare costs related to chronic stress or pain.Objective: This review summarize the most recent evidence on the microbiota-mediated beneficial effects of macro- and micronutrients, dietary-related factors, specific nutritional regimens and dietary intervention on these pathological conditions.
Collapse
Affiliation(s)
- Alessia Costa
- Department of Neuroscience, Psychology, Drug Area and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Elena Lucarini
- Department of Neuroscience, Psychology, Drug Area and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
Mthembu SXH, Mazibuko-Mbeje SE, Ziqubu K, Muvhulawa N, Marcheggiani F, Cirilli I, Nkambule BB, Muller CJF, Basson AK, Tiano L, Dludla PV. Potential regulatory role of PGC-1α within the skeletal muscle during metabolic adaptations in response to high-fat diet feeding in animal models. Pflugers Arch 2024; 476:283-293. [PMID: 38044359 PMCID: PMC10847180 DOI: 10.1007/s00424-023-02890-0] [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: 03/07/2023] [Revised: 11/13/2023] [Accepted: 11/23/2023] [Indexed: 12/05/2023]
Abstract
High-fat diet (HFD) feeding in rodents has become an essential tool to critically analyze and study the pathological effects of obesity, including mitochondrial dysfunction and insulin resistance. Peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) regulates cellular energy metabolism to influence insulin sensitivity, beyond its active role in stimulating mitochondrial biogenesis to facilitate skeletal muscle adaptations in response to HFD feeding. Here, some of the major electronic databases like PubMed, Embase, and Web of Science were accessed to update and critically discuss information on the potential role of PGC-1α during metabolic adaptations within the skeletal muscle in response to HFD feeding in rodents. In fact, available evidence suggests that partial exposure to HFD feeding (potentially during the early stages of disease development) is associated with impaired metabolic adaptations within the skeletal muscle, including mitochondrial dysfunction and reduced insulin sensitivity. In terms of implicated molecular mechanisms, these negative effects are partially associated with reduced activity of PGC-1α, together with the phosphorylation of protein kinase B and altered expression of genes involving nuclear respiratory factor 1 and mitochondrial transcription factor A within the skeletal muscle. Notably, metabolic abnormalities observed with chronic exposure to HFD (likely during the late stages of disease development) may potentially occur independently of PGC-1α regulation within the muscle of rodents. Summarized evidence suggests the causal relationship between PGC-1α regulation and effective modulations of mitochondrial biogenesis and metabolic flexibility during the different stages of disease development. It further indicates that prominent interventions like caloric restriction and physical exercise may affect PGC-1α regulation during effective modulation of metabolic processes.
Collapse
Affiliation(s)
- Sinenhlanhla X H Mthembu
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, Cape Town, 7505, South Africa
- Department of Biochemistry, North-West University, Mafikeng Campus, Mmabatho, 2735, South Africa
| | - Sithandiwe E Mazibuko-Mbeje
- Department of Biochemistry, North-West University, Mafikeng Campus, Mmabatho, 2735, South Africa
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 60131, Ancona, Italy
| | - Khanyisani Ziqubu
- Department of Biochemistry, North-West University, Mafikeng Campus, Mmabatho, 2735, South Africa
| | - Ndivhuwo Muvhulawa
- Department of Biochemistry, North-West University, Mafikeng Campus, Mmabatho, 2735, South Africa
| | - Fabio Marcheggiani
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 60131, Ancona, Italy
| | - Ilenia Cirilli
- Department of Clinical Sciences, Section of Biochemistry, Polytechnic University of Marche, 60131, Ancona, Italy
| | - Bongani B Nkambule
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, 4000, South Africa
| | - Christo J F Muller
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, Cape Town, 7505, South Africa
- Centre for Cardiometabolic Research Africa (CARMA), Division of Medical Physiology, Stellenbosch University, Tygerberg, Cape Town, 7505, South Africa
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, Empangeni, 3886, South Africa
| | - Albertus K Basson
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, Empangeni, 3886, South Africa
| | - Luca Tiano
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 60131, Ancona, Italy
| | - Phiwayinkosi V Dludla
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, Cape Town, 7505, South Africa.
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, Empangeni, 3886, South Africa.
- Cochrane South Africa, South African Medical Research Council, Tygerberg, 7505, South Africa.
| |
Collapse
|
8
|
Wang H, Shen Z, Wu CS, Ji P, Noh JY, Geoffroy CG, Kim S, Threadgill D, Li J, Zhou Y, Xiao X, Zheng H, Sun Y. Neuronal ablation of GHSR mitigates diet-induced depression and memory impairment via AMPK-autophagy signaling-mediated inflammation. Front Immunol 2024; 15:1339937. [PMID: 38464534 PMCID: PMC10920242 DOI: 10.3389/fimmu.2024.1339937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 02/06/2024] [Indexed: 03/12/2024] Open
Abstract
Obesity is associated with chronic inflammation in the central nervous system (CNS), and neuroinflammation has been shown to have detrimental effects on mood and cognition. The growth hormone secretagogue receptor (GHSR), the biologically relevant receptor of the orexigenic hormone ghrelin, is primarily expressed in the brain. Our previous study showed that neuronal GHSR deletion prevents high-fat diet-induced obesity (DIO). Here, we investigated the effect of neuronal GHSR deletion on emotional and cognitive functions in DIO. The neuron-specific GHSR-deficient mice exhibited reduced depression and improved spatial memory compared to littermate controls under DIO. We further examined the cortex and hippocampus, the major regions regulating cognitive and emotional behaviors, and found that the neuronal deletion of GHSR reduced DIO-induced neuroinflammation by suppressing proinflammatory chemokines/cytokines and decreasing microglial activation. Furthermore, our data showed that neuronal GHSR deletion suppresses neuroinflammation by downregulating AMPK-autophagy signaling in neurons. In conclusion, our data reveal that neuronal GHSR inhibition protects against DIO-induced depressive-like behavior and spatial cognitive dysfunction, at least in part, through AMPK-autophagy signaling-mediated neuroinflammation.
Collapse
Affiliation(s)
- Hongying Wang
- Department of Nutrition, Texas A&M University, College Station, TX, United States
- Department of Endocrinology, Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zheng Shen
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| | - Chia-Shan Wu
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| | - Pengfei Ji
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| | - Ji Yeon Noh
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| | - Cédric G. Geoffroy
- Department of Neuroscience & Experimental Therapeutics, Texas A&M University, College Station, TX, United States
| | - Sunja Kim
- Texas A&M Institute for Genome Sciences and Society, Texas A&M University, College Station, TX, United States
| | - David Threadgill
- Department of Nutrition, Texas A&M University, College Station, TX, United States
- Texas A&M Institute for Genome Sciences and Society, Texas A&M University, College Station, TX, United States
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, TX, United States
| | - Jianrong Li
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, United States
| | - Yu Zhou
- Department of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Xiaoqiu Xiao
- Department of Endocrinology, Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hui Zheng
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, United States
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| |
Collapse
|
9
|
Salvi J, Andreoletti P, Audinat E, Balland E, Ben Fradj S, Cherkaoui-Malki M, Heurtaux T, Liénard F, Nédélec E, Rovère C, Savary S, Véjux A, Trompier D, Benani A. Microgliosis: a double-edged sword in the control of food intake. FEBS J 2024; 291:615-631. [PMID: 35880408 DOI: 10.1111/febs.16583] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 06/30/2022] [Accepted: 07/25/2022] [Indexed: 02/16/2024]
Abstract
Maintaining energy balance is essential for survival and health. This physiological function is controlled by the brain, which adapts food intake to energy needs. Indeed, the brain constantly receives a multitude of biological signals that are derived from digested foods or that originate from the gastrointestinal tract, energy stores (liver and adipose tissues) and other metabolically active organs (muscles). These signals, which include circulating nutrients, hormones and neuronal inputs from the periphery, collectively provide information on the overall energy status of the body. In the brain, several neuronal populations can specifically detect these signals. Nutrient-sensing neurons are found in discrete brain areas and are highly enriched in the hypothalamus. In turn, specialized brain circuits coordinate homeostatic responses acting mainly on appetite, peripheral metabolism, activity and arousal. Accumulating evidence shows that hypothalamic microglial cells located at the vicinity of these circuits can influence the brain control of energy balance. However, microglial cells could have opposite effects on energy balance, that is homeostatic or detrimental, and the conditions for this shift are not totally understood yet. One hypothesis relies on the extent of microglial activation, and nutritional lipids can considerably change it.
Collapse
Affiliation(s)
- Juliette Salvi
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Pierre Andreoletti
- Laboratoire Bio-PeroxIL, Université Bourgogne Franche-Comté, Dijon, France
| | - Etienne Audinat
- IGF, Université de Montpellier, CNRS, Inserm, Montpellier, France
| | - Eglantine Balland
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Notting Hill, Australia
| | - Selma Ben Fradj
- IPMC, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Valbonne, France
| | | | - Tony Heurtaux
- Luxembourg Center of Neuropathology (LCNP), Dudelange, Luxembourg
- Department of Life Sciences and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Fabienne Liénard
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Emmanuelle Nédélec
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Carole Rovère
- IPMC, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Valbonne, France
| | - Stéphane Savary
- Laboratoire Bio-PeroxIL, Université Bourgogne Franche-Comté, Dijon, France
| | - Anne Véjux
- Laboratoire Bio-PeroxIL, Université Bourgogne Franche-Comté, Dijon, France
| | - Doriane Trompier
- Laboratoire Bio-PeroxIL, Université Bourgogne Franche-Comté, Dijon, France
| | - Alexandre Benani
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| |
Collapse
|
10
|
Zhan M, Liu X, Xia X, Yang Y, Xie Y, Zhang L, Lin C, Zhu J, Ding W, Xu S. Promotion of neuroinflammation by the glymphatic system: a new insight into ethanol extracts from Alisma orientale in alleviating obesity-associated cognitive impairment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 122:155147. [PMID: 37864890 DOI: 10.1016/j.phymed.2023.155147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 09/20/2023] [Accepted: 10/12/2023] [Indexed: 10/23/2023]
Abstract
BACKGROUND Obesity is one of the critical risk factors for cognitive dysfunction. The glymphatic system (GS) plays a key role in the pathogenesis of cognitive deficits. Alisma orientale has been shown to have anti-inflammatory and antihyperlipidemic effects, whereas its effects and underlying mechanisms on obesity-associated cognitive impairment (OACI) are unclear. PURPOSE This work aims to decipher the mechanism of ethanol extracts from Alisma orientale (EEAO) in restoring cognitive impairment in HFD-induced obese mice through a GS approach. METHODS The restoration of abnormal glucose/lipid metabolism and excess adipose deposition by EEAO were assayed by biochemical analysis and visually displayed by a micro-CT scanner and Oil Red O staining. Biochemical assays and Western blotting (WB) were used to measure cerebral blood flow (CBF), free fatty acid (FFAs) levels and the structural integrity of the blood-brain barrier (BBB). Microglial activation and neuroinflammation were assessed with immunohistochemistry staining, ELISA and WB. Moreover, GS function was determined by immunofluorescence staining, fluorescence tracer imaging and WB. Finally, the neuropathological features and cognitive functions were detested with immunohistochemistry staining, immunofluorescence and Morris Water Maze. RESULTS EEAO not only alleviated body weight, cerebral lipid accumulation and serum FFAs in HFD-induced obese mice, but also increased CBF and BBB integrity. EEAO suppressed microglial activation and lipid deposition in the hippocampus and reduced the level of inflammatory cytokines including IL-6, IL-1β and TNF-α in brain tissue. Interestingly, long-term HFD-induced GS dysfunction was significantly restored after EEAO intervention, and neuropathological lesions and cognitive deficits were also markedly rescued. CONCLUSION EEAO rescued the cognitive deficits of OACI by inhibiting neuroinflammation and restoring GS dysfunction, indicating a potential remedy for OACI.
Collapse
Affiliation(s)
- Meng Zhan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiao Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiuwen Xia
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Youjun Yang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ya Xie
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Lu Zhang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Chunqiao Lin
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jiushuang Zhu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Weijun Ding
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Shijun Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
11
|
Patel M, Braun J, Lambert G, Kameneva T, Keatch C, Lambert E. Central mechanisms in sympathetic nervous dysregulation in obesity. J Neurophysiol 2023; 130:1414-1424. [PMID: 37910522 DOI: 10.1152/jn.00254.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/16/2023] [Accepted: 10/26/2023] [Indexed: 11/03/2023] Open
Abstract
Cardiovascular and metabolic complications associated with excess adiposity are linked to chronic activation of the sympathetic nervous system, resulting in a high risk of mortality among obese individuals. Obesity-related positive energy balance underlies the progression of hypertension, end-organ damage, and insulin resistance, driven by increased sympathetic tone throughout the body. It is, therefore, important to understand the central network that drives and maintains sustained activation of the sympathetic nervous system in the obese state. Experimental and clinical studies have identified structural changes and altered dynamics in both grey and white matter regions in obesity. Aberrant activation in certain brain regions has been associated with altered reward circuitry and metabolic pathways including leptin and insulin signaling along with adiposity-driven systemic and central inflammation. The impact of these pathways on the brain via overactivity of the sympathetic nervous system has gained interest in the past decade. Primarily, the brainstem, hypothalamus, amygdala, hippocampus, and cortical structures including the insular, orbitofrontal, temporal, cingulate, and prefrontal cortices have been identified in this context. Although the central network involving these structures is much more intricate, this review highlights recent evidence identifying these regions in sympathetic overactivity in obesity.
Collapse
Affiliation(s)
- Mariya Patel
- School of Health Sciences, Swinburne University of Technology, Melbourne, Victoria, Australia
| | - Joe Braun
- School of Health Sciences, Swinburne University of Technology, Melbourne, Victoria, Australia
| | - Gavin Lambert
- School of Health Sciences, Swinburne University of Technology, Melbourne, Victoria, Australia
- Iverson Health Innovation Research Institute, Swinburne University of Technology, Melbourne, Victoria, Australia
| | - Tatiana Kameneva
- Iverson Health Innovation Research Institute, Swinburne University of Technology, Melbourne, Victoria, Australia
- School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Melbourne, Victoria, Australia
- Department of Biomedical Engineering, The University of Melbourne, Melbourne, Victoria, Australia
| | - Charlotte Keatch
- School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Melbourne, Victoria, Australia
| | - Elisabeth Lambert
- School of Health Sciences, Swinburne University of Technology, Melbourne, Victoria, Australia
- Iverson Health Innovation Research Institute, Swinburne University of Technology, Melbourne, Victoria, Australia
| |
Collapse
|
12
|
Shetty S, Duesman SJ, Patel S, Huyhn P, Shroff S, Das A, Chowhan D, Sebra R, Beaumont K, McAlpine CS, Rajbhandari P, Rajbhandari AK. Sexually dimorphic role of diet and stress on behavior, energy metabolism, and the ventromedial hypothalamus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.17.567534. [PMID: 38014350 PMCID: PMC10680837 DOI: 10.1101/2023.11.17.567534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Scientific evidence underscores the influence of biological sex on the interplay between stress and metabolic dysfunctions. However, there is limited understanding of how diet and stress jointly contribute to metabolic dysregulation in both males and females. To address this gap, our study aimed to investigate the combined effects of a high-fat diet (HFD) and repeated footshock stress on fear-related behaviors and metabolic outcomes in male and female mice. Using a robust rodent model that recapitulates key aspects of post-traumatic stress disorder (PTSD), we subjected mice to footshock stressor followed by weekly reminder footshock stressor or no stressor for 14 weeks while on either an HFD or chow diet. Our findings revealed that HFD impaired fear memory extinction in male mice that received initial stressor but not in female mice. Blood glucose levels were influenced by both diet and sex, with HFD-fed female mice displaying elevated levels that returned to baseline in the absence of stress, a pattern not observed in male mice. Male mice on HFD exhibited higher energy expenditure, while HFD-fed female mice showed a decreased respiratory exchange ratio (RER). Sex-specific alterations in pro-inflammatory markers and abundance of hematopoietic stem cells were observed in chronically stressed mice on an HFD in different peripheral tissues, indicating the manifestation of distinct comorbid disorders. Single-nuclei RNA sequencing of the ventromedial hypothalamus from stressed mice on an HFD provided insights into sex-specific glial cell activation and cell-type-specific transcriptomic changes. In conclusion, our study offers a comprehensive understanding of the intricate interactions between stress, diet, sex, and various physiological and behavioral outcomes, shedding light on a potential brain region coordinating these interactions.
Collapse
Affiliation(s)
- Sanutha Shetty
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, NY, New York 10029
| | - Samuel J. Duesman
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, NY, New York 10029
| | - Sanil Patel
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, NY, New York 10029
| | - Pacific Huyhn
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, NY, New York 10029
| | - Sanjana Shroff
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anika Das
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, NY, New York 10029
- Center for Excellence in Youth Education, Icahn School of Medicine at Mount Sinai, NY, New York 10029
| | - Disha Chowhan
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert Sebra
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kristin Beaumont
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cameron S. McAlpine
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, NY, New York 10029
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, NY, New York 10029
| | - Prashant Rajbhandari
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, NY, New York 10029
- Disease Mechanism and Therapeutics Program, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Senior authors
| | - Abha K. Rajbhandari
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, NY, New York 10029
- Senior authors
| |
Collapse
|
13
|
Ribeiro R, Silva EG, Moreira FC, Gomes GF, Cussat GR, Silva BSR, da Silva MCM, de Barros Fernandes H, de Sena Oliveira C, de Oliveira Guarnieri L, Lopes V, Ferreira CN, de Faria AMC, Maioli TU, Ribeiro FM, de Miranda AS, Moraes GSP, de Oliveira ACP, Vieira LB. Chronic hyperpalatable diet induces impairment of hippocampal-dependent memories and alters glutamatergic and fractalkine axis signaling. Sci Rep 2023; 13:16358. [PMID: 37773430 PMCID: PMC10541447 DOI: 10.1038/s41598-023-42955-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 09/16/2023] [Indexed: 10/01/2023] Open
Abstract
Chronic consumption of hyperpalatable and hypercaloric foods has been pointed out as a factor associated with cognitive decline and memory impairment in obesity. In this context, the integration between peripheral and central inflammation may play a significant role in the negative effects of an obesogenic environment on memory. However, little is known about how obesity-related peripheral inflammation affects specific neurotransmission systems involved with memory regulation. Here, we test the hypothesis that chronic exposure to a highly palatable diet may cause neuroinflammation, glutamatergic dysfunction, and memory impairment. For that, we exposed C57BL/6J mice to a high sugar and butter diet (HSB) for 12 weeks, and we investigated its effects on behavior, glial reactivity, blood-brain barrier permeability, pro-inflammatory features, glutamatergic alterations, plasticity, and fractalkine-CX3CR1 axis. Our results revealed that HSB diet induced a decrease in memory reconsolidation and extinction, as well as an increase in hippocampal glutamate levels. Although our data indicated a peripheral pro-inflammatory profile, we did not observe hippocampal neuroinflammatory features. Furthermore, we also observed that the HSB diet increased hippocampal fractalkine levels, a key chemokine associated with neuroprotection and inflammatory regulation. Then, we hypothesized that the elevation on glutamate levels may saturate synaptic communication, partially limiting plasticity, whereas fractalkine levels increase as a strategy to decrease glutamatergic damage.
Collapse
Affiliation(s)
- Roberta Ribeiro
- Department of Pharmacology, ICB, Federal University of Minas Gerais, Ave. Antonio Carlos 6627, Belo Horizonte, MG, CEP: 31270-901, Brazil
| | - Emanuele Guimarães Silva
- Department of Immunology and Biochemistry, ICB, University of Minas Gerais, Belo Horizonte, Brazil
| | - Felipe Caixeta Moreira
- Department of Immunology and Biochemistry, ICB, University of Minas Gerais, Belo Horizonte, Brazil
| | - Giovanni Freitas Gomes
- Center of Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Gabriela Reis Cussat
- Department of Pharmacology, ICB, Federal University of Minas Gerais, Ave. Antonio Carlos 6627, Belo Horizonte, MG, CEP: 31270-901, Brazil
| | - Barbara Stehling Ramos Silva
- Department of Pharmacology, ICB, Federal University of Minas Gerais, Ave. Antonio Carlos 6627, Belo Horizonte, MG, CEP: 31270-901, Brazil
| | - Maria Carolina Machado da Silva
- Department of Pharmacology, ICB, Federal University of Minas Gerais, Ave. Antonio Carlos 6627, Belo Horizonte, MG, CEP: 31270-901, Brazil
| | | | - Carolina de Sena Oliveira
- Department of Pharmacology, ICB, Federal University of Minas Gerais, Ave. Antonio Carlos 6627, Belo Horizonte, MG, CEP: 31270-901, Brazil
| | | | - Victoria Lopes
- Colégio Técnico, University of Minas Gerais, Belo Horizonte, Brazil
| | | | | | - Tatiani Uceli Maioli
- Department of Immunology and Biochemistry, ICB, University of Minas Gerais, Belo Horizonte, Brazil
| | - Fabíola Mara Ribeiro
- Department of Immunology and Biochemistry, ICB, University of Minas Gerais, Belo Horizonte, Brazil
| | | | | | | | - Luciene Bruno Vieira
- Department of Pharmacology, ICB, Federal University of Minas Gerais, Ave. Antonio Carlos 6627, Belo Horizonte, MG, CEP: 31270-901, Brazil.
| |
Collapse
|
14
|
Mukherjee S, Skrede S, Haugstøyl M, López M, Fernø J. Peripheral and central macrophages in obesity. Front Endocrinol (Lausanne) 2023; 14:1232171. [PMID: 37720534 PMCID: PMC10501731 DOI: 10.3389/fendo.2023.1232171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/28/2023] [Indexed: 09/19/2023] Open
Abstract
Obesity is associated with chronic, low-grade inflammation. Excessive nutrient intake causes adipose tissue expansion, which may in turn cause cellular stress that triggers infiltration of pro-inflammatory immune cells from the circulation as well as activation of cells that are residing in the adipose tissue. In particular, the adipose tissue macrophages (ATMs) are important in the pathogenesis of obesity. A pro-inflammatory activation is also found in other organs which are important for energy metabolism, such as the liver, muscle and the pancreas, which may stimulate the development of obesity-related co-morbidities, including insulin resistance, type 2 diabetes (T2D), cardiovascular disease (CVD) and non-alcoholic fatty liver disease (NAFLD). Interestingly, it is now clear that obesity-induced pro-inflammatory signaling also occurs in the central nervous system (CNS), and that pro-inflammatory activation of immune cells in the brain may be involved in appetite dysregulation and metabolic disturbances in obesity. More recently, it has become evident that microglia, the resident macrophages of the CNS that drive neuroinflammation, may also be activated in obesity and can be relevant for regulation of hypothalamic feeding circuits. In this review, we focus on the action of peripheral and central macrophages and their potential roles in metabolic disease, and how macrophages interact with other immune cells to promote inflammation during obesity.
Collapse
Affiliation(s)
- Sayani Mukherjee
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| | - Silje Skrede
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Martha Haugstøyl
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Miguel López
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| | - Johan Fernø
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
15
|
Li ZA, Cai Y, Taylor RL, Eisenstein SA, Barch DM, Marek S, Hershey T. Associations Between Socioeconomic Status, Obesity, Cognition, and White Matter Microstructure in Children. JAMA Netw Open 2023; 6:e2320276. [PMID: 37368403 DOI: 10.1001/jamanetworkopen.2023.20276] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/28/2023] Open
Abstract
Importance Lower neighborhood and household socioeconomic status (SES) are associated with negative health outcomes and altered brain structure in children. It is unclear whether such findings extend to white matter and via what mechanisms. Objective To assess whether and how neighborhood and household SES are independently associated with children's white matter microstructure and examine whether obesity and cognitive performance (reflecting environmental cognitive and sensory stimulation) are plausible mediators. Design, Setting, and Participants This cross-sectional study used baseline data from participants in the Adolescent Brain Cognitive Development (ABCD) study. Data were collected at 21 US sites, and school-based recruitment was used to represent the US population. Children aged 9 to 11 years and their parents or caregivers completed assessments between October 1, 2016, and October 31, 2018. After exclusions, 8842 of 11 875 children in the ABCD study were included in the analyses. Data analysis was conducted from July 11 to December 19, 2022. Exposures Neighborhood disadvantage was derived from area deprivation indices at participants' primary residence. Household SES factors were total income and highest parental educational attainment. Main Outcomes and Measures A restriction spectrum imaging (RSI) model was used to quantify restricted normalized directional (RND; reflecting oriented myelin organization) and restricted normalized isotropic (RNI; reflecting glial and neuronal cell bodies) diffusion in 31 major white matter tracts. The RSI measurements were scanner harmonized. Obesity was assessed through body mass index (BMI; calculated as weight in kilograms divided by height in meters squared), age- and sex-adjusted BMI z scores, and waist circumference, and cognition was assessed through the National Institutes of Health Toolbox Cognition Battery. Analyses were adjusted for age, sex, pubertal development stage, intracranial volume, mean head motion, and twin or siblingship. Results Among 8842 children, 4543 (51.4%) were boys, and the mean (SD) age was 9.9 (0.7) years. Linear mixed-effects models revealed that greater neighborhood disadvantage was associated with lower RSI-RND in the left superior longitudinal fasciculus (β = -0.055; 95% CI, -0.081 to -0.028) and forceps major (β = -0.040; 95% CI, -0.067 to -0.013). Lower parental educational attainment was associated with lower RSI-RND in the bilateral superior longitudinal fasciculus (eg, right hemisphere: β = 0.053; 95% CI, 0.025-0.080) and bilateral corticospinal or pyramidal tract (eg, right hemisphere: β = 0.042; 95% CI, 0.015-0.069). Structural equation models revealed that lower cognitive performance (eg, lower total cognition score and higher neighborhood disadvantage: β = -0.012; 95% CI, -0.016 to -0.009) and greater obesity (eg, higher BMI and higher neighborhood disadvantage: β = -0.004; 95% CI, -0.006 to -0.001) partially accounted for the associations between SES and RSI-RND. Lower household income was associated with higher RSI-RNI in most tracts (eg, right inferior longitudinal fasciculus: β = -0.042 [95% CI, -0.073 to -0.012]; right anterior thalamic radiations: β = -0.045 [95% CI, -0.075 to -0.014]), and greater neighborhood disadvantage had similar associations in primarily frontolimbic tracts (eg, right fornix: β = 0.046 [95% CI, 0.019-0.074]; right anterior thalamic radiations: β = 0.045 [95% CI, 0.018-0.072]). Lower parental educational attainment was associated with higher RSI-RNI in the forceps major (β = -0.048; 95% CI, -0.077 to -0.020). Greater obesity partially accounted for these SES associations with RSI-RNI (eg, higher BMI and higher neighborhood disadvantage: β = 0.015; 95% CI, 0.011-0.020). Findings were robust in sensitivity analyses and were corroborated using diffusion tensor imaging. Conclusions and Relevance In this cross-sectional study, both neighborhood and household contexts were associated with white matter development in children, and findings suggested that obesity and cognitive performance were possible mediators in these associations. Future research on children's brain health may benefit from considering these factors from multiple socioeconomic perspectives.
Collapse
Affiliation(s)
- Zhaolong Adrian Li
- Department of Psychiatry, Washington University in St Louis School of Medicine, St Louis, Missouri
- Department of Psychological and Brain Sciences, Washington University in St Louis, St Louis, Missouri
| | - Yuqi Cai
- Department of Psychological and Brain Sciences, Washington University in St Louis, St Louis, Missouri
- Now with Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Rita L Taylor
- Department of Psychological and Brain Sciences, Washington University in St Louis, St Louis, Missouri
| | - Sarah A Eisenstein
- Department of Psychiatry, Washington University in St Louis School of Medicine, St Louis, Missouri
- Mallinckrodt Institute of Radiology, Washington University in St Louis School of Medicine, St Louis, Missouri
| | - Deanna M Barch
- Department of Psychiatry, Washington University in St Louis School of Medicine, St Louis, Missouri
- Department of Psychological and Brain Sciences, Washington University in St Louis, St Louis, Missouri
- Mallinckrodt Institute of Radiology, Washington University in St Louis School of Medicine, St Louis, Missouri
| | - Scott Marek
- Department of Psychiatry, Washington University in St Louis School of Medicine, St Louis, Missouri
- Mallinckrodt Institute of Radiology, Washington University in St Louis School of Medicine, St Louis, Missouri
| | - Tamara Hershey
- Department of Psychiatry, Washington University in St Louis School of Medicine, St Louis, Missouri
- Department of Psychological and Brain Sciences, Washington University in St Louis, St Louis, Missouri
- Mallinckrodt Institute of Radiology, Washington University in St Louis School of Medicine, St Louis, Missouri
- Department of Neurology, Washington University in St Louis School of Medicine, St Louis, Missouri
| |
Collapse
|
16
|
Grover L, Sklioutovskaya-Lopez K, Parkman JK, Wang K, Hendricks E, Adams-Duffield J, Kim JH. Diet, sex, and genetic predisposition to obesity and type 2 diabetes modulate motor and anxiety-related behaviors in mice, and alter cerebellar gene expression. Behav Brain Res 2023; 445:114376. [PMID: 36868363 PMCID: PMC10065959 DOI: 10.1016/j.bbr.2023.114376] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 12/28/2022] [Accepted: 02/28/2023] [Indexed: 03/05/2023]
Abstract
Obesity and type 2 diabetes (T2D) are serious health problems linked to neurobehavioral alterations. We compared motor function, anxiety-related behavior, and cerebellar gene expression in TALLYHO/Jng (TH), a polygenic model prone to insulin resistance, obesity, and T2D, and normal C57BL/6 J (B6) mice. Male and female mice were weaned onto chow or high fat (HF) diet at 4 weeks of age (wk), and experiments conducted at young (5 wk) and old (14 - 20 wk) ages. In the open field, distance traveled was significantly lower in TH (vs. B6). For old mice, anxiety-like behavior (time in edge zone) was significantly increased for TH (vs B6), females (vs males), and for both ages HF diet (vs chow). In Rota-Rod testing, latency to fall was significantly shorter in TH (vs B6). For young mice, longer latencies to fall were observed for females (vs males) and HF (vs chow). Grip strength in young mice was greater in TH (vs B6), and there was a diet-strain interaction, with TH on HF showing increased strength, whereas B6 on HF showed decreased strength. For older mice, there was a strain-sex interaction, with B6 males (but not TH males) showing increased strength compared to the same strain females. There were significant sex differences in cerebellar mRNA levels, with Tnfα higher, and Glut4 and Irs2 lower in females (vs males). There were significant strain effects for Gfap and Igf1 mRNA levels with lower in TH (vs B6). Altered cerebellar gene expression may contribute to strain differences in coordination and locomotion.
Collapse
Affiliation(s)
- Lawrence Grover
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | | | - Jacaline K Parkman
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Katherine Wang
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Emily Hendricks
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Jessica Adams-Duffield
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Jung Han Kim
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA.
| |
Collapse
|
17
|
Li ZA, Samara A, Ray MK, Rutlin J, Raji CA, Shimony JS, Sun P, Song SK, Hershey T, Eisenstein SA. Childhood obesity is linked to putative neuroinflammation in brain white matter, hypothalamus, and striatum. Cereb Cortex Commun 2023; 4:tgad007. [PMID: 37207193 PMCID: PMC10191798 DOI: 10.1093/texcom/tgad007] [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: 11/17/2022] [Revised: 04/19/2023] [Accepted: 04/22/2023] [Indexed: 05/21/2023] Open
Abstract
Neuroinflammation is both a consequence and driver of overfeeding and weight gain in rodent obesity models. Advances in magnetic resonance imaging (MRI) enable investigations of brain microstructure that suggests neuroinflammation in human obesity. To assess the convergent validity across MRI techniques and extend previous findings, we used diffusion basis spectrum imaging (DBSI) to characterize obesity-associated alterations in brain microstructure in 601 children (age 9-11 years) from the Adolescent Brain Cognitive DevelopmentSM Study. Compared with children with normal-weight, greater DBSI restricted fraction (RF), reflecting neuroinflammation-related cellularity, was seen in widespread white matter in children with overweight and obesity. Greater DBSI-RF in hypothalamus, caudate nucleus, putamen, and, in particular, nucleus accumbens, correlated with higher baseline body mass index and related anthropometrics. Comparable findings were seen in the striatum with a previously reported restriction spectrum imaging (RSI) model. Gain in waist circumference over 1 and 2 years related, at nominal significance, to greater baseline RSI-assessed restricted diffusion in nucleus accumbens and caudate nucleus, and DBSI-RF in hypothalamus, respectively. Here we demonstrate that childhood obesity is associated with microstructural alterations in white matter, hypothalamus, and striatum. Our results also support the reproducibility, across MRI methods, of findings of obesity-related putative neuroinflammation in children.
Collapse
Affiliation(s)
- Zhaolong Adrian Li
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, United States
- Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, MO 63130, United States
| | - Amjad Samara
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, United States
- Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110 United States
| | - Mary Katherine Ray
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, United States
| | - Jerrel Rutlin
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, United States
| | - Cyrus A Raji
- Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110 United States
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, United States
| | - Joshua S Shimony
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, United States
| | - Peng Sun
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, United States
- Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Sheng-Kwei Song
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, United States
| | - Tamara Hershey
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, United States
- Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, MO 63130, United States
- Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110 United States
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, United States
| | - Sarah A Eisenstein
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, United States
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, United States
| |
Collapse
|
18
|
Gomes SV, Dias BV, Júnior PAM, Pereira RR, de Souza DMS, Breguez GS, de Lima WG, Magalhães CLDB, Cangussú SD, Talvani A, Queiroz KB, Calsavara AJC, Costa DC. High-fat diet increases mortality and intensifies immunometabolic changes in septic mice. J Nutr Biochem 2023; 116:109315. [PMID: 36921735 DOI: 10.1016/j.jnutbio.2023.109315] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 01/21/2023] [Accepted: 03/07/2023] [Indexed: 03/16/2023]
Abstract
Immunometabolic changes in the liver and white adipose tissue (WAT) caused by high-fat (HF) diet intake may worse metabolic adaptation and protection against pathogens in sepsis. We investigate the effect of chronic HF diet (15 weeks) on mortality and immunometabolic responses in female mice after sepsis induced by cecum ligation and perforation (CLP). At week 14, animals were divided into four groups: sham C diet (C-Sh), sepsis C diet (C-Sp), sham HF diet (HF-Sh) and sepsis HF diet (HF-Sp). The surviving animals were euthanised on the 7th day. The HF diet decreased survival rate (58.3% vs 76.2% C-Sp group), increased serum cytokine storm (IL-6 (1.41 ×; vs HF-Sh), IL-1β (1.37 ×; vs C-Sp), TNF (1.34 ×; vs C-Sp and 1.72 ×; vs HF-Sh), IL-17 (1.44 ×; vs HF-Sh), IL-10 (1.55 ×; vs C-Sp and 1.41 ×; HF-Sh), WAT inflammation (IL-6 (8.7 ×; vs C-Sp and 2.4 ×; vs HF-Sh), TNF (5 ×; vs C-Sp and 1.7 ×;vs HF-Sh), IL-17 (1.7 ×; vs C-Sp), IL-10 (7.4 ×; vs C-Sp and 1.3 ×; vs HF-Sh), and modulated lipid metabolism in septic mice. In the HF-Sp group liver's, we observed hepatomegaly, hydropic degeneration, necrosis, an increase in oxidative stress (reduction of CAT activity (-81.7%; vs HF-Sh); increase MDA levels (82.8%; vs HF-Sh), and hepatic IL-6 (1.9 ×; vs HF-Sh), and TNF (1.3 × %;vs HF-Sh) production. Furthermore, we found a decrease in the total number of inflammatory, mononuclear cells, and in the regenerative processes, and binucleated hepatocytes in a HF-Sp group liver's. Our results suggested that the organism under metabolic stress of a HF diet during sepsis may worsen the inflammatory landscape and hepatocellular injury and may harm the liver regenerative process.
Collapse
Affiliation(s)
- Sttefany Viana Gomes
- Laboratory of Metabolic Biochemistry (LBM), Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Bruna Vidal Dias
- Laboratory of Metabolic Biochemistry (LBM), Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Pedro Alves Machado Júnior
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Renata Rebeca Pereira
- Laboratory of Metabolic Biochemistry (LBM), Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Débora Maria Soares de Souza
- Laboratory of Immunobiology of Inflammation, Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Gustavo Silveira Breguez
- Multiuser Research Laboratory, School of Nutrition, School of Nutrition, Postgraduate Program in Health and Nutrition, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Wanderson Geraldo de Lima
- Morphopathology Laboratory, Department of Biological Sciences (DECBI), Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Cintia Lopes de Brito Magalhães
- Laboratory of Biology and Technology of Microorganisms (LBTM), Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Silvia Dantas Cangussú
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - André Talvani
- Laboratory of Immunobiology of Inflammation, Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Karina Barbosa Queiroz
- Laboratory of Experimental Nutrition (LABNEx), Department of Food, Postgraduate Program in Health and Nutrition, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Allan Jefferson Cruz Calsavara
- Laboratory of Cognition and Health (LACOS), School of Medicine, Department of Pediatric and Adult Clinics (DECPA), Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Daniela Caldeira Costa
- Laboratory of Metabolic Biochemistry (LBM), Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil.
| |
Collapse
|
19
|
Vints WA, Kušleikienė S, Sheoran S, Valatkevičienė K, Gleiznienė R, Himmelreich U, Pääsuke M, Česnaitienė VJ, Levin O, Verbunt J, Masiulis N. Body fat and components of sarcopenia relate to inflammation, brain volume and neurometabolism in older adults. Neurobiol Aging 2023; 127:1-11. [PMID: 37004309 DOI: 10.1016/j.neurobiolaging.2023.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 12/27/2022] [Accepted: 02/24/2023] [Indexed: 03/06/2023]
Abstract
Obesity and sarcopenia are associated with cognitive impairments at older age. Current research suggests that blood biomarkers may mediate this body-brain crosstalk, altering neurometabolism and brain structure eventually resulting in cognitive performance changes. Seventy-four older adults (60-85 years old) underwent bio-impedance body composition analysis, handgrip strength measurements, 8-Foot Up-and-Go (8UG) test, Montreal Cognitive Assessment (MoCA), blood analysis of interleukin-6 (IL-6), kynurenine, and insulin-like growth factor-1 (IGF-1), as well as brain magnetic resonance imaging (MRI) and proton magnetic resonance spectroscopy (1H-MRS), estimating neurodegeneration and neuroinflammation. Normal fat% or overweight was associated with larger total gray matter volume compared to underweight or obesity in older adults and obesity was associated with higher N-acetylaspartate/Creatine levels in the sensorimotor and dorsolateral prefrontal cortex. Muscle strength, not muscle mass/physical performance, corresponded to lower kynurenine and higher N-acetylaspartate/Creatine levels in the dorsal posterior cingulate and dorsolateral prefrontal cortex. The inflammatory and neurotrophic blood biomarkers did not significantly mediate these body-brain associations. This study used a multimodal approach to comprehensively assess the proposed mechanism of body-brain crosstalk.
Collapse
|
20
|
Li ZA, Cai Y, Taylor RL, Eisenstein SA, Barch DM, Marek S, Hershey T. Associations between socioeconomic status and white matter microstructure in children: indirect effects via obesity and cognition. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.02.09.23285150. [PMID: 36798149 PMCID: PMC9934783 DOI: 10.1101/2023.02.09.23285150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Importance Both neighborhood and household socioeconomic disadvantage relate to negative health outcomes and altered brain structure in children. It is unclear whether such findings extend to white matter development, and via what mechanisms socioeconomic status (SES) influences the brain. Objective To test independent associations between neighborhood and household SES indicators and white matter microstructure in children, and examine whether body mass index and cognitive function (a proxy of environmental cognitive/sensory stimulation) may plausibly mediate these associations. Design This cross-sectional study used baseline data from the Adolescent Brain Cognitive Development (ABCD) Study, an ongoing 10-year cohort study tracking child health. Setting School-based recruitment at 21 U.S. sites. Participants Children aged 9 to 11 years and their parents/caregivers completed baseline assessments between October 1 st , 2016 and October 31 st , 2018. Data analysis was conducted from July to December 2022. Exposures Neighborhood disadvantage was derived from area deprivation indices at primary residence. Household SES indicators were total income and the highest parental education attainment. Main Outcomes and Measures Thirty-one major white matter tracts were segmented from diffusion-weighted images. The Restriction Spectrum Imaging (RSI) model was implemented to measure restricted normalized directional (RND; reflecting oriented myelin organization) and isotropic (RNI; reflecting glial/neuronal cell bodies) diffusion in each tract. Obesity-related measures were body mass index (BMI), BMI z -scores, and waist circumference, and cognitive performance was assessed using the NIH Toolbox Cognition Battery. Linear mixed-effects models tested the associations between SES indicators and scanner-harmonized RSI metrics. Structural equation models examined indirect effects of obesity and cognitive performance in the significant associations between SES and white mater microstructure summary principal components. Analyses were adjusted for age, sex, pubertal development stage, intracranial volume, and head motion. Results The analytical sample included 8842 children (4299 [48.6%] girls; mean age [SD], 9.9 [0.7] years). Greater neighborhood disadvantage and lower parental education were independently associated with lower RSI-RND in forceps major and corticospinal/pyramidal tracts, and had overlapping associations in the superior longitudinal fasciculus. Lower cognition scores and greater obesity-related measures partially accounted for these SES associations with RSI-RND. Lower household income was related to higher RSI-RNI in almost every tract, and greater neighborhood disadvantage had similar effects in primarily frontolimbic tracts. Lower parental education was uniquely linked to higher RSI-RNI in forceps major. Greater obesity-related measures partially accounted for these SES associations with RSI-RNI. Findings were robust in sensitivity analyses and mostly corroborated using traditional diffusion tensor imaging (DTI). Conclusions and Relevance These cross-sectional results demonstrate that both neighborhood and household contexts are relevant to white matter development in children, and suggest cognitive performance and obesity as possible pathways of influence. Interventions targeting obesity reduction and improving cognition from multiple socioeconomic angles may ameliorate brain health in low-SES children. Key Points Question: Are neighborhood and household socioeconomic levels associated with children’s brain white matter microstructure, and if so, do obesity and cognitive performance (reflecting environmental stimulation) mediate the associations?Findings: In a cohort of 8842 children, higher neighborhood disadvantage, lower household income, and lower parental education had independent and overlapping associations with lower restricted directional diffusion and greater restricted isotropic diffusion in white matter. Greater body mass index and poorer cognitive performance partially mediated these associations.Meaning: Both neighborhood and household poverty may contribute to altered white matter development in children. These effects may be partially explained by obesity incidence and poorer cognitive performance.
Collapse
Affiliation(s)
- Zhaolong Adrian Li
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63130, USA
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Yuqi Cai
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Rita L. Taylor
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Sarah A. Eisenstein
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63130, USA
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Deanna M. Barch
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63130, USA
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO 63130, USA
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Scott Marek
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Tamara Hershey
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63130, USA
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO 63130, USA
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
21
|
Binayi F, Moslemi M, Khodagholi F, Hedayati M, Zardooz H. Long-term high-fat diet disrupts lipid metabolism and causes inflammation in adult male rats: possible intervention of endoplasmic reticulum stress. Arch Physiol Biochem 2023; 129:204-212. [PMID: 32907408 DOI: 10.1080/13813455.2020.1808997] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
This study investigated the effect of long-term high-fat diet (HFD) on plasma lipid profile and probability of inflammation in adult rats. After weaning, male offspring were divided into six groups based on diet type and medication. After 20 weeks of dietary intake, 4-PBA (endoplasmic reticulum (ER) stress inhibitor) was injected for three days. Then, blood samples were taken to measure plasma concentrations of low-density lipoprotein (LDL), triglyceride (TG), high-density lipoprotein (HDL), cholesterol, leptin and interleukin 1-β (IL 1-β). The HFD increased body weight and food intake and intra-abdominal fat and thymus weights, which were associated with elevated plasma leptin level. Moreover, HFD increased plasma concentrations of TG, LDL, cholesterol and IL 1-β and decreased HDL level. Injection of 4-PBA reversed the plasma parameters changes caused by HFD. It seems that long-term HFD feeding through inducing the ER stress, disrupted the lipid metabolism and resulted in inflammation.
Collapse
Affiliation(s)
- Fateme Binayi
- Department of Physiology, School of Medicine, Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Moslemi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- NeuroBiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Hedayati
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Homeira Zardooz
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Clyburn C, Carson KE, Smith CR, Travagli RA, Browning KN. Brainstem astrocytes control homeostatic regulation of caloric intake. J Physiol 2023; 601:801-829. [PMID: 36696965 PMCID: PMC10026361 DOI: 10.1113/jp283566] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 12/08/2022] [Indexed: 01/27/2023] Open
Abstract
Prolonged high-fat diet (HFD) exposure is associated with hyperphagia, excess caloric intake and weight gain. After initial exposure to a HFD, a brief (24-48 h) period of hyperphagia is followed by the regulation of caloric intake and restoration of energy balance within an acute (3-5 day) period. Previous studies have demonstrated this occurs via a vagally mediated signalling cascade that increases glutamatergic transmission via activation of NMDA receptors located on gastric-projecting neurons of the dorsal motor nucleus of the vagus (DMV). The present study used electrophysiological recordings from thin brainstem slice preparations, in vivo recordings of gastric motility and tone, measurement of gastric emptying rates, and food intake studies to investigate the hypothesis that activation of brainstem astrocytes in response to acute HFD exposure is responsible for the increased glutamatergic drive to DMV neurons and the restoration of caloric balance. Pharmacological and chemogenetic inhibition of brainstem astrocytes reduced glutamatergic signalling and DMV excitability, dysregulated gastric tone and motility, attenuated the homeostatic delay in gastric emptying, and prevented the decrease in food intake that is observed during the period of energy regulation following initial exposure to HFD. Understanding the mechanisms involved in caloric regulation may provide critical insights into energy balance as well as into the hyperphagia that develops as these mechanisms are overcome. KEY POINTS: Initial exposure to a high fat diet is associated with a brief period of hyperphagia before caloric intake and energy balance is restored. This period of homeostatic regulation is associated with a vagally mediated signalling cascade that increases glutamatergic transmission to dorsal motor nucleus of the vagus (DMV) neurons via activation of synaptic NMDA receptors. The present study demonstrates that pharmacological and chemogenetic inhibition of brainstem astrocytes reduced glutamatergic signalling and DMV neuronal excitability, dysregulated gastric motility and tone and emptying, and prevented the regulation of food intake following high-fat diet exposure. Astrocyte regulation of glutamatergic transmission to DMV neurons appears to involve release of the gliotransmitters glutamate and ATP. Understanding the mechanisms involved in caloric regulation may provide critical insights into energy balance as well as into the hyperphagia that develops as these mechanisms are overcome.
Collapse
Affiliation(s)
- Courtney Clyburn
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA
- Current position: Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, 97056
| | - Kaitlin E. Carson
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA
| | - Caleb R. Smith
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA
| | - R. Alberto Travagli
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA
- Current position: Neurobiology Research, Newport, NC 28570
| | - Kirsteen N. Browning
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA
| |
Collapse
|
23
|
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: 6] [Impact Index Per Article: 6.0] [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
|
24
|
Breach MR, Lenz KM. Sex Differences in Neurodevelopmental Disorders: A Key Role for the Immune System. Curr Top Behav Neurosci 2023; 62:165-206. [PMID: 35435643 PMCID: PMC10286778 DOI: 10.1007/7854_2022_308] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Sex differences are prominent defining features of neurodevelopmental disorders. Understanding the sex biases in these disorders can shed light on mechanisms leading to relative risk and resilience for the disorders, as well as more broadly advance our understanding of how sex differences may relate to brain development. The prevalence of neurodevelopmental disorders is increasing, and the two most common neurodevelopmental disorders, Autism Spectrum Disorder (ASD) and Attention-Deficit/Hyperactivity Disorder (ADHD) exhibit male-biases in prevalence rates and sex differences in symptomology. While the causes of neurodevelopmental disorders and their sex differences remain to be fully understood, increasing evidence suggests that the immune system plays a critical role in shaping development. In this chapter we discuss sex differences in prevalence and symptomology of ASD and ADHD, review sexual differentiation and immune regulation of neurodevelopment, and discuss findings from human and rodent studies of immune dysregulation and perinatal immune perturbation as they relate to potential mechanisms underlying neurodevelopmental disorders. This chapter will give an overview of how understanding sex differences in neuroimmune function in the context of neurodevelopmental disorders could lend insight into their etiologies and better treatment strategies.
Collapse
Affiliation(s)
- Michaela R Breach
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Kathryn M Lenz
- Department of Psychology, The Ohio State University, Columbus, OH, USA.
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA.
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
25
|
Melatonin decreases IRF-3 protein expression in the gastrocnemius muscle, reduces IL-1β and LPS plasma concentrations, and improves the lipid profile in rats with apical periodontitis fed on a high-fat diet. Odontology 2022:10.1007/s10266-022-00782-w. [PMID: 36567367 DOI: 10.1007/s10266-022-00782-w] [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: 09/24/2022] [Accepted: 12/14/2022] [Indexed: 12/26/2022]
Abstract
To evaluate the effects of melatonin (MEL) on the expression of toll-like receptor-4 (TLR4); myeloid differentiation primary response protein-88 (MyD88); TIR-domain-containing adapter-inducing interferon-β (TRIF); IFN regulatory-factor-3 (IRF-3); nuclear factor kappa-B (NF-κB); plasma concentrations of interleukin-1β (IL-1β) and lipopolysaccharide (LPS); and lipid profile of rats with apical periodontitis (AP) fed on a high-fat diet (HFD). Eighty 60-day-old rats were divided into eight groups: control, AP, HFD, HFDAP, CNMEL, APMEL, HFDMEL and HFDAPMEL. HFD groups were fed on a HFD for 107 days. On day 7, experimental AP was induced in the AP groups, and after 70 days, MEL (5 mg/kg) was administered to the MEL groups for 30 days. Plasma concentrations of LPS and IL-1β were analyzed using enzyme-linked immunosorbent assay, and the lipid profile was analyzed using biochemical tests. The expression of proteins involved in the TLR4 pathway (TLR4, MyD88, TRIF, IRF-3 and NF-κB) in the gastrocnemius muscle (GM) was evaluated using western blotting and qRT-PCR. Treatment with MEL decreased IRF-3 protein expression in GM and IL-1β plasma concentration in the APMEL and HFDMEL groups. Reduction in LPS plasma concentration was reported only in the HFDMEL group. Additionally, a decrease in LDL and an increase in HDL were observed in the HFDMEL and HFDAPMEL groups. Treatment with MEL exhibited anti-inflammatory and anti-hyperlipidemic effects attributed to HFD and AP by reducing the plasma concentrations of IL-1β and LPS in addition to reducing IRF-3 protein expression in the GM, which is associated with the production of inflammatory cytokines.
Collapse
|
26
|
Smith DC, Karahan H, Wijeratne HRS, Al-Amin M, McCord B, Moon Y, Kim J. Deletion of the Alzheimer's disease risk gene Abi3 locus results in obesity and systemic metabolic disruption in mice. Front Aging Neurosci 2022; 14:1035572. [PMID: 36620768 PMCID: PMC9813750 DOI: 10.3389/fnagi.2022.1035572] [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: 09/02/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022] Open
Abstract
Alzheimer's disease (AD) genetics studies have identified a coding variant within ABI3 gene that increases the risk of developing AD. Recently, we demonstrated that deletion of the Abi3 gene locus dramatically exacerbates AD neuropathology in a transgenic mouse model of amyloidosis. In the course of this AD project, we unexpectedly found that deletion of the Abi3 gene locus resulted in a dramatic obese phenotype in non-transgenic mice. Here, we report our investigation into this serendipitous metabolic finding. Specifically, we demonstrate that mice with deletion of the Abi3 gene locus (Abi3-/- ) have dramatically increased body weight and body fat. Further, we determined that Abi3-/- mice have impaired energy expenditure. Additionally, we found that deletion of the Abi3 gene locus altered gene expression within the hypothalamus, particularly within immune-related pathways. Subsequent immunohistological analysis of the central nervous system (CNS) revealed that microglia number and area were decreased specifically within the mediobasal hypothalamus of Abi3-/- mice. Altogether, this investigation establishes the functional importance of the Abi3 gene locus in the regulation of systemic metabolism and maintenance of healthy body weight. While our previous findings indicated the importance of Abi3 in neurodegeneration, this study indicates that Abi3 related functions are also essential for metabolic regulation.
Collapse
Affiliation(s)
- Daniel C. Smith
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Medical Scientist Training Program, Indiana University School of Medicine, Indianapolis, IN, United States
- Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Hande Karahan
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - H. R. Sagara Wijeratne
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Medical Scientist Training Program, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Mamun Al-Amin
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Brianne McCord
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Younghye Moon
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jungsu Kim
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
27
|
Yang J, Deng Y, Cai Y, Liu Y, Peng L, Luo Z, Li D. Mapping trends and hotspot regarding gastrointestinal microbiome and neuroscience: A bibliometric analysis of global research (2002-2022). Front Neurosci 2022; 16:1048565. [PMID: 36466165 PMCID: PMC9714683 DOI: 10.3389/fnins.2022.1048565] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 10/28/2022] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND Scholars have long understood that gastrointestinal microorganisms are intimately related to human disorders. The literature on research involving the gut microbiome and neuroscience is emerging. This study exposed the connections between gut microbiota and neuroscience methodically and intuitively using bibliometrics and visualization. This study's objectives were to summarize the knowledge structure and identify emerging trends and potential hotspots in this field. MATERIALS AND METHODS On October 18, 2022, a literature search was conducted utilizing the Web of Science Core Collection (WoSCC) database for studies on gut microbiota and neuroscience studies from 2002 to 2022 (August 20, 2022). VOSviewer and CiteSpace V software was used to conduct the bibliometrics and visualization analysis. RESULTS From 2002 to 2022 (August 20, 2022), 2,275 publications in the WoSCC database satisfied the criteria. The annual volume of publications has rapidly emerged in recent years (2016-2022). The most productive nation (n = 732, 32.18%) and the hub of inter-country cooperation (links: 38) were the United States. University College Cork had the most research papers published in this area, followed by McMaster University and Harvard Medical School. Cryan JF, Dinan TG, and Clarke G were key researchers with considerable academic influence. The journals with the most publications are "Neurogastroenterology and Motility" and "Brain Behavior and Immunity." The most cited article and co-cited reference was Cryan JF's 2012 article on the impact of gut microbiota on the brain and behavior. The current research hotspot includes gastrointestinal microbiome, inflammation, gut-brain axis, Parkinson's disease (PD), and Alzheimer's disease (AD). The research focus would be on the "gastrointestinal microbiome, inflammation: a link between obesity, insulin resistance, and cognition" and "the role of two important theories of the gut-brain axis and microbial-gut-brain axis in diseases." Burst detection analysis showed that schizophrenia, pathology, and psychiatric disorder may continue to be the research frontiers. CONCLUSION Research on "gastrointestinal microbiome, inflammation: a link between obesity, insulin resistance, and cognition" and "the role of two important theories of the gut-brain axis and microbial-gut-brain axis in diseases" will continue to be the hotspot. Schizophrenia and psychiatric disorder will be the key research diseases in the field of gut microbiota and neuroscience, and pathology is the key research content, which is worthy of scholars' attention.
Collapse
Affiliation(s)
- Jingjing Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Yihui Deng
- Hunan University of Chinese Medicine, Changsha, China
| | - Yuzhe Cai
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Yixuan Liu
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Lanyu Peng
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Zheng Luo
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Dingxiang Li
- Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
28
|
Kumar S, Raj VS, Ahmad A, Saini V. Amoxicillin modulates gut microbiota to improve short-term high-fat diet induced pathophysiology in mice. Gut Pathog 2022; 14:40. [PMID: 36229889 PMCID: PMC9563906 DOI: 10.1186/s13099-022-00513-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 08/02/2022] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND A high-fat diet (HFD) induced perturbation of gut microbiota is a major contributory factor to promote the pathophysiology of HFD-associated metabolic syndrome. The HFD could also increase the susceptibility to the microbial infections warranting the use of antibiotics which are independently capable of impacting both gut microbiota and metabolic syndrome. Further, the usage of antibiotics in individuals consuming HFD can impact mitochondrial function that can be associated with an elevated risk of chronic conditions like inflammatory bowel disease (IBD). Despite this high propensity to infections in individuals on HFD, the link between duration of HFD and antibiotic treatment, and its impact on diversity of the gut microbiome and features of metabolic syndrome is not well established. In this study, we have addressed these knowledge gaps by examining how the gut microbiota profile changes in HFD-fed mice receiving antibiotic intervention in the form of amoxicillin. We also determine whether antibiotic treatment in HFD-fed mice may adversely impact the ability of immune cells to clear microbial infections. METHODS AND RESULTS We have subjected mice to HFD and chow diet (CD) for 3 weeks, and a subset of these mice on both diets received antibiotic intervention in the form of amoxicillin in the 3rd week. Body weight and food intake were recorded for 3 weeks. After 21 days, all animals were weighted and sacrificed. Subsequently, these animals were evaluated for basic haemato-biochemical and histopathological attributes. We used 16S rRNA sequencing followed by bioinformatics analysis to determine changes in gut microbiota in these mice. We observed that a HFD, even for a short-duration, could successfully induce the partial pathophysiology typical of a metabolic syndrome, and substantially modulated the gut microbiota in mice. The short course of amoxicillin treatment to HFD-fed mice resulted in beneficial effects by significantly reducing fasting blood glucose and skewing the number of thrombocytes towards a normal range. Remarkably, we observed a significant remodelling of gut microbiota in amoxicillin-treated HFD-fed mice. Importantly, some gut microbes associated with improved insulin sensitivity and recovery from metabolic syndrome only appeared in amoxicillin-treated HFD-fed mice reinforcing the beneficial effects of antibiotic treatment in the HFD-associated metabolic syndrome. Moreover, we also observed the presence of gut-microbiota unique to amoxicillin-treated HFD-fed mice that are also known to improve the pathophysiology associated with metabolic syndrome. However, both CD-fed as well as HFD-fed mice receiving antibiotics showed an increase in intestinal pathogens as is typically observed for antibiotic treatment. Importantly though, infection studies with S. aureus and A. baumannii, revealed that macrophages isolated from amoxicillin-treated HFD-fed mice are comparable to those isolated from mice receiving only HFD or CD in terms of susceptibility, and progression of microbial infection. This finding clearly indicated that amoxicillin treatment does not introduce any additional deficits in the ability of macrophages to combat microbial infections. CONCLUSIONS Our results showed that amoxicillin treatment in HFD-fed mice exert a beneficial influence on the pathophysiological attributes of metabolic syndrome which correlates with a significant remodelling of gut microbiota. A novel observation was the increase in microbes known to improve insulin sensitivity following amoxicillin treatment during short-term intake of HFD. Even though there is a minor increase in gut-resistant intestinal pathogens in amoxicillin-treated groups, there is no adverse impact on macrophages with respect to their susceptibility and ability to control infections. Taken together, this study provides a proof of principle for the exploration of amoxicillin treatment as a potential therapy in the people affected with metabolic syndrome.
Collapse
Affiliation(s)
- Suresh Kumar
- National Institute of Biologicals, Ministry of Health & Family Welfare, Govt. of India, Noida, 201309, India.
| | - V Samuel Raj
- Center for Drug, Design, Discovery and Development (C4D), SRM University, Delhi-NCR, 131029, Sonepat, Haryana, India
| | - Ayaan Ahmad
- Laboratory of Infection Biology and Translational Research, Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| | - Vikram Saini
- Laboratory of Infection Biology and Translational Research, Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India.
- Biosafety Laboratory-3, Centralized Core Research Facility (CCRF), All India Institute of Medical Sciences (AIIMS), New Delhi, India.
| |
Collapse
|
29
|
Lim JZM, Burgess J, Ooi CG, Ponirakis G, Malik RA, Wilding JPH, Alam U. The Peripheral Neuropathy Prevalence and Characteristics Are Comparable in People with Obesity and Long-Duration Type 1 Diabetes. Adv Ther 2022; 39:4218-4229. [PMID: 35867275 PMCID: PMC9402741 DOI: 10.1007/s12325-022-02208-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 05/26/2022] [Indexed: 12/02/2022]
Abstract
INTRODUCTION Peripheral neuropathy is reported in obesity even in the absence of hyperglycaemia. OBJECTIVE To compare the prevalence and characterise the phenotype of peripheral neuropathy in people living with obesity (OB) and long-duration type 1 diabetes (T1D). PATIENTS AND METHODS We performed a prospective cross-sectional study of 130 participants including healthy volunteers (HV) (n = 28), people with T1D (n = 51), and OB (BMI 30-50 kg/m2) (n = 51). Participants underwent assessment of neuropathic symptoms (Neuropathy Symptom Profile, NSP), neurological deficits (Neuropathy Disability Score, NDS), vibration perception threshold (VPT) and evaluation of sural nerve conduction velocity and amplitude. RESULTS Peripheral neuropathy was present in 43.1% of people with T1D (age 49.9 ± 12.9 years; duration of diabetes 23.4 ± 13.5 years) and 33.3% of OB (age 48.2 ± 10.8 years). VPT for high risk of neuropathic foot ulceration (VPT ≥ 25 V) was present in 31.4% of T1D and 19.6% of OB. Participants living with OB were heavier (BMI 42.9 ± 3.5 kg/m2) and had greater centripetal adiposity with an increased body fat percentage (FM%) (P < 0.001) and waist circumference (WC) (P < 0.001) compared to T1D. The OB group had a higher NDS (P < 0.001), VAS for pain (P < 0.001), NSP (P < 0.001), VPT (P < 0.001) and reduced sural nerve conduction velocity (P < 0.001) and amplitude (P < 0.001) compared to HV, but these parameters were comparable in T1D. VPT was positively associated with increased WC (P = 0.011), FM% (P = 0.001) and HbA1c (P < 0.001) after adjusting for age (R2 = 0.547). Subgroup analysis of respiratory quotient (RQ) measured in the OB group did not correlate with VPT (P = 0.788), nerve conduction velocity (P = 0.743) or amplitude (P = 0.677). CONCLUSION The characteristics of peripheral neuropathy were comparable between normoglycaemic people living with obesity and people with long-duration T1D, suggesting that metabolic factors linked to obesity play a pivotal role in the development of peripheral neuropathy. Further studies are needed to investigate the mechanistic link between visceral adiposity and neuropathy.
Collapse
Affiliation(s)
- J Z M Lim
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- Division of Diabetes and Endocrinology, Aintree University Hospital, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - J Burgess
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - C G Ooi
- Division of Diabetes and Endocrinology, Aintree University Hospital, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - G Ponirakis
- Division of Medicine, Qatar Foundation, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - R A Malik
- Division of Medicine, Qatar Foundation, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - J P H Wilding
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- Division of Diabetes and Endocrinology, Aintree University Hospital, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Uazman Alam
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK.
- Division of Diabetes and Endocrinology, Aintree University Hospital, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK.
- Department of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
30
|
Food Reward Alterations during Obesity Are Associated with Inflammation in the Striatum in Mice: Beneficial Effects of Akkermansia muciniphila. Cells 2022; 11:cells11162534. [PMID: 36010611 PMCID: PMC9406832 DOI: 10.3390/cells11162534] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/06/2022] [Accepted: 08/09/2022] [Indexed: 11/17/2022] Open
Abstract
The reward system involved in hedonic food intake presents neuronal and behavioral dysregulations during obesity. Moreover, gut microbiota dysbiosis during obesity promotes low-grade inflammation in peripheral organs and in the brain contributing to metabolic alterations. The mechanisms underlying reward dysregulations during obesity remain unclear. We investigated if inflammation affects the striatum during obesity using a cohort of control-fed or diet-induced obese (DIO) male mice. We tested the potential effects of specific gut bacteria on the reward system during obesity by administrating Akkermansia muciniphila daily or a placebo to DIO male mice. We showed that dysregulations of the food reward are associated with inflammation and alterations in the blood–brain barrier in the striatum of obese mice. We identified Akkermansia muciniphila as a novel actor able to improve the dysregulated reward behaviors associated with obesity, potentially through a decreased activation of inflammatory pathways and lipid-sensing ability in the striatum. These results open a new field of research and suggest that gut microbes can be considered as an innovative therapeutic approach to attenuate reward alterations in obesity. This study provides substance for further investigations of Akkermansia muciniphila-mediated behavioral improvements in other inflammatory neuropsychiatric disorders.
Collapse
|
31
|
Near-infrared light reduces glia activation and modulates neuroinflammation in the brains of diet-induced obese mice. Sci Rep 2022; 12:10848. [PMID: 35761012 PMCID: PMC9237037 DOI: 10.1038/s41598-022-14812-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 06/13/2022] [Indexed: 12/06/2022] Open
Abstract
Neuroinflammation is a key event in neurodegenerative conditions such as Alzheimer's disease (AD) and characterizes metabolic pathologies like obesity and type 2 diabetes (T2D). Growing evidence in humans shows that obesity increases the risk of developing AD by threefold. Hippocampal neuroinflammation in rodents correlates with poor memory performance, suggesting that it contributes to cognitive decline. Here we propose that reducing obesity/T2D-driven neuroinflammation may prevent the progression of cognitive decline associated with AD-like neurodegenerative states. Near-infrared light (NIR) has attracted increasing attention as it was shown to improve learning and memory in both humans and animal models. We previously reported that transcranial NIR delivery reduced amyloid beta and Tau pathology and improved memory function in mouse models of AD. Here, we report the effects of NIR in preventing obesity-induced neuroinflammation in a diet-induced obese mouse model. Five-week-old wild-type mice were fed a high-fat diet (HFD) for 13 weeks to induce obesity prior to transcranial delivery of NIR for 4 weeks during 90-s sessions given 5 days a week. After sacrifice, brain slices were subjected to free-floating immunofluorescence for microglia and astrocyte markers to evaluate glial activation and quantitative real-time polymerase chain reaction (PCR) to evaluate expression levels of inflammatory cytokines and brain-derived neurotrophic factor (BDNF). The hippocampal and cortical regions of the HFD group had increased expression of the activated microglial marker CD68 and the astrocytic marker glial fibrillary acidic protein. NIR-treated HFD groups showed decreased levels of these markers. PCR revealed that hippocampal tissue from the HFD group had increased levels of pro-inflammatory interleukin (IL)-1β and tumor necrosis factor-α. Interestingly, the same samples showed increased levels of the anti-inflammatory IL-10. All these changes were attenuated by NIR treatment. Lastly, hippocampal levels of the neurotrophic factor BDNF were increased in NIR-treated HFD mice, compared to untreated HFD mice. The marked reductions in glial activation and pro-inflammatory cytokines along with elevated BDNF provide insights into how NIR could reduce neuroinflammation. These results support the use of NIR as a potential non-invasive and preventive therapeutic approach against chronic obesity-induced deficits that are known to occur with AD neuropathology.
Collapse
|
32
|
Dworzański W, Cholewińska E, Fotschki B, Juśkiewicz J, Ognik K. Oxidative, epigenetic changes and fermentation processes in the intestine of rats fed high-fat diets supplemented with various chromium forms. Sci Rep 2022; 12:9817. [PMID: 35701510 PMCID: PMC9198011 DOI: 10.1038/s41598-022-13328-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
The aim of the study was to determine how feeding rats a high-fat diet (F) supplemented with various forms of chromium affects the responses of the immune and redox systems, as well as epigenetic changes in the ileal tissue and the course of fermentation processes in the caecum. The rats received a pharmacologically relevant dose 0.3 mg Cr/kg body weight in form of chromium(III) picolinate (Cr-Pic), chromium (III)-methionine (Cr-Met), or chromium nanoparticles (Cr-NPs). The F increased DNA oxidation and raised the level of interleukin IL-6. The F was shown to reduce the intensity of fermentation processes in the caecum while increasing the activity of potentially harmful enzymes in the faeces. The addition of Cr in the form of Cr-NPs and Cr-Met in rats fed F beneficially increased mobilization of enzymes of the DNA repair pathway. All forms of Cr, but especially Cr-NPs, beneficially decreased the activity of caecal bacterial β-glucuronidase, faecal β-glucosidase and β-glucuronidase. However, due to the increase in level of cytokine IL-2 in small intestinal wall, induced by all tested forms of chromium, it is difficult to state conclusively that this element can mitigate unfavourable pro-inflammatory and oxidative changes induced by a F in the small intestinal wall.
Collapse
Affiliation(s)
- Wojciech Dworzański
- Chair and Department of Human Anatomy, Medical University of Lublin, Jaczewskiego 4, 20-090, Lublin, Poland
| | - Ewelina Cholewińska
- Department of Biochemistry and Toxicology, Faculty of Animal Sciences and Bioeconomy, University of Life Sciences in Lublin, Akademicka 13, 20-950, Lublin, Poland.
| | - Bartosz Fotschki
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Jerzy Juśkiewicz
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland.
| | - Katarzyna Ognik
- Department of Biochemistry and Toxicology, Faculty of Animal Sciences and Bioeconomy, University of Life Sciences in Lublin, Akademicka 13, 20-950, Lublin, Poland
| |
Collapse
|
33
|
Sambolín-Escobales L, Tirado-Castro L, Suarez C, Pacheco-Cruz D, Fonseca-Ferrer W, Deme P, Haughey N, Chompre G, Porter JT. High-Fat Diet and Short-Term Unpredictable Stress Increase Long-Chain Ceramides Without Enhancing Behavioral Despair. Front Mol Biosci 2022; 9:859760. [PMID: 35601829 PMCID: PMC9114865 DOI: 10.3389/fmolb.2022.859760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/22/2022] [Indexed: 11/28/2022] Open
Abstract
Clinical and preclinical studies suggest that increases in long-chain ceramides in blood may contribute to the development of depressive-like behavior. However, which factors contribute to these increases and whether the increases are sufficient to induce depressive-like behaviors is unclear. To begin to address this issue, we examined the effects of high fat diet (HFD) and short-term unpredictable (STU) stress on long-chain ceramides in the serum of male and female rats. We found that brief exposure to HFD or unpredictable stress was sufficient to induce selective increases in the serum concentrations of long-chain ceramides, associated with depression in people. Furthermore, combined exposure to HFD and unpredictable stress caused a synergistic increase in C16:0, C16:1, and C18:0 ceramides in both sexes and C18:1 and C24:1 in males. However, the increased peripheral long-chain ceramides were not associated with increases in depressive-like behaviors suggesting that increases in serum long-chain ceramides may not be associated with the development of depressive-like behaviors in rodents.
Collapse
Affiliation(s)
- Lubriel Sambolín-Escobales
- Division of Pharmacology, Basic Sciences Department, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| | - Lizmarie Tirado-Castro
- Division of Pharmacology, Basic Sciences Department, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| | - Cristina Suarez
- Division of Pharmacology, Basic Sciences Department, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| | - Dariangelly Pacheco-Cruz
- Biology and Biotechnology Department, Pontifical Catholic University of Puerto Rico, Ponce, Puerto Rico
| | | | - Pragney Deme
- Department of Neurology, John Hopkins University School of Medicine, Baltimore, MD, United States
| | - Norman Haughey
- Department of Neurology, John Hopkins University School of Medicine, Baltimore, MD, United States
| | - Gladys Chompre
- Biology and Biotechnology Department, Pontifical Catholic University of Puerto Rico, Ponce, Puerto Rico
| | - James T. Porter
- Division of Pharmacology, Basic Sciences Department, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
- *Correspondence: James T. Porter,
| |
Collapse
|
34
|
Lewandowski CT, Laham MS, Thatcher GR. Remembering your A, B, C's: Alzheimer's disease and ABCA1. Acta Pharm Sin B 2022; 12:995-1018. [PMID: 35530134 PMCID: PMC9072248 DOI: 10.1016/j.apsb.2022.01.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/27/2021] [Accepted: 01/07/2022] [Indexed: 12/24/2022] Open
Abstract
The function of ATP binding cassette protein A1 (ABCA1) is central to cholesterol mobilization. Reduced ABCA1 expression or activity is implicated in Alzheimer's disease (AD) and other disorders. Therapeutic approaches to boost ABCA1 activity have yet to be translated successfully to the clinic. The risk factors for AD development and progression, including comorbid disorders such as type 2 diabetes and cardiovascular disease, highlight the intersection of cholesterol transport and inflammation. Upregulation of ABCA1 can positively impact APOE lipidation, insulin sensitivity, peripheral vascular and blood–brain barrier integrity, and anti-inflammatory signaling. Various strategies towards ABCA1-boosting compounds have been described, with a bias toward nuclear hormone receptor (NHR) agonists. These agonists display beneficial preclinical effects; however, important side effects have limited development. In particular, ligands that bind liver X receptor (LXR), the primary NHR that controls ABCA1 expression, have shown positive effects in AD mouse models; however, lipogenesis and unwanted increases in triglyceride production are often observed. The longstanding approach, focusing on LXRβ vs. LXRα selectivity, is over-simplistic and has failed. Novel approaches such as phenotypic screening may lead to small molecule NHR modulators that elevate ABCA1 function without inducing lipogenesis and are clinically translatable.
Collapse
|
35
|
Church JS, Renzelman ML, Schwartzer JJ. Ten-week high fat and high sugar diets in mice alter gut-brain axis cytokines in a sex-dependent manner. J Nutr Biochem 2022; 100:108903. [PMID: 34748922 PMCID: PMC8761169 DOI: 10.1016/j.jnutbio.2021.108903] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 09/17/2021] [Accepted: 10/06/2021] [Indexed: 02/03/2023]
Abstract
Diets high in fat and sugar induce inflammation throughout the body, particularly along the gut-brain axis; however, the way these changes in immune signaling mediate one another remains unknown. We investigated cytokine changes in the brain and colon following prolonged high fat or sugar diet in female and male adult C57BL/6 mice. Ten weeks of high fat diet increased levels of TNFα, IL-1β, IL-6, IFNγ, and IL-10 in the female hippocampus and altered cytokines in the frontal cortex of both sexes. High sugar diet increased hippocampal cytokines and decreased cytokines in the diencephalon and frontal cortex. In the colon, high fat diet changed cytokine expression in both sexes, while high sugar diet only increased TNFα in males. Causal mediation analysis confirmed that colon IL-10 and IL-6 mediate high fat diet-induced neuroimmune changes in the female hippocampus and male frontal cortex. Additionally, high fat diet increased food consumption and weight gain in both sexes, while high sugar diet decreased male weight gain. These findings reveal a novel causal link between gut and brain inflammation specific to prolonged consumption of high fat, not high sugar, diet. Importantly, this work includes females which have been under-represented in diet research, and demonstrates that diet-induced neuroinflammation varies by brain region between sexes. Furthermore, our data suggest female brains are more vulnerable than males to inflammatory changes following excessive fat and sugar consumption, which may help explain the increased risk of inflammation-associated psychiatric conditions in women who eat a Western Diet rich in both dietary components.
Collapse
Affiliation(s)
- Jamie S. Church
- Program in Neuroscience and Behavior, Department of Psychology and Education, Mount Holyoke College, 50 College Street, South Hadley, MA 01075, USA
| | - Margaret L. Renzelman
- Program in Neuroscience and Behavior, Department of Psychology and Education, Mount Holyoke College, 50 College Street, South Hadley, MA 01075, USA
| | - Jared J. Schwartzer
- Program in Neuroscience and Behavior, Department of Psychology and Education, Mount Holyoke College, 50 College Street, South Hadley, MA 01075, USA
| |
Collapse
|
36
|
Wagner KM, Yang J, Morisseau C, Hammock BD. Soluble Epoxide Hydrolase Deletion Limits High-Fat Diet-Induced Inflammation. Front Pharmacol 2022; 12:778470. [PMID: 34975481 PMCID: PMC8719166 DOI: 10.3389/fphar.2021.778470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/08/2021] [Indexed: 01/28/2023] Open
Abstract
The soluble epoxide hydrolase (sEH) enzyme is a major regulator of bioactive lipids. The enzyme is highly expressed in liver and kidney and modulates levels of endogenous epoxy-fatty acids, which have pleiotropic biological effects including limiting inflammation, neuroinflammation, and hypertension. It has been hypothesized that inhibiting sEH has beneficial effects on limiting obesity and metabolic disease as well. There is a body of literature published on these effects, but typically only male subjects have been included. Here, we investigate the role of sEH in both male and female mice and use a global sEH knockout mouse model to compare the effects of diet and diet-induced obesity. The results demonstrate that sEH activity in the liver is modulated by high-fat diets more in male than in female mice. In addition, we characterized the sEH activity in high fat content tissues and demonstrated the influence of diet on levels of bioactive epoxy-fatty acids. The sEH KO animals had generally increased epoxy-fatty acids compared to wild-type mice but gained less body weight on higher-fat diets. Generally, proinflammatory prostaglandins and triglycerides were also lower in livers of sEH KO mice fed HFD. Thus, sEH activity, prostaglandins, and triglycerides increase in male mice on high-fat diet but are all limited by sEH ablation. Additionally, these changes also occur in female mice though at a different magnitude and are also improved by knockout of the sEH enzyme.
Collapse
Affiliation(s)
- Karen M Wagner
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA, United States
| | - Jun Yang
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA, United States
| | - Christophe Morisseau
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA, United States
| | - Bruce D Hammock
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA, United States
| |
Collapse
|
37
|
Bhusal A, Rahman MH, Suk K. Hypothalamic inflammation in metabolic disorders and aging. Cell Mol Life Sci 2021; 79:32. [PMID: 34910246 PMCID: PMC11071926 DOI: 10.1007/s00018-021-04019-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/01/2021] [Accepted: 10/29/2021] [Indexed: 12/15/2022]
Abstract
The hypothalamus is a critical brain region for the regulation of energy homeostasis. Over the years, studies on energy metabolism primarily focused on the neuronal component of the hypothalamus. Studies have recently uncovered the vital role of glial cells as an additional player in energy balance regulation. However, their inflammatory activation under metabolic stress condition contributes to various metabolic diseases. The recruitment of monocytes and macrophages in the hypothalamus helps sustain such inflammation and worsens the disease state. Neurons were found to actively participate in hypothalamic inflammatory response by transmitting signals to the surrounding non-neuronal cells. This activation of different cell types in the hypothalamus leads to chronic, low-grade inflammation, impairing energy balance and contributing to defective feeding habits, thermogenesis, and insulin and leptin signaling, eventually leading to metabolic disorders (i.e., diabetes, obesity, and hypertension). The hypothalamus is also responsible for the causation of systemic aging under metabolic stress. A better understanding of the multiple factors contributing to hypothalamic inflammation, the role of the different hypothalamic cells, and their crosstalks may help identify new therapeutic targets. In this review, we focus on the role of glial cells in establishing a cause-effect relationship between hypothalamic inflammation and the development of metabolic diseases. We also cover the role of other cell types and discuss the possibilities and challenges of targeting hypothalamic inflammation as a valid therapeutic approach.
Collapse
Affiliation(s)
- Anup Bhusal
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Md Habibur Rahman
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
- Division of Endocrinology, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA
| | - Kyoungho Suk
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea.
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea.
- Brain Science and Engineering Institute, Kyungpook National University, Daegu, 41944, Republic of Korea.
| |
Collapse
|
38
|
Ebrahim HA, El-Gamal R, Sherif RN. Intermittent Fasting Attenuates High Fat Diet-Induced Cerebellar Changes in Rats: Involvement of TNFα, autophagy and oxidative stress. Cells Tissues Organs 2021; 210:351-367. [PMID: 34551416 DOI: 10.1159/000519088] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 08/12/2021] [Indexed: 11/19/2022] Open
Affiliation(s)
- Hasnaa Ali Ebrahim
- Department of Basic Medical Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
- Department of Anatomy, Faculty of Medicine, University of Mansoura, Mansoura, Egypt
| | - Randa El-Gamal
- Department of Medical Biochemistry, Faculty of Medicine, University of Mansoura, Mansoura, Egypt
- Medical Experimental Research Center (MERC), Faculty of Medicine, University of Mansoura, Mansoura, Egypt
| | - Rania N Sherif
- Department of Anatomy, Faculty of Medicine, University of Mansoura, Mansoura, Egypt
- Department of Anatomy and Embryology, Faculty of Dentistry, Horus University, Damietta, Egypt
- Department of Anatomy and Embryology, Faculty of Medicine, Damietta University, Damietta, Egypt
| |
Collapse
|
39
|
Więckowska-Gacek A, Mietelska-Porowska A, Wydrych M, Wojda U. Western diet as a trigger of Alzheimer's disease: From metabolic syndrome and systemic inflammation to neuroinflammation and neurodegeneration. Ageing Res Rev 2021; 70:101397. [PMID: 34214643 DOI: 10.1016/j.arr.2021.101397] [Citation(s) in RCA: 143] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/10/2021] [Accepted: 06/24/2021] [Indexed: 02/06/2023]
Abstract
An excess of saturated fatty acids and simple sugars in the diet is a known environmental risk factor of Alzheimer's disease (AD) but the holistic view of the interacting processes through which such diet may contribute to AD pathogenesis is missing. We addressed this need through extensive analysis of published studies investigating the effects of western diet (WD) on AD development in humans and laboratory animals. We reviewed WD-induced systemic alterations comprising metabolic changes, induction of obesity and adipose tissue inflammation, gut microbiota dysbiosis and acceleration of systemic low-grade inflammation. Next we provide an overview of the evidence demonstrating that WD-associated systemic alterations drive impairment of the blood-brain barrier (BBB) and development of neuroinflammation paralleled by accumulation of toxic amyloid. Later these changes are followed by dysfunction of synaptic transmission, neurodegeneration and finally memory and cognitive impairment. We conclude that WD can trigger AD by acceleration of inflammaging, and that BBB impairment induced by metabolic and systemic inflammation play the central role in this process. Moreover, the concurrence of neuroinflammation and Aβ dyshomeostasis, which by reciprocal interactions drive the vicious cycle of neurodegeneration, contradicts Aβ as the primary trigger of AD. Given that in 2019 the World Health Organization recommended focusing on modifiable risk factors in AD prevention, this overview of the sequential, complex pathomechanisms initiated by WD, which can lead from peripheral disturbances to neurodegeneration, can support future prevention strategies.
Collapse
|
40
|
Hegab AE, Ozaki M, Kagawa S, Fukunaga K. Effect of High Fat Diet on the Severity and Repair of Lung Fibrosis in Mice. Stem Cells Dev 2021; 30:908-921. [PMID: 34269615 DOI: 10.1089/scd.2021.0050] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Lung fibrosis is a progressive fatal disease, and the underlying mechanisms remain unclear. These involve a combination of altered fibroblasts, excessive accumulation of extracellular matrix, inflammation, and aberrant activation of epithelial cells. Previously, we showed that high-fat diet (HFD) induces lung inflammation, aberrant activation of stem cells, and lung mitochondria impairment. Therefore, we hypothesized that HFD-induced changes would influence lung fibrosis. Mice were fed standard diet (SD) or HFD, administered bleomycin, then examined for fibrosis severity and the start of repair 3 weeks after injury, and for fibrosis repair/resolution 6-9 weeks after injury. At 3 weeks, no significant differences in inflammation and fibrosis severity were observed between SD- and HFD-fed mice. However, infiltration of alveolar type (AT)-2 cells and bronchioalveolar stem cells (BASCs) into the fibrotic areas (the start of repair) was impaired in HFD-fed mice. At 6 weeks, SD-fed mice showed near-complete resolution/repair of fibrosis and inflammation, while HFD-fed mice still showed residual fibrosis and inflammation. Infiltration of the fibrotic areas with AT2 cells was observed, but very few BASCs were detectable. At 9 weeks, mice from both groups showed complete resolution/repair of fibrosis and inflammation, indicating that HFD induced delayed, rather than failed, resolution of fibrosis and alveolar repair. To further confirm the direct role of enhanced fatty-acid oxidation (FAO) in delayed resolution/repair, we administered etomoxir, a FAO inhibitor, to HFD-fed mice for 3-6 weeks after bleomycin injury. Inhibition of FAO abolished the HFD-induced delay in alveolar repair and fibrosis resolution at both time points. In conclusion, after a fibrosis-inducing injury, HFD slows resolution of fibrosis/inflammation and delays alveolar repair by slowing the contribution of AT2 stem cells and abolishing the contribution of BASCs in the repair process. FAO activation appears to be involved in this delay mechanism; thus, inhibiting FAO may be useful in the treatment of lung injury and fibrosis.
Collapse
Affiliation(s)
- Ahmed E Hegab
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Shinjuku-ku, Japan.,Faculty of Medicine, Graduate School of Medicine, International University of Health and Welfare, Narita, Japan
| | - Mari Ozaki
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Shinjuku-ku, Japan
| | - Shizuko Kagawa
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Shinjuku-ku, Japan
| | - Koichi Fukunaga
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Shinjuku-ku, Japan
| |
Collapse
|
41
|
Yamada K, Watanabe M, Suzuki K, Suzuki Y. Cerebellar Volumes Associate with Behavioral Phenotypes in Prader-Willi Syndrome. THE CEREBELLUM 2021; 19:778-787. [PMID: 32661798 PMCID: PMC7588377 DOI: 10.1007/s12311-020-01163-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The objective of this study was to investigate lobule-specific cerebellar structural alterations relevant to clinical behavioral characteristics of Prader-Willi syndrome (PWS). We performed a case-control study of 21 Japanese individuals with PWS (age; median 21.0, range 13–50 years, 14 males, 7 females) and 40 age- and sex-matched healthy controls with typical development. Participants underwent 3-Tesla magnetic resonance imaging. Three-dimensional T1-weighted images were assessed for cerebellar lobular volume and adjusted for total intracerebellar volume (TIV) using a spatially unbiased atlas template to give a relative volume ratio. A region of interest analysis included the deep cerebellar nuclei. A correlation analysis was performed between the volumetric data and the clinical behavioral scores derived from the standard questionnaires (hyperphagia, autism, obsession, and maladaptive index) for global intelligence assessment in paired subgroups. In individuals with PWS, TIV was significantly reduced compared with that of controls (p < 0.05, family-wise error corrected; mean [standard deviation], 1014.1 [93.0] mm3). Decreased relative lobular volume ratios were observed in posterior inferior lobules with age, sex, and TIV as covariates (Crus I, Crus II, lobules VIIb, VIIIa, VIIIb, and IX). However, increased ratios were found in the dentate nuclei bilaterally in individuals with PWS (p < 0.01); the mean (standard deviation) × 10−3 was as follows: left, 1.58 (0.26); right, 1.67 (0.30). The altered lobular volume ratios showed negative correlations with hyperphagic and autistic characteristics and positive correlations with obsessive and intellectual characteristics. This study provides the first objective evidence of topographic patterns of volume differences in cerebellar structures consistent with clinical behavioral characteristics in individuals with PWS and strongly suggests a cerebellar contribution to altered functional brain connectivity in PWS.
Collapse
Affiliation(s)
- Kenichi Yamada
- Center for Integrated Human Brain Science, Brain Research Institute, University of Niigata, 1-757, Asahimachi, Chuo-ku, Niigata, 9518585, Japan.
| | - Masaki Watanabe
- Center for Integrated Human Brain Science, Brain Research Institute, University of Niigata, 1-757, Asahimachi, Chuo-ku, Niigata, 9518585, Japan
| | - Kiyotaka Suzuki
- Center for Integrated Human Brain Science, Brain Research Institute, University of Niigata, 1-757, Asahimachi, Chuo-ku, Niigata, 9518585, Japan
| | - Yuji Suzuki
- Center for Integrated Human Brain Science, Brain Research Institute, University of Niigata, 1-757, Asahimachi, Chuo-ku, Niigata, 9518585, Japan
| |
Collapse
|
42
|
Lam YY, Tsai SF, Chen PC, Kuo YM, Chen YW. Pioglitazone rescues high-fat diet-induced depression-like phenotypes and hippocampal astrocytic deficits in mice. Biomed Pharmacother 2021; 140:111734. [PMID: 34022606 DOI: 10.1016/j.biopha.2021.111734] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 10/21/2022] Open
Abstract
The prevalence of diabetes is rapidly increasing worldwide and is highly associated with the incidence of depression. Pioglitazone, a Peroxisome proliferator-activated receptor gamma (PPAR-γ) agonist, is widely used for treating patients with type 2 diabetes. However, whether pioglitazone alleviates metabolic disorder-related depression and astrocytic deficits remains unclear. Here we showed that 12 weeks of high-fat diet (HFD) feeding (from 8- to 20-week-old) induced not only obesity and insulin resistance, but also depression-like behaviors in mice. Astrocytic activation, a sign closely associated with depression, was also evident in the ventral hippocampus. Four weeks of pioglitazone (10 or 20 mg/kg, daily, from 20- to 24-week-old) treatment alleviated the HFD-induced glucose-metabolic dysfunctions, upregulation of ventral hippocampal GFAP, reduction of the total process lengths and the number of branch points of the ventral hippocampal CA1 GFAP-immunoreactive astrocytes and depressive phenotypes but had no effect on anxiety-like behaviors or hippocampus-related learning and memory in mice. These findings suggest that pioglitazone could be a potential therapeutic agent for metabolic disorders and associated depression.
Collapse
Affiliation(s)
- Ying-Yiu Lam
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Sheng-Feng Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Chun Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Min Kuo
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yun-Wen Chen
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
43
|
Bottemanne P, Guillemot-Legris O, Paquot A, Masquelier J, Malamas M, Makriyannis A, Alhouayek M, Muccioli GG. N-Acylethanolamine-Hydrolyzing Acid Amidase Inhibition, but Not Fatty Acid Amide Hydrolase Inhibition, Prevents the Development of Experimental Autoimmune Encephalomyelitis in Mice. Neurotherapeutics 2021; 18:1815-1833. [PMID: 34235639 PMCID: PMC8609003 DOI: 10.1007/s13311-021-01074-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2021] [Indexed: 02/06/2023] Open
Abstract
N-acylethanolamines (NAEs) are endogenous bioactive lipids reported to exert anti-inflammatory and neuroprotective effects mediated by cannabinoid receptors and peroxisome proliferator-activated receptors (PPARs), among others. Therefore, interfering with NAE signaling could be a promising strategy to decrease inflammation in neurological disorders such as multiple sclerosis (MS). Fatty acid amide hydrolase (FAAH) and N-acylethanolamine-hydrolyzing acid amidase (NAAA) are key modulators of NAE levels. This study aims to investigate and compare the effect of NAAA inhibition, FAAH inhibition, and dual inhibition of both enzymes in a mouse model of MS, namely the experimental autoimmune encephalomyelitis (EAE). Our data show that NAAA inhibition strongly decreased the hallmarks of the pathology. Interestingly, FAAH inhibition was less efficient in decreasing inflammatory hallmarks despite the increased NAE levels. Moreover, the inhibition of both NAAA and FAAH, using a dual-inhibitor or the co-administration of NAAA and FAAH inhibitors, did not show an added value compared to NAAA inhibition. Furthermore, our data suggest an important role of decreased activation of astrocytes and microglia in the effects of NAAA inhibition on EAE, while NAAA inhibition did not affect T cell recall. This work highlights the beneficial effects of NAAA inhibition in the context of central nervous system inflammation and suggests that the simultaneous inhibition of NAAA and FAAH has no additional beneficial effect in EAE.
Collapse
Affiliation(s)
- Pauline Bottemanne
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, B1.72.01, Av. E. Mounier 72, 1200, Bruxelles, Belgium
| | - Owein Guillemot-Legris
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, B1.72.01, Av. E. Mounier 72, 1200, Bruxelles, Belgium
| | - Adrien Paquot
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, B1.72.01, Av. E. Mounier 72, 1200, Bruxelles, Belgium
| | - Julien Masquelier
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, B1.72.01, Av. E. Mounier 72, 1200, Bruxelles, Belgium
| | - Michael Malamas
- Center for Drug Discovery and Departments of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, MA, 02115, USA
| | - Alexandros Makriyannis
- Center for Drug Discovery and Departments of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, MA, 02115, USA
| | - Mireille Alhouayek
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, B1.72.01, Av. E. Mounier 72, 1200, Bruxelles, Belgium
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, B1.72.01, Av. E. Mounier 72, 1200, Bruxelles, Belgium.
| |
Collapse
|
44
|
A high-fat diet, but not haloperidol or olanzapine administration, increases activated microglial expression in the rat brain. Neurosci Lett 2021; 757:135976. [PMID: 34023409 DOI: 10.1016/j.neulet.2021.135976] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 04/28/2021] [Accepted: 05/17/2021] [Indexed: 11/20/2022]
Abstract
This study examined the effects of chronic treatment of the antipsychotic drugs, haloperidol and olanzapine, on microglial activation in the brain. In addition, we explored the interaction of these antipsychotic drugs with normal and high-fat diet. In order to measure activated microglial expression, we used [3H] PK11195 in vitro autoradiography. Male Sprague Dawley rats were given a diet of either regular chow diet or a high-fat diet, and assigned either water, haloperidol drinking solution (1.5 mg/kg), or olanzapine drinking solution (10 mg/kg) for four weeks. Following treatment, rats were euthanized and brains extracted for [3H] PK11195 autoradiography. Rats on 4 weeks of a high-fat diet showed increased [3H] PK11195 binding compared to rats on a normal diet in the temporal association cortex (19 %), ectorhinal cortex (17 %), entorhinal cortex (18 %), and perirhinal cortex (18 %), irrespective of drug treatment. These are regions associated with memory, sensory, and visual processing. Rats treated with either haloperidol or olanzapine showed no differences in [3H] PK11195 binding compared to the control group. However, there were differences between the 2 different antipsychotic medications themselves. Haloperidol increased [3H] PK11195 binding in the amygdala (23 %), ectorhinal cortex (24 %), and perihinal cortex (29 %), compared to olanzapine. These results corroborate a known role of a high-fat diet and central inflammatory changes but suggest no role of these antipsychotic drugs in promoting neuroinflammation across 4 weeks compared to normal control rats.
Collapse
|
45
|
Banerjee A, Mukherjee S, Maji BK. Worldwide flavor enhancer monosodium glutamate combined with high lipid diet provokes metabolic alterations and systemic anomalies: An overview. Toxicol Rep 2021; 8:938-961. [PMID: 34026558 PMCID: PMC8120859 DOI: 10.1016/j.toxrep.2021.04.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 04/20/2021] [Accepted: 04/25/2021] [Indexed: 12/13/2022] Open
Abstract
Flavor enhancing high lipid diet acts as silent killer. Monosodium glutamate mixed with high lipid diet alters redox-status. Monosodium glutamate mixed with high lipid diet induces systemic anomalies.
In this fast-food era, people depend on ready-made foods and engage in minimal physical activities that ultimately change their food habits. Majorities of such foods have harmful effects on human health due to higher percentages of saturated fatty acids, trans-fatty acids, and hydrogenated fats in the form of high lipid diet (HLD). Moreover, food manufacturers add monosodium glutamate (MSG) to enhance the taste and palatability of the HLD. Both MSG and HLD induce the generation of reactive oxygen species (ROS) and thereby alter the redox-homeostasis to cause systemic damage. However, MSG mixed HLD (MH) consumption leads to dyslipidemia, silently develops non-alcoholic fatty liver disease followed by metabolic alterations and systemic anomalies, even malignancies, via modulating different signaling pathways. This comprehensive review formulates health care strategies to create global awareness about the harmful impact of MH on the human body and recommends the daily consumption of more natural foods rich in antioxidants instead of toxic ingredients to counterbalance the MH-induced systemic anomalies.
Collapse
|
46
|
Leocádio PCL, Lopes SC, Dias RP, Alvarez-Leite JI, Guerrant RL, Malva JO, Oriá RB. The Transition From Undernutrition to Overnutrition Under Adverse Environments and Poverty: The Risk for Chronic Diseases. Front Nutr 2021; 8:676044. [PMID: 33968973 PMCID: PMC8102690 DOI: 10.3389/fnut.2021.676044] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 03/26/2021] [Indexed: 12/29/2022] Open
Affiliation(s)
- Paola Caroline L Leocádio
- Laboratory of Atherosclerosis and Nutritional Biochemistry, Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Brazil.,Department of Nutrition, Nursing School, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Synara C Lopes
- Laboratory of Tissue Healing, Ontogeny, and Nutrition, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Ronaldo P Dias
- Laboratory of Tissue Healing, Ontogeny, and Nutrition, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Jacqueline I Alvarez-Leite
- Laboratory of Atherosclerosis and Nutritional Biochemistry, Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Richard L Guerrant
- Center for Global Health, University of Virginia, Charlottesville, VA, United States
| | - João O Malva
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - Reinaldo B Oriá
- Laboratory of Tissue Healing, Ontogeny, and Nutrition, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| |
Collapse
|
47
|
Syarif, Rasyid H, Aman M, Lawrence GS. High-fat diet increases the level of circulating Monocyte Chemoattractant Protein-1 in Wistar rats, independent of obesity. Ann Med Surg (Lond) 2021; 65:102266. [PMID: 33868686 PMCID: PMC8047162 DOI: 10.1016/j.amsu.2021.102266] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/24/2021] [Accepted: 03/24/2021] [Indexed: 11/26/2022] Open
Abstract
Introduction Low-grade chronic inflammation has emerged as a key pathogenic link between high-fat diet (HFD)-induced obesity and the increased risk of chronic diseases. Evidence has shown that HFDs may induce inflammation in the central nervous system and peripheral tissues. Monocyte Chemoattractant Protein-1 (MCP-1) is a product of various cells that is known to be an inflammatory marker. This study investigated whether a HFD could induce obesity and increase the level of MCP-1 in Wistar rats. Methods The Wistar rats were randomized into two groups: normal diet (ND) and HFD (n = 12 per group). Both groups were fed for 8 and 16 weeks, thus dividing the rats into 4 arms: ND8, ND16, HFD8, and HFD16 (n = 6 per sub-group). Obesity in rats was measured using the Lee index. Blood samples were taken to measure the level of MCP-1. Results Our results showed that obesity did not occur in the group with a normal diet (ND8 and ND16). However, in the HFD group (HFD8 and HFD16), 4 of the 6 rats became obese. However, MCP-1 was significantly higher among non-obese rats in the HFD group compared with the ND group (p < 0.001). Conclusion HFDs have been shown to increase the risk of obesity. In addition, increases in circulating MCP-1 were significantly different between Wistar rats given a HFD compared with the ND group.
Collapse
Affiliation(s)
- Syarif
- Division of Urology, Department of Surgery, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Haerani Rasyid
- Department of Internal Medicine, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Makbul Aman
- Department of Internal Medicine, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Gatot S Lawrence
- Department of Pathology Anatomy, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| |
Collapse
|
48
|
Mapping of Microglial Brain Region, Sex and Age Heterogeneity in Obesity. Int J Mol Sci 2021; 22:ijms22063141. [PMID: 33808700 PMCID: PMC8003547 DOI: 10.3390/ijms22063141] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 12/27/2022] Open
Abstract
The prevalence of obesity has increased rapidly in recent years and has put a huge burden on healthcare worldwide. Obesity is associated with an increased risk for many comorbidities, such as cardiovascular diseases, type 2 diabetes and hypertension. The hypothalamus is a key brain region involved in the regulation of food intake and energy expenditure. Research on experimental animals has shown neuronal loss, as well as microglial activation in the hypothalamus, due to dietary-induced obesity. Microglia, the resident immune cells in the brain, are responsible for maintaining the brain homeostasis and, thus, providing an optimal environment for neuronal function. Interestingly, in obesity, microglial cells not only get activated in the hypothalamus but in other brain regions as well. Obesity is also highly associated with changes in hippocampal function, which could ultimately result in cognitive decline and dementia. Moreover, changes have also been reported in the striatum and cortex. Microglial heterogeneity is still poorly understood, not only in the context of brain region but, also, age and sex. This review will provide an overview of the currently available data on the phenotypic differences of microglial innate immunity in obesity, dependent on brain region, sex and age.
Collapse
|
49
|
Cloos AS, Daenen LGM, Maja M, Stommen A, Vanderroost J, Van Der Smissen P, Rab M, Westerink J, Mignolet E, Larondelle Y, Terrasi R, Muccioli GG, Dumitru AC, Alsteens D, van Wijk R, Tyteca D. Impaired Cytoskeletal and Membrane Biophysical Properties of Acanthocytes in Hypobetalipoproteinemia - A Case Study. Front Physiol 2021; 12:638027. [PMID: 33708142 PMCID: PMC7940373 DOI: 10.3389/fphys.2021.638027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/25/2021] [Indexed: 12/03/2022] Open
Abstract
Familial hypobetalipoproteinemia is a metabolic disorder mainly caused by mutations in the apolipoprotein B gene. In its homozygous form it can lead without treatment to severe ophthalmological and neurological manifestations. In contrast, the heterozygous form is generally asymptomatic but associated with a low risk of cardiovascular disease. Acanthocytes or thorny red blood cells (RBCs) are described for both forms of the disease. However, those morphological changes are poorly characterized and their potential consequences for RBC functionality are not understood. Thus, in the present study, we asked whether, to what extent and how acanthocytes from a patient with heterozygous familial hypobetalipoproteinemia could exhibit altered RBC functionality. Acanthocytes represented 50% of the total RBC population and contained mitoTracker-positive surface patches, indicating the presence of mitochondrial fragments. While RBC osmotic fragility, calcium content and ATP homeostasis were preserved, a slight decrease of RBC deformability combined with an increase of intracellular free reactive oxygen species were observed. The spectrin cytoskeleton was altered, showing a lower density and an enrichment in patches. At the membrane level, no obvious modification of the RBC membrane fatty acids nor of the cholesterol content were detected but the ceramide species were all increased. Membrane stiffness and curvature were also increased whereas transversal asymmetry was preserved. In contrast, lateral asymmetry was highly impaired showing: (i) increased abundance and decreased functionality of sphingomyelin-enriched domains; (ii) cholesterol enrichment in spicules; and (iii) ceramide enrichment in patches. We propose that oxidative stress induces cytoskeletal alterations, leading to increased membrane stiffness and curvature and impaired lipid lateral distribution in domains and spicules. In addition, ceramide- and spectrin-enriched patches could result from a RBC maturation defect. Altogether, the data indicate that acanthocytes are associated with cytoskeletal and membrane lipid lateral asymmetry alterations, while deformability is only mildly impaired. In addition, familial hypobetalipoproteinemia might also affect RBC precursors leading to disturbed RBC maturation. This study paves the way for the potential use of membrane biophysics and lipid vital imaging as new methods for diagnosis of RBC disorders.
Collapse
Affiliation(s)
- Anne-Sophie Cloos
- CELL Unit & PICT Imaging Platform, de Duve Institute, UCLouvain, Brussels, Belgium
| | - Laura G M Daenen
- Department of Hematology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Mauriane Maja
- CELL Unit & PICT Imaging Platform, de Duve Institute, UCLouvain, Brussels, Belgium
| | - Amaury Stommen
- CELL Unit & PICT Imaging Platform, de Duve Institute, UCLouvain, Brussels, Belgium
| | - Juliette Vanderroost
- CELL Unit & PICT Imaging Platform, de Duve Institute, UCLouvain, Brussels, Belgium
| | | | - Minke Rab
- Central Diagnostic Laboratory - Research, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Jan Westerink
- Department of Vascular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Eric Mignolet
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Ottignies-Louvain-la-Neuve, Belgium
| | - Yvan Larondelle
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Ottignies-Louvain-la-Neuve, Belgium
| | - Romano Terrasi
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Brussels, Belgium
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Brussels, Belgium
| | - Andra C Dumitru
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Ottignies-Louvain-la-Neuve, Belgium
| | - David Alsteens
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Ottignies-Louvain-la-Neuve, Belgium
| | - Richard van Wijk
- Central Diagnostic Laboratory - Research, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Donatienne Tyteca
- CELL Unit & PICT Imaging Platform, de Duve Institute, UCLouvain, Brussels, Belgium
| |
Collapse
|
50
|
Swimming exercise improves gene expression of PPAR-γ and downregulates the overexpression of TLR4, MyD88, IL-6, and TNF-α after high-fat diet in rat skeletal muscle cells. Gene 2021; 775:145441. [PMID: 33482280 DOI: 10.1016/j.gene.2021.145441] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 12/26/2020] [Accepted: 01/13/2021] [Indexed: 02/07/2023]
Abstract
Exercise training with anti-inflammatory effects can improve insulin sensitivity in muscle tissue. This study investigated the effects of eight-week swimming exercises on lipid profile, toll-like receptor 4 (TLR4), myeloid differentiation factor 88 (MyD88), tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and peroxisome proliferator-activated receptor gamma (PPAR-γ) in gastrocnemius muscle of rats fed with high-fat diet (HFD). Thirty-two healthy male Wistar rats (8 weeks, 200 ± 20 g) were randomly divided into four groups (n = 8 each group): the control (C), aerobic exercise (E), HFD, and HFD + aerobic exercise (HFD & E). The exercise training protocol consisted of swimming 60 min/day, 5 days/week for eight weeks. Serum levels of glucose, insulin, and lipid profile were measured at end of the study. Protein expressions of TLR4, TNF-α, and IL-6 were determined by immunohistochemical method. Gene expression of TLR4/MyD88, TNF-α, IL-6, and PPAR-γ was evaluated by a real-time polymerase chain reaction in gastrocnemius muscle. HFD fed rats showed higher levels of cholesterol and LDL-c that were similar in weight gain. Meanwhile, the HFD group had a higher gene expression of TLR4, MyD88, TNF-α, IL-6, and lower gene expression of PPAR-γ compared to the control group (p < 0.05). Muscle protein expression of TLR4, TNF-α, IL-6 was lower in the E and HFD&E groups (especially when compared to HFD group, P < 0.05). We also showed a decrease in TLR4/MyD88 mRNA and an increase in PPAR-γ mRNA in gastrocnemius of E and HFD&E groups (compared to HFD group, p < 0.05). Insulin resistance in HFD&E groups show a significant decrease compared to the HFD group (p < 0.05). It seems that swimming aerobic exercise for eight weeks controlled the destructive effects of HFD on muscle inflammatory pathways along with the down-regulation of the TLR4/MyD88, inflammatory cytokine, and up-regulation PPAR-γ mRNA. It appears that the down-regulation in the expression of TLR4/MyD88 mRNA reduces the muscle pro-inflammatory cytokines, such as IL-6 and TNF-α, whose action may be caused by the adaptation of swimming aerobic exercise (an increase of PPAR-γ). Therefore, local and systemic inflammatory changes due to HFD and obesity may be affected by metabolic adaptations of aerobic exercise training, which requires further studies.
Collapse
|