1
|
Song M, Bai Y, Song F. High-fat diet and neuroinflammation: The role of mitochondria. Pharmacol Res 2025; 212:107615. [PMID: 39842474 DOI: 10.1016/j.phrs.2025.107615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 12/28/2024] [Accepted: 01/17/2025] [Indexed: 01/24/2025]
Abstract
In recent years, increasing evidence has supported that high-fat diet (HFD) can induce the chronic, low-grade neuroinflammation in the brain, which is closely associated with the impairment of cognitive function. As the key organelles responsible for energy metabolism in the cell, mitochondria are believed to involved in the pathogenesis of a variety of neurological disorders. This review summarizes the current progress in the field of the relationship between HFD exposure and neurodegenerative diseases, and outline the major routines of HFD induced neuroinflammation and its pathological significance in the pathogenesis of neurodegenerative diseases. Furthermore, the article highlights the pivotal role of mitochondrial dysfunction in driving the neuroinflammation in the setting of HFD. Danger-associated molecular patterns (DAMPs) from damaged mitochondria can activate innate immune signaling pathways, while mitochondrial dysfunction itself can lead to metabolic remodeling of inflammatory cells, thus inducing neuroinflammation. More importantly, mitochondrial damage, neuroinflammation, and insulin resistance caused by HFD form a mutually reinforcing vicious cycle, ultimately leading to the death of neurons and promoting the progression of neurodegenerative diseases. Thus, in-depth elucidation of the role and underlying mechanisms of mitochondrial dysfunction in HFD-induced metabolic disorders may not only expand our understanding of the mechanistic linkages between HFD and etiology of neurodegenerative diseases, but also help develop the specific strategies for the prevention and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Mingxue Song
- Department of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, China.
| | - Yao Bai
- NHC Key Laboratory of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing 100021, China.
| | - Fuyong Song
- Department of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, China.
| |
Collapse
|
2
|
Godos J, Lanza G, Ferri R, Caraci F, Cano SS, Elio I, Micek A, Castellano S, Grosso G. Relation between dietary inflammatory potential and sleep features: Systematic review of observational studies. MEDITERRANEAN JOURNAL OF NUTRITION AND METABOLISM 2024; 17:1-14. [DOI: 10.3233/mnm-240008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/26/2024]
Abstract
BACKGROUND: Diet and sleep represent key behavioral risk factors for major non-communicable diseases. Dietary factors may modulate systemic inflammation and potentially affect the human brain functionality, finally affecting the sleep-wake cycle. In this context, the Dietary Inflammatory Index (DII) has been studied as a tool to investigate the dietary inflammatory potential and its relationship with a variety of health conditions. OBJECTIVE: The aim of this study was to perform a systematic revision of observational studies on the inflammatory potential of the diet assessed through the DII and sleep features. METHODS: A systematic search of observational studies on DII and sleep features was conducted on Pubmed and EMBASE electronic databases. The Meta-analyses Of Observational Studies in Epidemiology (MOOSE) guidelines were used to plan the methodology. Studies identified through the search were selected according to inclusion/exclusion criteria. RESULTS: The systematic search and review led to the selection of 13 studies. Most studies including the general population reported an association between higher inflammatory potential of the diet and worse sleep quality and suboptimal sleep duration. Some studies also reported an association with daytime sleepiness and sleep apnea. However, some studies including younger individuals (i.e., college students or young employees) reported null findings. Four studies conducted in individuals with a variety of health conditions also showed some relation between DII and sleep features, although results were not fully consistent. CONCLUSIONS: Data published so far suggests an association between DII and sleep features. However, lack of randomized clinical trials and better designed observational studies, imply that further research on this matter is warranted to understand whether a causal relation could explain current findings.
Collapse
Affiliation(s)
- Justyna Godos
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Giuseppe Lanza
- Department of Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
- Clinical Neurophysiology Research Unit, Oasi Research Institute-IRCCS, Troina, Italy
| | - Raffaele Ferri
- Sleep Research Centre, Oasi Research Institute-IRCCS, Troina, Italy
| | - Filippo Caraci
- Department of Drug and Health Sciences, Universityof Catania, Catania, Italy
- Neuropharmacology and Translational Neurosciences Research Unit, Oasi Research Institute-IRCCS, Troina, Italy
| | - Sandra Sumalla Cano
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres, Santander, Spain
- Universidad Internacional Iberoamericana, Campeche, México
- Universidad de La Romana, LaRomana, República Dominicana
| | - Iñaki Elio
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres, Santander, Spain
- Universidad Internacional Iberoamericana, Campeche, México
- Fundación Universitaria Internacional de Colombia, Bogotá, Colombia
| | - Agnieszka Micek
- Statistical Laboratory, Faculty of Health Sciences, Jagiellonian University Medical College, Kraków, Poland
| | - Sabrina Castellano
- Department of Educational Sciences, University of Catania, Catania, Italy
| | - Giuseppe Grosso
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
- Center for Human Nutrition and Mediterranean Foods (NUTREA), University of Catania, Catania, Italy
| |
Collapse
|
3
|
Abbasi K, Zarezadeh R, Valizadeh A, Mehdizadeh A, Hamishehkar H, Nouri M, Darabi M. White-brown adipose tissue interplay in polycystic ovary syndrome: Therapeutic avenues. Biochem Pharmacol 2024; 220:116012. [PMID: 38159686 DOI: 10.1016/j.bcp.2023.116012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024]
Abstract
This study highlights the therapeutic potential of activating brown adipose tissue (BAT) for managing polycystic ovary syndrome (PCOS), a prevalent endocrine disorder associated with metabolic and reproductive abnormalities. BAT plays a crucial role in regulating energy expenditure and systemic insulin sensitivity, making it an attractive target for the treatment of obesity and metabolic diseases. Recent research suggests that impaired BAT function and mass may contribute to the link between metabolic disturbances and reproductive issues in PCOS. Additionally, abnormal white adipose tissue (WAT) can exacerbate these conditions by releasing adipokines and nonesterified fatty acids. In this review, we explored the impact of WAT changes on BAT function in PCOS and discussed the potential of BAT activation as a therapeutic strategy to improve PCOS symptoms. We propose that BAT activation holds promise for managing PCOS; however, further research is needed to confirm its efficacy and to develop clinically feasible methods for BAT activation.
Collapse
Affiliation(s)
- Khadijeh Abbasi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Zarezadeh
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Valizadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Hamishehkar
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Masoud Darabi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Division of Experimental Oncology, Department of Hematology and Oncology, University Medical Center Schleswig-Holstein, Campus Lübeck, Germany.
| |
Collapse
|
4
|
Liu Y, Guo YF, Peng H, Zhou HY, Su T, Yang M, Guo Q, Ye X, Huang Y, Jiang TJ. Hypothalamic Hnscr regulates glucose balance by mediating central inflammation and insulin signal. Cell Prolif 2023; 56:e13332. [PMID: 36042571 DOI: 10.1111/cpr.13332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/15/2022] [Accepted: 08/17/2022] [Indexed: 01/28/2023] Open
Abstract
OBJECTIVES Hypothalamic dysfunction leads to glucose metabolic imbalance; however, the mechanisms still need clarification. Our current study was to explore the role of hypothalamic Hnscr in glucose metabolism. MATERIALS AND METHODS Using Hnscr knockout or htNSC-specific Hnscr overexpression mice, we evaluated the effects of Hnscr on glucose metabolism through GTTs, ITTs, serum indicator measurements, etc. Immunofluorescence staining and Western blotting were performed to test inflammation levels and insulin signalling in hypothalamus. Conditioned medium intervene were used to investigate the effects of htNSCs on neuronal cell line. We also detected the glucose metabolism of mice with htNSCs implantation. RESULTS Hnscr expression decreased in the hypothalamus after high-fat diet feed. Hnscr-null mice displayed aggravated systematic insulin resistance, while mice with htNSC-specific Hnscr overexpression had the opposite phenotype. Notably, Hnscr-null mice had increased NF-κB signal in htNSCs, along with enhanced inflammation and damaged insulin signal in neurons located in arcuate nucleus of hypothalamus. The secretions, including sEVs, of Hnscr-deficient htNSCs mediated the detrimental effects on the CNS cell line. Locally implantation with Hnscr-depleted htNSCs disrupted glucose homeostasis. CONCLUSIONS This study demonstrated that decreased Hnscr in htNSCs led to systematic glucose imbalance through activating NF-κB signal and dampening insulin signal in hypothalamic neurons.
Collapse
Affiliation(s)
- Ya Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yi-Fan Guo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Hui Peng
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Hai-Yan Zhou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Tian Su
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Mi Yang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Qi Guo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xiao Ye
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yan Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Tie-Jian Jiang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
5
|
Hydrogen Sulfide Attenuates High-Fat Diet-Induced Obesity: Involvement of mTOR/IKK/NF-κB Signaling Pathway. Mol Neurobiol 2022; 59:6903-6917. [PMID: 36053437 DOI: 10.1007/s12035-022-03004-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 08/16/2022] [Indexed: 10/14/2022]
Abstract
Obesity has become a public health epidemic worldwide and is associated with many diseases with high mortality including hypertension, diabetes, and heart disease. High-fat diet (HFD)-induced energy imbalance is one of the primary causes of obesity, but the underlying mechanisms are not fully elucidated. Our study showed that HFD reduced the level of hydrogen sulfide (H2S) and its catalytic enzyme cystathionine β-synthase (CBS) in mouse hypothalamus and plasma. We found that HFD activated mTOR, IKK/NF-κB, the main pathway regulating inflammation. Activation of inflammatory pathway promoted the production of pro-inflammatory cytokines including IL-6, IL-1β, and TNF-α, which caused cell damage and loss in the hypothalamus. The disturbance of the hypothalamic neuron circuits resulted in body weight gain in HFD-induced mice. Importantly, we also showed that restoration of H2S level with NaHS or activation of CBS with SAMe attenuated HFD-induced activation of mTOR, IKK/NF-κB signaling, which reduced the inflammation and the neuronal cell loss in the hypothalamus, and also inhibited body weight gain in mice. The same effects were obtained by inhibiting mTOR or NF-κB, which suggested that mTOR and NF-κB were the critical molecular factors involved in hypothalamic inflammation. Taken together, this study identified that HFD-induced hypothalamus inflammation plays a critical role in the development of obesity. Moreover, the inhibition of hypothalamic inflammation by regaining H2S level could be a potential therapeutic to prevent the development of obesity.
Collapse
|
6
|
Shen B, Hernandez DG, Chitrala KN, Fanelli-Kuczmarski MT, Noren Hooten N, Pacheco NL, Mode NA, Zonderman AB, Ezike N, Evans MK. APOE gene region methylation is associated with cognitive performance in middle-aged urban adults. Neurobiol Aging 2022; 116:41-48. [PMID: 35561457 PMCID: PMC10878469 DOI: 10.1016/j.neurobiolaging.2022.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 11/17/2022]
Abstract
Apolipoprotein (APOE) ε4 allele is a strong risk factor for Alzheimer's disease (AD) and cognitive decline. Epigenetic modifications such as DNA methylation (DNAm) play a central role in cognition. This study sought to identify DNAm sites in the APOE genomic region associated with cognitive performance in a racially diverse middle-aged cohort (n = 411). Cognitive performance was measured by 11 standard neuropsychological tests. Two CpG sites were associated with the Card Rotation and Benton Visual Retention cognitive tests. The methylation level of the CpG site cg00397545 was associated with Card Rotation Test score (p = 0.000177) and a novel CpG site cg10178308 was associated with Benton Visual Retention Test score (p = 0.000084). Significant associations were observed among the dietary inflammatory index, which reflects the inflammatory potential of the diet, cognitive performance and the methylation level of several CpG sites. Our results indicate that DNAm in the APOE genomic area is correlated with cognitive performance and may presage cognitive decline.
Collapse
Affiliation(s)
- Botong Shen
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Dena G Hernandez
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Kumaraswamy Naidu Chitrala
- Fels Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Marie T Fanelli-Kuczmarski
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Natasha L Pacheco
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Nicolle A Mode
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Ngozi Ezike
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Michele K Evans
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|
7
|
Casagrande BP, Bueno AA, Pisani LP, Estadella D. Hepatic glycogen participates in the regulation of hypothalamic pAkt/Akt ratio in high-sugar/high-fat diet-induced obesity. Metab Brain Dis 2022; 37:1423-1434. [PMID: 35316448 DOI: 10.1007/s11011-022-00944-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/22/2022] [Indexed: 01/07/2023]
Abstract
The hypothalamus is a major integrating centre that controls energy homeostasis and plays a major role in hepatic glycogen (HGlyc) turnover. Not only do hypothalamic and hepatic Akt levels influence glucose homeostasis and glycogen synthesis, but exposure to high-sugar/high-fat diets (HSHF) can also lead to hypothalamic inflammation and HGlyc accumulation. HSHF withdrawal overall restores energy and glucose homeostasis, but the actual relationship between hypothalamic inflammation and HGlyc after short-term HSHF withdrawal has not yet been fully elucidated. Here we investigated the short-term effects of HSHF withdrawal preceded by a 30-day HSHF intake on the liver-hypothalamus crosstalk and glucose homeostasis. Sixty-day old male Wistar rats were fed for 30 days a control chow (n = 10) (Ct), or an HSHF diet (n = 20). On the 30th day of dietary intervention, a random HSHF subset (n = 10) had their diets switched to control chow for 48 h (Hw) whilst the remaining HSHF rats remained in the HSHF diet (n = 10) (Hd). All rats were anaesthetized and euthanized at the end of the protocol. We quantified HGlyc, Akt phosphorylation, inflammation and glucose homeostasis biomarkers. We also assessed the effect of propensity to obesity on those biomarkers, as detailed previously. Hd rats showed impaired glucose homeostasis, higher HGlyc and hypothalamic inflammation, and lower pAkt/Akt. Increased HGlyc was significantly associated with HSHF intake on pAkt/Akt lowered levels. We also found that HGlyc breakdown may have prevented a further pAkt/Akt drop after HSHF withdrawal. Propensity to obesity showed no apparent effect on hypothalamic inflammation or glucose homeostasis. Our findings suggest a comprehensive role of HGlyc as a structural and functional modulator of energy metabolism, and such roles may come into play relatively rapidly.
Collapse
Affiliation(s)
- Breno P Casagrande
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo-UNIFESP/BS, 1015-020, Santos, São Paulo, Brazil
| | - Allain A Bueno
- College of Health, Life and Environmental Sciences, University of Worcester, Henwick Grove, WR2 6AJ, Worcester, United Kingdom
| | - Luciana P Pisani
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo-UNIFESP/BS, 1015-020, Santos, São Paulo, Brazil
| | - Debora Estadella
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo-UNIFESP/BS, 1015-020, Santos, São Paulo, Brazil.
| |
Collapse
|
8
|
Depression and obesity among females, are sex specificities considered? Arch Womens Ment Health 2021; 24:851-866. [PMID: 33880649 DOI: 10.1007/s00737-021-01123-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 03/21/2021] [Indexed: 12/13/2022]
Abstract
This study aimed to systematically review the relationship of obesity-depression in the female sex. We carried out a systematic search (PubMed, MEDLINE, Embase) to quantify the articles (controlled trials and randomized controlled trials) regarding obesity and depression on a female population or a mixed sample. Successively, we established whether the sex specificities were studied by the authors and if they reported on collecting data regarding factors that may contribute to the evolution of obesity and depression and that could be responsible for the greater susceptibility of females to those conditions. After applying the inclusion and exclusion criteria, we found a total of 20 articles with a female sample and 54 articles with a mixed sample. More than half of all articles (51.35%, n = 38) evaluated the relationship between depression and obesity, but only 20 (27.03%) evaluated this relationship among females; still, 80% of those (n = 16) presented supporting results. However, few articles considered confounding factors related to female hormones (12.16%, n = 9) and none of the articles focused on factors responsible for the binomial obesity-depression in the female sex. The resulting articles also supported that depression (and related impairments) influencing obesity (and related impairments) is a two-way road. This systematic review supports the concurrency of obesity-depression in females but also shows how sex specificities are ultimately under-investigated. Female sex specificity is not being actively considered when studying the binomial obesity-depression, even within a female sample. Future studies should focus on trying to understand how the female sex and normal hormonal variations influence these conditions.
Collapse
|
9
|
Tran A, He W, Chen JTC, Wellhauser L, Hopperton KE, Bazinet RP, Belsham DD. Palmitate-mediated induction of neuropeptide Y expression occurs through intracellular metabolites and not direct exposure to proinflammatory cytokines. J Neurochem 2021; 159:574-589. [PMID: 34482548 DOI: 10.1111/jnc.15504] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 08/19/2021] [Accepted: 08/28/2021] [Indexed: 12/11/2022]
Abstract
A contributing factor to the development of obesity is the consumption of a diet high in saturated fatty acids, such as palmitate. These fats induce hypothalamic neuroinflammation, which dysregulates neuronal function and induces orexigenic neuropeptide Y (Npy) to promote food intake. An inflammatory cytokine array identified multiple candidates that could mediate palmitate-induced up-regulation of Npy mRNA levels. Of these, visfatin or nicotinamide phosphoribosyltransferase (NAMPT), macrophage migratory inhibitory factor (MIF), and IL-17F were chosen for further study. Direct treatment of the neuropeptide Y/agouti-related peptide (NPY/AgRP)-expressing mHypoE-46 neuronal cell line with the aforementioned cytokines demonstrated that visfatin could directly induce Npy mRNA expression. Preventing the intracellular metabolism of palmitate through long-chain acyl-CoA synthetase (ACSL) inhibition was sufficient to block the palmitate-mediated increase in Npy gene expression. Furthermore, thin-layer chromatography revealed that in neurons, palmitate is readily incorporated into ceramides and defined species of phospholipids. Exogenous C16 ceramide, dipalmitoyl-phosphatidylcholine, and dipalmitoyl-phosphatidylethanolamine were sufficient to significantly induce Npy expression. This study suggests that the intracellular metabolism of palmitate and elevation of metabolites, including ceramide and phospholipids, are responsible for the palmitate-mediated induction of the potent orexigen Npy. Furthermore, this suggests that the regulation of Npy expression is less reliant on inflammatory cytokines per se than palmitate metabolites in a model of NPY/AgRP neurons. These lipid species likely induce detrimental downstream cellular signaling events ultimately causing an increase in feeding, resulting in an overweight phenotype and/or obesity.
Collapse
Affiliation(s)
- Andy Tran
- Department of Physiology, University of Toronto, Ontario, Canada
| | - Wenyuan He
- Department of Physiology, University of Toronto, Ontario, Canada
| | - Jim T C Chen
- Department of Physiology, University of Toronto, Ontario, Canada
| | - Leigh Wellhauser
- Department of Physiology, University of Toronto, Ontario, Canada
| | | | | | - Denise D Belsham
- Department of Physiology, University of Toronto, Ontario, Canada.,Medicine, University of Toronto, Ontario, Canada.,Obstetrics and Gynaecology, University of Toronto, Ontario, Canada
| |
Collapse
|
10
|
Filbertone Protects Obesity-induced Hypothalamic Inflammation by Reduction of Microglia-mediated Inflammatory Responses. BIOTECHNOL BIOPROC E 2021. [DOI: 10.1007/s12257-020-0220-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
11
|
Dalvi P, Loganathan N, Mcilwraith EK, Tran A, Belsham DD. Hypothalamic Cell Models. CELLULAR ENDOCRINOLOGY IN HEALTH AND DISEASE 2021:27-77. [DOI: 10.1016/b978-0-12-819801-8.00002-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
12
|
Monounsaturated Fatty Acids in Obesity-Related Inflammation. Int J Mol Sci 2020; 22:ijms22010330. [PMID: 33396940 PMCID: PMC7795523 DOI: 10.3390/ijms22010330] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 12/23/2020] [Accepted: 12/25/2020] [Indexed: 12/14/2022] Open
Abstract
Obesity is an important aspect of the metabolic syndrome and is often associated with chronic inflammation. In this context, inflammation of organs participating in energy homeostasis (such as liver, adipose tissue, muscle and pancreas) leads to the recruitment and activation of macrophages, which secrete pro-inflammatory cytokines. Interleukin-1β secretion, sustained C-reactive protein plasma levels and activation of the NLRP3 inflammasome characterize this inflammation. The Stearoyl-CoA desaturase-1 (SCD1) enzyme is a central regulator of lipid metabolism and fat storage. This enzyme catalyzes the generation of monounsaturated fatty acids (MUFAs)-major components of triglycerides stored in lipid droplets-from saturated fatty acid (SFA) substrates. In this review, we describe the molecular effects of specific classes of fatty acids (saturated and unsaturated) to better understand the impact of different diets (Western versus Mediterranean) on inflammation in a metabolic context. Given the beneficial effects of a MUFA-rich Mediterranean diet, we also present the most recent data on the role of SCD1 activity in the modulation of SFA-induced chronic inflammation.
Collapse
|
13
|
Tran A, Loganathan N, McIlwraith EK, Belsham DD. Palmitate and Nitric Oxide Regulate the Expression of Spexin and Galanin Receptors 2 and 3 in Hypothalamic Neurons. Neuroscience 2020; 447:41-52. [DOI: 10.1016/j.neuroscience.2019.10.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/14/2019] [Accepted: 10/16/2019] [Indexed: 12/13/2022]
|
14
|
High fat diet-triggered non-alcoholic fatty liver disease: A review of proposed mechanisms. Chem Biol Interact 2020; 330:109199. [DOI: 10.1016/j.cbi.2020.109199] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/09/2020] [Accepted: 07/13/2020] [Indexed: 02/07/2023]
|
15
|
Figlewicz DP, Witkamp RF. FATTY ACIDS AS CELL SIGNALS IN INGESTIVE BEHAVIORS. Physiol Behav 2020; 223:112985. [PMID: 32473927 DOI: 10.1016/j.physbeh.2020.112985] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/04/2020] [Accepted: 05/23/2020] [Indexed: 12/17/2022]
|
16
|
Tran A, He W, Jiang N, Chen JTC, Belsham DD. NAMPT and BMAL1 Are Independently Involved in the Palmitate-Mediated Induction of Neuroinflammation in Hypothalamic Neurons. Front Endocrinol (Lausanne) 2020; 11:351. [PMID: 32595600 PMCID: PMC7303266 DOI: 10.3389/fendo.2020.00351] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/05/2020] [Indexed: 12/19/2022] Open
Abstract
Obesity is a prominent metabolic disease that predisposes individuals to multiple comorbidities, including type 2 diabetes mellitus, cardiovascular diseases, and cancer. Elevated circulating levels of fatty acids contribute to the development of obesity, in part, by targeting the hypothalamus. Palmitate, the most abundant circulating saturated fatty acid, has been demonstrated to dysregulate NAMPT and circadian clock proteins, as well as induce neuroinflammation. These effects ultimately result in hypothalamic dysregulation of feeding behavior and energy homeostasis. NAMPT is the rate-limiting enzyme of the NAD+ salvage pathway and its expression is under the control of the circadian clock. NAD+ produced from NAMPT can modulate the circadian clock, demonstrating bidirectional interactions between circadian and metabolic pathways. Using NPY/AgRP-expressing mHypoE-46 neurons as well as the novel mHypoA-BMAL1-WT/F and mHypoA-BMAL1-KO/F cell lines, we studied whether there were any interactions between NAMPT and the core circadian clock protein BMAL1 in the palmitate-mediated induction of neuroinflammation. We report that palmitate altered Nampt, Bmal1, Per2 and the inflammatory genes Nf-κb, IκBα, Il-6, and Tlr4. Contrary to studies performed with peripheral tissues, the palmitate-mediated induction in Nampt was independent of BMAL1, and basal Nampt levels did not appear to exhibit rhythmic expression. Palmitate-induced downregulation of Bmal1 and Per2 was independent of NAMPT. However, NAMPT and BMAL1 were both involved in the regulation of Nf-κb, IκBα, Il-6, and Tlr4, as NAMPT inhibition resulted in the repression of basal Nf-κb and IκBα and normalized palmitate-mediated increases in Il-6, and Tlr4. On the other hand, BMAL1 deletion repressed basal Nf-κb, but increased basal Il-6. We conclude that NAMPT and BMAL1 do not interact at the transcriptional level in hypothalamic neurons, but are independently involved in the expression of inflammatory genes.
Collapse
Affiliation(s)
- Andy Tran
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Wenyuan He
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Nan Jiang
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Jim T. C. Chen
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Denise D. Belsham
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
17
|
Jamar G, Ribeiro DA, Pisani LP. High-fat or high-sugar diets as trigger inflammation in the microbiota-gut-brain axis. Crit Rev Food Sci Nutr 2020; 61:836-854. [PMID: 32267169 DOI: 10.1080/10408398.2020.1747046] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Giovana Jamar
- Programa de Pós-Graduação Interdisciplinar em Ciências da Saúde, Universidade Federal de São Paulo, Santos, SP, Brazil
- Laboratório de Nutrição e Fisiologia Endócrina (LaNFE), Universidade Federal de São Paulo, Santos, SP, Brazil
| | - Daniel Araki Ribeiro
- Departamento de Biociências, Instituto de Saúde e Sociedade, Universidade Federal de São Paulo, Santos, SP, Brazil
| | - Luciana Pellegrini Pisani
- Laboratório de Nutrição e Fisiologia Endócrina (LaNFE), Universidade Federal de São Paulo, Santos, SP, Brazil
- Departamento de Biociências, Instituto de Saúde e Sociedade, Universidade Federal de São Paulo, Santos, SP, Brazil
| |
Collapse
|
18
|
Butler MJ, Perrini AA, Eckel LA. Estradiol treatment attenuates high fat diet-induced microgliosis in ovariectomized rats. Horm Behav 2020; 120:104675. [PMID: 31923417 PMCID: PMC7117977 DOI: 10.1016/j.yhbeh.2020.104675] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 11/20/2019] [Accepted: 12/28/2019] [Indexed: 01/03/2023]
Abstract
Consumption of a high fat diet (HFD) increases circulating free fatty acids, which can enter the brain and promote a state of microgliosis, as defined by a change in microglia number and/or morphology. Most studies investigating diet-induced microgliosis have been conducted in male rodents despite well-documented sex differences in the neural control of food intake and neuroimmune signaling. This highlights the need to investigate how sex hormones may modulate the behavioral and cellular response to HFD consumption. Estradiol is of particular interest since it exerts a potent anorexigenic effect and has both anti-inflammatory and neuroprotective effects in the brain. As such, the aim of the current study was to investigate whether estradiol attenuates the development of HFD-induced microgliosis in female rats. Estradiol- and vehicle-treated ovariectomized rats were fed either a low-fat chow diet or a 60% HFD for 4 days, after which they were perfused and brain sections were processed via immunohistochemistry for microglia-specific Iba1 protein. Four days of HFD consumption promoted microgliosis, as measured via an increase in the number of microglia in the arcuate nucleus (ARC) of the hypothalamus and nucleus of the solitary tract (NTS), and a decrease in microglial branching in the ARC, NTS, lateral hypothalamus (LH), and ventromedial hypothalamus. Estradiol replacement attenuated the HFD-induced changes in microglia accumulation and morphology in the ARC, LH, and NTS. We conclude that estradiol has protective effects against HFD-induced microgliosis in a region-specific manner in hypothalamic and hindbrain areas implicated in the neural control of food intake.
Collapse
Affiliation(s)
- Michael J Butler
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, United States; Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH, 43210, United States
| | - Alexis A Perrini
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, United States
| | - Lisa A Eckel
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, United States.
| |
Collapse
|
19
|
Stavrovskaya AV, Voronkov DN, Shestakova EA, Gushchina AS, Olshansky AS, Yamshikova NG. [Streptozocin-induced Alzheimer's disease as an independent risk factor for the development of hyperglycemia in Wistar rats]. ACTA ACUST UNITED AC 2019; 65:351-361. [PMID: 32202739 DOI: 10.14341/probl12126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/10/2019] [Accepted: 12/11/2019] [Indexed: 01/09/2023]
Abstract
BACKGROUND In recent years the theme of the relationship of Alzheimers disease (AD) and metabolic disorders has been widely discussed. Nevertheless, it remains unclear whether AD is a direct cause of carbohydrate metabolism disorders or it is the presence of classical risk factors for type 2 diabetes mellitus (DM 2), primarily obesity, that significantly increases the risk of AD. AIM To evaluate the separate contribution of two factors to the development of disorders of carbohydrate metabolism: (1) weight gain due to a high-calorie diet and (2) experimental-induced AD. METHODS Male Wistar rats were injected with streptozocin (STZ) in the lateral ventricles of the brain to induce AD or saline (sham operated animals - SO) during stereotactic operations. After 2 weeks, the animals were divided into four groups: 1) the SO group, which was assigned to the normal calorie (NCD) diet (SO NCD); 2) the SO group, which was assigned to the high-calorie diet (SO HCD); 3) the group to which the norm-calorie diet was prescribed after the administration of STZ into the lateral ventricles of the brain (STZ NCD); 4) the group to which the HCD was assigned after the administration of STZ (STZ HCD). The animals were on a diet for 3 months. Intraperitoneal glucose tolerance tests were carried out before the diet and after 3 months. At the end of the study, a morphological assessment of brain tissue, pancreas, and liver was performed. RESULTS 3 months after surgical interventions and the appointment of diets, the glycemic curves significantly differed in the 4 studied groups: normoglycemia persisted only in the SO + NCD group, while HCD and the STZ administration were accompanied by the development of hyperglycemia (p = 0.0001). The STZ + NСD group, which represented the isolated effect of AD, was also characterized by impaired carbohydrate metabolism. A morphological study showed that HCD leads to a more pronounced ectopic accumulation of fat in the liver and pancreas tissue than NCD. The administration of STZ, regardless of the diet, led to changes typical for the AD model an increase in the size of the ventricles of the brain, degeneration of white matter, and the accumulation of -amyloid in the hypothalamus. CONCLUSIONS The STZ-induced brain damage typical for AD led to impaired carbohydrate metabolism regardless of diet and was an independent risk factor for hyperglycemia.
Collapse
|
20
|
Liu Y, Yu J, Shi YC, Zhang Y, Lin S. The role of inflammation and endoplasmic reticulum stress in obesity-related cognitive impairment. Life Sci 2019; 233:116707. [PMID: 31374234 DOI: 10.1016/j.lfs.2019.116707] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 07/16/2019] [Accepted: 07/28/2019] [Indexed: 12/12/2022]
Abstract
The epidemiological investigations and animal model experiments have confirmed the impact of obesity on the brain, behavior, and cognition. However, the mechanism by which obesity affects cognitive function is not fully understood. With the development of an aging society, there is an increase in the economic and social burden caused by the decline in cognitive function. This manuscript reviews the effects of inflammation and endoplasmic reticulum stress (ERS) on the hypothalamus, hippocampus, and the possible impact on cognitive impairment. These findings provide new insights into the pathophysiological mechanisms that lead to the development of cognitive impairment in the context of obesity.
Collapse
Affiliation(s)
- Yilan Liu
- Quanzhou First Hospital, Fujian Medical University, China
| | - Jing Yu
- Quanzhou First Hospital, Fujian Medical University, China
| | - Yan-Chuan Shi
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), China
| | - Yi Zhang
- Quanzhou First Hospital, Fujian Medical University, China.
| | - Shu Lin
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), China; Illawarra Health and Medical Research Institute, Wollongong 2522, Australia.
| |
Collapse
|
21
|
Abstract
Under conditions leading to aging and metabolic syndrome, the hypothalamus atypically undergoes proinflammatory signaling activation leading to a chronic and stable background inflammation, referred to as "hypothalamic microinflammation." Through the past decade of research, progress has been made to causally link this hypothalamic inflammation to the mechanism of aging as well as metabolic syndrome, promoting the "hypothalamic microinflammation" theory, which helps characterize the consensus of these epidemic health problems. In general, it is consistently appreciated that hypothalamic microinflammation emerges during the early stages of aging and metabolic syndrome and evolves to be multifaceted and advanced alongside disease progression, while inhibition of key inflammatory components in the hypothalamus has a broad range of effects in counteracting these disorders. Herein, focusing on aging and metabolic syndrome, this writing aims to provide an overview of and insights into the mediators, signaling components, cellular impacts, and physiological significance of this hypothalamic microinflammation.
Collapse
|
22
|
Chen D, Cao S, Chang B, Ma T, Gao H, Tong Y, Li T, Han J, Yi X. Increasing hypothalamic nucleobindin 2 levels and decreasing hypothalamic inflammation in obese male mice via diet and exercise alleviate obesity-associated hypogonadism. Neuropeptides 2019; 74:34-43. [PMID: 30503692 DOI: 10.1016/j.npep.2018.10.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/26/2018] [Accepted: 10/30/2018] [Indexed: 02/08/2023]
Abstract
To explore the role of nesfatin-1 in regulating male reproductive function during energy balance variation, we employed an obese mouse model which was first induced by a high-fat diet (HFD) and followed by interventions of a normal diet (ND) and/or moderate exercise, and then serum reproductive hormones of male mice, hypothalamic nucleobindin 2 (NUCB2)/nesfatin-1, inflammatory factors, and gonadotropin-releasing hormone (GnRH) levels were tested. Our findings showed that both serum nesfatin-1, follicle-stimulating hormone (FSH), luteinizing hormone (LH), and testosterone (T) levels and hypothalamic NUCB2/nesfatin-1 and Gnrh mRNA levels were reduced, whereas, the mRNA and protein levels of hypothalamic tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, inhibitor kappa B kinase β (IKKβ), and nuclear factor (NF)-κB were increased in obese male mice. Diet, exercise, and diet combined with exercise interventions reversed the decreases in serum nesfatin-1, FSH, LH, and T levels; increased hypothalamic NUCB2/nesfatin-1 and Gnrh mRNA levels; and reduced hypothalamic TNF-α, IL-1β, IKKβ, and NF-κB levels. These changes were accompanied by reduced adiposity, and these effects were more obvious in the diet combined with exercise group. Overall, our findings suggested that the hypogonadotropic hypogonadism associated with obesity may be induced by reduced hypothalamic NUCB2/nesfatin-1 levels, which attenuated the stimulatory effect on GnRH directly or indirectly by suppressing its anti-inflammatory effect in the brain. Diet and/or exercise interventions were able to alleviate the hypogonadotropic hypogonadism associated with obesity, potentially by increasing hypothalamic NUCB2/nesfatin-1 levels.
Collapse
Affiliation(s)
- Dequan Chen
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, PR China; School of Physical Education, Minnan Normal Universtiy, Zhangzhou, Fujian 363000, PR China
| | - Shicheng Cao
- Department of Sport Medicine, School of Fundamental Sciences, China Medical University, Shenyang, Liaoning 110001, PR China
| | - Bo Chang
- School of Kinesiology, Shenyang Sport University, Shenyang, Liaoning 110102, PR China
| | - Tie Ma
- School of Kinesiology, Shenyang Sport University, Shenyang, Liaoning 110102, PR China
| | - Haining Gao
- School of Kinesiology, Shenyang Sport University, Shenyang, Liaoning 110102, PR China
| | - Yao Tong
- School of Kinesiology, Shenyang Sport University, Shenyang, Liaoning 110102, PR China
| | - Tao Li
- School of Kinesiology, Shenyang Sport University, Shenyang, Liaoning 110102, PR China
| | - Junchao Han
- School of Kinesiology, Shenyang Sport University, Shenyang, Liaoning 110102, PR China
| | - Xuejie Yi
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, PR China; School of Kinesiology, Shenyang Sport University, Shenyang, Liaoning 110102, PR China.
| |
Collapse
|
23
|
Clemenzi MN, Wellhauser L, Aljghami ME, Belsham DD. Tumour necrosis factor α induces neuroinflammation and insulin resistance in immortalised hypothalamic neurones through independent pathways. J Neuroendocrinol 2019; 31:e12678. [PMID: 30582235 DOI: 10.1111/jne.12678] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 11/26/2018] [Accepted: 12/05/2018] [Indexed: 12/27/2022]
Abstract
The links between obesity, inflammation and insulin resistance, which are all key characteristics of type 2 diabetes mellitus, are yet to be delineated in the brain. One of the key neuroinflammatory proteins detected in the hypothalamus with over-nutrition is tumour necrosis factor (TNF)α. Using immortalised embryonic rat and mouse hypothalamic cell lines (rHypoE-7 and mHypoE-46) that express orexigenic neuropeptide Y and agouti-related peptide, we investigated changes in insulin signalling and inflammatory gene marker mRNA expression after TNFα exposure. A quantitative polymerase chain reaction array of 84 inflammatory markers (cytokines, chemokines and receptors) demonstrated an increase in the expression of multiple genes encoding inflammatory markers upon exposure to 100 ng mL-1 TNFα for 4 hours. Furthermore, neurones pre-exposed to TNFα (50 ng mL-1 ) for 6 or 16 hours exhibited a significant reduction in phosphorylated Akt compared to control after insulin treatment, indicating the attenuation of insulin signalling. mRNA expression of insulin signalling-related genes was also decreased with exposure to TNFα. TNFα significantly increased mRNA expression of IκBα, Tnfrsf1a and IL6 at 4 and 24 hours, activating a pro-inflammatory state. An inhibitor study using an inhibitor of nuclear factor kappa B kinase subunit β (IKK-β) inhibitor, PS1145, demonstrated that TNFα-induced neuroinflammatory marker expression occurs through the IKK-β/nuclear factor-kappa B pathway, whereas oleate, a monounsaturated fatty acid, had no effect on inflammatory markers. To test the efficacy of anti-inflammatory treatment to reverse insulin resistance, neurones were treated with TNFα and PS1145, which did not significantly restore the TNFα-induced changes in cellular insulin sensitivity, indicating that an alternative pathway may be involved. In conclusion, exposure to the inflammatory cytokine TNFα causes cellular insulin resistance and inflammation marker expression in the rHypoE-7 and mHypoE-46 neurones, consistent with effects seen with TNFα in peripheral tissues. It also mimics insulin- and palmitate-induced insulin resistance in hypothalamic neurones. The present study provides further evidence that altered central energy metabolism may be caused by obesity-induced cytokine expression.
Collapse
Affiliation(s)
| | - Leigh Wellhauser
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Makram E Aljghami
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Denise D Belsham
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
24
|
Raikwar SP, Thangavel R, Dubova I, Selvakumar GP, Ahmed ME, Kempuraj D, Zaheer SA, Iyer SS, Zaheer A. Targeted Gene Editing of Glia Maturation Factor in Microglia: a Novel Alzheimer's Disease Therapeutic Target. Mol Neurobiol 2019; 56:378-393. [PMID: 29704201 PMCID: PMC6344368 DOI: 10.1007/s12035-018-1068-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 04/08/2018] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a devastating, progressive neurodegenerative disorder that leads to severe cognitive impairment in elderly patients. Chronic neuroinflammation plays an important role in the AD pathogenesis. Glia maturation factor (GMF), a proinflammatory molecule discovered in our laboratory, is significantly upregulated in various regions of AD brains. We have previously reported that GMF is predominantly expressed in the reactive glial cells surrounding the amyloid plaques (APs) in the mouse and human AD brain. Microglia are the major source of proinflammatory cytokines and chemokines including GMF. Recently clustered regularly interspaced short palindromic repeats (CRISPR) based genome editing has been recognized to study the functions of genes that are implicated in various diseases. Here, we investigated if CRISPR-Cas9-mediated GMF gene editing leads to inhibition of GMF expression and suppression of microglial activation. Confocal microscopy of murine BV2 microglial cell line transduced with an adeno-associated virus (AAV) coexpressing Staphylococcus aureus (Sa) Cas9 and a GMF-specific guide RNA (GMF-sgRNA) revealed few cells expressing SaCas9 while lacking GMF expression, thereby confirming successful GMF gene editing. To further improve GMF gene editing efficiency, we developed lentiviral vectors (LVs) expressing either Streptococcus pyogenes (Sp) Cas9 or GMF-sgRNAs. BV2 cells cotransduced with LVs expressing SpCas9 and GMF-sgRNAs revealed reduced GMF expression and the presence of indels in the exons 2 and 3 of the GMF coding sequence. Lipopolysaccharide (LPS) treatment of GMF-edited cells led to reduced microglial activation as shown by reduced p38 MAPK phosphorylation. We believe that targeted in vivo GMF gene editing has a significant potential for developing a unique and novel AD therapy.
Collapse
Affiliation(s)
- Sudhanshu P Raikwar
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
- Harry S. Truman Memorial Veteran's Hospital, US Department of Veterans Affairs, Columbia, MO, USA
| | - Ramasamy Thangavel
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
- Harry S. Truman Memorial Veteran's Hospital, US Department of Veterans Affairs, Columbia, MO, USA
| | - Iuliia Dubova
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
| | - Govindhasamy Pushpavathi Selvakumar
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
- Harry S. Truman Memorial Veteran's Hospital, US Department of Veterans Affairs, Columbia, MO, USA
| | - Mohammad Ejaz Ahmed
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
- Harry S. Truman Memorial Veteran's Hospital, US Department of Veterans Affairs, Columbia, MO, USA
| | - Duraisamy Kempuraj
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
- Harry S. Truman Memorial Veteran's Hospital, US Department of Veterans Affairs, Columbia, MO, USA
| | - Smita A Zaheer
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
| | - Shankar S Iyer
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA
- Harry S. Truman Memorial Veteran's Hospital, US Department of Veterans Affairs, Columbia, MO, USA
| | - Asgar Zaheer
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65211, USA.
- Harry S. Truman Memorial Veteran's Hospital, US Department of Veterans Affairs, Columbia, MO, USA.
| |
Collapse
|
25
|
Gaspar RC, Muñoz VR, Kuga GK, Nakandakari SCBR, Minuzzi LG, Botezelli JD, da Silva ASR, Cintra DE, de Moura LP, Ropelle ER, Pauli JR. Acute physical exercise increases leptin-induced hypothalamic extracellular signal-regulated kinase1/2 phosphorylation and thermogenesis of obese mice. J Cell Biochem 2018; 120:697-704. [PMID: 30206970 DOI: 10.1002/jcb.27426] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 07/12/2018] [Indexed: 11/12/2022]
Abstract
The obesity is a result of energy imbalance and the increase in thermogenesis seems an interesting alternative for the treatment of this disease. The mechanism of energy expenditure through thermogenesis is tightly articulated in the hypothalamus by leptin. The hypothalamic extracellular signal-regulated kinase-1/2 (ERK1/2) is a key mediator of the thermoregulatory effect of leptin and mediates the sympathetic signal to the brown adipose tissue (BAT). In this context, physical exercise is one of the main interventions for the treatment of obesity. Thus, this study aimed to verify the effects of acute physical exercise on leptin-induced hypothalamic ERK1/2 phosphorylation and thermogenesis in obese mice. Here we showed that acute physical exercise reduced the fasting glucose of obese mice and increased leptin-induced hypothalamic p-ERK1/2 and uncoupling protein 1 (UCP1) content in BAT ( P < 0.05). These molecular changes are accompanied by an increased oxygen uptake (VO 2 ) and heat production in obese exercised mice ( P < 0.05). The increased energy expenditure in the obese exercised animals occurred independently of changes in spontaneous activity. Thus, this is the first study demonstrating that acute physical exercise can increase leptin-induced hypothalamic ERK1/2 phosphorylation and energy expenditure of obese mice.
Collapse
Affiliation(s)
- Rafael Calais Gaspar
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Vitor Rosetto Muñoz
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Gabriel Keine Kuga
- Department of Physical Education, Post-graduate Program in Movement Sciences, São Paulo State University (UNESP), Rio Claro, São Paulo, Brazil
| | | | - Luciele Guerra Minuzzi
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.,Research Center for Sport and Physical Activity (IUD/DTP/04213/2016), Faculty of Sports Science and Physical Education, University of Coimbra, Portugal
| | - José Diego Botezelli
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Adelino S R da Silva
- School of Physical Education and Sports of Ribeirão Preto, Post-graduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, USP, Ribeirão Preto, São Paulo, Brazil
| | - Dennys Esper Cintra
- Laboratory of Nutritional Genomics (LabGeN), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.,Laboratory of Cell Signaling, Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, São Paulo, Brazil
| | - Leandro Pereira de Moura
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.,Laboratory of Nutritional Genomics (LabGeN), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.,Laboratory of Cell Signaling, Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, São Paulo, Brazil.,CEPECE - Center of Research in Sport Sciences, School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Eduardo Rochete Ropelle
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.,Laboratory of Cell Signaling, Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, São Paulo, Brazil.,CEPECE - Center of Research in Sport Sciences, School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - José Rodrigo Pauli
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.,Laboratory of Cell Signaling, Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, São Paulo, Brazil.,CEPECE - Center of Research in Sport Sciences, School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| |
Collapse
|
26
|
Tse EK, Salehi A, Clemenzi MN, Belsham DD. Role of the saturated fatty acid palmitate in the interconnected hypothalamic control of energy homeostasis and biological rhythms. Am J Physiol Endocrinol Metab 2018; 315:E133-E140. [PMID: 29631363 DOI: 10.1152/ajpendo.00433.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The brain, specifically the hypothalamus, controls whole body energy and glucose homeostasis through neurons that synthesize specific neuropeptides, whereas hypothalamic dysfunction is linked directly to insulin resistance, obesity, and type 2 diabetes mellitus. Nutrient excess, through overconsumption of a Western or high-fat diet, exposes the hypothalamus to high levels of free fatty acids, which induces neuroinflammation, endoplasmic reticulum stress, and dysregulation of neuropeptide synthesis. Furthermore, exposure to a high-fat diet also disrupts normal circadian rhythms, and conversely, clock gene knockout models have symptoms of metabolic disorders. While whole brain/animal studies have provided phenotypic end points and important clues to the genes involved, there are still major gaps in our understanding of the intracellular pathways and neuron-specific components that ultimately control circadian rhythms and energy homeostasis. Because of its complexity and heterogeneous nature, containing a diverse mix cell types, it is difficult to dissect the critical hypothalamic components involved in these processes. Of significance, we have the capacity to study these individual components using an extensive collection of both embryonic- and adult-derived, immortalized hypothalamic neuronal cell lines from rodents. These defined neuronal cell lines have been used to examine the impact of nutrient excess, such as palmitate, on circadian rhythms and neuroendocrine signaling pathways, as well as changes in vital neuropeptides, leading to the development of neuronal inflammation; the role of proinflammatory molecules in this process; and ultimately, restoration of normal signaling, clock gene expression, and neuropeptide synthesis in disrupted states by beneficial anti-inflammatory compounds in defined hypothalamic neurons.
Collapse
Affiliation(s)
- Erika K Tse
- Department of Physiology, University of Toronto , Toronto, Ontario , Canada
| | - Ashkan Salehi
- Department of Physiology, University of Toronto , Toronto, Ontario , Canada
| | - Matthew N Clemenzi
- Department of Physiology, University of Toronto , Toronto, Ontario , Canada
| | - Denise D Belsham
- Department of Physiology, University of Toronto , Toronto, Ontario , Canada
- Department Obstetrics and Gynaecology and Medicine, University of Toronto , Toronto, Ontario , Canada
| |
Collapse
|
27
|
Le Jemtel TH, Samson R, Milligan G, Jaiswal A, Oparil S. Visceral Adipose Tissue Accumulation and Residual Cardiovascular Risk. Curr Hypertens Rep 2018; 20:77. [PMID: 29992362 DOI: 10.1007/s11906-018-0880-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF THE REVIEW Low-grade systemic inflammation increases residual cardiovascular risk. The pathogenesis of low-grade systemic inflammation is not well understood. RECENT FINDINGS Visceral adipose tissue accumulates when the subcutaneous adipose tissue can no longer store excess nutrients. Visceral adipose tissue inflammation initially facilitates storage of nutrients but with time become maladaptive and responsible for low-grade systemic inflammation. Control of low-grade systemic inflammation requires reversal of visceral adipose tissue accumulation with intense and sustained aerobic exercise or bariatric surgery. Alternatively, pharmacologic inhibition of the inflammatory signaling pathway may be considered. Reversal visceral adipose tissue accumulation lowers residual cardiovascular risk.
Collapse
Affiliation(s)
- Thierry H Le Jemtel
- Division of Cardiology, Tulane University School of Medicine, 1430 Tulane Avenue, SL-48, New Orleans, LA, 70112, USA.
| | - Rohan Samson
- Division of Cardiology, Tulane University School of Medicine, 1430 Tulane Avenue, SL-48, New Orleans, LA, 70112, USA
| | - Gregory Milligan
- Division of Cardiology, Tulane University School of Medicine, 1430 Tulane Avenue, SL-48, New Orleans, LA, 70112, USA
| | - Abhishek Jaiswal
- Department of Cardiology, Hartford Hospital, 85 Jefferson Street, Suite 208, Hartford, CT, 06106, USA
| | - Suzanne Oparil
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, USA
| |
Collapse
|
28
|
Kim J, Kwon YH, Kim CS, Tu TH, Kim BS, Joe Y, Chung HT, Goto T, Kawada T, Park T, Choi MS, Kim MS, Yu R. The involvement of 4-1BB/4-1BBL signaling in glial cell-mediated hypothalamic inflammation in obesity. FEBS Open Bio 2018; 8:843-853. [PMID: 29744298 PMCID: PMC5929936 DOI: 10.1002/2211-5463.12426] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/09/2018] [Accepted: 03/29/2018] [Indexed: 01/12/2023] Open
Abstract
Obesity‐induced inflammation occurs not only in peripheral tissues but also in areas of the central nervous system. Glial cells such as astrocytes and microglia play crucial roles in obesity‐related hypothalamic inflammation, leading to the derangement of energy metabolism and neurodegenerative pathologies. Here, we show that the interaction of 4‐1BB/4‐1BBL between lipid‐laden astrocytes/microglia promotes hypothalamic inflammation in obesity. Stimulation of 4‐1BB, a member of the TNF receptor superfamily, and/or its ligand 4‐1BBL on astrocytes and/or microglia with a specific agonist resulted in activation of the inflammatory signaling pathway and enhanced production of inflammatory mediators. Contact coculture of lipid‐laden astrocytes and microglia increased the production of inflammatory mediators, and blockade of the 4‐1BB/4‐1BBL interaction reduced the inflammatory response. Moreover, deficiency of 4‐1BB reduced hypothalamic inflammation in obese mice fed an high‐fat diet. These findings suggest that 4‐1BBL/4‐1BB signaling enhances the glial cell‐mediated inflammatory cross talk and participates in obesity‐induced hypothalamic inflammation.
Collapse
Affiliation(s)
- Jiye Kim
- Department of Food Science and Nutrition University of Ulsan South Korea
| | - Yoon-Hee Kwon
- Department of Food Science and Nutrition University of Ulsan South Korea
| | - Chu-Sook Kim
- Department of Food Science and Nutrition University of Ulsan South Korea
| | - Thai H Tu
- Department of Food Science and Nutrition University of Ulsan South Korea
| | - Byung-Sam Kim
- Department of Biological Science University of Ulsan South Korea
| | - Yeonsoo Joe
- Department of Biological Science University of Ulsan South Korea
| | - Hun T Chung
- Department of Biological Science University of Ulsan South Korea
| | - Tsuyoshi Goto
- Graduate School of Agriculture Kyoto University Uji Japan
| | - Teruo Kawada
- Graduate School of Agriculture Kyoto University Uji Japan
| | - Taesun Park
- Department of Food and Nutrition Yonsei University Seoul South Korea
| | - Myung-Sook Choi
- Department of Food Science and Nutrition Center for Food and Nutritional Genomics Research Kyungpook National University Daegu South Korea
| | - Min-Seon Kim
- Division of Endocrinology and Metabolism University of Ulsan College of Medicine Seoul South Korea
| | - Rina Yu
- Department of Food Science and Nutrition University of Ulsan South Korea
| |
Collapse
|
29
|
Kim SM, McIlwraith EK, Chalmers JA, Belsham DD. Palmitate Induces an Anti-Inflammatory Response in Immortalized Microglial BV-2 and IMG Cell Lines that Decreases TNFα Levels in mHypoE-46 Hypothalamic Neurons in Co-Culture. Neuroendocrinology 2018; 107:387-399. [PMID: 30352432 DOI: 10.1159/000494759] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 10/23/2018] [Indexed: 11/19/2022]
Abstract
BACKGROUND AND OBJECTIVES Elevated levels of saturated fatty acids (SFA) induce a state of neuroinflammation in the hypothalamus. It has been suggested that microglia sense palmitate, a prevalent circulating SFA, and act as mediators of this inflammatory process by communicating with neurons, particularly those involved in appetite regulation. In this study, we examined the inflammatory response to palmitate in immortalized microglial cell lines, BV-2 and IMG, and the subsequent effects on inflammatory gene expression in a model of NPY/AgRP neurons, mHypoE-46. METHODS The BV-2 cells were treated with 50 µM palmitate for 4 and 24 h, and the transcriptional regulation of markers for inflammation and cellular stress was assessed using an RT2 Profiler PCR Array. Select genes were verified with qRT-PCR. The BV-2 and IMG cells were then co-cultured using 1.0-µm cell culture inserts with an immortalized hypothalamic cell line, mHypoE-46, to investigate potential intercellular communication between microglia and neurons. RESULTS We found that palmitate increased the mRNA levels of specific inflammatory genes, and a general anti-inflammatory profile was revealed in the microglia cells. The mRNA changes in TNFα at 4 and 24 h in BV-2 cells were abrogated with the toll-like receptor 4 (TLR4) inhibitor, TAK-242, indicating the involvement of TLR4. Co-culture of mHypoE-46 neurons with microglia pre-treated with palmitate resulted in repression of TNFα expression in the hypothalamic neurons. As palmitate significantly increased IL-13 expression in microglia, the effect of this cytokine was tested in mHypoE-46 neurons. The addition of IL-13 to neuronal cultures normalized the palmitate-mediated increase in IL-6 and AgRP expression, suggesting that microglia may protect surrounding neurons, at least in part, through the release of IL-13. CONCLUSIONS These results suggest a potential anti-inflammatory role of microglia towards the palmitate-induced neuroinflammation, and potentially energy homeostasis, in hypothalamic neurons.
Collapse
Affiliation(s)
- Stephanie M Kim
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Emma K McIlwraith
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jennifer A Chalmers
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Denise D Belsham
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario,
- Departments of Medicine and Obstetrics and Gynaecology, Faculty of Medicine, University of Toronto, Toronto, Ontario,
| |
Collapse
|
30
|
Alsabeeh N, Chausse B, Kakimoto PA, Kowaltowski AJ, Shirihai O. Cell culture models of fatty acid overload: Problems and solutions. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1863:143-151. [PMID: 29155055 DOI: 10.1016/j.bbalip.2017.11.006] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 11/09/2017] [Accepted: 11/14/2017] [Indexed: 12/17/2022]
Abstract
High plasma levels of fatty acids occur in a variety of metabolic diseases. Cellular effects of fatty acid overload resulting in negative cellular responses (lipotoxicity) are often studied in vitro, in an attempt to understand mechanisms involved in these diseases. Fatty acids are poorly soluble, and thus usually studied when complexed to albumins such as bovine serum albumin (BSA). The conjugation of fatty acids to albumin requires care pertaining to preparation of the solutions, effective free fatty acid concentrations, use of different fatty acid species, types of BSA, appropriate controls and ensuring cellular fatty acid uptake. This review discusses lipotoxicity models, the potential problems encountered when using these cellular models, as well as practical solutions for difficulties encountered.
Collapse
Affiliation(s)
- Nour Alsabeeh
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA; Department of Physiology and Biophysics, Boston University School of Medicine, Boston, MA 02118, USA; Department of Physiology, Faculty of Medicine, Kuwait University, Kuwait
| | - Bruno Chausse
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP 05508-000, Brazil
| | - Pamela A Kakimoto
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP 05508-000, Brazil
| | - Alicia J Kowaltowski
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP 05508-000, Brazil.
| | - Orian Shirihai
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| |
Collapse
|
31
|
Im DS. FFA4 (GPR120) as a fatty acid sensor involved in appetite control, insulin sensitivity and inflammation regulation. Mol Aspects Med 2017; 64:92-108. [PMID: 28887275 DOI: 10.1016/j.mam.2017.09.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 09/03/2017] [Accepted: 09/03/2017] [Indexed: 12/19/2022]
Abstract
Unsaturated long-chain fatty acids have been suggested to be beneficial in the context of cardiovascular disorders based in epidemiologic studies conducted in Greenland and Mediterranean. DHA and EPA are omega-3 polyunsaturated fatty acids that are plentiful in fish oil, and oleic acid is an omega-9 monounsaturated fatty acid, rich in olive oil. Dietary intake of these unsaturated long-chain fatty acids have been associated with insulin sensitivity and weight loss, which contrasts with the impairment of insulin sensitivity and weight gain associated with high intakes of saturated long-chain fatty acids. The recent discovery that free fatty acid receptor 4 (FFA4, also known as GPR120) acts as a sensor for unsaturated long-chain fatty acids started to unveil the molecular mechanisms underlying the beneficial functions played by these unsaturated long-chain fatty acids in various physiological processes, which include the secretions of gastrointestinal peptide hormones and glucose homeostasis. In this review, the physiological roles and therapeutic significance of FFA4 in appetite control, insulin sensitization, and inflammation reduction are discussed in relation to obesity and type 2 diabetes from pharmacological viewpoints.
Collapse
Affiliation(s)
- Dong-Soon Im
- Molecular Inflammation Research Center for Aging Intervention (MRCA), College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea.
| |
Collapse
|
32
|
Rodrigues BDA, Muñoz VR, Kuga GK, Gaspar RC, Nakandakari SCBR, Crisol BM, Botezelli JD, Pauli LSS, da Silva ASR, de Moura LP, Cintra DE, Ropelle ER, Pauli JR. Obesity Increases Mitogen-Activated Protein Kinase Phosphatase-3 Levels in the Hypothalamus of Mice. Front Cell Neurosci 2017; 11:313. [PMID: 29062272 PMCID: PMC5640777 DOI: 10.3389/fncel.2017.00313] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 09/21/2017] [Indexed: 11/21/2022] Open
Abstract
Mitogen-activated Protein Kinase Phosphatase 3 (MKP-3) has been involved in the negative regulation of insulin signaling. The absence of MKP-3 is also associated with reduced adiposity, increased energy expenditure and improved insulin sensitivity. The MKP-3 is known as the main Erk1/2 phosphatase and FoxO1 activator, which has repercussions on the gluconeogenesis pathway and hyperglycemia in obese mice. Recently, we showed that MKP-3 overexpression decreases FoxO1 phosphorylation in the hypothalamus of lean mice. However, the hypothalamic interaction between MKP-3 and FoxO1 during obesity was not investigated yet. Here, the MKP-3 expression and the effects on food intake and energy expenditure, were investigated in high-fat diet-induced obese mice. The results indicate that obesity in mice increased the MKP-3 protein content in the hypothalamus. This hypothalamic upregulation led to an increase of food intake, adiposity, and body weight. Furthermore, the obese mice with increased MKP-3 showed an insulin signaling impairment with reduction of insulin-induced FoxO1 and Erk1/2 phosphorylation in the hypothalamus. Moreover, a bioinformatics analysis of data demonstrated that hypothalamic MKP-3 mRNA levels were positively correlated with body weight and negatively correlated to oxygen consumption (VO2) in BXD mice. Taken together, our study reports that obesity is associated with increased protein levels of hypothalamic MKP-3, which is related to the reduction of FoxO1 and Erk1/2 phosphorylation in the hypothalamus as well as to an increase in body weight and a reduction in energy expenditure.
Collapse
Affiliation(s)
- Bárbara de A Rodrigues
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Vitor R Muñoz
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Gabriel K Kuga
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), São Paulo, Brazil.,Post-Graduate Program in Movement Sciences, São Paulo State University (Unesp), Institute of Biosciences, São Paulo, Brazil
| | - Rafael C Gaspar
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Susana C B R Nakandakari
- OCRC-Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Barbara M Crisol
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), São Paulo, Brazil
| | - José D Botezelli
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Luciana S S Pauli
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Adelino S R da Silva
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), São Paulo, Brazil
| | - Leandro P de Moura
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), São Paulo, Brazil.,OCRC-Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), São Paulo, Brazil.,CEPECE-Center of Research in Sport Sciences, School of Applied Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Dennys E Cintra
- OCRC-Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), São Paulo, Brazil.,Laboratory of Nutritional Genomics, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Eduardo R Ropelle
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), São Paulo, Brazil.,OCRC-Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), São Paulo, Brazil.,CEPECE-Center of Research in Sport Sciences, School of Applied Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
| | - José R Pauli
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), São Paulo, Brazil.,OCRC-Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), São Paulo, Brazil.,CEPECE-Center of Research in Sport Sciences, School of Applied Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
| |
Collapse
|
33
|
Totsch SK, Quinn TL, Strath LJ, McMeekin LJ, Cowell RM, Gower BA, Sorge RE. The impact of the Standard American Diet in rats: Effects on behavior, physiology and recovery from inflammatory injury. Scand J Pain 2017; 17:316-324. [PMID: 28927908 DOI: 10.1016/j.sjpain.2017.08.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 08/23/2017] [Accepted: 08/24/2017] [Indexed: 12/26/2022]
Abstract
BACKGROUND AND AIMS Obesity is a significant health concern in the Western world and the presence of comorbid conditions suggests an interaction. The overlapping distributions of chronic pain populations and obesity suggests that an interaction may exist. Poor quality diet (high carbohydrates, saturated fats, omega-6 polyunsaturated fatty acids) can lead to increased adiposity which can activate immune cells independent of the activating effect of the diet components themselves. This dual action can contribute to chronic inflammation that may alter susceptibility to chronic pain and prolong recovery from injury. However, traditional examinations of diet focus on high-fat diets that often contain a single source of fat, that is not reflective of an American diet. Thus, we examined the impact of a novel human-relevant (high-carbohydrate) American diet on measures of pain and inflammation in rats, as well as the effect on recovery and immune cell activation. METHODS We developed a novel, human-relevant Standard American Diet (SAD) to better model the kilocalorie levels and nutrient sources in an American population. Male and female rats were fed the SAD over the course of 20 weeks prior to persistent inflammatory pain induction with Complete Freund's Adjuvant (CFA). Mechanical and thermal sensitivity were measured weekly. Spontaneous pain, open field locomotion and blood glucose levels were measured during diet consumption. Body composition was assessed at 20 weeks. Following full recovery from CFA-induced hypersensitivity, blood was analyzed for inflammatory mediators and spinal cords were immunohistochemically processed for microglial markers. RESULTS Chronic consumption of the SAD increased fat mass, decreased lean mass and reduce bone mineral density. SAD-fed rats had increased leptin levels and pro-inflammatory cytokines in peripheral blood serum. Following CFA administration, mechanical sensitivity was assessed and recovery was delayed significantly in SAD-fed animals. Sex differences in the impact of the SAD were also observed. The SAD increased body weight and common T-cell related inflammatory mediators in female, but not male, animals. In males, the SAD had a greater effect on bone mineral density and body composition. Long-term consumption of the SAD resulted in elevated microglial staining in the dorsal horn of the spinal cord, but no sex differences were observed. CONCLUSIONS We demonstrate the negative effects of an American diet on physiology, behavior and recovery from injury. SAD consumption elevated pro-inflammatory mediators and increased microglial activation in the spinal cord. While there were sex differences in weight gain and inflammation, both sexes showed prolonged recovery from injury. IMPLICATIONS These data suggest that poor quality diet may increase susceptibility to chronic pain due to persistent peripheral and central immune system activation. Furthermore, consumption of a diet that is high in carbohydrates and omega-6 polyunsaturated fatty acid is likely to lead to protracted recovery following trauma or surgical procedures. These data suggest that recovery of a number of patients eating a poor quality diet may be expedited with a change in diet to one that is healthier.
Collapse
Affiliation(s)
- Stacie K Totsch
- Department of Psychology, University of Alabama at Birmingham, AL, USA
| | - Tammie L Quinn
- Department of Psychology, University of Alabama at Birmingham, AL, USA
| | - Larissa J Strath
- Department of Psychology, University of Alabama at Birmingham, AL, USA
| | - Laura J McMeekin
- Department of Psychiatry, University of Alabama at Birmingham, AL, USA
| | - Rita M Cowell
- Department of Psychiatry, University of Alabama at Birmingham, AL, USA
| | - Barbara A Gower
- Department of Nutrition Sciences, University of Alabama at Birmingham, AL, USA
| | - Robert E Sorge
- Department of Psychology, University of Alabama at Birmingham, AL, USA.
| |
Collapse
|
34
|
Wang L, Jacobs JP, Lagishetty V, Yuan PQ, Wu SV, Million M, Reeve JR, Pisegna JR, Taché Y. High-protein diet improves sensitivity to cholecystokinin and shifts the cecal microbiome without altering brain inflammation in diet-induced obesity in rats. Am J Physiol Regul Integr Comp Physiol 2017; 313:R473-R486. [PMID: 28724546 DOI: 10.1152/ajpregu.00105.2017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 07/14/2017] [Accepted: 07/14/2017] [Indexed: 12/14/2022]
Abstract
High-protein diet (HPD) curtails obesity and/or fat mass, but it is unknown whether it reverses neuroinflammation or alters glucose levels, CCK sensitivity, and gut microbiome in rats fed a Western diet (WD)-induced obesity (DIO). Male rats fed a WD (high fat and sugar) for 12 wk were switched to a HPD for 6 wk. Body composition, food intake, meal pattern, sensitivity to intraperitoneal CCK-8S, blood glucose, brain signaling, and cecal microbiota were assessed. When compared with a normal diet, WD increased body weight (9.3%) and fat mass (73.4%). CCK-8S (1.8 or 5.2 nmol/kg) did not alter food intake and meal pattern in DIO rats. Switching to a HPD for 6 wk reduced fat mass (15.7%) with a nonsignificantly reduced body weight gain, normalized blood glucose, and decreased feeding after CCK-8S. DIO rats on the WD or switched to a HPD showed comparable microbial diversity. However, in HPD versus WD rats, there was enrichment of 114 operational taxonomic units (OTUs) and depletion of 188 OTUs. Of those, Akkermansia muciniphila (enriched on a HPD), an unclassified Clostridiales, a member of the RF39 order, and a Phascolarctobacterium were significantly associated with fat mass. The WD increased cytokine expression in the hypothalamus and dorsal medulla that was unchanged by switching to HPD. These data indicate that HPD reduces body fat and restores glucose homeostasis and CCK sensitivity, while not modifying brain inflammation. In addition, expansion of cecal Akkermansia muciniphila correlated to fat mass loss may represent a potential peripheral mechanism of HPD beneficial effects.
Collapse
Affiliation(s)
- Lixin Wang
- CURE/Digestive Diseases Research Center and Center for Neurobiology of Stress and Resilience, Department of Medicine, Vatche and Tamar Manoukian Digestive Diseases Division, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California; and
| | - Jonathan P Jacobs
- CURE/Digestive Diseases Research Center and Center for Neurobiology of Stress and Resilience, Department of Medicine, Vatche and Tamar Manoukian Digestive Diseases Division, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California; and
| | - Venu Lagishetty
- CURE/Digestive Diseases Research Center and Center for Neurobiology of Stress and Resilience, Department of Medicine, Vatche and Tamar Manoukian Digestive Diseases Division, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California; and
| | - Pu-Qing Yuan
- CURE/Digestive Diseases Research Center and Center for Neurobiology of Stress and Resilience, Department of Medicine, Vatche and Tamar Manoukian Digestive Diseases Division, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California; and
| | - Shuping V Wu
- CURE/Digestive Diseases Research Center and Center for Neurobiology of Stress and Resilience, Department of Medicine, Vatche and Tamar Manoukian Digestive Diseases Division, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California; and
| | - Mulugeta Million
- CURE/Digestive Diseases Research Center and Center for Neurobiology of Stress and Resilience, Department of Medicine, Vatche and Tamar Manoukian Digestive Diseases Division, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California; and
| | - Joseph R Reeve
- CURE/Digestive Diseases Research Center and Center for Neurobiology of Stress and Resilience, Department of Medicine, Vatche and Tamar Manoukian Digestive Diseases Division, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California; and
| | - Joseph R Pisegna
- CURE/Digestive Diseases Research Center and Center for Neurobiology of Stress and Resilience, Department of Medicine, Vatche and Tamar Manoukian Digestive Diseases Division, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California; and.,Division of Gastroenterology, Hepatology and Parenteral Nutrition, and Research and Development, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California
| | - Yvette Taché
- CURE/Digestive Diseases Research Center and Center for Neurobiology of Stress and Resilience, Department of Medicine, Vatche and Tamar Manoukian Digestive Diseases Division, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California; and
| |
Collapse
|
35
|
Quercetin Protects Obesity-Induced Hypothalamic Inflammation by Reducing Microglia-Mediated Inflammatory Responses via HO-1 Induction. Nutrients 2017. [PMID: 28644409 PMCID: PMC5537770 DOI: 10.3390/nu9070650] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Obesity-induced hypothalamic inflammation is characterized by activation of microglia, which are resident macrophages of the central nervous system, and is implicated in the derangement of energy homeostasis, metabolic complications, and neurodegenerative diseases. Quercetin, a naturally occurring flavonoid, is known to protect against oxidative stress and inflammation-related metabolic complications. Here, we demonstrate that quercetin reduces obesity-induced hypothalamic inflammation by inhibiting microglia-mediated inflammatory responses, and the beneficial action of quercetin is associated with heme oxygenase (HO-1) induction. Quercetin markedly reduced the production of inflammatory mediators (monocyte chemoattractant protein (MCP)-1, interleukin (IL-6), IL-1β, nitric oxide) by microglia stimulated with saturated fatty acid palmitate and/or lipid-laden microglia-conditioned medium. Quercetin also upregulated the expression of HO-1 in palmitate-treated lipid-laden microglia, and the actions of quercetin against microglia activation accompanied by IκBα degradation were abolished by a HO-1 inhibitor. Moreover, quercetin supplementation reduced the levels of inflammatory cytokines and microglia activation markers in the hypothalamus of high fat diet (HFD)-fed obese mice, which was accompanied by upregulation of HO-1. These findings indicate that quercetin suppresses microglia-mediated inflammatory responses via the induction of HO-1, and hence protects against obesity-induced hypothalamic inflammation.
Collapse
|
36
|
Morin JP, Rodríguez-Durán LF, Guzmán-Ramos K, Perez-Cruz C, Ferreira G, Diaz-Cintra S, Pacheco-López G. Palatable Hyper-Caloric Foods Impact on Neuronal Plasticity. Front Behav Neurosci 2017; 11:19. [PMID: 28261067 PMCID: PMC5306218 DOI: 10.3389/fnbeh.2017.00019] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 01/23/2017] [Indexed: 01/01/2023] Open
Abstract
Neural plasticity is an intrinsic and essential characteristic of the nervous system that allows animals “self-tuning” to adapt to their environment over their lifetime. Activity-dependent synaptic plasticity in the central nervous system is a form of neural plasticity that underlies learning and memory formation, as well as long-lasting, environmentally-induced maladaptive behaviors, such as drug addiction and overeating of palatable hyper-caloric (PHc) food. In western societies, the abundance of PHc foods has caused a dramatic increase in the incidence of overweight/obesity and related disorders. To this regard, it has been suggested that increased adiposity may be caused at least in part by behavioral changes in the affected individuals that are induced by the chronic consumption of PHc foods; some authors have even drawn attention to the similarity that exists between over-indulgent eating and drug addiction. Long-term misuse of certain dietary components has also been linked to chronic neuroimmune maladaptation that may predispose individuals to neurodegenerative conditions such as Alzheimer’s disease. In this review article, we discuss recent evidence that shows how consumption of PHc food can cause maladaptive neural plasticity that converts short-term ingestive drives into compulsive behaviors. We also discuss the neural mechanisms of how chronic consumption of PHc foods may alter brain function and lead to cognitive impairments, focusing on prenatal, childhood and adolescence as vulnerable neurodevelopmental stages to dietary environmental insults. Finally, we outline a societal agenda for harnessing permissive obesogenic environments.
Collapse
Affiliation(s)
- Jean-Pascal Morin
- Department of Health Sciences, Metropolitan Autonomous University (UAM)Lerma, Mexico; Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-EssenEssen, Germany
| | - Luis F Rodríguez-Durán
- Department of Health Sciences, Metropolitan Autonomous University (UAM)Lerma, Mexico; Laboratory of Neurobiology of Learning and Memory, Division of Research and Graduate Studies, Faculty of Psychology, National Autonomous University of Mexico (UNAM)Mexico City, Mexico
| | - Kioko Guzmán-Ramos
- Department of Health Sciences, Metropolitan Autonomous University (UAM) Lerma, Mexico
| | - Claudia Perez-Cruz
- Department of Pharmacology, Center of Research and Advance Studies (CINVESTAV) Mexico City, Mexico
| | - Guillaume Ferreira
- Laboratory of Nutrition and Integrative Neurobiology, National Institute of Agricultural Research (INRA), UMR 1286Bordeaux, France; Laboratory of Nutrition and Integrative Neurobiology, Université de BordeauxBordeaux, France
| | - Sofia Diaz-Cintra
- Institute of Neurobiology, National Autonomous University of Mexico (UNAM) Queretaro, Mexico
| | - Gustavo Pacheco-López
- Department of Health Sciences, Metropolitan Autonomous University (UAM)Lerma, Mexico; Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) ZurichSchwerzenbach, Switzerland
| |
Collapse
|