1
|
Lv Q, Yang A, Han Z, Yu R, Zhu J, Shi Z, Yang C, Dai S, Hao M, Chen Y, Zhou JC. Selenoprotein H mediates low selenium-related cognitive decline through impaired oligodendrocyte myelination with disrupted hippocampal lipid metabolism in female mice. Food Funct 2024; 15:8544-8561. [PMID: 39072440 DOI: 10.1039/d4fo00888j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Low selenium levels are closely associated with reduced cognitive performance and lipid dysregulation, yet the mechanism of action remains unclear. The physiological function of selenium is primarily mediated by selenoproteins. Selenoprotein H (SELENOH), as one of the selenium-containing proteins, has an unelucidated role in regulating cognitive status and lipid metabolism. In this study, we established a Selenoh gene knockout (HKO) mouse model to investigate whether Selenoh mediates the impact of selenium on cognitive function. We found that HKO mice showed a significant decline in cognition compared with the wild-type (HWT) littermates, and were not affected by deficient or excessive selenium, while no differences in anxiety and depression behavior were observed. HKO mice showed reduced myelin basic protein expression in hippocampal oligodendrocytes, with decreased glycolipid levels and increased phospholipid and sphingolipid levels in the hippocampus. Furthermore, the high-fat diet (HFD) exerted no effect on cognition and limited impact on the gene profile in the hippocampus of HKO mice. Compared with those of HWT mice, the myelination pathways in the hippocampus of HKO mice were downregulated as revealed by RNA-seq, which was further confirmed by the reduced expression levels of myelin-related proteins. Finally, HKO increased the expression of hippocampal fatty acid transporter (FATP) 4, and HFD increased the FATP4 expression in HWT mice but not in HKO mice. In summary, our study demonstrated that HKO induced cognitive decline by impairing myelination in oligodendrocytes with disrupted hippocampal lipid metabolism, which provided a novel viewpoint on the selenoprotein-mediated neurodegenerative diseases of selenium.
Collapse
Affiliation(s)
- Qingqing Lv
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, Guangdong, China.
| | - Aolin Yang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, Guangdong, China.
| | - Ziyu Han
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, Guangdong, China.
| | - Ruirui Yu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, Guangdong, China.
| | - Junying Zhu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, Guangdong, China.
| | - Zhan Shi
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, Guangdong, China.
| | - Chenggang Yang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, Guangdong, China.
| | - Shimiao Dai
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, Guangdong, China.
| | - Mengru Hao
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, Guangdong, China.
| | - Yuqing Chen
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, Guangdong, China.
| | - Ji-Chang Zhou
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, Guangdong, China.
- Guangdong Province Engineering Laboratory for Nutrition Translation, Shenzhen 518107, Guangdong, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, Guangdong, China
| |
Collapse
|
2
|
Cruciani-Guglielmacci C, Le Stunff H, Magnan C. Brain lipid sensing and the neural control of energy balance. Biochimie 2024; 223:159-165. [PMID: 38825062 DOI: 10.1016/j.biochi.2024.05.020] [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: 04/11/2024] [Revised: 05/23/2024] [Accepted: 05/25/2024] [Indexed: 06/04/2024]
Abstract
The central nervous system continuously detects circulating concentrations of lipids such as fatty acids and troglycerides. Once information has been detected, the central nervous system can in turn participate in the control of energy balance and blood sugar levels and in particular regulate the secretion and action of insulin. Neurons capable of detecting circulating lipid variations are located in the hypothalamus and in other regions such as the nucleus accumbens, the striatum or the hippocampus. An excess of lipids will have deleterious effects and may induce central lipotoxicity, in particular following local production of ceramides and the appearance of neuroinflammation which may lead to metabolic diseases such as obesity and type 2 diabetes.
Collapse
Affiliation(s)
| | - Hervé Le Stunff
- Paris-Saclay Institute of Neuroscience, CNRS UMR 9197, Université Paris-Sud, University Paris Saclay, Orsay, France
| | | |
Collapse
|
3
|
Lee TH, Cota D, Quarta C. Yin-Yang control of energy balance by lipids in the hypothalamus: The endocannabinoids vs bile acids case. Biochimie 2024; 223:188-195. [PMID: 35863558 DOI: 10.1016/j.biochi.2022.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/22/2022] [Accepted: 07/12/2022] [Indexed: 11/02/2022]
Abstract
Obesity is a chronic and debilitating disorder that originates from alterations in energy-sensing brain circuits controlling body weight gain and food intake. The dysregulated syntheses and actions of lipid mediators in the hypothalamus induce weight gain and overfeeding, but the molecular and cellular underpinnings of these alterations remain elusive. In response to changes in the nutritional status, different lipid sensing pathways in the hypothalamus direct body energy needs in a Yin-Yang model. Endocannabinoids orchestrate the crosstalk between hypothalamic circuits and the sympathetic nervous system to promote food intake and energy accumulation during fasting, whereas bile acids act on the same top-down axis to reduce energy intake and possibly storage after the meal. In obesity, the bioavailability and downstream cellular actions of endocannabinoids and bile acids are altered in hypothalamic neurons involved in body weight and metabolic control. Thus, the onset and progression of this disease might result from an imbalance in hypothalamic sensing of multiple lipid signals, which are possibly integrated by common molecular nodes. In this viewpoint, we discuss a possible model that explains how bile acids and endocannabinoids may exert their effects on energy balance regulation via interconnected mechanisms at the level of the hypothalamic neuronal circuits. Therefore, we propose a new conceptual framework for understanding and treating central mechanisms of maladaptive lipid action in obesity.
Collapse
Affiliation(s)
- Thomas H Lee
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-3300, Bordeaux, France
| | - Daniela Cota
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-3300, Bordeaux, France
| | - Carmelo Quarta
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-3300, Bordeaux, France.
| |
Collapse
|
4
|
Xu DC, Sas-Nowosielska H, Donahue G, Huang H, Pourshafie N, Good CR, Berger SL. Histone acetylation in an Alzheimer's disease cell model promotes homeostatic amyloid-reducing pathways. Acta Neuropathol Commun 2024; 12:3. [PMID: 38167174 PMCID: PMC10759377 DOI: 10.1186/s40478-023-01696-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/21/2023] [Indexed: 01/05/2024] Open
Abstract
Alzheimer's Disease (AD) is a disorder characterized by cognitive decline, neurodegeneration, and accumulation of amyloid plaques and tau neurofibrillary tangles in the brain. Dysregulation of epigenetic histone modifications may lead to expression of transcriptional programs that play a role either in protecting against disease genesis or in worsening of disease pathology. One such histone modification, acetylation of histone H3 lysine residue 27 (H3K27ac), is primarily localized to genomic enhancer regions and promotes active gene transcription. We previously discovered H3K27ac to be more abundant in AD patient brain tissue compared to the brains of age-matched non-demented controls. In this study, we use iPSC-neurons derived from familial AD patients with an amyloid precursor protein (APP) duplication (APPDup neurons) as a model to study the functional effect of lowering CBP/P300 enzymes that catalyze H3K27ac. We found that homeostatic amyloid-reducing genes were upregulated in the APPDup neurons compared to non-demented controls. We lowered CBP/P300 to reduce H3K27ac, which led to decreased expression of numerous of these homeostatic amyloid-reducing genes, along with increased extracellular secretion of a toxic amyloid-β species, Aβ(1-42). Our findings suggest that epigenomic histone acetylation, including H3K27ac, drives expression of compensatory genetic programs in response to AD-associated insults, specifically those resulting from APP duplication, and thus may play a role in mitigating AD pathology in neurons.
Collapse
Affiliation(s)
- Daniel C Xu
- Department of Cell and Developmental Biology, Perelman School of Medicine Philadelphia, Penn Institute of Epigenetics, Philadelphia, PA, 19104, USA
| | - Hanna Sas-Nowosielska
- Department of Cell and Developmental Biology, Perelman School of Medicine Philadelphia, Penn Institute of Epigenetics, Philadelphia, PA, 19104, USA
| | - Greg Donahue
- Department of Cell and Developmental Biology, Perelman School of Medicine Philadelphia, Penn Institute of Epigenetics, Philadelphia, PA, 19104, USA
| | - Hua Huang
- Department of Cell and Developmental Biology, Perelman School of Medicine Philadelphia, Penn Institute of Epigenetics, Philadelphia, PA, 19104, USA
| | - Naemeh Pourshafie
- Department of Cell and Developmental Biology, Perelman School of Medicine Philadelphia, Penn Institute of Epigenetics, Philadelphia, PA, 19104, USA
| | - Charly R Good
- Department of Cell and Developmental Biology, Perelman School of Medicine Philadelphia, Penn Institute of Epigenetics, Philadelphia, PA, 19104, USA
| | - Shelley L Berger
- Department of Cell and Developmental Biology, Perelman School of Medicine Philadelphia, Penn Institute of Epigenetics, Philadelphia, PA, 19104, USA.
| |
Collapse
|
5
|
Colquitt BM, Li K, Green F, Veline R, Brainard MS. Neural circuit-wide analysis of changes to gene expression during deafening-induced birdsong destabilization. eLife 2023; 12:e85970. [PMID: 37284822 PMCID: PMC10259477 DOI: 10.7554/elife.85970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 04/17/2023] [Indexed: 06/08/2023] Open
Abstract
Sensory feedback is required for the stable execution of learned motor skills, and its loss can severely disrupt motor performance. The neural mechanisms that mediate sensorimotor stability have been extensively studied at systems and physiological levels, yet relatively little is known about how disruptions to sensory input alter the molecular properties of associated motor systems. Songbird courtship song, a model for skilled behavior, is a learned and highly structured vocalization that is destabilized following deafening. Here, we sought to determine how the loss of auditory feedback modifies gene expression and its coordination across the birdsong sensorimotor circuit. To facilitate this system-wide analysis of transcriptional responses, we developed a gene expression profiling approach that enables the construction of hundreds of spatially-defined RNA-sequencing libraries. Using this method, we found that deafening preferentially alters gene expression across birdsong neural circuitry relative to surrounding areas, particularly in premotor and striatal regions. Genes with altered expression are associated with synaptic transmission, neuronal spines, and neuromodulation and show a bias toward expression in glutamatergic neurons and Pvalb/Sst-class GABAergic interneurons. We also found that connected song regions exhibit correlations in gene expression that were reduced in deafened birds relative to hearing birds, suggesting that song destabilization alters the inter-region coordination of transcriptional states. Finally, lesioning LMAN, a forebrain afferent of RA required for deafening-induced song plasticity, had the largest effect on groups of genes that were also most affected by deafening. Combined, this integrated transcriptomics analysis demonstrates that the loss of peripheral sensory input drives a distributed gene expression response throughout associated sensorimotor neural circuitry and identifies specific candidate molecular and cellular mechanisms that support the stability and plasticity of learned motor skills.
Collapse
Affiliation(s)
- Bradley M Colquitt
- Howard Hughes Medical InstituteChevy ChaseUnited States
- Department of Physiology, University of California, San FranciscoSan FranciscoUnited States
| | - Kelly Li
- Howard Hughes Medical InstituteChevy ChaseUnited States
- Department of Physiology, University of California, San FranciscoSan FranciscoUnited States
| | - Foad Green
- Howard Hughes Medical InstituteChevy ChaseUnited States
- Department of Physiology, University of California, San FranciscoSan FranciscoUnited States
| | - Robert Veline
- Howard Hughes Medical InstituteChevy ChaseUnited States
- Department of Physiology, University of California, San FranciscoSan FranciscoUnited States
| | - Michael S Brainard
- Howard Hughes Medical InstituteChevy ChaseUnited States
- Department of Physiology, University of California, San FranciscoSan FranciscoUnited States
| |
Collapse
|
6
|
Campbell K, Cawley NX, Luke R, Scott KEJ, Johnson N, Farhat NY, Alexander D, Wassif CA, Li W, Cologna SM, Berry-Kravis E, Do AD, Dale RK, Porter FD. Identification of cerebral spinal fluid protein biomarkers in Niemann-Pick disease, type C1. Biomark Res 2023; 11:14. [PMID: 36721240 PMCID: PMC9887810 DOI: 10.1186/s40364-023-00448-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 01/03/2023] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Niemann-Pick disease, type C1 (NPC1) is an ultrarare, recessive, lethal, lysosomal disease characterized by progressive cerebellar ataxia and cognitive impairment. Although the NPC1 phenotype is heterogeneous with variable age of onset, classical NPC1 is a pediatric disorder. Currently there are no therapies approved by the FDA and therapeutics trials for NPC1 are complicated by disease rarity, heterogeneity, and the relatively slow rate of neurological decline. Thus, identification of disease relevant biomarkers is necessary to provide tools that can support drug development efforts for this devastating neurological disease. METHODS Proximal extension assays (O-link® Explore 1536) were used to compare cerebrospinal fluid (CSF) samples from individuals with NPC1 enrolled in a natural history study and non-NPC1 comparison samples. Relative expression levels of 1467 proteins were determined, and candidate protein biomarkers were identified by evaluating fold-change and adjusted Kruskal-Wallis test p-values. Selected proteins were orthogonally confirmed using ELISA. To gain insight into disease progression and severity we evaluated the altered protein expression with respect to clinically relevant phenotypic aspects: NPC Neurological Severity Score (NPC1 NSS), Annual Severity Increment Score (ASIS) and age of neurological onset. RESULTS This study identified multiple proteins with altered levels in CSF from individuals with NPC1 compared to non-NPC1 samples. These included proteins previously shown to be elevated in NPC1 (NEFL, MAPT, CHIT1, CALB1) and additional proteins confirmed by orthogonal assays (PARK7, CALB2/calretinin, CHI3L1/YKL-40, MIF, CCL18 and ENO2). Correlations with clinically relevant phenotypic parameters demonstrated moderate negative (p = 0.0210, r = -0.41) and possible moderate positive (p = 0.0631, r = 0.33) correlation of CSF CALB2 levels with age of neurological onset and ASIS, respectively. CSF CHI3L1 levels showed a moderate positive (p = 0.0183, r = 0.40) correlation with the concurrent NPC1 NSS. A strong negative correlation (p = 0.0016, r = -0.648) was observed between CSF CCL18 and age of neurological onset for childhood/adolescent cases. CSF CCL18 levels also showed a strong positive correlation (p = 0.0017, r = 0.61) with ASIS. CONCLUSION Our study identified and validated multiple proteins in CSF from individuals with NPC1 that are candidates for further investigation in a larger cohort. These analytes may prove to be useful as supportive data in therapeutic trials. TRIAL REGISTRATIONS NCT00344331, NCT00001721, NCT02931682.
Collapse
Affiliation(s)
- Kiersten Campbell
- grid.420089.70000 0000 9635 8082Bioinformatics and Scientific Programming Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 10CRC, Rm. 1-3330, 10 Center Dr., Bethesda, MD 20879 USA
| | - Niamh X. Cawley
- grid.420089.70000 0000 9635 8082Section On Molecular Dysmorphology, Division of Translational Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD USA
| | - Rachel Luke
- grid.420089.70000 0000 9635 8082Section On Molecular Dysmorphology, Division of Translational Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD USA
| | - Katelin E. J. Scott
- grid.420089.70000 0000 9635 8082Section On Molecular Dysmorphology, Division of Translational Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD USA
| | - Nicholas Johnson
- grid.420089.70000 0000 9635 8082Bioinformatics and Scientific Programming Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 10CRC, Rm. 1-3330, 10 Center Dr., Bethesda, MD 20879 USA
| | - Nicole Y. Farhat
- grid.420089.70000 0000 9635 8082Section On Molecular Dysmorphology, Division of Translational Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD USA
| | - Derek Alexander
- grid.420089.70000 0000 9635 8082Section On Molecular Dysmorphology, Division of Translational Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD USA
| | - Christopher A. Wassif
- grid.420089.70000 0000 9635 8082Section On Molecular Dysmorphology, Division of Translational Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD USA
| | - Wenping Li
- grid.185648.60000 0001 2175 0319Department of Chemistry and Laboratory of Integrative Neuroscience, University of Illinois Chicago, Chicago, IL USA
| | - Stephanie M. Cologna
- grid.185648.60000 0001 2175 0319Department of Chemistry and Laboratory of Integrative Neuroscience, University of Illinois Chicago, Chicago, IL USA
| | | | - An Dang Do
- grid.420089.70000 0000 9635 8082Unit On Cellular Stress in Development and Diseases, Division of Translational Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD USA
| | - Ryan K. Dale
- grid.420089.70000 0000 9635 8082Bioinformatics and Scientific Programming Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 10CRC, Rm. 1-3330, 10 Center Dr., Bethesda, MD 20879 USA
| | - Forbes D. Porter
- grid.420089.70000 0000 9635 8082Section On Molecular Dysmorphology, Division of Translational Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD USA
| |
Collapse
|
7
|
Hu W, Liu J, Hu Y, Xu Q, Deng T, Wei M, Lu L, Mi J, Bergquist J, Xu F, Tian G. Transcriptome-wide association study reveals cholesterol metabolism gene Lpl is a key regulator of cognitive dysfunction. Front Mol Neurosci 2022; 15:1044022. [PMID: 36590920 PMCID: PMC9798092 DOI: 10.3389/fnmol.2022.1044022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 11/16/2022] [Indexed: 12/23/2022] Open
Abstract
Cholesterol metabolism in the brain plays a crucial role in normal physiological function, and its aberrations are associated with cognitive dysfunction. The present study aimed to determine which cholesterol-related genes play a vital role in cognitive dysfunction and to dissect its underlying molecular mechanisms using a systems genetics approach in the BXD mice family. We first systematically analyzed the association of expression of 280 hippocampal genes related to cholesterol metabolism with cognition-related traits and identified lipoprotein lipase (Lpl) as a critical regulator. This was further confirmed by phenome-wide association studies that indicate Lpl associated with hippocampus volume residuals and anxiety-related traits. By performing expression quantitative trait locus mapping, we demonstrate that Lpl is strongly cis-regulated in the BXD hippocampus. We also identified ∼3,300 genes significantly (p < 0.05) correlated with the Lpl expression. Those genes are mainly involved in the regulation of neuron-related traits through the MAPK signaling pathway, axon guidance, synaptic vesicle cycle, and NF-kappa B signaling pathway. Furthermore, a protein-protein interaction network analysis identified several direct interactors of Lpl, including Rab3a, Akt1, Igf1, Crp, and Lrp1, which indicates that Lpl involves in the regulation of cognitive dysfunction through Rab3a-mediated synaptic vesicle cycle and Akt1/Igf1/Crp/Lrp1-mediated MAPK signaling pathway. Our findings demonstrate the importance of the Lpl, among the cholesterol-related genes, in regulating cognitive dysfunction and highlighting the potential signaling pathways, which may serve as novel therapeutic targets for the treatment of cognitive dysfunction.
Collapse
Affiliation(s)
- Wei Hu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China,Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, School of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
| | - Jian Liu
- Department of Plastic Surgery, The First Affiliated Hospital of Shandong First Medical University, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China,Jinan Clinical Research Center for Tissue Engineering Skin Regeneration and Wound Repair, Jinan, Shandong, China
| | - Yaorui Hu
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, School of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
| | - Qingling Xu
- Department of Ultrasound, Yantai Affiliated Hospital, Binzhou Medical University, Yantai, Shandong, China
| | - Tingzhi Deng
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, School of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
| | - Mengna Wei
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, School of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
| | - Lu Lu
- Department of Genetics, Genomics, and Informatics, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Jia Mi
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, School of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
| | - Jonas Bergquist
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, School of Pharmacy, Binzhou Medical University, Yantai, Shandong, China,Department of Chemistry - BMC, Uppsala University, Uppsala, Sweden,*Correspondence: Jonas Bergquist,
| | - Fuyi Xu
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, School of Pharmacy, Binzhou Medical University, Yantai, Shandong, China,Fuyi Xu,
| | - Geng Tian
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, School of Pharmacy, Binzhou Medical University, Yantai, Shandong, China,Geng Tian,
| |
Collapse
|
8
|
Electrochemical determination of glucose and H2O2 using Co(II), Ni(II), Cu(II) complexes of novel 2-(1,3-benzothiazol-2-ylamino)–N-(5-chloro-2-hydroxyphenyl)acetamide: Synthesis, structural characterization, antimicrobial, anticancer activity and docking studies. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.134483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
9
|
Leng L, Yuan Z, Pan R, Su X, Wang H, Xue J, Zhuang K, Gao J, Chen Z, Lin H, Xie W, Li H, Chen Z, Ren K, Zhang X, Wang W, Jin ZB, Wu S, Wang X, Yuan Z, Xu H, Chow HM, Zhang J. Microglial hexokinase 2 deficiency increases ATP generation through lipid metabolism leading to β-amyloid clearance. Nat Metab 2022; 4:1287-1305. [PMID: 36203054 DOI: 10.1038/s42255-022-00643-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 08/18/2022] [Indexed: 01/20/2023]
Abstract
Microglial cells consume adenosine triphosphate (ATP) during phagocytosis to clear neurotoxic β-amyloid in Alzheimer's disease (AD). However, the contribution of energy metabolism to microglial function in AD remains unclear. Here, we demonstrate that hexokinase 2 (HK2) is elevated in microglia from an AD mouse model (5xFAD) and AD patients. Genetic deletion or pharmacological inhibition of HK2 significantly promotes microglial phagocytosis, lowers the amyloid plaque burden and attenuates cognitive impairment in male AD mice. Notably, the ATP level is dramatically increased in HK2-deficient or inactive microglia, which can be attributed to a marked upregulation in lipoprotein lipase (LPL) expression and subsequent increase in lipid metabolism. We further show that two downstream metabolites of HK2, glucose-6-phosphate and fructose-6-phosphate, can reverse HK2-deficiency-induced upregulation of LPL, thus supporting ATP production and microglial phagocytosis. Our findings uncover a crucial role for HK2 in phagocytosis through regulation of microglial energy metabolism, suggesting a potential therapeutic strategy for AD by targeting HK2.
Collapse
Affiliation(s)
- Lige Leng
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China.
| | - Ziqi Yuan
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Ruiyuan Pan
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Xiao Su
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Han Wang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Jin Xue
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Kai Zhuang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Ju Gao
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Zhenlei Chen
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Hui Lin
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Wenting Xie
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Huifang Li
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Zhenyi Chen
- Department of Anesthesiology, First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Keke Ren
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medcial University, Xi'an, Shaanxi, China
| | - Xiao Zhang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Lab, Beijing, China
| | - Wenting Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medcial University, Xi'an, Shaanxi, China
| | - Zi-Bing Jin
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Lab, Beijing, China
| | - Shengxi Wu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medcial University, Xi'an, Shaanxi, China
| | - Xinglong Wang
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Zengqiang Yuan
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Huaxi Xu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Hei-Man Chow
- School of Life Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - Jie Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China.
- Department of Anesthesiology, First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China.
- Institute of Neuroscience, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
10
|
Sterol and lipid analyses identifies hypolipidemia and apolipoprotein disorders in autism associated with adaptive functioning deficits. Transl Psychiatry 2021; 11:471. [PMID: 34504056 PMCID: PMC8429516 DOI: 10.1038/s41398-021-01580-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/03/2021] [Accepted: 08/18/2021] [Indexed: 12/30/2022] Open
Abstract
An improved understanding of sterol and lipid abnormalities in individuals with autism spectrum disorder (ASD) could lead to personalized treatment approaches. Toward this end, in blood, we identified reduced synthesis of cholesterol in families with ≥2 children with ASD participating with the Autism Genetic Resource Exchange (AGRE), as well as reduced amounts of high-density lipoprotein cholesterol (HDL), apolipoprotein A1 (ApoA1) and apolipoprotein B (ApoB), with 19.9% of the subjects presenting with apolipoprotein patterns similar to hypolipidemic clinical syndromes and 30% with either or both ApoA1 and ApoB less than the fifth centile. Subjects with levels less than the fifth centile of HDL or ApoA1 or ApoA1 + ApoB had lower adaptive functioning than other individuals with ASD, and hypocholesterolemic subjects had apolipoprotein deficits significantly divergent from either typically developing individuals participating in National Institutes of Health or the National Health and Nutrition Examination Survey III.
Collapse
|
11
|
Lam M, Chen CY, Ge T, Xia Y, Hill DW, Trampush JW, Yu J, Knowles E, Davies G, Stahl EA, Huckins L, Liewald DC, Djurovic S, Melle I, Christoforou A, Reinvang I, DeRosse P, Lundervold AJ, Steen VM, Espeseth T, Räikkönen K, Widen E, Palotie A, Eriksson JG, Giegling I, Konte B, Hartmann AM, Roussos P, Giakoumaki S, Burdick KE, Payton A, Ollier W, Chiba-Falek O, Koltai DC, Need AC, Cirulli ET, Voineskos AN, Stefanis NC, Avramopoulos D, Hatzimanolis A, Smyrnis N, Bilder RM, Freimer NB, Cannon TD, London E, Poldrack RA, Sabb FW, Congdon E, Conley ED, Scult MA, Dickinson D, Straub RE, Donohoe G, Morris D, Corvin A, Gill M, Hariri AR, Weinberger DR, Pendleton N, Bitsios P, Rujescu D, Lahti J, Le Hellard S, Keller MC, Andreassen OA, Deary IJ, Glahn DC, Huang H, Liu C, Malhotra AK, Lencz T. Identifying nootropic drug targets via large-scale cognitive GWAS and transcriptomics. Neuropsychopharmacology 2021; 46:1788-1801. [PMID: 34035472 PMCID: PMC8357785 DOI: 10.1038/s41386-021-01023-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 02/22/2021] [Accepted: 04/12/2021] [Indexed: 02/05/2023]
Abstract
Broad-based cognitive deficits are an enduring and disabling symptom for many patients with severe mental illness, and these impairments are inadequately addressed by current medications. While novel drug targets for schizophrenia and depression have emerged from recent large-scale genome-wide association studies (GWAS) of these psychiatric disorders, GWAS of general cognitive ability can suggest potential targets for nootropic drug repurposing. Here, we (1) meta-analyze results from two recent cognitive GWAS to further enhance power for locus discovery; (2) employ several complementary transcriptomic methods to identify genes in these loci that are credibly associated with cognition; and (3) further annotate the resulting genes using multiple chemoinformatic databases to identify "druggable" targets. Using our meta-analytic data set (N = 373,617), we identified 241 independent cognition-associated loci (29 novel), and 76 genes were identified by 2 or more methods of gene identification. Actin and chromatin binding gene sets were identified as novel pathways that could be targeted via drug repurposing. Leveraging our transcriptomic and chemoinformatic databases, we identified 16 putative genes targeted by existing drugs potentially available for cognitive repurposing.
Collapse
Affiliation(s)
- Max Lam
- Division of Psychiatry Research, The Zucker Hillside Hospital, Glen Oaks, NY, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Institute for Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Institute of Mental Health, Singapore, Singapore
| | - Chia-Yen Chen
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Biogen, Inc, Cambridge, MA, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Tian Ge
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Yan Xia
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
- Psychiatry Department, SUNY Upstate Medical University, Syracuse, NY, USA
| | - David W Hill
- Lothian Birth Cohorts, University of Edinburgh, Edinburgh, Scotland, UK
- Lothian Birth Cohorts group, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Joey W Trampush
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jin Yu
- Division of Psychiatry Research, The Zucker Hillside Hospital, Glen Oaks, NY, USA
| | - Emma Knowles
- Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Olin Neuropsychic Research Center, Institute of Living, Hartford Hospital, Hartford, CT, USA
| | - Gail Davies
- Lothian Birth Cohorts, University of Edinburgh, Edinburgh, Scotland, UK
- Lothian Birth Cohorts group, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Eli A Stahl
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science and Institute for Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Laura Huckins
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science and Institute for Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David C Liewald
- Lothian Birth Cohorts group, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
- NORMENT, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ingrid Melle
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Andrea Christoforou
- Spaulding Rehabilitation Hospital Boston, Charlestown, MA, USA
- Dr. Einar Martens Research Group for Biological Psychiatry, Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Ivar Reinvang
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Pamela DeRosse
- Division of Psychiatry Research, The Zucker Hillside Hospital, Glen Oaks, NY, USA
- Institute for Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Department of Psychiatry, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Astri J Lundervold
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
| | - Vidar M Steen
- NORMENT, Department of Clinical Science, University of Bergen, Bergen, Norway
- Dr. Einar Martens Research Group for Biological Psychiatry, Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Thomas Espeseth
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Katri Räikkönen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Elisabeth Widen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, UK
- Department of Medical Genetics, University of Helsinki and University Central Hospital, Helsinki, Finland
| | - Johan G Eriksson
- Department of General Practice, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Folkhälsan Research Center, Helsinki, Finland
| | - Ina Giegling
- Department of Psychiatry, Martin Luther University of Halle-Wittenberg, Halle, Germany
| | - Bettina Konte
- Department of Psychiatry, Martin Luther University of Halle-Wittenberg, Halle, Germany
| | - Annette M Hartmann
- Department of Psychiatry, Martin Luther University of Halle-Wittenberg, Halle, Germany
| | - Panos Roussos
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science and Institute for Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research, Education, and Clinical Center (VISN 2), James J. Peters VA Medical Center, Bronx, NY, USA
| | | | - Katherine E Burdick
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research, Education, and Clinical Center (VISN 2), James J. Peters VA Medical Center, Bronx, NY, USA
- Department of Psychiatry - Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Antony Payton
- Division of Informatics, Imaging & Data Sciences, School of Health Sciences, The University of Manchester, Manchester, UK
| | - William Ollier
- Centre for Epidemiology, Division of Population Health, Health Services Research & Primary Care, The University of Manchester, Manchester, UK
- School of Healthcare Sciences, Manchester Metropolitan University, Manchester, United Kingdom
| | - Ornit Chiba-Falek
- Division of Translational Brain Sciences, Department of Neurology, Bryan Alzheimer's Disease Research Center, and Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, USA
| | - Deborah C Koltai
- Psychiatry and Behavioral Sciences, Division of Medical Psychology, and Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Anna C Need
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | | | - Aristotle N Voineskos
- Campbell Family Mental Health Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, ON, Canada
| | - Nikos C Stefanis
- 2nd Department of Psychiatry, National and Kapodistrian University of Athens Medical School, University General Hospital "ATTIKON", Athens, Greece
- University Mental Health Research Institute, Athens, Greece
- Neurobiology Research Institute, Theodor-Theohari Cozzika Foundation, Athens, Greece
| | - Dimitrios Avramopoulos
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alex Hatzimanolis
- 2nd Department of Psychiatry, National and Kapodistrian University of Athens Medical School, University General Hospital "ATTIKON", Athens, Greece
- University Mental Health Research Institute, Athens, Greece
- Neurobiology Research Institute, Theodor-Theohari Cozzika Foundation, Athens, Greece
| | - Nikolaos Smyrnis
- 2nd Department of Psychiatry, National and Kapodistrian University of Athens Medical School, University General Hospital "ATTIKON", Athens, Greece
- University Mental Health Research Institute, Athens, Greece
| | - Robert M Bilder
- UCLA Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA, USA
| | - Nelson B Freimer
- UCLA Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA, USA
| | - Tyrone D Cannon
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychology, Yale University, New Haven, CT, USA
| | - Edythe London
- UCLA Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA, USA
| | | | - Fred W Sabb
- Robert and Beverly Lewis Center for Neuroimaging, University of Oregon, Eugene, OR, USA
| | - Eliza Congdon
- UCLA Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA, USA
| | | | - Matthew A Scult
- Weill Cornell Psychiatry at NewYork-Presbyterian, Weill Cornell Medical Center, New York, NY, USA
- Laboratory of NeuroGenetics, Department of Psychology & Neuroscience, Duke University, Durham, NC, USA
| | - Dwight Dickinson
- Clinical and Translational Neuroscience Branch, Intramural Research Program, National Institute of Mental Health, National Institute of Health, Bethesda, MD, USA
| | - Richard E Straub
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
| | - Gary Donohoe
- Neuroimaging, Cognition & Genomics (NICOG) Centre, School of Psychology and Discipline of Biochemistry, National University of Ireland, Galway, Ireland
| | - Derek Morris
- Neuroimaging, Cognition & Genomics (NICOG) Centre, School of Psychology and Discipline of Biochemistry, National University of Ireland, Galway, Ireland
| | - Aiden Corvin
- Neuropsychiatric Genetics Research Group, Department of Psychiatry and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Michael Gill
- Neuropsychiatric Genetics Research Group, Department of Psychiatry and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Ahmad R Hariri
- Laboratory of NeuroGenetics, Department of Psychology & Neuroscience, Duke University, Durham, NC, USA
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
| | - Neil Pendleton
- Division of Neuroscience and Experimental Psychology/School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, Salford Royal NHS Foundation Trust, University of Manchester, Manchester, UK
| | - Panos Bitsios
- Department of Psychiatry and Behavioral Sciences, Faculty of Medicine, University of Crete, Heraklion, Crete, GR, Greece
| | - Dan Rujescu
- Department of Psychiatry, Martin Luther University of Halle-Wittenberg, Halle, Germany
| | - Jari Lahti
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Helsinki Collegium for Advanced Studies, University of Helsinki, Helsinki, Finland
| | - Stephanie Le Hellard
- NORMENT, Department of Clinical Science, University of Bergen, Bergen, Norway
- Dr. Einar Martens Research Group for Biological Psychiatry, Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Matthew C Keller
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO, USA
| | - Ole A Andreassen
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ian J Deary
- Lothian Birth Cohorts, University of Edinburgh, Edinburgh, Scotland, UK
- Lothian Birth Cohorts group, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - David C Glahn
- Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Olin Neuropsychic Research Center, Institute of Living, Hartford Hospital, Hartford, CT, USA
| | - Hailiang Huang
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Chunyu Liu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
- Psychiatry Department, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Anil K Malhotra
- Division of Psychiatry Research, The Zucker Hillside Hospital, Glen Oaks, NY, USA
- Institute for Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Department of Psychiatry, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Todd Lencz
- Division of Psychiatry Research, The Zucker Hillside Hospital, Glen Oaks, NY, USA.
- Institute for Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY, USA.
- Department of Psychiatry, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA.
| |
Collapse
|
12
|
Kadri S, El Ayed M, Kadri A, Limam F, Aouani E, Mokni M. Protective effect of grape seed extract and orlistat co-treatment against stroke: Effect on oxidative stress and energy failure. Biomed Pharmacother 2021; 136:111282. [PMID: 33485068 DOI: 10.1016/j.biopha.2021.111282] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/10/2021] [Accepted: 01/12/2021] [Indexed: 11/17/2022] Open
Abstract
Ischemic stroke is a major health concern and a leading cause of mortality worldwide. Oxidative stress is an early event in the course of stroke inducing neuro-inflammation and cell death. Grape seed extract (GSE) is a natural phytochemical mixture exhibiting antioxidant, anti-inflammatory and neuroprotective properties. Orlistat (ORL) is an anti-obesity agent and a gastro-intestinal lipase inhibitor which showed recently beneficial effects on brain lipotoxicity. Recent studies reported the increase of lipase activity upon stroke which led us to investigate the neuroprotective effect of ORL on rat brain I/R injury as well as the putative synergism with GSE. I/R insult infarcted the brain parenchyma as assessed by TTC staining, induced an oxidative stress as revealed by increased lipoperoxidation along with alteration of antioxidant enzymes activities which was corrected using the cotreatment of ORL + GSE. I/R also disturbed the main metabolic pathways involved in brain fueling as glycolysis, neoglucogenesis, glycogenolysis, TCA cycle and electron transfer chain (ETC) complexes. These disturbances were also corrected with the cotreatment ORL + GSE which maintained energetic activities near to the control level. I/R also disrupted transition metals distribution, along with associated enzymes as tyrosinase, LDH or glutamine synthetase activities and induced hippocampal inflammation as revealed by glycogen depletion from dentate gyrus area along with depressed anti-inflammatory IL1β cytokine and increased pro-inflammatory CD68 antigen. Interestingly almost all I/R-induced disturbances were corrected either partially upon ORL and GSE on their own and the best neuroprotection was obtained in the presence of both drugs (ORL + GSE) enabling robust neuroprotection of the sub granular zone within hippocampal dentate gyrus area.
Collapse
Affiliation(s)
- Safwen Kadri
- Bioactive Substances Laboratory, Biotechnology Centre, Technopolis Borj-Cedria, BP-901, 2050, Hammam-Lif, Tunisia.
| | - Mohamed El Ayed
- Bioactive Substances Laboratory, Biotechnology Centre, Technopolis Borj-Cedria, BP-901, 2050, Hammam-Lif, Tunisia
| | - Amal Kadri
- Bioactive Substances Laboratory, Biotechnology Centre, Technopolis Borj-Cedria, BP-901, 2050, Hammam-Lif, Tunisia
| | - Ferid Limam
- Bioactive Substances Laboratory, Biotechnology Centre, Technopolis Borj-Cedria, BP-901, 2050, Hammam-Lif, Tunisia
| | - Ezzedine Aouani
- Bioactive Substances Laboratory, Biotechnology Centre, Technopolis Borj-Cedria, BP-901, 2050, Hammam-Lif, Tunisia
| | - Meherzia Mokni
- Bioactive Substances Laboratory, Biotechnology Centre, Technopolis Borj-Cedria, BP-901, 2050, Hammam-Lif, Tunisia
| |
Collapse
|
13
|
Abstract
OBJECTIVES The current study examines the placental and maternal lipid profile and expression of genes involved in placental lipid metabolism in women with preeclampsia. METHODS The current study includes normotensive control women (n = 40) and women with preeclampsia (n = 39). Preeclampsia women were further classified into women delivering at term preeclampsia (T-PE; n = 15) and preterm preeclampsia (PT-PE; n = 24). RESULTS There were no significant differences in maternal lipid profile between the T-PE and normotensive control groups. Maternal plasma VLDL (P < 0.05) and ratios of total cholesterol : HDL (P < 0.05), atherogenic index [log (triglycerides/HDL)] (P < 0.01) and apolipoprotein B : apolipoprotein A (P < 0.05) were higher in the PT-PE group as compared with the normotensive control group. Placental total cholesterol and HDL levels were higher (P < 0.05) in the T-PE as compared with the normotensive control group. Higher placental triglycerides (P < 0.05) were observed in PT-PE group compared with T-PE group. Placental mRNA levels of peroxisome proliferator activated receptor α, carnitine palmitoyl transferase-1, cluster of differentiation 36 and lipoprotein lipases were lower (P < 0.05) in the PT-PE than normotensive control group. A negative association of mRNA levels of peroxisome proliferator activated receptor α (r = -0.246, P = 0.032; r = -0.308, P = 0.007, respectively), carnitine palmitoyl transferase-1 (r = -0.292, P = 0.011; r = -0.366, P = 0.001), lipoprotein lipases (r = -0.296, P = 0.010; r = -0.254, P = 0.028) with SBP and DBP was observed. There was a positive association of placental triglycerides (r = 0.244, P = 0.031) with DBP. CONCLUSION Women with preeclampsia exhibit higher lipid : lipoprotein ratios suggesting an atherogenic state particularly in women delivering preterm. Lower expression of genes involved in placental fatty acid oxidation and transport was also observed in preeclampsia.
Collapse
Affiliation(s)
- Amrita A Khaire
- Mother and Child Health, Interactive Research School for Health Affairs, Bharati Vidyapeeth (Deemed to be University)
| | - Shivani R Thakar
- Mother and Child Health, Interactive Research School for Health Affairs, Bharati Vidyapeeth (Deemed to be University)
| | - Girija N Wagh
- Department of Obstetrics and Gynecology, Bharati Medical College and Hospital, Bharati Vidyapeeth University, Pune, India
| | - Sadhana R Joshi
- Mother and Child Health, Interactive Research School for Health Affairs, Bharati Vidyapeeth (Deemed to be University)
| |
Collapse
|
14
|
Mizuno S, Hirota JN, Ishii C, Iwasaki H, Sano Y, Furuichi T. Comprehensive Profiling of Gene Expression in the Cerebral Cortex and Striatum of BTBRTF/ArtRbrc Mice Compared to C57BL/6J Mice. Front Cell Neurosci 2020; 14:595607. [PMID: 33362469 PMCID: PMC7758463 DOI: 10.3389/fncel.2020.595607] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 11/09/2020] [Indexed: 12/19/2022] Open
Abstract
Mouse line BTBR T+ Iptr3tf/J (hereafter referred as to BTBR/J) is a mouse strain that shows lower sociability compared to the C57BL/6J mouse strain (B6) and thus is often utilized as a model for autism spectrum disorder (ASD). In this study, we utilized another subline, BTBRTF/ArtRbrc (hereafter referred as to BTBR/R), and analyzed the associated brain transcriptome compared to B6 mice using microarray analysis, quantitative RT-PCR analysis, various bioinformatics analyses, and in situ hybridization. We focused on the cerebral cortex and the striatum, both of which are thought to be brain circuits associated with ASD symptoms. The transcriptome profiling identified 1,280 differentially expressed genes (DEGs; 974 downregulated and 306 upregulated genes, including 498 non-coding RNAs [ncRNAs]) in BTBR/R mice compared to B6 mice. Among these DEGs, 53 genes were consistent with ASD-related genes already established. Gene Ontology (GO) enrichment analysis highlighted 78 annotations (GO terms) including DNA/chromatin regulation, transcriptional/translational regulation, intercellular signaling, metabolism, immune signaling, and neurotransmitter/synaptic transmission-related terms. RNA interaction analysis revealed novel RNA–RNA networks, including 227 ASD-related genes. Weighted correlation network analysis highlighted 10 enriched modules including DNA/chromatin regulation, neurotransmitter/synaptic transmission, and transcriptional/translational regulation. Finally, the behavioral analyses showed that, compared to B6 mice, BTBR/R mice have mild but significant deficits in social novelty recognition and repetitive behavior. In addition, the BTBR/R data were comprehensively compared with those reported in the previous studies of human subjects with ASD as well as ASD animal models, including BTBR/J mice. Our results allow us to propose potentially important genes, ncRNAs, and RNA interactions. Analysis of the altered brain transcriptome data of the BTBR/R and BTBR/J sublines can contribute to the understanding of the genetic underpinnings of autism susceptibility.
Collapse
Affiliation(s)
- Shota Mizuno
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Japan
| | - Jun-Na Hirota
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Japan
| | - Chiaki Ishii
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Japan
| | - Hirohide Iwasaki
- Department of Anatomy, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yoshitake Sano
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Japan
| | - Teiichi Furuichi
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Japan
| |
Collapse
|
15
|
Erukainure OL, Salau VF, Alabi OO, Ebuehi OAT, Koorbanally NA, Islam MS. Casein micelles from bovine Milk exerts Neuroprotection by stalling metabolic complications linked to oxidative brain injury. Metab Brain Dis 2020; 35:1417-1428. [PMID: 32990928 DOI: 10.1007/s11011-020-00621-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 09/22/2020] [Indexed: 11/30/2022]
Abstract
Caseins are the most abundant milk proteins in mammalian species and are assembled in supra-macromolecular structures called micelles. In this study, the microstructural properties, particle size, and elemental composition of isolated casein from bovine milk and its therapeutic effect on oxidative and cholinergic activities linked to dementia in oxidative brain injury were investigated. SEM analysis of the isolated casein micelles from skimmed fresh bovine milk revealed spherical colloid aggregates, while TEM analysis revealed dispersed spherical particles with a mean size of 63.15 ± 4.77 nm. SEM-EDX analysis revealed clusters of carbon, oxygen, sulfur, copper, sodium, magnesium, potassium, iron, and selenium. Treatment of oxidative brain injury with the isolated casein micelles led to elevated levels of GSH, SOD, catalase, ENTPDase, 5'NTPase activities, while concomitantly suppressing MDA, cholesterol, HDL-c levels, acetylcholinesterase and lipase activities. Treatment with the isolated casein micelles led to complete depletion of oxidative generated lipid metabolites, while deactivating oxidative-activated lipid metabolic pathways. These results indicate the microstructural properties, particle size, elemental composition, and antioxidant neuroprotective effect of casein micelles from bovine milk. Thus, demonstrating the nutraceutical properties of milk in the management of oxidative induced cognitive impairment.
Collapse
Affiliation(s)
- Ochuko L Erukainure
- Department of Pharmacology, University of the Free State, Bloemfontein, 9300, South Africa.
| | - Veronica F Salau
- Department of Biochemistry, University of KwaZulu-Natal, Westville Campus, Durban, 4000, South Africa
| | - Opeyemi O Alabi
- Department of Food Technology, Federal University, Oye-Ekiti, Ekiti, Nigeria
| | | | - Neil A Koorbanally
- School of Chemistry and Physics, University of KwaZulu-Natal, Westville Campus, Durban, 4000, South Africa
| | - Md Shahidul Islam
- Department of Biochemistry, University of KwaZulu-Natal, Westville Campus, Durban, 4000, South Africa
| |
Collapse
|
16
|
Cross-sectional analysis of plasma and CSF metabolomic markers in Huntington's disease for participants of varying functional disability: a pilot study. Sci Rep 2020; 10:20490. [PMID: 33235276 PMCID: PMC7686309 DOI: 10.1038/s41598-020-77526-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 11/04/2020] [Indexed: 01/24/2023] Open
Abstract
Huntington’s Disease (HD) is a progressive, fatal neurodegenerative condition. While generally considered for its devastating neurological phenotype, disturbances in other organ systems and metabolic pathways outside the brain have attracted attention for possible relevance to HD pathology, potential as therapeutic targets, or use as biomarkers of progression. In addition, it is not established how metabolic changes in the HD brain correlate to progression across the full spectrum of early to late-stage disease. In this pilot study, we sought to explore the metabolic profile across manifest HD from early to advanced clinical staging through metabolomic analysis by mass spectrometry in plasma and cerebrospinal fluid (CSF). With disease progression, we observed nominally significant increases in plasma arginine, citrulline, and glycine, with decreases in total and d-serine, cholesterol esters, diacylglycerides, triacylglycerides, phosphatidylcholines, phosphatidylethanolamines, and sphingomyelins. In CSF, worsening disease was associated with nominally significant increases in NAD+, arginine, saturated long chain free fatty acids, diacylglycerides, triacylglycerides, and sphingomyelins. Notably, diacylglycerides and triacylglyceride species associated with clinical progression were different between plasma and CSF, suggesting different metabolic preferences for these compartments. Increasing NAD+ levels strongly correlating with disease progression was an unexpected finding. Our data suggest that defects in the urea cycle, glycine, and serine metabolism may be underrecognized in the progression HD pathology, and merit further study for possible therapeutic relevance.
Collapse
|
17
|
Kadri S, El Ayed M, Limam F, Aouani E, Mokni M. Protective effect of (Xenical+GSF) against I/R-induced blood brain barrier disruption, ionic edema, lipid deregulation and neuroinflammation. Microvasc Res 2020; 132:104054. [PMID: 32768464 DOI: 10.1016/j.mvr.2020.104054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/03/2020] [Accepted: 08/03/2020] [Indexed: 12/09/2022]
Abstract
Ischemic stroke is a leading cause of mortality worldwide that occurs following the reduction or interruption of blood brain supply, characterized by a cascade of early events as oxidative stress and ensuing neuro-inflammation, energy failure and the burst of intracellular Ca++ resulting in activation of phospholipases and large increase in FFA including arachidonic acid, ultimately leading to nervous cell death. Grape Seed Flour (GSF) is a complex polyphenolic mixture harboring antioxidant, anti-inflammatory and neuroprotective properties. Orlistat (Xenical ™,Xe) is a gastro-intestinal lipase inhibitor and an anti-obesity agent. In an earlier study we reported the higher efficiency in neuroprotection against HFD-induced brain lipotoxicity when combining the two drugs (GSF + Xe). As a result repurposing Xe as an adjunct to GSF therapy against stroke appeared relevant and worthy of investigation. I/R insult disrupted the blood brain barrier (BBB) as assessed by EB dye extravasation, increased water and Na+ within the brain. Ultrastructurally I/R altered the brain blood capillaries at the vicinity of hippocampus dentate gyrus area as assessed by transmission and scanning electron microscopy. I/R altered lipid metabolism as revealed by LDL/HDL ratio, lipase activity, and FFA profiles. Moreover, I/R induced neuro-inflammation as assessed by down-regulation of anti-inflammatory CD 56 and up-regulation of pro-inflammatory CD 68 antigen. Importantly almost all I/R-induced disturbances were retrieved partially upon Xe or GSF on their own, and optimally when combining the two drugs. Xe per se is protective against I/R injury and the best neuroprotection was obtained when associating low dosage Xe with high dosage GSF, enabling neuroprevention and cell survival within hippocampus dentate gyrus area as revealed by increased staining of Ki 67 proliferation biomarker.
Collapse
Affiliation(s)
- Safwen Kadri
- Bioactive Substances Laboratory, Biotechnology Centre, Technopolis Borj-Cedria, BP-901, 2050 Hammam-Lif, Tunis, Tunisia.
| | - Mohamed El Ayed
- Bioactive Substances Laboratory, Biotechnology Centre, Technopolis Borj-Cedria, BP-901, 2050 Hammam-Lif, Tunis, Tunisia
| | - Ferid Limam
- Bioactive Substances Laboratory, Biotechnology Centre, Technopolis Borj-Cedria, BP-901, 2050 Hammam-Lif, Tunis, Tunisia
| | - Ezzedine Aouani
- Bioactive Substances Laboratory, Biotechnology Centre, Technopolis Borj-Cedria, BP-901, 2050 Hammam-Lif, Tunis, Tunisia
| | - Meherzia Mokni
- Bioactive Substances Laboratory, Biotechnology Centre, Technopolis Borj-Cedria, BP-901, 2050 Hammam-Lif, Tunis, Tunisia
| |
Collapse
|
18
|
Zhang J, Liu Y, Wang S, Que R, Zhao W, An L. Exploration of the Molecular Mechanism for Lipoprotein Lipase Expression Variations in SH-SY5Y Cells Exposed to Different Doses of Amyloid-Beta Protein. Front Aging Neurosci 2020; 12:132. [PMID: 32477101 PMCID: PMC7235190 DOI: 10.3389/fnagi.2020.00132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 04/20/2020] [Indexed: 11/13/2022] Open
Abstract
Progressive accumulation of amyloid-β (Aβ) plaques in the brain is a characteristic pathological change in Alzheimer's disease (AD). We previously found the expression of lipoprotein lipase (LPL) was increased in SH-SY5Y cells exposed to low-dose Aβ and decreased in cells with high-dose Aβ exposure, but the molecular mechanism is still unclear. Based on previous studies, the opposite regulation of histone deacetylase2 (HDAC2) and HDAC3 on LPL expression probably explain the above molecular mechanism, in which microRNA-29a and peroxisome proliferator-activated receptor γ (PPARγ) may be involved. This study further revealed the mechanism of HDAC2 and HDAC3 on conversely regulating LPL expression. The results showed that HDAC2 down-regulated microRNA-29a by decreasing histone acetylation (Ace-H3K9) level in its promoter region, subsequently increasing LPL expression directly or through PPARγ/LPL pathway; HDAC3 decreased LPL expression through inhibiting Ace-H3K9 levels in LPL and PPARγ promoter regions and up-regulating microRNA-29a. This study also found that with increasing concentrations of Aβ in cells, HDAC2 and HDAC3 expression were gradually increased, and Ace-H3K9 levels in LPL and PPARγ promoter region regulated by HDAC3 were decreased correspondingly, while Ace-H3K9 levels in microRNA-29a promoter region modulated by HDAC2 were not decreased gradually but presented a U-shaped trend. These may lead to the results that a U-shaped alteration in microRNA-29a expression, subsequently leading to an inverse U-shaped alteration in PPARγ or LPL expression. In conclusion, HDAC2 and HDAC3 at least partly mediate LPL expression variations in different concentrations of Aβ exposed SH-SY5Y cells, in which microRNA-29a and PPARγ are involved, and the histone acetylation level in microRNA-29a promoter region plays a key role.
Collapse
Affiliation(s)
- Jingzhu Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, China Medical University, Shenyang, China
| | - Yufan Liu
- China Medical University-The Queen's University of Belfast Joint College, China Medical University, Shenyang, China
| | - Sihui Wang
- Department of Nutrition and Food Hygiene, School of Public Health, China Medical University, Shenyang, China
| | - Ran Que
- Department of Nutrition and Food Hygiene, School of Public Health, China Medical University, Shenyang, China
| | - Weidong Zhao
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, China Medical University, Shenyang, China.,Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Li An
- Department of Nutrition and Food Hygiene, School of Public Health, China Medical University, Shenyang, China
| |
Collapse
|
19
|
Alam J, Sharma L. Potential Enzymatic Targets in Alzheimer's: A Comprehensive Review. Curr Drug Targets 2020; 20:316-339. [PMID: 30124150 DOI: 10.2174/1389450119666180820104723] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 06/23/2018] [Accepted: 08/15/2018] [Indexed: 12/13/2022]
Abstract
Alzheimer's, a degenerative cause of the brain cells, is called as a progressive neurodegenerative disease and appears to have a heterogeneous etiology with main emphasis on amyloid-cascade and hyperphosphorylated tau-cascade hypotheses, that are directly linked with macromolecules called enzymes such as β- & γ-secretases, colinesterases, transglutaminases, and glycogen synthase kinase (GSK-3), cyclin-dependent kinase (cdk-5), microtubule affinity-regulating kinase (MARK). The catalytic activity of the above enzymes is the result of cognitive deficits, memory impairment and synaptic dysfunction and loss, and ultimately neuronal death. However, some other enzymes also lead to these dysfunctional events when reduced to their normal activities and levels in the brain, such as α- secretase, protein kinase C, phosphatases etc; metabolized to neurotransmitters, enzymes like monoamine oxidase (MAO), catechol-O-methyltransferase (COMT) etc. or these abnormalities can occur when enzymes act by other mechanisms such as phosphodiesterase reduces brain nucleotides (cGMP and cAMP) levels, phospholipase A2: PLA2 is associated with reactive oxygen species (ROS) production etc. On therapeutic fronts, several significant clinical trials are underway by targeting different enzymes for development of new therapeutics to treat Alzheimer's, such as inhibitors for β-secretase, GSK-3, MAO, phosphodiesterase, PLA2, cholinesterases etc, modulators of α- & γ-secretase activities and activators for protein kinase C, sirtuins etc. The last decades have perceived an increasing focus on findings and search for new putative and novel enzymatic targets for Alzheimer's. Here, we review the functions, pathological roles, and worth of almost all the Alzheimer's associated enzymes that address to therapeutic strategies and preventive approaches for treatment of Alzheimer's.
Collapse
Affiliation(s)
- Jahangir Alam
- School of Pharmaceutical Sciences, Shoolini University, Solan, H.P., Pin 173229, India
| | - Lalit Sharma
- School of Pharmaceutical Sciences, Shoolini University, Solan, H.P., Pin 173229, India
| |
Collapse
|
20
|
Schartl M, Kneitz S, Volkoff H, Adolfi M, Schmidt C, Fischer P, Minx P, Tomlinson C, Meyer A, Warren WC. The Piranha Genome Provides Molecular Insight Associated to Its Unique Feeding Behavior. Genome Biol Evol 2020; 11:2099-2106. [PMID: 31282935 PMCID: PMC6681833 DOI: 10.1093/gbe/evz139] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2019] [Indexed: 12/27/2022] Open
Abstract
The piranha enjoys notoriety due to its infamous predatory behavior but much is still not understood about its evolutionary origins and the underlying molecular mechanisms for its unusual feeding biology. We sequenced and assembled the red-bellied piranha (Pygocentrus nattereri) genome to aid future phenotypic and genetic investigations. The assembled draft genome is similar to other related fishes in repeat composition and gene count. Our evaluation of genes under positive selection suggests candidates for adaptations of piranhas’ feeding behavior in neural functions, behavior, and regulation of energy metabolism. In the fasted brain, we find genes differentially expressed that are involved in lipid metabolism and appetite regulation as well as genes that may control the aggression/boldness behavior of hungry piranhas. Our first analysis of the piranha genome offers new insight and resources for the study of piranha biology and for feeding motivation and starvation in other organisms.
Collapse
Affiliation(s)
- Manfred Schartl
- Physiologische Chemie, Biozentrum, University of Würzburg, Germany.,Comprehensive Cancer Center Mainfranken, University Clinic Würzburg, Germany.,Hagler Institute for Advanced Study, Texas A&M University.,Department of Biology, Texas A&M University
| | - Susanne Kneitz
- Physiologische Chemie, Biozentrum, University of Würzburg, Germany
| | - Helene Volkoff
- Department of Biology, Memorial University of Newfoundland, St John's, Canada.,Department of Biochemistry, Memorial University of Newfoundland, St John's, Canada
| | - Mateus Adolfi
- Physiologische Chemie, Biozentrum, University of Würzburg, Germany
| | - Cornelia Schmidt
- Physiologische Chemie, Biozentrum, University of Würzburg, Germany
| | - Petra Fischer
- Physiologische Chemie, Biozentrum, University of Würzburg, Germany
| | - Patrick Minx
- McDonnell Genome Institute, Washington University School of Medicine
| | - Chad Tomlinson
- McDonnell Genome Institute, Washington University School of Medicine
| | - Axel Meyer
- Chair in Zoology and Evolutionary Biology, University of Konstanz, Germany
| | - Wesley C Warren
- McDonnell Genome Institute, Washington University School of Medicine.,Bond Life Sciences Center, University of Missouri
| |
Collapse
|
21
|
Murali N, Browne RW, Fellows Maxwell K, Bodziak ML, Jakimovski D, Hagemeier J, Bergsland N, Weinstock-Guttman B, Zivadinov R, Ramanathan M. Cholesterol and neurodegeneration: longitudinal changes in serum cholesterol biomarkers are associated with new lesions and gray matter atrophy in multiple sclerosis over 5 years of follow-up. Eur J Neurol 2019; 27:188-e4. [PMID: 31369181 DOI: 10.1111/ene.14055] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 07/22/2019] [Indexed: 01/25/2023]
Abstract
BACKGROUND AND PURPOSE Cholesterol is an important structural component of myelin and essential for brain homeostasis. Our objective was to investigate whether longitudinal changes in cholesterol biomarkers are associated with neurodegeneration in multiple sclerosis (MS). METHODS This prospective, longitudinal study (n = 154) included 41 healthy controls, 76 relapsing-remitting MS subjects and 37 progressive MS subjects. Neurological examination, brain magnetic resonance imaging and blood samples were obtained at baseline and at 5-year follow-up visits. Cholesterol biomarkers measured included plasma total cholesterol, high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol and the apolipoproteins ApoA-I, Apo-II, ApoB, ApoC-II and ApoE. Key cholesterol pathway single nucleotide polymorphisms were genotyped. RESULTS Greater percentage increases in HDL-C and ApoA-I levels were associated with a lower rate of gray matter and cortical volume loss. Greater percentage increases in low-density lipoprotein cholesterol were associated with increases in new T2 lesions. The percentage increases in HDL-C (P = 0.032) and ApoA-I (P = 0.007) were smaller in patients with relapsing-remitting MS at baseline who converted to secondary progressive MS during the 5-year follow-up period. Changes in HDL-C and ApoA-I were associated with lipoprotein lipase rs328 genotype status. CONCLUSIONS Increases in HDL-C and ApoA-I have protective associations with magnetic resonance imaging measures of neurodegeneration in MS.
Collapse
Affiliation(s)
- N Murali
- Departments of Pharmaceutical Sciences, State University of New York, Buffalo, NY, USA
| | - R W Browne
- Biotechnical and Clinical Laboratory Sciences, State University of New York, Buffalo, NY, USA
| | - K Fellows Maxwell
- Departments of Pharmaceutical Sciences, State University of New York, Buffalo, NY, USA
| | - M L Bodziak
- Biotechnical and Clinical Laboratory Sciences, State University of New York, Buffalo, NY, USA
| | - D Jakimovski
- Buffalo Neuroimaging Analysis Center, State University of New York, Buffalo, NY, USA
| | - J Hagemeier
- Buffalo Neuroimaging Analysis Center, State University of New York, Buffalo, NY, USA
| | - N Bergsland
- Buffalo Neuroimaging Analysis Center, State University of New York, Buffalo, NY, USA
| | | | - R Zivadinov
- Buffalo Neuroimaging Analysis Center, State University of New York, Buffalo, NY, USA.,Neurology, State University of New York, Buffalo, NY, USA
| | - M Ramanathan
- Departments of Pharmaceutical Sciences, State University of New York, Buffalo, NY, USA.,Neurology, State University of New York, Buffalo, NY, USA
| |
Collapse
|
22
|
Le Foll C. Hypothalamic Fatty Acids and Ketone Bodies Sensing and Role of FAT/CD36 in the Regulation of Food Intake. Front Physiol 2019; 10:1036. [PMID: 31474875 PMCID: PMC6702519 DOI: 10.3389/fphys.2019.01036] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 07/29/2019] [Indexed: 12/19/2022] Open
Abstract
The obesity and type-2 diabetes epidemic is escalating and represents one of the costliest biomedical challenges confronting modern society. Moreover, the increasing consumption of high fat food is often correlated with an increase in body mass index. In people predisposed to be obese or already obese, the impaired ability of the brain to monitor and respond to alterations in fatty acid (FA) metabolism is increasingly recognized as playing a role in the pathophysiological development of these disorders. The brain senses and regulates metabolism using highly specialized nutrient-sensing neurons located mainly in the hypothalamus. The same neurons are able to detect variation in the extracellular levels of glucose, FA and ketone bodies as a way to monitor nutrient availability and to alter its own activity. In addition, glial cells such as astrocytes create major connections to neurons and form a tight relationship to closely regulate nutrient uptake and metabolism. This review will examine the different pathways by which neurons are able to detect free fatty acids (FFA) to alter its activity and how high fat diet (HFD)-astrocytes induced ketone bodies production interplays with neuronal FA sensing. The role of HFD-induced inflammation and how FA modulate the reward system will also be investigated here.
Collapse
Affiliation(s)
- Christelle Le Foll
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
23
|
Kamermans A, Rijnsburger M, Chakraborty A, van der Pol S, de Vries HE, van Horssen J. Reduced Angiopoietin-Like 4 Expression in Multiple Sclerosis Lesions Facilitates Lipid Uptake by Phagocytes via Modulation of Lipoprotein-Lipase Activity. Front Immunol 2019; 10:950. [PMID: 31130950 PMCID: PMC6509157 DOI: 10.3389/fimmu.2019.00950] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 04/12/2019] [Indexed: 12/17/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disorder of the central nervous system (CNS) characterized by the presence of focal demyelinated plaques. Sufficient clearance of myelin and cellular debris is one of the requirements for proper tissue repair and remyelination. The mechanisms underlying the clearance of such debris by phagocytes are not fully understood, but recent findings suggest a prominent role for lipoprotein-lipase (LPL) in this process. Here, we demonstrate that angiopoietin-like 4 (ANGPTL4), a potent inhibitor of LPL, is abundantly expressed in astrocytes in control white matter tissue and its expression is markedly reduced in active MS lesions. We provide evidence that ANGPTL4 inhibits the uptake of myelin-derived lipids by LPL-immunoreactive phagocytes. Taken together, our data suggest that the strong reduction in astrocytic ANGPTL4 expression in active demyelinating MS lesions enables phagocytes to adequately clear myelin debris, setting the stage for remyelination.
Collapse
Affiliation(s)
- Alwin Kamermans
- Department of Molecular Cell Biology and Immunology, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Merel Rijnsburger
- Department of Molecular Cell Biology and Immunology, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Ananya Chakraborty
- Department of Molecular Cell Biology and Immunology, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Susanne van der Pol
- Department of Molecular Cell Biology and Immunology, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Jack van Horssen
- Department of Molecular Cell Biology and Immunology, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
24
|
Souza DG, Almeida RF, Souza DO, Zimmer ER. The astrocyte biochemistry. Semin Cell Dev Biol 2019; 95:142-150. [PMID: 30951895 DOI: 10.1016/j.semcdb.2019.04.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 03/19/2019] [Accepted: 04/01/2019] [Indexed: 02/06/2023]
Abstract
Astrocytes are a unique and dynamic subtype of glial cells in the central nervous system (CNS). Understanding their biochemical reactions and their influence in the surrounding cells is extremely important in the neuroscience field. They exert important influence in the neurotransmission, ionic homeostasis and also release neuroactive molecules termed gliotransmitters. Additionally, they metabolize, store and release metabolic substrates to meet high brain energy requirements. In this review, we highlight the main biochemical reactions regarding energy metabolism that take place in astrocytes. Special attention is given to synthesis, storage and catabolism of glucose, release of lactate, oxidation of fatty acids, production of ketone bodies, and metabolism of the main neurotransmitters, glutamate and GABA. The recent findings allow proposing these cells as key players controlling the energetic homeostasis in the CNS.
Collapse
Affiliation(s)
- Débora G Souza
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Roberto F Almeida
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Exact and Biological Sciences Institute, Biological Sciences Department, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Diogo O Souza
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Biochemistry, UFRGS, Porto Alegre, Brazil
| | - Eduardo R Zimmer
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Pharmacology, UFRGS, Porto Alegre, Brazil; Graduate Program in Biological Sciences: Pharmacology and Therapeutics, UFRGS, Porto Alegre, Brazil; Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil.
| |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW Lipoprotein lipase (LpL) is well known for its lipolytic action in blood lipoprotein triglyceride catabolism. This article summarizes the recent mechanistic and molecular studies on elucidating the 'unconventional' roles of LpL in mediating biological events related to immune cell response and lipid transport in the pathogenesis of cardiovascular disease (CVD) and tissue degenerative disorders. RECENT FINDINGS Several approaches to inactivate the inhibitors that block LpL enzymatic activity have reestablished the importance of systemic LpL activity in reducing CVD risk. On the other hand, increasing evidence suggests that focal arterial expression of LpL relates to aortic macrophage levels and inflammatory processes. In the hematopoietic origin, LpL also plays a role in modulating hematopoietic stem cell proliferation and circulating blood cell levels and phenotypes. Finally, building upon the strong genetic evidence on the association with assorted brain disorders, a new era in exploring the mechanistic insights into the functions and activity of LpL in brain that impacts central nerve systems has begun. SUMMARY A better understanding of the molecular action of LpL will help to devise novel strategies for intervention of a number of diseases, including blood cell or metabolic disorders, as well to inhibit pathways related to CVD and tissue degenerative processes.
Collapse
Affiliation(s)
- Chuchun L Chang
- Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| |
Collapse
|
26
|
Luquet SH, Vaudry H, Granata R. Editorial: Neuroendocrine Control of Feeding Behavior. Front Endocrinol (Lausanne) 2019; 10:399. [PMID: 31297088 PMCID: PMC6593060 DOI: 10.3389/fendo.2019.00399] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 06/05/2019] [Indexed: 11/13/2022] Open
Affiliation(s)
- Serge H. Luquet
- Paris Diderot University, Paris, France
- *Correspondence: Serge H. Luquet
| | - Hubert Vaudry
- Université de Rouen, Mont-Saint-Aignan, France
- Hubert Vaudry
| | - Riccarda Granata
- Department of Medical Sciences, University of Turin, Turin, Italy
- Riccarda Granata
| |
Collapse
|
27
|
He PP, Jiang T, OuYang XP, Liang YQ, Zou JQ, Wang Y, Shen QQ, Liao L, Zheng XL. Lipoprotein lipase: Biosynthesis, regulatory factors, and its role in atherosclerosis and other diseases. Clin Chim Acta 2018; 480:126-137. [PMID: 29453968 DOI: 10.1016/j.cca.2018.02.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 02/06/2018] [Accepted: 02/07/2018] [Indexed: 01/20/2023]
|
28
|
Zhang J, Li X, Ren Y, Zhao Y, Xing A, Jiang C, Chen Y, An L. Intermittent Fasting Alleviates the Increase of Lipoprotein Lipase Expression in Brain of a Mouse Model of Alzheimer's Disease: Possibly Mediated by β-hydroxybutyrate. Front Cell Neurosci 2018; 12:1. [PMID: 29386999 PMCID: PMC5776118 DOI: 10.3389/fncel.2018.00001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Accepted: 01/01/2018] [Indexed: 12/13/2022] Open
Abstract
Intermittent fasting has been demonstrated to protect against Alzheimer's disease (AD), however, the mechanism is unclear. Histone acetylation and lipoprotein lipase (LPL) are involved in AD progression. Importantly, LPL has been documented to be regulated by histone deacetylases (HDACs) inhibitors (increase histone acetylation level) in adipocyte and mesenchymal stem cells, or by fasting in adipose and muscle tissues. In brain, however, whether histone acetylation or fasting regulates LPL expression is unknown. This study was designed to demonstrate intermittent fasting may protect against AD through increasing β-hydroxybutyrate, a HDACs inhibitor, to regulate LPL. We also investigated microRNA-29a expression associating with regulation of LPL and histone acetylation. The results showed LPL mRNA expression was increased and microRNA-29a expression was decreased in the cerebral cortex of AD model mice (APP/PS1), which were alleviated by intermittent fasting. No significant differences were found in the total expression of LPL protein (brain-derived and located in capillary endothelial cells from peripheral tissues) in the cerebral cortex of APP/PS1 mice. Further study indicated that LPL located in capillary endothelial cells was decreased in the cerebral cortex of APP/PS1 mice, which was alleviated by intermittent fasting. LPL and microRNA-29a expression were separately increased and down-regulated in 2 μM Aβ25−35-exposed SH-SY5Y cells, but respectively decreased and up-regulated in 10 μM Aβ25−35-exposed cells, which were all reversed by β-hydroxybutyrate. The increase of HDAC2/3 expression and the decrease of acetylated H3K9 and H4K12 levels were alleviated in APP/PS1 mice by intermittent fasting treatment, as well in 2 or 10 μM Aβ25−35-exposed cells by β-hydroxybutyrate treatment. These findings above suggested the results from APP/PS1 mice were consistent with those from cells treated with 2 μM Aβ25−35. Interestingly, LPL expression was reduced (0.2-folds) and microRNA-29a expression was up-regulated (1.7-folds) in HDAC2-silenced cells, but respectively increased (1.3-folds) and down-regulated (0.8-folds) in HDAC3-silenced cells. Furthermore, LPL expression was decreased in cells treated with microRNA-29a mimic and increased with inhibitor treatment. In conclusion, intermittent fasting inhibits the increase of brain-derived LPL expression in APP/PS1 mice partly through β-hydroxybutyrate-mediated down-regulation of microRNA-29a expression. HDAC2/3 may be implicated in the effect of β-hydroxybutyrate on microRNA-29a expression.
Collapse
Affiliation(s)
- Jingzhu Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, China Medical University, Shenyang, China
| | - Xinhui Li
- Department of Nutrition and Food Hygiene, School of Public Health, China Medical University, Shenyang, China
| | - Yahao Ren
- Department of Nutrition and Food Hygiene, School of Public Health, China Medical University, Shenyang, China
| | - Yue Zhao
- Department of Nutrition and Food Hygiene, School of Public Health, China Medical University, Shenyang, China
| | - Aiping Xing
- Department of Nutrition and Food Hygiene, School of Public Health, China Medical University, Shenyang, China
| | - Congmin Jiang
- Department of Nutrition and Food Hygiene, School of Public Health, China Medical University, Shenyang, China
| | - Yanqiu Chen
- Department of Nutrition and Food Hygiene, School of Public Health, China Medical University, Shenyang, China
| | - Li An
- Department of Nutrition and Food Hygiene, School of Public Health, China Medical University, Shenyang, China
| |
Collapse
|
29
|
Laperrousaz E, Denis RG, Kassis N, Contreras C, López M, Luquet S, Cruciani-Guglielmacci C, Magnan C. Lipoprotein Lipase Expression in Hypothalamus Is Involved in the Central Regulation of Thermogenesis and the Response to Cold Exposure. Front Endocrinol (Lausanne) 2018; 9:103. [PMID: 29593657 PMCID: PMC5861133 DOI: 10.3389/fendo.2018.00103] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 03/02/2018] [Indexed: 11/15/2022] Open
Abstract
Lipoprotein lipase (LPL) is expressed in different areas of the brain, including the hypothalamus and plays an important role in neural control of the energy balance, including feeding behavior and metabolic fluxes. This study tested the hypothesis that hypothalamic LPL participates in the control of body temperature. We first showed that cold exposure induces decreased activity and expression of LPL in the mouse hypothalamus. We then selectively deleted LPL in the mediobasal hypothalamus (MBH) through an adeno-associated virus approach in LPL-floxed mice and generated MBHΔ Lpl mice with 30-35% decrease in hypothalamic LPL activity. Results showed a decrease in body temperature in MBHΔ Lpl mice when compared with controls at 22°C. Exposure to cold (4°C for 4 h) decreased the body temperature of the control mice while that of the MBHΔ Lpl mice remained similar to that observed at 22°C. MBHΔ Lpl mice also showed increased energy expenditure during cold exposure, when compared to controls. Finally, the selective MBH deletion of LPL also increased the expression of the thermogenic PRMD16 and Dio2 in subcutaneous and perigonadal adipose tissues. Thus, the MBH LPL deletion seems to favor thermogenesis. These data demonstrate that for the first time hypothalamic LPL appears to function as a regulator of body temperature and cold-induced thermogenesis.
Collapse
Affiliation(s)
- Elise Laperrousaz
- Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Sorbonne Paris Cité, Université Denis Diderot, Paris, France
| | - Raphaël G Denis
- Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Sorbonne Paris Cité, Université Denis Diderot, Paris, France
| | - Nadim Kassis
- Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Sorbonne Paris Cité, Université Denis Diderot, Paris, France
| | - Cristina Contreras
- NeurObesity Group, Department of Physiology, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| | - Miguel López
- NeurObesity Group, Department of Physiology, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| | - Serge Luquet
- Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Sorbonne Paris Cité, Université Denis Diderot, Paris, France
| | - Céline Cruciani-Guglielmacci
- Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Sorbonne Paris Cité, Université Denis Diderot, Paris, France
| | - Christophe Magnan
- Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Sorbonne Paris Cité, Université Denis Diderot, Paris, France
| |
Collapse
|
30
|
Frago LM, Canelles S, Freire-Regatillo A, Argente-Arizón P, Barrios V, Argente J, Garcia-Segura LM, Chowen JA. Estradiol Uses Different Mechanisms in Astrocytes from the Hippocampus of Male and Female Rats to Protect against Damage Induced by Palmitic Acid. Front Mol Neurosci 2017; 10:330. [PMID: 29114202 PMCID: PMC5660686 DOI: 10.3389/fnmol.2017.00330] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 09/29/2017] [Indexed: 01/22/2023] Open
Abstract
An excess of saturated fatty acids can be toxic for tissues, including the brain, and this has been associated with the progression of neurodegenerative diseases. Since palmitic acid (PA) is a free fatty acid that is abundant in the diet and circulation and can be harmful, we have investigated the effects of this fatty acid on lipotoxicity in hippocampal astrocytes and the mechanism involved. Moreover, as males and females have different susceptibilities to some neurodegenerative diseases, we accessed the responses of astrocytes from both sexes, as well as the possible involvement of estrogens in the protection against fatty acid toxicity. PA increased endoplasmic reticulum stress leading to cell death in astrocytes from both males and females. Estradiol (E2) increased the levels of protective factors, such as Hsp70 and the anti-inflammatory cytokine interleukin-10, in astrocytes from both sexes. In male astrocytes, E2 decreased pJNK, TNFα, and caspase-3 activation. In contrast, in female astrocytes E2 did not affect the activation of JNK or TNFα levels, but decreased apoptotic cell death. Hence, although E2 exerted protective effects against the detrimental effects of PA, the mechanisms involved appear to be different between male and female astrocytes. This sexually dimorphic difference in the protective mechanisms induced by E2 could be involved in the different susceptibilities of males and females to some neurodegenerative processes.
Collapse
Affiliation(s)
- Laura M Frago
- Departamento de Pediatría, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Endocrinología, Hospital Infantil Universitario Niño Jesús, Madrid, Spain.,Instituto de Investigación Sanitaria Princesa, Madrid, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Sandra Canelles
- Departamento de Endocrinología, Hospital Infantil Universitario Niño Jesús, Madrid, Spain.,Instituto de Investigación Sanitaria Princesa, Madrid, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Alejandra Freire-Regatillo
- Departamento de Pediatría, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Endocrinología, Hospital Infantil Universitario Niño Jesús, Madrid, Spain.,Instituto de Investigación Sanitaria Princesa, Madrid, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Pilar Argente-Arizón
- Departamento de Pediatría, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Endocrinología, Hospital Infantil Universitario Niño Jesús, Madrid, Spain.,Instituto de Investigación Sanitaria Princesa, Madrid, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Vicente Barrios
- Departamento de Endocrinología, Hospital Infantil Universitario Niño Jesús, Madrid, Spain.,Instituto de Investigación Sanitaria Princesa, Madrid, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Argente
- Departamento de Pediatría, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Endocrinología, Hospital Infantil Universitario Niño Jesús, Madrid, Spain.,Instituto de Investigación Sanitaria Princesa, Madrid, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain.,IMDEA Food Institute, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Luis M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,CIBER de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Julie A Chowen
- Departamento de Endocrinología, Hospital Infantil Universitario Niño Jesús, Madrid, Spain.,Instituto de Investigación Sanitaria Princesa, Madrid, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
31
|
Cruciani-Guglielmacci C, López M, Campana M, le Stunff H. Brain Ceramide Metabolism in the Control of Energy Balance. Front Physiol 2017; 8:787. [PMID: 29075199 PMCID: PMC5643460 DOI: 10.3389/fphys.2017.00787] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 09/26/2017] [Indexed: 01/11/2023] Open
Abstract
The regulation of energy balance by the central nervous system (CNS) is a key actor of energy homeostasis in mammals, and deregulations of the fine mechanisms of nutrient sensing in the brain could lead to several metabolic diseases such as obesity and type 2 diabetes (T2D). Indeed, while neuronal activity primarily relies on glucose (lactate, pyruvate), the brain expresses at high level enzymes responsible for the transport, utilization and storage of lipids. It has been demonstrated that discrete neuronal networks in the hypothalamus have the ability to detect variation of circulating long chain fatty acids (FA) to regulate food intake and peripheral glucose metabolism. During a chronic lipid excess situation, this physiological lipid sensing is impaired contributing to type 2 diabetes in predisposed subjects. Recently, different studies suggested that ceramides levels could be involved in the regulation of energy balance in both hypothalamic and extra-hypothalamic areas. Moreover, under lipotoxic conditions, these ceramides could play a role in the dysregulation of glucose homeostasis. In this review we aimed at describing the potential role of ceramides metabolism in the brain in the physiological and pathophysiological control of energy balance.
Collapse
Affiliation(s)
- Céline Cruciani-Guglielmacci
- Unité de Biologie Fonctionnelle et Adaptative, Centre National de la Recherche Scientifique, Unité Mixte de Recherche, Université Paris Diderot, Université Sorbonne Paris Cité, Paris, France
| | - Miguel López
- Department of Physiology, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Instituto de Investigación Sanitaria de Santiago de Compostela, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Mélanie Campana
- Unité de Biologie Fonctionnelle et Adaptative, Centre National de la Recherche Scientifique, Unité Mixte de Recherche, Université Paris Diderot, Université Sorbonne Paris Cité, Paris, France
| | - Hervé le Stunff
- Unité de Biologie Fonctionnelle et Adaptative, Centre National de la Recherche Scientifique, Unité Mixte de Recherche, Université Paris Diderot, Université Sorbonne Paris Cité, Paris, France.,UMR9197 Institut des Neurosciences Paris Saclay (Neuro-PSI), Université Paris-Saclay, Saclay, France
| |
Collapse
|
32
|
Wu W, Yin Y, Zhong J, Peng Y, Li S, Zheng L, Cao H, Zhang J. Cell therapy could be a potential way to improve lipoprotein lipase deficiency. Lipids Health Dis 2017; 16:189. [PMID: 28969646 PMCID: PMC5625700 DOI: 10.1186/s12944-017-0577-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 09/22/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Lipoprotein lipase (LPL) deficiency is an autosomal recessive genetic disorder characterized by extreme hypertriglyceridemia, with no cure presently available. The purpose of this study was to test the possibility of using cell therapy to alleviate LPL deficiency. METHODS The LPL coding sequence was cloned into the MSCV retrovirus vector, after which MSCV-hLPL and MSCV (empty construct without LPL coding sequence) virion suspensions were made using the calcium chloride method. A muscle cell line (C2C12), kidney cell line (HEK293T) and pre-adipocyte cell line (3 T3-L1) were transfected with the virus in order to express recombinant LPL in vitro. Finally, each transfected cell line was injected subcutaneously into nude mice to identify the cell type which could secret recombinant LPL in vivo. Control cells were transfected with the MSCV empty vector. LPL activity was analyzed using a radioimmunoassay. RESULTS After virus infection, the LPL activity at the cell surface of each cell type was significantly higher than in the control cells, which indicates that all three cell types can be used to generate functional LPL. The transfected cells were injected subcutaneously into nude mice, and the LPL activity of the nearby muscle tissue at the injection site in mice injected with 3 T3-L1 cells was more than 5 times higher at the injection sites than at non-injected control sites. The other two types of cells did not show this trend. CONCLUSION The subcutaneous injection of adipocytes overexpressing LPL can improve the LPL activity of the adjacent tissue of nude mice. This is a ground-breaking preliminary study for the treatment of LPL deficiency, and lays a good foundation for using cell therapy to correct LPL deficiency.
Collapse
Affiliation(s)
- Wenjing Wu
- College of Biological and Chemical Science and Engineering, Jiaxing University, Lianglin Campus,118 Jiahang Road, Jiaxing, 314001, China
| | - Yajun Yin
- College of Biological and Chemical Science and Engineering, Jiaxing University, Lianglin Campus,118 Jiahang Road, Jiaxing, 314001, China
| | - Jie Zhong
- College of life science and biotechnology, Hebei Normal University of Science and Technology, Qinhuangdao, 066004, China
| | - Yongjia Peng
- College of Biological and Chemical Science and Engineering, Jiaxing University, Lianglin Campus,118 Jiahang Road, Jiaxing, 314001, China
| | - Shuncai Li
- College of life science and biotechnology, Hebei Normal University of Science and Technology, Qinhuangdao, 066004, China
| | - Libin Zheng
- College of Biological and Chemical Science and Engineering, Jiaxing University, Lianglin Campus,118 Jiahang Road, Jiaxing, 314001, China
| | - Hong Cao
- College of Biological and Chemical Science and Engineering, Jiaxing University, Lianglin Campus,118 Jiahang Road, Jiaxing, 314001, China
| | - Jin Zhang
- College of Biological and Chemical Science and Engineering, Jiaxing University, Lianglin Campus,118 Jiahang Road, Jiaxing, 314001, China. .,College of life science and biotechnology, Hebei Normal University of Science and Technology, Qinhuangdao, 066004, China.
| |
Collapse
|
33
|
Gao Y, Layritz C, Legutko B, Eichmann TO, Laperrousaz E, Moullé VS, Cruciani-Guglielmacci C, Magnan C, Luquet S, Woods SC, Eckel RH, Yi CX, Garcia-Caceres C, Tschöp MH. Disruption of Lipid Uptake in Astroglia Exacerbates Diet-Induced Obesity. Diabetes 2017; 66:2555-2563. [PMID: 28710138 PMCID: PMC6463752 DOI: 10.2337/db16-1278] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 07/04/2017] [Indexed: 02/06/2023]
Abstract
Neuronal circuits in the brain help to control feeding behavior and systemic metabolism in response to afferent nutrient and hormonal signals. Although astrocytes have historically been assumed to have little relevance for such neuroendocrine control, we investigated whether lipid uptake via lipoprotein lipase (LPL) in astrocytes is required to centrally regulate energy homeostasis. Ex vivo studies with hypothalamus-derived astrocytes showed that LPL expression is upregulated by oleic acid, whereas it is decreased in response to palmitic acid or triglycerides. Likewise, astrocytic LPL deletion reduced the accumulation of lipid droplets in those glial cells. Consecutive in vivo studies showed that postnatal ablation of LPL in glial fibrillary acidic protein-expressing astrocytes induced exaggerated body weight gain and glucose intolerance in mice exposed to a high-fat diet. Intriguingly, astrocytic LPL deficiency also triggered increased ceramide content in the hypothalamus, which may contribute to hypothalamic insulin resistance. We conclude that hypothalamic LPL functions in astrocytes to ensure appropriately balanced nutrient sensing, ceramide distribution, body weight regulation, and glucose metabolism.
Collapse
Affiliation(s)
- Yuanqing Gao
- Helmholtz Diabetes Center (HDC) and German Center for Diabetes Research (DZD), Helmholtz Zentrum München and Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Clarita Layritz
- Helmholtz Diabetes Center (HDC) and German Center for Diabetes Research (DZD), Helmholtz Zentrum München and Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany
| | - Beata Legutko
- Helmholtz Diabetes Center (HDC) and German Center for Diabetes Research (DZD), Helmholtz Zentrum München and Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany
| | - Thomas O Eichmann
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Elise Laperrousaz
- Unité de Biologie Fonctionnelle et Adaptative, Sorbonne Paris Cité, CNRS UMR 8251, University of Paris Diderot, Paris, France
| | - Valentine S Moullé
- Unité de Biologie Fonctionnelle et Adaptative, Sorbonne Paris Cité, CNRS UMR 8251, University of Paris Diderot, Paris, France
| | - Celine Cruciani-Guglielmacci
- Unité de Biologie Fonctionnelle et Adaptative, Sorbonne Paris Cité, CNRS UMR 8251, University of Paris Diderot, Paris, France
| | - Christophe Magnan
- Unité de Biologie Fonctionnelle et Adaptative, Sorbonne Paris Cité, CNRS UMR 8251, University of Paris Diderot, Paris, France
| | - Serge Luquet
- Unité de Biologie Fonctionnelle et Adaptative, Sorbonne Paris Cité, CNRS UMR 8251, University of Paris Diderot, Paris, France
| | - Stephen C Woods
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH
| | - Robert H Eckel
- Division of Endocrinology, Metabolism, & Diabetes, University of Colorado at Denver, Denver, CO
| | - Chun-Xia Yi
- Helmholtz Diabetes Center (HDC) and German Center for Diabetes Research (DZD), Helmholtz Zentrum München and Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Cristina Garcia-Caceres
- Helmholtz Diabetes Center (HDC) and German Center for Diabetes Research (DZD), Helmholtz Zentrum München and Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany
| | - Matthias H Tschöp
- Helmholtz Diabetes Center (HDC) and German Center for Diabetes Research (DZD), Helmholtz Zentrum München and Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany
| |
Collapse
|
34
|
Periyasamy S, Sathya M, Karthick C, Kandasamy M, Shanmugaapriya S, Tamilselvan J, Jayachandran KS, Anusuyadevi M. Association Studies of Specific Cholesterol Related Genes (APOE, LPL, and CETP) with Lipid Profile and Memory Function: A Correlative Study Among Rural and Tribal Population of Dharmapuri District, India. J Alzheimers Dis 2017; 60:S195-S207. [PMID: 28777751 DOI: 10.3233/jad-170272] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Sabapathy Periyasamy
- Department of Biochemistry, Molecular Gerontology Laboratory, Bharathidasan University, Tiruchirappalli, India
| | - Mohan Sathya
- Department of Biochemistry, Molecular Gerontology Laboratory, Bharathidasan University, Tiruchirappalli, India
| | - Chennakesavan Karthick
- Department of Biochemistry, Molecular Gerontology Laboratory, Bharathidasan University, Tiruchirappalli, India
| | - Mahesh Kandasamy
- UGC-Faculty Recharge Program (UGC-FRP), University Grant Commission, Laboratory of Stem cells and Neuroregeneration, Department of Animal Science, Bharathidasan University, Tiruchirappalli, Tamilnadu, India
| | | | | | | | - Muthuswamy Anusuyadevi
- Department of Biochemistry, Molecular Gerontology Laboratory, Bharathidasan University, Tiruchirappalli, India
| |
Collapse
|
35
|
Laperrousaz E, Moullé VS, Denis RG, Kassis N, Berland C, Colsch B, Fioramonti X, Philippe E, Lacombe A, Vanacker C, Butin N, Bruce KD, Wang H, Wang Y, Gao Y, Garcia-Caceres C, Prévot V, Tschöp MH, Eckel RH, Le Stunff H, Luquet S, Magnan C, Cruciani-Guglielmacci C. Lipoprotein lipase in hypothalamus is a key regulator of body weight gain and glucose homeostasis in mice. Diabetologia 2017; 60:1314-1324. [PMID: 28456865 DOI: 10.1007/s00125-017-4282-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 03/16/2017] [Indexed: 01/04/2023]
Abstract
AIMS/HYPOTHESIS Regulation of energy balance involves the participation of many factors, including nutrients, among which are circulating lipids, acting as peripheral signals informing the central nervous system of the energy status of the organism. It has been shown that neuronal lipoprotein lipase (LPL) participates in the control of energy balance by hydrolysing lipid particles enriched in triacylglycerols. Here, we tested the hypothesis that LPL in the mediobasal hypothalamus (MBH), a well-known nucleus implicated in the regulation of metabolic homeostasis, could also contribute to the regulation of body weight and glucose homeostasis. METHODS We injected an adeno-associated virus (AAV) expressing Cre-green fluorescent protein into the MBH of Lpl-floxed mice (and wild-type mice) to specifically decrease LPL activity in the MBH. In parallel, we injected an AAV overexpressing Lpl into the MBH of wild-type mice. We then studied energy homeostasis and hypothalamic ceramide content. RESULTS The partial deletion of Lpl in the MBH in mice led to an increase in body weight compared with controls (37.72 ± 0.7 g vs 28.46 ± 0.12, p < 0.001) associated with a decrease in locomotor activity. These mice developed hyperinsulinaemia and glucose intolerance. This phenotype also displayed reduced expression of Cers1 in the hypothalamus as well as decreased concentration of several C18 species of ceramides and a 3-fold decrease in total ceramide intensity. Conversely, overexpression of Lpl specifically in the MBH induced a decrease in body weight. CONCLUSIONS/INTERPRETATION Our study shows that LPL in the MBH is an important regulator of body weight and glucose homeostasis.
Collapse
Affiliation(s)
- Elise Laperrousaz
- Unité de Biologie Fonctionnelle et Adaptative, Sorbonne Paris Cité, CNRS UMR 8251, Université Paris Diderot, Bâtiment Buffon, P. O. box 7126, 4, rue Marie-Andrée Lagroua Weill-Halle, 75205, Paris Cedex 13, France
| | - Valentine S Moullé
- Unité de Biologie Fonctionnelle et Adaptative, Sorbonne Paris Cité, CNRS UMR 8251, Université Paris Diderot, Bâtiment Buffon, P. O. box 7126, 4, rue Marie-Andrée Lagroua Weill-Halle, 75205, Paris Cedex 13, France
| | - Raphaël G Denis
- Unité de Biologie Fonctionnelle et Adaptative, Sorbonne Paris Cité, CNRS UMR 8251, Université Paris Diderot, Bâtiment Buffon, P. O. box 7126, 4, rue Marie-Andrée Lagroua Weill-Halle, 75205, Paris Cedex 13, France
| | - Nadim Kassis
- Unité de Biologie Fonctionnelle et Adaptative, Sorbonne Paris Cité, CNRS UMR 8251, Université Paris Diderot, Bâtiment Buffon, P. O. box 7126, 4, rue Marie-Andrée Lagroua Weill-Halle, 75205, Paris Cedex 13, France
| | - Chloé Berland
- Unité de Biologie Fonctionnelle et Adaptative, Sorbonne Paris Cité, CNRS UMR 8251, Université Paris Diderot, Bâtiment Buffon, P. O. box 7126, 4, rue Marie-Andrée Lagroua Weill-Halle, 75205, Paris Cedex 13, France
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum, Munich, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Benoit Colsch
- CEA-Centre d'Etude de Saclay, Laboratoire d'étude du Métabolisme des Médicaments, Gif-sur-Yvette, France
| | - Xavier Fioramonti
- Centre des Sciences du Goût et de l'Alimentation, Unité Mixte de Recherche CNRS, INRA, Université de Bourgogne, Dijon, France
| | - Erwann Philippe
- Unité de Biologie Fonctionnelle et Adaptative, Sorbonne Paris Cité, CNRS UMR 8251, Université Paris Diderot, Bâtiment Buffon, P. O. box 7126, 4, rue Marie-Andrée Lagroua Weill-Halle, 75205, Paris Cedex 13, France
| | - Amélie Lacombe
- Unité de Biologie Fonctionnelle et Adaptative, Sorbonne Paris Cité, CNRS UMR 8251, Université Paris Diderot, Bâtiment Buffon, P. O. box 7126, 4, rue Marie-Andrée Lagroua Weill-Halle, 75205, Paris Cedex 13, France
| | - Charlotte Vanacker
- Development and Plasticity of the Neuroendocrine Brain, Neurobese International Associated Laboratory, Jean-Pierre Aubert Research Center, Inserm U1172, University of Lille, Lille, France
| | - Noémie Butin
- CEA-Centre d'Etude de Saclay, Laboratoire d'étude du Métabolisme des Médicaments, Gif-sur-Yvette, France
| | - Kimberley D Bruce
- Division of Endocrinology, Metabolism, & Diabetes, Department of Medicine, University of Colorado, Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Hong Wang
- Division of Endocrinology, Metabolism, & Diabetes, Department of Medicine, University of Colorado, Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Yongping Wang
- Division of Endocrinology, Metabolism, & Diabetes, Department of Medicine, University of Colorado, Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Yuanqing Gao
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum, Munich, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Cristina Garcia-Caceres
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum, Munich, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Vincent Prévot
- Centre des Sciences du Goût et de l'Alimentation, Unité Mixte de Recherche CNRS, INRA, Université de Bourgogne, Dijon, France
| | - Matthias H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum, Munich, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Division of Metabolic Diseases, Technische Universität München, Munich, Germany
| | - Robert H Eckel
- Division of Endocrinology, Metabolism, & Diabetes, Department of Medicine, University of Colorado, Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Hervé Le Stunff
- Unité de Biologie Fonctionnelle et Adaptative, Sorbonne Paris Cité, CNRS UMR 8251, Université Paris Diderot, Bâtiment Buffon, P. O. box 7126, 4, rue Marie-Andrée Lagroua Weill-Halle, 75205, Paris Cedex 13, France
| | - Serge Luquet
- Unité de Biologie Fonctionnelle et Adaptative, Sorbonne Paris Cité, CNRS UMR 8251, Université Paris Diderot, Bâtiment Buffon, P. O. box 7126, 4, rue Marie-Andrée Lagroua Weill-Halle, 75205, Paris Cedex 13, France
| | - Christophe Magnan
- Unité de Biologie Fonctionnelle et Adaptative, Sorbonne Paris Cité, CNRS UMR 8251, Université Paris Diderot, Bâtiment Buffon, P. O. box 7126, 4, rue Marie-Andrée Lagroua Weill-Halle, 75205, Paris Cedex 13, France.
| | - Céline Cruciani-Guglielmacci
- Unité de Biologie Fonctionnelle et Adaptative, Sorbonne Paris Cité, CNRS UMR 8251, Université Paris Diderot, Bâtiment Buffon, P. O. box 7126, 4, rue Marie-Andrée Lagroua Weill-Halle, 75205, Paris Cedex 13, France.
| |
Collapse
|
36
|
Freire-Regatillo A, Argente-Arizón P, Argente J, García-Segura LM, Chowen JA. Non-Neuronal Cells in the Hypothalamic Adaptation to Metabolic Signals. Front Endocrinol (Lausanne) 2017; 8:51. [PMID: 28377744 PMCID: PMC5359311 DOI: 10.3389/fendo.2017.00051] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 03/03/2017] [Indexed: 12/19/2022] Open
Abstract
Although the brain is composed of numerous cell types, neurons have received the vast majority of attention in the attempt to understand how this organ functions. Neurons are indeed fundamental but, in order for them to function correctly, they rely on the surrounding "non-neuronal" cells. These different cell types, which include glia, epithelial cells, pericytes, and endothelia, supply essential substances to neurons, in addition to protecting them from dangerous substances and situations. Moreover, it is now clear that non-neuronal cells can also actively participate in determining neuronal signaling outcomes. Due to the increasing problem of obesity in industrialized countries, investigation of the central control of energy balance has greatly increased in attempts to identify new therapeutic targets. This has led to interesting advances in our understanding of how appetite and systemic metabolism are modulated by non-neuronal cells. For example, not only are nutrients and hormones transported into the brain by non-neuronal cells, but these cells can also metabolize these metabolic factors, thus modifying the signals reaching the neurons. The hypothalamus is the main integrating center of incoming metabolic and hormonal signals and interprets this information in order to control appetite and systemic metabolism. Hence, the factors transported and released from surrounding non-neuronal cells will undoubtedly influence metabolic homeostasis. This review focuses on what is known to date regarding the involvement of different cell types in the transport and metabolism of nutrients and hormones in the hypothalamus. The possible involvement of non-neuronal cells, in particular glial cells, in physiopathological outcomes of poor dietary habits and excess weight gain are also discussed.
Collapse
Affiliation(s)
- Alejandra Freire-Regatillo
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, Madrid, Spain
- Department of Pediatrics, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red: Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Madrid, Spain
| | - Pilar Argente-Arizón
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, Madrid, Spain
- Department of Pediatrics, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red: Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Madrid, Spain
| | - Jesús Argente
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, Madrid, Spain
- Department of Pediatrics, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red: Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Madrid, Spain
- IMDEA Food Institute, Campus of International Excellence (CEI) UAM + CSIC, Madrid, Spain
| | - Luis Miguel García-Segura
- Laboratory of Neuroactive Steroids, Department of Functional and Systems Neurobiology, Instituto Cajal, CSIC (Consejo Superior de Investigaciones Científicas), Madrid, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Julie A. Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red: Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Madrid, Spain
| |
Collapse
|
37
|
Niot I, Besnard P. Appetite control by the tongue-gut axis and evaluation of the role of CD36/SR-B2. Biochimie 2017; 136:27-32. [PMID: 28238842 DOI: 10.1016/j.biochi.2017.02.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 02/22/2017] [Indexed: 12/31/2022]
Abstract
Understanding the mechanisms governing food intake is a public health issue given the dramatic rise of obesity over the world. The overconsumption of tasty energy-dense foods rich in lipids is considered to be one of the nutritional causes of this epidemic. Over the last decade, the identification of fatty acid receptors in strategic places in the body (i.e. oro-intestinal tract and brain) has provided a major progress in the deciphering of regulatory networks involved in the control of dietary intake. Among these lipid sensors, CD36/SR-B2 appears to play a significant role since this membrane protein, known to bind long-chain fatty acid with a high affinity, was specifically found both in enterocytes and in a subset of taste bud cells and entero-endocrine cells. After a short overview on CD36/SR-B2 structure, function and regulation, this mini-review proposes to analyze the key findings about the role of CD36/SR-B2 along of the tongue-gut axis in relation to appetite control. In addition, we discuss whether obesogenic diets might impair lipid sensing mediated by CD36/SR-B2 along this axis.
Collapse
Affiliation(s)
- Isabelle Niot
- Physiologie de la Nutrition et Toxicologie (NUTox), LCN UMR 1231, INSERM/AgroSupDijon/Univ. Bourgogne Franche-Comté, F-21000, Dijon, France.
| | - Philippe Besnard
- Physiologie de la Nutrition et Toxicologie (NUTox), LCN UMR 1231, INSERM/AgroSupDijon/Univ. Bourgogne Franche-Comté, F-21000, Dijon, France.
| |
Collapse
|
38
|
Lee LL, Aung HH, Wilson DW, Anderson SE, Rutledge JC, Rutkowsky JM. Triglyceride-rich lipoprotein lipolysis products increase blood-brain barrier transfer coefficient and induce astrocyte lipid droplets and cell stress. Am J Physiol Cell Physiol 2017; 312:C500-C516. [PMID: 28077357 DOI: 10.1152/ajpcell.00120.2016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 01/06/2017] [Accepted: 01/09/2017] [Indexed: 12/19/2022]
Abstract
Elevation of blood triglycerides, primarily as triglyceride-rich lipoproteins (TGRL), has been linked to cerebrovascular inflammation, vascular dementia, and Alzheimer's disease (AD). Brain microvascular endothelial cells and astrocytes, two cell components of the neurovascular unit, participate in controlling blood-brain barrier (BBB) permeability and regulating neurovascular unit homeostasis. Our studies showed that infusion of high physiological concentrations of TGRL lipolysis products (TGRL + lipoprotein lipase) activate and injure brain endothelial cells and transiently increase the BBB transfer coefficient (Ki = permeability × surface area/volume) in vivo. However, little is known about how blood lipids affect astrocyte lipid accumulation and inflammation. To address this, we first demonstrated TGRL lipolysis products increased lipid droplet formation in cultured normal human astrocytes. We then evaluated the transcriptional pathways activated in astrocytes by TGRL lipolysis products and found upregulated stress and inflammatory-related genes including activating transcription factor 3 (ATF3), macrophage inflammatory protein-3α (MIP-3α), growth differentiation factor-15 (GDF15), and prostaglandin-endoperoxide synthase 2 (COX2). TGRL lipolysis products also activated the JNK/cJUN/ATF3 pathway, induced endoplasmic reticulum stress protein C/EBP homologous protein (CHOP), and the NF-κB pathway, while increasing secretion of MIP-3α, GDF15, and IL-8. Thus our results demonstrate TGRL lipolysis products increase the BBB transfer coefficient (Ki), induce astrocyte lipid droplet formation, activate cell stress pathways, and induce secretion of inflammatory cytokines. Our observations are consistent with evidence for lipid-induced neurovascular injury and inflammation, and we, therefore, speculate that lipid-induced astrocyte injury could play a role in cognitive decline.
Collapse
Affiliation(s)
- Linda L Lee
- Department of Internal Medicine, University of California, Davis, California
| | - Hnin H Aung
- Department of Internal Medicine, University of California, Davis, California
| | - Dennis W Wilson
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, California; and
| | - Steven E Anderson
- Department of Physiology and Membrane Biology, University of California, Davis, California
| | - John C Rutledge
- Department of Internal Medicine, University of California, Davis, California
| | | |
Collapse
|
39
|
Dong W, Gong H, Zhang G, Vuletic S, Albers J, Zhang J, Liang H, Sui Y, Zheng J. Lipoprotein lipase and phospholipid transfer protein overexpression in human glioma cells and their effect on cell growth, apoptosis, and migration. Acta Biochim Biophys Sin (Shanghai) 2017; 49:62-73. [PMID: 27864281 DOI: 10.1093/abbs/gmw117] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/26/2016] [Indexed: 01/01/2023] Open
Abstract
Glioma is one of the common tumors in brain. The expression level of lipoprotein lipase (LPL) or phospholipid transfer protein (PLTP) may influence glioma progression and its relationship with clinical and pathological parameters. The clinical significance of LPL or PLTP expression in glioma has not been established. In the present study, the LPL and PLTP levels in glioma tumors were investigated and the relationship between the LPL and PLTP level and the grade of malignant glioma was analyzed, with the aim to provide new ideas for the diagnosis and treatment of gliomas in clinical and basic research settings. LPL and PLTP mRNA and protein levels were significantly higher in Grade IV glioma than those in the lower grade tumors (P < 0.01). Double immunofluorescent staining showed that the levels of LPL and PLTP were significantly associated with the pathological grade of glioma (P = 0.005). The levels of LPL and PLTP were increased with the shortened survival of glioma patients (P < 0.001). Knockdown of LPL and PLTP led to decreased cell growth and migration but increased apoptosis in vitro Additionally, cell cycle-related cyclins and their partners were found to be down-regulated while cyclin-dependent kinase inhibitors p16, p21, and Rb were up-regulated. Furthermore, knockdown of LPL or PLTP resulted in the up-regulation of pro-apoptotic molecules and the down-regulation of anti-apoptotic molecules. Ablation of LPL or PLTP in U251 cells resulted in the down-regulation of epithelial mesenchymal transition markers and invasion molecules matrix metalloproteinases. LPL and PLTP appear to be novel glioma-associated proteins and play a role in the progression of human glioma.
Collapse
Affiliation(s)
- Weijiang Dong
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Huilin Gong
- Department of Pathology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Guanjun Zhang
- Department of Pathology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Simona Vuletic
- Northwest Lipid Metabolism and Diabetes Research Laboratories, Department of Medicine, School of Medicine, University of Washington, Seattle, 98109 WA
| | - John Albers
- Northwest Lipid Metabolism and Diabetes Research Laboratories, Department of Medicine, School of Medicine, University of Washington, Seattle, 98109 WA
| | - Jiaojiao Zhang
- Department of Pathology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Hua Liang
- Department of Pathology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Yanxia Sui
- Department of Pathology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Jin Zheng
- Hospital of Nephrology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
40
|
Chen H, Jia J, Ni Z, Vastermark A, Wu B, Le Y, Jawad U. Orlistat response to missense mutations in lipoprotein lipase. Biotechnol Appl Biochem 2016; 64:464-470. [DOI: 10.1002/bab.1500] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 04/14/2016] [Indexed: 11/09/2022]
Affiliation(s)
- Huayou Chen
- Institute of Life Sciences; Jiangsu University; Zhenjiang People's Republic of China
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering; Chinese Academy of Sciences; Beijing People's Republic of China
- Division of Biological Sciences; University of California at San Diego; La Jolla CA USA
| | - Jinru Jia
- Institute of Life Sciences; Jiangsu University; Zhenjiang People's Republic of China
| | - Zhong Ni
- Institute of Life Sciences; Jiangsu University; Zhenjiang People's Republic of China
| | - Ake Vastermark
- Division of Biological Sciences; University of California at San Diego; La Jolla CA USA
| | - Bangguo Wu
- Institute of Life Sciences; Jiangsu University; Zhenjiang People's Republic of China
| | - Yilin Le
- Institute of Life Sciences; Jiangsu University; Zhenjiang People's Republic of China
| | - Ullah Jawad
- Institute of Life Sciences; Jiangsu University; Zhenjiang People's Republic of China
| |
Collapse
|
41
|
Genetic Restoration of Plasma ApoE Improves Cognition and Partially Restores Synaptic Defects in ApoE-Deficient Mice. J Neurosci 2016; 36:10141-50. [PMID: 27683909 DOI: 10.1523/jneurosci.1054-16.2016] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 08/16/2016] [Indexed: 12/23/2022] Open
Abstract
UNLABELLED Alzheimer's disease (AD) is the most common form of dementia in individuals over the age of 65 years. The most prevalent genetic risk factor for AD is the ε4 allele of apolipoprotein E (ApoE4), and novel AD treatments that target ApoE are being considered. One unresolved question in ApoE biology is whether ApoE is necessary for healthy brain function. ApoE knock-out (KO) mice have synaptic loss and cognitive dysfunction; however, these findings are complicated by the fact that ApoE knock-out mice have highly elevated plasma lipid levels, which may independently affect brain function. To bypass the effect of ApoE loss on plasma lipids, we generated a novel mouse model that expresses ApoE normally in peripheral tissues, but has severely reduced ApoE in the brain, allowing us to study brain ApoE loss in the context of a normal plasma lipid profile. We found that these brain ApoE knock-out (bEKO) mice had synaptic loss and dysfunction similar to that of ApoE KO mice; however, the bEKO mice did not have the learning and memory impairment observed in ApoE KO mice. Moreover, we found that the memory deficit in the ApoE KO mice was specific to female mice and was fully rescued in female bEKO mice. Furthermore, while the AMPA/NMDA ratio was reduced in ApoE KO mice, it was unchanged in bEKO mice compared with controls. These findings suggest that plasma lipid levels can influence cognition and synaptic function independent of ApoE expression in the brain. SIGNIFICANCE STATEMENT One proposed treatment strategy for Alzheimer's disease (AD) is the reduction of ApoE, whose ε4 isoform is the most common genetic risk factor for the disease. A major concern of this strategy is that an animal model of ApoE deficiency, the ApoE knock-out (KO) mouse, has reduced synapses and cognitive impairment; however, these mice also develop dyslipidemia and severe atherosclerosis. Here, we have shown that genetic restoration of plasma ApoE to wild-type levels normalizes plasma lipids in ApoE KO mice. While this does not rescue synaptic loss, it does completely restore learning and memory in the mice, suggesting that both CNS and plasma ApoE are independent parameters that affect brain health.
Collapse
|
42
|
Hodges JK, Tan L, Green MH, Ross AC. Vitamin A Supplementation Increases the Uptake of Chylomicron Retinyl Esters into the Brain of Neonatal Rats Raised under Vitamin A-Marginal Conditions. J Nutr 2016; 146:1677-83. [PMID: 27511933 PMCID: PMC4997285 DOI: 10.3945/jn.116.233692] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 06/22/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The most rapid phase of brain development occurs during the neonatal period. Vitamin A (VA; retinol) is critical for many aspects of this process, including neurogenesis, synaptic plasticity, learning, and memory formation. However, the metabolism of retinol in the neonatal brain has not been extensively explored. OBJECTIVE We examined the uptake of VA into the brain in neonatal rats raised under VA-marginal conditions (control group) and assessed the effect of VA supplementation on the uptake of VA into the brain. METHODS Sprague-Dawley neonatal rats (n = 104) nursed by mothers fed a VA-marginal diet were randomly assigned and treated on postnatal day 4 with an oral dose of either VA (6 μg retinyl palmitate/g body weight) or canola oil as the control, both of which contained 1.8 μCi [(3)H]retinol. Pups (n = 4/group at a time) were killed at 13 sampling times from 30 min to 24 d after dosing. The uptake of total retinol, chylomicron-associated retinyl esters (REs), and retinol bound to retinol-binding protein (RBP) was estimated with the use of WinSAAM version 3.0.8. RESULTS Total retinol mass in the brain was closely dependent on its mass in plasma over time (r = 0.91; P < 0.001). The uptake of retinol into the brain involved both postprandial chylomicrons and RBP, with RBP delivering most of the retinol in the control group [0.27 nmol/d (RBP) compared with 0.01 nmol/d (chylomicrons)]. VA supplementation increased the fractional uptake of chylomicron REs from 0.3% to 1.2% of plasma pool/d, decreased that of RBP retinol from 0.5% to 0.2% of plasma pool/d, and increased the transfer rate of chylomicron REs from nearly zero to 0.7 nmol/d, causing a day-long elevation in the brain mass of total retinol. CONCLUSION Postprandial chylomicrons may be a primary mechanism for delivering a recently ingested large dose of VA to the brain of neonatal rats raised under VA-marginal conditions.
Collapse
Affiliation(s)
- Joanna K Hodges
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA
| | - Libo Tan
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA
| | - Michael H Green
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA
| | - A Catharine Ross
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA
| |
Collapse
|
43
|
Wang H, Taussig MD, DiPatrizio NV, Bruce K, Piomelli D, Eckel RH. Obesity development in neuron-specific lipoprotein lipase deficient mice is not responsive to increased dietary fat content or change in fat composition. Metabolism 2016; 65:987-97. [PMID: 27282869 DOI: 10.1016/j.metabol.2016.01.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 01/25/2016] [Accepted: 01/29/2016] [Indexed: 11/16/2022]
Abstract
We have previously reported that mice with neuron-specific LPL deficiency (NEXLPL-/-) become obese by 16weeks of age on chow. Moreover, these mice had reduced uptake of triglyceride (TG)-rich lipoprotein-derived fatty acids and lower levels of n-3 long chain polyunsaturated fatty acids (n-3 PUFAs) in the hypothalamus. Here, we asked whether increased dietary fat content or altered dietary composition could modulate obesity development in NEXLPL-/- mice. Male NEXLPL-/- mice and littermate controls (WT) were randomly assigned one of three synthetic diets; a high carbohydrate diet (HC, 10% fat), a high-fat diet (HF, 45% fat), or a HC diet supplemented with n-3 PUFAs (HCn-3, 10% fat, Lovaza, GSK®). After 42weeks of HC feeding, body weight and fat mass were increased in the NEXLPL-/- mice compared to WT. WT mice fed a HF diet displayed typical diet-induced obesity, but weight gain was only marginal in HF-fed NEXLPL-/- mice, with no significant difference in body composition. Dietary n-3 PUFA supplementation did not prevent obesity in NEXLPL-/- mice, but was associated with differential modifications in hypothalamic gene expression and PUFA concentration compared to WT mice. Our findings suggest that neuronal LPL is involved in the regulation of body weight and composition in response to either the change in quantity (HF feeding) or quality (n-3 PUFA-enriched) of dietary fat. The precise role of LPL in lipid sensing in the brain requires further investigation.
Collapse
Affiliation(s)
- Hong Wang
- Division of Endocrinology, Metabolism, & Diabetes, Department of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Matthew D Taussig
- Division of Endocrinology, Metabolism, & Diabetes, Department of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Nicholas V DiPatrizio
- Department of Pharmacology, University of California, Irvine, Irvine, CA 92617, USA; School of Medicine, University of California, Riverside, Riverside, CA 92521, USA
| | - Kimberley Bruce
- Division of Endocrinology, Metabolism, & Diabetes, Department of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Daniele Piomelli
- Department of Pharmacology, University of California, Irvine, Irvine, CA 92617, USA; Drug Discovery and Development, Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Robert H Eckel
- Division of Endocrinology, Metabolism, & Diabetes, Department of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
44
|
Wang H, Wang Y, Taussig MD, Eckel RH. Sex differences in obesity development in pair-fed neuronal lipoprotein lipase deficient mice. Mol Metab 2016; 5:1025-1032. [PMID: 27689015 PMCID: PMC5034494 DOI: 10.1016/j.molmet.2016.05.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 05/24/2016] [Accepted: 05/24/2016] [Indexed: 02/08/2023] Open
Abstract
Objective Compared to men, postmenopausal women suffer from a disproportionate burden of many co-morbidities associated with obesity, e.g. cardiovascular disease, cancer, and dementia. The underlying mechanism for this sex difference is not well understood but is believed to relate to absence of the protective effect of estrogen through the action of estrogen receptor alpha (ERα) in the central nervous system. With the recently developed neuron-specific lipoprotein lipase deficient mice (NEXLPL−/−) (Wang et al., Cell Metabolism, 2011 [15]), we set to explore the possible role of lipid sensing in sex differences in obesity development. Methods Both male and female NEXLPL−/− mice and littermate WT controls were subjected to pair feeding (pf) where daily food amount given was adjusted according to body weight to match the food intake of ad libitum (ad) fed control WT mice. Food intake and body weight were measured daily, and pair feeding was maintained to 42 wk in male mice and to 38 wk in female mice. Various brain regions of the mice were harvested, and ERα gene expression was examined in both male and female NEXLPL−/− and WT control mice under both ad- and pf-fed conditions. Results Although both male and female NEXLPL−/− mice developed obesity similarly on standard chow, male NEXLPL−/− mice still developed obesity under with pair feeding, but on a much delayed time course, while female NEXLPL−/− mice were protected from extra body weight and fat mass gain compared to pair-fed WT control mice. Pair feeding alone induced extra fat mass gain in both male and female WT mice, and this was mostly driven by the reduction in physical activity. LPL deficiency resulted in an increase in ERα mRNA in the hypothalamus of ad-fed female mice, while pair feeding alone also resulted in an increase of ERα in both female WT control and NEXLPL−/− mice. The effect on increasing ERα by pair feeding and LPL deficiency was additive in pair-fed female NEXLPL−/− mice. ERα mRNA levels were not significantly modified in other brain regions examined, nor in the hypothalamus of male NEXLPL−/− mice compared to control mice. Conclusions These results suggest that the mechanism underlying ERα regulation of body weight interacts with the LPL-dependent lipid processing in the hypothalamus in a sex specific way. ERα could provide the link between brain lipid sensing and sex differences in obesity development. This study has the potential important clinical implication to provide better management for women who suffer from obesity and obesity-related co-morbidities. Male neuronal lipoprotein lipase deficient mice are still obese with pair feeding. Female neuronal lipoprotein lipase deficient mice are not obese with pair feeding. Neuronal LPL deficiency results in an increase in ERa expression in female mice. Pair feeding alone also results in an increase in ERa in both male and female mice. ERa provides the link between brain lipid sensing and sex differences in obesity.
Collapse
Affiliation(s)
- Hong Wang
- Division of Endocrinology, Metabolism, & Diabetes, Department of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Yongping Wang
- Division of Endocrinology, Metabolism, & Diabetes, Department of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Matthew D Taussig
- Division of Endocrinology, Metabolism, & Diabetes, Department of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Robert H Eckel
- Division of Endocrinology, Metabolism, & Diabetes, Department of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
45
|
Dijk W, Beigneux AP, Larsson M, Bensadoun A, Young SG, Kersten S. Angiopoietin-like 4 promotes intracellular degradation of lipoprotein lipase in adipocytes. J Lipid Res 2016; 57:1670-83. [PMID: 27034464 DOI: 10.1194/jlr.m067363] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Indexed: 01/17/2023] Open
Abstract
LPL hydrolyzes triglycerides in triglyceride-rich lipoproteins along the capillaries of heart, skeletal muscle, and adipose tissue. The activity of LPL is repressed by angiopoietin-like 4 (ANGPTL4) but the underlying mechanisms have not been fully elucidated. Our objective was to study the cellular location and mechanism for LPL inhibition by ANGPTL4. We performed studies in transfected cells, ex vivo studies, and in vivo studies with Angptl4(-/-) mice. Cotransfection of CHO pgsA-745 cells with ANGPTL4 and LPL reduced intracellular LPL protein levels, suggesting that ANGPTL4 promotes LPL degradation. This conclusion was supported by studies of primary adipocytes and adipose tissue explants from wild-type and Angptl4(-/-) mice. Absence of ANGPTL4 resulted in accumulation of the mature-glycosylated form of LPL and increased secretion of LPL. Blocking endoplasmic reticulum (ER)-Golgi transport abolished differences in LPL abundance between wild-type and Angptl4(-/-) adipocytes, suggesting that ANGPTL4 acts upon LPL after LPL processing in the ER. Finally, physiological changes in adipose tissue ANGPTL4 expression during fasting and cold resulted in inverse changes in the amount of mature-glycosylated LPL in wild-type mice, but not Angptl4(-/-) mice. We conclude that ANGPTL4 promotes loss of intracellular LPL by stimulating LPL degradation after LPL processing in the ER.
Collapse
Affiliation(s)
- Wieneke Dijk
- Nutrition, Metabolism, and Genomics Group, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
| | - Anne P Beigneux
- Departments of Medicine David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Mikael Larsson
- Departments of Medicine David Geffen School of Medicine, University of California, Los Angeles, CA
| | - André Bensadoun
- Division of Nutritional Sciences, Cornell University, Ithaca, NY
| | - Stephen G Young
- Departments of Medicine David Geffen School of Medicine, University of California, Los Angeles, CA Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Sander Kersten
- Nutrition, Metabolism, and Genomics Group, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands Division of Nutritional Sciences, Cornell University, Ithaca, NY
| |
Collapse
|
46
|
Berland C, Cansell C, Hnasko TS, Magnan C, Luquet S. Dietary triglycerides as signaling molecules that influence reward and motivation. Curr Opin Behav Sci 2016; 9:126-135. [PMID: 28191490 DOI: 10.1016/j.cobeha.2016.03.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The reinforcing and motivational aspects of food are tied to the release of the dopamine in the mesolimbic system (ML). Free fatty acids from triglyceride (TG)-rich particles are released upon action of TG-lipases found at high levels in peripheral oxidative tissue (muscle, heart), but also in the ML. This suggests that local TG-hydrolysis in the ML might regulate food seeking and reward. Indeed, evidence now suggests that dietary TG directly target the ML to regulate amphetamine-induced locomotion and reward seeking behavior. Though the cellular mechanisms of TG action are unresolved, TG act in part through ML lipoprotein lipase, upstream of dopamine 2 receptor (D2R), and show desensitization in conditions of chronically elevated plasma TG as occur in obesity. TG sensing in the ML therefore represents a new mechanism by which chronic consumption of dietary fat might lead to adaptations in the ML and dysregulated feeding behaviors.
Collapse
Affiliation(s)
- Chloé Berland
- Univ Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, F-75205 Paris, France; Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health, München/Neuherberg, Germany; Div. of Metabolic Diseases, Dept. of Medicine, Technische Universität München, Germany
| | - Céline Cansell
- Université de Nice Sophia Antipolis, IPMC, Sophia Antipolis, F-06560, France; CNRS, IPMC, Sophia Antipolis, F-06560, France
| | - Thomas S Hnasko
- Department of Neurosciences, University of California, San Diego, La Jolla CA, USA
| | - Christophe Magnan
- Univ Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, F-75205 Paris, France
| | - Serge Luquet
- Univ Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, F-75205 Paris, France
| |
Collapse
|
47
|
Cansell C, Luquet S. Triglyceride sensing in the reward circuitry: A new insight in feeding behaviour regulation. Biochimie 2016; 120:75-80. [DOI: 10.1016/j.biochi.2015.07.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 07/03/2015] [Indexed: 12/17/2022]
|
48
|
Magnan C, Levin BE, Luquet S. Brain lipid sensing and the neural control of energy balance. Mol Cell Endocrinol 2015; 418 Pt 1:3-8. [PMID: 26415589 DOI: 10.1016/j.mce.2015.09.019] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Revised: 09/14/2015] [Accepted: 09/22/2015] [Indexed: 12/29/2022]
Abstract
Fatty acid (FA) -sensitive neurons are present in the brain, especially the hypothalamus, and play a key role in the neural control of energy and glucose homeostasis including feeding behavior, secretion insulin and action. Subpopulations of neurons in the arcuate and ventromedial hypothalamic nuclei are selectively either activated or inhibited by FA. Molecular effectors of these FA effects include ion channels such as chloride, potassium or calcium. In addition, at least half of the responses in the hypothalamic ventromedial FA neurons are mediated through interaction with the FA translocator/receptor, FAT/CD36, that does not require metabolism to activate intracellular signaling downstream. Recently, an important role of lipoprotein lipase in FA detection has also been demonstrated not only in the hypothalamus, but also in the hippocampus and striatum. Finally, FA could overload energy homeostasis via increased hypothalamic ceramide synthesis which could, in turn, contribute to the pathogenesis of diabetes of obesity and/or type 2 in predisposed individuals by disrupting the endocrine signaling pathways of insulin and/or leptin.
Collapse
Affiliation(s)
- Christophe Magnan
- Univ Paris Diderot, Sorbonne Paris Cité, CNRS UMR 8251, F-75205, Paris, France.
| | - Barry E Levin
- Neurology Service, VA Medical Center, East Orange, NJ, USA; Department of Neurology, Rutgers, NJ Medical School, Newark, NJ, USA
| | - Serge Luquet
- Univ Paris Diderot, Sorbonne Paris Cité, CNRS UMR 8251, F-75205, Paris, France
| |
Collapse
|
49
|
Ng KF, Anderson S, Mayo P, Aung HH, Walton JH, Rutledge JC. Characterizing blood-brain barrier perturbations after exposure to human triglyceride-rich lipoprotein lipolysis products using MRI in a rat model. Magn Reson Med 2015; 76:1246-51. [PMID: 26485349 PMCID: PMC4838551 DOI: 10.1002/mrm.25985] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 08/11/2015] [Accepted: 08/21/2015] [Indexed: 12/11/2022]
Abstract
PURPOSE Previous studies indicated hyperlipidemia may be a risk factor for Alzheimer's disease, but the contributions of postprandial triglyceride-rich lipoprotein (TGRL) are not known. In this study, changes in blood-brain barrier diffusional transport following exposure to human TGRL lipolysis products were studied using MRI in a rat model. METHODS Male Sprague-Dawley rats (∼180-250 g) received an i.v. injection of lipoprotein lipase (LpL)-hydrolyzed TGRL (n = 8, plasma concentration ≈ 150 mg human TGRL/dL). Controls received i.v. injection of either saline (n = 6) or LpL only (n = 6). The (1) H longitudinal relaxation rate R1 = 1/T1 was measured over 18 min using a rapid-acquired refocus-echo (RARE) sequence after each of three injections of the contrast agent Gd-DTPA. Patlak plots were generated for each pixel yielding blood-to-brain transfer coefficients, Ki , chosen for best fit to impermeable, uni-directional influx or bi-directional flux models using the F-test. RESULTS Analysis from a 2-mm slice, 2-mm rostral to the bregma showed a 275% increase of mean Ki during the first 20 min after infusion of human TGRL lipolysis product that differed significantly compared with saline and LpL controls. This difference disappeared by 40 min mark. CONCLUSION These results suggest human TGRL lipolysis products can lead to a transient increase in rat BBB permeability. Magn Reson Med 76:1246-1251, 2016. © 2015 The Authors. Magnetic Resonance in Medicine published by Wiley Periodicals, Inc. on behalf of International Society for Magnetic Resonance in Medicine. This is an open access article under the terms of the Creative Commons Attribution NonCommercial License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited and is not used for commercial purposes.
Collapse
Affiliation(s)
- Kit Fai Ng
- School of Medicine, Division of Cardiovascular Medicine, University of California, Davis, California, USA
| | - Steve Anderson
- School of Medicine, Department of Physiology and Membrane Biology, University of California, Davis, California, USA
| | - Patrice Mayo
- School of Medicine, Division of Cardiovascular Medicine, University of California, Davis, California, USA
| | - Hnin Hnin Aung
- School of Medicine, Division of Cardiovascular Medicine, University of California, Davis, California, USA
| | - Jeffrey H Walton
- NMR Facility and Biomedical Engineering Graduate Group, University of California, Davis, California, USA
| | - John C Rutledge
- School of Medicine, Division of Cardiovascular Medicine, University of California, Davis, California, USA.
| |
Collapse
|
50
|
Fasting for 21days leads to changes in adipose tissue and liver physiology in juvenile checkered garter snakes (Thamnophis marcianus). Comp Biochem Physiol A Mol Integr Physiol 2015; 190:68-74. [PMID: 26358832 DOI: 10.1016/j.cbpa.2015.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 09/02/2015] [Accepted: 09/02/2015] [Indexed: 12/17/2022]
Abstract
Snakes often undergo periods of prolonged fasting and, under certain conditions, can survive years without food. Despite this unique phenomenon, there are relatively few reports of the physiological adaptations to fasting in snakes. At post-prandial day 1 (fed) or 21 (fasting), brain, liver, and adipose tissues were collected from juvenile checkered garter snakes (Thamnophis marcianus). There was greater glycerol-3-phosphate dehydrogenase (G3PDH)-specific activity in the liver of fasted than fed snakes (P=0.01). The mRNA abundance of various fat metabolism-associated factors was measured in brain, liver, and adipose tissue. Lipoprotein lipase (LPL) mRNA was greater in fasted than fed snakes in the brain (P=0.04). Adipose triglyceride lipase (ATGL; P=0.006) mRNA was greater in the liver of fasted than fed snakes. In adipose tissue, expression of peroxisome proliferator-activated receptor (PPAR)γ (P=0.01), and fatty acid binding protein 4 (P=0.03) was greater in fed than fasted snakes. Analysis of adipocyte morphology revealed that cross-sectional area (P=0.095) and diameter (P=0.27) were not significantly different between fed and fasted snakes. Results suggest that mean adipocyte area can be preserved during fasting by dampening lipid biosynthesis while not changing rates of lipid hydrolysis. In the liver, however, extensive lipid remodeling may provide energy and lipoproteins to maintain lipid structural integrity during energy restriction. Because the duration of fasting was not sufficient to change adipocyte size, results suggest that the liver is important as a short-term provider of energy in the snake.
Collapse
|