1
|
Fongsaran C, Jirakanwisal K, Peng BH, Fracassi A, Taglialatela G, Dineley KT, Paessler S, Cisneros IE. Arbovirus infection increases the risk for the development of neurodegenerative disease pathology in the murine model. Brain Behav Immun Health 2024; 38:100780. [PMID: 38706571 PMCID: PMC11067009 DOI: 10.1016/j.bbih.2024.100780] [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: 11/20/2023] [Revised: 03/04/2024] [Accepted: 04/23/2024] [Indexed: 05/07/2024] Open
Abstract
Alzheimer's disease is classified as a progressive disorder resulting from protein misfolding, also known as proteinopathies. Proteinopathies include synucleinopathies triggered by misfolded amyloid α-synuclein, tauopathies triggered by misfolded tau, and amyloidopathies triggered by misfolded amyloid of which Alzheimer's disease (β-amyloid) is most prevalent. Most neurodegenerative diseases (>90%) are not due to dominantly inherited genetic causes. Instead, it is thought that the risk for disease is a complicated interaction between inherited and environmental risk factors that, with age, drive pathology that ultimately results in neurodegeneration and disease onset. Since it is increasingly appreciated that encephalitic viral infections can have profoundly detrimental neurological consequences long after the acute infection has resolved, we tested the hypothesis that viral encephalitis exacerbates the pathological profile of protein-misfolding diseases. Using a robust, reproducible, and well-characterized mouse model for β-amyloidosis, Tg2576, we studied the contribution of alphavirus-induced encephalitis (TC-83 strain of VEEV to model alphavirus encephalitis viruses) on the progression of neurodegenerative pathology. We longitudinally evaluated neurological, neurobehavioral, and cognitive levels, followed by a post-mortem analysis of brain pathology focusing on neuroinflammation. We found more severe cognitive deficits and brain pathology in Tg2576 mice inoculated with TC-83 than in their mock controls. These data set the groundwork to investigate sporadic Alzheimer's disease and treatment interventions for this infectious disease risk factor.
Collapse
Affiliation(s)
- Chanida Fongsaran
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
- Neuroinfectious Diseases, University of Texas Medical Branch, Galveston, TX, USA
| | - Krit Jirakanwisal
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
- Neuroinfectious Diseases, University of Texas Medical Branch, Galveston, TX, USA
| | - Bi-Hung Peng
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Anna Fracassi
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Giulio Taglialatela
- Neuroinfectious Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Kelly T. Dineley
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
- Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, TX, USA
| | - Slobodan Paessler
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Irma E. Cisneros
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
- Neuroinfectious Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
2
|
Six genetically linked mutations in the CD36 gene significantly delay the onset of Alzheimer's disease. Sci Rep 2022; 12:10994. [PMID: 35768560 PMCID: PMC9243110 DOI: 10.1038/s41598-022-15299-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/22/2022] [Indexed: 11/08/2022] Open
Abstract
The risk of Alzheimer’s disease (AD) has a strong genetic component, also in the case of late-onset AD (LOAD). Attempts to sequence whole genome in large populations of subjects have identified only a few mutations common to most of the patients with AD. Targeting smaller well-characterized groups of subjects where specific genetic variations in selected genes could be related to precisely defined psychological traits typical of dementia is needed to better understand the heritability of AD. More than one thousand participants, categorized according to cognitive deficits, were assessed using 14 psychometric tests evaluating performance in five cognitive domains (attention/working memory, memory, language, executive functions, visuospatial functions). CD36 was selected as a gene previously shown to be implicated in the etiology of AD. A total of 174 polymorphisms were tested for associations with cognition-related traits and other AD-relevant data using the next generation sequencing. Several associations between single nucleotide polymorphisms (SNP’s) and the cognitive deficits have been found (rs12667404 with language performance, rs3211827 and rs41272372 with executive functions, rs137984792 with visuospatial performance). The most prominent association was found between a group of genotypes in six genetically linked and the age at which the AD patients presented with, or developed, a full-blown dementia. The identified alleles appear to be associated with a delay in the onset of LOAD. In silico studies suggested that the SNP’s alter the expression of CD36 thus potentially affecting CD36-related neuroinflammation and other molecular and cellular mechanisms known to be involved in the neuronal loss leading to AD. The main outcome of the study is an identification of a set of six new mutations apparently conferring a distinct protection against AD and delaying the onset by about 8 years. Additional mutations in CD36 associated with certain traits characteristic of the cognitive decline in AD have also been found.
Collapse
|
3
|
Fu H, Hsu J, Li T, Yeh S, Chen C. Safety assessment of HEA-enriched Cordyceps cicadae mycelia on the central nervous system (CNS), cardiovascular system, and respiratory system in ICR male mice. Food Sci Nutr 2021; 9:4905-4915. [PMID: 34532002 PMCID: PMC8441276 DOI: 10.1002/fsn3.2440] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 06/01/2021] [Accepted: 06/11/2021] [Indexed: 11/06/2022] Open
Abstract
Cordyceps cicadae, an entomopathogenic fungus, is a source of traditional Chinese medicine in China. Due to the low yield of wild C. cicadae, artificial cultivation approaches will be needed to meet the increasing market demand. Using bioreactor culture can increase mass production and the abundance of the active component, N6-(2-hydroxyethyl)-adenosine (HEA). Here, we describe a safety assessment for a novel mycelium preparation method. Many studies have confirmed the safety of C. cicadae mycelia. However, the acute safety pharmacology of the C. cicadae enriched with the high HEA (3.90 mg/g) compound has not been evaluated. This study evaluated the central nervous system (CNS), cardiovascular system, and respiratory system in ICR male mice via oral gavage administration. For each requested item, two batches of eight mice tested on a vehicle (0.5% carboxymethyl cellulose, CMC) and C. cicadae mycelia (1,000 mg/kg) were performed. The heart rate at 60 min for the vehicle and C. cicadae mycelium treatment was 700.3 ± 55.4 and 603.0 ± 42.3 bpm, respectively (p = .4279). For echocardiographic analysis, the LV mass of the vehicle and drug treatment was 86.7 ± 6.4 and 80.2 ± 7.7, respectively (p = .0933). In the respiratory test, the tidal volume of the vehicle and drug treatments was 0.11 ± 0.01 and 0.14 ± 0.01 at 60 min, respectively (p = .4262). These results demonstrate that the oral administration of HEA-enriched C. cicadae mycelia is safe for the CNS, cardiovascular, and respiratory systems.
Collapse
Affiliation(s)
- Hsin‐I Fu
- Biotech Research InstituteGrape King Bio LtdTaoyuan CityTaiwan
| | - Jui‐Hsia Hsu
- Biotech Research InstituteGrape King Bio LtdTaoyuan CityTaiwan
| | - Tsung‐Ju Li
- Biotech Research InstituteGrape King Bio LtdTaoyuan CityTaiwan
| | - Shu‐Hsing Yeh
- Biotech Research InstituteGrape King Bio LtdTaoyuan CityTaiwan
| | - Chin‐Chu Chen
- Biotech Research InstituteGrape King Bio LtdTaoyuan CityTaiwan
- Institute of Food Science and TechnologyNational Taiwan UniversityTaipei CityTaiwan
- Department of Food Science, Nutrition and Nutraceutical BiotechnologyShih Chien UniversityTaipei CityTaiwan
| |
Collapse
|
4
|
Abstract
The SmithKline, Harwell, Imperial College, Royal Hospital, Phenotype Assessment (SHIRPA) is a rapid battery of tests comprising 42 measurements of motor activity, coordination, postural control, muscle tone, autonomic functions, and emotional reactivity, as well as reflexes dependent on visual, auditory, and tactile modalities. Individual scores in SHIRPA are sensitive in detecting phenotypes of several experimental models of neural disease, especially cerebellar degeneration and Alzheimer disease, and combined subscores have been useful in estimating the impact of vascular anomalies and exposure to infectious agents. In cerebellar degeneration, weak forelimb grip, impaired wire maneuver and air righting, and negative geotaxis appear as prevalent features. Most of the measures in the battery are susceptible to change after gene modifications or physiological alterations. SHIRPA can be used both in adult mice and mice in the preweaning period to screen for sensorimotor function and emotional reactivity, not selective attention or memory. © 2021 Wiley Periodicals LLC Basic Protocol: Step-by-step procedure for SHIRPA.
Collapse
Affiliation(s)
- Robert Lalonde
- Department of Psychology, University of Rouen, Mont-Saint-Aignan, France
- Laboratory of Stress, Immunity, Pathogens, Medical School, University of Lorraine, Vandœuvre-les-Nancy, France
| | | | - Catherine Strazielle
- Laboratory of Stress, Immunity, Pathogens, Medical School, University of Lorraine, Vandœuvre-les-Nancy, France
- CHRU Nancy, Vandœuvre-les-Nancy, France
| |
Collapse
|
5
|
CD36 deficiency affects depressive-like behaviors possibly by modifying gut microbiota and the inflammasome pathway in mice. Transl Psychiatry 2021; 11:16. [PMID: 33414380 PMCID: PMC7791141 DOI: 10.1038/s41398-020-01130-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 11/29/2020] [Accepted: 12/01/2020] [Indexed: 02/07/2023] Open
Abstract
Both inflammatory processes and gut microbiota have been implicated in the pathophysiology of depressive disorders. The class B scavenger receptor CD36 is involved in the cytotoxicity associated with inflammation. However, its role in depression has not yet been examined. In this study, we investigated whether CD36 affects depression by modulating the microbiota-gut-inflammasome-brain axis. We used CD36-/- (knockout) mice subjected to chronic social defeat stress, and measured the expression of CD36 in these depressed mice and in patients with depression. The hippocampus of CD36-/- mice was used to investigate changes in the NLRP3 inflammasome signaling pathway. The 16S rRNA gene sequence-based approach was used to compare the cecal microbial communities in CD36-/- and WT mice. The CD36 deficiency in CD36-/- mice alleviated chronic stress-induced depression-like behaviors. CD36 was upregulated in depressed mice as well as in depressed patients. Furthermore, the NLRP3 inflammasome signaling pathway was downregulated in the hippocampus of CD36-/- mice. The Simpson Diversity Index revealed increased cecal bacterial alpha-diversity in the CD36-/- mice. Among genera, Bacteroides, Rikenella, and Alloprevotella were significantly more abundant in the CD36-/- mice, whereas Allobaculum was less abundant, consistent with the attenuated inflammation in the hippocampus of CD36-/- mice. Our findings suggest that CD36 deficiency changes the gut microbiota composition, which in turn may impact depressive-like behaviors by affecting the inflammasome pathway.
Collapse
|
6
|
Dobri AM, Dudău M, Enciu AM, Hinescu ME. CD36 in Alzheimer's Disease: An Overview of Molecular Mechanisms and Therapeutic Targeting. Neuroscience 2020; 453:301-311. [PMID: 33212223 DOI: 10.1016/j.neuroscience.2020.11.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/17/2020] [Accepted: 11/03/2020] [Indexed: 12/11/2022]
Abstract
CD36 is a membrane protein with wide distribution in the human body, is enriched in the monocyte-macrophage system and endothelial cells, and is involved in the cellular uptake of long chain fatty acids (LCFA) and oxidized low-density lipoproteins. It is also a scavenger receptor, binding hydrophobic amyloid fibrils found in the Alzheimer's disease (AD) brain. In neurobiology research, it has been mostly studied in relationship with chronic ischemia and stroke, but it was also related to amyloid clearance by microglial phagocytosis. In AD animal models, amyloid binding to CD36 has been consistently correlated with a pro-inflammatory response. Therapeutic approaches have two main focuses: CD36 blockade with monoclonal antibodies or small molecules, which is beneficial in terms of the inflammatory milieu, and upregulation of CD36 for increased amyloid clearance. The balance of the two approaches, centered on microglia, is poorly understood. Furthermore, CD36 evaluation in AD clinical studies is still at a very early stage and there is a gap in the knowledge regarding the impact of LCFA on AD progression and CD36 expression and genetic phenotype. This review summarizes the role played by CD36 in the pathogenic amyloid cascade and explore the translatability of preclinical data towards clinical research.
Collapse
Affiliation(s)
- Ana-Maria Dobri
- "Victor Babes" National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania; "Carol Davila" University of Medicine and Pharmacy, 5 Eroilor Sanitari Blvd, 050047 Bucharest, Romania.
| | - Maria Dudău
- "Victor Babes" National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania; "Carol Davila" University of Medicine and Pharmacy, 5 Eroilor Sanitari Blvd, 050047 Bucharest, Romania.
| | - Ana-Maria Enciu
- "Victor Babes" National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania; "Carol Davila" University of Medicine and Pharmacy, 5 Eroilor Sanitari Blvd, 050047 Bucharest, Romania.
| | - Mihail Eugen Hinescu
- "Victor Babes" National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania; "Carol Davila" University of Medicine and Pharmacy, 5 Eroilor Sanitari Blvd, 050047 Bucharest, Romania
| |
Collapse
|
7
|
Qin Y, Jiang X, Li W, Li J, Tian T, Zang G, Fang L, Zhou C, Xu B, Gong X, Huang C, Yang X, Bai M, Fan L, Xie P. Chronic mild stress leads to aberrant glucose energy metabolism in depressed Macaca fascicularis models. Psychoneuroendocrinology 2019; 107:59-69. [PMID: 31108306 DOI: 10.1016/j.psyneuen.2019.05.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 05/05/2019] [Accepted: 05/09/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Major depressive disorder (MDD) is a pathophysiologically uncharacterized mental illness with complex etiology and clinical manifestations. Rodent depression-like models have been widely used to mimic the morbid state of depression. However, research on emotional disorders can also benefit from the use of models in non-human primates, which share a wide range of genetic and social similarities with humans. METHODS To investigate the pathophysiological mechanisms of depression, we established two models, naturally occurring depression cynomolgus (NOD) and social plus visual isolation-induced depression cynomolgus (SVC), imitating chronic mild or acute intense stress, respectively. We used i-TRAQ (isobaric tags for relative and absolute quantitation)-based quantitative proteomics and shotgun proteomics to identify differentially expressed proteins in cerebrospinal fluid (CSF) of the two monkey models and human MDD patients. We also used DAVID and ingenuity pathway analysis (IPA) for further bioinformatic investigation. RESULTS In behavioral tests, NOD monkeys achieved higher scores in depression-like and anxiety-like behavioral measures, and spent more time on ingesting, thermoregulatory, and locomotive actions than SVC monkeys. A total of 902 proteins were identified by i-TRAQ, and 40 differentially expressed proteins were identified in each of the NOD-CON1 and SVC-CON2 groups. Application of DAVID revealed dysregulation of energy metabolism in the NOD group, whereas lipid metabolism and inflammatory response pathways were significantly altered in the SVC group. Use of IPA and Cytoscape showed that the oxygen species metabolic process glycolysis I/gluconeogenesis I, accompanied by downregulation of tubulin beta 3 class III (TUBB3), RAC-alpha serine/threonine-protein kinase (AKT1), and glyceraldehyde-3-phosphate dehydrogenase (GAPDH), was the most significantly affected pathway in the NOD group. Furthermore, 152 differentially expressed proteins in human MDD patients also revealed disruption of glucose energy metabolism. Significantly aberrant energy metabolism in various brain regions and the plasma and liver of chronic unpredictable mild stress rodent samples were also observed in a previous study. CONCLUSIONS Our results reveal for the first time the overall CSF protein profiles of two cynomolgus monkey models of depression. We propose that chronic mild stress may affect the disruption of glucose energy metabolism in NOD cynomolgus monkeys and rodents. These findings promote our understanding of the pathophysiology of MDD and may help to identify novel therapeutic targets.
Collapse
Affiliation(s)
- Yinhua Qin
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China; Key Laboratory of Laboratory Medical Diagnostics of Education, Department of Laboratory Medicine, Chongqing 400016, China
| | - XiaoFeng Jiang
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Wei Li
- Department of Neurology, Army Medical Center of PLA, Chongqing 400042, China
| | - Jie Li
- Clinical Medicine Research Center, Dazhou Central Hospital, Sichuan 635000, China
| | - Tian Tian
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Guangchao Zang
- Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China; Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center of Chongqing Medical University, Chongqing 400016, China
| | - Liang Fang
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402460, China
| | - Chanjuan Zhou
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402460, China
| | - Bin Xu
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xue Gong
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Cheng Huang
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xun Yang
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Mengge Bai
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China; Key Laboratory of Laboratory Medical Diagnostics of Education, Department of Laboratory Medicine, Chongqing 400016, China
| | - Li Fan
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Peng Xie
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
8
|
He Y, Li W, Tian Y, Chen X, Cheng K, Xu K, Li C, Wang H, Qu C, Wang C, Li P, Chen H, Xie P. iTRAQ-based proteomics suggests LRP6, NPY and NPY2R perturbation in the hippocampus involved in CSDS may induce resilience and susceptibility. Life Sci 2018; 211:102-117. [DOI: 10.1016/j.lfs.2018.09.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 09/05/2018] [Accepted: 09/06/2018] [Indexed: 12/12/2022]
|
9
|
Zhang W, Chen R, Yang T, Xu N, Chen J, Gao Y, Stetler RA. Fatty acid transporting proteins: Roles in brain development, aging, and stroke. Prostaglandins Leukot Essent Fatty Acids 2018; 136:35-45. [PMID: 28457600 PMCID: PMC5650946 DOI: 10.1016/j.plefa.2017.04.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 04/16/2017] [Accepted: 04/20/2017] [Indexed: 12/18/2022]
Abstract
Polyunsaturated fatty acids are required for the brain development and significantly impact aging and stroke. Due to the hydrophobicity of fatty acids, fatty acids transportation related proteins that include fatty acid binding proteins (FABPs), long chain acyl-coA synthase (ACS), fatty acid transportation proteins (FATPs), fatty acid translocase (FAT/CD36) and newly reported major facilitator superfamily domain-containing protein (Mfsd2a) play critical roles in the uptake of various fatty acids, especially polyunsaturated fatty acids. They are not only involved in neurodevelopment, but also have great impact on neurological disease, such as aging related dementia and stroke.
Collapse
Affiliation(s)
- Wenting Zhang
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Ruiying Chen
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Tuo Yang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Na Xu
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Jun Chen
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China; Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Education and Clinical Center Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China; Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| | - R Anne Stetler
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China; Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Education and Clinical Center Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA.
| |
Collapse
|
10
|
Lee S, Tsuzuki S, Amitsuka T, Masuda D, Yamashita S, Inoue K. CD36 involvement in the olfactory perception of oleic aldehyde, an odour-active volatile compound, in mice. Biomed Res 2018. [PMID: 28637956 DOI: 10.2220/biomedres.38.207] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Cluster of differentiation 36 (CD36) is a broadly expressed transmembrane receptor that has multiple ligands. It has been found to occur abundantly on the surface of the olfactory epithelium in mice and postulated to play a role in mammalian olfaction. However, there have been no ethological analyses of the mammalian behaviour showing CD36 involvement in the olfactory perception of a distinct odour-active volatile compound. In this study, we aimed to assess whether mammals perceive oleic aldehyde, an odour-active volatile that serves as a potential CD36 ligand, and if so, whether CD36 is involved in the sensing by following measurements using CD36-knockout mice and their wild-type littermates. In a two-bottle choice test, wild-type mice, but not CD36-knockout mice, discriminated a sucrose solution containing oleic aldehyde from the sucrose solution alone. To assess the importance of the olfactory system in the oleic aldehyde perception, we conducted an exploration test where the animals could rely primarily on the odour of test volatiles for recognition. We found that the wild-type, but not CD36-knockout mice, were aware of the compound. Our results provide behavioural evidence that CD36 plays a role in the perception of specific odour-active volatile compounds in the nasal cavity.
Collapse
Affiliation(s)
- Shinhye Lee
- Laboratory of Nutrition Chemistry, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University
| | - Satoshi Tsuzuki
- Laboratory of Nutrition Chemistry, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University
| | | | - Daisaku Masuda
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Shizuya Yamashita
- Department of Community Medicine, Osaka University Graduate School of Medicine
| | - Kazuo Inoue
- Laboratory of Nutrition Chemistry, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University
| |
Collapse
|
11
|
Huo R, Zeng B, Zeng L, Cheng K, Li B, Luo Y, Wang H, Zhou C, Fang L, Li W, Niu R, Wei H, Xie P. Microbiota Modulate Anxiety-Like Behavior and Endocrine Abnormalities in Hypothalamic-Pituitary-Adrenal Axis. Front Cell Infect Microbiol 2017; 7:489. [PMID: 29250490 PMCID: PMC5715198 DOI: 10.3389/fcimb.2017.00489] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 11/13/2017] [Indexed: 12/14/2022] Open
Abstract
Intestinal microbes are an important system in the human body, with significant effects on behavior. An increasing body of research indicates that intestinal microbes affect brain function and neurogenesis, including sensitivity to stress. To investigate the effects of microbial colonization on behavior, we examined behavioral changes associated with hormones and hormone receptors in the hypothalamic-pituitary-adrenal (HPA) axis under stress. We tested germ-free (GF) mice and specific pathogen-free (SPF) mice, divided into four groups. A chronic restraint stress (CRS) protocol was utilized to induce external pressure in two stress groups by restraining mice in a conical centrifuge tube for 4 h per day for 21 days. After CRS, Initially, GF restraint-stressed mice explored more time than SPF restraint-stressed mice in the center and total distance of the OFT. Moreover, the CRH, ACTH, CORT, and ALD levels in HPA axis of GF restraint-stressed mice exhibited a significantly greater increase than those of SPF restraint-stressed mice. Finally, the Crhr1 mRNA levels of GF CRS mice were increased compared with SPF CRS mice. However, the Nr3c2 mRNA levels of GF CRS mice were decreased compared with SPF CRS mice. All results revealed that SPF mice exhibited more anxiety-like behavior than GF mice under the same external stress. Moreover, we also found that GF mice exhibited significant differences in, hormones, and hormone receptors compared with SPF mice. In conclusion, Imbalances of the HPA axis caused by intestinal microbes could affect the neuroendocrine system in the brain, resulting in an anxiety-like behavioral phenotype. This study suggested that intervention into intestinal microflora may provide a new approach for treating stress-related diseases.
Collapse
Affiliation(s)
- Ran Huo
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, China.,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China.,Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Benhua Zeng
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Li Zeng
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China.,Department of Neurology, First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Ke Cheng
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, China.,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Bo Li
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, China.,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China.,Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Yuanyuan Luo
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, China.,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Haiyang Wang
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Chanjuan Zhou
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Liang Fang
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Wenxia Li
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Rong Niu
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Hong Wei
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Peng Xie
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, China.,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China.,Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China.,Department of Neurology, First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| |
Collapse
|
12
|
Bröker-Lai J, Kollewe A, Schindeldecker B, Pohle J, Nguyen Chi V, Mathar I, Guzman R, Schwarz Y, Lai A, Weißgerber P, Schwegler H, Dietrich A, Both M, Sprengel R, Draguhn A, Köhr G, Fakler B, Flockerzi V, Bruns D, Freichel M. Heteromeric channels formed by TRPC1, TRPC4 and TRPC5 define hippocampal synaptic transmission and working memory. EMBO J 2017; 36:2770-2789. [PMID: 28790178 DOI: 10.15252/embj.201696369] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 07/01/2017] [Accepted: 07/07/2017] [Indexed: 12/30/2022] Open
Abstract
Canonical transient receptor potential (TRPC) channels influence various neuronal functions. Using quantitative high-resolution mass spectrometry, we demonstrate that TRPC1, TRPC4, and TRPC5 assemble into heteromultimers with each other, but not with other TRP family members in the mouse brain and hippocampus. In hippocampal neurons from Trpc1/Trpc4/Trpc5-triple-knockout (Trpc1/4/5-/-) mice, lacking any TRPC1-, TRPC4-, or TRPC5-containing channels, action potential-triggered excitatory postsynaptic currents (EPSCs) were significantly reduced, whereas frequency, amplitude, and kinetics of quantal miniature EPSC signaling remained unchanged. Likewise, evoked postsynaptic responses in hippocampal slice recordings and transient potentiation after tetanic stimulation were decreased. In vivo, Trpc1/4/5-/- mice displayed impaired cross-frequency coupling in hippocampal networks and deficits in spatial working memory, while spatial reference memory was unaltered. Trpc1/4/5-/- animals also exhibited deficiencies in adapting to a new challenge in a relearning task. Our results indicate the contribution of heteromultimeric channels from TRPC1, TRPC4, and TRPC5 subunits to the regulation of mechanisms underlying spatial working memory and flexible relearning by facilitating proper synaptic transmission in hippocampal neurons.
Collapse
Affiliation(s)
- Jenny Bröker-Lai
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Astrid Kollewe
- Institute of Physiology, University of Freiburg, Freiburg, Germany
| | - Barbara Schindeldecker
- Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Jörg Pohle
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany.,Physiology of Neural Networks, Psychiatry/Psychopharmacology, Central Institute of Mental Health, J5, Heidelberg University, Mannheim, Germany
| | - Vivan Nguyen Chi
- Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Ilka Mathar
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Raul Guzman
- Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Yvonne Schwarz
- Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Alan Lai
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Petra Weißgerber
- Experimental and Clinical Pharmacology and Toxicology, Saarland University, Homburg, Germany
| | | | - Alexander Dietrich
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, München, Germany
| | - Martin Both
- Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Rolf Sprengel
- Max Planck Research Group of the Max Planck Institute for Medical Research at the Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Andreas Draguhn
- Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Georg Köhr
- Physiology of Neural Networks, Psychiatry/Psychopharmacology, Central Institute of Mental Health, J5, Heidelberg University, Mannheim, Germany
| | - Bernd Fakler
- Institute of Physiology, University of Freiburg, Freiburg, Germany.,BIOSS, Center for Biological Signaling Studies, University of Freiburg, Freiburg, Germany‡
| | - Veit Flockerzi
- Experimental and Clinical Pharmacology and Toxicology, Saarland University, Homburg, Germany
| | - Dieter Bruns
- Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Marc Freichel
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
13
|
Psychosocial stress on neuroinflammation and cognitive dysfunctions in Alzheimer's disease: the emerging role for microglia? Neurosci Biobehav Rev 2017; 77:148-164. [PMID: 28185874 DOI: 10.1016/j.neubiorev.2017.01.046] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 01/20/2017] [Accepted: 01/31/2017] [Indexed: 01/22/2023]
Abstract
Chronic psychosocial stress is increasingly recognized as a risk factor for late-onset Alzheimer's disease (LOAD) and associated cognitive deficits. Chronic stress also primes microglia and induces inflammatory responses in the adult brain, thereby compromising synapse-supportive roles of microglia and deteriorating cognitive functions during aging. Substantial evidence demonstrates that failure of microglia to clear abnormally accumulating amyloid-beta (Aβ) peptide contributes to neuroinflammation and neurodegeneration in AD. Moreover, genome-wide association studies have linked variants in several immune genes, such as TREM2 and CD33, the expression of which in the brain is restricted to microglia, with cognitive dysfunctions in LOAD. Thus, inflammation-promoting chronic stress may create a vicious cycle of aggravated microglial dysfunction accompanied by increased Aβ accumulation, collectively exacerbating neurodegeneration. Surprisingly, however, little is known about whether and how chronic stress contributes to microglia-mediated neuroinflammation that may underlie cognitive impairments in AD. This review aims to summarize the currently available clinical and preclinical data and outline potential molecular mechanisms linking stress, microglia and neurodegeneration, to foster future research in this field.
Collapse
|
14
|
Zapata I, Serpell JA, Alvarez CE. Genetic mapping of canine fear and aggression. BMC Genomics 2016; 17:572. [PMID: 27503363 PMCID: PMC4977763 DOI: 10.1186/s12864-016-2936-3] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 07/13/2016] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Fear/anxiety and anger/aggression greatly influence health, quality of life and social interactions. They are a huge burden to wellbeing, and personal and public economics. However, while much is known about the physiology and neuroanatomy of such emotions, little is known about their genetics - most importantly, why some individuals are more susceptible to pathology under stress. RESULTS We conducted genomewide association (GWA) mapping of breed stereotypes for many fear and aggression traits across several hundred dogs from diverse breeds. We confirmed those findings using GWA in a second cohort of partially overlapping breeds. Lastly, we used the validated loci to create a model that effectively predicted fear and aggression stereotypes in a third group of dog breeds that were not involved in the mapping studies. We found that i) known IGF1 and HMGA2 loci variants for small body size are associated with separation anxiety, touch-sensitivity, owner directed aggression and dog rivalry; and ii) two loci, between GNAT3 and CD36 on chr18, and near IGSF1 on chrX, are associated with several traits, including touch-sensitivity, non-social fear, and fear and aggression that are directed toward unfamiliar dogs and humans. All four genome loci are among the most highly evolutionarily-selected in dogs, and each of those was previously shown to be associated with morphological traits. We propose that the IGF1 and HMGA2 loci are candidates for identical variation being associated with both behavior and morphology. In contrast, we show that the GNAT3-CD36 locus has distinct variants for behavior and morphology. The chrX region is a special case due to its extensive linkage disequilibrium (LD). Our evidence strongly suggests that sociability (which we propose is associated with HS6ST2) and fear/aggression are two distinct GWA loci within this LD block on chrX, but there is almost perfect LD between the peaks for fear/aggression and animal size. CONCLUSIONS We have mapped many canine fear and aggression traits to single haplotypes at the GNAT3-CD36 and IGSF1 loci. CD36 is widely expressed, but areas of the amygdala and hypothalamus are among the brain regions with highest enrichment; and CD36-knockout mice are known to have significantly increased anxiety and aggression. Both of the other genes have very high tissue-specificity and are very abundantly expressed in brain regions that comprise the core anatomy of fear and aggression - the amygdala to hypothalamic-pituitary-adrenal (HPA) axis. We propose that reduced-fear variants at these loci may have been involved in the domestication process.
Collapse
Affiliation(s)
- Isain Zapata
- Center for Molecular and Human Genetics, The Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205 USA
| | - James A. Serpell
- Center for the Interaction of Animals and Society, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Carlos E. Alvarez
- Center for Molecular and Human Genetics, The Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205 USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210 USA
- Department of Veterinary Clinical Sciences, The Ohio State University College of Veterinary Medicine, Columbus, OH 43210 USA
| |
Collapse
|
15
|
Chen X, Chen X, Xu Y, Yang W, Wu N, Ye H, Yang JY, Hong Q, Xin Y, Yang MQ, Deng Y, Duan S. Association of six CpG-SNPs in the inflammation-related genes with coronary heart disease. Hum Genomics 2016; 10 Suppl 2:21. [PMID: 27461004 PMCID: PMC4965732 DOI: 10.1186/s40246-016-0067-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Background Chronic inflammation has been widely considered to be the major risk factor of coronary heart disease (CHD). The goal of our study was to explore the possible association with CHD for inflammation-related single nucleotide polymorphisms (SNPs) involved in cytosine-phosphate-guanine (CpG) dinucleotides. A total of 784 CHD patients and 739 non-CHD controls were recruited from Zhejiang Province, China. Using the Sequenom MassARRAY platform, we measured the genotypes of six inflammation-related CpG-SNPs, including IL1B rs16944, IL1R2 rs2071008, PLA2G7 rs9395208, FAM5C rs12732361, CD40 rs1800686, and CD36 rs2065666). Allele and genotype frequencies were compared between CHD and non-CHD individuals using the CLUMP22 software with 10,000 Monte Carlo simulations. Results Allelic tests showed that PLA2G7 rs9395208 and CD40 rs1800686 were significantly associated with CHD. Moreover, IL1B rs16944, PLA2G7 rs9395208, and CD40 rs1800686 were shown to be associated with CHD under the dominant model. Further gender-based subgroup tests showed that one SNP (CD40 rs1800686) and two SNPs (FAM5C rs12732361 and CD36 rs2065666) were associated with CHD in females and males, respectively. And the age-based subgroup tests indicated that PLA2G7 rs9395208, IL1B rs16944, and CD40 rs1800686 were associated with CHD among individuals younger than 55, younger than 65, and over 65, respectively. Conclusions In conclusion, all the six inflammation-related CpG-SNPs (rs16944, rs2071008, rs12732361, rs2065666, rs9395208, and rs1800686) were associated with CHD in the combined or subgroup tests, suggesting an important role of inflammation in the risk of CHD.
Collapse
Affiliation(s)
- Xiaomin Chen
- Cardiovascular Center of Ningbo First Hospital, Ningbo University, Ningbo, Zhejiang, 315010, China
| | - Xiaoying Chen
- School of Medicine, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Yan Xu
- School of Medicine, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - William Yang
- Texas Advanced Computing Center, University of Texas at Austin, 10100 Burnet Road (R8700), Austin, TX, 78758-4497, USA
| | - Nan Wu
- Cardiovascular Center of Ningbo First Hospital, Ningbo University, Ningbo, Zhejiang, 315010, China.,School of Medicine, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Huadan Ye
- School of Medicine, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Jack Y Yang
- MidSouth Bioinformatics Center, Department of Information Science, George Washington Donaghey College of Engineering and Information Science, and Joint Bioinformatics Graduate Program, University of Arkansas at Little Rock and University of Arkansas for Medical Sciences, 2881 S. University Ave, Little Rock, AR, 72204, USA
| | - Qingxiao Hong
- School of Medicine, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Yanfei Xin
- Center of Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, 310007, China
| | - Mary Qu Yang
- MidSouth Bioinformatics Center, Department of Information Science, George Washington Donaghey College of Engineering and Information Science, and Joint Bioinformatics Graduate Program, University of Arkansas at Little Rock and University of Arkansas for Medical Sciences, 2881 S. University Ave, Little Rock, AR, 72204, USA
| | - Youping Deng
- Medical College, Wuhan University of Science and Technology, Wuhan, 430064, China.,Department of Internal Medicine and Biochemistry, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Shiwei Duan
- School of Medicine, Ningbo University, Ningbo, Zhejiang, 315211, China.
| |
Collapse
|
16
|
Quantitative proteomics analysis of the liver reveals immune regulation and lipid metabolism dysregulation in a mouse model of depression. Behav Brain Res 2016; 311:330-339. [PMID: 27247144 DOI: 10.1016/j.bbr.2016.05.057] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/25/2016] [Accepted: 05/27/2016] [Indexed: 11/20/2022]
Abstract
Major depressive disorder (MDD) is a highly prevalent and debilitating mental illness with substantial impairments in quality of life and functioning. However, the pathophysiology of major depression remains poorly understood. Combining the brain and body should provide a comprehensive understanding of the etiology of MDD. As the largest internal organ of the human body, the liver has an important function, yet no proteomic study has assessed liver protein expression in a preclinical model of depression. Using the chronic unpredictable mild stress (CUMS) mouse model of depression, differential protein expression between CUMS and control (CON) mice was examined in the liver proteome using isobaric tag for relative and absolute quantitation (iTRAQ) coupled with tandem mass spectrometry. More than 4000 proteins were identified and 66 most significantly differentiated proteins were used for further bioinformatic analysis. According to the ingenuity pathway analysis (IPA), we found that proteins related to the inflammation response, immune regulation, lipid metabolism and NFκB signaling network were altered by CUMS. Moreover, four proteins closely associated with these processes, hemopexin, haptoglobin, cytochrome P450 2A4 (CYP2A4) and bile salt sulfotransferase 1 (SULT2A1), were validated by western blotting. In conclusion, we report, for the first time, the liver protein expression profile in the CUMS mouse model of depression. Our findings provide novel insight (liver-brain axis) into the multifaceted mechanisms of major depressive disorder.
Collapse
|