1
|
Qi X, He X, Peng Y, He X, Yang Q, Jiao K, Liu H. Roles of osteocalcin in the central nervous system. CNS Neurosci Ther 2024; 30:e70016. [PMID: 39252492 PMCID: PMC11386255 DOI: 10.1111/cns.70016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/04/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND Bone-derived protein osteocalcin, which has beneficial effects on brain function, may be a future research direction for neurological disorders. A growing body of evidence suggests a link between osteocalcin and neurological disorders, but the exact relationship is contradictory and unclear. SCOPE OF REVIEW The aim of this review is to summarize the current research on the interaction between osteocalcin and the central nervous system and to propose some speculative future research directions. MAJOR CONCLUSIONS In the normal central nervous system, osteocalcin is involved in neuronal structure, neuroprotection, and the regulation of cognition and anxiety. Studies on osteocalcin-related abnormalities in the central nervous system are divided into animal model studies and human studies, depending on the subject. In humans, the link between osteocalcin and brain function is inconsistent. These conflicting data may be due to methodological inconsistencies. By reviewing the related literature on osteocalcin, some comorbidities of the bone and nervous system and future research directions related to osteocalcin are proposed.
Collapse
Affiliation(s)
- Xiao‐Shan Qi
- Department of RadiologyAffiliated Hospital of Zunyi Medical University, Engineering Research Center of Intelligent Medical Imaging in Guizhou Higher Education lnstitutions, Medical Imaging Center of Guizhou ProvinceZunyiChina
- The First Clinical Medical CollegeZunyi Medical UniversityZunyiChina
| | - Xin He
- Department of RadiologyAffiliated Hospital of Zunyi Medical University, Engineering Research Center of Intelligent Medical Imaging in Guizhou Higher Education lnstitutions, Medical Imaging Center of Guizhou ProvinceZunyiChina
| | - Ying Peng
- Department of RadiologyAffiliated Hospital of Zunyi Medical University, Engineering Research Center of Intelligent Medical Imaging in Guizhou Higher Education lnstitutions, Medical Imaging Center of Guizhou ProvinceZunyiChina
| | - Xing‐Hong He
- Department of RadiologyAffiliated Hospital of Zunyi Medical University, Engineering Research Center of Intelligent Medical Imaging in Guizhou Higher Education lnstitutions, Medical Imaging Center of Guizhou ProvinceZunyiChina
| | - Qian‐Yu Yang
- The First Clinical Medical CollegeZunyi Medical UniversityZunyiChina
| | - Kai Jiao
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, School of StomatologyThe Fourth Military Medical UniversityXi‘anChina
| | - Heng Liu
- Department of RadiologyAffiliated Hospital of Zunyi Medical University, Engineering Research Center of Intelligent Medical Imaging in Guizhou Higher Education lnstitutions, Medical Imaging Center of Guizhou ProvinceZunyiChina
| |
Collapse
|
2
|
Qi W, Guan W. GPR56: A potential therapeutic target for neurological and psychiatric disorders. Biochem Pharmacol 2024; 226:116395. [PMID: 38942087 DOI: 10.1016/j.bcp.2024.116395] [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: 03/21/2024] [Revised: 06/17/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024]
Abstract
GPR56, also known as GPR56/ADGRG1, is a member of the ADGRG subgroup belonging to adhesion G protein-coupled receptors (aGPCRs). aGPCRs are the second largest subfamily of the GPCR superfamily, which is the largest family of membrane protein receptors in the human genome. Studies in recent years have demonstrated that GPR56 is integral to the normal development of the brain and functions as an important player in cortical development, suggesting that GPR56 is involved in many physiological processes. Indeed, aberrant expression of GPR56 has been implicated in multiple neurological and psychiatric disorders, including bilateral frontoparietal polymicrogyria (BFPP), depression and epilepsy. In a recent study, it was found that upregulated expression of GPR56 reduced depressive-like behaviours in an animal model of depression, indicating that GPR56 plays an important role in the antidepressant response. Given the link of GPR56 with the antidepressant response, the function of GPR56 has become a focus of research. Although GPR56 may be a potential target for the development of antidepressants, the underlying molecular mechanisms remain largely unknown. Therefore, in this review, we will summarize the latest findings of GPR56 function in neurological and psychiatric disorders (depression, epilepsy, autism, and BFPP) and emphasize the mechanisms of GPR56 in activation and signalling in those conditions. After reviewing several studies, attributing to its significant biological functions and exceptionally long extracellular N-terminus that interacts with multiple ligands, we draw a conclusion that GPR56 may serve as an important drug target for neuropsychological diseases.
Collapse
Affiliation(s)
- Wang Qi
- Department of Pharmacology, The First People's Hospital of Yancheng, China
| | - Wei Guan
- Department of Pharmacology, Pharmacy College, Nantong University, China.
| |
Collapse
|
3
|
Kim B, Hong S, Lee J, Kang S, Kim JS, Jung C, Shin T, Youn B, Moon C. Identifying candidate genes associated with hippocampal dysfunction in a hemiparkinsonian rat model by transcriptomic profiling. Anim Cells Syst (Seoul) 2024; 28:198-215. [PMID: 38693920 PMCID: PMC11062273 DOI: 10.1080/19768354.2024.2348671] [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: 01/29/2024] [Accepted: 03/12/2024] [Indexed: 05/03/2024] Open
Abstract
Parkinson's disease (PD) often results in hippocampal dysfunction, which leads to cognitive and emotional challenges and synaptic irregularities. This study attempted to assess behavioral anomalies and identify differentially expressed genes (DEGs) within the hippocampus of a hemiparkinsonian rat model to potentially uncover novel genetic candidates linked to hippocampal dysfunction. Striatal 6-hydroxydopamine (6-OHDA) infusions were performed unilaterally in the brains of adult SD rats, while dopaminergic impairments were verified in rats with 6-OHDA-lesioned striata. RNA sequencing and gene expression analysis unveiled 1018 DEGs in the ipsilateral rat hippocampus following 6-OHDA infusion: 631 genes exhibited upregulation, while 387 genes were downregulated (with FDR-adjusted p-value < 0.05 and absolute fold-change > 1.5). Gene ontology analysis of DEGs indicated that alterations in the hippocampi of 6-OHDA-lesioned rats were primarily associated with synaptic signaling, axon development, behavior, postsynaptic membrane, synaptic membrane, neurotransmitter receptor activity, and peptide receptor activity. The Kyoto Encyclopedia of Genes and Genomes analysis of DEGs demonstrated significant enrichment of the neuroactive ligand-receptor interaction, calcium signaling pathway, cAMP signaling pathway, axon guidance, and notch signaling pathway in rat hippocampi that had been subjected to striatal 6-OHDA infusion. STRING analysis confirmed a notable upregulation of eight hub genes (Notch3, Gng4, Itga3, Grin2d, Hgf, Fgf11, Htr3a, and Col6a2), along with a significant downregulation of two hub genes (Itga11 and Plp1), as validated by reverse transcription-quantitative polymerase chain reaction. This study provides a comprehensive transcriptomic profile of the hippocampi in a hemiparkinsonian rat model, thereby offering insights into the signaling pathways underlying hippocampal dysfunction.
Collapse
Affiliation(s)
- Bohye Kim
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, Republic of Korea
| | - Sungmoo Hong
- Department of Veterinary Anatomy, College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju, Republic of Korea
| | - Jeongmin Lee
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, Republic of Korea
| | - Sohi Kang
- Department of Anatomy and Convergence Medical Science, College of Medicine, Institute of Health Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Joong-Sun Kim
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, Republic of Korea
| | - Chaeyong Jung
- Department of Anatomy, Chonnam National University Medical School, Gwangju, Korea
| | - Taekyun Shin
- Department of Veterinary Anatomy, College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju, Republic of Korea
| | - BuHyun Youn
- Department of Biological Science, Pusan National University, Busan, Republic of Korea
| | - Changjong Moon
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, Republic of Korea
| |
Collapse
|
4
|
Dean B, Scarr E. Common changes in rat cortical gene expression after antidepressant drug treatment: Impacts on metabolism of polyamines, mRNA splicing, regulation of RAS by GAPs, neddylation and GPCR ligand binding. World J Biol Psychiatry 2024; 25:200-213. [PMID: 38349617 DOI: 10.1080/15622975.2024.2312475] [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: 11/07/2023] [Accepted: 01/27/2024] [Indexed: 03/02/2024]
Abstract
OBJECTIVES This study sought to identify pathways affected by rat cortical RNA that were changed after treatment with fluoxetine or imipramine. METHODS We measured levels of cortical RNA in male rats using GeneChip® Rat Exon 1.0 ST Array after treatment with vehicle (0.9% NaCl), fluoxetine (10 mg/kg/day) or imipramine (20 mg/kg/day) for 28 days. Levels of coding and non-coding RNA in vehicle treated rats were compared to those in treated rats using ANOVA in JMP Genomics 13 and the Panther Gene Ontology Classification System was used to identify pathways involving the changed RNAs. RESULTS 18,876 transcripts were detected; there were highly correlated changes in 1010 levels of RNA after both drug treatments that would principally affect the metabolism of polyamines, mRNA splicing, regulation of RAS by GAPs, neddylation and GPCR ligand binding. Using our previously published data, we compared changes in transcripts after treatment with antipsychotic and mood stabilising drugs. CONCLUSIONS Our study shows there are common, correlated, changes in coding and non-coding RNA in the rat cortex after treatment with fluoxetine or imipramine; we propose the pathways affected by these changes are involved in the therapeutic mechanisms of action of antidepressant drugs.
Collapse
Affiliation(s)
- Brian Dean
- The Molecular Psychiatry Laboratory, The Florey Institute for Neuroscience and Mental Health, Parkville, Australia
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Elizabeth Scarr
- The Department of Psychiatry, The University of Melbourne, Parkville, Australia
| |
Collapse
|
5
|
Shin D, Lee J, Kim Y, Park J, Shin D, Song Y, Joo EJ, Roh S, Lee KY, Oh S, Ahn YM, Rhee SJ, Kim Y. Evaluation of a Nondepleted Plasma Multiprotein-Based Model for Discriminating Psychiatric Disorders Using Multiple Reaction Monitoring-Mass Spectrometry: Proof-of-Concept Study. J Proteome Res 2024; 23:329-343. [PMID: 38063806 DOI: 10.1021/acs.jproteome.3c00580] [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] [Indexed: 01/06/2024]
Abstract
Psychiatric evaluation relies on subjective symptoms and behavioral observation, which sometimes leads to misdiagnosis. Despite previous efforts to utilize plasma proteins as objective markers, the depletion method is time-consuming. Therefore, this study aimed to enhance previous quantification methods and construct objective discriminative models for major psychiatric disorders using nondepleted plasma. Multiple reaction monitoring-mass spectrometry (MRM-MS) assays for quantifying 453 peptides in nondepleted plasma from 132 individuals [35 major depressive disorder (MDD), 47 bipolar disorder (BD), 23 schizophrenia (SCZ) patients, and 27 healthy controls (HC)] were developed. Pairwise discriminative models for MDD, BD, and SCZ, and a discriminative model between patients and HC were constructed by machine learning approaches. In addition, the proteins from nondepleted plasma-based discriminative models were compared with previously developed depleted plasma-based discriminative models. Discriminative models for MDD versus BD, BD versus SCZ, MDD versus SCZ, and patients versus HC were constructed with 11 to 13 proteins and showed reasonable performances (AUROC = 0.890-0.955). Most of the shared proteins between nondepleted and depleted plasma models had consistent directions of expression levels and were associated with neural signaling, inflammatory, and lipid metabolism pathways. These results suggest that multiprotein markers from nondepleted plasma have a potential role in psychiatric evaluation.
Collapse
Affiliation(s)
- Dongyoon Shin
- Proteomics Research Team, CHA Institute of Future Medicine, Seongnam 13520, Republic of Korea
| | - Jihyeon Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Yeongshin Kim
- Department of Life Science, General Graduate School, CHA University, Seongnam 13488, Republic of Korea
| | - Junho Park
- Proteomics Research Team, CHA Institute of Future Medicine, Seongnam 13520, Republic of Korea
- Department of Life Science, General Graduate School, CHA University, Seongnam 13488, Republic of Korea
| | - Daun Shin
- Department of Psychiatry, Korea University Anam Hospital, Seoul 02841, Republic of Korea
| | - Yoojin Song
- Department of Psychiatry, Kangwon National University Hospital, Chuncheon 24289, Republic of Korea
| | - Eun-Jeong Joo
- Department of Neuropsychiatry, School of Medicine, Eulji University, Daejeon 34824, Republic of Korea
- Department of Psychiatry, Uijeongbu Eulji Medical Center, Eulji University, Uijeongbu 11759, Republic of Korea
| | - Sungwon Roh
- Department of Psychiatry, Hanyang University Hospital and Hanyang University College of Medicine, Seoul 04763, Republic of Korea
| | - Kyu Young Lee
- Department of Neuropsychiatry, School of Medicine, Eulji University, Daejeon 34824, Republic of Korea
- Department of Psychiatry, Nowon Eulji University Hospital, Seoul 01830, Republic of Korea
| | - Sanghoon Oh
- Department of Neuropsychiatry, School of Medicine, Eulji University, Daejeon 34824, Republic of Korea
- Department of Psychiatry, Uijeongbu Eulji Medical Center, Eulji University, Uijeongbu 11759, Republic of Korea
| | - Yong Min Ahn
- Department of Psychiatry, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul 03080, Republic of Korea
- Institute of Human Behavioral Medicine, Seoul National University Medical Research Center, Seoul 03080, Republic of Korea
| | - Sang Jin Rhee
- Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Youngsoo Kim
- Proteomics Research Team, CHA Institute of Future Medicine, Seongnam 13520, Republic of Korea
- Department of Life Science, General Graduate School, CHA University, Seongnam 13488, Republic of Korea
| |
Collapse
|
6
|
Bolinger AA, Frazier A, La JH, Allen JA, Zhou J. Orphan G Protein-Coupled Receptor GPR37 as an Emerging Therapeutic Target. ACS Chem Neurosci 2023; 14:3318-3334. [PMID: 37676000 PMCID: PMC11144446 DOI: 10.1021/acschemneuro.3c00479] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are successful druggable targets, making up around 35% of all FDA-approved medications. However, a large number of receptors remain orphaned, with no known endogenous ligand, representing a challenging but untapped area to discover new therapeutic targets. Among orphan GPCRs (oGPCRs) of interest, G protein-coupled receptor 37 (GPR37) is highly expressed in the central nervous system (CNS), particularly in the spinal cord and oligodendrocytes. While its cellular signaling mechanisms and endogenous receptor ligands remain elusive, GPR37 has been implicated in several important neurological conditions, including Parkinson's disease (PD), inflammation, pain, autism, and brain tumors. GPR37 structure, signaling, emerging physiology, and pharmacology are reviewed while integrating a discussion on potential therapeutic indications and opportunities.
Collapse
Affiliation(s)
- Andrew A. Bolinger
- Department of Pharmacology and Toxicology, Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Andrew Frazier
- Department of Pharmacology and Toxicology, Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Jun-Ho La
- Department of Neurobiology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - John A. Allen
- Department of Pharmacology and Toxicology, Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Jia Zhou
- Department of Pharmacology and Toxicology, Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, Texas 77555, United States
| |
Collapse
|
7
|
He L, Xu Z, Niu X, Li R, Wang F, You Y, Gao J, Zhao L, Shah KM, Fan J, Liu M, Luo J. GPRC5B protects osteoarthritis by regulation of autophagy signaling. Acta Pharm Sin B 2023; 13:2976-2989. [PMID: 37521864 PMCID: PMC10372909 DOI: 10.1016/j.apsb.2023.05.014] [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: 11/30/2022] [Revised: 01/17/2023] [Accepted: 03/14/2023] [Indexed: 08/01/2023] Open
Abstract
Osteoarthritis (OA) is one of the most common chronic diseases in the world. However, current treatment modalities mainly relieve pain and inhibit cartilage degradation, but do not promote cartilage regeneration. In this study, we show that G protein-coupled receptor class C group 5 member B (GPRC5B), an orphan G-protein-couple receptor, not only inhibits cartilage degradation, but also increases cartilage regeneration and thereby is protective against OA. We observed that Gprc5b deficient chondrocytes had an upregulation of cartilage catabolic gene expression, along with downregulation of anabolic genes in vitro. Furthermore, mice deficient in Gprc5b displayed a more severe OA phenotype in the destabilization of the medial meniscus (DMM) induced OA mouse model, with upregulation of cartilage catabolic factors and downregulation of anabolic factors, consistent with our in vitro findings. Overexpression of Gprc5b by lentiviral vectors alleviated the cartilage degeneration in DMM-induced OA mouse model by inhibiting cartilage degradation and promoting regeneration. We also assessed the molecular mechanisms downstream of Gprc5b that may mediate these observed effects and identify the role of protein kinase B (AKT)-mammalian target of rapamycin (mTOR)-autophagy signaling pathway. Thus, we demonstrate an integral role of GPRC5B in OA pathogenesis, and activation of GPRC5B has the potential in preventing the progression of OA.
Collapse
Affiliation(s)
- Liang He
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai 201613, China
| | - Ziwei Xu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xin Niu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Rong Li
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai 201613, China
| | - Fanhua Wang
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai 201613, China
| | - Yu You
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jingduo Gao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Lei Zhao
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai 201613, China
| | - Karan M. Shah
- Department of Oncology and Metabolism, the Medical School, the University of Sheffield, Sheffield S10 2TN, UK
| | - Jian Fan
- Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jian Luo
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai 201613, China
| |
Collapse
|
8
|
Yu H, Ni P, Zhao L, Tian Y, Li M, Li X, Wei W, Wei J, Deng W, Du X, Wang Q, Guo W, Ma X, Coid J, Li T. Decreased plasma neuropeptides in first-episode schizophrenia, bipolar disorder, major depressive disorder: associations with clinical symptoms and cognitive function. Front Psychiatry 2023; 14:1180720. [PMID: 37275985 PMCID: PMC10235770 DOI: 10.3389/fpsyt.2023.1180720] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/02/2023] [Indexed: 06/07/2023] Open
Abstract
Background There is an urgent need to identify differentiating and disease-monitoring biomarkers of schizophrenia, bipolar disorders (BD), and major depressive disorders (MDD) to improve treatment and management. Methods We recruited 54 first-episode schizophrenia (FES) patients, 52 BD patients, 35 MDD patients, and 54 healthy controls from inpatient and outpatient clinics. α-Melanocyte Stimulating Hormone (α-MSH), β-endorphin, neurotensin, orexin-A, oxytocin, and substance P were investigated using quantitative multiplex assay method. Psychotic symptoms were measured using the Brief Psychiatric Rating Scale (BPRS) and Positive and Negative Syndrome Scale (PANSS), manic symptoms using the Young Mania Rating Scale (YMRS), and depressive symptoms using 17 item-Hamilton Depression Rating Scale (HAMD). We additionally measured cognitive function by using a battery of tests given to all participants. Results α-MSH, neurotensin, orexin-A, oxytocin, and substance P were decreased in the three patient groups compared with controls. Neurotensin outperformed all biomarkers in differentiating patient groups from controls. There were no significant differences for 6 neuropeptides in their ability to differentiate between the three patient groups. Higher neurotensin was associated with better executive function across the entire sample. Lower oxytocin and higher substance p were associated with more psychotic symptoms in FES and BD groups. β-endorphin was associated with early morning wakening symptom in all three patient groups. Conclusion Our research shows decreased circulating neuropeptides have the potential to differentiate severe mental illnesses from controls. These neuropeptides are promising treatment targets for improving clinical symptoms and cognitive function in FES, BD, and MDD.
Collapse
Affiliation(s)
- Hua Yu
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Peiyan Ni
- The Psychiatric Laboratory and Mental Health Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Liansheng Zhao
- The Psychiatric Laboratory and Mental Health Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yang Tian
- The Psychiatric Laboratory and Mental Health Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Mingli Li
- The Psychiatric Laboratory and Mental Health Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaojing Li
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wei Wei
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jinxue Wei
- The Psychiatric Laboratory and Mental Health Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wei Deng
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiangdong Du
- Suzhou Psychiatry Hospital, Affiliated Guangji Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qiang Wang
- The Psychiatric Laboratory and Mental Health Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wanjun Guo
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaohong Ma
- The Psychiatric Laboratory and Mental Health Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jeremy Coid
- The Psychiatric Laboratory and Mental Health Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tao Li
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
9
|
Chen J, Long MD, Sribenja S, Ma SJ, Yan L, Hu Q, Liu S, Khoury T, Hong CC, Bandera E, Singh AK, Repasky EA, Bouchard EG, Higgins M, Ambrosone CB, Yao S. An epigenome-wide analysis of socioeconomic position and tumor DNA methylation in breast cancer patients. Clin Epigenetics 2023; 15:68. [PMID: 37101222 PMCID: PMC10131486 DOI: 10.1186/s13148-023-01470-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 03/21/2023] [Indexed: 04/28/2023] Open
Abstract
BACKGROUND Disadvantaged socioeconomic position (SEP), including lower educational attainment and household income, may influence cancer risk and outcomes. We hypothesized that DNA methylation could function as an intermediary epigenetic mechanism that internalizes and reflects the biological impact of SEP. METHODS Based on tumor DNA methylation data from the Illumina 450 K array from 694 breast cancer patients in the Women's Circle of Health Study, we conducted an epigenome-wide analysis in relation to educational attainment and household income. Functional impact of the identified CpG sites was explored in silico using data from publicly available databases. RESULTS We identified 25 CpG sites associated with household income at an array-wide significance level, but none with educational attainment. Two of the top CpG sites, cg00452016 and cg01667837, were in promoter regions of NNT and GPR37, respectively, with multiple epigenetic regulatory features identified in each region. NNT is involved in β-adrenergic stress signaling and inflammatory responses, whereas GPR37 is involved in neurological and immune responses. For both loci, gene expression was inversely correlated to the levels of DNA methylation. The associations were consistent between Black and White women and did not differ by tumor estrogen receptor (ER) status. CONCLUSIONS In a large breast cancer patient population, we discovered evidence of the significant biological impact of household income on the tumor DNA methylome, including genes in the β-adrenergic stress and immune response pathways. Our findings support biological effects of socioeconomic status on tumor tissues, which might be relevant to cancer development and progression.
Collapse
Affiliation(s)
- Jianhong Chen
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Mark D Long
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Sirinapa Sribenja
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Sung Jun Ma
- Department of Radiation Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Li Yan
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Qiang Hu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Thaer Khoury
- Department of Pathology and Laboratory Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Chi-Chen Hong
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Elisa Bandera
- Cancer Prevention and Control Program, Rutgers Cancer Institute of New Jersey, The State University of New Jersey, New Brunswick, NJ, USA
| | - Anurag K Singh
- Department of Radiation Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Elizabeth A Repasky
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Elizabeth G Bouchard
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Michael Higgins
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Christine B Ambrosone
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Song Yao
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
| |
Collapse
|
10
|
Impaired Aversive Memory Formation in GPR37L1KO Mice. Int J Mol Sci 2022; 23:ijms232214290. [PMID: 36430766 PMCID: PMC9696904 DOI: 10.3390/ijms232214290] [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: 09/27/2022] [Revised: 11/12/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
GPR37L1 is an orphan G-protein-coupled receptor, which is implicated in neurological disorders, but its normal physiological role is poorly understood. Its close homologue, GPR37, is implicated in Parkinson's disease and affective disorders. In this study, we set out to characterize adult and middle-aged global GPR37L1 knock-out (KO) mice regarding emotional behaviors. Our results showed that GPR37L1KO animals, except adult GPR37L1KO males, exhibited impaired retention of aversive memory formation as assessed by the shorter retention latency in a passive avoidance task. Interestingly, the viral-mediated deletion of GPR37L1 in conditional knockout mice in the hippocampus of middle-aged mice also showed impaired retention in passive avoidance tasks, similar to what was observed in global GPR37L1KO mice, suggesting that hippocampal GPR37L1 is involved in aversive learning processes. We also observed that middle-aged GPR37L1KO male and female mice exhibited a higher body weight than their wild-type counterparts. Adult and middle-aged GPR37L1KO female mice exhibited a reduced level of serum corticosterone and middle-aged GPR37L1KO females showed a reduced level of epinephrine in the dorsal hippocampus in the aftermath of passive avoidance task, with no such effects observed in GPR37L1KO male mice, suggesting that lack of GPR37L1 influences behavior and biochemical readouts in age- and sex-specific manners.
Collapse
|
11
|
Su T, Guan Q, Cheng H, Zhu Z, Jiang C, Guo P, Tai Y, Sun H, Wang M, Wei W, Wang Q. Functions of G protein-coupled receptor 56 in health and disease. Acta Physiol (Oxf) 2022; 236:e13866. [PMID: 35959520 DOI: 10.1111/apha.13866] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 08/05/2022] [Accepted: 08/08/2022] [Indexed: 01/29/2023]
Abstract
Human G protein-coupled receptor 56 (GPR56) is encoded by gene ADGRG1 from chromosome 16q21 and is homologously encoded in mice, at chromosome 8. Both 687 and 693 splice forms are present in humans and mice. GPR56 has a 381 amino acid-long N-terminal extracellular segment and a GPCR proteolysis site upstream from the first transmembrane domain. GPR56 is mainly expressed in the heart, brain, thyroid, platelets, and peripheral blood mononuclear cells. Accumulating evidence indicates that GPR56 promotes the formation of myelin sheaths and the development of oligodendrocytes in the cerebral cortex of the central nervous system. Moreover, GPR56 contributes to the development and differentiation of hematopoietic stem cells, induces adipogenesis, and regulates the function of immune cells. The lack of GPR56 leads to nervous system dysfunction, platelet disorders, and infertility. Abnormal expression of GPR56 is related to the malignant transformation and tumor metastasis of several cancers including melanoma, neuroglioma, and gastrointestinal cancer. Metabolic disorders and cardiovascular diseases are also associated with dysregulation of GPR56 expression, and GPR56 is involved in the pharmacological resistance to some antidepressant and cancer drug treatments. In this review, the molecular structure, expression profile, and signal transduction of GPR56 are introduced, and physiological and pathological functions of GRP56 are comprehensively summarized. Attributing to its significant biological functions and its long N-terminal extracellular region that interacts with multiple ligands, GPR56 is becoming an attractive therapeutic target in treating neurological and hematopoietic diseases.
Collapse
Affiliation(s)
- Tiantian Su
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui Province, China
| | - Qiuyun Guan
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui Province, China
| | - Huijuan Cheng
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui Province, China
| | - Zhenduo Zhu
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui Province, China
| | - Chunru Jiang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui Province, China
| | - Paipai Guo
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui Province, China
| | - Yu Tai
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui Province, China
| | - Hanfei Sun
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui Province, China
| | - Manman Wang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui Province, China
| | - Wei Wei
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui Province, China
| | - Qingtong Wang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui Province, China
| |
Collapse
|
12
|
Xie X, Cai X, Zhou F, Li Y, Liu Q, Cai L, Zhu W, Wei J, Jin C, Liu Z, Jiang C, Zhao H, Yang L, Zhao C, Huang X. GPR37 promotes cancer growth by binding to CDK6 and represents a new theranostic target in lung adenocarcinoma. Pharmacol Res 2022; 183:106389. [PMID: 35934193 DOI: 10.1016/j.phrs.2022.106389] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 08/02/2022] [Accepted: 08/03/2022] [Indexed: 01/11/2023]
Abstract
Lung adenocarcinoma (LUAD) is associated with poor prognosis. Identifying novel cancer targets and helpful therapeutic strategies remains a serious clinical challenge. This study detected differentially expressed genes in The Cancer Genome Atlas (TCGA) LUAD data collection. We also identified a predictive DNA biomarker, G protein-coupled receptor 37 (GPR37), which was verified as a prognostic biomarker with a critical role in tumor progression. In human LUAD specimens and microarray analyses, we determined that GPR37 was significantly upregulated and associated with a poor prognosis. GPR37 downregulation markedly inhibited the proliferation and migration of LUAD both in vitro and in vivo. Mechanistically, GPR37 could bind to CDK6, thereby facilitating tumor progression in LUAD by inducing cell cycle arrest at the G1 phase. GPR37 also facilitates tumorigenesis in xenograft tumors in vivo. High-throughput screening for GPR37-targeted drugs was performed using the Natural Products Library, which revealed the potential of Hypocrellin B to inhibit GPR37 and cell growth in LUAD. We demonstrated that Hypocrellin B suppressed LUAD cell proliferation and migration both in vitro and in vivo via GPR37 inhibition. Collectively, our findings reveal the role of GPR37 in LUAD progression and migration and the potential of GPR37 as a target for the treatment of LUAD. Thus, the specific inhibition of GPR37 by the natural product Hypocrellin B may possess the potential for the treatment of LUAD.
Collapse
Affiliation(s)
- Xiaona Xie
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, China; Department of Medical Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xueding Cai
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, China
| | - Feng Zhou
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, China
| | - Yaozhe Li
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, China
| | - Qianzi Liu
- The Institute of Life Sciences, Wenzhou University, University Town, Wenzhou, Zhejiang 325035, China
| | - Luqiong Cai
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, China
| | - Wenjing Zhu
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, China
| | - Jinqiu Wei
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, China
| | - Chenying Jin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Zitian Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chunhui Jiang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Haiyang Zhao
- The Institute of Life Sciences, Wenzhou University, University Town, Wenzhou, Zhejiang 325035, China
| | - Lehe Yang
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, China.
| | - Chengguang Zhao
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China.
| | - Xiaoying Huang
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
13
|
Zakaria FH, Samhani I, Mustafa MZ, Shafin N. Pathophysiology of Depression: Stingless Bee Honey Promising as an Antidepressant. Molecules 2022; 27:molecules27165091. [PMID: 36014336 PMCID: PMC9416360 DOI: 10.3390/molecules27165091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 07/30/2022] [Accepted: 08/06/2022] [Indexed: 11/16/2022] Open
Abstract
Depression is a debilitating psychiatric disorder impacting an individual’s quality of life. It is the most prevalent mental illness across all age categories, incurring huge socio-economic impacts. Most depression treatments currently focus on the elevation of neurotransmitters according to the monoamine hypothesis. Conventional treatments include tricyclic antidepressants (TCAs), norepinephrine–dopamine reuptake inhibitors (NDRIs), monoamine oxidase inhibitors (MAOIs), and serotonin reuptake inhibitors (SSRIs). Despite numerous pharmacological strategies utilising conventional drugs, the discovery of alternative medicines from natural products is a must for safer and beneficial brain supplement. About 30% of patients have been reported to show resistance to drug treatments coupled with functional impairment, poor quality of life, and suicidal ideation with a high relapse rate. Hence, there is an urgency for novel discoveries of safer and highly effective depression treatments. Stingless bee honey (SBH) has been proven to contain a high level of antioxidants compared to other types of honey. This is a comprehensive review of the potential use of SBH as a new candidate for antidepressants from the perspective of the monoamine, inflammatory and neurotrophin hypotheses.
Collapse
Affiliation(s)
- Fatin Haniza Zakaria
- Department of Neuroscience, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kota Bharu 16150, Malaysia
| | - Ismail Samhani
- Faculty of Medicine, Universiti Sultan Zainal Abidin (UniSZA), Medical Campus, Jalan Sultan Mahmud, Kuala Terengganu 20400, Malaysia
| | - Mohd Zulkifli Mustafa
- Department of Neuroscience, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kota Bharu 16150, Malaysia
- Correspondence: (M.Z.M.); (N.S.); Tel.: +609-7673000 (M.Z.M. & N.S.)
| | - Nazlahshaniza Shafin
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kota Bharu 16150, Malaysia
- Correspondence: (M.Z.M.); (N.S.); Tel.: +609-7673000 (M.Z.M. & N.S.)
| |
Collapse
|
14
|
Integrating variant functional annotation scores have varied abilities to improve power of genome-wide association studies. Sci Rep 2022; 12:10720. [PMID: 35750789 PMCID: PMC9232605 DOI: 10.1038/s41598-022-14924-1] [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: 02/27/2022] [Accepted: 06/15/2022] [Indexed: 11/12/2022] Open
Abstract
Functional annotations have the potential to increase power of genome-wide association studies (GWAS) by prioritizing variants according to their biological function, but this potential has not been well studied. We comprehensively evaluated all 1132 traits in the UK Biobank whose SNP-heritability estimates were given “medium” or “high” labels by Neale’s lab. For each trait, we integrated GWAS summary statistics of close to 8 million common variants (minor allele frequency \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$>1\%$$\end{document}>1%) with either their 75 individual functional scores or their meta-scores, using three different data-integration methods. Overall, the number of new genome-wide significant findings after data-integration increases as a trait SNP-heritability estimate increases. However, there is a trade-off between new findings and loss of baseline GWAS findings, resulting in similar total numbers of significant findings between using GWAS alone and integrating GWAS with functional scores, across all 1132 traits analyzed and all three data-integration methods considered. Our findings suggest that, even with the current biobank-level sample size, more informative functional scores and/or new data-integration methods are needed to further improve the power of GWAS of common variants. For example, studying variants in coding sequence and obtaining cell-type-specific scores are potential future directions.
Collapse
|
15
|
Fu W, Franchini L, Orlandi C. Comprehensive Spatial Profile of the Orphan G Protein Coupled Receptor GPRC5B Expression in Mouse Brain. Front Neurosci 2022; 16:891544. [PMID: 35812210 PMCID: PMC9259939 DOI: 10.3389/fnins.2022.891544] [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: 03/07/2022] [Accepted: 06/07/2022] [Indexed: 11/13/2022] Open
Abstract
Orphan G Protein Coupled Receptors (GPCRs) are GPCRs whose endogenous ligands are unknown or still debated. Due to the lack of pharmacological modulators, the physiological function of orphan GPCRs is understudied. However, relevant physiological roles associated with orphan GPCRs have been revealed by analysis of animal models and genome wide association studies illuminating an untapped potential for drug discovery. G Protein Coupled Receptor class C Group 5 Member B (GPRC5B) is among the most expressed GPCRs in the central nervous system. Thus, the expression profiling of GPRC5B is an essential step toward understanding GPRC5B function in health and disease. In this study, we generated new GPRC5B polyclonal antibodies and investigated the expression levels of GPRC5B across different organs and brain regions. We identified high levels of GPRC5B glycosylation both in transfected cells and in mouse brain. Moreover, in situ hybridization imaging analysis indicated that Gprc5b was expressed at the highest level in olfactory bulb, hippocampus, cerebellum, and pons. To dissect expression within various neuronal populations, we conducted a comprehensive spatial profiling of Gprc5b across excitatory and inhibitory neuronal types in medial prefrontal cortex, motor cortex, hippocampal regions, hypothalamus, and cerebellum. Overall, we discovered that GABAergic neurons displayed higher Gprc5b expression levels than glutamatergic neurons in most of the analyzed regions with the important exception of the hippocampal dentate gyrus. Overall, the expression analysis of GPRC5B in mouse brain will guide functional studies ultimately positioning GPRC5B in pathophysiological mechanisms and drug discovery.
Collapse
|
16
|
Osteocalcin Alleviates Lipopolysaccharide-Induced Acute Inflammation via Activation of GPR37 in Macrophages. Biomedicines 2022; 10:biomedicines10051006. [PMID: 35625743 PMCID: PMC9138386 DOI: 10.3390/biomedicines10051006] [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: 03/25/2022] [Revised: 04/24/2022] [Accepted: 04/24/2022] [Indexed: 11/16/2022] Open
Abstract
The G protein-coupled receptor 37 (GPR37) has been reported to be expressed in macrophages and the activation of GPR37 by its ligand/agonist, and it can regulate macrophage-associated functions and inflammatory responses. Since our previous work identified that osteocalcin (OCN) acts as an endogenous ligand for GPR37 and can elicit various intracellular signals by interacting with GPR37, we thus hypothesized that OCN may also play a functional role in macrophage through the activation of GPR37. To verify the hypothesis, we conducted a series of in vivo and in vitro studies in lipopolysaccharide (LPS)-challenged mice and primary cultured macrophages. Our results reveal that the OCN gene deletion (OCN−/−) and wild type (WT) mice showed comparable death rates and inflammatory cytokines productions in response to a lethal dose of LPS exposure. However, the detrimental effects caused by LPS were significantly ameliorated by exogenous OCN treatments in both WT and OCN−/− mice. Notably, the protective effects of OCN were absent in GPR37−/− mice. In coordination with the in vivo results, our in vitro studies further illustrated that OCN triggered intracellular responses via GPR37 in peritoneal macrophages by regulating the release of inflammatory factors and macrophage phagocytic function. Finally, we exhibited that the adoptive transfer of OCN-treated macrophages from WT mice significantly inhibits the release of pro-inflammatory cytokines in GPR37−/− mice exposed to LPS. Taken together, these findings suggest a protective role of OCN against LPS-caused acute inflammation, by the activation of GPR37 in macrophages, and provide a potential application of the activation of the OCN/GPR37 regulatory axis as a therapeutic strategy for inflammatory diseases.
Collapse
|
17
|
The role of orphan receptor GPR139 in neuropsychiatric behavior. Neuropsychopharmacology 2022; 47:902-913. [PMID: 33479510 PMCID: PMC8882194 DOI: 10.1038/s41386-021-00962-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 01/01/2021] [Accepted: 01/05/2021] [Indexed: 01/30/2023]
Abstract
Orphan G protein Coupled Receptors (GPCRs) present attractive targets both for understanding neuropsychiatric diseases and for development of novel therapeutics. GPR139 is an orphan GPCR expressed in select brain circuits involved in controlling movement, motivation and reward. It has been linked to the opioid and dopamine neuromodulatory systems; however, its role in animal behavior and neuropsychiatric processes is poorly understood. Here we present a comprehensive behavioral characterization of a mouse model with a GPR139 null mutation. We show that loss of GPR139 in mice results in delayed onset hyperactivity and prominent neuropsychiatric manifestations including elevated stereotypy, increased anxiety-related traits, delayed acquisition of operant responsiveness, disruption of cued fear conditioning and social interaction deficits. Furthermore, mice lacking GPR139 exhibited complete loss of pre-pulse inhibition and developed spontaneous 'hallucinogenic' head-twitches, altogether suggesting schizophrenia-like symptomatology. Remarkably, a number of these behavioral deficits could be rescued by the administration of μ-opioid and D2 dopamine receptor (D2R) antagonists: naltrexone and haloperidol, respectively, suggesting that loss of neuropsychiatric manifestations in mice lacking GPR139 are driven by opioidergic and dopaminergic hyper-functionality. The inhibitory influence of GPR139 on D2R signaling was confirmed in cell-based functional assays. These observations define the role of GPR139 in controlling behavior and implicate in vivo actions of this receptor in the neuropsychiatric process with schizophrenia-like pathology.
Collapse
|
18
|
Qin Y, Havulinna AS, Liu Y, Jousilahti P, Ritchie SC, Tokolyi A, Sanders JG, Valsta L, Brożyńska M, Zhu Q, Tripathi A, Vázquez-Baeza Y, Loomba R, Cheng S, Jain M, Niiranen T, Lahti L, Knight R, Salomaa V, Inouye M, Méric G. Combined effects of host genetics and diet on human gut microbiota and incident disease in a single population cohort. Nat Genet 2022; 54:134-142. [PMID: 35115689 PMCID: PMC9883041 DOI: 10.1038/s41588-021-00991-z] [Citation(s) in RCA: 183] [Impact Index Per Article: 91.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 11/19/2021] [Indexed: 01/31/2023]
Abstract
Human genetic variation affects the gut microbiota through a complex combination of environmental and host factors. Here we characterize genetic variations associated with microbial abundances in a single large-scale population-based cohort of 5,959 genotyped individuals with matched gut microbial metagenomes, and dietary and health records (prevalent and follow-up). We identified 567 independent SNP-taxon associations. Variants at the LCT locus associated with Bifidobacterium and other taxa, but they differed according to dairy intake. Furthermore, levels of Faecalicatena lactaris associated with ABO, and suggested preferential utilization of secreted blood antigens as energy source in the gut. Enterococcus faecalis levels associated with variants in the MED13L locus, which has been linked to colorectal cancer. Mendelian randomization analysis indicated a potential causal effect of Morganella on major depressive disorder, consistent with observational incident disease analysis. Overall, we identify and characterize the intricate nature of host-microbiota interactions and their association with disease.
Collapse
Affiliation(s)
- Youwen Qin
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- School of BioSciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - Aki S Havulinna
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Institute for Molecular Medicine Finland, FIMM-HiLIFE, Helsinki, Finland
| | - Yang Liu
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Clinical Pathology, Melbourne Medical School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Pekka Jousilahti
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Scott C Ritchie
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
| | - Alex Tokolyi
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Jon G Sanders
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, NY, USA
- Cornell Institute for Host-Microbe Interaction and Disease, Cornell University, Ithaca, NY, USA
| | - Liisa Valsta
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Marta Brożyńska
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Qiyun Zhu
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Anupriya Tripathi
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
- Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Yoshiki Vázquez-Baeza
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Department of Computer Science & Engineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, USA
| | - Rohit Loomba
- NAFLD Research Center, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Susan Cheng
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Mohit Jain
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
| | - Teemu Niiranen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Leo Lahti
- Department of Computing, University of Turku, Turku, Finland
| | - Rob Knight
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Department of Computer Science & Engineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, USA
| | - Veikko Salomaa
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Michael Inouye
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.
- School of BioSciences, The University of Melbourne, Melbourne, Victoria, Australia.
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK.
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- Health Data Research UK Cambridge, Wellcome Genome Campus & University of Cambridge, Cambridge, UK.
- The Alan Turing Institute, London, UK.
| | - Guillaume Méric
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.
- Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
19
|
Veenit V, Zhang X, Ambrosini A, Sousa V, Svenningsson P. The Effect of Early Life Stress on Emotional Behaviors in GPR37KO Mice. Int J Mol Sci 2021; 23:410. [PMID: 35008836 PMCID: PMC8745300 DOI: 10.3390/ijms23010410] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 12/23/2021] [Accepted: 12/28/2021] [Indexed: 12/14/2022] Open
Abstract
GPR37 is an orphan G-protein-coupled receptor, a substrate of parkin which is linked to Parkinson's disease (PD) and affective disorders. In this study, we sought to address the effects of early life stress (ELS) by employing the paradigm of limited nesting material on emotional behaviors in adult GPR37 knockout (KO) mice. Our results showed that, while there was an adverse effect of ELS on various domains of emotional behaviors in wild type (WT) mice in a sex specific manner (anxiety in females, depression and context-dependent fear memory in males), GPR37KO mice subjected to ELS exhibited less deteriorated emotional behaviors. GPR37KO female mice under ELS conditions displayed reduced anxiety compared to WT mice. This was paralleled by lower plasma corticosterone in GPR37KO females and a lower increase in P-T286-CaMKII by ELS in the amygdala. GPR37KO male mice, under ELS conditions, showed better retention of hippocampal-dependent emotional processing in the passive avoidance behavioral task. GPR37KO male mice showed increased immobility in the forced swim task and increased P-T286-CaMKII in the ventral hippocampus under baseline conditions. Taken together, our data showed overall long-term effects of ELS-deleterious or beneficial depending on the genotype, sex of the mice and the emotional context.
Collapse
Affiliation(s)
- Vandana Veenit
- Neuro Svenningsson, Department of Clinical Neuroscience, Karolinska Institutet, 171 76 Stockholm, Sweden; (X.Z.); (A.A.); (V.S.)
| | | | | | | | - Per Svenningsson
- Neuro Svenningsson, Department of Clinical Neuroscience, Karolinska Institutet, 171 76 Stockholm, Sweden; (X.Z.); (A.A.); (V.S.)
| |
Collapse
|
20
|
Monfared RV, Alhassen W, Truong TM, Gonzales MAM, Vachirakorntong V, Chen S, Baldi P, Civelli O, Alachkar A. Transcriptome Profiling of Dysregulated GPCRs Reveals Overlapping Patterns across Psychiatric Disorders and Age-Disease Interactions. Cells 2021; 10:2967. [PMID: 34831190 PMCID: PMC8616384 DOI: 10.3390/cells10112967] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/24/2021] [Accepted: 10/25/2021] [Indexed: 12/29/2022] Open
Abstract
G-protein-coupled receptors (GPCRs) play an integral role in the neurobiology of psychiatric disorders. Almost all neurotransmitters involved in psychiatric disorders act through GPCRs, and GPCRs are the most common targets of therapeutic drugs currently used in the treatment of psychiatric disorders. However, the roles of GPCRs in the etiology and pathophysiology of psychiatric disorders are not fully understood. Using publically available datasets, we performed a comprehensive analysis of the transcriptomic signatures of G-protein-linked signaling across the major psychiatric disorders: autism spectrum disorder (ASD), schizophrenia (SCZ), bipolar disorder (BP), and major depressive disorder (MDD). We also used the BrainSpan transcriptomic dataset of the developing human brain to examine whether GPCRs that exhibit chronological age-associated expressions have a higher tendency to be dysregulated in psychiatric disorders than age-independent GPCRs. We found that most GPCR genes were differentially expressed in the four disorders and that the GPCR superfamily as a gene cluster was overrepresented in the four disorders. We also identified a greater amplitude of gene expression changes in GPCRs than other gene families in the four psychiatric disorders. Further, dysregulated GPCRs overlapped across the four psychiatric disorders, with SCZ exhibiting the highest overlap with the three other disorders. Finally, the results revealed a greater tendency of age-associated GPCRs to be dysregulated in ASD than random GPCRs. Our results substantiate the central role of GPCR signaling pathways in the etiology and pathophysiology of psychiatric disorders. Furthermore, our study suggests that common GPCRs' signaling may mediate distinct phenotypic presentations across psychiatric disorders. Consequently, targeting these GPCRs could serve as a common therapeutic strategy to treat specific clinical symptoms across psychiatric disorders.
Collapse
Affiliation(s)
- Roudabeh Vakil Monfared
- Department of Pharmaceutical Sciences, School of Pharmacy, University of California Irvine, Irvine, CA 92697, USA; (R.V.M.); (W.A.); (T.M.T.); (M.A.M.G.); (V.V.); (O.C.)
| | - Wedad Alhassen
- Department of Pharmaceutical Sciences, School of Pharmacy, University of California Irvine, Irvine, CA 92697, USA; (R.V.M.); (W.A.); (T.M.T.); (M.A.M.G.); (V.V.); (O.C.)
| | - Tri Minh Truong
- Department of Pharmaceutical Sciences, School of Pharmacy, University of California Irvine, Irvine, CA 92697, USA; (R.V.M.); (W.A.); (T.M.T.); (M.A.M.G.); (V.V.); (O.C.)
| | - Michael Angelo Maglalang Gonzales
- Department of Pharmaceutical Sciences, School of Pharmacy, University of California Irvine, Irvine, CA 92697, USA; (R.V.M.); (W.A.); (T.M.T.); (M.A.M.G.); (V.V.); (O.C.)
| | - Vincent Vachirakorntong
- Department of Pharmaceutical Sciences, School of Pharmacy, University of California Irvine, Irvine, CA 92697, USA; (R.V.M.); (W.A.); (T.M.T.); (M.A.M.G.); (V.V.); (O.C.)
| | - Siwei Chen
- Department of Computer Science, School of Information and Computer Sciences, University of California Irvine, Irvine, CA 92697, USA; (S.C.); (P.B.)
- Institute for Genomics and Bioinformatics, School of Information and Computer Sciences, University of California Irvine, Irvine, CA 92697, USA
| | - Pierre Baldi
- Department of Computer Science, School of Information and Computer Sciences, University of California Irvine, Irvine, CA 92697, USA; (S.C.); (P.B.)
- Institute for Genomics and Bioinformatics, School of Information and Computer Sciences, University of California Irvine, Irvine, CA 92697, USA
| | - Olivier Civelli
- Department of Pharmaceutical Sciences, School of Pharmacy, University of California Irvine, Irvine, CA 92697, USA; (R.V.M.); (W.A.); (T.M.T.); (M.A.M.G.); (V.V.); (O.C.)
| | - Amal Alachkar
- Department of Pharmaceutical Sciences, School of Pharmacy, University of California Irvine, Irvine, CA 92697, USA; (R.V.M.); (W.A.); (T.M.T.); (M.A.M.G.); (V.V.); (O.C.)
- Institute for Genomics and Bioinformatics, School of Information and Computer Sciences, University of California Irvine, Irvine, CA 92697, USA
- Center for the Neurobiology of Learning and Memory, University of California Irvine, Irvine, CA 92697, USA
| |
Collapse
|
21
|
Mattila SO, Tuhkanen HE, Lackman JJ, Konzack A, Morató X, Argerich J, Saftig P, Ciruela F, Petäjä-Repo UE. GPR37 is processed in the N-terminal ectodomain by ADAM10 and furin. FASEB J 2021; 35:e21654. [PMID: 34042202 DOI: 10.1096/fj.202002385rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 04/12/2021] [Accepted: 04/26/2021] [Indexed: 11/11/2022]
Abstract
GPR37 is an orphan G protein-coupled receptor (GPCR) implicated in several neurological diseases and important physiological pathways in the brain. We previously reported that its long N-terminal ectodomain undergoes constitutive metalloprotease-mediated cleavage and shedding, which have been rarely described for class A GPCRs. Here, we demonstrate that the protease that cleaves GPR37 at Glu167↓Gln168 is a disintegrin and metalloprotease 10 (ADAM10). This was achieved by employing selective inhibition, RNAi-mediated downregulation, and genetic depletion of ADAM10 in cultured cells as well as in vitro cleavage of the purified receptor with recombinant ADAM10. In addition, the cleavage was restored in ADAM10 knockout cells by overexpression of the wild type but not the inactive mutant ADAM10. Finally, postnatal conditional depletion of ADAM10 in mouse neuronal cells was found to reduce cleavage of the endogenous receptor in the brain cortex and hippocampus, confirming the physiological relevance of ADAM10 as a GPR37 sheddase. Additionally, we discovered that the receptor is subject to another cleavage step in cultured cells. Using site-directed mutagenesis, the site (Arg54↓Asp55) was localized to a highly conserved region at the distal end of the ectodomain that contains a recognition site for the proprotein convertase furin. The cleavage by furin was confirmed by using furin-deficient human colon carcinoma LoVo cells and proprotein convertase inhibitors. GPR37 is thus the first multispanning membrane protein that has been validated as an ADAM10 substrate and the first GPCR that is processed by both furin and ADAM10. The unconventional N-terminal processing may represent an important regulatory element for GPR37.
Collapse
Affiliation(s)
- S Orvokki Mattila
- Medical Research Center Oulu, Research Unit of Biomedicine, University of Oulu, Oulu, Finland
| | - Hanna E Tuhkanen
- Medical Research Center Oulu, Research Unit of Biomedicine, University of Oulu, Oulu, Finland
| | - Jarkko J Lackman
- Medical Research Center Oulu, Research Unit of Biomedicine, University of Oulu, Oulu, Finland
| | - Anja Konzack
- Medical Research Center Oulu, Research Unit of Biomedicine, University of Oulu, Oulu, Finland
| | - Xavier Morató
- Unitat de Farmacologia, Departament de Patologia i Terapèutica Experimental, Facultat de Medicina i Ciències de la Salut, IDIBELL, Universitat de Barcelona, Barcelona, Spain
| | - Josep Argerich
- Unitat de Farmacologia, Departament de Patologia i Terapèutica Experimental, Facultat de Medicina i Ciències de la Salut, IDIBELL, Universitat de Barcelona, Barcelona, Spain
| | - Paul Saftig
- Institute of Biochemistry, Kiel University, Kiel, Germany
| | - Francisco Ciruela
- Unitat de Farmacologia, Departament de Patologia i Terapèutica Experimental, Facultat de Medicina i Ciències de la Salut, IDIBELL, Universitat de Barcelona, Barcelona, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Ulla E Petäjä-Repo
- Medical Research Center Oulu, Research Unit of Biomedicine, University of Oulu, Oulu, Finland
| |
Collapse
|
22
|
Olgun G, Nabi A, Tastan O. NoRCE: non-coding RNA sets cis enrichment tool. BMC Bioinformatics 2021; 22:294. [PMID: 34078267 PMCID: PMC8170991 DOI: 10.1186/s12859-021-04112-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 03/30/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND While some non-coding RNAs (ncRNAs) are assigned critical regulatory roles, most remain functionally uncharacterized. This presents a challenge whenever an interesting set of ncRNAs needs to be analyzed in a functional context. Transcripts located close-by on the genome are often regulated together. This genomic proximity on the sequence can hint at a functional association. RESULTS We present a tool, NoRCE, that performs cis enrichment analysis for a given set of ncRNAs. Enrichment is carried out using the functional annotations of the coding genes located proximal to the input ncRNAs. Other biologically relevant information such as topologically associating domain (TAD) boundaries, co-expression patterns, and miRNA target prediction information can be incorporated to conduct a richer enrichment analysis. To this end, NoRCE includes several relevant datasets as part of its data repository, including cell-line specific TAD boundaries, functional gene sets, and expression data for coding & ncRNAs specific to cancer. Additionally, the users can utilize custom data files in their investigation. Enrichment results can be retrieved in a tabular format or visualized in several different ways. NoRCE is currently available for the following species: human, mouse, rat, zebrafish, fruit fly, worm, and yeast. CONCLUSIONS NoRCE is a platform-independent, user-friendly, comprehensive R package that can be used to gain insight into the functional importance of a list of ncRNAs of any type. The tool offers flexibility to conduct the users' preferred set of analyses by designing their own pipeline of analysis. NoRCE is available in Bioconductor and https://github.com/guldenolgun/NoRCE .
Collapse
Affiliation(s)
- Gulden Olgun
- Department of Computer Engineering, Bilkent University, Ankara, Turkey.,Cancer Data Science Lab, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Afshan Nabi
- Faculty of Engineering and Natural Sciences, Sabanci University, 34956, Istanbul, Turkey
| | - Oznur Tastan
- Faculty of Engineering and Natural Sciences, Sabanci University, 34956, Istanbul, Turkey. .,Cancer Data Science Lab, National Cancer Institute, National Institute of Health, Bethesda, MD, USA.
| |
Collapse
|
23
|
Li X, Su X, Liu J, Li H, Li M, Li W, Luo XJ. Transcriptome-wide association study identifies new susceptibility genes and pathways for depression. Transl Psychiatry 2021; 11:306. [PMID: 34021117 PMCID: PMC8140098 DOI: 10.1038/s41398-021-01411-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 04/22/2021] [Accepted: 04/30/2021] [Indexed: 12/11/2022] Open
Abstract
Depression is the most prevalent mental disorder with substantial morbidity and mortality. Although genome-wide association studies (GWASs) have identified multiple risk variants for depression, due to the complicated gene regulatory mechanisms and complexity of linkage disequilibrium (LD), the biological mechanisms by which the risk variants exert their effects on depression remain largely unknown. Here, we perform a transcriptome-wide association study (TWAS) of depression by integrating GWAS summary statistics from 807,553 individuals (246,363 depression cases and 561,190 controls) and summary-level gene-expression data (from the dorsolateral prefrontal cortex (DLPFC) of 1003 individuals). We identified 53 transcriptome-wide significant (TWS) risk genes for depression, of which 23 genes were not implicated in risk loci of the original GWAS. Seven out of 53 risk genes (B3GALTL, FADS1, TCTEX1D1, XPNPEP3, ZMAT2, ZNF501 and ZNF502) showed TWS associations with depression in two independent brain expression quantitative loci (eQTL) datasets, suggesting that these genes may represent promising candidates. We further conducted conditional analyses and identified the potential risk genes that driven the TWAS association signal in each locus. Finally, pathway enrichment analysis revealed biologically pathways relevant to depression. Our study identified new depression risk genes whose expression dysregulation may play a role in depression. More importantly, we translated the GWAS associations into risk genes and relevant pathways. Further mechanistic study and functional characterization of the TWS depression risk genes will facilitate the diagnostics and therapeutics for depression.
Collapse
Affiliation(s)
- Xiaoyan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, 650204, Kunming, Yunnan, China
- Key Laboratory of Intelligent Computing and Signal Processing of Ministry of Education, Institutes of Physical Science and Information Technology, Anhui University, 230601, Hefei, Anhui, China
| | - Xi Su
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, Henan, China
| | - Jiewei Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, 650204, Kunming, Yunnan, China
| | - Huijuan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, 650204, Kunming, Yunnan, China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, 650204, Kunming, Yunnan, China
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, 650204, Kunming, Yunnan, China
| | - Wenqiang Li
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China.
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, Henan, China.
| | - Xiong-Jian Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, 650204, Kunming, Yunnan, China.
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, 650204, Kunming, Yunnan, China.
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, 650204, Kunming, China.
| |
Collapse
|
24
|
Protective Effects of p-Coumaric Acid Isolated from Vaccinium bracteatum Thunb. Leaf Extract on Corticosterone-Induced Neurotoxicity in SH-SY5Y Cells and Primary Rat Cortical Neurons. Processes (Basel) 2021. [DOI: 10.3390/pr9050869] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Corticosterone (CORT)-induced oxidative stress and neurotoxicity can cause neuronal dysfunction and mental disorders. In the present study, we investigated the effects and mechanism of the HP-20 resin fraction of the water extract of Vaccinium bracteatum leaves (NET-D1602) and its bioactive compound p-coumaric acid on neuronal cell damage in SH-SY5Y cells and primary culture of rat cortical cells. NET-D1602 and p-coumaric acid significantly improved cell viability in CORT-induced neurotoxicity in SH-SY5Y cells and primary cultures of rat cortical cells, and increased the activities of antioxidant enzymes (superoxide dismutase and catalase) against CORT-induced neurotoxicity in SH-SY5Y cells. NET-D1602 and p-coumaric acid increased the phosphorylation levels of ERK1/2 and cAMP response element-binding protein (CREB) in cortical neurons. In addition, CREB phosphorylation by NET-D1602 and p-coumaric acid was dramatically reversed by PKA, c-Raf/ERK, PI3K, and mTOR inhibitors. Lastly, we demonstrated the neuroprotective effects of NET-D1602 (3 and 10 μg/mL) and p-coumaric acid (3 and 10 μM) via increased CREB phosphorylation in CORT-induced neurotoxicity mediated via the ERK1/2, Akt, and mTOR pathways. These results suggest that p-coumaric acid is a potential neuroprotective component of NET-D1602, with the ability to protect against CORT-induced neurotoxicity by regulating ERK1/2, Akt, and mTOR-mediated CREB phosphorylation.
Collapse
|
25
|
Huang X, Li W, You B, Tang W, Gan T, Feng C, Li C, Yang R. Serum Metabonomic Study on the Antidepressant-like Effects of Ellagic Acid in a Chronic Unpredictable Mild Stress-Induced Mouse Model. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:9546-9556. [PMID: 32786855 DOI: 10.1021/acs.jafc.0c02895] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
As a polyphenol, ellagic acid (EA) has shown potential antidepressant activity. In this study, the effects and serum metabolomic analysis of EA against depression were investigated using a chronic unpredictable mild stress-induced (CUMS) model. EA (20 or 100 mg/kg body weight) significantly ameliorated the CUMS-induced depression-like behaviors, including reduced body weight, decreased sucrose preference, and increased immobility time in both the tail suspension test and the forced swimming test. Furthermore, EA attenuated the CUMS-induced hippocampal damage and significantly increased the brain-derived neurotrophic factor (BDNF) and the serotonin (5-HT) levels as well as suppressed the inflammatory response. The metabolomics analysis showed that the disturbance of glycerophospholipid (phosphatidylethanolamine and phosphatidylinositol), amino acid (l-arginine and N-stearoyl serine), and purine (uric acid) metabolism induced by CUMS was attenuated by the EA treatment. Furthermore, the correlation analysis indicated that the metabolite changes were strongly correlated with behavioral disorders, BDNF, 5-HT, and inflammatory cytokines levels. This study provided new insights for the antidepressant effects of EA and suggests that EA may be a potential nutraceutical for improving the management of depression.
Collapse
Affiliation(s)
- Xiaoxia Huang
- Hainan Key Laboratory of Food Nutrition and Functional Food, College of Food Science and Engineering, Hainan University, Haikou 570228, China
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Wu Li
- Hainan Key Laboratory of Food Nutrition and Functional Food, College of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Bangyan You
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Wanpei Tang
- Hainan Key Laboratory of Food Nutrition and Functional Food, College of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Tingsheng Gan
- Hainan Key Laboratory of Food Nutrition and Functional Food, College of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Chao Feng
- Hainan Key Laboratory of Food Nutrition and Functional Food, College of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Congfa Li
- Hainan Key Laboratory of Food Nutrition and Functional Food, College of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Ruili Yang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
26
|
Watkins LR, Orlandi C. Orphan G Protein Coupled Receptors in Affective Disorders. Genes (Basel) 2020; 11:E694. [PMID: 32599826 PMCID: PMC7349732 DOI: 10.3390/genes11060694] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/20/2020] [Accepted: 06/21/2020] [Indexed: 12/12/2022] Open
Abstract
G protein coupled receptors (GPCRs) are the main mediators of signal transduction in the central nervous system. Therefore, it is not surprising that many GPCRs have long been investigated for their role in the development of anxiety and mood disorders, as well as in the mechanism of action of antidepressant therapies. Importantly, the endogenous ligands for a large group of GPCRs have not yet been identified and are therefore known as orphan GPCRs (oGPCRs). Nonetheless, growing evidence from animal studies, together with genome wide association studies (GWAS) and post-mortem transcriptomic analysis in patients, pointed at many oGPCRs as potential pharmacological targets. Among these discoveries, we summarize in this review how emotional behaviors are modulated by the following oGPCRs: ADGRB2 (BAI2), ADGRG1 (GPR56), GPR3, GPR26, GPR37, GPR50, GPR52, GPR61, GPR62, GPR88, GPR135, GPR158, and GPRC5B.
Collapse
Affiliation(s)
| | - Cesare Orlandi
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642, USA;
| |
Collapse
|
27
|
Hirabayashi Y, Kim YJ. Roles of GPRC5 family proteins: focusing on GPRC5B and lipid-mediated signalling. J Biochem 2020; 167:541-547. [DOI: 10.1093/jb/mvaa030] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 02/26/2020] [Indexed: 12/15/2022] Open
Abstract
Abstract
In the past decade, physiological roles and molecular functions of GPRC5 family receptors, originally identified as retinoic acid-induced gene products, have been uncovered, even though their intrinsic agonists are still a mystery. They are differentially distributed in certain tissues and cells in the body suggesting that cell-type-specific regulations and functions are significant. Molecular biological approaches and knockout mouse studies reveal that GPRC5 family proteins have pivotal roles in cancer progression and control of metabolic homeostasis pathways. Remarkably, GPRC5B-mediated tyrosine-phosphorylation signalling cascades play a critical role in development of obesity and insulin resistance through dynamic sphingolipid metabolism.
Collapse
Affiliation(s)
| | - Yeon-Jeong Kim
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan
| |
Collapse
|
28
|
McCrary MR, Jiang MQ, Giddens MM, Zhang JY, Owino S, Wei ZZ, Zhong W, Gu X, Xin H, Hall RA, Wei L, Yu SP. Protective effects of GPR37 via regulation of inflammation and multiple cell death pathways after ischemic stroke in mice. FASEB J 2019; 33:10680-10691. [PMID: 31268736 PMCID: PMC6766648 DOI: 10.1096/fj.201900070r] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 06/04/2019] [Indexed: 12/15/2022]
Abstract
GPCR 37 (GPR37) is a GPCR expressed in the CNS; its physiological and pathophysiological functions are largely unknown. We tested the role of GPR37 in the ischemic brain of GPR37 knockout (KO) mice, exploring the idea that GPR37 might be protective against ischemic damage. In an ischemic stroke model, GPR37 KO mice exhibited increased infarction and cell death compared with wild-type (WT) mice, measured by 2,3,5-triphenyl-2H-tetrazolium chloride and TUNEL staining 24 h after stroke. Moreover, more severe functional deficits were detected in GPR37 KO mice in the adhesive-removal and corner tests. In the peri-infarct region of GPR37 KO mice, there was significantly more apoptotic and autophagic cell death accompanied by caspase-3 activation and attenuated mechanistic target of rapamycin signaling. GPR37 deletion attenuated astrocyte activation and astrogliosis compared with WT stroke controls 24-72 h after stroke. Immunohistochemical staining showed more ionized calcium-binding adapter molecule 1-positive cells in the ischemic cortex of GPR37 KO mice, and RT-PCR identified an enrichment of M1-type microglia or macrophage markers in the GPR37 KO ischemic cortex. Western blotting demonstrated higher levels of inflammatory factors IL-1β, IL-6, monocyte chemoattractant protein, and macrophage inflammatory protein-1α in GPR37-KO mice after ischemia. Thus, GPR37 plays a multifaceted role after stroke, suggesting a novel target for stroke therapy.-McCrary, M. R., Jiang, M. Q., Giddens, M. M., Zhang, J. Y., Owino, S., Wei, Z. Z., Zhong, W., Gu, X., Xin, H., Hall, R. A., Wei, L., Yu, S. P. Protective effects of GPR37 via regulation of inflammation and multiple cell death pathways after ischemic stroke in mice.
Collapse
Affiliation(s)
- Myles R. McCrary
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Michael Q. Jiang
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Michelle M. Giddens
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - James Y. Zhang
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Sharon Owino
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Zheng Z. Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Weiwei Zhong
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Xiaohuan Gu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Huang Xin
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Randy A. Hall
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Shan P. Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, Georgia, USA
| |
Collapse
|
29
|
Song C, Orlandi C, Sutton LP, Martemyanov KA. The signaling proteins GPR158 and RGS7 modulate excitability of L2/3 pyramidal neurons and control A-type potassium channel in the prelimbic cortex. J Biol Chem 2019; 294:13145-13157. [PMID: 31311860 DOI: 10.1074/jbc.ra119.007533] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 07/11/2019] [Indexed: 12/18/2022] Open
Abstract
Stress profoundly affects physiological properties of neurons across brain circuits and thereby increases the risk for depression. However, the molecular and cellular mechanisms mediating these effects are poorly understood. In this study, we report that chronic physical restraint stress in mice decreases excitability specifically in layer 2/3 of pyramidal neurons within the prelimbic subarea of the prefrontal cortex (PFC) accompanied by the induction of depressive-like behavioral states. We found that a complex between G protein-coupled receptor (GPCR) 158 (GPR158) and regulator of G protein signaling 7 (RGS7), a regulatory GPCR signaling node recently discovered to be a key modulator of affective behaviors, plays a key role in controlling stress-induced changes in excitability in this neuronal population. Deletion of GPR158 or RGS7 enhanced excitability of layer 2/3 PFC neurons and prevented the impact of stress. Investigation of the underlying molecular mechanisms revealed that the A-type potassium channel Kv4.2 subunit is a molecular target of the GPR158-RGS7 complex. We further report that GPR158 physically associates with Kv4.2 channel and promotes its function by suppressing inhibitory modulation by cAMP-protein kinase A (PKA)-mediated phosphorylation. Taken together, our observations reveal a critical mechanism that adjusts neuronal excitability in L2/3 pyramidal neurons of the PFC and may thereby modulate the effects of stress on depression.
Collapse
Affiliation(s)
- Chenghui Song
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458
| | - Cesare Orlandi
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458
| | - Laurie P Sutton
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458.
| |
Collapse
|
30
|
Luminescence- and Fluorescence-Based Complementation Assays to Screen for GPCR Oligomerization: Current State of the Art. Int J Mol Sci 2019; 20:ijms20122958. [PMID: 31213021 PMCID: PMC6627893 DOI: 10.3390/ijms20122958] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/05/2019] [Accepted: 06/12/2019] [Indexed: 01/22/2023] Open
Abstract
G protein-coupled receptors (GPCRs) have the propensity to form homo- and heterodimers. Dysfunction of these dimers has been associated with multiple diseases, e.g., pre-eclampsia, schizophrenia, and depression, among others. Over the past two decades, considerable efforts have been made towards the development of screening assays for studying these GPCR dimer complexes in living cells. As a first step, a robust in vitro assay in an overexpression system is essential to identify and characterize specific GPCR–GPCR interactions, followed by methodologies to demonstrate association at endogenous levels and eventually in vivo. This review focuses on protein complementation assays (PCAs) which have been utilized to study GPCR oligomerization. These approaches are typically fluorescence- and luminescence-based, making identification and localization of protein–protein interactions feasible. The GPCRs of interest are fused to complementary fluorescent or luminescent fragments that, upon GPCR di- or oligomerization, may reconstitute to a functional reporter, of which the activity can be measured. Various protein complementation assays have the disadvantage that the interaction between the reconstituted split fragments is irreversible, which can lead to false positive read-outs. Reversible systems offer several advantages, as they do not only allow to follow the kinetics of GPCR–GPCR interactions, but also allow evaluation of receptor complex modulation by ligands (either agonists or antagonists). Protein complementation assays may be used for high throughput screenings as well, which is highly relevant given the growing interest and effort to identify small molecule drugs that could potentially target disease-relevant dimers. In addition to providing an overview on how PCAs have allowed to gain better insights into GPCR–GPCR interactions, this review also aims at providing practical guidance on how to perform PCA-based assays.
Collapse
|
31
|
Wong BCF, Chau CKL, Ao FK, Mo CH, Wong SY, Wong YH, So HC. Differential associations of depression-related phenotypes with cardiometabolic risks: Polygenic analyses and exploring shared genetic variants and pathways. Depress Anxiety 2019; 36:330-344. [PMID: 30521077 DOI: 10.1002/da.22861] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 06/11/2018] [Accepted: 10/20/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Numerous studies have suggested associations between depression and cardiometabolic (CM) diseases. However, little is known about the mechanism underlying this comorbidity, and whether the relationship differs by depression subtypes. METHODS Using polygenic risk scores (PRS) and linkage disequilibrium (LD) score regression, we investigated the genetic overlap of various depression-related phenotypes with a comprehensive panel of 20 CM traits. GWAS results for major depressive disorder (MDD) were taken from the PGC and CONVERGE studies, with the latter focusing on severe melancholic depression. GWAS results on general depressive symptoms (DS) and neuroticism were also included. We identified the shared genetic variants and inferred enriched pathways. We also looked for drugs over-represented among the top-shared genes, with an aim to finding repositioning opportunities for comorbidities. RESULTS We found significant genetic overlap between MDD, DS, and neuroticism with cardiometabolic traits. In general, positive polygenic associations with CM abnormalities were observed except for MDD-CONVERGE. Counterintuitively, PRS representing severe melancholic depression was associated with reduced CM risks. Enrichment analyses of shared SNPs revealed many interesting pathways such as those related to inflammation that underlie the comorbidity of depressive and CM traits. Using a gene-set analysis approach, we also revealed several repositioning candidates with literature support (e.g., bupropion). CONCLUSIONS Our study highlights shared genetic bases of depression with CM traits, and suggests the associations vary by depression subtypes, which may have implications in targeted prevention of cardiovascular events for patients. Identification of shared genetic factors may also guide drug discovery for the comorbidities.
Collapse
Affiliation(s)
- Brian Chi-Fung Wong
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Carlos Kwan-Long Chau
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Fu-Kiu Ao
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Cheuk-Hei Mo
- Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Sze-Yung Wong
- Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Yui-Hang Wong
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Hon-Cheong So
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong.,KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Kunming Institute of Zoology and The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
32
|
Yang LN, Pu JC, Liu LX, Wang GW, Zhou XY, Zhang YQ, Liu YY, Xie P. Integrated Metabolomics and Proteomics Analysis Revealed Second Messenger System Disturbance in Hippocampus of Chronic Social Defeat Stress Rat. Front Neurosci 2019; 13:247. [PMID: 30983951 PMCID: PMC6448023 DOI: 10.3389/fnins.2019.00247] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 03/01/2019] [Indexed: 12/17/2022] Open
Abstract
Depression is a common and disabling mental disorder characterized by high disability and mortality, but its physiopathology remains unclear. In this study, we combined a non-targeted gas chromatography-mass spectrometry (GC-MS)-based metabolomic approach and isobaric tags for relative and absolute quantitation (iTRAQ)-based proteomic analysis to elucidate metabolite and protein alterations in the hippocampus of rat after chronic social defeat stress (CSDS), an extensively used animal model of depression. Ingenuity pathway analysis (IPA) was conducted to integrate underlying relationships among differentially expressed metabolites and proteins. Twenty-five significantly different expressed metabolites and 234 differentially expressed proteins were identified between CSDS and control groups. IPA canonical pathways and network analyses revealed that intracellular second messenger/signal transduction cascades were most significantly altered in the hippocampus of CSDS rats, including cyclic adenosine monophosphate (cAMP), phosphoinositol, tyrosine kinase, and arachidonic acid systems. These results provide a better understanding of biological mechanisms underlying depression, and may help identify potential targets for novel antidepressants.
Collapse
Affiliation(s)
- Li-Ning Yang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Jun-Cai Pu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Lan-Xiang Liu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Guo-Wei Wang
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Xin-Yu Zhou
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yu-Qing Zhang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yi-Yun Liu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Peng Xie
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| |
Collapse
|
33
|
Pazini FL, Cunha MP, Rodrigues ALS. The possible beneficial effects of creatine for the management of depression. Prog Neuropsychopharmacol Biol Psychiatry 2019; 89:193-206. [PMID: 30193988 DOI: 10.1016/j.pnpbp.2018.08.029] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 08/17/2018] [Accepted: 08/28/2018] [Indexed: 01/23/2023]
Abstract
Depression, a highly prevalent neuropsychiatric disorder worldwide, causes a heavy burden for the society and is associated with suicide risk. The treatment of this disorder remains a challenge, since currently available antidepressants provide a slow and, often, incomplete response and cause several side effects that contribute to diminish the adhesion of patients to treatment. In this context, several nutraceuticals have been investigated regarding their possible beneficial effects for the management of this neuropsychiatric disorder. Creatine stands out as a supplement frequently used for ergogenic purpose, but it also is a neuroprotective compound with potential to treat or mitigate a broad range of central nervous systems diseases, including depression. This review presents preclinical and clinical evidence that creatine may exhibit antidepressant properties. The focus is given on the possible molecular mechanisms underlying its effects based on the results obtained with different animal models of depression. Finally, evidence obtained in animal models of depression addressing the possibility that creatine may produce rapid antidepressant effect, similar to ketamine, are also presented and discussed.
Collapse
Affiliation(s)
- Francis L Pazini
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Campus Universitário, Trindade, 88040-900 Florianópolis, SC, Brazil
| | - Mauricio P Cunha
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Campus Universitário, Trindade, 88040-900 Florianópolis, SC, Brazil
| | - Ana Lúcia S Rodrigues
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Campus Universitário, Trindade, 88040-900 Florianópolis, SC, Brazil.
| |
Collapse
|
34
|
Huang C, Yang X, Zeng B, Zeng L, Gong X, Zhou C, Xia J, Lian B, Qin Y, Yang L, Liu L, Xie P. Proteomic analysis of olfactory bulb suggests CACNA1E as a promoter of CREB signaling in microbiota-induced depression. J Proteomics 2019; 194:132-147. [DOI: 10.1016/j.jprot.2018.11.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 11/24/2018] [Accepted: 11/26/2018] [Indexed: 12/18/2022]
|
35
|
Orlandi C, Sutton LP, Muntean BS, Song C, Martemyanov KA. Homeostatic cAMP regulation by the RGS7 complex controls depression-related behaviors. Neuropsychopharmacology 2019; 44:642-653. [PMID: 30546127 PMCID: PMC6333837 DOI: 10.1038/s41386-018-0238-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 09/25/2018] [Accepted: 10/02/2018] [Indexed: 01/27/2023]
Abstract
Affective disorders arise from abnormal responses of the brain to prolonged exposure to challenging environmental stimuli. Recent work identified the orphan receptor GPR158 as a molecular link between chronic stress and depression. Here we reveal a non-canonical mechanism by which GPR158 exerts its effects on stress-induced depression by the complex formation with Regulator of G protein Signaling 7 (RGS7). Chronic stress promotes membrane recruitment of RGS7 via GPR158 in the medial prefrontal cortex (mPFC). The resultant complex suppresses homeostatic regulation of cAMP by inhibitory GPCRs in the region. Accordingly, RGS7 loss in mice induces an antidepressant-like phenotype and resiliency to stress, whereas its restoration within the mPFC is sufficient to rescue this phenotype in a GPR158-dependent way. These findings mechanistically link the unusual orphan receptor-RGS complex to a major stress mediator, the cAMP system and suggest new avenues for pharmacological interventions in affective disorders.
Collapse
Affiliation(s)
- Cesare Orlandi
- 0000000122199231grid.214007.0Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458 USA
| | - Laurie P. Sutton
- 0000000122199231grid.214007.0Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458 USA
| | - Brian S. Muntean
- 0000000122199231grid.214007.0Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458 USA
| | - Chenghui Song
- 0000000122199231grid.214007.0Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458 USA
| | - Kirill A. Martemyanov
- 0000000122199231grid.214007.0Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458 USA
| |
Collapse
|
36
|
Senese NB, Rasenick MM, Traynor JR. The Role of G-proteins and G-protein Regulating Proteins in Depressive Disorders. Front Pharmacol 2018; 9:1289. [PMID: 30483131 PMCID: PMC6244039 DOI: 10.3389/fphar.2018.01289] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/22/2018] [Indexed: 11/29/2022] Open
Abstract
Progress toward new antidepressant therapies has been relatively slow over the past few decades, with the result that individuals suffering from depression often struggle to find an effective treatment – a process often requiring months. Furthermore, the neural factors that contribute to depression remain poorly understood, and there are many open questions regarding the mechanism of action of existing antidepressants. A better understanding of the molecular processes that underlie depression and contribute to antidepressant efficacy is therefore badly needed. In this review we highlight research investigating the role of G-proteins and the regulators of G-protein signaling (RGS) proteins, two protein families that are intimately involved in both the genesis of depressive states and the action of antidepressant drugs. Many antidepressants are known to indirectly affect the function of these proteins. Conversely, dysfunction of the G-protein and RGS systems can affect antidepressant efficacy. However, a great deal remains unknown about how these proteins interact with antidepressants. Findings pertinent to each individual G-protein and RGS protein are summarized from in vitro, in vivo, and clinical studies.
Collapse
Affiliation(s)
- Nicolas B Senese
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, United States.,Jesse Brown VA Medical Center, Chicago, IL, United States.,Department of Pharmacology and Edward F. Domino Research Center, University of Michigan, Ann Arbor, MI, United States
| | - Mark M Rasenick
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, United States.,Jesse Brown VA Medical Center, Chicago, IL, United States.,Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, United States
| | - John R Traynor
- Department of Pharmacology and Edward F. Domino Research Center, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
37
|
Hertz E, Terenius L, Vukojević V, Svenningsson P. GPR37 and GPR37L1 differently interact with dopamine 2 receptors in live cells. Neuropharmacology 2018; 152:51-57. [PMID: 30423289 PMCID: PMC6599889 DOI: 10.1016/j.neuropharm.2018.11.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 10/26/2018] [Accepted: 11/08/2018] [Indexed: 02/09/2023]
Abstract
Receptor-receptor interactions are essential to fine tune receptor responses and new techniques enable closer characterization of the interactions between involved proteins directly in the plasma membrane. Fluorescence cross-correlation spectroscopy (FCCS), which analyses concurrent movement of bound molecules with single-molecule detection limit, was here used to, in live N2a cells, study interactions between the Parkinson's disease (PD) associated orphan receptor GPR37, its homologue GPR37L1, and the two splice variants of the dopamine 2 receptor (D2R). An interaction between GPR37 and both splice forms of D2R was detected. 4-phenylbutyrate (4-PBA), a neuroprotective chemical chaperone known to increase GPR37 expression at the cell surface, increased the fraction of interacting molecules. The interaction was also increased by pramipexole, a D2R agonist commonly used in the treatment of PD, indicating a possible clinically relevance. Cross-correlation, indicating interaction between GPR37L1 and the short isoform of D2R, was also detected. However, this interaction was not changed with 4-PBA or pramipexole treatment. Overall, these data provide further evidence that heteromeric GPR37-D2R exist and can be pharmacologically modulated, which is relevant for the treatment of PD. This article is part of the Special Issue entitled ‘Receptor heteromers and their allosteric receptor-receptor interactions’. GPCR interaction is studied with fluorescence cross-correlation spectroscopy. Interaction between GPR37 and both isoforms of D2R is detected in live cells. GPR37's homologue GPR37L1 is detected to interact with D2RS in live cells. GPR37-D2R interaction is increased by D2-like agonist and 4-PBA treatment.
Collapse
Affiliation(s)
- E Hertz
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden.
| | - L Terenius
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - V Vukojević
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - P Svenningsson
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
38
|
Arabnejad M, Dawkins BA, Bush WS, White BC, Harkness AR, McKinney BA. Transition-transversion encoding and genetic relationship metric in ReliefF feature selection improves pathway enrichment in GWAS. BioData Min 2018; 11:23. [PMID: 30410580 PMCID: PMC6215626 DOI: 10.1186/s13040-018-0186-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 10/22/2018] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND ReliefF is a nearest-neighbor based feature selection algorithm that efficiently detects variants that are important due to statistical interactions or epistasis. For categorical predictors, like genotypes, the standard metric used in ReliefF has been a simple (binary) mismatch difference. In this study, we develop new metrics of varying complexity that incorporate allele sharing, adjustment for allele frequency heterogeneity via the genetic relationship matrix (GRM), and physicochemical differences of variants via a new transition/transversion encoding. METHODS We introduce a new two-dimensional transition/transversion genotype encoding for ReliefF, and we implement three ReliefF attribute metrics: 1.) genotype mismatch (GM), which is the ReliefF standard, 2.) allele mismatch (AM), which accounts for heterozygous differences and has not been used previously in ReliefF, and 3.) the new transition/transversion metric. We incorporate these attribute metrics into the ReliefF nearest neighbor calculation with a Manhattan metric, and we introduce GRM as a new ReliefF nearest-neighbor metric to adjust for allele frequency heterogeneity. RESULTS We apply ReliefF with each metric to a GWAS of major depressive disorder and compare the detection of genes in pathways implicated in depression, including Axon Guidance, Neuronal System, and G Protein-Coupled Receptor Signaling. We also compare with detection by Random Forest and Lasso as well as random/null selection to assess pathway size bias. CONCLUSIONS Our results suggest that using more genetically motivated encodings, such as transition/transversion, and metrics that adjust for allele frequency heterogeneity, such as GRM, lead to ReliefF attribute scores with improved pathway enrichment.
Collapse
Affiliation(s)
- M. Arabnejad
- Tandy School of Computer Science, The University of Tulsa, 800 S. Tucker Dr, Tulsa, OK 74104 USA
| | - B. A. Dawkins
- Department of Mathematics, The University of Tulsa, Tulsa, OK 74104 USA
| | - W. S. Bush
- Institute for Computational Biology, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH 44106 USA
| | - B. C. White
- Tandy School of Computer Science, The University of Tulsa, 800 S. Tucker Dr, Tulsa, OK 74104 USA
| | - A. R. Harkness
- Department of Psychology, The University of Tulsa, Tulsa, OK 74104 USA
| | - B. A. McKinney
- Tandy School of Computer Science, The University of Tulsa, 800 S. Tucker Dr, Tulsa, OK 74104 USA
- Department of Mathematics, The University of Tulsa, Tulsa, OK 74104 USA
| |
Collapse
|
39
|
Sano T, Kohyama-Koganeya A, Kinoshita MO, Tatsukawa T, Shimizu C, Oshima E, Yamada K, Le TD, Akagi T, Tohyama K, Nagao S, Hirabayashi Y. Loss of GPRC5B impairs synapse formation of Purkinje cells with cerebellar nuclear neurons and disrupts cerebellar synaptic plasticity and motor learning. Neurosci Res 2018; 136:33-47. [DOI: 10.1016/j.neures.2018.02.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 02/22/2018] [Accepted: 02/22/2018] [Indexed: 12/30/2022]
|
40
|
Oh DR, Yoo JS, Kim Y, Kang H, Lee H, Lm SJ, Choi EJ, Jung MA, Bae D, Oh KN, Hong JA, Jo A, Shin J, Kim J, Kim YR, Cho SS, Lee BJ, Choi CY. Vaccinium bracteatum Leaf Extract Reverses Chronic Restraint Stress-Induced Depression-Like Behavior in Mice: Regulation of Hypothalamic-Pituitary-Adrenal Axis, Serotonin Turnover Systems, and ERK/Akt Phosphorylation. Front Pharmacol 2018; 9:604. [PMID: 30038568 PMCID: PMC6047486 DOI: 10.3389/fphar.2018.00604] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 05/21/2018] [Indexed: 01/02/2023] Open
Abstract
The leaves of Vaccinium bracteatum Thunb. are a source of traditional herbal medicines found in East Asia. The present study aimed to evaluate the mechanisms underlying the antidepressant-like effects of water extract of V. bracteatum Thunb. leaves (VBLW) in a mouse model of chronic restraint stress (CRS) and to identify the possible molecular in vitro mechanisms of the neuroprotective effects. The CRS-exposed mice were orally administered VBLW (100 and 200 mg/kg) daily for 21 days consecutively. The behavioral effects of VBLW were assessed through the forced swim test (FST) and the open field test (OFT). The levels of serum corticosterone (CORT), corticotropin releasing hormone (CRH), and adrenocorticotropin hormone (ACTH), brain monoamines, such as serotonin, dopamine, and norepinephrine, and serotonin turnover by tryptophan hydroxylase 2 (TPH2), serotonin reuptake (SERT), and monoamine oxidase A (MAO-A) were evaluated, in addition to the extracellular signal-regulated kinases (ERKs)/protein kinase B (Akt) signaling pathway. CRS-exposed mice treated with VBLW (100 and 200 mg/kg) showed significantly reduced immobility time and increased swimming and climbing times in the FST, and increased locomotor activity in the OFT. Moreover, CRS mice treated with VBLW exhibited significantly decreased CORT and ACTH, but enhanced brain monoamine neurotransmitters. In addition, CRS mice treated with VBLW had dramatically decreased protein levels of MAO-A and SERT, but increased TPH2 protein levels in the hippocampus and the PFC. Similarly, VBLW significantly upregulated the ERKs/Akt signaling pathway in the hippocampus and the PFC. Furthermore, VBLW showed neuroprotective effects via increased CREB phosphorylation in CORT-induced cell injury that were mediated through the ERK/Akt/mTOR signaling pathways. These results suggested that the antidepressant-like effects of VBLW might be mediated by the regulation of the HPA axis, glucocorticoids, and serotonin turnover, such as TPH2, SERT, and MAO-A, as well as the concentration of monoamine neurotransmitters, and the activities of ERK and Akt phosphorylation, which were possibly associated with neuroprotective effects.
Collapse
Affiliation(s)
- Dool-Ri Oh
- Jeonnam Bioindustry Foundation, Jeonnam Institute of Natural Resources Research, Jeollanamdo, South Korea.,College of Pharmacy and Research Institute of Drug Development, Chonnam National University, Gwangju, South Korea
| | - Ji-Seok Yoo
- Bioavailability Control Laboratory, College of Pharmacy, Ajou University, Suwon, South Korea
| | - Yujin Kim
- Jeonnam Bioindustry Foundation, Jeonnam Institute of Natural Resources Research, Jeollanamdo, South Korea
| | - Huwon Kang
- Jeonnam Bioindustry Foundation, Jeonnam Institute of Natural Resources Research, Jeollanamdo, South Korea
| | - Hunmi Lee
- Jeonnam Bioindustry Foundation, Jeonnam Institute of Natural Resources Research, Jeollanamdo, South Korea
| | - So J Lm
- Jeonnam Bioindustry Foundation, Jeonnam Institute of Natural Resources Research, Jeollanamdo, South Korea
| | - Eun-Jin Choi
- Jeonnam Bioindustry Foundation, Jeonnam Institute of Natural Resources Research, Jeollanamdo, South Korea
| | - Myung-A Jung
- Jeonnam Bioindustry Foundation, Jeonnam Institute of Natural Resources Research, Jeollanamdo, South Korea
| | - Donghyuck Bae
- Jeonnam Bioindustry Foundation, Jeonnam Institute of Natural Resources Research, Jeollanamdo, South Korea
| | - Kyo-Nyeo Oh
- Jeonnam Bioindustry Foundation, Jeonnam Institute of Natural Resources Research, Jeollanamdo, South Korea
| | - Ji-Ae Hong
- Jeonnam Bioindustry Foundation, Jeonnam Institute of Natural Resources Research, Jeollanamdo, South Korea
| | - Ara Jo
- Jeonnam Bioindustry Foundation, Jeonnam Institute of Natural Resources Research, Jeollanamdo, South Korea
| | - Jawon Shin
- Jeonnam Bioindustry Foundation, Jeonnam Institute of Natural Resources Research, Jeollanamdo, South Korea
| | - Jaeyong Kim
- Jeonnam Bioindustry Foundation, Jeonnam Institute of Natural Resources Research, Jeollanamdo, South Korea
| | - Young R Kim
- College of Pharmacy and Research Institute of Drug Development, Chonnam National University, Gwangju, South Korea
| | - Seung S Cho
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan, South Korea
| | - Beom-Jin Lee
- Bioavailability Control Laboratory, College of Pharmacy, Ajou University, Suwon, South Korea
| | - Chul Yung Choi
- Jeonnam Bioindustry Foundation, Jeonnam Institute of Natural Resources Research, Jeollanamdo, South Korea
| |
Collapse
|
41
|
Serum lipidomic analysis for the discovery of biomarkers for major depressive disorder in drug-free patients. Psychiatry Res 2018; 265:174-182. [PMID: 29719272 DOI: 10.1016/j.psychres.2018.04.029] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 02/19/2018] [Accepted: 04/10/2018] [Indexed: 12/13/2022]
Abstract
Lipidomic analysis can be used to efficiently identify hundreds of lipid molecular species in biological materials and has been recently established as an important tool for biomarker discovery in various neuropsychiatric disorders including major depressive disorder (MDD). In this study, quantitative targeted serum lipidomic profiling was performed on female subjects using liquid chromatography-mass spectrometry. Global lipid profiling of pooled serum samples from 10 patients currently with MDD (cMDD), 10 patients with remitted MDD (rMDD), and 10 healthy controls revealed 37 differentially regulated lipids (DRLs). DRLs were further verified using multiple-reaction monitoring (MRM) in each of the 25 samples from the three groups of independent cohorts. Using multivariate analysis and MRM data we identified serum biomarker panels of discriminatory lipids that differentiated between pairs of groups: lysophosphatidic acid (LPA)(16:1), triglycerides (TG)(44:0), and TG(54:8) distinguished cMDD from controls with 76% accuracy; lysophosphatidylcholines(16:1), TG(44:0), TG(46:0), and TG(50:1) distinguished between cMDD and rMDD at 65% accuracy; and LPA(16:1), TG(52:6), TG(54:8), and TG(58:10) distinguished between rMDD and controls with 60% accuracy. Our lipidomic analysis identified peripheral lipid signatures of MDD, which thereby provides providing important biomarker candidates for MDD.
Collapse
|
42
|
Oh DR, Kim Y, Choi EJ, Jung MA, Oh KN, Hong JA, Bae D, Kim K, Kang H, Kim J, Kim YR, Cho SS, Choi CY. Antidepressant-Like Effects of Vaccinium bracteatum in Chronic Restraint Stress Mice: Functional Actions and Mechanism Explorations. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2018; 46:357-387. [DOI: 10.1142/s0192415x18500180] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The fruit of Vaccinium bracteatum Thunb. (VBF) is commonly known as the oriental blueberry in Korea. The aim of this study was to evaluate the antidepressant-like effects of water VBF extract (VBFW) in a mouse model of chronic restraint stress (CRS) and to identify the underlying mechanisms of its action. The behavioral effects of VBFW were assessed in the forced swim test (FST) and open field test (OFT). The levels of serum corticosterone (CORT), brain monoamines, in addition to the extracellular signal-regulated kinases (ERKs)/protein kinase B (Akt) signaling pathway were evaluated. VBFW treatment significantly reduced the immobility time and increased swimming time in FST without altering the locomotor activity in unstressed mice. Furthermore, CRS mice treated with VBFW exhibited a significantly decreased immobility time in FST and serum CORT, increased locomotor activity in OFT, and enhanced brain monoamine neurotransmitters. Similarly, VBFW significantly upregulated the ERKs/Akt signaling pathway in the hippocampus and PFC. In addition, VBFW may reverse CORT-induced cell death by enhancing cyclic AMP-responsive element-binding protein expression through the up-regulation of ERKs/Akt signaling pathways. In addition, VBFW showed the strong antagonistic effect of the 5-HT[Formula: see text] receptor by inhibiting 5-HT-induced intracellular Ca[Formula: see text] and ERK1/2 phosphorylation. Our study provides evidence that antidepressant-like effects of VBFW might be mediated by the regulation of monoaminergic systems and glucocorticoids, which is possibly associated with neuroprotective effects and antagonism of 5-HT[Formula: see text] receptor.
Collapse
Affiliation(s)
- Dool-Ri Oh
- Jeonnam Bioindustry Foundation, Jeonnam Institute of Natural Resources Research (JINR), Jeollanamdo 59338, Republic of Korea
- College of Pharmacy and Research Institute of Drug Development, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Yujin Kim
- Jeonnam Bioindustry Foundation, Jeonnam Institute of Natural Resources Research (JINR), Jeollanamdo 59338, Republic of Korea
| | - Eun-Jin Choi
- Jeonnam Bioindustry Foundation, Jeonnam Institute of Natural Resources Research (JINR), Jeollanamdo 59338, Republic of Korea
| | - Myung-A Jung
- Jeonnam Bioindustry Foundation, Jeonnam Institute of Natural Resources Research (JINR), Jeollanamdo 59338, Republic of Korea
| | - Kyo-Nyeo Oh
- Jeonnam Bioindustry Foundation, Jeonnam Institute of Natural Resources Research (JINR), Jeollanamdo 59338, Republic of Korea
| | - Ji-Ae Hong
- Jeonnam Bioindustry Foundation, Jeonnam Institute of Natural Resources Research (JINR), Jeollanamdo 59338, Republic of Korea
| | - Donghyuck Bae
- Jeonnam Bioindustry Foundation, Jeonnam Institute of Natural Resources Research (JINR), Jeollanamdo 59338, Republic of Korea
| | - Kwangsu Kim
- Jeonnam Bioindustry Foundation, Jeonnam Institute of Natural Resources Research (JINR), Jeollanamdo 59338, Republic of Korea
| | - Huwon Kang
- Jeonnam Bioindustry Foundation, Jeonnam Institute of Natural Resources Research (JINR), Jeollanamdo 59338, Republic of Korea
| | - Jaeyong Kim
- Jeonnam Bioindustry Foundation, Jeonnam Institute of Natural Resources Research (JINR), Jeollanamdo 59338, Republic of Korea
| | - Young Ran Kim
- College of Pharmacy and Research Institute of Drug Development, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Seung Sik Cho
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan, Jeollanamdo 58554, Republic of Korea
| | - Chul-Young Choi
- Jeonnam Bioindustry Foundation, Jeonnam Institute of Natural Resources Research (JINR), Jeollanamdo 59338, Republic of Korea
| |
Collapse
|
43
|
Sutton LP, Orlandi C, Song C, Oh WC, Muntean BS, Xie K, Filippini A, Xie X, Satterfield R, Yaeger JDW, Renner KJ, Young SM, Xu B, Kwon H, Martemyanov KA. Orphan receptor GPR158 controls stress-induced depression. eLife 2018; 7:33273. [PMID: 29419376 PMCID: PMC5823542 DOI: 10.7554/elife.33273] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 02/06/2018] [Indexed: 01/23/2023] Open
Abstract
Stress can be a motivational force for decisive action and adapting to novel environment; whereas, exposure to chronic stress contributes to the development of depression and anxiety. However, the molecular mechanisms underlying stress-responsive behaviors are not fully understood. Here, we identified the orphan receptor GPR158 as a novel regulator operating in the prefrontal cortex (PFC) that links chronic stress to depression. GPR158 is highly upregulated in the PFC of human subjects with major depressive disorder. Exposure of mice to chronic stress also increased GPR158 protein levels in the PFC in a glucocorticoid-dependent manner. Viral overexpression of GPR158 in the PFC induced depressive-like behaviors. In contrast GPR158 ablation, led to a prominent antidepressant-like phenotype and stress resiliency. We found that GPR158 exerts its effects via modulating synaptic strength altering AMPA receptor activity. Taken together, our findings identify a new player in mood regulation and introduce a pharmacological target for managing depression.
Collapse
Affiliation(s)
- Laurie P Sutton
- Department of Neuroscience, The Scripps Research Institute, Jupiter, United States
| | - Cesare Orlandi
- Department of Neuroscience, The Scripps Research Institute, Jupiter, United States
| | - Chenghui Song
- Department of Neuroscience, The Scripps Research Institute, Jupiter, United States
| | - Won Chan Oh
- Max Planck Florida Institute for Neuroscience, Jupiter, United States
| | - Brian S Muntean
- Department of Neuroscience, The Scripps Research Institute, Jupiter, United States
| | - Keqiang Xie
- Department of Neuroscience, The Scripps Research Institute, Jupiter, United States
| | - Alice Filippini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Xiangyang Xie
- Department of Neuroscience, The Scripps Research Institute, Jupiter, United States
| | | | - Jazmine D W Yaeger
- Center for Brain and Behavior Research, University of South Dakota, Vermillion, United States.,Department of Biology, University of South Dakota, Vermillion, United States
| | - Kenneth J Renner
- Center for Brain and Behavior Research, University of South Dakota, Vermillion, United States.,Department of Biology, University of South Dakota, Vermillion, United States
| | - Samuel M Young
- Max Planck Florida Institute for Neuroscience, Jupiter, United States.,Department of Anatomy and Cell Biology, University of Iowa, Iowa, United States.,Aging Mind and Brain Initiative, University of Iowa, Iowa, United States.,Department of Otolaryngology, Carver College of Medicine, University of Iowa, Iowa, United States
| | - Baoji Xu
- Department of Neuroscience, The Scripps Research Institute, Jupiter, United States
| | - Hyungbae Kwon
- Max Planck Florida Institute for Neuroscience, Jupiter, United States.,Max Planck Institute of Neurobiology, Martinsried, Germany
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, United States
| |
Collapse
|
44
|
Lichawska-Cieslar A, Pietrzycka R, Ligeza J, Kulecka M, Paziewska A, Kalita A, Dolicka DD, Wilamowski M, Miekus K, Ostrowski J, Mikula M, Jura J. RNA sequencing reveals widespread transcriptome changes in a renal carcinoma cell line. Oncotarget 2018; 9:8597-8613. [PMID: 29492220 PMCID: PMC5823589 DOI: 10.18632/oncotarget.24269] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 12/30/2017] [Indexed: 12/12/2022] Open
Abstract
We used RNA sequencing (RNA-Seq) technology to investigate changes in the transcriptome profile in the Caki-1 clear cell renal cell carcinoma (ccRCC) cells, which overexpress monocyte chemoattractant protein-induced protein 1 (MCPIP1). RNA-Seq data showed changes in 11.6% and 41.8% of the global transcriptome of Caki-1 cells overexpressing wild-type MCPIP1 or its D141N mutant, respectively. Gene ontology and KEGG pathway functional analyses showed that these transcripts encoded proteins involved in cell cycle progression, protein folding in the endoplasmic reticulum, hypoxia response and cell signalling. We identified 219 downregulated transcripts in MCPIP1-expressing cells that were either unchanged or upregulated in D141N-expressing cells. We validated downregulation of 15 transcripts belonging to different functional pathways by qRT-PCR. The growth and viability of MCPIP1-expressing cells was reduced because of elevated p21Cip1 levels. MCPIP1-expressing cells also showed reduced levels of DDB1 transcript that encodes component of the E3 ubiquitin ligase that degrades p21Cip1. These results demonstrate that MCPIP1 influences the growth and viability of ccRCC cells by increasing or decreasing the transcript levels for proteins involved in cell cycle progression, protein folding, hypoxia response, and cell signaling.
Collapse
Affiliation(s)
- Agata Lichawska-Cieslar
- Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Roza Pietrzycka
- Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Janusz Ligeza
- Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Maria Kulecka
- Departments of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Agnieszka Paziewska
- Departments of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Agata Kalita
- Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Dobrochna D. Dolicka
- Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Mateusz Wilamowski
- Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Katarzyna Miekus
- Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Jerzy Ostrowski
- Departments of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Department of Genetics, Maria Sklodowska-Curie Memorial Cancer Centre and Institute of Oncology, Warsaw, Poland
| | - Michal Mikula
- Department of Genetics, Maria Sklodowska-Curie Memorial Cancer Centre and Institute of Oncology, Warsaw, Poland
| | - Jolanta Jura
- Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
45
|
Chlis NK, Bei ES, Zervakis M. Introducing a Stable Bootstrap Validation Framework for Reliable Genomic Signature Extraction. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2018; 15:181-190. [PMID: 27913357 DOI: 10.1109/tcbb.2016.2633267] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
The application of machine learning methods for the identification of candidate genes responsible for phenotypes of interest, such as cancer, is a major challenge in the field of bioinformatics. These lists of genes are often called genomic signatures and their linkage to phenotype associations may form a significant step in discovering the causation between genotypes and phenotypes. Traditional methods that produce genomic signatures from DNA Microarray data tend to extract significantly different lists under relatively small variations of the training data. That instability hinders the validity of research findings and raises skepticism about the reliability of such methods. In this study, a complete framework for the extraction of stable and reliable lists of candidate genes is presented. The proposed methodology enforces stability of results at the validation step and as a result, it is independent of the feature selection and classification methods used. Furthermore, two different statistical tests are performed in order to assess the statistical significance of the observed results. Moreover, the consistency of the signatures extracted by independent executions of the proposed method is also evaluated. The results of this study highlight the importance of stability issues in genomic signatures, beyond their prediction capabilities.
Collapse
|
46
|
Alavi MS, Shamsizadeh A, Azhdari-Zarmehri H, Roohbakhsh A. Orphan G protein-coupled receptors: The role in CNS disorders. Biomed Pharmacother 2017; 98:222-232. [PMID: 29268243 DOI: 10.1016/j.biopha.2017.12.056] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 12/12/2017] [Accepted: 12/14/2017] [Indexed: 12/20/2022] Open
Abstract
There are various types of receptors in the central nervous system (CNS). G protein-coupled receptors (GPCRs) have the highest expression with a wide range of physiological functions. A newer sub group of these receptors namely orphan GPCRs have been discovered. GPR3, GPR6, GPR17, GPR26, GPR37, GPR39, GPR40, GPR50, GPR52, GPR54, GPR55, GPR85, GPR88, GPR103, and GPR139 are the selected orphan GPCRs for this article. Their roles in the central nervous system have not been understood well so far. However, recent studies show that they may have very important functions in the CNS. Hence, in the present study, we reviewed most recent findings regarding the physiological roles of the selected orphan GPCRs in the CNS. After a brief presentation of each receptor, considering the results from genetic and pharmacological manipulation of the receptors, their roles in the pathophysiology of different diseases and disorders including anxiety, depression, schizophrenia, epilepsy, Alzheimer's disease, Parkinson's disease, and substance abuse will be discussed. At present, our knowledge regarding the role of GPCRs in the brain is very limited. However, previous limited studies show that orphan GPCRs have an important place in psychopharmacology and these receptors are potential new targets for the treatment of major CNS diseases.
Collapse
Affiliation(s)
- Mohaddeseh Sadat Alavi
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Shamsizadeh
- Physiology-Pharmacology Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hassan Azhdari-Zarmehri
- Department of Basic Medical Sciences and Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Ali Roohbakhsh
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
47
|
Liu B, Teschemacher AG, Kasparov S. Astroglia as a cellular target for neuroprotection and treatment of neuro-psychiatric disorders. Glia 2017; 65:1205-1226. [PMID: 28300322 PMCID: PMC5669250 DOI: 10.1002/glia.23136] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 02/15/2017] [Accepted: 02/17/2017] [Indexed: 12/12/2022]
Abstract
Astrocytes are key homeostatic cells of the central nervous system. They cooperate with neurons at several levels, including ion and water homeostasis, chemical signal transmission, blood flow regulation, immune and oxidative stress defense, supply of metabolites and neurogenesis. Astroglia is also important for viability and maturation of stem-cell derived neurons. Neurons critically depend on intrinsic protective and supportive properties of astrocytes. Conversely, all forms of pathogenic stimuli which disturb astrocytic functions compromise neuronal functionality and viability. Support of neuroprotective functions of astrocytes is thus an important strategy for enhancing neuronal survival and improving outcomes in disease states. In this review, we first briefly examine how astrocytic dysfunction contributes to major neurological disorders, which are traditionally associated with malfunctioning of processes residing in neurons. Possible molecular entities within astrocytes that could underpin the cause, initiation and/or progression of various disorders are outlined. In the second section, we explore opportunities enhancing neuroprotective function of astroglia. We consider targeting astrocyte-specific molecular pathways which are involved in neuroprotection or could be expected to have a therapeutic value. Examples of those are oxidative stress defense mechanisms, glutamate uptake, purinergic signaling, water and ion homeostasis, connexin gap junctions, neurotrophic factors and the Nrf2-ARE pathway. We propose that enhancing the neuroprotective capacity of astrocytes is a viable strategy for improving brain resilience and developing new therapeutic approaches.
Collapse
Affiliation(s)
- Beihui Liu
- School of Physiology, Pharmacology and NeuroscienceUniversity of Bristol, University WalkBS8 1TDUnited Kingdom
| | - Anja G. Teschemacher
- School of Physiology, Pharmacology and NeuroscienceUniversity of Bristol, University WalkBS8 1TDUnited Kingdom
| | - Sergey Kasparov
- School of Physiology, Pharmacology and NeuroscienceUniversity of Bristol, University WalkBS8 1TDUnited Kingdom
- Institute for Chemistry and BiologyBaltic Federal UniversityKaliningradRussian Federation
| |
Collapse
|
48
|
Grammatopoulos DK. Regulation of G-protein coupled receptor signalling underpinning neurobiology of mood disorders and depression. Mol Cell Endocrinol 2017; 449:82-89. [PMID: 28229904 DOI: 10.1016/j.mce.2017.02.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 02/09/2017] [Indexed: 02/08/2023]
Abstract
G-protein coupled receptors (GPCRs) have long been at the center of investigations of the neurobiology of depression and mood disorders. Different facets of GPCR signalling pathways, including those controlling monoaminergic and neuropeptidergic hormonal systems are believed to be dysregulated in major depressive and bipolar disorders. Although these receptors are key molecular targets for a variety of therapeutic agents and continue to be the focus of intense pharmaceutical development, the molecular mechanisms activated by these GPCRs and underpin the pathological basis of mood disorders remain poorly understood. This review will discuss some of the emerging regulatory mechanisms of GPCR signaling in the central nervous system (CNS) involving protein-protein interactions, downstream effectors and cross-talk with other signaling molecules and their potential involvement in the neurobiology of psychiatric disease.
Collapse
Affiliation(s)
- Dimitris K Grammatopoulos
- Translational Medicine, Warwick Medical School & Clinical Biochemistry, Coventry and Warwickshire Pathology Service, United Kingdom.
| |
Collapse
|
49
|
Morris G, Walder K, McGee SL, Dean OM, Tye SJ, Maes M, Berk M. A model of the mitochondrial basis of bipolar disorder. Neurosci Biobehav Rev 2017; 74:1-20. [DOI: 10.1016/j.neubiorev.2017.01.014] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 01/08/2017] [Accepted: 01/10/2017] [Indexed: 12/11/2022]
|
50
|
Hagihara H, Horikawa T, Nakamura HK, Umemori J, Shoji H, Kamitani Y, Miyakawa T. Circadian Gene Circuitry Predicts Hyperactive Behavior in a Mood Disorder Mouse Model. Cell Rep 2016; 14:2784-96. [PMID: 27028761 DOI: 10.1016/j.celrep.2016.02.067] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 01/07/2016] [Accepted: 02/22/2016] [Indexed: 11/26/2022] Open
Abstract
Bipolar disorder, also known as manic-depressive illness, causes swings in mood and activity levels at irregular intervals. Such changes are difficult to predict, and their molecular basis remains unknown. Here, we use infradian (longer than a day) cyclic activity levels in αCaMKII (Camk2a) mutant mice as a proxy for such mood-associated changes. We report that gene-expression patterns in the hippocampal dentate gyrus could retrospectively predict whether the mice were in a state of high or low locomotor activity (LA). Expression of a subset of circadian genes, as well as levels of cAMP and pCREB, possible upstream regulators of circadian genes, were correlated with LA states, suggesting that the intrinsic molecular circuitry changes concomitant with infradian oscillatory LA. Taken together, these findings shed light onto the molecular basis of how irregular biological rhythms and behavior are controlled by the brain.
Collapse
Affiliation(s)
- Hideo Hagihara
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Kawaguchi, Saitama 332-0012, Japan
| | - Tomoyasu Horikawa
- ATR Computational Neuroscience Laboratories, Soraku-gun, Kyoto 619-0288, Japan
| | - Hironori K Nakamura
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Kawaguchi, Saitama 332-0012, Japan
| | - Juzoh Umemori
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Kawaguchi, Saitama 332-0012, Japan
| | - Hirotaka Shoji
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Kawaguchi, Saitama 332-0012, Japan
| | - Yukiyasu Kamitani
- ATR Computational Neuroscience Laboratories, Soraku-gun, Kyoto 619-0288, Japan; Graduate School of Informatics, Kyoto University, Kyoto, Kyoto 606-8501, Japan
| | - Tsuyoshi Miyakawa
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Kawaguchi, Saitama 332-0012, Japan; Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan.
| |
Collapse
|