1
|
Bi M, Chen Y, Tian Z. Serum N-glycoproteomics characterization of differential N-glycosylation in schizophrenia. J Proteomics 2025; 316:105434. [PMID: 40118250 DOI: 10.1016/j.jprot.2025.105434] [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: 12/12/2024] [Revised: 03/17/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025]
Abstract
Glycosylation plays a crucial role in neurotransmission and signaling in schizophrenia; however, comprehensive characterization at the glycoproteome level is still lacking. Here we report our site- and structure-specific quantitative N-glycoproteomics characterization of differential N-glycosylation in the sera of schizophrenia patients at the molecular level of intact N-glycopeptide, where comprehensive qualitative (N-glycosite, monosaccharide composition and sequence structures of N-glycans) and quantitative (fold change) information are obtained. With tandem mass tag labeling, liquid chromatography tandem mass spectrometry analysis and site- and structure-specific DB search using GPSeeker, 7855 intact N-glycopeptides were identified corresponding to 1914 peptide backbones, 1997 N-glycosites and 1671 N-glycoprotein; where 1088 intact N-glycopeptides were differentially expressed in the sera of schizophrenia patients (relative to healthy control) with fold change of no less than 1.5. Function annotation of the corresponding N-glycoproteins was carried out. Neurodegeneration and complement pathway were enriched. These findings provide a comprehensive site- and structure-specific picture of aberrant N-glycosylation in schizophrenia and may foster further function and mechanism studies. SIGNIFICANCE: Schizophrenia, as a complex mental disorder, is affecting an increasing number of individuals globally, yet clinical research has struggled to clearly elucidate its pathogenesis. Current diagnostic and treatment approaches largely depend on patient symptoms and behavior, which lack precision. N-glycoproteomics offers a new dimension of understanding by exploring how schizophrenia alters protein glycosylation patterns in the body. Investigating N-glycoproteins not only contributes to the identification of novel early diagnostic biomarkers but also enhances our knowledge of disease pathogenesis. These molecular insights could pave the way for more accurate diagnostic tools and targeted therapies.
Collapse
Affiliation(s)
- Ming Bi
- School of Chemical Science & Engineering, Tongji University, Shanghai 200092, China
| | - Yun Chen
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China.
| | - Zhixin Tian
- School of Chemical Science & Engineering, Tongji University, Shanghai 200092, China.
| |
Collapse
|
2
|
Zhang H, Li H, Yu M, Yu M, Feng S, Tingting W, Yu Y, Zhang J, Liu K, Tan Y, Xiang B. Modified Electroconvulsive Therapy Normalizes Plasma GNA13 Following Schizophrenic Relapse. J ECT 2024; 40:286-292. [PMID: 39121017 DOI: 10.1097/yct.0000000000001050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/11/2024]
Abstract
OBJECTIVE GNA13 is an important member of the G protein family, and its coding gene GNA13 has been identified as one of the risk genes for schizophrenia (SCZ). This study aimed to investigate the relationship between GNA13 levels and the clinical symptoms of SCZ following treatment with modified electroconvulsive therapy (MECT). METHODS This study recruited 82 SCZ patients and 86 healthy controls (HCs). Each SCZ patient received 6 sessions of MECT. The Positive and Negative Syndrome Scale (PANSS) was used to assess SCZ symptom severity. Plasma levels of GNA13 were measured by enzyme-linked immunosorbent assay. RESULTS Pretreatment, SCZ patients had a higher GNA13 level than HC ( t = 8.199, P < 0.001). MECT reduced the GNA13 level significantly ( t = 11.13, P < 0.001) and normalized the difference between SCZ and HC ( t = 0.219, P = 0.827). After treatment, the downregulation of GNA13 (ΔGNA13) was negatively correlated with the positive symptoms score reduction rate (ΔP) ( r = -0.379, P = 0.027) and positively correlated with the negative score reduction rate (ΔN) ( r = 0.480, P = 0.004) in females. In both males and females, the receiver operating characteristic curve revealed that the pretreatment GNA13 level could help differentiate SCZ from HC (male: area under the curve = 0.792, P < 0.001; female: area under the curve = 0.814, P < 0.001). CONCLUSION The reduced expression of GNA13 after MECT may be related to the exhibition of both negative and positive symptoms of SCZ in female patients.
Collapse
Affiliation(s)
| | | | | | | | - Shuangshuang Feng
- From the Department of Psychiatry, Fundamental and Clinical Research on Mental Disorders Key Laboratory of Luzhou, Medical Laboratory Center, Laboratory of Neurological Diseases & Brain Function, Affiliated Hospital of Southwest Medical University, Luzhou
| | - Wang Tingting
- From the Department of Psychiatry, Fundamental and Clinical Research on Mental Disorders Key Laboratory of Luzhou, Medical Laboratory Center, Laboratory of Neurological Diseases & Brain Function, Affiliated Hospital of Southwest Medical University, Luzhou
| | | | | | | | | | | |
Collapse
|
3
|
Li J, Chen R, Zhou J, Wang Y, Zhao X, Liu C, Zhou P, Chen Y, Song L, Li N, Yan H, Zhao H. Atherosclerotic Autoantigen ALDH4A1 as a Novel Immunological Indicator for Plaque Erosion in Patients with ST Segment Elevated Myocardial Infarction. Thromb Haemost 2024; 124:584-594. [PMID: 38109905 DOI: 10.1055/s-0043-1777265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
OBJECTIVE Aldehyde dehydrogenase 4A1 (ALDH4A1) was recently reported to be a novel autoantigen of atherosclerosis. However, its role in different phenotypes of acute coronary syndrome remains unclear. Herein, we planned to explore the circulating and regional expression of ALDH4A1 in patients with plaque rupture (PR) and plaque erosion (PE) determined by optical coherence tomography (OCT). METHODS AND RESULTS After applying the inclusion and exclusion criteria, a prospective series of 312 patients with ST segment elevated myocardial infarction (STEMI), including 161 patients with PR and 151 patients with PE determined by OCT, were enrolled for plasma ALDH4A1 testing. In addition, ALDH4A1 was quantified using immunofluorescence in aspirated coronary thrombus samples obtained from 31 patients with PR and 25 patients with PE. In addition, we established an atherosclerosis mouse model and analyzed the distribution of ALDH4A1 expression in different mouse organs. Furthermore, we compared the level of ALDH4A1 in the spleen and carotid artery between Apoe-/- and C57 mice. The results showed that the plasma level of ALDH4A1 was significantly higher in STEMI patients with PE than in those with PR (4.6 ng/mL [2.2-8.7] vs. 3.5 ng/mL [1.6-5.6] p = 0.005). The expression of ALDH4A1 in aspirated coronary thrombi was also significantly higher in patients with PE than in those with PR (mean gray value: 32.0 [23.6-40.6] vs. 16.8 [14.0-24.5], p < 0.001). In animal models, the expression of ALDH4A1 is much higher in the spleen than in other organs, and the level of ALDH4A1 is significantly elevated in the spleen and carotid artery of Apoe-/- mice compared with C57 mice. CONCLUSION The high levels of ALDH4A1 in the plasma and aspirated coronary thrombi independently correlated with PE in patients with STEMI. These results suggested that ALDH4A1 is involved in the mechanism of PE and serves as a promising biomarker and treatment target for patients with PE.
Collapse
Affiliation(s)
- Jiannan Li
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Department of Cardiology, Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzhen, China
| | - Runzhen Chen
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jinying Zhou
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Ying Wang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoxiao Zhao
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Chen Liu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Department of Cardiology, Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzhen, China
| | - Peng Zhou
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yi Chen
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Li Song
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Coronary Heart Disease Center, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Nan Li
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Hongbing Yan
- Department of Cardiology, Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzhen, China
- Coronary Heart Disease Center, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Hanjun Zhao
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Coronary Heart Disease Center, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
4
|
Yazla E, Cetin I, Kayadibi H. Assessing the relationship between antipsychotic drug use and prolidase enzyme activity and oxidative stress in schizophrenia patients: a case-control study. Int Clin Psychopharmacol 2023; 38:394-401. [PMID: 37490605 DOI: 10.1097/yic.0000000000000491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
BACKGROUND The relationship between proline, its association with oxidative stress, and its connection to schizophrenia is a subject that has not been sufficiently investigated. OBJECTIVE The aim of this study is to evaluate the possible effects of atypical and combined (typical and atypical) antipsychotic use on serum prolidase enzyme activity (SPEA) and serum oxidative stress parameters, and to assess the relationship between SPEA and oxidative stress in patients with schizophrenia. METHODS A total of 57 patients with schizophrenia, of which 34 were using atypical (AAPG) and 23 were using combined (typical and atypical) (CAPG) antipsychotic therapy, and 28 healthy volunteers (control group) were included in this case-control study. RESULTS SPEA levels of AAPG and CAPG were significantly lower than that of control group ( P = 0.003). The oxidative stress index (OSI) value of AAPG was significantly higher than the other two groups ( P = 0.001). SPEA (<1860 U/l) and OSI (≥0.54) could discriminate schizophrenia patients with antipsychotic therapy from control groups ( P = 0.001 and P = 0.007, respectively). Lower SPEA levels were associated with antipsychotic use ( P = 0.007). CONCLUSION The SPEA values of patients with schizophrenia on antipsychotics were significantly lower compared to controls. OSI values were significantly higher in atypical antipsychotic recipients compared to those on combined antipsychotics and healthy controls.
Collapse
Affiliation(s)
- Ece Yazla
- Department of Psychiatry, Hitit University Faculty of Medicine
| | - Ihsan Cetin
- Department of Medical Biochemistry, Hitit University Faculty of Medicine, Corum
| | - Huseyin Kayadibi
- Department of Biochemistry, Eskisehir Osmangazi University Faculty of Medicine, Eskisehir, Turkey
| |
Collapse
|
5
|
Vaiana AM, Chen Y, Gelfond J, Johnson-Pais TL, Leach RJ, Ramamurthy C, Thompson IM, Morilak DA. Effects of vortioxetine on hippocampal-related cognitive impairment induced in rats by androgen deprivation as a model of prostate cancer treatment. Transl Psychiatry 2023; 13:307. [PMID: 37788996 PMCID: PMC10547695 DOI: 10.1038/s41398-023-02600-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 09/15/2023] [Accepted: 09/20/2023] [Indexed: 10/05/2023] Open
Abstract
Advances in prostate cancer treatment have significantly improved survival, but quality of life for survivors remains an under-studied area of research. Androgen deprivation therapy (ADT) is a foundational treatment for advanced prostate cancer and is used as an adjuvant for prolonged periods in many high-risk, localized tumors. More than half of patients treated with ADT experience debilitating cognitive impairments in domains such as spatial learning and working memory. In this study, we investigated the effects of androgen deprivation on hippocampal-mediated cognition in rats. Vortioxetine, a multimodal antidepressant, has been shown to improve cognition in depressed patients. Thus, we also tested the potential efficacy of vortioxetine in restoring impaired cognition after ADT. We further investigated mechanisms that might contribute to these effects, measuring changes in the circuitry and gene expression within the dorsal hippocampus. ADT via surgical castration induced impairments in visuospatial cognition on the novel object location test and attenuated afferent-evoked local field potentials recorded in the CA1 region of the dorsal hippocampus. Chronic dietary administration of vortioxetine effectively reversed these deficits. Castration significantly altered gene expression in the hippocampus, whereas vortioxetine had little effect. Pathway analysis revealed that androgen depletion altered pathways related to synaptic plasticity. These results suggest that the hippocampus may be vulnerable to ADT, contributing to cognitive impairment in prostate cancer patients. Further, vortioxetine may be a candidate to improve cognition in patients who experience cognitive decline after androgen deprivation therapy for prostate cancer and may do so by restoring molecular and circuit-level plasticity-related mechanisms compromised by ADT.
Collapse
Affiliation(s)
- Alexandra M Vaiana
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, TX, 78229, USA
- Center for Biomedical Neuroscience, University of Texas Health Science Center, San Antonio, TX, 78229, USA
- Mays Cancer Center, University of Texas Health Science Center, San Antonio, TX, 78229, USA
| | - Yidong Chen
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center, San Antonio, TX, 78229, USA
- Department of Population Health Sciences, University of Texas Health Science Center, San Antonio, TX, 78229, USA
| | - Jonathan Gelfond
- Department of Population Health Sciences, University of Texas Health Science Center, San Antonio, TX, 78229, USA
| | - Teresa L Johnson-Pais
- Mays Cancer Center, University of Texas Health Science Center, San Antonio, TX, 78229, USA
- Department of Urology, University of Texas Health Science Center, San Antonio, TX, 78229, USA
| | - Robin J Leach
- Mays Cancer Center, University of Texas Health Science Center, San Antonio, TX, 78229, USA
- Department of Cell Systems & Anatomy, University of Texas Health Science Center, San Antonio, TX, 78229, USA
| | - Chethan Ramamurthy
- Mays Cancer Center, University of Texas Health Science Center, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX, 78229, USA
| | - Ian M Thompson
- Department of Urology, University of Texas Health Science Center, San Antonio, TX, 78229, USA
| | - David A Morilak
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, TX, 78229, USA.
- Center for Biomedical Neuroscience, University of Texas Health Science Center, San Antonio, TX, 78229, USA.
- Mays Cancer Center, University of Texas Health Science Center, San Antonio, TX, 78229, USA.
- South Texas Veterans Health Care System, San Antonio, TX, 78229, USA.
| |
Collapse
|
6
|
Kunii Y, Hino M, Tomita H. Editorial: Molecular pathology in psychiatric diseases: frontiers of postmortem brain research. Front Psychiatry 2023; 14:1286182. [PMID: 37829758 PMCID: PMC10565467 DOI: 10.3389/fpsyt.2023.1286182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 09/13/2023] [Indexed: 10/14/2023] Open
Affiliation(s)
- Yasuto Kunii
- Department of Disaster Psychiatry, International Research Institute of Disaster Science, Tohoku University, Sendai, Japan
- Department of Psychiatry, Tohoku University Hospital, Miyagi, Japan
- Department of Psychiatry, Graduate School of Medicine, Tohoku University, Miyagi, Japan
| | - Mizuki Hino
- Department of Disaster Psychiatry, International Research Institute of Disaster Science, Tohoku University, Sendai, Japan
| | - Hiroaki Tomita
- Department of Disaster Psychiatry, International Research Institute of Disaster Science, Tohoku University, Sendai, Japan
- Department of Psychiatry, Tohoku University Hospital, Miyagi, Japan
- Department of Psychiatry, Graduate School of Medicine, Tohoku University, Miyagi, Japan
| |
Collapse
|
7
|
Arihisa W, Kondo T, Yamaguchi K, Matsumoto J, Nakanishi H, Kunii Y, Akatsu H, Hino M, Hashizume Y, Sato S, Sato S, Niwa S, Yabe H, Sasaki T, Shigenobu S, Setou M. Lipid-correlated alterations in the transcriptome are enriched in several specific pathways in the postmortem prefrontal cortex of Japanese patients with schizophrenia. Neuropsychopharmacol Rep 2023; 43:403-413. [PMID: 37498306 PMCID: PMC10496066 DOI: 10.1002/npr2.12368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/21/2023] [Accepted: 06/26/2023] [Indexed: 07/28/2023] Open
Abstract
AIMS Schizophrenia is a chronic relapsing psychiatric disorder that is characterized by many symptoms and has a high heritability. There were studies showing that the phospholipid abnormalities in subjects with schizophrenia (Front Biosci, S3, 2011, 153; Schizophr Bull, 48, 2022, 1125; Sci Rep, 7, 2017, 6; Anal Bioanal Chem, 400, 2011, 1933). Disturbances in prefrontal cortex phospholipid and fatty acid composition have been reported in subjects with schizophrenia (Sci Rep, 7, 2017, 6; Anal Bioanal Chem, 400, 2011, 1933; Schizophr Res, 215, 2020, 493; J Psychiatr Res, 47, 2013, 636; Int J Mol Sci, 22, 2021). For exploring the signaling pathways contributing to the lipid changes in previous study (Sci Rep, 7, 2017, 6), we performed two types of transcriptome analyses in subjects with schizophrenia: an unbiased transcriptome analysis solely based on RNA-seq data and a correlation analysis between levels of gene expression and lipids. METHODS RNA-Seq analysis was performed in the postmortem prefrontal cortex from 10 subjects with schizophrenia and 5 controls. Correlation analysis between the transcriptome and lipidome from 9 subjects, which are the same samples in the previous lipidomics study (Sci Rep, 7, 2017, 6). RESULTS Extraction of differentially expressed genes (DEGs) and further sequence and functional group analysis revealed changes in gene expression levels in phosphoinositide 3-kinase (PI3K)-Akt signaling and the complement system. In addition, a correlation analysis clarified alterations in ether lipid metabolism pathway, which is not found as DEGs in transcriptome analysis alone. CONCLUSIONS This study provided results of the integrated analysis of the schizophrenia-associated transcriptome and lipidome within the PFC and revealed that lipid-correlated alterations in the transcriptome are enriched in specific pathways including ether lipid metabolism pathway.
Collapse
Affiliation(s)
- Wataru Arihisa
- Department of Cellular and Molecular AnatomyHamamatsu University School of MedicineShizuokaJapan
| | - Takeshi Kondo
- Department of Cellular and Molecular AnatomyHamamatsu University School of MedicineShizuokaJapan
- International Mass Imaging CenterHamamatsu University School of MedicineShizuokaJapan
- Department of Biochemistry, Faculty of Medicine and Graduate School of MedicineHokkaido UniversitySapporoJapan
| | | | - Junya Matsumoto
- Department of Neuropsychiatry, School of MedicineFukushima Medical UniversityFukushimaJapan
| | | | - Yasuto Kunii
- Department of Neuropsychiatry, School of MedicineFukushima Medical UniversityFukushimaJapan
- Department of Disaster PsychiatryInternational Research Institute of Disaster Science, Tohoku UniversitySendaiJapan
| | - Hiroyasu Akatsu
- Choju Medical Institute, Fukushimura HospitalToyohashiJapan
- Department of Community‐based Medical Education/Department of Community‐based MedicineNagoya City University Graduate School of Medical ScienceNagoyaJapan
| | - Mizuki Hino
- Department of Neuropsychiatry, School of MedicineFukushima Medical UniversityFukushimaJapan
- Department of Disaster PsychiatryInternational Research Institute of Disaster Science, Tohoku UniversitySendaiJapan
| | | | - Shumpei Sato
- RIKEN Center for Biosystems Dynamics ResearchOsakaJapan
| | - Shinji Sato
- Business Development, Otsuka Pharmaceutical Co., Ltd. Shinagawa Grand Central TowerTokyoJapan
| | - Shin‐Ichi Niwa
- Department of Psychiatry, Aizu Medical CenterFukushima Medical UniversityFukushimaJapan
| | - Hirooki Yabe
- Department of Neuropsychiatry, School of MedicineFukushima Medical UniversityFukushimaJapan
| | - Takehiko Sasaki
- Department of Biochemical PathophysiologyMedical Research Institute, Tokyo Medical and Dental UniversityTokyoJapan
| | | | - Mitsutoshi Setou
- Department of Cellular and Molecular AnatomyHamamatsu University School of MedicineShizuokaJapan
- International Mass Imaging CenterHamamatsu University School of MedicineShizuokaJapan
- Preeminent Medical Photonics Education & Research CenterHamamatsu University School of MedicineShizuokaJapan
- Department of AnatomyThe University of Hong KongHong KongChina
| |
Collapse
|
8
|
Yi Q, Huang M, Zhang X, Xu Z, Sun J, Wang S, Xu H, Du Z, Liu M. GNA13 inhibits glioblastoma metastasis via the ERKs/FOXO3 signaling pathway. Cell Signal 2023:110789. [PMID: 37392861 DOI: 10.1016/j.cellsig.2023.110789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 06/21/2023] [Accepted: 06/28/2023] [Indexed: 07/03/2023]
Abstract
Glioblastoma (GBM) is a malignant tumor characterized by poor prognosis and low overall survival (OS) rate. Identification of novel biological markers for the diagnosis and treatment of GBM is crucial to developing interventions to improve patient survival. GNA13, a member of the G12 family, has been reported to play important roles in a variety of biological processes involved in tumorigenesis and development. However, its role in GBM is currently unknown. Here, we explored the expression patterns and functions of GNA13 in GBM, as wells its impact on metastasis process. Results showed that GNA13 was downregulated in GBM tissues and correlated with poor prognosis of GBM. Downregulation of GNA13 promoted the migration, invasion and proliferation of GBM cells; whereas its overexpression abolished these effects. Western blots revealed that GNA13 knockdown and overexpression upregulated and inhibited the phosphorylation of ERKs, respectively. Moreover, GNA13 was the upstream of ERKs signaling to regulating ERKs phosphorylation level. Furthermore, U0126 alleviated the metastasis effect induced by GNA13 knockdown. Bioinformatics analyses and qRT-PCR experiments demonstrated that GNA13 could regulate FOXO3, a downstream signaling molecule of ERKs pathway. Overall, our results demonstrate that GNA13 expression is negatively correlated with GBM and can suppress tumor metastasis by inhibiting the ERKs signaling pathway and upregulating FOXO3 expression.
Collapse
Affiliation(s)
- Qingfeng Yi
- Clinical Research Center, Shantou Central Hospital, Shantou 515031, Guangdong, China; Department of Neurosurgery, Shantou Central Hospital, Shantou 515031, Guangdong, China
| | - Meihui Huang
- Department of Central Laboratory, Shantou Central Hospital, Shantou 515031, Guangdong, China
| | - Xiaona Zhang
- Department of Central Laboratory, Shantou Central Hospital, Shantou 515031, Guangdong, China
| | - Zhennan Xu
- Department of Neurosurgery, Shantou Central Hospital, Shantou 515031, Guangdong, China
| | - Jianhong Sun
- Department of Pathology, Shantou Central Hospital, Shantou 515031, Guangdong, China
| | - Shaohong Wang
- Department of Pathology, Shantou Central Hospital, Shantou 515031, Guangdong, China
| | - Haixiong Xu
- Department of Neurosurgery, Shantou Central Hospital, Shantou 515031, Guangdong, China; Shantou Academy of Medical Sciences, Shantou 515031, Guangdong, China.
| | - Zepeng Du
- Department of Central Laboratory, Shantou Central Hospital, Shantou 515031, Guangdong, China; Department of Pathology, Shantou Central Hospital, Shantou 515031, Guangdong, China.
| | - Mingfa Liu
- Department of Neurosurgery, Shantou Central Hospital, Shantou 515031, Guangdong, China.
| |
Collapse
|
9
|
Biswal SR, Singh M, Dwibedy SLL, Kumari S, Muthuswamy S, Kumar A, Kumar S. Deciphering the RNA-binding protein interaction with the mRNAs encoded from human chromosome 15q11.2 BP1-BP2 microdeletion region. Funct Integr Genomics 2023; 23:174. [PMID: 37219715 DOI: 10.1007/s10142-023-01105-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 05/24/2023]
Abstract
Microdeletion of the 15q11.2 BP1-BP2 region, also known as Burnside-Butler susceptibility region, is associated with phenotypes like delayed developmental language abilities along with motor skill disabilities, combined with behavioral and emotional problems. The 15q11.2 microdeletion region harbors four evolutionarily conserved and non-imprinted protein-coding genes: NIPA1, NIPA2, CYFIP1, and TUBGCP5. This microdeletion is a rare copy number variation frequently associated with several pathogenic conditions in humans. The aim of this study is to investigate the RNA-binding proteins binding with the four genes present in 15q11.2 BP1-BP2 microdeletion region. The results of this study will help to better understand the molecular intricacies of the Burnside-Butler Syndrome and also the possible involvement of these interactions in the disease aetiology. Our results of enhanced crosslinking and immunoprecipitation data analysis indicate that most of the RBPs interacting with the 15q11.2 region are involved in the post-transcriptional regulation of the concerned genes. The RBPs binding to this region are found from the in silico analysis, and the interaction of RBPs like FASTKD2 and EFTUD2 with exon-intron junction sequence of CYFIP1 and TUBGCP5 has also been validated by combined EMSA and western blotting experiment. The exon-intron junction binding nature of these proteins suggests their potential involvement in splicing process. This study may help to understand the intricate relationship of RBPs with mRNAs within this region, along with their functional significance in normal development, and lack thereof, in neurodevelopmental disorders. This understanding will help in the formulation of better therapeutic approaches.
Collapse
Affiliation(s)
- Smruti Rekha Biswal
- Department of Life Science, National Institute of Technology (NIT), Rourkela, Odisha, 769008, India
| | - Mandakini Singh
- Department of Life Science, National Institute of Technology (NIT), Rourkela, Odisha, 769008, India
| | | | - Subhadra Kumari
- Department of Life Science, National Institute of Technology (NIT), Rourkela, Odisha, 769008, India
| | - Srinivasan Muthuswamy
- Department of Life Science, National Institute of Technology (NIT), Rourkela, Odisha, 769008, India
| | - Ajay Kumar
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| | - Santosh Kumar
- Department of Life Science, National Institute of Technology (NIT), Rourkela, Odisha, 769008, India.
| |
Collapse
|
10
|
Brown JS. Comparison of Oncogenes, Tumor Suppressors, and MicroRNAs Between Schizophrenia and Glioma: The Balance of Power. Neurosci Biobehav Rev 2023; 151:105206. [PMID: 37178944 DOI: 10.1016/j.neubiorev.2023.105206] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 04/25/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023]
Abstract
The risk of cancer in schizophrenia has been controversial. Confounders of the issue are cigarette smoking in schizophrenia, and antiproliferative effects of antipsychotic medications. The author has previously suggested comparison of a specific cancer like glioma to schizophrenia might help determine a more accurate relationship between cancer and schizophrenia. To accomplish this goal, the author performed three comparisons of data; the first a comparison of conventional tumor suppressors and oncogenes between schizophrenia and cancer including glioma. This comparison determined schizophrenia has both tumor-suppressive and tumor-promoting characteristics. A second, larger comparison between brain-expressed microRNAs in schizophrenia with their expression in glioma was then performed. This identified a core carcinogenic group of miRNAs in schizophrenia offset by a larger group of tumor-suppressive miRNAs. This proposed "balance of power" between oncogenes and tumor suppressors could cause neuroinflammation. This was assessed by a third comparison between schizophrenia, glioma and inflammation in asbestos-related lung cancer and mesothelioma (ALRCM). This revealed that schizophrenia shares more oncogenic similarity to ALRCM than glioma.
Collapse
|
11
|
Sano F, Kikushima K, Benner S, Xu L, Kahyo T, Yamasue H, Setou M. Associations between prefrontal PI (16:0/20:4) lipid, TNC mRNA, and APOA1 protein in schizophrenia: A trans-omics analysis in post-mortem brain. Front Psychiatry 2023; 14:1145437. [PMID: 37143779 PMCID: PMC10151580 DOI: 10.3389/fpsyt.2023.1145437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/24/2023] [Indexed: 05/06/2023] Open
Abstract
Background Though various mechanisms have been proposed for the pathophysiology of schizophrenia, the full extent of these mechanisms remains unclear, and little is known about the relationships among them. We carried out trans-omics analyses by comparing the results of the previously reported lipidomics, transcriptomics, and proteomics analyses; all of these studies used common post-mortem brain samples. Methods We collected the data from three aforementioned omics studies on 6 common post-mortem samples (3 schizophrenia patients and 3 controls), and analyzed them as a whole group sample. Three correlation analyses were performed for each of the two of three omics studies in these samples. In order to discuss the strength of the correlations in a limited sample size, the p-values of each correlation coefficient were confirmed using the Student's t-test. In addition, partial correlation analysis was also performed for some correlations, to verify the strength of the impact of each factor on the correlations. Results The following three factors were strongly correlated with each other: the lipid level of phosphatidylinositol (PI) (16:0/20:4), the amount of TNC mRNA, and the quantitative signal intensity of APOA1 protein. PI (16:0/20:4) and TNC showed a positive correlation, while PI (16:0/20:4) and APOA1, and TNC and APOA1 showed negative correlations. All of these correlations reached at p < 0.01. PI (16:0/20:4) and TNC were decreased in the prefrontal cortex of schizophrenia samples, while APOA1 was increased. Partial correlation analyses among them suggested that PI (16:0/20:4) and TNC have no direct correlation, but their relationships are mediated by APOA1. Conclusion The current results suggest that these three factors may provide new clues to elucidate the relationships among the candidate mechanisms of schizophrenia, and support the potential of trans-omics analyses as a new analytical method.
Collapse
Affiliation(s)
- Fumito Sano
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kenji Kikushima
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
- International Mass Imaging Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Integrative Anatomy, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Seico Benner
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Lili Xu
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tomoaki Kahyo
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hidenori Yamasue
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Mitsutoshi Setou
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
- International Mass Imaging Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Systems Molecular Anatomy, Institute for Medical Photonics Research, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
12
|
Identification of potentially functional modules and diagnostic genes related to amyotrophic lateral sclerosis based on the WGCNA and LASSO algorithms. Sci Rep 2022; 12:20144. [PMID: 36418457 PMCID: PMC9684499 DOI: 10.1038/s41598-022-24306-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 11/14/2022] [Indexed: 11/24/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a genetically and phenotypically heterogeneous disease results in the loss of motor neurons. Mounting information points to involvement of other systems including cognitive impairment. However, neither the valid biomarker for diagnosis nor effective therapeutic intervention is available for ALS. The present study is aimed at identifying potentially genetic biomarker that improves the diagnosis and treatment of ALS patients based on the data of the Gene Expression Omnibus. We retrieved datasets and conducted a weighted gene co-expression network analysis (WGCNA) to identify ALS-related co-expression genes. Functional enrichment analysis was performed to determine the features and pathways of the main modules. We then constructed an ALS-related model using the least absolute shrinkage and selection operator (LASSO) regression analysis and verified the model by the receiver operating characteristic (ROC) curve. Besides we screened the non-preserved gene modules in FTD and ALS-mimic disorders to distinct ALS-related genes from disorders with overlapping genes and features. Altogether, 4198 common genes between datasets with the most variation were analyzed and 16 distinct modules were identified through WGCNA. Blue module had the most correlation with ALS and functionally enriched in pathways of neurodegeneration-multiple diseases', 'amyotrophic lateral sclerosis', and 'endocytosis' KEGG terms. Further, some of other modules related to ALS were enriched in 'autophagy' and 'amyotrophic lateral sclerosis'. The 30 top of hub genes were recruited to a LASSO regression model and 5 genes (BCLAF1, GNA13, ARL6IP5, ARGLU1, and YPEL5) were identified as potentially diagnostic ALS biomarkers with validating of the ROC curve and AUC value.
Collapse
|
13
|
Jeong Y, Bae HJ, Park K, Bae HJ, Yang X, Cho YJ, Jung SY, Jang DS, Ryu JH. 4-Methoxycinnamic acid attenuates schizophrenia-like behaviors induced by MK-801 in mice. JOURNAL OF ETHNOPHARMACOLOGY 2022; 285:114864. [PMID: 34822958 DOI: 10.1016/j.jep.2021.114864] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/13/2021] [Accepted: 11/21/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Scrophularia buergeriana has been used for traditional medicine as an agent for reducing heat in the blood and for nourishing kidney 'Yin'. Therefore, S. buergeriana might be a potential treatment for mental illness, especially schizophrenia, which may be attenuated by supplying kidney Yin and reducing blood heat. In a pilot study, we found that S. buergeriana alleviated sensorimotor gating dysfunction induced by MK-801. AIM OF THE STUDY In the present study, we attempted to reveal the active component(s) of S. buergeriana as a candidate for treating sensorimotor gating dysfunction, and we identified 4-methoxycinnamic acid. We explored whether 4-methoxycinnamic acid could affect schizophrenia-like behaviors induced by hypofunction of the glutamatergic neurotransmitter system. MATERIALS AND METHODS Mice were treated with 4-methoxycinnamic acid (3, 10, or 30 mg/kg, i.g.) under MK-801-induced schizophrenia-like conditions. The effect of 4-methoxycinnamic acid on schizophrenia-like behaviors were explored using several behavioral tasks. We also used Western blotting to investigate which signaling pathway(s) is involved in the pharmacological activities of 4-methoxycinnamic acid. RESULTS 4-Methoxycinnamic acid ameliorated MK-801-induced prepulse inhibition deficits, social interaction disorders and cognitive impairment by regulating the phosphorylation levels of PI3K, Akt and GSK-3β signaling in the prefrontal cortex. And there were no adverse effects in terms of catalepsy and motor coordination impairments. CONCLUSION Collectively, 4-methoxycinnamic acid would be a potential candidate for treating schizophrenia with fewer adverse effects, especially the negative symptoms and cognitive dysfunctions.
Collapse
Affiliation(s)
- Yongwoo Jeong
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Ho Jung Bae
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Keontae Park
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Hyo Jeoung Bae
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Xingquan Yang
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Young-Jin Cho
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Seo Yun Jung
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Dae Sik Jang
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Jong Hoon Ryu
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea; Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
14
|
Afia AB, Vila È, MacDowell KS, Ormazabal A, Leza JC, Haro JM, Artuch R, Ramos B, Garcia-Bueno B. Kynurenine pathway in post-mortem prefrontal cortex and cerebellum in schizophrenia: relationship with monoamines and symptomatology. J Neuroinflammation 2021; 18:198. [PMID: 34511126 PMCID: PMC8436477 DOI: 10.1186/s12974-021-02260-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 08/30/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The cortico-cerebellar-thalamic-cortical circuit has been implicated in the emergence of psychotic symptoms in schizophrenia (SZ). The kynurenine pathway (KP) has been linked to alterations in glutamatergic and monoaminergic neurotransmission and to SZ symptomatology through the production of the metabolites quinolinic acid (QA) and kynurenic acid (KYNA). METHODS This work describes alterations in KP in the post-mortem prefrontal cortex (PFC) and cerebellum (CB) of 15 chronic SZ patients and 14 control subjects in PFC and 13 control subjects in CB using immunoblot for protein levels and ELISA for interleukins and QA and KYNA determinations. Monoamine metabolites were analysed by high-performance liquid chromatography and SZ symptomatology was assessed by Positive and Negative Syndrome Scale (PANSS). The association of KP with inflammatory mediators, monoamine metabolism and SZ symptomatology was explored. RESULTS In the PFC, the presence of the anti-inflammatory cytokine IL-10 together with IDO2 and KATII enzymes decreased in SZ, while TDO and KMO enzyme expression increased. A network interaction analysis showed that in the PFC IL-10 was coupled to the QA branch of the kynurenine pathway (TDO-KMO-QA), whereas IL-10 associated with KMO in CB. KYNA in the CB inversely correlated with negative and general PANSS psychopathology. Although there were no changes in monoamine metabolite content in the PFC in SZ, a network interaction analysis showed associations between dopamine and methoxyhydroxyphenylglycol degradation metabolite. Direct correlations were found between general PANSS psychopathology and the serotonin degradation metabolite, 5-hydroxyindoleacetic acid. Interestingly, KYNA in the CB inversely correlated with 5-hydroxyindoleacetic acid in the PFC. CONCLUSIONS Thus, this work found alterations in KP in two brain areas belonging to the cortico-cerebellar-thalamic-cortical circuit associated with SZ symptomatology, with a possible impact across areas in 5-HT degradation.
Collapse
Affiliation(s)
- Amira Ben Afia
- Laboratory of Genetics, Biodiversity and Bioresource Valorization, Higher Institute of Biotechnology of Monastir, University of Monastir, Monastir, Tunisia
| | - Èlia Vila
- Psiquiatria Molecular, Parc Sanitari Sant Joan de Déu, Institut de Recerca Sant Joan de Déu, Dr. Antoni Pujadas, 42, 08830 Sant Boi de Llobregat, Barcelona, Spain
| | - Karina S MacDowell
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Instituto Universitario de Investigación en Neuroquímica UCM, Avda. Complutense s/n, 28040, Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM (Biomedical Network Research Center of Mental Health), Ministry of Economy, Industry and Competitiveness Institute of Health Carlos III, Madrid, Spain
| | - Aida Ormazabal
- Clinical Chemistry Department, Institut de recerca Sant Joan de Déu and CIBERER-ISCIII, Passeig Sant Joan de Déu, 2. 08950, Esplugues de Llobregat, Barcelona, Spain
| | - Juan C Leza
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Instituto Universitario de Investigación en Neuroquímica UCM, Avda. Complutense s/n, 28040, Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM (Biomedical Network Research Center of Mental Health), Ministry of Economy, Industry and Competitiveness Institute of Health Carlos III, Madrid, Spain
| | - Josep M Haro
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM (Biomedical Network Research Center of Mental Health), Ministry of Economy, Industry and Competitiveness Institute of Health Carlos III, Madrid, Spain.,Parc Sanitari Sant Joan de Déu, Dr. Antoni Pujadas, 42, 08830 Sant Boi de Llobregat, Barcelona, Spain
| | - Rafael Artuch
- Clinical Chemistry Department, Institut de recerca Sant Joan de Déu and CIBERER-ISCIII, Passeig Sant Joan de Déu, 2. 08950, Esplugues de Llobregat, Barcelona, Spain
| | - Belén Ramos
- Psiquiatria Molecular, Parc Sanitari Sant Joan de Déu, Institut de Recerca Sant Joan de Déu, Dr. Antoni Pujadas, 42, 08830 Sant Boi de Llobregat, Barcelona, Spain. .,Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM (Biomedical Network Research Center of Mental Health), Ministry of Economy, Industry and Competitiveness Institute of Health Carlos III, Madrid, Spain. .,Dept. de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain.
| | - Borja Garcia-Bueno
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Instituto Universitario de Investigación en Neuroquímica UCM, Avda. Complutense s/n, 28040, Madrid, Spain. .,Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM (Biomedical Network Research Center of Mental Health), Ministry of Economy, Industry and Competitiveness Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
15
|
Gebicke-Haerter PJ, Leonardi-Essmann F, Haerter JO, Rossner MJ, Falkai P, Schmitt A, Raabe FJ. Differential gene regulation in the anterior cingulate cortex and superior temporal cortex in schizophrenia: A molecular network approach. Schizophr Res 2021; 232:1-10. [PMID: 34004381 DOI: 10.1016/j.schres.2021.04.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 04/01/2021] [Accepted: 04/25/2021] [Indexed: 12/17/2022]
Abstract
The closely connected anterior cingulate cortex (ACC) and superior temporal cortex (STC) are important for higher cognitive functions. Both brain regions are disturbed in schizophrenia, i.e., functional and structural alterations have been reported. This postmortem investigation in brains from patients with schizophrenia and controls compared gene expression in the left ACC and left STC. Most differentially expressed genes were unique to each brain region, but some clusters of genes were equally dysregulated in both, giving rise to a more general disease-specific pattern of gene regulation. The data was used to construct a molecular network of the genes identically expressed in both regions as primary nodes and the metabolically connected genes as secondary nodes. The network analysis identified downregulated clusters of immune-associated gene products and upregulated clusters belonging to the ubiquitin-proteasome system. These findings could help to identify new potential therapeutic targets for future approaches.
Collapse
Affiliation(s)
- Peter J Gebicke-Haerter
- Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Av. Independencia 1027, Santiago, Chile; Institute of Psychopharmacology, Central Institute of Mental Health, Faculty of Medicine, University of Heidelberg, J 5, 68159 Mannheim, Germany
| | - Fernando Leonardi-Essmann
- Institute of Psychopharmacology, Central Institute of Mental Health, Faculty of Medicine, University of Heidelberg, J 5, 68159 Mannheim, Germany
| | - Jan O Haerter
- Niels Bohr Institute, University of Copenhagen, Copenhagen, Denmark
| | - Moritz J Rossner
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Nußbaumstrasse 7, 80336 Munich, Germany
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Nußbaumstrasse 7, 80336 Munich, Germany
| | - Andrea Schmitt
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Nußbaumstrasse 7, 80336 Munich, Germany; Laboratory of Neuroscience (LIM27), Institute of Psychiatry, University of Sao Paulo, Rua Dr. Ovidio Pires de Campos 785, 05453-010 São Paulo, SP, Brazil.
| | - Florian J Raabe
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Nußbaumstrasse 7, 80336 Munich, Germany; International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Kraepelinstr. 2-10, 80804 Munich, Germany
| |
Collapse
|
16
|
Santa Cruz EC, Zandonadi FDS, Fontes W, Sussulini A. A pilot study indicating the dysregulation of the complement and coagulation cascades in treated schizophrenia and bipolar disorder patients. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2021; 1869:140657. [PMID: 33839315 DOI: 10.1016/j.bbapap.2021.140657] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/15/2022]
Abstract
A better understanding of the proteome profile after bipolar disorder (BD) and schizophrenia (SCZ) treatment, besides monitoring disease progression, may assist on the development of novel therapeutic strategies with the ability to reduce or control possible side effects. In this pilot study, proteomics analysis employing nano liquid chromatography coupled to mass spectrometry (nLC-MS) and bioinformatic tools were applied to identify differentially abundant proteins in serum of treated BD and SCZ patients. In total, 10 BD patients, 10 SCZ patients, and 14 healthy controls (HC) were included in this study. 24 serum proteins were significantly altered (p < 0.05) in BD and SCZ treated patients and, considering log2FC > 0.58, 8 proteins presented lower abundance in the BD group, while 7 proteins presented higher abundance and 2 lower abundance in SCZ group when compared against HC. Bioinformatics analysis based on these 24 proteins indicated two main altered pathways previously described in the literature; furthermore, it revealed that opposite abundances of the complement and coagulation cascades were the most significant biological processes involved in these pathologies. Moreover, we describe disease-related proteins and pathways associations suggesting the necessity of clinical follow-up improvement besides treatment, as a precaution or safety measure, along with the disease progression. Further biological validation and investigations are required to define whether there is a correlation between complement and coagulation cascade expression for BD and SCZ and cardiovascular diseases.
Collapse
Affiliation(s)
- Elisa Castañeda Santa Cruz
- Laboratory of Bioanalytics and Integrated Omics (LaBIOmics), Department of Analytical Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), 13083-970 Campinas, SP, Brazil
| | - Flávia da Silva Zandonadi
- Laboratory of Bioanalytics and Integrated Omics (LaBIOmics), Department of Analytical Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), 13083-970 Campinas, SP, Brazil
| | - Wagner Fontes
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasilia (UnB), 70910-900 Brasilia, DF, Brazil
| | - Alessandra Sussulini
- Laboratory of Bioanalytics and Integrated Omics (LaBIOmics), Department of Analytical Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), 13083-970 Campinas, SP, Brazil; National Institute of Science and Technology for Bioanalytics - INCTBio, Institute of Chemistry, University of Campinas (UNICAMP), 13083-970 Campinas, SP, Brazil.
| |
Collapse
|
17
|
Ito S, Lee W, Park JE, Yasunaga M, Mori A, Ohtsuki S, Sugiyama Y. Transient, Tunable Expression of NTCP and BSEP in MDCKII Cells for Kinetic Delineation of the Rate-Determining Process and Inhibitory Effects of Rifampicin in Hepatobiliary Transport of Taurocholate. J Pharm Sci 2020; 110:365-375. [PMID: 33159914 DOI: 10.1016/j.xphs.2020.10.064] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/14/2020] [Accepted: 10/30/2020] [Indexed: 11/16/2022]
Abstract
In predicting the hepatic elimination of compounds, the extended clearance concept has proven useful. Yet, its experimental proof was scarce partly due to the lack of models with the controlled expression of transporters. Here, the uptake and efflux transporters [NTCP (SLC10A1) and BSEP (ABCB11), respectively] were doubly and transiently expressed in MDCKII cells by electroporation-based transfection (with the BSEP plasmid amount varied and with the NTCP plasmid fixed), achieving the activity levels of NTCP and BSEP comparable to those of sandwich cultured human hepatocytes. The biliary excretion clearance for taurocholate increased proportionally to the BSEP expression level. Under the same conditions, the basal-to-apical transcellular clearance of taurocholate displayed an initial increase, and a subsequent plateau, indicating that the basolateral uptake of taurocholate became rate-limiting. The doubly transfected MDCKII cells were also used to kinetically analyze the inhibitory effects of rifampicin on BSEP and NTCP. The obtained results showed a bell-shaped profile for cell-to-medium concentration ratios over a range of rifampicin concentrations, which were quantitatively captured by kinetic modeling based on the extended clearance concept. The present study highlights the utility of the transient, tunable transporter expression system in delineating the rate-determining process and providing mechanistic insights into intracellular substrate accumulation.
Collapse
Affiliation(s)
- Sumito Ito
- GenoMembrane Co., Ltd, 2-3-18 Namamugi, Tsurumi-ku, Yokohama, Kanagawa 230-0052, Japan.
| | - Wooin Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Ji Eun Park
- Sugiyama Laboratory, RIKEN Baton Zone Program, RIKEN Cluster for Science, RIKEN, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan; Pharmacokinetics, Dynamics and Metabolism, Translational Medicine and Early Development, R&D, Sanofi K.K., 3 Chome-20-2, Nishishinjuku, Tokyo 160-0023, Japan
| | - Masa Yasunaga
- GenoMembrane Co., Ltd, 2-3-18 Namamugi, Tsurumi-ku, Yokohama, Kanagawa 230-0052, Japan
| | - Ayano Mori
- Department of Pharmaceutical Microbiology, School of Pharmacy, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Sumio Ohtsuki
- Department of Pharmaceutical Microbiology, School of Pharmacy, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Baton Zone Program, RIKEN Cluster for Science, RIKEN, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| |
Collapse
|
18
|
Clifton NE, Thomas KL, Wilkinson LS, Hall J, Trent S. FMRP and CYFIP1 at the Synapse and Their Role in Psychiatric Vulnerability. Complex Psychiatry 2020; 6:5-19. [PMID: 34883502 PMCID: PMC7673588 DOI: 10.1159/000506858] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 02/27/2020] [Indexed: 12/23/2022] Open
Abstract
There is increasing awareness of the role genetic risk variants have in mediating vulnerability to psychiatric disorders such as schizophrenia and autism. Many of these risk variants encode synaptic proteins, influencing biological pathways of the postsynaptic density and, ultimately, synaptic plasticity. Fragile-X mental retardation 1 (FMR1) and cytoplasmic fragile-X mental retardation protein (FMRP)-interacting protein 1 (CYFIP1) contain 2 such examples of highly penetrant risk variants and encode synaptic proteins with shared functional significance. In this review, we discuss the biological actions of FMRP and CYFIP1, including their regulation of (i) protein synthesis and specifically FMRP targets, (ii) dendritic and spine morphology, and (iii) forms of synaptic plasticity such as long-term depression. We draw upon a range of preclinical studies that have used genetic dosage models of FMR1 and CYFIP1 to determine their biological function. In parallel, we discuss how clinical studies of fragile X syndrome or 15q11.2 deletion patients have informed our understanding of FMRP and CYFIP1, and highlight the latest psychiatric genomic findings that continue to implicate FMRP and CYFIP1. Lastly, we assess the current limitations in our understanding of FMRP and CYFIP1 biology and how they must be addressed before mechanism-led therapeutic strategies can be developed for psychiatric disorders.
Collapse
Affiliation(s)
- Nicholas E. Clifton
- Neuroscience & Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Kerrie L. Thomas
- Neuroscience & Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Lawrence S. Wilkinson
- Neuroscience & Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
- School of Psychology, Cardiff University, Cardiff, United Kingdom
| | - Jeremy Hall
- Neuroscience & Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, United Kingdom
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Simon Trent
- Neuroscience & Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
- School of Life Sciences, Faculty of Natural Sciences, Keele University, Keele, United Kingdom
| |
Collapse
|
19
|
Masuda T, Mori A, Ito S, Ohtsuki S. Quantitative and targeted proteomics-based identification and validation of drug efficacy biomarkers. Drug Metab Pharmacokinet 2020; 36:100361. [PMID: 33097418 DOI: 10.1016/j.dmpk.2020.09.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/25/2020] [Accepted: 09/28/2020] [Indexed: 12/25/2022]
Abstract
Proteomics refers to the large-scale study of proteins, providing comprehensive and quantitative information on proteins in tissue, blood, and cell samples. In many studies, proteomics utilizes liquid chromatography-mass spectrometry. Proteomics has developed from a qualitative methodology of protein identification to a quantitative methodology for comparing protein expression, and it is currently classified into two distinct methodologies: quantitative and targeted proteomics. Quantitative proteomics comprehensively identifies proteins in samples, providing quantitative information on large-scale comparative profiles of protein expression. Targeted proteomics simultaneously quantifies only target proteins with high sensitivity and specificity. Therefore, in biomarker research, quantitative proteomics is used for the identification of biomarker candidates, and targeted proteomics is used for the validation of biomarkers. Understanding the specific characteristics of each method is important for conducting appropriate proteomics studies. In this review, we introduced the different characteristics and applications of quantitative and targeted proteomics, and then discussed the results of our recent proteomics studies that focused on the identification and validation of biomarkers of drug efficacy. These findings may enable us to predict the outcomes of cancer therapy and drug-drug interactions with antibiotics through changes in the intestinal microbiome.
Collapse
Affiliation(s)
- Takeshi Masuda
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Department of Pharmaceutical Microbiology, School of Pharmacy, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan.
| | - Ayano Mori
- Department of Pharmaceutical Microbiology, School of Pharmacy, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan.
| | - Shingo Ito
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Department of Pharmaceutical Microbiology, School of Pharmacy, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan.
| | - Sumio Ohtsuki
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Department of Pharmaceutical Microbiology, School of Pharmacy, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan.
| |
Collapse
|
20
|
An N, Bassil K, Al Jowf GI, Steinbusch HWM, Rothermel M, de Nijs L, Rutten BPF. Dual-specificity phosphatases in mental and neurological disorders. Prog Neurobiol 2020; 198:101906. [PMID: 32905807 DOI: 10.1016/j.pneurobio.2020.101906] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 08/26/2020] [Accepted: 09/01/2020] [Indexed: 01/01/2023]
Abstract
The dual-specificity phosphatase (DUSP) family includes a heterogeneous group of protein phosphatases that dephosphorylate both phospho-tyrosine and phospho-serine/phospho-threonine residues within a single substrate. These protein phosphatases have many substrates and modulate diverse neural functions, such as neurogenesis, differentiation, and apoptosis. DUSP genes have furthermore been associated with mental disorders such as depression and neurological disorders such as Alzheimer's disease. Herein, we review the current literature on the DUSP family of genes concerning mental and neurological disorders. This review i) outlines the structure and general functions of DUSP genes, and ii) overviews the literature on DUSP genes concerning mental and neurological disorders, including model systems, while furthermore providing perspectives for future research.
Collapse
Affiliation(s)
- Ning An
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands; European Graduate School of Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Katherine Bassil
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands; European Graduate School of Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Ghazi I Al Jowf
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands; College of Applied Medical Sciences, Department of Public Health, King Faisal University, Al-Ahsa, Saudi Arabia; European Graduate School of Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Harry W M Steinbusch
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands; European Graduate School of Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Markus Rothermel
- European Graduate School of Neuroscience, Maastricht University, Maastricht, the Netherlands; Department of Chemosensation - AG Neuromodulation, RWTH Aachen University, Aachen, Germany
| | - Laurence de Nijs
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands; European Graduate School of Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Bart P F Rutten
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands; European Graduate School of Neuroscience, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
21
|
Reis-de-Oliveira G, Zuccoli GS, Fioramonte M, Schimitt A, Falkai P, Almeida V, Martins-de-Souza D. Digging deeper in the proteome of different regions from schizophrenia brains. J Proteomics 2020; 223:103814. [PMID: 32389842 DOI: 10.1016/j.jprot.2020.103814] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/21/2020] [Accepted: 05/05/2020] [Indexed: 02/07/2023]
Abstract
Schizophrenia is a psychiatric disorder that affects 21 million people worldwide. Despite several studies having been shown that some brain regions may play a critical role in the pathophysiology of schizophrenia, the molecular basis to explain this diversity is still lacking. The cerebellum (CER), caudate nucleus (CAU), and posterior cingulate cortex (PCC) are areas associated with negative and cognitive symptoms in schizophrenia. In this study, we performed shotgun proteomics of the aforementioned brain regions, collected postmortem from patients with schizophrenia and compared with the mentally healthy group. In addition, we performed a proteomic analysis of nuclear and mitochondrial fractions of these same regions. Our results presented 106, 727 and 135 differentially regulated proteins in the CAU, PCC, and CER, respectively. Pathway enrichment analysis revealed dysfunctions associated with synaptic processes in the CAU, transport in the CER, and in energy metabolism in the PCC. In all brain areas, we found that proteins related to oligodendrocytes and the metabolic processes were dysregulated in schizophrenia. SIGNIFICANCE: Schizophrenia is a complex and heterogeneous psychiatric disorder. Despite much research having been done to increase the knowledge about the role of each region in the pathophysiology of this disorder, the molecular mechanisms underlying it are still lacking. We performed shotgun proteomics in the postmortem cerebellum (CER), caudate nucleus (CAU) and posterior cingulate cortex (PCC) from patients with schizophrenia and compared with healthy controls. Our findings suggest that each aforementioned region presents dysregulations in specific molecular pathways, such as energy metabolism in the PCC, transport in the CER, and synaptic process in the CAU. Additionally, these areas presented dysfunctions in oligodendrocytes and metabolic processes. Our results may highlight future directions for the development of novel clinical approaches for specific therapeutic targets.
Collapse
Affiliation(s)
- G Reis-de-Oliveira
- Lab of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - G S Zuccoli
- Lab of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - M Fioramonte
- Lab of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - A Schimitt
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University (LMU), Munich, Germany; Laboratory of Neurosciences (LIM-27), Institute of Psychiatry, University of São Paulo, São Paulo, Brazil
| | - P Falkai
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University (LMU), Munich, Germany
| | - V Almeida
- Lab of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - D Martins-de-Souza
- Lab of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil; Experimental Medicine Research Cluster (EMRC), University of Campinas, Campinas, Brazil; D'Or Institute for Research and Education (IDOR), São Paulo, Brazil; Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBION), Conselho Nacional de Desenvolvimento Científico e Tecnológico, São Paulo, Brazil.
| |
Collapse
|
22
|
Sultana R, Shrestha A, Lee CC, Ogundele OM. Disc1 Carrier Mice Exhibit Alterations in Neural pIGF-1Rβ and Related Kinase Expression. Front Cell Neurosci 2020; 14:94. [PMID: 32431597 PMCID: PMC7214624 DOI: 10.3389/fncel.2020.00094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 03/30/2020] [Indexed: 12/20/2022] Open
Abstract
Mutation of the disc1 gene underlies a broad range of developmental neuropsychiatric defects, including schizophrenia, depression, and bipolar disorder. The pathophysiological phenotypes linked with disc1 mutation are due to the truncation of the DISC1 primary protein structure. This leads to a defective post-synaptic scaffolding and kinase—GSK3β and Erk1/2—signaling. As a result, synaptic function and maintenance are significantly impaired in the disc1 mutant brain. Among several other pathways, GSK3β and Erk1/2 are involved in insulin-like growth factor 1 receptor (IGF-1Rβ) kinase signaling. Although disc1 mutation alters these kinases, it is unclear if the mutation impacts IGF-1R expression and activity in the brain. Here, we demonstrate that the expression of active IGF-1Rβ (pIGF-1Rβ) is altered in the hippocampus and prefrontal cortex (PFC) of disc1 mutant mice and vary with the dose of the mutation (homozygous and heterozygous). The expression of pIGF-1Rβ decreased significantly in 129S (hom, disc1−/−) brains. In contrast, 129S:B6 (het, disc1+/−) brains were characterized by an increase in pIGF-1Rβ when compared with the C57BL/6 (disc1+/+) level. The decrease in pIGF-1Rβ level for the 129S brains was accompanied by the loss of Akt activity (S473 pAkt) and decreased Ser9 phosphorylation of GSK3β (increased basal GSK3β). Additionally, hippocampal and cortical pErk1/2 activity increased in the 129S hippocampus and cortex. Although 129S:B6 recorded alterations in pIGF-1Rβ-pAkt-GSK3β (like 129S), there was no observable change in pErk1/2 activity for the heterozygote (disc1+/−) mutant. In addition to GSK3β inhibition, we conclude that pIGF-1R, pAkt, and pErk1/2 are potential targets in disc1−/− mutant brain. On the other hand, pIGF-1R and pAkt can be further explored in disc1+/− brain.
Collapse
Affiliation(s)
- Razia Sultana
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Amita Shrestha
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Charles C Lee
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Olalekan M Ogundele
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| |
Collapse
|
23
|
Petralia MC, Ciurleo R, Saraceno A, Pennisi M, Basile MS, Fagone P, Bramanti P, Nicoletti F, Cavalli E. Meta-Analysis of Transcriptomic Data of Dorsolateral Prefrontal Cortex and of Peripheral Blood Mononuclear Cells Identifies Altered Pathways in Schizophrenia. Genes (Basel) 2020; 11:genes11040390. [PMID: 32260267 PMCID: PMC7230488 DOI: 10.3390/genes11040390] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/13/2020] [Accepted: 04/02/2020] [Indexed: 12/11/2022] Open
Abstract
Schizophrenia (SCZ) is a psychiatric disorder characterized by both positive and negative symptoms, including cognitive dysfunction, decline in motivation, delusion and hallucinations. Antipsychotic agents are currently the standard of care treatment for SCZ. However, only about one-third of SCZ patients respond to antipsychotic medications. In the current study, we have performed a meta-analysis of publicly available whole-genome expression datasets on Brodmann area 46 of the brain dorsolateral prefrontal cortex in order to prioritize potential pathways underlying SCZ pathology. Moreover, we have evaluated whether the differentially expressed genes in SCZ belong to specific subsets of cell types. Finally, a cross-tissue comparison at both the gene and functional level was performed by analyzing the transcriptomic pattern of peripheral blood mononuclear cells of SCZ patients. Our study identified a robust disease-specific set of dysfunctional biological pathways characterizing SCZ patients that could in the future be exploited as potential therapeutic targets.
Collapse
Affiliation(s)
| | - Rosella Ciurleo
- IRCCS Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124 Messina, Italy; (R.C.); (P.B.)
| | - Andrea Saraceno
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.S.); (M.P.); (M.S.B.); (F.N.); (E.C.)
| | - Manuela Pennisi
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.S.); (M.P.); (M.S.B.); (F.N.); (E.C.)
| | - Maria Sofia Basile
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.S.); (M.P.); (M.S.B.); (F.N.); (E.C.)
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.S.); (M.P.); (M.S.B.); (F.N.); (E.C.)
- Correspondence: ; Tel.: +39-095-4781284
| | - Placido Bramanti
- IRCCS Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124 Messina, Italy; (R.C.); (P.B.)
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.S.); (M.P.); (M.S.B.); (F.N.); (E.C.)
| | - Eugenio Cavalli
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.S.); (M.P.); (M.S.B.); (F.N.); (E.C.)
| |
Collapse
|
24
|
Nagaoka A, Kunii Y, Hino M, Izumi R, Nagashima C, Takeshima A, Sainouchi M, Nawa H, Kakita A, Yabe H. ALDH4A1 expression levels are elevated in postmortem brains of patients with schizophrenia and are associated with genetic variants in enzymes related to proline metabolism. J Psychiatr Res 2020; 123:119-127. [PMID: 32065947 DOI: 10.1016/j.jpsychires.2020.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 01/28/2020] [Accepted: 02/03/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND The molecular mechanisms underlying schizophrenia remain largely unclear, and we recently identified multiple proteins significantly altered in the postmortem prefrontal cortex (PFC) of schizophrenia patients amongst which aldehyde dehydrogenase 4 family member A1 (ALDH4A1) was especially elevated. In this study, we aimed to investigate the expression of ALDH4A1 in the PFC and superior temporal gyrus (STG) and to elucidate functional correlations between schizophrenia risk alleles and molecular expression profiles in the postmortem brains of patients with schizophrenia. METHODS The levels of ALDH4A1 protein expression in the PFC and STG in postmortem brains from 24 patients with schizophrenia, 8 patients with bipolar disorder, and 32 controls were assessed using enzyme-linked immunosorbent assay. Moreover, we explored the associations between ALDH4A1 expression and genetic variants in enzymes associated with proline metabolism, including ALDH4A1 (schizophrenia [n = 22], bipolar disorder [n = 6], controls [n = 11]). RESULTS ALDH4A1 levels were significantly elevated in both the PFC and STG in patients with schizophrenia and tended to elevate in patients with bipolar disorder. Furthermore, ALDH4A1 expression levels in the PFC were significantly associated with the following three single-nucleotide polymorphisms: rs10882639, rs33823, rs153508. We also found partial coexpression of ALDH4A1 in mitochondria in a subset of putative astrocytes of postmortem brain. LIMITATIONS Our study population was relatively small, particularly for a genetic study. CONCLUSION These findings indicate that altered expression of ALDH4A1 may reflect the potential molecular mechanisms underlying the pathogenesis of schizophrenia and bipolar disorder, and may aid in the development of novel drug therapies.
Collapse
Affiliation(s)
- Atsuko Nagaoka
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, 960-1295, Fukushima, Japan
| | - Yasuto Kunii
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, 960-1295, Fukushima, Japan; Department of Psychiatry, Aizu Medical Center, Fukushima Medical University, 969-3492, Fukushima, Japan.
| | - Mizuki Hino
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, 960-1295, Fukushima, Japan
| | - Ryuta Izumi
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, 960-1295, Fukushima, Japan
| | - Chisato Nagashima
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, 960-1295, Fukushima, Japan
| | - Akari Takeshima
- Department of Pathology, Brain Research Institute, Niigata University, 951-8585, Niigata, Japan
| | - Makoto Sainouchi
- Department of Pathology, Brain Research Institute, Niigata University, 951-8585, Niigata, Japan
| | - Hiroyuki Nawa
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, 951-8585, Niigata, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, 951-8585, Niigata, Japan
| | - Hirooki Yabe
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, 960-1295, Fukushima, Japan
| |
Collapse
|
25
|
Balan S, Toyoshima M, Yoshikawa T. Contribution of induced pluripotent stem cell technologies to the understanding of cellular phenotypes in schizophrenia. Neurobiol Dis 2019; 131:104162. [DOI: 10.1016/j.nbd.2018.04.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 04/23/2018] [Accepted: 04/28/2018] [Indexed: 02/07/2023] Open
|
26
|
Woo YJ, Kanellopoulos AK, Hemati P, Kirschen J, Nebel RA, Wang T, Bagni C, Abrahams BS. Domain-Specific Cognitive Impairments in Humans and Flies With Reduced CYFIP1 Dosage. Biol Psychiatry 2019; 86:306-314. [PMID: 31202490 PMCID: PMC6679746 DOI: 10.1016/j.biopsych.2019.04.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 03/19/2019] [Accepted: 04/03/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND Deletions encompassing a four-gene region on chromosome 15 (BP1-BP2 at 15q11.2), seen at a population frequency of 1 in 500, are associated with increased risk for schizophrenia, epilepsy, and other common neurodevelopmental disorders. However, little is known in terms of how these common deletions impact cognition. METHODS We used a Web-based tool to characterize cognitive function in a novel cohort of adult carriers and their noncarrier family members. Results from 31 carrier and 38 noncarrier parents from 40 families were compared with control data from 6530 individuals who self-registered on the Lumosity platform and opted in to participate in research. We then examined aspects of sensory and cognitive function in flies harboring a mutation in Cyfip, the homologue of one of the genes within the deletion. For the fly studies, 10 or more groups of 50 individuals per genotype were included. RESULTS Our human studies revealed profound deficits in grammatical reasoning, arithmetic reasoning, and working memory in BP1-BP2 deletion carriers. No such deficits were observed in noncarrier spouses. Our fly studies revealed deficits in associative and nonassociative learning despite intact sensory perception. CONCLUSIONS Our results provide new insights into outcomes associated with BP1-BP2 deletions and call for a discussion on how to appropriately communicate these findings to unaffected carriers. Findings also highlight the utility of an online tool in characterizing cognitive function in a geographically distributed population.
Collapse
Affiliation(s)
- Young Jae Woo
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | - Parisa Hemati
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York; Human Genetics Program, Sarah Lawrence College, Yonkers, New York
| | - Jill Kirschen
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Rebecca A Nebel
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York
| | - Tao Wang
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | - Claudia Bagni
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland; Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Brett S Abrahams
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York.
| |
Collapse
|
27
|
Domínguez-Iturza N, Lo AC, Shah D, Armendáriz M, Vannelli A, Mercaldo V, Trusel M, Li KW, Gastaldo D, Santos AR, Callaerts-Vegh Z, D'Hooge R, Mameli M, Van der Linden A, Smit AB, Achsel T, Bagni C. The autism- and schizophrenia-associated protein CYFIP1 regulates bilateral brain connectivity and behaviour. Nat Commun 2019; 10:3454. [PMID: 31371726 PMCID: PMC6672001 DOI: 10.1038/s41467-019-11203-y] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 06/20/2019] [Indexed: 12/26/2022] Open
Abstract
Copy-number variants of the CYFIP1 gene in humans have been linked to autism spectrum disorders (ASD) and schizophrenia (SCZ), two neuropsychiatric disorders characterized by defects in brain connectivity. Here, we show that CYFIP1 plays an important role in brain functional connectivity and callosal functions. We find that Cyfip1-heterozygous mice have reduced functional connectivity and defects in white matter architecture, similar to phenotypes found in patients with ASD, SCZ and other neuropsychiatric disorders. Cyfip1-deficient mice also present decreased myelination in the callosal axons, altered presynaptic function, and impaired bilateral connectivity. Finally, Cyfip1 deficiency leads to abnormalities in motor coordination, sensorimotor gating and sensory perception, which are also known neuropsychiatric disorder-related symptoms. These results show that Cyfip1 haploinsufficiency compromises brain connectivity and function, which might explain its genetic association to neuropsychiatric disorders.
Collapse
Affiliation(s)
- Nuria Domínguez-Iturza
- Department of Fundamental Neurosciences, University of Lausanne, 1005, Lausanne, Switzerland
- Department of Human Genetics KU Leuven, VIB Center for Brain & Disease Research, 3000, Leuven, Belgium
| | - Adrian C Lo
- Department of Fundamental Neurosciences, University of Lausanne, 1005, Lausanne, Switzerland
| | - Disha Shah
- Department of Biomedical Sciences, Bio-Imaging Laboratory, University of Antwerp, 2610, Antwerp, Belgium
- Department of Neuroscience KU Leuven, VIB Center for Brain & Disease Research, 3000, Leuven, Belgium
| | - Marcelo Armendáriz
- Department of Neurosciences, Laboratory of Neuro- and Psychophysiology, KU Leuven, 3000, Leuven, Belgium
| | - Anna Vannelli
- Department of Fundamental Neurosciences, University of Lausanne, 1005, Lausanne, Switzerland
| | - Valentina Mercaldo
- Department of Fundamental Neurosciences, University of Lausanne, 1005, Lausanne, Switzerland
| | - Massimo Trusel
- Department of Fundamental Neurosciences, University of Lausanne, 1005, Lausanne, Switzerland
| | - Ka Wan Li
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, 1081, Amsterdam, The Netherlands
| | - Denise Gastaldo
- Department of Fundamental Neurosciences, University of Lausanne, 1005, Lausanne, Switzerland
| | - Ana Rita Santos
- Department of Human Genetics KU Leuven, VIB Center for Brain & Disease Research, 3000, Leuven, Belgium
- VIB Discovery Sciences, Bioincubator, 3001, Heverlee, Belgium
| | - Zsuzsanna Callaerts-Vegh
- Faculty of Psychology and Educational Sciences, KU Leuven, Laboratory of Biological Psychology, 3000, Leuven, Belgium
| | - Rudi D'Hooge
- Faculty of Psychology and Educational Sciences, KU Leuven, Laboratory of Biological Psychology, 3000, Leuven, Belgium
| | - Manuel Mameli
- Department of Fundamental Neurosciences, University of Lausanne, 1005, Lausanne, Switzerland
| | - Annemie Van der Linden
- Department of Biomedical Sciences, Bio-Imaging Laboratory, University of Antwerp, 2610, Antwerp, Belgium
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, 1081, Amsterdam, The Netherlands
| | - Tilmann Achsel
- Department of Fundamental Neurosciences, University of Lausanne, 1005, Lausanne, Switzerland
- Department of Human Genetics KU Leuven, VIB Center for Brain & Disease Research, 3000, Leuven, Belgium
| | - Claudia Bagni
- Department of Fundamental Neurosciences, University of Lausanne, 1005, Lausanne, Switzerland.
- Department of Human Genetics KU Leuven, VIB Center for Brain & Disease Research, 3000, Leuven, Belgium.
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133, Rome, Italy.
| |
Collapse
|
28
|
Szczurkowska J, Pischedda F, Pinto B, Managò F, Haas CA, Summa M, Bertorelli R, Papaleo F, Schäfer MK, Piccoli G, Cancedda L. NEGR1 and FGFR2 cooperatively regulate cortical development and core behaviours related to autism disorders in mice. Brain 2019; 141:2772-2794. [PMID: 30059965 PMCID: PMC6113639 DOI: 10.1093/brain/awy190] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 06/04/2018] [Indexed: 12/15/2022] Open
Abstract
Autism spectrum disorders are neurodevelopmental conditions with diverse aetiologies, all characterized by common core symptoms such as impaired social skills and communication, as well as repetitive behaviour. Cell adhesion molecules, receptor tyrosine kinases and associated downstream signalling have been strongly implicated in both neurodevelopment and autism spectrum disorders. We found that downregulation of the cell adhesion molecule NEGR1 or the receptor tyrosine kinase fibroblast growth factor receptor 2 (FGFR2) similarly affects neuronal migration and spine density during mouse cortical development in vivo and results in impaired core behaviours related to autism spectrum disorders. Mechanistically, NEGR1 physically interacts with FGFR2 and modulates FGFR2-dependent extracellular signal-regulated kinase (ERK) and protein kinase B (AKT) signalling by decreasing FGFR2 degradation from the plasma membrane. Accordingly, FGFR2 overexpression rescues all defects due to Negr1 knockdown in vivo. Negr1 knockout mice present phenotypes similar to Negr1-downregulated animals. These data indicate that NEGR1 and FGFR2 cooperatively regulate cortical development and suggest a role for defective NEGR1-FGFR2 complex and convergent downstream ERK and AKT signalling in autism spectrum disorders.
Collapse
Affiliation(s)
- Joanna Szczurkowska
- Local Micro-environment and Brain Development Laboratory, Italian Institute of Technology, Genoa, Italy.,Università degli Studi di Genova, Via Balbi, 5, Genoa, Italy
| | - Francesca Pischedda
- Laboratory of Biology of Synapse. Center for Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Bruno Pinto
- Local Micro-environment and Brain Development Laboratory, Italian Institute of Technology, Genoa, Italy.,Bio@SNS, Scuola Normale Superiore, Pisa, Italy
| | - Francesca Managò
- Genetics of Cognition Laboratory, Italian Institute of Technology, Genoa, Italy
| | - Carola A Haas
- Experimental Epilepsy Research, Department of Neurosurgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Maria Summa
- Department of Drug Discovery and Development, Italian Institute of Technology, Genoa, Italy
| | - Rosalia Bertorelli
- Department of Drug Discovery and Development, Italian Institute of Technology, Genoa, Italy
| | - Francesco Papaleo
- Genetics of Cognition Laboratory, Italian Institute of Technology, Genoa, Italy
| | - Michael K Schäfer
- Department of Anesthesiology and Focus Program Translational Neurosciences, University Medical Center of the Johannes Gutenberg-University Mainz, Germany
| | - Giovanni Piccoli
- Laboratory of Biology of Synapse. Center for Integrative Biology (CIBIO), University of Trento, Trento, Italy.,Dulbecco Telethon Institute, Varese Street 16b - 00185 Rome, Italy
| | - Laura Cancedda
- Local Micro-environment and Brain Development Laboratory, Italian Institute of Technology, Genoa, Italy.,Dulbecco Telethon Institute, Varese Street 16b - 00185 Rome, Italy
| |
Collapse
|
29
|
Nanoemulsions in CNS drug delivery: recent developments, impacts and challenges. Drug Discov Today 2019; 24:1104-1115. [PMID: 30914298 DOI: 10.1016/j.drudis.2019.03.021] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 03/03/2019] [Accepted: 03/20/2019] [Indexed: 02/06/2023]
Abstract
Despite enormous efforts, treatment of CNS diseases remains challenging. One of the main issues causing this situation is limited CNS access for the majority of drugs used as part of the therapeutic regimens against life-threatening CNS diseases. Regarding the inarguable position of the nanocarrier systems in neuropharmacokinetic enhancement of the CNS drugs, this review discusses the latest findings on nanoemulsions (NEs) as one of the most promising candidates of this type, to overcome the challenges of CNS drug delivery. Future development of NE-based CNS drug delivery needs to consider so many aspects not only from a physicochemical point of view but also related to the biointerface of these very small droplets before achieving clinical value.
Collapse
|
30
|
Enhanced Molecular Appreciation of Psychiatric Disorders Through High-Dimensionality Data Acquisition and Analytics. Methods Mol Biol 2019; 2011:671-723. [PMID: 31273728 DOI: 10.1007/978-1-4939-9554-7_39] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The initial diagnosis, molecular investigation, treatment, and posttreatment care of major psychiatric disorders (schizophrenia and bipolar depression) are all still significantly hindered by the current inability to define these disorders in an explicit molecular signaling manner. High-dimensionality data analytics, using large datastreams from transcriptomic, proteomic, or metabolomic investigations, will likely advance both the appreciation of the molecular nature of major psychiatric disorders and simultaneously enhance our ability to more efficiently diagnose and treat these debilitating conditions. High-dimensionality data analysis in psychiatric research has been heterogeneous in aims and methods and limited by insufficient sample sizes, poorly defined case definitions, methodological inhomogeneity, and confounding results. All of these issues combine to constrain the conclusions that can be extracted from them. Here, we discuss possibilities for overcoming methodological challenges through the implementation of transcriptomic, proteomic, or metabolomics signatures in psychiatric diagnosis and offer an outlook for future investigations. To fulfill the promise of intelligent high-dimensionality data-based differential diagnosis in mental disease diagnosis and treatment, future research will need large, well-defined cohorts in combination with state-of-the-art technologies.
Collapse
|
31
|
Miao J, Liu L, Yan C, Zhu X, Fan M, Yu P, Ji K, Huang Y, Wang Y, Zhu G. Association between ADORA2A gene polymorphisms and schizophrenia in the North Chinese Han population. Neuropsychiatr Dis Treat 2019; 15:2451-2458. [PMID: 31695381 PMCID: PMC6718062 DOI: 10.2147/ndt.s205014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 07/09/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND A large number of studies have shown a close relationship between ADORA2A and the pathological mechanism of schizophrenia. However, to our knowledge, there has been no studies examining the association between the ADORA2A gene and schizophrenia in Chinese Han population. PURPOSE The objective of this study was to examine the relationship between adenosine A2A receptor (ADORA2A) single nucleotide polymorphisms and schizophrenia in the North Chinese Han population. PATIENTS AND METHODS We detected ADORA2A single nucleotide polymorphisms (SNPs) using polymerase chain reaction-restriction fragment length polymorphism analyses and summarized our results using SPSS statistical software and Haploview in schizophrenia case group (n=398) and healthy control group (n=535). RESULTS The frequency of the CC homozygote genotype of SNP rs2298383T/C were significantly higher in the case than the control group (p=0.005, OR=1.712, 95% CI=1.172-2.502). After linkage disequilibrium analysis, SNPs rs5996696A/C and rs2298383T/C displayed strong linkage disequilibrium. We found that the frequencies of haplotypes TA (χ2=6.268, p=0.0123) and CA (χ2=7.012, p=0.0081) were significantly higher in the case group than in the control group. CONCLUSION In conclusion, SNPs in the ADORA2A gene may be associated with schizophrenia in the northern Chinese Han population.
Collapse
Affiliation(s)
- Junxiao Miao
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Lu Liu
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China.,Department of Psychiatry, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, People's Republic of China
| | - Ci Yan
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Xiaotong Zhu
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Mengqi Fan
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Peitong Yu
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Keming Ji
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Yinglin Huang
- Department of Psychiatry, Shengjing Hospital of China Medical University, Shenyang 110020, People's Republic of China
| | - Yuan Wang
- Department of Psychiatry, Shengjing Hospital of China Medical University, Shenyang 110020, People's Republic of China
| | - Gang Zhu
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| |
Collapse
|
32
|
Involvement of an Orphan Transporter, SLC22A18, in Cell Growth and Drug Resistance of Human Breast Cancer MCF7 Cells. J Pharm Sci 2018; 107:3163-3170. [DOI: 10.1016/j.xphs.2018.08.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 08/06/2018] [Accepted: 08/07/2018] [Indexed: 01/05/2023]
|
33
|
Abdul W, Aliyu SR, Lin L, Sekete M, Chen X, Otieno FJ, Yang T, Lin Y, Norvienyeku J, Wang Z. Family-Four Aldehyde Dehydrogenases Play an Indispensable Role in the Pathogenesis of Magnaporthe oryzae. FRONTIERS IN PLANT SCIENCE 2018; 9:980. [PMID: 30135691 PMCID: PMC6092734 DOI: 10.3389/fpls.2018.00980] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 06/15/2018] [Indexed: 05/22/2023]
Abstract
The oxidative degradation of lipids through lipid peroxidation processes results in the generation of free fatty acid radicals. These free radicals including reactive oxygen species (ROS) serve as a substrate for generating reactive aldehydes. The accumulation of free fatty acid radicals, ROS, and reactive aldehydes in cell compartments beyond physiological threshold levels tends to exert a damaging effect on proximal membranes and distal tissues. Living organisms deploy a wide array of efficient enzymes including superoxide dismutase (SOD), catalase (CAT), peroxidase (POD), and aldehyde dehydrogenases (ALDHs) for scavenging reactive molecules and intermediates produced from membrane lipid peroxidation events. Although the contributions of SOD, CAT, and POD to the pathogenesis of microbial plant pathogens are well known, the influence of ALDH genes on the morphological and infectious development of plant pathogenic microbes is not well understood. In this study, we deployed RNA interference (RNAi) techniques and successfully silenced two putative family-four aldehyde dehydrogenase genes potassium-activated aldehyde dehydrogenase (MoKDCDH) and delta-1-pyrrorine-5-carboxylate dehydrogenase (MoP5CDH) in the rice blast pathogen Magnaporthe oryzae. The results obtained from the phenotypic analysis of individual knock-down strains showed that the RNAi-mediated inactivation of MoKDCDH and MoP5CDH triggered a significant reduction in conidiogenesis and vegetative growth of ΔMokdcdh and ΔMop5cdh strains. We further observed that downregulating the expression of MoKDCDH and MoP5CDH severely compromised the pathogenesis of the rice blast fungus. Also, the disruption of MoKDCDH and MoP5CDH M. oryzae undermined membrane integrity and rendered the mutant strains highly sensitive to membrane stress inducing osmolytes. However, the MoKDCDH and MoP5CDH knock-down strains generated in this study displayed unaltered cell wall integrity and thus suggested that family-four ALDHs play a dispensable role in enforcing cell wall-directed stress tolerance in M. oryzae. From these results, we deduced that family-four ALDHs play a conserved role in fostering membrane integrity in M. oryzae possibly by scavenging reactive aldehydes, fatty acid radicals, and other alcohol derivatives. The observation that downregulating the expression activities of MoKDCDH had a lethal effect on potential mutants further emphasized the need for comprehensive and holistic evaluation of the numerous ALDHs amassed by the rice blast fungus for their possible engagement as suitable targets as antiblast agents.
Collapse
Affiliation(s)
- Waheed Abdul
- Fujian University Key Laboratory for Functional Genomics of Plant Fungal Pathogens, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
- Fujian and Taiwan Joint Center for Ecological Control of Crop Pests, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Sami R. Aliyu
- Fujian and Taiwan Joint Center for Ecological Control of Crop Pests, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Lili Lin
- Fujian and Taiwan Joint Center for Ecological Control of Crop Pests, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Malota Sekete
- Department of Crop Science, Faculty of Agriculture, National University of Lesotho, Lesotho, Southern Africa
| | - Xiaomin Chen
- Fujian and Taiwan Joint Center for Ecological Control of Crop Pests, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Frankline J. Otieno
- Fujian University Key Laboratory for Functional Genomics of Plant Fungal Pathogens, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Tao Yang
- Fujian and Taiwan Joint Center for Ecological Control of Crop Pests, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yahong Lin
- Fujian University Key Laboratory for Functional Genomics of Plant Fungal Pathogens, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Justice Norvienyeku
- Fujian University Key Laboratory for Functional Genomics of Plant Fungal Pathogens, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
- Fujian and Taiwan Joint Center for Ecological Control of Crop Pests, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
- *Correspondence: Justice Norvienyeku, Zonghua Wang,
| | - Zonghua Wang
- Fujian University Key Laboratory for Functional Genomics of Plant Fungal Pathogens, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
- Fujian and Taiwan Joint Center for Ecological Control of Crop Pests, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
- Institute of Oceanography, Minjiang University, Fuzhou, China
- *Correspondence: Justice Norvienyeku, Zonghua Wang,
| |
Collapse
|
34
|
MacDowell KS, Pinacho R, Leza JC, Costa J, Ramos B, García-Bueno B. Differential regulation of the TLR4 signalling pathway in post-mortem prefrontal cortex and cerebellum in chronic schizophrenia: Relationship with SP transcription factors. Prog Neuropsychopharmacol Biol Psychiatry 2017; 79:481-492. [PMID: 28803924 DOI: 10.1016/j.pnpbp.2017.08.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 07/28/2017] [Accepted: 08/06/2017] [Indexed: 12/21/2022]
Abstract
Alterations in innate immunity may underlie the pathophysiology of schizophrenia (SZ). Toll-like receptor-4 (TLR4) is a master element of innate immunity. The specificity proteins (SPs), transcription factors recently implicated in SZ, are putative regulatory agents of this. This work was aimed at describing alterations in the TLR4 signalling pathway in postmortem brain prefrontal cortex (PFC) and cerebellum (CB) of 16 chronic SZ patients and 14 controls. The possible association of TLR4 pathway with SP1 and SP4 and SZ negative symptomatology is explored. In PFC, TLR4/myeloid differentiation factor 88 (MyD88)/inhibitory subunit of nuclear factor kappa B alpha (IκBα) protein levels were lower in SZ patients, while nuclear transcription factor-κB (NFκB) activity, cyclooxygenase-2 (COX-2) expression and the lipid peroxidation index malondialdehyde (MDA) appeared increased. The pattern of changes in CB is opposite, except for COX-2 expression that remained augmented and MDA levels unaltered. Network interaction analysis showed that TLR4/MyD88/IκBα/NFκB/COX-2 pathway was coupled in PFC and uncoupled in CB. SP4 co-expressed with TLR4 and NFκB in PFC and both SP1 and SP4 co-expressed with NFκB in CB. In PFC, correlation analysis found an inverse relationship between NFκB and negative symptoms. In summary, we found brain region-specific alterations in the TLR4 signalling pathway in chronic SZ, in which SP transcription factors could participate at different levels. Further studies are required to elucidate the regulatory mechanisms of innate immunity in SZ and its relationship with symptoms.
Collapse
Affiliation(s)
- Karina S MacDowell
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain; Dept. of Pharmacology, Faculty of Medicine, Hospital 12 de Octubre Imas12, IUINQ, University Complutense, 28040 Madrid, Spain
| | - Raquel Pinacho
- Psiquiatria Molecular, Institut de Recerca Sant Joan de Déu, Santa Rosa 39-57, 08950 Esplugues de Llobregat, Spain
| | - Juan C Leza
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain; Dept. of Pharmacology, Faculty of Medicine, Hospital 12 de Octubre Imas12, IUINQ, University Complutense, 28040 Madrid, Spain
| | - Joan Costa
- Banc de Teixits Neurologics, Parc Sanitari Sant Joan de Déu, Sant Boi de Llobregat, 08830 Barcelona, Spain; Parc Sanitari Sant Joan de Déu, Dr. Antoni Pujadas, 42, 08830 Sant Boi de Llobregat, Spain
| | - Belén Ramos
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain; Psiquiatria Molecular, Institut de Recerca Sant Joan de Déu, Santa Rosa 39-57, 08950 Esplugues de Llobregat, Spain; Parc Sanitari Sant Joan de Déu, Dr. Antoni Pujadas, 42, 08830 Sant Boi de Llobregat, Spain; Dept. de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain.
| | - Borja García-Bueno
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain; Dept. of Pharmacology, Faculty of Medicine, Hospital 12 de Octubre Imas12, IUINQ, University Complutense, 28040 Madrid, Spain.
| |
Collapse
|