1
|
Gan X, Li X, Cai Y, Yin B, Pan Q, Teng T, He Y, Tang H, Wang T, Li J, Zhu Z, Zhou X, Li J. Metabolic features of adolescent major depressive disorder: A comparative study between treatment-resistant depression and first-episode drug-naive depression. Psychoneuroendocrinology 2024; 167:107086. [PMID: 38824765 DOI: 10.1016/j.psyneuen.2024.107086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 04/12/2024] [Accepted: 05/21/2024] [Indexed: 06/04/2024]
Abstract
Major depressive disorder (MDD) is a psychiatric illness that can jeopardize the normal growth and development of adolescents. Approximately 40% of adolescent patients with MDD exhibit resistance to conventional antidepressants, leading to the development of Treatment-Resistant Depression (TRD). TRD is associated with severe impairments in social functioning and learning ability and an elevated risk of suicide, thereby imposing an additional societal burden. In this study, we conducted plasma metabolomic analysis on 53 adolescents diagnosed with first-episode drug-naïve MDD (FEDN-MDD), 53 adolescents with TRD, and 56 healthy controls (HCs) using hydrophilic interaction liquid chromatography-mass spectrometry (HILIC-MS) and reversed-phase liquid chromatography-mass spectrometry (RPLC-MS). We established a diagnostic model by identifying differentially expressed metabolites and applying cluster analysis, metabolic pathway analysis, and multivariate linear support vector machine (SVM) algorithms. Our findings suggest that adolescent TRD shares similarities with FEDN-MDD in five amino acid metabolic pathways and exhibits distinct metabolic characteristics, particularly tyrosine and glycerophospholipid metabolism. Furthermore, through multivariate receiver operating characteristic (ROC) analysis, we optimized the area under the curve (AUC) and achieved the highest predictive accuracy, obtaining an AUC of 0.903 when comparing FEDN-MDD patients with HCs and an AUC of 0.968 when comparing TRD patients with HCs. This study provides new evidence for the identification of adolescent TRD and sheds light on different pathophysiologies by delineating the distinct plasma metabolic profiles of adolescent TRD and FEDN-MDD.
Collapse
Affiliation(s)
- Xieyu Gan
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xuemei Li
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuping Cai
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Bangmin Yin
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qiyuan Pan
- The First People's Hospital of Zaoyang City, Hubei, China
| | - Teng Teng
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuqian He
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Han Tang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ting Wang
- Department of Psychology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jie Li
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhengjiang Zhu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China; Shanghai Key Laboratory of Aging Studies, Shanghai, China.
| | - Xinyu Zhou
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Jinfang Li
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
2
|
Xia N, Wang J, Guo Q, Duan J, Wang X, Zhou P, Li J, Tang T, Li T, Li H, Wu Z, Yang M, Sun J, Guo D, Chang X, Zhang X. Deciphering the antidepressant effects of Rosa damascena essential oil mediated through the serotonergic synapse signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:118007. [PMID: 38492791 DOI: 10.1016/j.jep.2024.118007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/08/2024] [Accepted: 03/02/2024] [Indexed: 03/18/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Rosa damascena is an ancient plant with significance in both medicine and perfumery that have a variety of therapeutic properties, including antidepressant, anti-anxiety, and anti-stress effects. Rose damascena essential oil (REO) has been used to treat depression, anxiety and other neurological related disorders in Iranian traditional medicine. However, its precise mechanism of action remains elusive. AIM OF THE STUDY The aim of this study was to investigate the impact and mechanism underlying the influence of REO on chronic unpredictable mild stress (CUMS) rats. MATERIALS AND METHODS Gas chromatography-mass spectrometry (GC-MS) technique coupling was used to analyze of the components of REO. A CUMS rat model was replicated to assess the antidepressant effects of varying doses of REO. This assessment encompassed behavioral evaluations, biochemical index measurements, and hematoxylin-eosin staining. For a comprehensive analysis of hippocampal tissues, we employed transcriptomics and incorporated weighting coefficients by means of network pharmacology. These measures allowed us to explore differentially expressed genes and biofunctional pathways affected by REO in the context of depression treatment. Furthermore, GC-MS metabolomics was employed to assess metabolic profiles, while a joint analysis in Metscape facilitated the construction of a network elucidating the links between differentially expressed genes and metabolites, thereby elucidating potential relationships and clarifying key pathways regulated by REO. Finally, the expression of relevant proteins in the key pathways was determined through immunohistochemistry and Western blot analysis. Molecular docking was utilized to investigate the interactions between active components and key targets, thereby validating the experimental results. RESULTS REO alleviated depressive-like behavior, significantly elevated levels of the neurotransmitter 5-hydroxytryptamine (5-HT), and reduced hippocampal neuronal damage in CUMS rats. This therapeutic effect may be associated with the modulation of the serotonergic synapse signaling pathway. Furthermore, REO rectified metabolic disturbances, primarily through the regulation of amino acid metabolic pathways. Joint analysis revealed five differentially expressed genes (EEF1A1, LOC729197, ATP8A2, NDST4, and GAD2), suggesting their potential in alleviating depressive symptoms by modulating the serotonergic synapse signaling pathway and tryptophan metabolism. REO also modulated the 5-HT2A-mediated extracellular regulated protein kinases-cAMP-response element binding protein-brain-derived neurotrophic factor (ERK-CREB-BDNF) pathway. In addition, molecular docking results indicated that citronellol, geraniol and (E,E)-farnesol in REO may serve as key active ingredients responsible for its antidepressant effects. CONCLUSIONS This study is the first to report that REO can effectively alleviate CUMS-induced depression-like effects in rats. Additionally, the study offers a comprehensive understanding of its intricate antidepressant mechanism from a multi-omics and multi-level perspective. Our findings hold promise for the clinical application and further development of this essential oil.
Collapse
Affiliation(s)
- Ning Xia
- Key Laboratory of Basic and New Drug Research in Chinese Medicine, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| | - Jie Wang
- Key Laboratory of Basic and New Drug Research in Chinese Medicine, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| | - Qiuting Guo
- Xianyang Polytechnic Institute, Xianyang, 712000, Shaanxi, China
| | - Jiawei Duan
- Key Laboratory of Basic and New Drug Research in Chinese Medicine, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| | - Xuan Wang
- Key Laboratory of Basic and New Drug Research in Chinese Medicine, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| | - Peijie Zhou
- Key Laboratory of Basic and New Drug Research in Chinese Medicine, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| | - Jinkai Li
- Key Laboratory of Basic and New Drug Research in Chinese Medicine, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| | - Tiantian Tang
- Key Laboratory of Basic and New Drug Research in Chinese Medicine, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| | - Taotao Li
- Key Laboratory of Basic and New Drug Research in Chinese Medicine, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| | - Huiting Li
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi, China
| | - Zhenfeng Wu
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi, China
| | - Ming Yang
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi, China
| | - Jing Sun
- Key Laboratory of Basic and New Drug Research in Chinese Medicine, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| | - Dongyan Guo
- Key Laboratory of Basic and New Drug Research in Chinese Medicine, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| | - Xing Chang
- Key Laboratory of Basic and New Drug Research in Chinese Medicine, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China.
| | - Xiaofei Zhang
- Key Laboratory of Basic and New Drug Research in Chinese Medicine, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China.
| |
Collapse
|
3
|
Lin S, Zhou Z, Qi Y, Chen J, Xu G, Shi Y, Yu Z, Li M, Chai K. Depression promotes breast cancer progression by regulating amino acid neurotransmitter metabolism and gut microbial disturbance. Clin Transl Oncol 2024; 26:1407-1418. [PMID: 38194019 DOI: 10.1007/s12094-023-03367-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 12/04/2023] [Indexed: 01/10/2024]
Abstract
INTRODUCTION Breast cancer (BC) is the most prevalent type of cancer and has the highest mortality among women worldwide. BC patients have a high risk of depression, which has been recognized as an independent factor in the progression of BC. However, the potential mechanism has not been clearly demonstrated. METHODS To explore the correlation and mechanism between depression and BC progression, we induced depression and tumor in BC mouse models. Depression was induced via chronic unpredictable mild stress (CUMS) and chronic restraint stress (CRS). Amino acid (AA) neurotransmitter-targeted metabonomics and gut microbiota 16S rDNA gene sequencing were employed in the mouse model after evaluation with behavioral tests and pathological analysis. RESULTS The tumors in cancer-depression (CD) mice grew faster than those in cancer (CA) mice, and lung metastasis was observed in CD mice. Metabonomics revealed that the neurotransmitters and plasma AAs in CD mice were dysregulated, namely the tyrosine and tryptophan pathways and monoamine neurotransmitters in the brain. Gut microbiota analysis displayed an increased ratio of Firmicutes/Bacteroides. In detail, the abundance of f_Lachnospiraceae and s_Lachnospiraceae increased, whereas the abundance of o_Bacteroidales and s_Bacteroides_caecimuris decreased. Moreover, the gut microbiota was more closely associated with AA neurotransmitters than with plasma AA. CONCLUSION Depression promoted the progression of BC by modulating the abundance of s_Lachnospiraceae and s_Bacteroides_caecimuris, which affected the metabolism of monoamine neurotransmitters in the brain and AA in the blood.
Collapse
Affiliation(s)
- Sisi Lin
- The Second School of Clinical Medical, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Cancer Prevention and Treatment Technology of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, Zhejiang, China
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Zhe Zhou
- The Second School of Clinical Medical, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Cancer Prevention and Treatment Technology of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, Zhejiang, China
| | - Yiming Qi
- The Second School of Clinical Medical, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Cancer Prevention and Treatment Technology of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, Zhejiang, China
| | - Jiabing Chen
- The Second School of Clinical Medical, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Cancer Prevention and Treatment Technology of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, Zhejiang, China
| | - Guoshu Xu
- The Second School of Clinical Medical, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Cancer Prevention and Treatment Technology of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, Zhejiang, China
| | - Yunfu Shi
- The Second School of Clinical Medical, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Cancer Prevention and Treatment Technology of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, Zhejiang, China
| | - Zhihong Yu
- The Second School of Clinical Medical, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Cancer Prevention and Treatment Technology of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, Zhejiang, China
| | - Mingqian Li
- Zhejiang Provincial Key Laboratory of Cancer Prevention and Treatment Technology of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, Zhejiang, China.
| | - Kequn Chai
- The Second School of Clinical Medical, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
- Zhejiang Provincial Key Laboratory of Cancer Prevention and Treatment Technology of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, Zhejiang, China.
| |
Collapse
|
4
|
Chang X, Zhang Y, Chen X, Li S, Mei H, Xiao H, Ma X, Liu Z, Li R. Gut microbiome and serum amino acid metabolome alterations in autism spectrum disorder. Sci Rep 2024; 14:4037. [PMID: 38369656 PMCID: PMC10874930 DOI: 10.1038/s41598-024-54717-2] [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: 10/05/2023] [Accepted: 02/15/2024] [Indexed: 02/20/2024] Open
Abstract
Gut microbiota and their metabolic products might play important roles in regulating the pathogenesis of autism spectrum disorder (ASD). The purpose of this study was to characterize gut microbiota and serum amino acid metabolome profiles in children with ASD. A non-randomized controlled study was carried out to analyze the alterations in the intestinal microbiota and their metabolites in patients with ASD (n = 30) compared with neurotypical controls (NC) (n = 30) by metagenomic sequencing to define the gut microbiota community and liquid chromatography/mass spectrometry (LC/MS) analysis to characterize the metabolite profiles. Compared with children in the NC group, those in the ASD group showed lower richness, higher evenness, and an altered microbial community structure. At the class level, Deinococci and Holophagae were significantly lower in children with ASD compared with TD. At the phylum level, Deinococcus-Thermus was significantly lower in children with ASD compared with TD. In addition, the functional properties (such as galactose metabolism) displayed significant differences between the ASD and NC groups. Five dominant altered species were identified and analyzed (LDA score > 2.0, P < 0.05), including Subdoligranulum, Faecalibacterium_praushitzii, Faecalibacterium, Veillonellaceae, and Rumminococcaceae. The peptides/nickel transport system was the main metabolic pathway involved in the differential species in the ASD group. Decreased ornithine levels and elevated valine levels may increase the risk of ASD through a metabolic pathway known as the nickel transport system. The microbial metabolism in diverse environments was negatively correlated with phascolarctobacterium succinatutens. Our study provides novel insights into compositional and functional alterations in the gut microbiome and metabolite profiles in ASD and the underlying mechanisms between metabolite and ASD.
Collapse
Affiliation(s)
- Xuening Chang
- Department of Child Health Care, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430016, China
| | - Yuchen Zhang
- School of Medicine, Wuhan University of Science and Technology, Wuhan, 430081, China
| | - Xue Chen
- School of Medicine, Wuhan University of Science and Technology, Wuhan, 430081, China
| | - Shihan Li
- Department of Child Health Care, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430016, China
| | - Hong Mei
- Department of Maternal and Child Health, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430016, China
| | - Han Xiao
- Department of Maternal and Child Health, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430016, China.
| | - Xinyu Ma
- Department of Radiology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430016, China.
| | - Zhisheng Liu
- Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430016, China.
| | - Ruizhen Li
- Department of Child Health Care, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430016, China.
| |
Collapse
|
5
|
Yang R, Lin Z, Cai Y, Chen N, Zhou Y, Zhang J, Hong G. Assessing the risk of prenatal depressive symptoms in Chinese women: an integrated evaluation of serum metabolome, multivitamin supplement intake, and clinical blood indicators. Front Psychiatry 2024; 14:1234461. [PMID: 38274432 PMCID: PMC10808622 DOI: 10.3389/fpsyt.2023.1234461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 12/11/2023] [Indexed: 01/27/2024] Open
Abstract
Background Prenatal depressive symptoms (PDS) is a serious public health problem. This study aimed to develop an integrated panel and nomogram to assess at-risk populations by examining the association of PDS with the serum metabolome, multivitamin supplement intake, and clinical blood indicators. Methods This study comprised 221 pregnant women, categorized into PDS and non-PDS groups based on the Edinburgh postnatal depression scale. The participants were divided into training and test sets according to their enrollment time. We conducted logistic regression analysis to identify risk factors, and employed liquid chromatography/high resolution mass spectrometry-based serum metabolome analysis to identify metabolic biomarkers. Multiple factor analysis was used to combine risk factors, clinical blood indicators and key metabolites, and then a nomogram was developed to estimate the probability of PDS. Results We identified 36 important differential serum metabolites as PDS biomarkers, mainly involved in amino acid metabolism and lipid metabolism. Multivitamin intake works as a protective factor for PDS. The nomogram model, including multivitamin intake, HDL-C and three key metabolites (histidine, estrone and valylasparagine), exhibited an AUC of 0.855 in the training set and 0.774 in the test set, and the calibration curves showed good agreement, indicating that the model had good stability. Conclusion Our approach integrates multiple models to identify metabolic biomarkers for PDS, ensuring their robustness. Furthermore, the inclusion of dietary factors and clinical blood indicators allows for a comprehensive characterization of each participant. The analysis culminated in an intuitive nomogram based on multimodal data, displaying potential performance in initial PDS risk assessment.
Collapse
Affiliation(s)
- Rongrong Yang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen, China
| | - Zhenguo Lin
- Department of Clinical Medicine, Xiamen Medical College, Xiamen, China
| | - Yanhua Cai
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen, China
| | - Nan Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen, China
| | - Ying Zhou
- Department of Obstetrics and Gynecology, Clinical Medical Research Center for Obstetrics and Gynecology Diseases, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Jie Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen, China
| | - Guolin Hong
- Department of Laboratory Medicine, Xiamen Key Laboratory of Genetic Testing, The First Affiliated Hospital of Xiamen University, School of Public Health, Xiamen University, Xiamen, China
| |
Collapse
|
6
|
Papageorgiou MP, Theodoridou D, Nussbaumer M, Syrrou M, Filiou MD. Deciphering the Metabolome under Stress: Insights from Rodent Models. Curr Neuropharmacol 2024; 22:884-903. [PMID: 37448366 PMCID: PMC10845087 DOI: 10.2174/1570159x21666230713094843] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/22/2022] [Accepted: 01/09/2023] [Indexed: 07/15/2023] Open
Abstract
Despite intensive research efforts to understand the molecular underpinnings of psychological stress and stress responses, the underlying molecular mechanisms remain largely elusive. Towards this direction, a plethora of stress rodent models have been established to investigate the effects of exposure to different stressors. To decipher affected molecular pathways in a holistic manner in these models, metabolomics approaches addressing altered, small molecule signatures upon stress exposure in a high-throughput, quantitative manner provide insightful information on stress-induced systemic changes in the brain. In this review, we discuss stress models in mice and rats, followed by mass spectrometry (MS) and nuclear magnetic resonance (NMR) metabolomics studies. We particularly focus on acute, chronic and early life stress paradigms, highlight how stress is assessed at the behavioral and molecular levels and focus on metabolomic outcomes in the brain and peripheral material such as plasma and serum. We then comment on common metabolomics patterns across different stress models and underline the need for unbiased -omics methodologies and follow-up studies of metabolomics outcomes to disentangle the complex pathobiology of stress and pertinent psychopathologies.
Collapse
Affiliation(s)
- Maria P. Papageorgiou
- Laboratory of Biochemistry, Department of Biological Applications and Technology, School of Health Sciences, University of Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas (BRI-FORTH), Ioannina, Greece
| | - Daniela Theodoridou
- Laboratory of Biology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Greece
| | - Markus Nussbaumer
- Laboratory of Biochemistry, Department of Biological Applications and Technology, School of Health Sciences, University of Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas (BRI-FORTH), Ioannina, Greece
| | - Maria Syrrou
- Laboratory of Biology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Greece
| | - Michaela D. Filiou
- Laboratory of Biochemistry, Department of Biological Applications and Technology, School of Health Sciences, University of Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas (BRI-FORTH), Ioannina, Greece
- Ιnstitute of Biosciences, University of Ioannina, Greece
| |
Collapse
|
7
|
Qi X, Fang J, Sun Y, Xu W, Li G. Altered Functional Brain Network Structure between Patients with High and Low Generalized Anxiety Disorder. Diagnostics (Basel) 2023; 13:1292. [PMID: 37046509 PMCID: PMC10093329 DOI: 10.3390/diagnostics13071292] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/26/2023] [Accepted: 03/27/2023] [Indexed: 04/01/2023] Open
Abstract
To investigate the differences in functional brain network structures between patients with a high level of generalized anxiety disorder (HGAD) and those with a low level of generalized anxiety disorder (LGAD), a resting-state electroencephalogram (EEG) was recorded in 30 LGAD patients and 21 HGAD patients. Functional connectivity between all pairs of brain regions was determined by the Phase Lag Index (PLI) to construct a functional brain network. Then, the characteristic path length, clustering coefficient, and small world were calculated to estimate functional brain network structures. The results showed that the PLI values of HGAD were significantly increased in alpha2, and significantly decreased in the theta and alpha1 rhythms, and the small-world attributes for both HGAD patients and LGAD patients were less than one for all the rhythms. Moreover, the small-world values of HGAD were significantly lower than those of LGAD in the theta and alpha2 rhythms, which indicated that the brain functional network structure would deteriorate with the increase in generalized anxiety disorder (GAD) severity. Our findings may play a role in the development and understanding of LGAD and HGAD to determine whether interventions that target these brain changes may be effective in treating GAD.
Collapse
Affiliation(s)
- Xuchen Qi
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
- Department of Neurosurgery, Shaoxing People’s Hospital, Shaoxing 312000, China
| | - Jiaqi Fang
- College of Engineering, Zhejiang Normal University, Jinhua 321004, China
| | - Yu Sun
- Key Laboratory for Biomedical Engineering of Ministry of Education of China, Department of Biomedical Engineering, Zhejiang University, Hangzhou 310000, China
| | - Wanxiu Xu
- College of Engineering, Zhejiang Normal University, Jinhua 321004, China
| | - Gang Li
- College of Mathematical Medicine, Zhejiang Normal University, Jinhua 321004, China
| |
Collapse
|