1
|
Bindi VE, Hones KM, Schoch BS, Hampton HL, Wright TW, King JJ, Hao KA. The influence of depression on clinical outcomes of total shoulder arthroplasty: a systematic Review. EUROPEAN JOURNAL OF ORTHOPAEDIC SURGERY & TRAUMATOLOGY : ORTHOPEDIE TRAUMATOLOGIE 2024; 34:1757-1763. [PMID: 38526619 DOI: 10.1007/s00590-024-03911-z] [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: 12/02/2023] [Accepted: 03/07/2024] [Indexed: 03/27/2024]
Abstract
PURPOSE Much of the current literature on total shoulder arthroplasty (TSA) has assessed the impact of preoperative medical comorbidities on postoperative clinical outcomes. The literature concerning the impact of psychological disorders such as depression on TSA has increased in popularity in recent years, but there lacks a thorough review of the influence of depression on postoperative pain and functional outcomes. METHODS We queried PubMed/MEDLINE and identified six clinical studies that evaluated the influence of a psychiatric diagnosis of depression on patient outcomes after TSA. Studies that discussed the impacts of depression on TSA, including PROs or adverse events in adults, were included. Studies focused on other psychologic pathology, non-TSA shoulder treatments, or TSA not for primary osteoarthritis were excluded. Non-clinical studies, systematic reviews, letters to the editor, commentaries, dissertations, books, and book chapters were excluded. RESULTS Three cohort studies described patient-reported pain and functional outcomes and three database studies assessed the risk of postoperative complications. Cohort studies demonstrated that the prevalence of depression in patients undergoing TSA decreased from preoperatively to 12-months postoperatively. Two studies demonstrated that depression is an independent predictor of less pre- to postoperative improvement in the ASES score at minimum 2-year follow-up; however, one study found the difference between patients with and without depression did not exceed the minimum clinically important difference. Database studies demonstrated that depression was associated with higher rates of blood transfusion (n = 1, OR = 1.8), anemia (n = 1, OR = 1.65), wound infection (n = 2, OR = 1.41-2.09), prosthetic revision (n = 1, OR = 1.92), and length of hospital stay (n = 3, LOS = 2.5-3 days). CONCLUSION Although patients with a preoperative diagnosis of depression undergoing TSA can achieve satisfactory relief of shoulder pain and restoration of function, they may experience poorer patient-reported outcomes and a higher risk of postoperative adverse events compared to their peers. Surgeons should be cognizant of the influence of depression in their patients to facilitate proper patient selection that maximizes patient satisfaction, function, and minimizes the risk of adverse events following TSA. LEVEL OF EVIDENCE Level IV.
Collapse
Affiliation(s)
- Victoria E Bindi
- College of Medicine, University of Florida, Gainesville, FL, USA
| | - Keegan M Hones
- Department of Orthopaedic Surgery & Sports Medicine, University of Florida, 3450 Hull Road, Gainesville, FL, 32611, USA
| | - Bradley S Schoch
- Department of Orthopedic Surgery, Mayo Clinic, Jacksonville, FL, USA
| | - Hailey L Hampton
- Department of Orthopaedic Surgery & Sports Medicine, University of Florida, 3450 Hull Road, Gainesville, FL, 32611, USA
| | - Thomas W Wright
- Department of Orthopaedic Surgery & Sports Medicine, University of Florida, 3450 Hull Road, Gainesville, FL, 32611, USA
| | - Joseph J King
- Department of Orthopaedic Surgery & Sports Medicine, University of Florida, 3450 Hull Road, Gainesville, FL, 32611, USA.
| | - Kevin A Hao
- College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
2
|
Khan M, Baussan Y, Hebert-Chatelain E. Connecting Dots between Mitochondrial Dysfunction and Depression. Biomolecules 2023; 13:695. [PMID: 37189442 PMCID: PMC10135685 DOI: 10.3390/biom13040695] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/10/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
Mitochondria are the prime source of cellular energy, and are also responsible for important processes such as oxidative stress, apoptosis and Ca2+ homeostasis. Depression is a psychiatric disease characterized by alteration in the metabolism, neurotransmission and neuroplasticity. In this manuscript, we summarize the recent evidence linking mitochondrial dysfunction to the pathophysiology of depression. Impaired expression of mitochondria-related genes, damage to mitochondrial membrane proteins and lipids, disruption of the electron transport chain, higher oxidative stress, neuroinflammation and apoptosis are all observed in preclinical models of depression and most of these parameters can be altered in the brain of patients with depression. A deeper knowledge of the depression pathophysiology and the identification of phenotypes and biomarkers with respect to mitochondrial dysfunction are needed to help early diagnosis and the development of new treatment strategies for this devastating disorder.
Collapse
Affiliation(s)
- Mehtab Khan
- Department of Biology, University of Moncton, Moncton, NB E1A 3E9, Canada
- Mitochondrial Signaling and Pathophysiology, University of Moncton, Moncton, NB E1A 3E9, Canada
| | - Yann Baussan
- Department of Biology, University of Moncton, Moncton, NB E1A 3E9, Canada
- Mitochondrial Signaling and Pathophysiology, University of Moncton, Moncton, NB E1A 3E9, Canada
| | - Etienne Hebert-Chatelain
- Department of Biology, University of Moncton, Moncton, NB E1A 3E9, Canada
- Mitochondrial Signaling and Pathophysiology, University of Moncton, Moncton, NB E1A 3E9, Canada
| |
Collapse
|
3
|
Aerobic Exercise Prevents Depression via Alleviating Hippocampus Injury in Chronic Stressed Depression Rats. Brain Sci 2020; 11:brainsci11010009. [PMID: 33374661 PMCID: PMC7822431 DOI: 10.3390/brainsci11010009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/17/2020] [Accepted: 12/17/2020] [Indexed: 02/01/2023] Open
Abstract
(1) Background: Depression is one of the overwhelming public health problems. Alleviating hippocampus injury may prevent depression development. Herein, we established the chronic unpredictable mild stress (CUMS) model and aimed to investigate whether aerobic exercise (AE) could alleviate CUMS induced depression-like behaviors and hippocampus injury. (2) Methods: Forty-eight healthy male Sprague-Dawley rats (200 ± 20 g) were randomly divided into 4 groups (control, CUMS, CUMS + 7 days AE, CUMS + 14 days AE). Rats with AE treatments were subjected to 45 min treadmill per day. (3) Results: AE intervention significantly improved CUMS-induced depressive behaviors, e.g., running square numbers and immobility time assessed by the open field and forced swimming test, suppressed hippocampal neuron apoptosis, reduced levels of phosphorylation of NMDA receptor and homocysteine in hippocampus, as well as serum glucocorticoids, compared to the CUMS rats. In contrast, AE upregulated phosphorylation of AMPAR receptor and brain-derived neurotrophic factor (BDNF) hippocampus in CUMS depression rats. The 14 day-AE treatment exhibited better performance than 7 day-AE on the improvement of the hippocampal function. (4) Conclusion: AE might be an efficient strategy for prevention of CUMS-induced depression via ameliorating hippocampus functions. Underlying mechanisms may be related with glutamatergic system, the neurotoxic effects of homocysteine, and/or influences in glucocorticoids-BDNF expression interaction.
Collapse
|
4
|
He Y, Wang Y, Wu Z, Lan T, Tian Y, Chen X, Li Y, Dang R, Bai M, Cheng K, Xie P. Metabolomic abnormalities of purine and lipids implicated olfactory bulb dysfunction of CUMS depressive rats. Metab Brain Dis 2020; 35:649-659. [PMID: 32152797 DOI: 10.1007/s11011-020-00557-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 02/24/2020] [Indexed: 12/27/2022]
Abstract
Major depressive disorder (MDD) is a serious mood disorder and leads to a high suicide rate as well as financial burden. The volume and function (the sensitivity and neurogenesis) of the olfactory bulb (OB) were reported to be altered among the MDD patients and rodent models of depression. In addition, the olfactory epithelium was newly reported to decrease its volume and function under chronic unpredictable mild stress (CUMS) treatment. However, the underlying molecular mechanism still remains unclear. Herein, we conducted the non-targeted metabolomics method based on gas chromatography-mass spectrometry (GC-MS) coupled with multivariate statistical analysis to characterize the differential metabolites in OB of CUMS rats. Our results showed that 19 metabolites were categorized into two perturbed pathways: purine metabolism and lipid metabolism, which were regarded as the vital pathways concerned with dysfunction of OB. These findings indicated that the turbulence of metabolic pathways may be partly responsible for the dysfunction of OB in MDD.
Collapse
Affiliation(s)
- Yong He
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400016, China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016, China
| | - Yue Wang
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400016, China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016, China
- Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Zhonghao Wu
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400016, China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016, China
- College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Tianlan Lan
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400016, China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016, China
- College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Yu Tian
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400016, China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xi Chen
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400016, China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016, China
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing, 402460, China
| | - Yan Li
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400016, China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Ruozhi Dang
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400016, China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Mengge Bai
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400016, China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016, China
- Key Laboratory of Laboratory Medical Diagnostics of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Ke Cheng
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400016, China.
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016, China.
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Peng Xie
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400016, China.
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016, China.
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing, 402460, China.
| |
Collapse
|
5
|
Kang SY, Kim HB, Sunwoo S. Association between anemia and maternal depression: A systematic review and meta-analysis. J Psychiatr Res 2020; 122:88-96. [PMID: 31945502 DOI: 10.1016/j.jpsychires.2020.01.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 11/22/2019] [Accepted: 01/06/2020] [Indexed: 12/24/2022]
Abstract
Previous observational epidemiological studies have reported inconsistent findings regarding the association between anemia and the risk of maternal depression. In the present study, we investigated the relationship between anemia and the risk of maternal depression using a meta-analysis. We searched PubMed, EMBASE, and the bibliographies of relevant articles in May 2019. Three evaluators independently reviewed and selected the eligible studies based on the predetermined selection criteria. A random-effects model was employed to calculate meta-estimates of the association between anemia and maternal depression. Of the 1305 articles, 15 observational epidemiological studies (five case-control studies and 10 cohort studies) were included in the final analysis. A total of 32,792,378 women were included. Anemia was significantly associated with an increased risk of maternal depression in the random-effects meta-analysis of 15 studies (OR/RR: 1.53, 95% CI: 1.32-1.78). The association was consistent in both antepartum (OR/RR: 1.36, 95% CI: 1.07-1.72) and postpartum depression (OR/RR: 1.53, 95% CI: 1.32-1.78). Subgroup meta-analyses based on definition of anemia, definition of depression, and methodological quality reported consistent findings. The current meta-analysis showed that anemia was associated with an increased risk of maternal depression.
Collapse
Affiliation(s)
- Seo Young Kang
- International Healthcare Center, Asan Medical Center, Republic of Korea
| | - Hong-Bae Kim
- Department of Family Medicine, Myoungji Hospital, Hanyang University College of Medicine, Republic of Korea.
| | - Sung Sunwoo
- Department of Family Medicine, Asan Medical Center, University of Ulsan College of Medicine, Republic of Korea
| |
Collapse
|
6
|
Yang X, Wang G, Gong X, Huang C, Mao Q, Zeng L, Zheng P, Qin Y, Ye F, Lian B, Zhou C, Wang H, Zhou W, Xie P. Effects of chronic stress on intestinal amino acid pathways. Physiol Behav 2019; 204:199-209. [PMID: 30831184 DOI: 10.1016/j.physbeh.2019.03.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 02/22/2019] [Accepted: 03/01/2019] [Indexed: 01/23/2023]
Abstract
Major depressive disorder (MDD) is a debilitating mental disorder with a high prevalence and severe impacts on quality of life. However, the pathophysiological mechanisms underlying MDD remain poorly understood. Here, we used high-performance liquid chromatography with ultraviolet detection-based targeted metabolomics to identify amino acid changes in the small intestine, in a rat model of chronic unpredictable mild stress (CUMS). Pearson's correlation analysis was conducted to investigate the correlations between amino acid changes and behavioral outcomes. Western blot analysis was employed to verify intestinal amino acid transport function. Moreover, we performed an integrated analysis of related differential amino acids in the hippocampus, peripheral blood mononuclear cells (PBMCs), urine and cerebellum identified in our previous studies using the CUMS rat model to further our understanding of amino acid metabolism in depression. Decreased concentrations of glutamine and glycine and upregulation of aspartic acid were found in CUMS model rats. These changes were significantly correlated with depressive-like behaviors. Western blot analysis revealed that CUMS rats exhibited a reduction in the expression levels of amino acid transporters ASCT2 and B0AT1, as well as an increase in LAT1 expression. Impaired transport of glycine and glutamine into the small intestine may contribute to a central deficiency. The current findings suggest that the glycine and glutamine uptake systems may be potential therapeutic targets for depression. The integrated analysis strategy used in the current study may provide new insight into the cellular and molecular mechanisms underlying the gut-brain axis, and help to elucidate the pathophysiological changes in central and peripheral systems in depression.
Collapse
Affiliation(s)
- Xun Yang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China
| | - Guowei Wang
- Ning Xia Medical University, Yin Chuan, Ning Xia 750004, China
| | - Xue Gong
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China
| | - Cheng Huang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China
| | - Qiang Mao
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China; The Second Affiliated Hospital of Chongqing Medical University, Department of Pharmacy, Chongqing, China
| | - Li Zeng
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China; Department of Nephrology, Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Peng Zheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China
| | - Yinhua Qin
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China
| | - Fei Ye
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China
| | - Bin Lian
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China
| | - Chanjuan Zhou
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China; Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402460, China
| | - Haiyang Wang
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China
| | - Wei Zhou
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China
| | - Peng Xie
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China.
| |
Collapse
|
7
|
Woods AG, Wormwood KL, Iosifescu DV, Murrough J, Darie CC. Protein Biomarkers in Major Depressive Disorder: An Update. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1140:585-600. [DOI: 10.1007/978-3-030-15950-4_35] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
8
|
Pan JX, Xia JJ, Deng FL, Liang WW, Wu J, Yin BM, Dong MX, Chen JJ, Ye F, Wang HY, Zheng P, Xie P. Diagnosis of major depressive disorder based on changes in multiple plasma neurotransmitters: a targeted metabolomics study. Transl Psychiatry 2018; 8:130. [PMID: 29991685 PMCID: PMC6039504 DOI: 10.1038/s41398-018-0183-x] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 05/11/2018] [Accepted: 06/05/2018] [Indexed: 12/13/2022] Open
Abstract
Major depressive disorder (MDD) is a debilitating psychiatric illness. However, there is currently no objective laboratory-based diagnostic tests for this disorder. Although, perturbations in multiple neurotransmitter systems have been implicated in MDD, the biochemical changes underlying the disorder remain unclear, and a comprehensive global evaluation of neurotransmitters in MDD has not yet been performed. Here, using a GC-MS coupled with LC-MS/MS-based targeted metabolomics approach, we simultaneously quantified the levels of 19 plasma metabolites involved in GABAergic, catecholaminergic, and serotonergic neurotransmitter systems in 50 first-episode, antidepressant drug-naïve MDD subjects and 50 healthy controls to identify potential metabolite biomarkers for MDD (training set). Moreover, an independent sample cohort comprising 49 MDD patients, 30 bipolar disorder (BD) patients and 40 healthy controls (testing set) was further used to validate diagnostic generalizability and specificity of these candidate biomarkers. Among the 19 plasma neurotransmitter metabolites examined, nine were significantly changed in MDD subjects. These metabolites were mainly involved in GABAergic, catecholaminergic and serotonergic systems. The GABAergic and catecholaminergic had better diagnostic value than serotonergic pathway. A panel of four candidate plasma metabolite biomarkers (GABA, dopamine, tyramine, kynurenine) could distinguish MDD subjects from health controls with an AUC of 0.968 and 0.953 in the training and testing set, respectively. Furthermore, this panel distinguished MDD subjects from BD subjects with high accuracy. This study is the first to globally evaluate multiple neurotransmitters in MDD plasma. The altered plasma neurotransmitter metabolite profile has potential differential diagnostic value for MDD.
Collapse
Affiliation(s)
- Jun-Xi Pan
- 0000 0000 8653 0555grid.203458.8Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, 402460 China ,Chongqing Key Laboratory of Neurobiology, Chongqing, 400016 China ,0000 0000 8653 0555grid.203458.8Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016 China ,0000 0000 8653 0555grid.203458.8The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016 China
| | - Jin-Jun Xia
- 0000 0000 8653 0555grid.203458.8Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, 402460 China ,Chongqing Key Laboratory of Neurobiology, Chongqing, 400016 China ,0000 0000 8653 0555grid.203458.8Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016 China ,0000 0000 8653 0555grid.203458.8The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016 China
| | - Feng-Li Deng
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400016 China ,0000 0000 8653 0555grid.203458.8Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016 China
| | - Wei-Wei Liang
- 0000 0000 8653 0555grid.203458.8Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, 402460 China ,Chongqing Key Laboratory of Neurobiology, Chongqing, 400016 China ,0000 0000 8653 0555grid.203458.8Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016 China
| | - Jing Wu
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400016 China ,0000 0000 8653 0555grid.203458.8Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016 China
| | - Bang-Min Yin
- 0000 0000 8653 0555grid.203458.8Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, 402460 China ,Chongqing Key Laboratory of Neurobiology, Chongqing, 400016 China ,0000 0000 8653 0555grid.203458.8Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016 China
| | - Mei-Xue Dong
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400016 China ,0000 0000 8653 0555grid.203458.8Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016 China ,grid.452206.7Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jian-Jun Chen
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400016 China ,0000 0000 8653 0555grid.203458.8Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016 China
| | - Fei Ye
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400016 China ,0000 0000 8653 0555grid.203458.8Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016 China ,grid.452206.7Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hai-Yang Wang
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400016 China ,0000 0000 8653 0555grid.203458.8Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016 China
| | - Peng Zheng
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400016, China. .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016, China. .,Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Peng Xie
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, 402460, China. .,Chongqing Key Laboratory of Neurobiology, Chongqing, 400016, China. .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
9
|
Reig-Viader R, Sindreu C, Bayés À. Synaptic proteomics as a means to identify the molecular basis of mental illness: Are we getting there? Prog Neuropsychopharmacol Biol Psychiatry 2018; 84:353-361. [PMID: 28941771 DOI: 10.1016/j.pnpbp.2017.09.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 09/05/2017] [Accepted: 09/15/2017] [Indexed: 12/31/2022]
Abstract
Synapses are centrally involved in many brain disorders, particularly in psychiatric and neurodevelopmental ones. However, our current understanding of the proteomic alterations affecting synaptic performance in the majority of mental illnesses is limited. As a result, novel pharmacotherapies with improved neurological efficacy have been scarce over the past decades. The main goal of synaptic proteomics in the context of mental illnesses is to identify dysregulated molecular mechanisms underlying these conditions. Here we reviewed and performed a meta-analysis of previous neuroproteomic research to identify proteins that may be consistently dysregulated in one or several mental disorders. Notably, we found very few proteins reproducibly altered among independent experiments for any given condition or between conditions, indicating that we are still far from identifying key pathophysiological mechanisms of mental illness. We suggest that future research in the field will require higher levels of standardization and larger-scale experiments to address the challenge posed by biological and methodological variability. We strongly believe that more resources should be placed in this field as the need to identify the molecular roots of mental illnesses is highly pressing.
Collapse
Affiliation(s)
- Rita Reig-Viader
- Molecular Physiology of the Synapse Laboratory, Biomedical Research Institute Sant Pau (IIB Sant Pau), Sant Antoni Mª Claret 167, 08025 Barcelona, Spain; Universitat Autònoma de Barcelona, 08193, Bellaterra, Cerdanyola del Vallès, Spain\
| | - Carlos Sindreu
- Department of Clinical Foundations, University of Barcelona, Barcelona 08036, Spain; Institute of Neuroscience UB, Barcelona 08035, Spain
| | - Àlex Bayés
- Molecular Physiology of the Synapse Laboratory, Biomedical Research Institute Sant Pau (IIB Sant Pau), Sant Antoni Mª Claret 167, 08025 Barcelona, Spain; Universitat Autònoma de Barcelona, 08193, Bellaterra, Cerdanyola del Vallès, Spain\.
| |
Collapse
|
10
|
Perić I, Costina V, Stanisavljević A, Findeisen P, Filipović D. Proteomic characterization of hippocampus of chronically socially isolated rats treated with fluoxetine: Depression-like behaviour and fluoxetine mechanism of action. Neuropharmacology 2018; 135:268-283. [DOI: 10.1016/j.neuropharm.2018.03.034] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 03/22/2018] [Accepted: 03/24/2018] [Indexed: 12/20/2022]
|
11
|
Zhang Y, Yuan S, Pu J, Yang L, Zhou X, Liu L, Jiang X, Zhang H, Teng T, Tian L, Xie P. Integrated Metabolomics and Proteomics Analysis of Hippocampus in a Rat Model of Depression. Neuroscience 2018; 371:207-220. [DOI: 10.1016/j.neuroscience.2017.12.001] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 11/29/2017] [Accepted: 12/02/2017] [Indexed: 02/06/2023]
|
12
|
Bai S, Zhang X, Chen Z, Wang W, Hu Q, Liang Z, Shen P, Gui S, Zeng L, Liu Z, Chen J, Xie X, Huang H, Han Y, Wang H, Xie P. Insight into the metabolic mechanism of Diterpene Ginkgolides on antidepressant effects for attenuating behavioural deficits compared with venlafaxine. Sci Rep 2017; 7:9591. [PMID: 28852120 PMCID: PMC5575021 DOI: 10.1038/s41598-017-10391-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 08/09/2017] [Indexed: 02/05/2023] Open
Abstract
Depression is a severe and chronic mental disorder, affecting about 322 million individuals worldwide. A recent study showed that diterpene ginkgolides (DG) have antidepressant-like effects on baseline behaviours in mice. Here, we examined the effects of DG and venlafaxine (VLX) in a chronic social defeat stress model of depression. Both DG and VLX attenuated stress-induced social deficits, despair behaviour and exploratory behaviour. To elucidate the metabolic changes underlying the antidepressive effects of DG and VLX, we investigated candidate functional pathways in the prefrontal cortex using a GC-MS-based metabolomics approach. Metabolic functions and pathways analysis revealed that DG and VLX affect protein biosynthesis and nucleotide metabolism to enhance cell proliferation, with DG having a weaker impact than VLX. Glutamate and aspartate metabolism played important roles in the antidepressant effects of DG and VLX. Tyrosine degradation and cell-to-cell signaling and interaction helped discriminate the two antidepressants. L-glutamic acid was negatively correlated, while hypoxanthine was positively correlated, with the social interaction ratio. Understanding the metabolic changes produced by DG and VLX should provide insight into the mechanisms of action of these drugs and aid in the development of novel therapies for depression.
Collapse
Affiliation(s)
- Shunjie Bai
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
- Key Laboratory of Laboratory Medical Diagnostics of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Xiaodong Zhang
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhi Chen
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
- Key Laboratory of Laboratory Medical Diagnostics of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Wei Wang
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Qingchuan Hu
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
- Key Laboratory of Laboratory Medical Diagnostics of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Zihong Liang
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
- Department of Neurology, The Inner Mongolia Autonomous Region people's Hospital, Hohhot, Inner Mongolia, China
| | - Peng Shen
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Siwen Gui
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Li Zeng
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhao Liu
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianjun Chen
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Xiongfei Xie
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hua Huang
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yu Han
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haiyang Wang
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Peng Xie
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, China.
- Chongqing Key Laboratory of Neurobiology, Chongqing, China.
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China.
- Key Laboratory of Laboratory Medical Diagnostics of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China.
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
13
|
Mao Q, Gong X, Zhou C, Tu Z, Zhao L, Wang L, Wang X, Sun L, Xia J, Lian B, Chen J, Mu J, Yang D, Xie P. Up-regulation of SIRT6 in the hippocampus induced rats with depression-like behavior via the block Akt/GSK3β signaling pathway. Behav Brain Res 2017; 323:38-46. [DOI: 10.1016/j.bbr.2017.01.035] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 01/20/2017] [Accepted: 01/21/2017] [Indexed: 12/22/2022]
|
14
|
Carboni L, Nguyen TP, Caberlotto L. Systems biology integration of proteomic data in rodent models of depression reveals involvement of the immune response and glutamatergic signaling. Proteomics Clin Appl 2016; 10:1254-1263. [PMID: 27612656 DOI: 10.1002/prca.201500149] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 08/31/2016] [Accepted: 09/07/2016] [Indexed: 01/29/2023]
Affiliation(s)
- Lucia Carboni
- Department of Pharmacy and Biotechnology; Alma Mater Studiorum University of Bologna; Bologna Italy
| | | | - Laura Caberlotto
- Centre for Computational and Systems Biology (COSBI); The Microsoft Research-University of Trento; Trento Italy
- Aptuit (Verona); Verona Italy
| |
Collapse
|
15
|
Abstract
Major depressive disorder (MDD: unipolar depression) is widely distributed in the USA and world-wide populations and it is one of the leading causes of disability in both adolescents and adults. Traditional diagnostic approaches for MDD are based on patient interviews, which provide a subjective assessment of clinical symptoms which are frequently shared with other maladies. Reliance upon clinical assessments and patient interviews for diagnosing MDD is frequently associated with misdiagnosis and suboptimal treatment outcomes. As such, there is increasing interest in the identification of objective methods for the diagnosis of depression. Newer technologies from genomics, transcriptomics, proteomics, metabolomics and imaging are technically sophisticated and objective but their application to diagnostic tests in psychiatry is still emerging. This brief overview evaluates the technical basis for these technologies and discusses how the extension of their clinical performance can lead to an objective diagnosis of MDD.
Collapse
Affiliation(s)
- John A Bilello
- Ridge Diagnostics Laboratories, Research & Development, Research Triangle Park, NC, USA
| |
Collapse
|
16
|
Sethi S, Chourasia D, Parhar IS. Approaches for targeted proteomics and its potential applications in neuroscience. J Biosci 2016; 40:607-27. [PMID: 26333406 DOI: 10.1007/s12038-015-9537-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
An extensive guide on practicable and significant quantitative proteomic approaches in neuroscience research is important not only because of the existing overwhelming limitations but also for gaining valuable understanding into brain function and deciphering proteomics from the workbench to the bedside. Early methodologies to understand the functioning of biological systems are now improving with high-throughput technologies, which allow analysis of various samples concurrently, or of thousand of analytes in a particular sample. Quantitative proteomic approaches include both gel-based and non-gel-based methods that can be further divided into different labelling approaches. This review will emphasize the role of existing technologies, their advantages and disadvantages, as well as their applications in neuroscience. This review will also discuss advanced approaches for targeted proteomics using isotope-coded affinity tag (ICAT) coupled with laser capture microdissection (LCM) followed by liquid chromatography tandem mass spectrometric (LC-MS/MS) analysis. This technology can further be extended to single cell proteomics in other areas of biological sciences and can be combined with other 'omics' approaches to reveal the mechanism of a cellular alterations. This approach may lead to further investigation in basic biology, disease analysis and surveillance, as well as drug discovery. Although numerous challenges still exist, we are confident that this approach will increase the understanding of pathological mechanisms involved in neuroendocrinology, neuropsychiatric and neurodegenerative disorders by delivering protein biomarker signatures for brain dysfunction.
Collapse
Affiliation(s)
- Sumit Sethi
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, MONASH University, Selangor Darul Ehsan, Malaysia,
| | | | | |
Collapse
|
17
|
Rao C, Shi H, Zhou C, Zhu D, Zhao M, Wang Z, Yang Y, Chen J, Liao L, Tang J, Wu Y, Zhou J, Cheng K, Xie P. Hypothalamic Proteomic Analysis Reveals Dysregulation of Glutamate Balance and Energy Metabolism in a Mouse Model of Chronic Mild Stress-Induced Depression. Neurochem Res 2016; 41:2443-56. [DOI: 10.1007/s11064-016-1957-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 05/07/2016] [Accepted: 05/11/2016] [Indexed: 01/21/2023]
|
18
|
Shao WH, Chen JJ, Fan SH, Lei Y, Xu HB, Zhou J, Cheng PF, Yang YT, Rao CL, Wu B, Liu HP, Xie P. Combined Metabolomics and Proteomics Analysis of Major Depression in an Animal Model: Perturbed Energy Metabolism in the Chronic Mild Stressed Rat Cerebellum. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2016; 19:383-92. [PMID: 26134254 DOI: 10.1089/omi.2014.0164] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Major depressive disorder (MDD) is a highly prevalent, debilitating mental illness of importance for global health. However, its molecular pathophysiology remains poorly understood. Combined proteomics and metabolomics approaches should provide a comprehensive understanding of MDD's etiology. The present study reports novel "-omics" insights from a rodent model of MDD. Cerebellar samples from chronic mild stressed (CMS)-treated depressed rats and controls were compared with a focus on the differentially expressed proteins and metabolites using isobaric tags for relative and absolute quantitation (iTRAQ)-based proteomics and gas chromotography/mass spectrometry (GC-MS) metabolomics techniques, respectively. The combined analyses found significant alterations associated with cerebellar energy metabolism, as indicated by (1) abnormal amino acid metabolism accompanied by corresponding metabolic enzymatic alterations and disturbed protein turnover, (2) increased glycolytic and tricarboxylic acid (TCA) cycle enzyme levels paralleled by changes in the concentrations of associated metabolites, and (3) perturbation of ATP biosynthesis through adenosine accompanied by perturbation of the mitochondrial respiratory chain. To the best of our knowledge, this study is the first to integrate proteomics and metabolomics analyses to examine the pathophysiological mechanism(s) underlying MDD in a CMS rodent model of depression. These results can offer important insights into the pathogenesis of MDD.
Collapse
Affiliation(s)
- Wei-hua Shao
- 1 Department of Respiratory Medicine, First Affiliated Hospital of Chongqing Medical University , Chongqing, China .,3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Jian-jun Chen
- 3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Song-hua Fan
- 2 Department of Neurology, First Affiliated Hospital of Chongqing Medical University , Chongqing, China .,3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Yang Lei
- 2 Department of Neurology, First Affiliated Hospital of Chongqing Medical University , Chongqing, China .,3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Hong-bo Xu
- 2 Department of Neurology, First Affiliated Hospital of Chongqing Medical University , Chongqing, China .,3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Jian Zhou
- 3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Peng-fei Cheng
- 2 Department of Neurology, First Affiliated Hospital of Chongqing Medical University , Chongqing, China .,3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Yong-tao Yang
- 2 Department of Neurology, First Affiliated Hospital of Chongqing Medical University , Chongqing, China .,3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Cheng-long Rao
- 3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Bo Wu
- 3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Hai-peng Liu
- 2 Department of Neurology, First Affiliated Hospital of Chongqing Medical University , Chongqing, China .,3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Peng Xie
- 2 Department of Neurology, First Affiliated Hospital of Chongqing Medical University , Chongqing, China .,3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| |
Collapse
|
19
|
Rong R, Yang H, Rong L, Wei X, Li Q, Liu X, Gao H, Xu Y, Zhang Q. Proteomic analysis of PSD-93 knockout mice following the induction of ischemic cerebral injury. Neurotoxicology 2016; 53:1-11. [DOI: 10.1016/j.neuro.2015.12.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Revised: 12/04/2015] [Accepted: 12/05/2015] [Indexed: 01/09/2023]
|
20
|
Wang Z, Li W, Chen J, Shi H, Zhao M, You H, Rao C, Zhan Y, Yang Y, Xie P. Proteomic analysis reveals energy metabolic dysfunction and neurogenesis in the prefrontal cortex of a lipopolysaccharide-induced mouse model of depression. Mol Med Rep 2016; 13:1813-20. [PMID: 26718926 DOI: 10.3892/mmr.2015.4741] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 09/09/2015] [Indexed: 11/05/2022] Open
Abstract
Substantial evidence from previous studies has suggested an association between major depressive disorder (MDD) and inflammation, and previous studies have associated prefrontal cortex (PFC) dysfunction with MDD. Systemic administration of bacterial lipopolysaccharide has been used to study inflammation-associated behavioral changes in rodents. However, proteomic studies investigating PFC protein expression in an LPS-induced mouse model of depression have yet to be conducted. Using two-dimensional electrophoresis coupled with matrix-assisted laser desorption ionization-time of flight-tandem mass spectrometry, PFC proteomes were comparatively assessed in LPS-induced acute inflammation reaction mice, LPS-induced depressive-like behavior mice (Dep), and control mice. A total of 26 differentially expressed proteins were identified, two of which were selected for western blot analysis, the results of which revealed a significant increase in the expression levels of creatine kinase B and dihydropyrimidinase-like 3 in Dep mice, suggesting that changes in energy metabolism and neuro-genesis occur in the PFC of Dep mice. Further investigation on these processes and on the proteins of the PFC are required in order to elucidate the pathophysiological mechanism underlying MDD.
Collapse
Affiliation(s)
- Ziye Wang
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402460, P.R. China
| | - Wenwen Li
- Chongqing Key Laboratory of Neurobiology, Chongqing 400016, P.R. China
| | - Jin Chen
- Chongqing Key Laboratory of Neurobiology, Chongqing 400016, P.R. China
| | - Haiyang Shi
- Chongqing Key Laboratory of Neurobiology, Chongqing 400016, P.R. China
| | - Mingjun Zhao
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402460, P.R. China
| | - Hongmin You
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402460, P.R. China
| | - Chenglong Rao
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402460, P.R. China
| | - Yuan Zhan
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402460, P.R. China
| | - Yongtao Yang
- Chongqing Key Laboratory of Neurobiology, Chongqing 400016, P.R. China
| | - Peng Xie
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402460, P.R. China
| |
Collapse
|
21
|
van Zyl PJ, Dimatelis JJ, Russell VA. Behavioural and biochemical changes in maternally separated Sprague-Dawley rats exposed to restraint stress. Metab Brain Dis 2016; 31:121-33. [PMID: 26555398 DOI: 10.1007/s11011-015-9757-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 10/27/2015] [Indexed: 01/26/2023]
Abstract
Early life adversity has been associated with the development of various neuropsychiatric disorders in adulthood such as depression and anxiety. The aim of this study was to determine if stress during adulthood can exaggerate the depression-/anxiety-like behaviour observed in the widely accepted maternally separated (MS) Sprague-Dawley (SD) rat model of depression. A further aim was to determine whether the behavioural changes were accompanied by changes in hippocampal brain-derived neurotrophic factor (BDNF) and the protein profile of the prefrontal cortex (PFC). Depression-/anxiety-like behaviour was measured in the elevated plus maze, open field and forced swim test (FST) in the MS SD rats exposed to chronic restraint stress in adulthood. As expected, MS increased immobility of SD rats in the FST but restraint stress did not enhance this effect of MS on SD rats. A proteomic analysis of the PFC revealed a decrease in actin-related proteins in MS and non-separated rats subjected to restraint stress as well as a decrease in mitochondrial energy-related proteins in the stressed rat groups. Since MS during early development causes a disruption in the hypothalamic-pituitary-adrenal axis and long-term changes in the response to subsequent stress, it may have prevented restraint stress from exerting its effects on behaviour. Moreover, the decrease in proteins related to mitochondrial energy metabolism in MS rats with or without subsequent restraint stress may be related to stress per se and not depression-like behaviour, because rats subjected to restraint stress displayed similar decreases in energy-related proteins and spent less time immobile in the FST than control rats.
Collapse
Affiliation(s)
- P J van Zyl
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Observatory, 7925, South Africa.
| | - J J Dimatelis
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Observatory, 7925, South Africa
| | - V A Russell
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Observatory, 7925, South Africa
| |
Collapse
|
22
|
The efficacy of early iron supplementation on postpartum depression, a randomized double-blind placebo-controlled trial. Eur J Nutr 2015; 56:901-908. [DOI: 10.1007/s00394-015-1140-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 12/15/2015] [Indexed: 10/22/2022]
|
23
|
Pan Y, Lü P, Yin L, Chen K, He Y. Effect of fluoride on the proteomic profile of the hippocampus in rats. ACTA ACUST UNITED AC 2015; 70:151-7. [PMID: 26075534 DOI: 10.1515/znc-2014-4158] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 05/19/2015] [Indexed: 11/15/2022]
Abstract
Two-dimensional gel electrophoresis (2-DE) was used to detect fluoride-induced alterations in the proteome of the rat hippocampus. Male Sprague-Dawley rats (n=30) were subjected to treatments three weeks after weaning. Animals of the first group were injected intraperitoneally (i.p.) with aqueous NaF (20 mg/kg/body weight/day), the second group, injected with physiological saline, served as the control. After 30 days, the body weight of the fluoride-treated rats was lower than that of the control, and F- levels in serum were higher than in the control. The hippocampus was subjected to proteomic analysis, and the fluoride-treated group was found to contain 19 up-regulated and eight down-regulated proteins. The proteins, identified by mass-spectroscopic analysis of their fragments obtained after digestion, were found to be involved in amino acid biosynthesis, the insulin signaling pathway and various other crucial functions. Our results also provide useful information on the mechanism of the reduction of the learning ability and memory induced by F.
Collapse
|
24
|
Li Q, Yang D, Wang J, Liu L, Feng G, Li J, Liao J, Wei Y, Li Z. Reduced amount of olfactory receptor neurons in the rat model of depression. Neurosci Lett 2015; 603:48-54. [PMID: 26170245 DOI: 10.1016/j.neulet.2015.07.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 06/29/2015] [Accepted: 07/03/2015] [Indexed: 11/17/2022]
Abstract
Reduced olfactory sensitivity has been reported in depressive disorder. However, the pathological mechanism is still unclear. The reduced olfactory bulb (OB) volume and reduced hippocampal neurogenesis has been unraveled in major depressive disorder (MDD). However, changes in olfactory epithelium (OE) have not been reported, which might contribute to olfactory deficits in MDD. In the context, we investigated the thickness of OE in a chronic unpredictable mild stress (CUMS) rat model of depression using hematoxylin and eosin (HE) staining. Simultaneously, the basal cells (labeled by nerve growth factor receptor (p75NGFR)), immature olfactory receptor neurons (ORNs) (marked by growth-associated protein 43 (GAP43)) and mature ORNs (labeled by olfactory marker protein (OMP)) in OE were detected by immunohistochemistry. The results showed that the thickness of OE, the number of basal cells, immature ORNs as well as mature ORNs decreased dramatically in the OE of CUMS rats. Those findings indicate that the reduced number of ORNs might induce OE atrophy in CUMS rats and the abnormalities of the OE may be partially responsible for the reduced olfactory sensitivity in MDD.
Collapse
Affiliation(s)
- Qianlu Li
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, China; Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Deyu Yang
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, China; Central Laboratory, Yongchuan Hospital, Chongqing Medical University, Chongqing, China; Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Juan Wang
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, China; Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Li Liu
- Department of Health Management, Yongchuan Hospital, Chongqing Medical University, Chongqing, China; Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Guibo Feng
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, China; Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Juan Li
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Juan Liao
- Central Laboratory, Yongchuan Hospital, Chongqing Medical University, Chongqing, China; Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Youdong Wei
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Zhiwei Li
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, China; Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
25
|
Han X, Shao W, Liu Z, Fan S, Yu J, Chen J, Qiao R, Zhou J, Xie P. iTRAQ-based quantitative analysis of hippocampal postsynaptic density-associated proteins in a rat chronic mild stress model of depression. Neuroscience 2015; 298:220-92. [DOI: 10.1016/j.neuroscience.2015.04.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Revised: 03/20/2015] [Accepted: 04/02/2015] [Indexed: 01/26/2023]
|
26
|
Carboni L. The contribution of proteomic studies in humans, animal models, and after antidepressant treatments to investigate the molecular neurobiology of major depression. Proteomics Clin Appl 2015; 9:889-98. [PMID: 25488430 DOI: 10.1002/prca.201400139] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 11/03/2014] [Accepted: 12/02/2014] [Indexed: 11/07/2022]
Abstract
The neurobiological basis of major depressive disorder (MDD) is only partially understood. The proposed hypotheses postulate dysregulations of monoaminergic and other neurotransmitter pathways, impaired stress responses, insufficient neurogenetic and neurotrophic processes generating maladaptive neuroplasticity, inappropriate inflammatory and metabolic responses. Proteomic approaches can provide useful contributions to the investigation of the molecular neurobiology of MDD due to their open-ended nature. Studies performed in brain regions of MDD patients which had received antidepressant (AD) treatment showed that affected proteins mainly belonged to energy pathways, transport of molecules, signaling, and synaptic transmission. Studies performed in animal models offer the advantage of more controlled experimental conditions at the expense of potential loss in relevance. The design of proteomic investigations included experiments carried out in MDD models, in naive animals treated with ADs, and in animal models subjected to AD treatments. A comparison of results suggested an overlap of several modulated pathways between MDD patients and animal models. Examples include the regulation of energy metabolism, especially oxidative phosphorylation and glycolysis, signal transduction pathways, including calcium-calmodulin kinase II, synaptic proteins, and cytoskeletal proteins. Nevertheless, the paucity of studies performed in human brains requires additional studies to confirm the correspondence.
Collapse
Affiliation(s)
- Lucia Carboni
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, Bologna, Italy
| |
Collapse
|
27
|
Mallei A, Failler M, Corna S, Racagni G, Mathé AA, Popoli M. Synaptoproteomic analysis of a rat gene-environment model of depression reveals involvement of energy metabolism and cellular remodeling pathways. Int J Neuropsychopharmacol 2015; 18:pyu067. [PMID: 25522407 PMCID: PMC4360251 DOI: 10.1093/ijnp/pyu067] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Major depression is a severe mental illness that causes heavy social and economic burdens worldwide. A number of studies have shown that interaction between individual genetic vulnerability and environmental risk factors, such as stress, is crucial in psychiatric pathophysiology. In particular, the experience of stressful events in childhood, such as neglect, abuse, or parental loss, was found to increase the risk for development of depression in adult life. Here, to reproduce the gene x environment interaction, we employed an animal model that combines genetic vulnerability with early-life stress. METHODS The Flinders Sensitive Line rats (FSL), a validated genetic animal model of depression, and the Flinders Resistant Line (FRL) rats, their controls, were subjected to a standard protocol of maternal separation (MS) from postnatal days 2 to 14. A basal comparison between the two lines for the outcome of the environmental manipulation was performed at postnatal day 73, when the rats were into adulthood. We carried out a global proteomic analysis of purified synaptic terminals (synaptosomes), in order to study a subcellular compartment enriched in proteins involved in synaptic function. Two-dimensional gel electrophoresis (2-DE), mass spectrometry, and bioinformatic analysis were used to analyze proteins and related functional networks that were modulated by genetic susceptibility (FSL vs. FRL) or by exposure to early-life stress (FRL + MS vs. FRL and FSL + MS vs. FSL) RESULTS We found that, at a synaptic level, mainly proteins and molecular pathways related to energy metabolism and cellular remodeling were dysregulated. CONCLUSIONS The present results, in line with previous works, suggest that dysfunction of energy metabolism and cytoskeleton dynamics at a synaptic level could be features of stress-related pathologies, in particular major depression.
Collapse
Affiliation(s)
- Alessandra Mallei
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and Center of Excellence on Neurodegenerative Diseases, University of Milano, Milano, Italy (Drs Mallei, Failler, Corna, Racagni, and Popoli); Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden (Dr Mathé).
| | - Marion Failler
- *Present address: Université Paris Descartes, Inserm U1163, Imagine Institute, Necker Hospital, Paris.
| | | | | | | | | |
Collapse
|
28
|
Amino acid metabolic dysfunction revealed in the prefrontal cortex of a rat model of depression. Behav Brain Res 2014; 278:286-92. [PMID: 24861712 DOI: 10.1016/j.bbr.2014.05.027] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 05/04/2014] [Accepted: 05/07/2014] [Indexed: 12/30/2022]
Abstract
Major depressive disorder (MDD) is a debilitating mood disorder. However, the molecular mechanism(s) underlying depression remain largely unknown. Here, we applied a GC-MS-based metabonomic approach in the chronic unpredictable mild stress (CUMS) model, a well-established rodent model of depression, to investigate significant metabolic changes in the rat prefrontal cortex (PFC). Multivariate statistical analysis - including principal component analysis, partial least squares-discriminate analysis, and pair-wise orthogonal projections to latent structures discriminant - was applied to identify differential PFC metabolites between CUMS rats and healthy controls. As compared to healthy control rats, CUMS rats were characterized by lower levels of isoleucine and glycerol in combination with higher levels of N-acetylaspartate and β-alanine. These findings should provide insight into the pathophysiological mechanism(s) underlying MDD and preliminary leads relevant to diagnostic biomarker discovery for depression.
Collapse
|
29
|
|
30
|
Biomarkers in major depressive disorder: the role of mass spectrometry. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 806:545-60. [PMID: 24952202 DOI: 10.1007/978-3-319-06068-2_27] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Major depressive disorder (MDD) is common. Despite numerous available treatments, many individuals fail to improve clinically. MDD continues to be diagnosed exclusively via behavioral rather than biological methods. Biomarkers-which include measurements of genes, proteins, and patterns of brain activity-may provide an important objective tool for the diagnosis of MDD or in the rational selection of treatments. Proteomic analysis and validation of its results as biomarkers is less explored than other areas of biomarker research in MDD. Mass spectrometry (MS) is a comprehensive, unbiased means of proteomic analysis, which can be complemented by directed protein measurements, such as Western Blotting. Prior studies have focused on MS analysis of several human biomaterials in MDD, including human post-mortem brain, cerebrospinal fluid (CSF), blood components, and urine. Further studies utilizing MS and proteomic analysis in MDD may help solidify and establish biomarkers for use in diagnosis, identification of new treatment targets, and understanding of the disorder. The ultimate goal is the validation of a biomarker or a biomarker signature that facilitates a convenient and inexpensive predictive test for depression treatment response and helps clinicians in the rational selection of next-step treatments.
Collapse
|
31
|
The potential of biomarkers in psychiatry: focus on proteomics. J Neural Transm (Vienna) 2013; 122 Suppl 1:S9-18. [DOI: 10.1007/s00702-013-1134-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 12/02/2013] [Indexed: 02/06/2023]
|
32
|
Proteomics reveals energy and glutathione metabolic dysregulation in the prefrontal cortex of a rat model of depression. Neuroscience 2013; 247:191-200. [DOI: 10.1016/j.neuroscience.2013.05.031] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Revised: 05/02/2013] [Accepted: 05/19/2013] [Indexed: 10/26/2022]
|
33
|
Chronic Exposure to Light Reverses the Effect of Maternal Separation on Proteins in the Prefrontal Cortex. J Mol Neurosci 2013; 51:835-43. [DOI: 10.1007/s12031-013-0071-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2013] [Accepted: 07/08/2013] [Indexed: 10/26/2022]
|
34
|
Ngounou Wetie AG, Sokolowska I, Wormwood K, Beglinger K, Michel TM, Thome J, Darie CC, Woods AG. Mass spectrometry for the detection of potential psychiatric biomarkers. J Mol Psychiatry 2013; 1:8. [PMID: 25408901 PMCID: PMC4223884 DOI: 10.1186/2049-9256-1-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Accepted: 02/12/2013] [Indexed: 12/20/2022] Open
Abstract
The search for molecules that can act as potential biomarkers is increasing in the scientific community, including in the field of psychiatry. The field of proteomics is evolving and its indispensability for identifying biomarkers is clear. Among proteomic tools, mass spectrometry is the core technique for qualitative and quantitative identification of protein markers. While significant progress has been made in the understanding of biomarkers for neurodegenerative diseases such as Alzheimer's disease, multiple sclerosis and Parkinson's disease, psychiatric disorders have not been as extensively investigated. Recent and successful applications of mass spectrometry-based proteomics in fields such as cardiovascular disease, cancer, infectious diseases and neurodegenerative disorders suggest a similar path for psychiatric disorders. In this brief review, we describe mass spectrometry and its use in psychiatric biomarker research and highlight some of the possible challenges of undertaking this type of work. Further, specific examples of candidate biomarkers are highlighted. A short comparison of proteomic with genomic methods for biomarker discovery research is presented. In summary, mass spectrometry-based techniques may greatly facilitate ongoing efforts to understand molecular mechanisms of psychiatric disorders.
Collapse
Affiliation(s)
- Armand G Ngounou Wetie
- Biochemistry and Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Avenue, Potsdam, NY 13699-5810 USA
| | - Izabela Sokolowska
- Biochemistry and Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Avenue, Potsdam, NY 13699-5810 USA
| | - Kelly Wormwood
- Biochemistry and Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Avenue, Potsdam, NY 13699-5810 USA
| | - Katherine Beglinger
- Biochemistry and Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Avenue, Potsdam, NY 13699-5810 USA
| | - Tanja Maria Michel
- Department of Psychiatry, University of Rostock, Rostock, Gehlsheimer Straße 20, D-18147 Germany
| | - Johannes Thome
- Department of Psychiatry, University of Rostock, Rostock, Gehlsheimer Straße 20, D-18147 Germany ; College of Medicine, Swansea University, Singleton Park, Swansea, SA2 8PP UK
| | - Costel C Darie
- Biochemistry and Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Avenue, Potsdam, NY 13699-5810 USA
| | - Alisa G Woods
- Biochemistry and Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Avenue, Potsdam, NY 13699-5810 USA ; Neuropsychology Clinic and Psychoeducation Services, SUNY Plattsburgh, 101 Broad St, Plattsburgh, 12901 NY USA
| |
Collapse
|
35
|
Hu Y, Zhou J, Fang L, Liu H, Zhan Q, Luo D, Zhou C, Chen J, Li Q, Xie P. Hippocampal synaptic dysregulation of exo/endocytosis-associated proteins induced in a chronic mild-stressed rat model. Neuroscience 2013; 230:1-12. [DOI: 10.1016/j.neuroscience.2012.08.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 08/04/2012] [Accepted: 08/14/2012] [Indexed: 10/28/2022]
|
36
|
Bousette N, Gramolini AO, Kislinger T. Proteomics-based investigations of animal models of disease. Proteomics Clin Appl 2012; 2:638-53. [PMID: 21136864 DOI: 10.1002/prca.200780043] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cells contain a large yet, constant genome, which contains all the coding information necessary to sustain cellular physiology. However, proteins are the end products of genes, and hence dictate the phenotype of cells and tissues. Therefore, proteomics can provide key information for the elucidation of physiological and pathophysiological mechanisms by identifying the protein profile from cells and tissues. The relatively novel techniques used for the study of proteomics thus have the potential to improve diagnostic, prognostic, as well as therapeutic avenues. In this review, we first discuss the benefits of animal models over the use of human samples for the proteomic analysis of human disease. Next, we aim to demonstrate the potential of proteomics in the elucidation of disease mechanisms that may not be possible by other conventional technologies. Following this, we describe the use of proteomics for the analysis of PTM and protein interactions in animal models and their relevance to the study of human disease. Finally, we discuss the development of clinical biomarkers for the early diagnosis of disease via proteomic analysis of animal models. We also discuss the development of standard proteomes and relate how this data will benefit future proteomic research. A comprehensive review of all animal models used in conjunction with proteomics is beyond the scope of this manuscript. Therefore, we aimed to cover a large breadth of topics, which together, demonstrate the potential of proteomics as a powerful tool in biomedical research.
Collapse
Affiliation(s)
- Nicolas Bousette
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada; Heart and Stroke/Richard Lewar Centre of Cardiovascular Excellence, Toronto, Ontario, Canada
| | | | | |
Collapse
|
37
|
Zhang R, Yang D, Zhou C, Cheng K, Liu Z, Chen L, Fang L, Xie P. β-actin as a loading control for plasma-based Western blot analysis of major depressive disorder patients. Anal Biochem 2012; 427:116-20. [PMID: 22617797 DOI: 10.1016/j.ab.2012.05.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Revised: 05/09/2012] [Accepted: 05/13/2012] [Indexed: 12/14/2022]
Abstract
Western blot analysis is a commonly used technique for determining specific protein levels in clinical samples. For normalization of protein levels in Western blot, a suitable loading control is required. On account of its relatively high and constant expression, β-actin has been widely employed in Western blot of cell cultures and tissue extracts. However, β-actin's presence in human plasma and this protein's putative role as a plasma-based loading control for Western blot analysis remain unknown. In this study, an enzyme-linked immunosorbent assay was used to determine the concentration of β-actin in human plasma, which is 6.29±0.54 ng/ml. In addition, the linearity of β-actin immunostaining and loaded protein amount was evaluated by Western blot, and a fine linearity (R²=0.974±0.012) was observed. Furthermore, the expression of plasma β-actin in major depressive disorder subjects and healthy controls was compared. The data revealed no statistically significant difference between these two groups. Moreover, the total coefficient of variation for β-actin expression in the two groups was 9.2±1.2%. These findings demonstrate that β-actin is present in human plasma and may possibly be used as a suitable loading control for plasma-based Western blot analysis in major depressive disorder.
Collapse
Affiliation(s)
- Rufang Zhang
- Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China; Institute of Neuroscience, Chongqing Medical University, Chongqing 400016, China
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Prunet P, Øverli Ø, Douxfils J, Bernardini G, Kestemont P, Baron D. Fish welfare and genomics. FISH PHYSIOLOGY AND BIOCHEMISTRY 2012; 38:43-60. [PMID: 21671026 DOI: 10.1007/s10695-011-9522-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Accepted: 05/31/2011] [Indexed: 05/30/2023]
Abstract
There is a considerable public and scientific debate concerning welfare of fish in aquaculture. In this review, we will consider fish welfare as an integration of physiological, behavioral, and cognitive/emotional responses, all of which are essentially adaptative responses to stressful situations. An overview of fish welfare in this context suggests that understanding will rely on knowledge of all components of allostatic responses to stress and environmental perturbations. The development of genomic technologies provides new approaches to this task, exemplified by how genome-wide analysis of genetic structures and corresponding expression patterns can lead to the discovery of new aspects of adaptative responses. We will illustrate how the genomic approach may give rise to new biomarkers for fish welfare and also increase our understanding of the interaction between physiological, behavioral, and emotional responses. In a first part, we present data on expression of candidate genes selected a priori. This is a common avenue to develop molecular biomarkers capable of diagnosing a stress condition at its earliest onset, in order to allow quick corrective intervention in an aquaculture setting. However, most of these studies address isolated physiological functions and stress responses that may not be truly indicative of animal welfare, and there is only rudimentary understanding of genes related to possible cognitive and emotional responses in fish. We also present an overview on transcriptomic analysis related to the effect of aquaculture stressors, environmental changes (temperature, salinity, hypoxia), or concerning specific behavioral patterns. These studies illustrate the potential of genomic approaches to characterize the complexity of the molecular mechanisms which underlies not only physiological but also behavioral responses in relation to fish welfare. Thirdly, we address proteomic studies on biological responses to stressors such as salinity change and hypoxia. We will also consider proteomic studies developed in mammals in relation to anxiety and depressive status which may lead to new potential candidates in fish. Finally, in the conclusion, we will suggest new developments to facilitate an integrated view of fish welfare. This includes use of laser microdissection in the transcriptomic/proteomic studies, development of meta-analysis methods for extracting information from genomic data sets, and implementation of technological advances for high-throughput proteomic studies. Development of these new approaches should be as productive for our understanding of the biological processes underlying fish welfare as it has been for the progress of pathophysiological research.
Collapse
Affiliation(s)
- P Prunet
- UR1037 SCRIBE, IFR140, INRA, Campus de Beaulieu, Rennes, France.
| | | | | | | | | | | |
Collapse
|
39
|
Filiou MD, Martins-de-Souza D, Guest PC, Bahn S, Turck CW. To label or not to label: Applications of quantitative proteomics in neuroscience research. Proteomics 2012; 12:736-47. [DOI: 10.1002/pmic.201100350] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Revised: 10/18/2011] [Accepted: 10/24/2011] [Indexed: 01/09/2023]
|
40
|
Increased apoptosis and different regulation of pro-apoptosis protein bax and anti-apoptosis protein bcl-2 in the olfactory bulb of a rat model of depression. Neurosci Lett 2011; 504:18-22. [DOI: 10.1016/j.neulet.2011.08.046] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2011] [Revised: 08/07/2011] [Accepted: 08/22/2011] [Indexed: 11/23/2022]
|
41
|
Reduced neurogenesis and pre-synaptic dysfunction in the olfactory bulb of a rat model of depression. Neuroscience 2011; 192:609-18. [DOI: 10.1016/j.neuroscience.2011.06.043] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Revised: 05/20/2011] [Accepted: 06/15/2011] [Indexed: 01/19/2023]
|
42
|
Christensen T, Bisgaard CF, Wiborg O. Biomarkers of anhedonic-like behavior, antidepressant drug refraction, and stress resilience in a rat model of depression. Neuroscience 2011; 196:66-79. [PMID: 21889970 DOI: 10.1016/j.neuroscience.2011.08.024] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 08/04/2011] [Accepted: 08/12/2011] [Indexed: 01/08/2023]
Abstract
The aim of the present study was to identify potential biomarkers for depression in the search for novel disease targets and treatment regimens. Furthermore, the study includes a search for biomarkers involved in treatment resistance and stress resilience in order to investigate mechanisms underlying antidepressant drug refraction and stress-coping strategies. Depression-related transcriptomic changes in gene expression profiles were investigated in laser-captured microdissected (LCM) rat hippocampal granular cell layers (GCL) using the chronic mild stress (CMS) rat model of depression and chronic administration of two selective serotonin reuptake inhibitors (SSRIs), escitalopram and sertraline. CMS rats were segregated into diverging groups according to behavioral readouts, and under stringent constraints, the associated differential gene regulations were analyzed. Accordingly, we identified four genes associated with recovery, two genes implicated in treatment resistance, and three genes involved in stress resilience. The identified genes associated with mechanisms of cellular plasticity, including signal transduction, cell proliferation, cell differentiation, and synaptic release. Hierarchical clustering analysis confirmed the subgroup segregation pattern in the CMS model. Thus antidepressant treatment refractors cluster with anhedonic-like rats, and, interestingly, stress-resilient rats cluster with rats undergoing antidepressant-mediated recovery from anhedonia, suggesting antidepressant mechanisms of action to emulate endogenous stress-coping strategies.
Collapse
Affiliation(s)
- T Christensen
- Centre for Psychiatric Research, Aarhus University Hospital, Risskov, Denmark
| | | | | |
Collapse
|
43
|
Yoo DY, Yoo KY, Choi JW, Kim W, Lee CH, Choi JH, Park JH, Won MH, Hwang IK. Time course of postnatal distribution of doublecortin immunoreactive developing/maturing neurons in the somatosensory cortex and hippocampal CA1 region of C57BL/6 mice. Cell Mol Neurobiol 2011; 31:729-36. [PMID: 21360195 PMCID: PMC11498416 DOI: 10.1007/s10571-011-9670-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Accepted: 02/18/2011] [Indexed: 11/25/2022]
Abstract
In this study, we observed neuroblast differentiation in the somatosensory cortex (SSC) and hippocampal CA1 region (CA1), which is vulnerable to oxidative stress, of the mouse at various early postnatal days (P) 1, 7, 14, and 21 using doublecortin (DCX, a marker for neuroblasts). Cresyl violet and NeuN (Neuronal Nuclei) staining showed development of layers as well as neurons in the SSC and CA1. At P1, DCX-positive neuroblasts expressed strong DCX immunoreactivity in both the SSC and CA1. Thereafter, DCX immunoreactivity was decreased with time. At P7, many DCX-immunoreactive neuroblasts were well detected in the SSC and CA1. At P14, some DCX-positive neuroblasts were found in the SSC and CA1: The immunoreactivity was weak. At P21, DCX immunoreactivity was hardly found in cells in the SSC and CA1. These results suggest that DCX-positive neuroblasts were significantly decreased in the mouse SSC and CA1 from P14.
Collapse
Affiliation(s)
- Dae Young Yoo
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 151-742 South Korea
| | - Ki-Yeon Yoo
- Department of Oral Anatomy, School of Dentistry, Gangneung-Wonju National University, Gangneung, 210-702 South Korea
| | - Ji Won Choi
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 151-742 South Korea
| | - Woosuk Kim
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 151-742 South Korea
| | - Choong Hyun Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 200-701 South Korea
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 151-742 South Korea
| | - Jung Hoon Choi
- Department of Anatomy, College of Veterinary Medicine, Kangwon National University, Chuncheon, 200-701 South Korea
| | - Jeong Ho Park
- Division of Applied Chemistry and Biotechnology, Hanbat National University, Daejeon, 305-719 South Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 200-701 South Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 151-742 South Korea
| |
Collapse
|
44
|
Liu Y, Yang N, Hao W, Zhao Q, Ying T, Liu S, Li Q, Liang Y, Wang T, Dong Y, Ji C, Zuo P. Dynamic proteomic analysis of protein expression profiles in whole brain of Balb/C mice subjected to unpredictable chronic mild stress: implications for depressive disorders and future therapies. Neurochem Int 2011; 58:904-13. [PMID: 21349309 DOI: 10.1016/j.neuint.2011.02.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Revised: 02/11/2011] [Accepted: 02/16/2011] [Indexed: 01/26/2023]
Abstract
The etiology and pathophysiology of depression remain unknown. Previous works were mostly performed on single observation time-point which might be insufficiently to reveal the molecular events changed during the disease development. Adult BALB/c mice were exposed to unpredictable chronic mild stress (UCMS) for different periods and differential 2D gel electrophoresis (DIGE) approach was employed to the brain tissue to explore the molecular disease signatures. Sustained elevation of corticosterone level was observed, suggesting the hyperactivity of hypothalamic-pituitary-adrenal (HPA) axis when the mice were subjected to the stressful situation. The behavioral results indicated the depressive alterations of the mice exposing to UCMS. The altered proteins identified by proteomics showed that abnormal energy mobilization under stress condition was accompanied by overproduction of reactive oxygen species (ROS) and endoplasmic reticulum (ER) stress. Cytoskeleton protein and anti-oxidant enzymes were also changed by UCMS treatment. The results of biochemical and immunohistochemical assay confirmed the changes identified by DIGE analysis. These results indicated that the insufficiency of ATP synthesis, overwhelming ROS production and ER stress subsequently contributed to the cytoskeletal damage and inhibition to expression of some anti-oxidant proteins, which might ultimately bring functional neuron to apoptosis or death. Proteins whose expression is affected may provide tools for potential treatment strategies.
Collapse
Affiliation(s)
- Yanyong Liu
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 5 Dongdansantiao, Beijing 100005, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Whittle N, Li L, Chen WQ, Yang JW, Sartori SB, Lubec G, Singewald N. Changes in brain protein expression are linked to magnesium restriction-induced depression-like behavior. Amino Acids 2011; 40:1231-48. [DOI: 10.1007/s00726-010-0758-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2010] [Accepted: 01/31/2011] [Indexed: 01/22/2023]
|
46
|
Taurines R, Dudley E, Grassl J, Warnke A, Gerlach M, Coogan AN, Thome J. Proteomic research in psychiatry. J Psychopharmacol 2011; 25:151-96. [PMID: 20142298 DOI: 10.1177/0269881109106931] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Psychiatric disorders such as Alzheimer's disease, schizophrenia and mood disorders are severe and disabling conditions of largely unknown origin and poorly understood pathophysiology. An accurate diagnosis and treatment of these disorders is often complicated by their aetiological and clinical heterogeneity. In recent years proteomic technologies based on mass spectrometry have been increasingly used, especially in the search for diagnostic and prognostic biomarkers in neuropsychiatric disorders. Proteomics enable an automated high-throughput protein determination revealing expression levels, post-translational modifications and complex protein-interaction networks. In contrast to other methods such as molecular genetics, proteomics provide the opportunity to determine modifications at the protein level thereby possibly being more closely related to pathophysiological processes underlying the clinical phenomenology of specific psychiatric conditions. In this article we review the theoretical background of proteomics and its most commonly utilized techniques. Furthermore the current impact of proteomic research on diverse psychiatric diseases, such as Alzheimer's disease, schizophrenia, mood and anxiety disorders, drug abuse and autism, is discussed. Proteomic methods are expected to gain crucial significance in psychiatric research and neuropharmacology over the coming decade.
Collapse
Affiliation(s)
- Regina Taurines
- Academic Unit of Psychiatry, The School of Medicine, Institute of Life Science, Swansea University, Singleton Park, Swansea SA2 8PP, UK
| | | | | | | | | | | | | |
Collapse
|
47
|
Filiou MD, Turck CW, Martins-de-Souza D. Quantitative proteomics for investigating psychiatric disorders. Proteomics Clin Appl 2010; 5:38-49. [DOI: 10.1002/prca.201000060] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2010] [Revised: 09/09/2010] [Accepted: 09/20/2010] [Indexed: 12/21/2022]
|
48
|
Intranasal administration of nerve growth factor produces antidepressant-like effects in animals. Neurochem Res 2010; 35:1302-14. [PMID: 20521102 DOI: 10.1007/s11064-010-0183-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2010] [Indexed: 12/15/2022]
Abstract
Many works showed that nerve growth factor (NGF) injected into the brain of animal model emerges potential antidepressant effects. However, this route of administration significantly restricts the application of NGF clinically. Here, we reported that intranasal NGF could provide an alternative to intraventricular injection. The behavioral analysis showed that intranasal administration of NGF reduced the immobility time in forced swimming test (FST) and tail suspension test (TST) in mice. Likewise, intranasal NGF increased the sucrose intake and the locomotor activity in rats after unpredictable chronic mild stress (UCMS). Furthermore, intranasal NGF increased the levels of monoamine neurotransmitters (norepinephrine, dopamine) in the frontal cortex and hippocampus and affected the number of 5-bromodeoxyuridine (BrdU), c-fos and caspase-3 positive neurons in dentate gyrus of hippocampus in rats after UCMS. In summary, intranasal NGF had significant antidepressant effects on animal models of depression and this route of administration may provide a promising way to deliver NGF to brain in a therapeutic perspective.
Collapse
|
49
|
Tian WM, Zhang KR, Zhang J, Shen Y, Xu Q. Association between the Epidermal Growth Factor Gene and Intelligence in Major Depression Patients. ACTA ACUST UNITED AC 2010; 25:105-8. [DOI: 10.1016/s1001-9294(10)60031-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
50
|
The effects of perceived emotional distress on language performance in intractable epilepsy. Epilepsy Behav 2010; 18:64-73. [PMID: 20478748 DOI: 10.1016/j.yebeh.2010.02.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Revised: 02/08/2010] [Accepted: 02/19/2010] [Indexed: 11/22/2022]
Abstract
We evaluated the potential moderating effect of emotional distress (Minnesota Multiphasic Personality Inventory 2, scales D and Pt) on language functioning (i.e., Boston Naming Test, phonemic paraphasic error production on the Boston Naming Test, Controlled Oral Word Association Task, Animal Naming, Token Test) in patients with left (N=43) and right (N=34) mesial temporal lobe epilepsy (MTLE) and frontal lobe epilepsy (FLE) (N=30). Video/EEG and brain imaging results confirmed localization. Logistic regression models revealed that perceived emotional distress moderated language performance. Performance of patients with left MTLE and that of patients with FLE were equally poor across language measures. Performance of patients with right MTLE was intact. Depression and anxiety differentially moderated performance. Anxiety was associated with better performance in patients with FLE on classically temporal lobe-mediated tasks (Boston Naming Test). Depression was associated with worse language performance on measures for which impaired performance was traditionally intrinsic to the underlying epileptogenic lesion (word fluency in FLE). Emotional distress influences language performance. Adequate treatment of mood should be considered when managing pharmacoresistant epilepsy.
Collapse
|