Back to Journals » International Journal of Nanomedicine » Volume 19
Investigating Neuroplasticity Changes Reflected by BDNF Levels in Astrocyte-Derived Extracellular Vesicles in Patients with Depression
Authors Li K , Wang K, Xu SX , Xie XH, Tang Y, Zhang L, Liu Z
Received 12 May 2024
Accepted for publication 28 August 2024
Published 2 September 2024 Volume 2024:19 Pages 8971—8985
DOI https://doi.org/10.2147/IJN.S477482
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 2
Editor who approved publication: Prof. Dr. RDK Misra
Kun Li,1,2 Kun Wang,3 Shu-Xian Xu,1 Xin-Hui Xie,1 Yan Tang,3 Lihong Zhang,2 Zhongchun Liu1,4
1Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China; 2Clinical Laboratory, Affiliated Hospital of West Anhui Health Vocational College, Lu’an, Anhui, People’s Republic of China; 3Department of Psychiatry, Affied Hospital of West Anhui Health Vocational College, Lu’an, Anhui, People’s Republic of China; 4Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, People’s Republic of China
Correspondence: Zhongchun Liu, Department of Psychiatry, Renmin Hospital of Wuhan University, No. 99 Jiefang Road, Wuchang District, Wuhan, Hubei, 430060, People’s Republic of China, Email [email protected]
Purpose: To investigate the neuroplasticity hypothesis of depression by measuring brain-derived neurotrophic factor (BDNF) levels in plasma astrocyte-derived extracellular vesicles (ADEVs) and to evaluate their potential as biomarkers for depression compared with plasma BDNF levels.
Patients and Methods: Thirty-five patients with major depressive disorder (MDD) and 35 matched healthy controls (HCs) were enrolled. Plasma ADEVs were isolated using a combination of ultracentrifugation and immunoaffinity capture. Isolated ADEVs were validated using transmission electron microscopy, nanoparticle tracking analysis, and Western blotting. BDNF levels were quantified in both ADEVs and plasma. ALG-2-interacting protein X (Alix) and cluster of differentiation 81 (CD81) levels, two established extracellular vesicle markers, were measured in ADEVs.
Results: After false discovery rate correction, patients with MDD exhibited higher CD81 levels (PFDR = 0.040) and lower BDNF levels (PFDR = 0.043) in ADEVs than HCs at baseline. BDNF levels in ADEVs normalized to CD81 (PFDR = 0.002) and Alix (PFDR = 0.040) remained consistent with this finding. Following four weeks of selective serotonin reuptake inhibitor treatment (n=10), CD81 levels in ADEVs decreased (PFDR = 0.046), while BDNF levels normalized to CD81 increased (PFDR = 0.022). BDNF levels in ADEVs were more stable than in plasma. Exploratory analysis revealed no correlation between BDNF levels in ADEVs and plasma (ρ=0.117, P = 0.334).
Conclusion: This study provides human in vivo evidence supporting the neuroplasticity hypothesis of depression by demonstrating altered BDNF levels in ADEVs. ADEVs may be more suitable for developing biomarkers of depression than plasma-derived biomarkers.
Keywords: major depressive disorder, ADEV, BDNF, biomarker, treatment
Introduction
Major depressive disorder (MDD) is a severe mental illness characterized by persistent low mood, loss of interest, and anhedonia.1 MDD increases the risk of suicide and represents a significant global public health concern.2 Approximately 5% of the global population experiences depression each year, with the highest prevalence observed in young individuals.3 Despite this significant health burden, depression remains a largely neglected health crisis. The global prevalence of depression reached 279.6 million individuals in 2019, incurring a substantial impact on individuals, families, and society.4
The pathogenesis of depression is very complex, and various hypotheses have been proposed to explain its development. Of these, the neuroplasticity hypothesis is a widely studied and recognized theory.5–7 Neuroplasticity refers to the nervous system’s ability to reorganize its structure, function, and connections in response to intrinsic or extrinsic stimuli8 and regulated by neurotrophic factors and their associated signaling pathways.9,10 Neurotrophic factors not only support the metabolic needs of neurons but also play vital roles in their proliferation and survival. Functionally, they can act as signaling molecules that mediate learning and memory processes in the brain.11 The main neurotrophic factors include brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), neurotrophin 3 (NT-3), and neurotrophin 4/5 (NT-4/5), with previous studies on the neuroplasticity hypothesis of MDD mainly focusing on BDNF. A meta-analysis of 1095 drug-naïve first-episode MDD patients and 1399 healthy controls (HCs) revealed a trend towards decreased BDNF levels in the peripheral blood of MDD patients.12 Furthermore, the neuroplasticity hypothesis may explain the delayed onset of efficacy of antidepressant drugs.13 Therefore, neurotrophic factors and their signaling pathways are considered as primary therapeutic targets in depression.14
Despite the data implicating neurotrophic factors in depression, the results are heterogeneous due to variations in clinical samples and cohorts, treatment modalities, detection techniques, and analytical methods. To minimize the potential influence of clinical factors, Chiou et al conducted a six-year longitudinal study of 71 first-episode drug-naïve patients with MDD rigorously matched with HCs.15 They observed significantly lower baseline serum BDNF levels in patients with MDD compared with HCs. Conversely, Hung et al found no significant differences in serum BDNF levels in patients with MDD using age-adjusted covariance analysis.16 Several longitudinal studies have shown no correlation between changes in BDNF levels during treatment and improvements in depressive symptoms.17–19 Studies of other peripheral blood neurotrophic factors such as NGF, glial cell-derived neurotrophic factor (GDNF), and NT-4/5 in depression similarly show considerable heterogeneity.20–23 A recent systematic review and meta-analysis further indicated that candidate biomarker levels in the peripheral blood do not correlate significantly with those in brain tissue nor cerebrospinal fluid (CSF).24 Nevertheless, depression is a disorder characterized by structural and functional changes in the brain,25,26 and it remains unclear whether detecting peripheral circulating neurotrophic factors accurately reflect their state within the brain. Therefore, it is essential to obtain direct evidence of neurotrophic factor alterations within the brains of patients with MDD.
Astrocytes, the most abundant glial cells in the brain, play vital roles in providing nutritional support to neurons, mediating synaptic formation and function, transmitting signals across brain regions to regulate neuronal activity, and maintaining central nervous system (CNS) homeostasis.27 Astrocytes are recognized as key neuroglial cells implicated in the pathophysiology of depression,28,29 serving as crucial regulators of brain function via mechanisms such as neuroplasticity and neuroinflammation.30 In vitro, antidepressants can induce the expression and secretion of neurotrophic factors including BDNF, fibroblast growth factor 2 (FGF2), and GDNF in primary astrocytes.31 A recent meta-analysis suggested that selective serotonin reuptake inhibitors (SSRIs) exert antidepressant effects by modulating the expression of specific molecules in astrocytes.32 However, the exact mechanisms, transport, and distribution of astrocyte-related BDNF expression in patients with depression remain uncertain.
Studying astrocytes directly in the human brain is challenging due to methodological limitations; brain tissues and biofluids are not usually available from patients with MDD. Recent advances in extracellular vesicle (EV) extraction techniques offer a non-invasive approach to assessing brain status through the isolation of CNS-derived EVs.33,34 Almost all types of cells can release EVs, small vesicles that carry various biological substances such as proteins, lipids, and nucleic acids that target recipient cells as a form of intercellular communication.35,36 Since EVs can cross the blood-brain barrier (BBB) and can be isolated from various peripheral fluids,37 CNS-derived EVs may provide insights into the pathophysiology of brain-related diseases. Our previous research showed that astrocyte-derived extracellular vesicles (ADEVs) from patients with MDD contain significantly higher levels of proinflammatory factors, with higher diagnostic values compared with serum.38 This suggests that brain-derived EVs such as ADEVs can serve as a “window” into the health of the CNS39 and provide more information about the brain than peripheral blood values.
Here we aimed to investigate: (1) whether BDNF levels in ADEVs differ in patients with MDD and HCs; (2) correlations between BDNF levels in ADEVs and plasma; (3) associations between BDNF levels in ADEVs and depression severity and cognitive function; and (4) whether BDNF measured in ADEVs offers diagnostic advantages over plasma BDNF levels in patients with depression.
Materials and Methods
This observational study was conducted jointly at the Renmin Hospital of Wuhan University and the Affiliated Hospital of West Anhui Health Vocational College. All experiments were performed at the Psychiatric Center Laboratory of Renmin Hospital of Wuhan University.
Study Design
To minimize potential interference from psychotropic medications, we recruited 35 patients with MDD between April 1, 2023 to March 31, 2024 who were not taking any antidepressant medications. Drug-naïve MDD patients were prescribed selective serotonin reuptake inhibitors (SSRIs) and were followed up for four weeks, ten of whom completed four-week follow-up. Inclusion criteria were: (1) meeting ICD-10 criteria for MDD by using the Mini-International Neuropsychiatry Interview;40 (2) Hamilton Depression Rating Scale (HAMD-17)41 total score ≥ 17; (3) drug-naïve or off antidepressant medications for more than three months; (4) Han Chinese ethnicity, aged between 18 and 65; and (5) provided written informed consent. Exclusion criteria were: (1) severe physical illness, especially neurological disorders; (2) infection, fever, or allergies within the previous two weeks; (3) pregnant or lactating; (4) presence of other psychiatric disorders; and (5) deemed unsuitable for participation upon assessment by the researchers. Additionally, we recruited 35 age- and sex-matched HCs through offline advertisements during the same period. HCs were required to be physically healthy with no personal nor first-degree family history of psychiatric illness. Other exclusion criteria for HCs were identical to those for MDD patients. All participants were assessed by two experienced and rigorously trained psychiatrists to ensure consistency. Additionally, HCs underwent a comprehensive health examination at enrollment to ensure the absence of any psychiatric or significant physical illnesses.
Sample Size Calculation
Due to the absence of studies investigating neurotrophic factors in ADEVs isolated from patients with MDD, we used findings from a clinical study examining BDNF and pro-BDNF in total exosomes isolated from patients with MDD as a reference for our sample size calculation.42 In that study, the effect size of BDNF in serum total exosomes between MDD and control groups was 0.78, while pro-BDNF had an effect size of 1.16.42 We therefore set the effect size for this study at 0.75, with a power of 0.85 (1-β) and a significance level of 0.05 (two-tailed). Sample size calculations were performed using G*Power software (v3.1.9.2),43 which indicated that the study would need 66 subjects (33 per group). Finally, to account for unforeseen factors and dropout, we set the sample size to 70 (35 per group).
Clinical and Cognitive Measurements
The HAMD-17 and its subscales were used to assess depressive symptoms. The Montreal Cognitive Assessment (MoCA)44 and the Digit Symbol Substitution Test (DSST)45 were used to measure objective cognitive functioning. In our study, we conducted targeted symptom analysis of MDD patients using four subscales of the HAMD-17: Maier (items 1, 2, 7, 8, 9, and 10), anxiety/somatization (items 10, 11, 12, 13, 15, and 17), retardation (items 1, 7, 8, and 14), and sleep (items 4, 5, and 6).46 The MoCA subscales contain five domains: visuospatial, frontal functioning, language, memory, and orientation.47
Plasma and ADEV Collection
All participants fasted for at least 8 h before blood collection. Five-milliliter venous blood samples were collected from patients and HCs between 7:00 and 10:00 am into Vacutainer tubes containing EDTA-K2 anticoagulant (BD Biosciences, Franklin Lakes, NJ, USA). Blood samples were centrifuged at 2000 g for 15 min at 4°C within 2 h of collection to separate plasma. After excluding hemolysis and lipemia, plasma samples were divided into 500 μL aliquots and stored at −80°C until use. Two days before testing, plasma samples stored at the Affiliated Hospital of West Anhui Health Vocational College were transported on dry ice to the Psychiatric Center Laboratory of Renmin Hospital of Wuhan University and immediately stored in a −80°C freezer.
ADEVs were separated using a two-step method combining ultracentrifugation with immunoaffinity capture.48–50 First, total EVs (TEVs) were isolated from plasma samples using ultracentrifugation (Optima XPN-100, SW60Ti horizontal rotor; Beckman Coulter, Brea, CA, USA). Subsequently, biotinylated glutamate-aspartate transporter (GLAST) antibodies (Miltenyi Biotec, Bergisch Gladbach, Germany) targeting an astrocyte surface marker were used to isolate GLAST+ EVs, which ultimately constituted the ADEV samples (Figure 1). All samples were assayed simultaneously on the testing day. A detailed protocol describing the isolation of ADEVs is provided in the Supplementary Material 1.
![]() |
Figure 1 Protocol for the isolation of ADEVs by ultracentrifugation and immunoaffinity capture. |
Confirmation of ADEVs
Transmission Electron Microscopy
Twenty microliters of ADEV samples were added dropwise to 200-mesh grids and incubated at room temperature for 10 min. Then, grids were negatively stained with 2% phosphotungstic acid for 3 min. The remaining liquid was removed with filter paper and ADEVs observed on a JEM1400 transmission electron microscope (JEOL Ltd., Tokyo, Japan).
Nanoparticle Tracking Analysis (NTA)
A Zetaview-PMX120-Z (Particle Metrix, Meerbusch, Germany) was used for NTA along with its corresponding software ZetaView (v8.05.14 SP7) to measure particle diameter and concentration.
Western Blotting
Three EV markers [ALG-2 interacting protein-X (Alix) (Proteintech, Rosemont, IL, USA), heat shock protein 70 (HSP70) (Abcam, Cambridge, UK), cluster of differentiation (CD) 9 (Abcam)] and one astrocyte marker [glial fibrillary acidic protein (GFAP) (Abcam)] were detected, and TEV supernatant was used as a negative control.
Protein Measurements
Enzyme-linked immunosorbent assay (ELISA) kits were used to measure BDNF (Elabscience, Wuhan, China) levels in ADEVs and plasma. Additionally, levels of CD81 (CUSABIO, Wuhan, China) and Alix (CUSABIO) in ADEVs were measured.
Statistical Analyses
In the baseline descriptive data analysis, continuous numerical variables were compared using Welch’s two-sample t-tests. For categorical variable such as sex, we used Pearson’s chi-squared test. General linear models (GLMs) were used to compare differences in target proteins between the two groups, setting the group as the independent variable and target proteins as the dependent variables, while sex, age, and BMI were added as covariates. The model-fitted estimated marginal means (EMMs) and their associated 95% confidence intervals (CIs) were used to describe differences between the two groups. Spearman coefficients were employed to examine correlations between BDNF levels in plasma and ADEVs as well as with clinical and cognitive characteristics. Finally, receiver operating characteristic (ROC) curves were used to further explore the diagnostic value of BDNF in ADEVs compared with plasma in patients with MDD. Pre- and post-treatment values were compared using paired sample t-tests.
In the main analysis, we used a false discovery rate (FDR) correction (Benjamini and Hochberg method)51 to detect possible false positive results. A two-tailed P-value < 0.05 indicated a significant difference. All statistical analyses were performed in R v4.3.3, and the ggplot2 package was used for visualizations.
Results
Confirmation of ADEVs
Isolated ADEVs were validated using three methods: transmission electron microscopy, NTA, and Western blotting (Figure 2). Small vesicles were clearly observed by transmission electron microscopy, and NTA results showed that the diameter distribution of these small EVs corresponded to previous studies.52 Western blotting further confirmed the expression of three EV markers, Alix, HSP70, and CD9, as well as the astrocyte marker GFAP in ADEV samples. These results confirm successful collection of ADEVs.
Descriptive Clinical Data
The two groups of participants showed no significant differences in sex, age, BMI, and education (Table 1).
![]() |
Table 1 Comparison of Baseline Demographic and Clinical Characteristics |
Detection of Plasma Lipoproteins
Pre-isolation plasma lipid levels were quantified to evaluate potential interference in ADEV isolation. There were no significant differences in total cholesterol (TC) (Roche, Basel, Switzerland), triglycerides (TG) (Roche), high-density lipoprotein cholesterol (HDL-C) (YONGHE-SUN, Changsha, China), and low-density lipoprotein cholesterol (LDL-C) (YONGHE-SUN) levels between the two groups (Table 2).
![]() |
Table 2 Comparison of Plasma Lipid Levels Between MDD and HC Groups |
Results of the Main Analyses
At baseline, BDNF levels in ADEVs from the MDD group were significantly lower, while CD81 levels in ADEVs were significantly higher, than in controls. There were no significant differences in BDNF levels in plasma and Alix levels in ADEVs between the two groups. Furthermore, BDNF levels normalized to CD81 and Alix in ADEVs were significantly lower in the MDD group compared with the HC group, and the result remained consistent when normalized to plasma volume (Figure 3A–F). Detailed results are presented in Table 3. In ROC analysis, the area under curve (AUC) values ranged from 0.647 to 0.744 (Figure 3G; see detailed results in the figure caption).
![]() |
Table 3 Comparison of BDNF, CD81, and Alix Between MDD and HC at Baseline Based on General Linear Model |
Ten drug-naïve patients with MDD completed four weeks of follow-up after SSRI treatment. Among them, seven patients were treated with escitalopram (10 mg/day) and the other three patients were treated with sertraline (50 mg/day). Following treatment, MDD patients exhibited significant improvements in depressive symptoms. Similarly, significant improvements were observed in all cognitive domains except orientation and language (Table 4).
![]() |
Table 4 Clinical Features and Protein Changes in MDD Patients After 4 Weeks of SSRI Treatment (n = 10) |
In the longitudinal analysis, CD81 levels in ADEVs significant decreased, while BDNF levels in plasma and ADEVs, as well as Alix in ADEVs, did not significantly change after treatment. Furthermore, BDNF levels normalized to CD81 in ADEVs significantly increased after treatment, while there were no significant differences when BDNF levels were normalized to Alix in ADEVs (Figure 4A–F and Table 4).
Results of Exploratory Analyses
First, there was no significant correlation between BDNF levels in ADEVs and plasma (ρ=0.117, P=0.334) (Figure 5A). Second, there was a positive correlation between CD81 and Alix in ADEVs (ρ=0.699, P<0.001) (Figure 5B). Third, in patients with MDD, BDNF levels in ADEVs were positively correlated with CD81 (ρ=0.452, P=0.006) and Alix (ρ=0.454, P=0.006) (Figure 5C and D). Finally, there were no significant correlations between these target proteins and the severity of depression nor cognitive function (Supplementary Figure 1).
Discussion
Here, by comparing BDNF levels in ADEVs and plasma from patients with MDD and HCs, we demonstrate for the first time that detecting neurotrophic factors in ADEVs has greater value than measuring these factors in plasma. Patients with MDD had significantly higher CD81 levels and lower BDNF levels than HCs. Notably, these changes were not observed in plasma. Our exploratory follow-up study revealed that after four weeks of SSRI treatment, patients with MDD had a significant decrease in ADEV CD81 levels and a significant increase in ADEV BDNF levels normalized to CD81.
Quality Control of ADEV Isolation
We used ultracentrifugation to separate TEVs in the first purification step, which achieves highest separation purity than other size-based or precipitation-based methods.53 We also measured the concentration of plasma lipoproteins prior to ADEV extraction and detected no significant difference in plasma lipoprotein levels between the two groups. This quality control step was necessary, as lipoproteins and EVs share some physical properties, such as particle size and density,54 especially with low-density lipoproteins,55 which can lead to their co-separation during ultracentrifugation. Third, we strictly followed standardized operating procedures during ADEV isolation to minimize the introduction of human error. Together, these procedures enhanced the robustness of our results.
Astrocytes, Neuroplasticity, and Depression
Astrocytes are key participants in neuroplasticity, playing crucial roles in synaptic development and remodeling through dynamic and bidirectional interactions with neurons.56 Astrocytes regulate neuroplasticity through various mechanisms, with neurotrophic factors a pivotal component. Astrocytes activate intracellular responses by sensing extracellular BDNF through TrkB receptors. They also facilitate synaptic plasticity by taking up and circulating BDNF/pro-BDNF at synapses.57 Mesencephalic astrocyte-derived neurotrophic factor has been shown to prevent neuronal loss,58 and astrocytes can release EVs containing neurotrophic factors such as GDNF and BDNF in vitro and necroptotic astrocytes can induce neuronal apoptosis through EV-derived pro-BDNF.59 In animal experiments, conditional BDNF delivery from astrocytes contributes to the restoration of neural plasticity.60 Notably, ADEVs are crucial mediators of cellular communication and signal transduction in astrocytes, and detecting changes in the ADEV cargo provides direct evidence of astrocyte activity or dysfunction.61 Therefore, ADEV-derived BDNF may play an important role in the regulation of neural plasticity. We found that patients with MDD had lower levels of BDNF in ADEVs, consistent with the findings of a recent meta-analysis of CSF in patients with depression.62 A reduction in BDNF in ADEVs may be an important feature of impaired neural plasticity in patients with depression.
Neurotrophic Factors, Depression, and ADEVs
Previous studies on neurotrophic factors in depression have mainly focused on their levels in peripheral blood, with little data on EVs. However, the results of peripheral blood studies have yielded inconsistent and even contradictory results with respect to state or trait changes in patients with depression and whether these changes serve as predictors of treatment response. We detected no significant difference in BDNF levels in plasma between patients with MDD and HCs but we did detect significantly lower BDNF levels in ADEVs from patients with MDD. This difference may be because neurotrophic factors are not solely expressed in the nervous system. For example, platelets can store and release BDNF, and their levels are much higher than in the brain.63 A previous study reported lower serum and plasma BDNF levels in patients with depression compared with HCs, but this change was not observed in whole blood,64 suggesting that the observed differences may not be due to platelet BDNF content but rather their ability to release BDNF. It remains unknown whether MDD affects the release of platelet BDNF. Many studies have assumed that peripheral blood BDNF is a biomarker reflecting brain diseases, but there is no direct evidence of an association between peripheral blood BDNF levels and BDNF expression in the brain. Moreover, the penetration of neurotrophic factors through the BBB is generally poor, and their plasma half-life is short.9 Our study also showed no significant correlation between BDNF levels in ADEVs and plasma. The assessment of biomarkers such as neurotrophic factors in plasma/serum may be influenced by other peripheral processes in individuals, and CNS-derived EVs may largely reduce the effects of such interference. Consistent with this, previous studies have shown large inter-individual differences in plasma/serum BDNF levels.19,65 Similarly, our study revealed that the coefficient of variation (CV) of BDNF in plasma was 1.20 for MDD and 1.43 for HCs, while in ADEVs, the CVs were 0.15 and 0.19, respectively. Additionally, there is evidence that brain-derived exosomes in the peripheral blood have a similar diagnostic efficacy to CSF biomarkers, and the levels of various biomarkers in exosomes are highly correlated with those in CSF.66 ROC curve analysis showed that the AUC for diagnosing MDD using BDNF normalized to CD81 concentration in ADEVs was 0.744. Therefore, we believe that ADEVs, as a “liquid biopsy” of the CNS,67 have significant advantages over peripheral fluids for future biomarker development for psychiatric disorders like MDD.
We found that levels of BDNF normalized to CD81 in ADEVs within patients with MDD significantly increased after four weeks of SSRI treatment, while plasma BDNF levels did not significantly alter. This is consistent with previous research suggesting that peripheral measurements of neurotrophic factors may not predict treatment responses in patients with MDD.22,68 However, it is interesting to note that, compared with untreated patients with MDD, those receiving antidepressant treatment showed increased BDNF expression in the post-mortem hippocampus.69 Although we observed some changes in ADEVs from patients with MDD after antidepressant treatment, which may help to explain the delayed effects of SSRIs, the molecular characteristics of antidepressant responses are often highly heterogeneous. Considering that ADEVs originate from the CNS, it is more accurate to say that they may have an advantage in reflecting treatment responses in patients with MDD.
Exploratory Analyses
A common mechanism of astrocyte activation in many neurological disorders is an increase of reactive astrocytes, which can exert neuroprotective or neurotoxic effects.70 Astrocyte activation may be accompanied by an increase in ADEV release.71 CD81 and Alix are established biomarkers of EVs, with CD81 a transmembrane protein and Alix a cytoplasmic protein.72–74 In this study, we observed an increase in CD81 levels within ADEVs, consistent with our previous findings.50 However, Alix levels in ADEVs were not significantly different between the two groups. This might be due to variations in vesicle size. A clinical study revealed that both TEVs and CNS-EVs were smaller in patients with MDD than in HCs.75 Assuming that ADEVs are spherical, CD81 levels within ADEVs could theoretically correlate with their surface area, while Alix levels might be associated with their volume. As the radius changes, the volume of a sphere changes more than its surface area. In our exploratory analysis, we found a significant positive correlation between CD81 and Alix in ADEVs. Furthermore, at baseline, BDNF levels in ADEVs were positively correlated with both CD81 and Alix in ADEVs in patients with MDD. These findings also confirm the high purity of our ADEVs, and the release of BDNF from ADEVs might exhibit particle-dependent characteristics. This could explain why the differences in BDNF levels within ADEVs were more pronounced when normalized to CD81 and Alix. In EV research, different normalization methods may amplify or mask differences, so we suggest employing appropriate and standardized normalization methods to ensure reliability and comparability.
Limitations
Our study has several limitations. First, due to the limited volume of extracted ADEVs, we only measured BDNF levels, a representative neurotrophic factor. A comprehensive assessment of known neurotrophins and growth factors would be valuable to obtain a full profile of such factors in the brain. Second, while ultracentrifugation is considered the “gold standard” for EV separation due to its high purity,53 the recovery rate of EVs is low.76 A single-step ultracentrifugation protocol (100,000 g, 2 h) for EV isolation has a reported recovery rate of 40%, and repeating the ultracentrifugation process under the same conditions results in a significantly lower recovery rate of only 16%.53 To increase recovery of EVs, we modified the two-step ultracentrifugation to a one-step process and extended the centrifugation time.77 This adjustment may have increased co-isolated protein aggregates and lipoproteins. However, in the second separation step, we utilized immunocapture with astrocyte-specific antibodies, which mitigated against the introduction of errors. Third, due to the uncontrollable nature of clinical follow-up, only ten drug-naïve patients with MDD completed the four-week follow-up on SSRI treatment. Consequently, the sample size for longitudinal observations in this study was small, limiting our ability to conduct further analysis based on the specific SSRI used. Fourth, we attempted to quantify residual platelets in the pre-isolation plasma of ADEVs. However, the count fell below the lower detection limit of the clinical blood cell analyzer (BC-7500, Mindray, China). Fifth, lifestyle factors such as diet and exercise can also affect BDNF levels,78,79 but this study did not strictly control for these variables. Sixth, while we observed changes in CD81 levels in ADEVs from patients with MDD, we did not perform NTA on all samples. Therefore, we cannot confirm whether the quantity of ADEVs in patients with MDD also changed. The relationship between EV biomarker levels and particle counts warrants further investigation. Finally, although we detected a significant decrease in BDNF levels in ADEVs from patients with MDD, suggesting potential changes in neuroplasticity, this observation cannot establish a causal relationship between neuroplasticity and depression.
Conclusions
In conclusion, our results indicate that BDNF levels are modified in MDD patient-derived plasma ADEVs compared with HCs and after SSRI treatment, potentially reflecting changes in neuroplasticity. This study provides human in vivo evidence supporting the neuroplasticity hypothesis of depression by demonstrating altered BDNF levels in ADEVs. Furthermore, ADEVs are emerging as a novel tool for investigating astrocyte status in the brain and assessing hypotheses related to depression.
Abbreviations
MDD, Major depressive disorder; HCs, healthy controls; BDNF, brain-derived neurotrophic factor; NGF, nerve growth factor; NT-3, neurotrophin 3; NT-4/5, neurotrophin 4/5; FGF2, fibroblast growth factor 2; GDNF, glial cell-derived neurotrophic factor; SSIR, selective serotonin reuptake inhibitor; EVs, extracellular vesicles; BBB, blood-brain barrier; CNS, central nervous system; ADEVs, astrocyte-derived extracellular vesicles; CSF, cerebrospinal fluid; HAMD-17, Hamilton Depression Rating Scale; MoCA, Montreal Cognitive Assessment; DSST, Digit Symbol Substitution Test; TEVs, total Evs; NTA, Nanoparticle tracking analysis; Alix, ALG-2 interacting protein-X; CD, cluster of differentiation; HSP70, heat shock protein 70; GFAP, glial fibrillary acidic protein; ELISA, Enzyme-linked immunosorbent assay; BMI, body mass index; TC, total cholesterol; TG, triglyceride; HDL-C, high-density lipoprotein cholesterol; LDL-C, low-density lipoprotein cholesterol; GLMs, general linear models; EMMs, Estimated Marginal Means; CIs, confidence intervals; ROC, receiver operating characteristic; AUC, area under curve; FDR, false discovery rate.
Data Sharing Statement
The data that support the findings of this study are available from the corresponding author upon reasonable request.
Ethics Approval
The study followed the Declaration of Helsinki (2013 revision) and obtained approval from the Ethics Committees of Renmin Hospital of Wuhan University (WDRY2020-K191) and Affiliated Hospital of West Anhui Health Vocational College (LAEY-2023-008). Informed consent was obtained from all participants or their legal guardians before they participated in the study.
Author Contributions
All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.
Funding
This work was supported by the National Natural Science Foundation of China (U21A20364) and the National Key Research and Development Program of China (2018YFC1314600).
Disclosure
The authors report no conflicts of interest in this work.
References
1. Monroe SM, Harkness KL. Major depression and its recurrences: life course matters. Annu Rev Clin Psycho. 2022;18(1):329–357. doi:10.1146/annurev-clinpsy-072220-021440
2. Marwaha S, Palmer E, Suppes T, et al. Novel and emerging treatments for major depression. Lancet. 2023;401(10371):141–153. doi:10.1016/S0140-6736(22)02080-3
3. Herrman H, Patel V, Kieling C, et al. Time for united action on depression: a lancet-world psychiatric association commission. Lancet. 2022;399(10328):957–1022. doi:10.1016/S0140-6736(21)02141-3
4. GBD 2019 Mental Disorders Collaborators. Global, regional, and national burden of 12 mental disorders in 204 countries and territories, 1990-2019: a systematic analysis for the global burden of disease study 2019. Lancet Psychiat. 2022;9(2):137–150. doi:10.1016/S2215-0366(21)00395-3
5. Malhi GS, Mann JJ. Depression. Lancet. 2018;392(10161):2299–2312. doi:10.1016/S0140-6736(18)31948-2
6. Boku S, Nakagawa S, Toda H, et al. Neural basis of major depressive disorder: beyond monoamine hypothesis. Psychiat Clin Neuros. 2018;72(1):3–12. doi:10.1111/pcn.12604
7. Esalatmanesh S, Kashani L, Akhondzadeh S. Effects of antidepressant medication on brain-derived neurotrophic factor concentration and neuroplasticity in depression: a review of preclinical and clinical studies. Avicenna J Med Biotechnol. 2023;15(3):129–138. doi:10.18502/ajmb.v15i3.12921
8. Cramer SC, Sur M, Dobkin BH, et al. Harnessing neuroplasticity for clinical applications. Brain. 2011;134(Pt 6):1591–1609. doi:10.1093/brain/awr039
9. Levy M, Boulle F, Steinbusch HW, et al. Neurotrophic factors and neuroplasticity pathways in the pathophysiology and treatment of depression. Psychopharmacology. 2018;235(8):2195–2220. doi:10.1007/s00213-018-4950-4
10. Castren E, Monteggia LM. Brain-derived neurotrophic factor signaling in depression and antidepressant action. Biol Psychiat. 2021;90(2):128–136. doi:10.1016/j.biopsych.2021.05.008
11. Xue Y, Liang H, Yang R, et al. The role of pro- and mature neurotrophins in the depression. Behav Brain Res. 2021;404:113162. doi:10.1016/j.bbr.2021.113162
12. Cakici N, Sutterland AL, Penninx B, et al. Altered peripheral blood compounds in drug-naive first-episode patients with either schizophrenia or major depressive disorder: a meta-analysis. Brain Behav Immun. 2020;88:547–558. doi:10.1016/j.bbi.2020.04.039
13. Tartt AN, Mariani MB, Hen R, et al. Dysregulation of adult hippocampal neuroplasticity in major depression: pathogenesis and therapeutic implications. Mol Psychiatr. 2022;27(6):2689–2699. doi:10.1038/s41380-022-01520-y
14. Joshi R, Salton S. Neurotrophin crosstalk in the etiology and treatment of neuropsychiatric and neurodegenerative disease. Front Mol Neurosci. 2022;15:932497. doi:10.3389/fnmol.2022.932497
15. Chiou YJ, Huang TL. Serum brain-derived neurotrophic factors in Taiwanese patients with drug-naive first-episode major depressive disorder: effects of antidepressants. Int J Neuropsychoph. 2017;20(3):213–218. doi:10.1093/ijnp/pyw096
16. Hung YY, Lin CJ, Huang TL. Higher serum tropomyosin-related kinase B protein level in major depression. Prog Neuro-Psychoph. 2010;34(4):610–612. doi:10.1016/j.pnpbp.2010.02.021
17. Mikoteit T, Beck J, Eckert A, et al. High baseline BDNF serum levels and early psychopathological improvement are predictive of treatment outcome in major depression. Psychopharmacology. 2014;231(15):2955–2965. doi:10.1007/s00213-014-3475-8
18. Brunoni AR, Machado-Vieira R, Zarate CJ, et al. BDNF plasma levels after antidepressant treatment with sertraline and transcranial direct current stimulation: results from a factorial, randomized, sham-controlled trial. Eur Neuropsychopharm. 2014;24(7):1144–1151. doi:10.1016/j.euroneuro.2014.03.006
19. Meshkat S, Alnefeesi Y, Jawad MY, et al. Brain-derived neurotrophic factor (BDNF) as a biomarker of treatment response in patients with treatment resistant depression (TRD): a systematic review & meta-analysis. Psychiat Res. 2022;317:114857. doi:10.1016/j.psychres.2022.114857
20. Yang J, Tan J, Zheng L, et al. Plasma BDNF and TrkB mRNA in PBMCs are correlated with anti-depressive effects of 12-weeks supervised exercise during protracted methamphetamine abstinence. Front Mol Neurosci. 2020;13:20. doi:10.3389/fnmol.2020.00020
21. Lin PY, Tseng PT. Decreased glial cell line-derived neurotrophic factor levels in patients with depression: a meta-analytic study. J Psychiatr Res. 2015;63:20–27. doi:10.1016/j.jpsychires.2015.02.004
22. Brunoni AR, Machado-Vieira R, Zarate CJ, et al. Assessment of non-BDNF neurotrophins and GDNF levels after depression treatment with sertraline and transcranial direct current stimulation in a factorial, randomized, sham-controlled trial (SELECT-TDCS): an exploratory analysis. Prog Neuro-Psychoph. 2015;56:91–96. doi:10.1016/j.pnpbp.2014.08.009
23. Shi Y, Luan D, Song R, et al. Value of peripheral neurotrophin levels for the diagnosis of depression and response to treatment: a systematic review and meta-analysis. Eur Neuropsychopharm. 2020;41:40–51. doi:10.1016/j.euroneuro.2020.09.633
24. Enache D, Pariante CM, Mondelli V. Markers of central inflammation in major depressive disorder: a systematic review and meta-analysis of studies examining cerebrospinal fluid, positron emission tomography and post-mortem brain tissue. Brain Behav Immun. 2019;81:24–40. doi:10.1016/j.bbi.2019.06.015
25. Zhang FF, Peng W, Sweeney JA, et al. Brain structure alterations in depression: psychoradiological evidence. Cns Neurosci Ther. 2018;24(11):994–1003. doi:10.1111/cns.12835
26. Enneking V, Klug M, Borgers T, et al. Changes in brain function during negative emotion processing in the long-term course of depression. Brit J Psychiat. 2022;221(2):476–484. doi:10.1192/bjp.2021.223
27. Hasel P, Liddelow SA. Astrocytes. Curr Biol. 2021;31(7):R326–R327. doi:10.1016/j.cub.2021.01.056
28. Peng L, Verkhratsky A, Gu L, et al. Targeting astrocytes in major depression. Expert Rev Neurother. 2015;15(11):1299–1306. doi:10.1586/14737175.2015.1095094
29. Rajkowska G, Stockmeier CA. Astrocyte pathology in major depressive disorder: insights from human postmortem brain tissue. Curr Drug Targets. 2013;14(11):1225–1236. doi:10.2174/13894501113149990156
30. Jing D, Hou X, Guo X, et al. Astrocytes in post-stroke depression: roles in inflammation, neurotransmission, and neurotrophin signaling. Cell Mol Neurobiol. 2023;43(7):3301–3313. doi:10.1007/s10571-023-01386-w
31. Kajitani N, Hisaoka-Nakashima K, Morioka N, et al. Antidepressant acts on astrocytes leading to an increase in the expression of neurotrophic/growth factors: differential regulation of FGF-2 by noradrenaline. PLoS One. 2012;7(12):e51197. doi:10.1371/journal.pone.0051197
32. Chen B, Zhang M, Ji M, et al. The association between antidepressant effect of SSRIs and astrocytes: conceptual overview and meta-analysis of the literature. Neurochem Res. 2021;46(10):2731–2745. doi:10.1007/s11064-020-03225-6
33. Kumar A, Nader MA, Deep G. Emergence of extracellular vesicles as ”liquid biopsy” for neurological disorders: boom or bust. Pharmacol Rev. 2024;76(2):199–227. doi:10.1124/pharmrev.122.000788
34. Mansur RB, Delgado-Peraza F, Subramaniapillai M, et al. Exploring brain insulin resistance in adults with bipolar depression using extracellular vesicles of neuronal origin. J Psychiatr Res. 2021;133:82–92. doi:10.1016/j.jpsychires.2020.12.007
35. Li Y, Gui Y, Zhao M, et al. The roles of extracellular vesicles in major depressive disorder. Front Psychiatry. 2023;14:1138110. doi:10.3389/fpsyt.2023.1138110
36. Budnik V, Ruiz-Canada C, Wendler F. Extracellular vesicles round off communication in the nervous system. Nat Rev Neurosci. 2016;17(3):160–172. doi:10.1038/nrn.2015.29
37. Kong L, Zhang D, Huang S, et al. Extracellular vesicles in mental disorders: a state-of-art review. Int J Biol Sci. 2023;19(4):1094–1109. doi:10.7150/ijbs.79666
38. Xie XH, Lai WT, Xu SX, et al. Hyper-inflammation of astrocytes in patients of major depressive disorder: evidence from serum astrocyte-derived extracellular vesicles. Brain Behav Immun. 2023;109:51–62. doi:10.1016/j.bbi.2022.12.014
39. Luarte A, Nardocci G, Chakraborty A, et al. Astrocyte-derived extracellular vesicles in stress-associated mood disorders. Does the immune system get astrocytic? Pharmacol Res. 2023;194:106833. doi:10.1016/j.phrs.2023.106833
40. Sheehan DV, Lecrubier Y, Sheehan KH, et al. The mini-international neuropsychiatric interview (M.I.N.I.): the development and validation of a structured diagnostic psychiatric interview for DSM-IV and ICD-10. J Clin Psychiat. 1998;59(Suppl 20):22–33.
41. Hamilton M. Development of a rating scale for primary depressive illness. Br J Soc Clin Psychol. 1967;6(4):278–296. doi:10.1111/j.2044-8260.1967.tb00530.x
42. Gelle T, Samey RA, Plansont B, et al. BDNF and pro-BDNF in serum and exosomes in major depression: evolution after antidepressant treatment. Prog Neuro-Psychoph. 2021;109:110229. doi:10.1016/j.pnpbp.2020.110229
43. Faul F, Erdfelder E, Buchner A, et al. Statistical power analyses using G*Power 3.1: tests for correlation and regression analyses. Behav Res Methods. 2009;41(4):1149–1160. doi:10.3758/BRM.41.4.1149
44. Nasreddine ZS, Phillips NA, Bedirian V, et al. The Montreal Cognitive Assessment, MoCA: a brief screening tool for mild cognitive impairment. J Am Geriatr Soc. 2005;53(4):695–699. doi:10.1111/j.1532-5415.2005.53221.x
45. Jaeger J. Digit symbol substitution test: the case for sensitivity over specificity in neuropsychological testing. J Clin Psychopharm. 2018;38(5):513–519. doi:10.1097/JCP.0000000000000941
46. Mallinckrodt CH, Watkin JG, Liu C, et al. Duloxetine in the treatment of major depressive disorder: a comparison of efficacy in patients with and without melancholic features. Bmc Psychiatry. 2005;5(1):1. doi:10.1186/1471-244X-5-1
47. Tsai CF, Lee WJ, Wang SJ, et al. Psychometrics of the Montreal Cognitive Assessment (MoCA) and its subscales: validation of the Taiwanese version of the MoCA and an item response theory analysis. Int Psychogeriatr. 2012;24(4):651–658. doi:10.1017/S1041610211002298
48. Xue T, Liu W, Wang L, et al. Extracellular vesicle biomarkers for complement dysfunction in schizophrenia. Brain. 2024;147(3):1075–1086. doi:10.1093/brain/awad341
49. Bang OY, Kim EH, Cho YH, et al. Circulating extracellular vesicles in stroke patients treated with mesenchymal stem cells: a biomarker analysis of a randomized trial. Stroke. 2022;53(7):2276–2286. doi:10.1161/STROKEAHA.121.036545
50. Xu SX, Xie XH, Yao L, et al. Human in vivo evidence of reduced astrocyte activation and neuroinflammation in patients with treatment-resistant depression following electroconvulsive therapy. Psychiat Clin Neuros. 2023;77(12):653–664. doi:10.1111/pcn.13596
51. Glickman ME, Rao SR, Schultz MR. False discovery rate control is a recommended alternative to Bonferroni-type adjustments in health studies. J Clin Epidemiol. 2014;67(8):850–857. doi:10.1016/j.jclinepi.2014.03.012
52. Zhang W, Huang P, Lin J, et al. The role of extracellular vesicles in osteoporosis: a scoping review. Membranes-Basel. 2022;12(3). doi:10.3390/membranes12030324
53. Tian Y, Gong M, Hu Y, et al. Quality and efficiency assessment of six extracellular vesicle isolation methods by nano-flow cytometry. J Extracell Vesicles. 2020;9(1):1697028. doi:10.1080/20013078.2019.1697028
54. Simonsen JB. What are we looking at? Extracellular vesicles, lipoproteins, or both? Circ Res. 2017;121(8):920–922. doi:10.1161/CIRCRESAHA.117.311767
55. Sodar BW, Kittel A, Paloczi K, et al. Low-density lipoprotein mimics blood plasma-derived exosomes and microvesicles during isolation and detection. Sci Rep-UK. 2016;6(1):24316. doi:10.1038/srep24316
56. Allen NJ, Eroglu C. Cell Biology of Astrocyte-Synapse Interactions. Neuron. 2017;96(3):697–708. doi:10.1016/j.neuron.2017.09.056
57. Albini M, Krawczun-Rygmaczewska A, Cesca F. Astrocytes and brain-derived neurotrophic factor (BDNF). Neurosci Res. 2023;197:42–51. doi:10.1016/j.neures.2023.02.001
58. Yang W, Shen Y, Chen Y, et al. Mesencephalic astrocyte-derived neurotrophic factor prevents neuron loss via inhibiting ischemia-induced apoptosis. J Neurol Sci. 2014;344(1–2):129–138. doi:10.1016/j.jns.2014.06.042
59. Chen Z, Tang HB, Kang JJ, et al. Necroptotic astrocytes induced neuronal apoptosis partially through EVs-derived pro-BDNF. Brain Res Bull. 2021;177:73–80. doi:10.1016/j.brainresbull.2021.09.014
60. de Pins B, Cifuentes-Diaz C, Farah AT, et al. Conditional BDNF delivery from astrocytes rescues memory deficits, spine density, and synaptic properties in the 5xFAD mouse model of Alzheimer disease. J Neurosci. 2019;39(13):2441–2458. doi:10.1523/JNEUROSCI.2121-18.2019
61. Upadhya R, Zingg W, Shetty S, et al. Astrocyte-derived extracellular vesicles: neuroreparative properties and role in the pathogenesis of neurodegenerative disorders. J Control Release. 2020;323:225–239. doi:10.1016/j.jconrel.2020.04.017
62. Mousten IV, Sorensen NV, Christensen R, et al. Cerebrospinal fluid biomarkers in patients with unipolar depression compared with healthy control individuals: a systematic review and meta-analysis. JAMA Psychiat. 2022;79(6):571–581. doi:10.1001/jamapsychiatry.2022.0645
63. Chacon-Fernandez P, Sauberli K, Colzani M, et al. Brain-derived neurotrophic factor in megakaryocytes. J Biol Chem. 2016;291(19):9872–9881. doi:10.1074/jbc.M116.720029
64. Karege F, Bondolfi G, Gervasoni N, et al. Low brain-derived neurotrophic factor (BDNF) levels in serum of depressed patients probably results from lowered platelet BDNF release unrelated to platelet reactivity. Biol Psychiat. 2005;57(9):1068–1072. doi:10.1016/j.biopsych.2005.01.008
65. Bocchio-Chiavetto L, Bagnardi V, Zanardini R, et al. Serum and plasma BDNF levels in major depression: a replication study and meta-analyses. World J Biol Psychia. 2010;11(6):763–773. doi:10.3109/15622971003611319
66. Jia L, Qiu Q, Zhang H, et al. Concordance between the assessment of Abeta42, T-tau, and P-T181-tau in peripheral blood neuronal-derived exosomes and cerebrospinal fluid. Alzheimers Dement. 2019;15(8):1071–1080. doi:10.1016/j.jalz.2019.05.002
67. Oraki KM, Papiol S, Delalle I, et al. Extracellular vesicle approach to major psychiatric disorders. Eur Arch Psy Clin N. 2023;273(6):1279–1293. doi:10.1007/s00406-022-01497-3
68. Wu HE, Teixeira AL, Barroso L, et al. Epidermal growth factor and fibroblast growth factor-2 circulating levels in elderly with major depressive disorder. Psychiat Res. 2019;272:141–143. doi:10.1016/j.psychres.2018.12.084
69. Chen B, Dowlatshahi D, Macqueen GM, et al. Increased hippocampal BDNF immunoreactivity in subjects treated with antidepressant medication. Biol Psychiat. 2001;50(4):260–265. doi:10.1016/s0006-3223(01)01083-6
70. Liddelow SA, Barres BA. Reactive astrocytes: production, function, and therapeutic potential. Immunity. 2017;46(6):957–967. doi:10.1016/j.immuni.2017.06.006
71. Wallensten J, Nager A, Asberg M, et al. Leakage of astrocyte-derived extracellular vesicles in stress-induced exhaustion disorder: a cross-sectional study. Sci Rep-UK. 2021;11(1):2009. doi:10.1038/s41598-021-81453-8
72. Mansur RB, Delgado-Peraza F, Subramaniapillai M, et al. Extracellular vesicle biomarkers reveal inhibition of neuroinflammation by infliximab in association with antidepressant response in adults with bipolar depression. Cells-Basel. 2020;9(4). doi:10.3390/cells9040895
73. Saftics A, Abuelreich S, Romano E, et al. Single extracellular vesicle nanoscopy. J Extracell Vesicles. 2023;12(7):e12346. doi:10.1002/jev2.12346
74. Ha D, Yang N, Nadithe V. Exosomes as therapeutic drug carriers and delivery vehicles across biological membranes: current perspectives and future challenges. Acta Pharm Sin B. 2016;6(4):287–296. doi:10.1016/j.apsb.2016.02.001
75. Saeedi S, Nagy C, Ibrahim P, et al. Neuron-derived extracellular vesicles enriched from plasma show altered size and miRNA cargo as a function of antidepressant drug response. Mol Psychiatr. 2021;26(12):7417–7424. doi:10.1038/s41380-021-01255-2
76. Lobb RJ, Becker M, Wen SW, et al. Optimized exosome isolation protocol for cell culture supernatant and human plasma. J Extracell Vesicles. 2015;4:27031. doi:10.3402/jev.v4.27031
77. Coumans F, Brisson AR, Buzas EI, et al. Methodological Guidelines to Study Extracellular Vesicles. Circ Res. 2017;120(10):1632–1648. doi:10.1161/CIRCRESAHA.117.309417
78. Paoli A, Cenci L, Pompei P, et al. Effects of two months of very low carbohydrate ketogenic diet on body composition, muscle strength, muscle area, and blood parameters in competitive natural body builders. Nutrients. 2021;13(2):374. doi:10.3390/nu13020374
79. Szuhany KL, Otto MW. Assessing BDNF as a mediator of the effects of exercise on depression. J Psychiatr Res. 2020;123:114–118. doi:10.1016/j.jpsychires.2020.02.003
© 2024 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 3.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.
Recommended articles
Serum CC Chemokines as Potential Biomarkers for the Diagnosis of Major Depressive Disorder
Gao W, Xu Y, Liang J, Sun Y, Zhang Y, Shan F, Ge J, Xia Q
Psychology Research and Behavior Management 2022, 15:2971-2978
Published Date: 11 October 2022

Individuals with Bipolar Disorder Have a Higher Level of Homocysteine Than Major Depressive Disorder: A Retrospective Chart Review and Observational Study
Zhao M, Liu T, Qi S, Li W, Liu X, Li X, Xun G
Neuropsychiatric Disease and Treatment 2022, 18:2671-2682
Published Date: 15 November 2022