You are on page 1of 12

Brain, Behavior, and Immunity 109 (2023) 51–62

Contents lists available at ScienceDirect

Brain Behavior and Immunity


journal homepage: www.elsevier.com/locate/ybrbi

Hyper-inflammation of astrocytes in patients of major depressive disorder:


Evidence from serum astrocyte-derived extracellular vesicles
Xin-hui Xie a, b, Wen-tao Lai a, Shu-xian Xu b, Marta Di Forti c, Jing-ya Zhang d, Mian-mian Chen b,
Li-hua Yao b, Peilin Wang a, Ke-ke Hao b, Han Rong a, e, *
a
Department of Psychiatry, Shenzhen Kangning Hospital, and Shenzhen Mental Health Center, Shenzhen, Guangdong, China
b
Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
c
Department of Social Genetics and Developmental Psychiatry, Institute of Psychiatry Psychology and Neuroscience, King’s College London, London, United Kingdom
d
Department of Clinical Psychology, Second People’s Hospital of Huizhou, Huizhou, Guangdong, China
e
Affiliated Shenzhen Clinical College of Psychiatry, Jining Medical University, Jining, Shandong, China

A R T I C L E I N F O A B S T R A C T

Keywords: Astrocyte-derived extracellular vesicles (ADEs) allow the in vivo probing of the inflammatory status of astrocytes
Major depressive disorder practical. Serum sample and ADEs were used to test the inflammatory hypothesis in 70 patients with major
Astrocyte depressive disorder (MDD) and 70 matched healthy controls (HCs). In serum, tumor necrosis factor α (TNF-α)
Extracellular vesicle
and interleukin (IL)-17A were significantly increased, where as IL-12p70 was significantly reduced in the MDD
Astrocyte-derived extracellular vesicles
Inflammation
patients compared with HCs. In ADEs, all inflammatory markers (Interferon-γ, IL-12p70, IL-1β, IL-2, IL-4, IL-6,
Central nervous system TNF-α, and IL-17A) except IL-10 were significantly increased in the MDD patients, the Hedge’s g values of
elevated inflammatory markers varied from 0.48 to 1.07. However, there were no differences of all inflammatory
markers whether in serum or ADEs between MDD-drug free and medicated subgroups. The association of in­
flammatory biomarkers between ADEs and serum did not reach statistically significance after multi-comparison
correction neither in the HCs nor MDD patients. The spearman coefficients between inflammatory factors and
clinical characteristics in the MDD patients, such as onset age, disease course, current episode duration, and
severity of depression, were nonsignificant after multi-comparison correction. In the receiver operating char­
acteristic curves analysis, the corrected partial area under the curve (pAUC) of each inflammatory markers in
ADEs ranged from 0.522 to 0.696, and the combination of these inflammatory factors achieved a high pAUC
(>0.9). Our findings support the inflammatory glial hypothesis of depression, and suggests that in human ADEs
could be a useful tool to probe the in vivo astrocyte status.

1. Introduction inflammatory factors were found in depressed patients compared with


healthy controls (HCs) (Kohler et al., 2017; Liu et al., 2012; Osimo et al.,
Major depressive disorder (MDD) which carrying a high prevalence, 2020), while these factors had a significant association with the subse­
a high recurrence rate, and also a suicidal tendency (Belmaker and quent development of depressive symptoms (Valkanova et al., 2013) and
Agam, 2008) is among the leading causes of burden and disability the poor outcome of first-line antidepressant treatments (Arteaga-Hen­
worldwide (Collins et al., 2011). Multiple factors are believed to play riquez et al., 2019). Likewise, depression was highly prevalent in the
important roles in the pathologies of MDD, and the new major candi­ individuals with inflammatory disorders, such as rheumatoid arthritis
dates are the inflammation hypothesis (Najjar et al., 2013; Troubat et al., (Matcham et al., 2013), inflammatory bowel disease (Barberio et al.,
2021; Woelfer et al., 2019) in which the inflamed glia, as well as 2021), and systemic lupus erythematosus (Moustafa et al., 2020). Lon­
microglia and astrocyte played a critical role (Leng et al., 2018). gitudinal cohorts also revealed that the higher levels of serum inter­
Inflammation has been linked to depression for a long time. Multiple leukin 6 (IL-6) and C-reactive protein (CRP) in childhood are associated
peripheral cytokine concentrations are connected to brain function, with an increased risk of developing depression in young adulthood
wellbeing and cognition (Bollen et al., 2017). Elevated peripheral (Khandaker et al., 2014). The prior presence of inflammation implies its

* Corresponding author at: Department of Psychiatry, Shenzhen Kangning Hospital, and Shenzhen Mental Health Center, Shenzhen, Guangdong, China.
E-mail address: ronghansz@126.com (H. Rong).

https://doi.org/10.1016/j.bbi.2022.12.014
Received 6 September 2022; Received in revised form 3 December 2022; Accepted 16 December 2022
Available online 29 December 2022
0889-1591/© 2022 Elsevier Inc. All rights reserved.
X.-h. Xie et al. Brain Behavior and Immunity 109 (2023) 51–62

probability of biological cause of depression. However, inflammatory 2021) and lipid composition (Losito et al., 2013; Vidal et al., 1989) with
markers were considered to be the sensitive rather than specific in­ their source cells. Most importantly, EVs could cross the blood–brain
dicators of various somatic illnesses, unhealthy status such as obesity barrier (BBB) from both directions (Saint-Pol et al., 2020; Terstappen
(Milaneschi et al., 2019) and of exposure to stressful life events (Her­ et al., 2021), and further, CNS-derived EVs could be detected and
berth et al., 2008), and could result in a significant heterogeneity in collected in the peripheral blood and reveal the status of their source
related studies. To eliminate potential biases and test the inflammatory cells without brain biopsy or lumbar puncture (Mustapic et al., 2017; Shi
hypothesis, Cassano et al. (2017) rigorously recruited MDD patients et al., 2019). For some target molecules, the correlation coefficients
without significant medical comorbidity, and finally found no signifi­ between the concentration level in the CNS-derived EV and CSF excee­
cant alternations of 22 plasma cytokines in MDD patients compared with ded 0.9 (Jia et al., 2019). Specifically, for astrocyte-derived EVs (ADEs),
well-matched HCs. On the opposite, some studies and meta-analyses Delgado-Peraza et al. (2021) found that ADEs C1q had strong correla­
have provided evidence to the contrary, showing that patients with tions with C1q in the cortex brain homogenate and the correlation co­
depression had higher levels of inflammatory markers than controls efficient reached higher than 0.85, this suggested that ADEs could be
after accounting for various confounders (Lindqvist et al., 2017; Osimo used as a “liquid biopsy” for CNS. Therefore, to contribute to clarify the
et al., 2020). Pitharouli et al. further showed an association between dual role of astrocytes in the pathology of depression (Wang et al., 2017)
peripheral inflammation and depression after adjusting for confounders and CNS inflammatory process (Linnerbauer et al., 2020), we use ADEs
in a very large sample (Pitharouli et al., 2021). Nevertheless, depression as the “window of brain” to test the in vivo inflammatory status of as­
is a brain disease, and the relationship of inflammation between the trocytes in CNS.
central nervous system (CNS) and the periphery is currently under In the present study, the primary aim was to investigate if in patients
debate. The results of some studies support the existence of a significant with MDD compared with HCs, there are alterations of inflammatory
relationship between the CNS and the periphery (Felger et al., 2020; biomarkers in ADEs and serum. Secondly, we aimed to carry out
Lindqvist et al., 2013; Miller et al., 2017), while others suggested that exploratory analyses to investigate: 1) whether there are differences in
the peripheral inflammatory status may not accurately represent the these inflammatory biomarkers between the drug-free (MDD-DF) and
inflammatory status in the CNS (Enache et al., 2019; Hannestad et al., medicated (MDD-M) subgroup of MDD patients; 2) the relations of these
2013; Holmes et al., 2018; Lindqvist et al., 2009; Richards et al., 2018; inflammatory biomarkers between ADEs and serum; 3) the relations
Sasayama et al., 2013). The many studies that directly focused on between these inflammatory biomarkers and clinical features and finally
measuring CNS inflammation markers in depressed patients, still found to 4) the potential of inflammatory markers in ADEs as a biomarker for
heterogeneous results. A comprehensive meta-analysis (Enache et al., MDD.
2019) summarized relevant markers of central inflammation in MDD
patients and suggested a significant higher pro-inflammatory levels of 2. Material and methods
IL-6, IL-8, IL-1β, and tumor necrosis factor α (TNF-α) in cerebrospinal
fluid (CSF), and reduction of astrocytes that may result from the CNS This observational study was conducted at the Shenzhen Kangning
inflammation. These findings suggest that we should concentrate on the Hospital (Mental Health Center of Shenzhen, Shenzhen, Guangdong, PR
glia most closely associated with neuroinflammation, namely microglia China) from June 2018 to July 2019 in accordance with the Declaration
and astrocytes, the latter of which is the target of this study. of Helsinki (revised edition, 2008). This study was approved by the
Astrocytes are considered important glial cells implicated in the Institutional Review Board of Shenzhen Kangning Hospital. Written
pathology of MDD (Peng et al., 2015; Rajkowska and Stockmeier, 2013). informed consents were obtained from all participants or their legal
Impaired astrocytes were found in several post-mortem studies on sub­ guardians. The present report followed the STROBE statement (von Elm
jects with depression (Cotter et al., 2002; Medina et al., 2016; Nagy et al., 2007).
et al., 2015; Ongur et al., 1998; Rajkowska et al., 1999). As the second
abundant glial cells, astrocytes not only maintain the synaptic function, 2.1. Sample size calculation
nerve fiber integrity, neurogenesis, neurotrophic and energy meta­
bolism (Khakh and Deneen, 2019), but also play a vital role in CNS A most widely measured peripheral inflammatory marker—IL-6 was
inflammation regulation (Linnerbauer et al., 2020). Astrocytes produce chosen as the reference for our sample size calculation according to the
both pro- and anti-inflammatory cytokines as well as other chemokines lack of studies on inflammatory markers in ADEs in MDD patients. In the
to influence the CNS inflammatory status (Dong and Benveniste, 2001; recent meta-analyses, effect size of IL-6 between MDD and control group
Falsig et al., 2006). Inflamed astrocytes could lead to depression-like were 0.62 (Cakici et al., 2020) and 0.76 (Goldsmith et al., 2016).
behaviors in rodent models (Leng et al., 2018), and a postmortem Therefore, effect size of 0.65 with a power (1-β) of 0.95, α of 0.05 (two
investigation (Torres-Platas et al., 2011) found that, compared with sided) were set and then 126 (63 per group) subjects were needed in our
sudden-death controls, astrocytes in anterior cingulate cortex had study. Taking the other unexpected factors into account, the final sample
significantly larger cell bodies, longer and ramified processes in size was set at 140 (70 per group). Sample size calculation was per­
depressed suicides, these results may reflect a state of chronic inflam­ formed by using G*Power software (Faul et al., 2009) (ver. 3.1.9.7).
mation in limbic region and support the neuroinflammatory hypothesis
of depression. 2.2. Participants
Despite emerging evidence support the hypothesis of inflammatory
astrocytes in depression, little is known about the in vivo astrocytes Inclusion criteria for the MDD group were: 1) aged between 18 and
status in individuals suffering from MDD, because probing the astrocytes 60 years; 2) meeting ICD-10(Organization, 1992) criteria for the diag­
status in CNS on human individuals in vivo is challenging due to the nosis of MDD using the Mini International Neuropsychiatric Interview
limitations of methodology. Fortunately, the development of technology (Sheehan et al., 1998); 3) Hamilton Depression Rating Scale (HAMD-17)
in the field of the CNS-derived extracellular vesicles (EVs) extraction (Hamilton, 1960) > 17; 4) ability to provide informed consent. Using
have recently made it possible to measure the in vivo status of the CNS several mode of online and offline advertising HCs were recruited to
without performing a brain biopsy (Goetzl et al., 2021; Shi et al., 2019). match for sex- and age the MDD group. HCs were required to be in good
Same as other EVs, CNS-derived EVs, which were secreted by almost all physical health, and with no history of personal, or first-degree family,
types of cells and contained the surface biomarkers of their source cells, psychiatric disorder.
including neurons, oligodendrocytes, microglia and astrocytes, could Considering the inflammatory markers are sensitive to several so­
help to identify their parent cells (Mustapic et al., 2017). EVs showed matic conditions which also may have interaction with depression sta­
close relationship of protein (Casella et al., 2020; Delgado-Peraza et al., tus, a rigorous exclusion criteria for both groups included: 1) with

52
X.-h. Xie et al. Brain Behavior and Immunity 109 (2023) 51–62

current or chronic inflammatory disorders; 2) antibiotic use within the microscope (FEI Company, Hillsboro, USA) was used for imaging.
prior one month; 3) history of neurological disorders such as epilepsy,
multiple sclerosis, stroke, and neurodegenerative diseases; 4) history of 2.4.2. Nanoparticle tracking analysis
metabolic syndromes, such as obesity (BMI > 30), diabetes, hyperlip­ The diameter (nm) and concentration (particles/ml) of ADEs was
idemia; 5) cancer; 6) cardiovascular illness; 7) presence of severe stress determined using the Nanosight NS500 system (Malvern Panalytical,
events, such as severe injuries or surgeries within the prior one month; Malvern, UK) with a G532 nm laser module and NTA 3.4.003 nano­
8) current pregnancy; 9) other unstable, serious comorbidities; 10) particle tracking software (Malvern Instruments, Malvern, UK).
psychoactive substance abuse in the last year.
2.4.3. Western blotting
2.3. Collection of serum and ADEs Western blot was performed to detect three exosomal marker (cluster
of differentiation (CD) 63, CD81 and tumor susceptibility (TSG) 101),
Two milliliters peripheral fasting blood samples were collected after and astrocyte marker glial fibrillary acidic protein (GFAP) using corre­
a 12-h fast from each subject between 06:00–09:00 in the morning and sponding anti-human antibodies according to the manufacturer’s in­
stored in a serum separator tube and was centrifuged at 3000×g for 10 structions (CD63, Abcam, Cambridge, UK, Catalog number: ab134045;
min to obtain the serum. The serum samples were divided into 500 μl per CD81, Abcam, Catalog number: ab109201; TSG101, Abcam, Catalog
tube and stored at − 80 ℃. On the test day, all samples were tested number: ab125011; GFAP, Abcam, Catalog number: ab68428).
simultaneously. For each participant, a tube of 500 μl of serum was
thawed, of which 250 μl of serum was used to test for peripheral 2.5. Protein measurements
inflammation markers. The remaining 250 μl of serum was used to
extract ADEs. ADEs were separated according to a two-step widely For protein measurements, 365 ul of mammalian protein extraction
tested and well certified protocol (Chen et al., 2019; Fiandaca et al., reagent (M-PER, Thermo Fisher Scientific, Catalog number: 78503) was
2015; Goetzl et al., 2016; Goetzl et al., 2018; Goetzl et al., 2020; Jia added to an ADE sample. The levels of interferon-γ (IFN-γ), IL-12p70, IL-
et al., 2019; Mustapic et al., 2017; Winston et al., 2019a; Winston et al., 1β, IL-2, IL-4, IL-6, TNF-α, IL-10, and IL-17A in both serum and ADE
2019b). In brief, 250 ul of serum was gently mixed with 250 ul calciu­ were measured with the High Sensitivity 9-Plex Human ProcartaPlex™
mand magnesium-free Dulbecco’s phosphate-buffered saline (DPBS, Panel (Thermo Fisher Scientific, Catalog number: EPXS090-12199-901).
Thermo Fisher Scientific, Catalog number: 14190136) with protease and The levels of CD81 protein in ADEs were measured with the enzyme-
phosphatase inhibitor cocktails (Roche Life Science, Catalog number: linked immunosorbent assay (ELISA, American Research Products Inc.,
4,693116001 and 4906837001) at 4 ℃, and followed the centrifugation Catalog number: CSB-EL004960HU). The amount of CD81 protein
at 3000×g for 30 min to remove possible cells or cell debris. After that, concentrations in ADEs were used to normalize the EV content as
the supernatants were mixed with 126 ul of ExoQuick exosome pre­ following, the mean value of CD81 levels in each group was set to 1.00,
cipitation solution (EXOQ; System Biosciences, CA, Catalog number: and the relative values for each sample were used to normalize their
EXOQ20A) and incubated at 4 ◦ C for 60 min. After centrifugation at recovery (Fiandaca et al., 2015; Goetzl et al., 2016; Goetzl et al., 2018;
1500×g for 30 min, the pellets (total EVs) were then resuspended in 350 Goetzl et al., 2020; Jia et al., 2019; Winston et al., 2019a). All samples
ul of DPBS and placed in a rotating mixer for 2 h at 4 ◦ C. Each sample were tested in duplicate at the same time.
was incubated for 1 h at room temperature with 1.5 μg of mouse anti-
human glutamine aspartate transporter (ACSA-1) biotinylated anti­ 2.6. Statistical analyses
body (Miltenyi Biotec, Auburn, CA, Catalog number: 130-118-984) after
being combined with 50 ul 3 % bovine serum albumin (Sangon Biotech Descriptive analyses for categorical data, such as sex, were analyzed
(Shanghai) Co., ltd., Catalog number: A600332). After that, 10 μl of using the Fisher exact test. Numerical data were first tested for equal
streptavidinagarose resin (Thermo Fisher Scientific, Catalog number: variance and normality (Shapiro-Wilk test). Welch’s two-sample t-tests
53117) and 40 μl of 3 % bovine serum albumin was added and then were used for normally distributed data, and Mann-Whitney U tests were
incubated for 30 min at room temperature with continuous mixing, used for non-normality numerical data. To test for differences of in­
followed by centrifugation at 800×g for 10 min at 4 ℃ and removal the flammatory markers between two groups, general linear models (GLM)
supernatant. Each sample was then resuspended using 100 μl cold 0.05 were used. Inflammatory markers were set as dependent variables and
M glycine-HCl (pH = 3.0) and was centrifugated at 4000×g for 10 min. the group was set as the independent variable; sex, age, and BMI were
The supernatant was recovered and then mixed with 25 ul DPBS con­ added as covariates. The model-fitted estimated marginal means
taining 3 % bovine serum albumin, and adjust the pH to 7.0 by adding (EMMs) and their associated 95 % CI were used to represent the dif­
10 ul 1 M Tris-HCl (pH = 8.0). The samples were then incubated at 37 ℃ ferences between two groups. For multiple comparisons correction, after
for 10 min and then the ADEs were isolated for further confirmation and considering the advantages and disadvantages of different methods
protein measurements. (Perneger, 1998), the false discovery rate (FDR) correction (Benjamini
and Yekutieli method) (Benjamini and Yekutieli, 2001) was conducted
2.4. Confirmation of ADE to accommodate false positive results. Considering that inflammation
markers in peripheral and CNS may affect each other, sensitivity anal­
An ADE sample of MDD-M patient was used for the ADE confirma­ ysis was conducted as follow: when comparing the group differences of
tion, and the supernatants of total exosomes and ADE were used as the inflammatory markers in ADEs, the values of the same inflammatory
negative controls. The ADE were confirmed by three different well- marker in serum was added as a covariate, and vice versa. Hedges’ g was
established methods, namely, the transmission election microscopy, also used to present the original effect size between two groups. In the
nanoparticle tracking analysis, and western blotting. exploratory analyses, similar methods were used, namely, GLMs were
used to test the differences of inflammatory markers between two sub­
2.4.1. Transmission electron microscopy groups, the MDD-DF and MDD-M subgroups; spearman coefficients were
Transmission electron microscopy scanning was performed to used for correlation analyses of the inflammatory markers’ concentra­
observe the shape of ADEs. In brief, after immunoprecipitation, 10 ul tion levels between the serum and the ADEs, and clinical features.
ADE sample was added dropwise to 200-mesh grids and incubated at Receiver operating characteristic curves (ROCs) and corrected partial
room temperature for 5 min, then were negatively stained with uranium area under the curve (pAUC) were used to further explore the potential
dioxide acetate solution for 1 min, and the remaining liquid was of inflammatory markers in ADEs as biomarkers of MDD; and FDR
removed by filter paper. A FEI Tecnai G2 Spirit transmission electron correction was also conducted to control false positive results. A two-

53
X.-h. Xie et al. Brain Behavior and Immunity 109 (2023) 51–62

tailed p-values <0.05 was considered significant. All statistical analyses significantly increased, where as IL-12p70 was significantly reduced in
were performed using R version 4.1.0 (R Project for Statistical the MDD group compared with HCs. The differences of other six in­
Computing) within RStudio version 1.4.1106 (RStudio). flammatory markers between MDD and HCs were non-significant,
detailed results are demonstrated in Table 2 and Fig. 2.
3. Results For inflammatory markers in ADEs, all inflammatory markers except
IL-10 were significantly increased in the MDD group compared with
3.1. Participant characteristics HCs, the Hedge’s g value of these eight inflammatory markers varied
from 0.48 to 1.07, detailed results are demonstrated in Table 2 and
A total of 70 patients with MDD and 70 HCs were recruited. Table 1 Fig. 3.
reports the characteristics of the participants. As to be expected there In the sensitivity analyses, the results were generally similar to the
were no differences in sex and age between the two groups and the same main analysis, except that the IL-12p70 in ADEs became statistically
was for BMI. Similarly, there were no differences in sex ratio, age, BMI, non-significant (PFDR = 0.073) after the IL-12p70 level in serum was
age of onset, total disease course, current episode duration, and HAMD- added as a covariate, and the IL-17A in serum became statistically non-
17 score between the MDD-DF and the MDD-M subgroups (see sTable 1). significant (PFDR = 1.000) after the IL-17A level in ADEs was added as a
covariate. Detailed results are demonstrated in sTable 2.
3.2. Confirmation of ADEs
3.4. Exploratory objective 1: Subgroup differences
ADEs were confirmed by transmission electron microscopy, nano­
particle tracking analysis and western blotting (Fig. 1). The transmission In total, the subgroup differences of all inflammatory markers
electron microscopy image of an MDD-DF patient’s ADEs clearly shows whether in serum or ADEs between MDD-DF and MDD-M subgroups
the exosomes-like shape. The results of nanoparticle tracking analysis were non-significant (all PFDR values > 0.05). Detailed results are
show the diameter distribution of the EVs, which also corroborate with demonstrated in sTable 3.
the prior knowledge of exosomes-like small EVs. Western blotting results
showed that three exosomes marker CD63, CD81, TSG101, and an 3.5. Exploratory objective 2: Relations of inflammatory biomarkers
astrocyte marker GFAP was expressed in the ADE samples (The original between ADEs and serum
whole piece pictures of western blotting are shown in sFigure 1). Based
on these results, we confirmed that ADEs were successfully collected. No spearman coefficients of inflammatory biomarkers between ADEs
In ADEs, the distribution of CD81 was not norm. As shown in Fig. 1e, and serum of neither in HCs nor MDD group reached statistically sig­
the difference of CD81 was non-significant between the MDD (median nificance after the FDR correction (all PFDR values > 0.05). Detailed
(inter quartile range (IQR)) = 1013 (1698) pg/ml serum) group and HC results are demonstrated in sTable 4.
(median (IQR) = 993 (675) pg/ml serum) group (Mann-Whitney U test,
W = 2108, p = 0.154). In subgroups, the difference of CD81 was non- 3.6. Exploratory objective 3: Relations between inflammatory biomarkers
significant between the MDD-DF (median (IQR) = 1010 (1956) pg/ml and clinical features.
serum) subgroup and MDD-M (median (IQR) = 1016 (1684) pg/ml
serum) subgroup (Mann-Whitney U test, W = 540, p = 0.783, In analyses exploring the correlation between inflammatory factors
sFigure 3a). and clinical characteristics, such as onset age, disease course, current
episode duration, and HAMD-17 score, no correlation coefficients
3.3. Main objective: Group differences of inflammatory markers in serum remained significant after FDR correction, although some original P
and ADEs values <0.05. Detailed results are demonstrated in sTable 5.

For inflammatory markers in serum, TNF-α and IL-17A were 3.7. Exploratory objective 4: Potentiality of inflammatory markers in
ADEs as biomarkers for MDD.
Table 1
Descriptive data of included participants.
The corrected pAUCs of each inflammatory markers ranged from
0.522 to 0.696, while IL-6 being the most differentiated (pAUC = 0.696,
Parameters MDD HC pa
95 % CI = (0.605, 0.792)), further, the combination of these inflam­
Subject, n 70 70 – matory factors had a very high discriminatory effect on MDD group and
Sex, Female/Male 42/28 42/28 1.000 HCs (pAUC = 0.919, 95 % CI = (0.860, 0.974)). Detailed results are
Age, years, median (IQR) 41.0 (18.5) 37.0 (15.8) 0.398
BMI, kg/m2, median (IQR) 21.6 (3.65) 22.2 (3.63) 0.079
demonstrated in Fig. 4 and sTable 6.
Clinical characteristics
Medication, yes/no 45/25 NA – 3.8. Data availability statement
SSRIsb, yes/no 28/17 NA –
SNRIsb, yes/no 19/26 NA –
The data that support the findings of this study are available from the
Other antidepressantsb, yes/no 7/38 NA –
Mood stabilizerb, yes/no 2/43 NA – corresponding author, upon reasonable request.
Atypical antipsychoticsb, yes/no 10/35 NA –
Equivalent fluoxetine doseb, mg, mean (SD) 33.2 (16.6) NA – 4. Discussion
Age of onset, years, mean (SD) 32.5 (12.4) NA –
Total disease course, years, mean (SD) 6.7 (7.8) NA –
Current episode duration, months, mean (SD) 2.4 (2.3) NA –
This is the first study to show significantly higher inflammatory
HAMD-17, mean (SD) 25.7 (5.5) NA – markers in ADEs in MDD patients compared with HCs. Our results
support the neuroinflammatory hypothesis of depression via a unique
Note: a: For categorical data, Fisher exact test was used, for numerical data; the
and direct perspective. Further, the inflammatory markers in ADEs
distribution of age and BMI are not normal, therefore, the Mann-Whitney U tests
were used. b: Only medicated patients were calculated (n = 45).
demonstrated to be potential new biological markers of MDD.
Abbreviations: MDD = major depressive disorder; HC = health control; SD =
standard deviation; BMI = body mass index; IQR = inter quartile range; SSRI = 4.1. Inflammatory status in serum
selective serotonin reuptake inhibitor; SNRI = serotonin and norepinephrine
reuptake inhibitor; HAMD-17 = Hamilton’s Depression Scale (17-item version). We found only two significant higher levels of serum pro-

54
X.-h. Xie et al. Brain Behavior and Immunity 109 (2023) 51–62

Fig. 1. Confirmation of astrocyte-derived extracellular vesicles (ADEs). a) The results of nanoparticle tracking analysis show the diameter distribution of the EVs,
which also corroborate with the prior knowledge of exosomes-like small EVs (diameter: 118.4 ± 45.5 nm, Concentration: 2.01 ± 0.12 109 particles/ml); b–c) The
transmission electron microscopy image of ADEs isolated from a drug-free patient with major depressive disorder (MDD) patient, the images clearly shows the
exosomes-like shape (red arrows); d) Western blotting results of ADE sample: three exosomes marker cluster of differentiation (CD) 63, CD81, tumor susceptibility
(TSG) 101, and an astrocyte marker glial fibrillary acidic protein (GFAP) was identified; e) Boxplot of CD81 between the MDD (median (inter quartile range (IQR)) =
1013 (1698) pg/ml serum) group and HC (median (IQR) = 993 (675) pg/ml serum) group (Mann-Whitney U test, p = 0.154). (For interpretation of the references to
colour in this figure legend, the reader is referred to the web version of this article.)

Table 2
Comparisons of EMMs of inflammatory markers in serum and ADEs based on general linear model.
HC MDD Difference P PFDR Hedge’s g (95 % CI)
EMM (95 % CI) EMM (95 % CI) MDD - HC (95 % CI)

In Serum
IFN-γ 1.36 (1.21, 1.52) 1.53 (1.38, 1.69) 0.17 (− 0.04, 0.39) 0.118 0.601 0.30 (− 0.03, 0.64)
IL-12p70 0.65 (0.60, 0.71) 0.49 (0.44, 0.54) − 0.16 (− 0.23, − 0.09) <0.001 <0.001 − 0.73 (− 1.09, − 0.40)
IL-1β 0.12 (0.09, 0.14) 0.15 (0.12, 0.18) 0.03 (− 0.01, 0.07) 0.118 0.601 0.34 (0.01, 0.68)
IL-2 1.46 (1.08, 1.84) 1.72 (1.35, 2.09) 0.26 (− 0.27, 0.79) 0.333 1.000 0.14 (− 0.19, 0.48)
IL-4 1.55 (1.25, 1.86) 1.69 (1.39, 1.99) 0.14 (− 0.29, 0.57) 0.512 1.000 0.13 (− 0.21, 0.46)
IL-6 1.35 (0.99, 1.70) 1.13 (0.78, 1.48) − 0.22 (− 0.72, 0.28) 0.389 1.000 − 0.22 (− 0.56, 0.11)
TNF-α 0.84 (0.54, 1.13) 1.47 (1.18, 1.76) 0.63 (0.22, 1.05) 0.003 0.038 0.52 (0.18, 0.86)
IL-10 0.21 (0.18, 0.25) 0.24 (0.20, 0.27) 0.03 (− 0.02, 0.07) 0.268 1.000 0.16 (− 0.17, 0.50)
IL-17A 0.06 (0.02, 0.11) 0.15 (0.11, 0.19) 0.08 (0.02, 0.14) 0.005 0.045 0.45 (0.12, 0.80)
In ADEs
IFN-γ 9.17 (7.31, 11.0) 16.73 (14.90,18.60) 7.56 (4.95, 10.17) <0.001 <0.001 0.99 (0.65, 1.36)
IL-12p70 5.14 (4.17, 6.11) 7.14 (6.18, 8.10) 2.00 (0.63, 3.36) 0.004 0.001 0.48 (0.14, 0.82)
IL-1β 3.73 (2.88, 4.59) 7.16 (6.32, 8.00) 3.42 (2.23, 4.62) <0.001 <0.001 0.93 (0.60, 1.30)
IL-2 20.7 (15.1, 26.4) 44.8 (39.2, 50.4) 24.10 (16.12, 32.08) <0.001 <0.001 0.99 (0.65, 1.36)
IL-4 14.3 (11.3, 17.2) 22.6 (19.7, 25.5) 8.36 (4.18, 12.53) <0.001 <0.001 0.70 (0.36, 1.05)
IL-6 13.2 (10.1, 16.4) 27.5 (24.4, 30.6) 14.29 (9.89, 18.69) <0.001 <0.001 1.07 (0.73, 1.44)
TNF-α 15.70 (12.40, 19.10) 26.90 (23.60, 30.20) 11.19 (6.46, 15.92) <0.001 <0.001 0.77 (0.44, 1.13)
IL-10 1.11 (0.81, 1.40) 1.57 (1.29, 1.86) 0.47 (0.06, 0.88) 0.026 0.073 0.37 (0.04, 0.71)
IL-17A 0.89 (0.58, 1.21) 1.81 (1.50, 2.13) 0.92 (0.48, 1.37) <0.001 <0.001 0.63 (0.30, 0.98)

Note: Age, sex, and BMI are added as covariates, and the concentrations of inflammatory markers were normalized to CD81 concentration level.
Abbreviations: MDD = major depressive disorder; HC = health control; ES = effect size; EMM = estimated marginal mean; CI = confidence interval; ADEs = astrocyte-
derived extracellular vesicles; IFN = interferon; IL = interleukin; TNF = tumour necrosis factor; BMI = body mass index; FDR = false discovery rate.

55
X.-h. Xie et al. Brain Behavior and Immunity 109 (2023) 51–62

Fig. 2. Boxplots of inflammatory markers in serum between major depressive disorder (MDD) and healthy control (HC) group. Note: *Pfalse discovery rate (FDR) < 0.05;
**PFDR < 0.01; ***PFDR < 0.001.

56
X.-h. Xie et al. Brain Behavior and Immunity 109 (2023) 51–62

Fig. 3. Boxplots of inflammatory markers in astrocyte-derived extracellular vesicles (ADEs) between major depressive disorder (MDD) and healthy control (HC)
group. Note: *Pfalse discovery rate (FDR) < 0.05; **PFDR < 0.01; ***PFDR < 0.001.

57
X.-h. Xie et al. Brain Behavior and Immunity 109 (2023) 51–62

Fig. 4. Receiver operating characteristic curves (ROCs) of inflammatory markers in astrocyte-derived extracellular vesicles (ADEs). The corrected partial area under
the curves (pAUCs) of each inflammatory marker ranged from 0.522 to 0.696 (interferon (IFN)-γ: 0.624; interleukin (IL)-12p70: 0.558; IL-1β: 0.607; IL-2: 0.642; IL-4:
0.603; IL-6: 0.696; tumor necrosis factor (TNF)-α: 0.593; IL-10: 0.548; IL-17A: 0.522; Combination of above inflammatory markers: 0.919.).

inflammatory cytokines (TNF-α and IL-17A) in MDD group, and inter­ patients, we cannot yet make any trustworthy conclusions based on the
estingly, one serum pro-inflammatory cytokine higher in HC group. No sparse and inconsistent clinical data. We hope to see more pertinent
other significant alternations of cytokines were found between MDD and studies in the future.
HC groups. Significant heterogeneity of inflammatory markers in pe­ However, it should be noted that, the rigorous exclusion of somatic
riphery is a lasting issue in MDD studies. Confounding by somatic co­ co-morbidities may excluded a subtype of depression, in which somatic
morbidity is one potential reason. The prevalence of depression in co-morbidities may be a causal factor of inflammation or a key mediator
medically ill patients is significantly higher than in the general popu­ of depression. A more ideal approach would be to perform a subgroup
lation (Cassano and Fava, 2002), especially in patients with analysis of MDD individuals who do not have somatic co-morbidities in a
inflammation-related diseases (Barberio et al., 2021; Matcham et al., much larger MDD sample, but this would result in a markedly larger
2013; Moustafa et al., 2020), and with some unhealthy lifestyles (Chang sample size. We expect that the findings of this investigation will provide
et al., 2019; Kohno et al., 2019; Milaneschi et al., 2019). As a result, a priori data for the next study with larger sample size. Hitherto, absence
some associations between elevated peripheral inflammatory markers or inconsistent peripheral inflammatory status do not falsify the in­
and depression in prior studies that did not rigorously control for these flammatory hypothesis of MDD which is partly due to the poor corre­
potential confounders may have been coincidental (Cassano et al., lation of inflammatory markers between peripheral serum and the CNS
2017), or, these elevated peripheral inflammatory markers and in participants with depression(Enache et al., 2019).
depressive symptoms could be a common result of some pathology
source of depression. To control these confounding factors, Cassano
4.2. Neuro-inflammation, astrocytes, and depression
et al. rigorously recruited a group of depressed patients with no sub­
stantial medical comorbidities, obesity and problem substance use, as
Astrocyte is a protagonist in neuro-inflammation. About 20–40 %
well as a group of one-by-one matched HCs, and observed no significant
glial cells in CNS are astrocytes (Herculano-Houzel, 2014; Rowitch and
differences in 22 cytokine levels between MDD patients and HCs (Cas­
Kriegstein, 2010). Cytokines are not only produced in the periphery by
sano et al., 2017). Their findings suggested that some shared con­
immune cells, but recent research has shown that glia, such as astro­
founding factors associated with both depression and inflammatory
cytes, can also produce cytokines within the CNS (Rothhammer and
markers may have influenced the findings in the inflammatory studies in
Quintana, 2015). In the inflammatory status, the total number of as­
MDD patients. Likewise, to reduce the confounding factors as much as
trocytes increases, and they differentiate into the reactive type (Ben
possible, we applied the similar rigorous criteria for somatic illnesses
Haim et al., 2015; Goetzl and Miller, 2017; Liddelow and Barres, 2017).
that may increase inflammation levels in our study, it might explain why
These reactive astrocytes lose their neuronal trophic potential and in­
our proinflammatory markers in serum were not widely raised in the
crease the expression of proinflammatory pathways(Ceyzeriat et al.,
MDD group. Unexpectedly, the higher serum level of IL-12p70 was
2016; Liddelow and Barres, 2017; Sofroniew, 2014; Zamanian et al.,
found in the HC group, not the MDD group. This is consistent with
2012), and produce IL-6, TNF, IL-1β, IFN-γ, and other pro-inflammatory
Cassano et al.(2017), who reported an approximately 3-fold higher level
cytokines(Ben Haim et al., 2015; John et al., 2003; Krumbholz et al.,
of IL-12p70 in HC group compared to MDD group, but did not reach a
2005; Lau and Yu, 2001; Rothhammer and Quintana, 2015; Van
statistically significant level may due to the low detectable rate. Un­
Wagoner et al., 1999). In fact, for IL-6, astrocytes are the major gener­
fortunately, due to the lack of relevant research on IL-12 in MDD
ator in CNS inflammation status (Gruol and Nelson, 1997), the elevated

58
X.-h. Xie et al. Brain Behavior and Immunity 109 (2023) 51–62

IL-6 and other pro-inflammatory cytokines such as TNF-α, IL-1β, IL-17A among the various studies. Of course, this requires further replication.
may further promote IL-6 production in astrocytes (Ma et al., 2010; Van
Wagoner et al., 1999). As a result, these widely elevated levels of in­ 4.3. Inconsistent inflammatory status in serum and ADEs
flammatory molecules in their producer—astrocytes could be detected
in ADEs. Our study failed to find significant relations of cytokines between
Furthermore, the inflammation hypothesis (Beurel et al., 2020; ADEs and serum. Further, in the sensitive analysis, when analysis the
Miller and Raison, 2016) suggests that the inflammatory cytokine may cytokines in ADEs, we add the concentration of the same cytokine in
passage through leaky regions in the BBB, such as the circumventricular serum as the control variable, and vice versa, the results remained.
organs, and the binding of cytokines to saturable transport molecules on Although, the immune dysregulation is present in both periphery-CNS
the BBB, then drive inflammation in the brain leading to depression. and CNS-peripheral directions (Miller et al., 2017), the issue of the
Hence, measuring the same inflammatory markers in periphery and CNS correlation between the specific inflammatory markers in CNS and pe­
simultaneously may provide a more integrated picture. Because of the riphery is debatable. Among the most commonly tested inflammatory
various measurement techniques used and the absence of unstandard­ markers, the relationships for each inflammatory markers are various. In
ized effect sizes, it is difficult to compare the results of different studies clinical studies, CRP was found has the strongest significant correlation
directly. Therefore, we compared the differences of the same cytokines between periphery and CSF (Felger et al., 2020; Lindqvist et al., 2013),
in periphery and CNS in the same participants, using the same kits from whereas the periphery-CNS correlations of IL-1β (Felger et al., 2020;
the same batch at the same time, and to get a comparable result, we use Lindqvist et al., 2009), IL-6 (Felger et al., 2020; Sasayama et al., 2013),
Hedges’ g as the effect size index. In this study, Hedges’ g values in ADEs IL-8 (Lindqvist et al., 2009), and TNF-α (Felger et al., 2020; Lindqvist
are more consistent and relatively larger, on the contrary, the g values in et al., 2013; Lindqvist et al., 2009) were found nonsignificant, similar to
serum are smaller and more heterogeneous. Furthermore, no significant our results (sTable 4). For IL-1 receptor antagonist (IL-1ra) (r = 0.298)
correlations of cytokines between serum and ADEs were found. This and IL-6 soluble receptor (IL-6sr) (r = 0.418), moderate peripheral-CNS
suggests that inflammatory differences are more significant in ADEs. correlations were found in 73 unmedicated adult outpatients with MDD
Additionally, it appears that CNS inflammation is a process independent (Felger et al., 2020). For some inflammatory chemokines, several small
from peripheral inflammation. It also should be noted that a main sample studies found significant correlations between periphery and
function of EVs is intercellular communication (Kalluri and LeBleu, CSF (Hestad et al., 2016; Levine et al., 1999). This lack of association is
2020; Meldolesi, 2018), and inflammatory factors are cargos in these also shown in preclinical studies including both peripheral and central
vesicles; therefore, when we detect the cargos, namely these cytokines, inflammatory status (Bay-Richter et al., 2011; McColl et al., 2016; Qin
EVs may play a role of enrichment amplification of these cargos. On one et al., 2007; Thomson et al., 2014). In brief, due to the heterogeneity of
hand, this possible amplification effect may result in an overestimation inflammatory markers and the methodological restriction, immune-to-
of the concentration of inflammatory markers in CNS, and subsequently brain communication’s processes have yet to be fully explored. Thus,
lead to an overestimation of the between-group effect size. On the other considering the higher effect sizes of cytokines in ADEs and the non-
hand, if this assumption is correct, we may be able to employ ADEs as a significant periphery–central coefficients, our findings suggest a possi­
natural bio-amplifier to detect changes in inflammation in CNS, which bility that, these cytokines may originate from the CNS, where glia as
warrants additional exploration. Whatever the case, this work offers a astrocytes are known as the major cytokines producer (Gruol and
fresh perspective on the inflammation in the CNS, and raises new Nelson, 1997; John et al., 2003; Krumbholz et al., 2005; Lau and Yu,
questions about the importance of measuring inflammation directly in 2001; Lindqvist et al., 2009; Rothhammer and Quintana, 2015; Van
the CNS in vivo. Wagoner et al., 1999). Fortunately, ADEs demonstrated their potential
The inflammatory markers in ADEs also demonstrated their potential as the in vivo “liquid biopsy” for CNS (Delgado-Peraza et al., 2021), and
as biomarkers for MDD. For single cytokine, the pAUC is approximate allow us to test related hypotheses in the future.
0.6, but the combination of these cytokines can achieve a powerful
distinguishing capability for MDD patients from HCs. We speculated that 4.4. Limitations
the combination of these pro- and anti-inflammatory markers may better
depict a more comprehensive picture of the inflammatory status in CNS Our study has several limitations. First, the amount of ADEs is rela­
and therefore enhanced the distinguishing ability. tively small and insufficient for some high-throughput omics tests, such
Another intriguing finding was that while we found significant in­ as proteomics, transcriptomics, lipidomics, and makes it difficult to gain
creases in a variety of pro-inflammatory factors in ADEs in the MDD a comprehensive understanding of ADEs. Second, we chose a highly
group, an essential anti-inflammatory cytokine, IL-10, whose levels were sensitive multi-factor assay kit for nine cytokines because the small
found to be non-significantly different between MDD and HC groups amounts of ADEs and low-grade levels of peripheral/central inflamma­
neither in serum nor in ADEs. Clinical studies on peripheral IL-10 are tory markers, but the limited number of cytokines targeted of kits limits
highly heterogeneous (Hiles et al., 2012), a recent meta-analysis(Kohler our ability to explore cytokines extensively. The findings of this study,
et al., 2017) demonstrated no significant alternation of peripheral IL-10 however, suggest that future studies should broaden the scope of testing
in antidepressant-free participants with MDD, but significantly elevated in order to gain an integrity picture of important inflammation markers.
in medicated patients. In CNS, IL-10 could reduce the expression of pro- Third, we did not isolate neuron-derived EVs and microglia-derived EVs
inflammatory cytokine and chemokine in astrocytes (Kenison et al., due to serum sample volume and funding constraints, so there is no way
2020; Mayo et al., 2016; Zhang et al., 2020). Production of IL-10 after to estimate the cross-talk among the three major cells in brain, we will
different types of CNS injuries has been demonstrated to have protective continue to investigate the interactions in the next study. Forth, neu­
anti-inflammatory functions (Arimoto et al., 2007; Park et al., 2007; Xin roimaging studies have revealed cytokine-induced alterations in
et al., 2011), and in addition, IL-10 administration could also rescue regional brain activity which involved regulating motivation and motor
depression-like behavior (Mesquita et al., 2008; Zhang et al., 2020) and activity as well as arousal, anxiety and alarm. However, we are currently
associated learning and memory deficits in depression animal model unable to precisely localize the source brain regions for these ADEs due
(Worthen et al., 2020). In line to this study, widespread increased pro- to the methodology restriction. We will develop new protocols,
inflammatory cytokines but no elevation of anti-inflammatory IL-10 in combining neuroimaging methods and EVs, and anticipate it providing
ADEs provided another postulate for the inflammation hypothesis of some coupled position information. Fifth, in the exploratory analysis, no
depression. The insufficient responsive anti-inflammatory cytokines significant correlations between ADEs and clinical characteristics were
production in CNS might provide a concise interpretation of the highly observed, limiting our ability to draw further inferences between the
heterogeneous low-grade inflammatory status in depression population biological process and clinical features. We plan to enhance the

59
X.-h. Xie et al. Brain Behavior and Immunity 109 (2023) 51–62

collection of clinical characteristics for more detailed exploration in the Ben Haim, L., Carrillo-de Sauvage, M.A., Ceyzeriat, K., Escartin, C., 2015. Elusive roles
for reactive astrocytes in neurodegenerative diseases. Front. Cell. Neurosci. 9, 278.
next studies.
Benjamini, Y., Yekutieli, D., 2001. The control of the false discovery rate in multiple
testing under dependency. Ann. Stat. 1165–1188.
5. Conclusions Beurel, E., Toups, M., Nemeroff, C.B., 2020. The bidirectional relationship of depression
and inflammation: double trouble. Neuron 107, 234–256.
Bollen, J., Trick, L., Llewellyn, D., Dickens, C., 2017. The effects of acute inflammation
Our findings on the overall evidence of nine cytokines both in serum on cognitive functioning and emotional processing in humans: A systematic review
and ADEs support the inflammatory glia hypothesis of depression, and of experimental studies. J. Psychosom. Res. 94, 47–55.
suggests that ADEs could be a useful tool to test the in vivo astrocyte Cakici, N., Sutterland, A.L., Penninx, B., Dalm, V.A., de Haan, L., van Beveren, N.J.M.,
2020. Altered peripheral blood compounds in drug-naive first-episode patients with
status in human participants and their role in Depression. Most impor­ either schizophrenia or major depressive disorder: a meta-analysis. Brain Behav.
tantly, our study suggests that in vivo CNS probing via brain-derived EVs Immun. 88, 547–558.
on human participants is feasible and attractive for evaluating existing Casella, G., Rasouli, J., Boehm, A., Zhang, W., Xiao, D., Ishikawa, L.L.W., Thome, R., Li,
X., Hwang, D., Porazzi, P., Molugu, S., Tang, H.Y., Zhang, G.X., Ciric, B., Rostami, A.,
hypotheses or developing new ones. 2020. Oligodendrocyte-derived extracellular vesicles as antigen-specific therapy for
autoimmune neuroinflammation in mice. Sci. Transl. Med., 12.
CRediT authorship contribution statement Cassano, P., Fava, M., 2002. Depression and public health: an overview. J. Psychosom.
Res. 53, 849–857.
Cassano, P., Bui, E., Rogers, A.H., Walton, Z.E., Ross, R., Zeng, M., Nadal-Vicens, M.,
Xin-hui Xie: Conceptualization, Methodology, Formal analysis, Mischoulon, D., Baker, A.W., Keshaviah, A., Worthington, J., Hoge, E.A., Alpert, J.,
Writing – original draft. Wen-tao Lai: Investigation, Data curation, Fava, M., Wong, K.K., Simon, N.M., 2017. Inflammatory cytokines in major
depressive disorder: A case-control study. Aust. N. Z. J. Psychiatry 51, 23–31.
Writing – review & editing. Shu-xian Xu: Data curation, Writing – re­ Ceyzeriat, K., Abjean, L., Carrillo-de Sauvage, M.A., Ben Haim, L., Escartin, C., 2016. The
view & editing. Marta Di Forti: Writing – review & editing. Jing-ya complex STATes of astrocyte reactivity: How are they controlled by the JAK-STAT3
Zhang: Formal analysis, Writing – review & editing. Mian-mian Chen: pathway? Neuroscience 330, 205–218.
Chang, H.B., Munroe, S., Gray, K., Porta, G., Douaihy, A., Marsland, A., Brent, D.,
Formal analysis, Visualization. Li-hua Yao: Writing – review & editing. Melhem, N.M., 2019. The role of substance use, smoking, and inflammation in risk
Peilin Wang: Writing – review & editing. Ke-ke Hao: Writing – review for suicidal behavior. J. Affect. Disord. 243, 33–41.
& editing. Han Rong: Project administration, Supervision, Funding Chen, Y., Xia, K., Chen, L., Fan, D., 2019. Increased interleukin-6 levels in the astrocyte-
derived exosomes of sporadic amyotrophic lateral sclerosis patients. Front. Neurosci.
acquisition.
13, 574.
Collins, P.Y., Patel, V., Joestl, S.S., March, D., Insel, T.R., Daar, A.S., Scientific
Advisory, B., the Executive Committee of the Grand Challenges on Global Mental, H.,
Declaration of Competing Interest
Anderson, W., Dhansay, M.A., Phillips, A., Shurin, S., Walport, M., Ewart, W.,
Savill, S.J., Bordin, I.A., Costello, E.J., Durkin, M., Fairburn, C., Glass, R.I., Hall, W.,
The authors declare that they have no known competing financial Huang, Y., Hyman, S.E., Jamison, K., Kaaya, S., Kapur, S., Kleinman, A.,
interests or personal relationships that could have appeared to influence Ogunniyi, A., Otero-Ojeda, A., Poo, M.M., Ravindranath, V., Sahakian, B.J.,
Saxena, S., Singer, P.A., Stein, D.J., 2011. Grand challenges in global mental health.
the work reported in this paper. Nature 475, 27–30.
Cotter, D., Mackay, D., Chana, G., Beasley, C., Landau, S., Everall, I.P., 2002. Reduced
Data availability neuronal size and glial cell density in area 9 of the dorsolateral prefrontal cortex in
subjects with major depressive disorder. Cereb. Cortex 12, 386–394.
Delgado-Peraza, F., Nogueras-Ortiz, C.J., Volpert, O., Liu, D., Goetzl, E.J., Mattson, M.P.,
Data will be made available on request. Greig, N.H., Eitan, E., Kapogiannis, D., 2021. Neuronal and astrocytic extracellular
vesicle biomarkers in blood reflect brain pathology in mouse models of Alzheimer’s
disease. Cells 10.
Acknowledgements Dong, Y., Benveniste, E.N., 2001. Immune function of astrocytes. Glia 36, 180–190.
Enache, D., Pariante, C.M., Mondelli, V., 2019. Markers of central inflammation in major
depressive disorder: A systematic review and meta-analysis of studies examining
This work was supported by the Sanming Project of Medicine in
cerebrospinal fluid, positron emission tomography and post-mortem brain tissue.
Shenzhen (Grant Number: SZSM201812052), Natural Science Founda­ Brain Behav. Immun. 81, 24–40.
tion of Guangdong Province (Grant Number: 2020A1515011290), Sci­ Falsig, J., Porzgen, P., Lund, S., Schrattenholz, A., Leist, M., 2006. The inflammatory
transcriptome of reactive murine astrocytes and implications for their innate
ence and Technology Program of Shenzhen (Grant Numbers:
immune function. J. Neurochem. 96, 893–907.
JCYJ20190809155403596), and Chinese National Natural Science Faul, F., Erdfelder, E., Buchner, A., Lang, A.G., 2009. Statistical power analyses using
Foundation (Grant Number: 81201047). This work has not received G*Power 3.1: tests for correlation and regression analyses. Behav. Res. Methods 41,
funding/assistance from any commercial organizations. 1149–1160.
Felger, J.C., Haroon, E., Patel, T.A., Goldsmith, D.R., Wommack, E.C., Woolwine, B.J.,
Le, N.A., Feinberg, R., Tansey, M.G., Miller, A.H., 2020. What does plasma CRP tell
Role of the funding sources us about peripheral and central inflammation in depression? Mol. Psychiatry 25,
1301–1311.
Fiandaca, M.S., Kapogiannis, D., Mapstone, M., Boxer, A., Eitan, E., Schwartz, J.B.,
The funding sources had no roles in the design of this study and will Abner, E.L., Petersen, R.C., Federoff, H.J., Miller, B.L., Goetzl, E.J., 2015.
not have any roles during the execution, analyses, interpretation of the Identification of preclinical Alzheimer’s disease by a profile of pathogenic proteins in
neurally derived blood exosomes: A case-control study. Alzheimers Dement. 11
data, or decision to submit results. (600–607), e601.
Goetzl, E.J., Miller, B.L., 2017. Multicellular hypothesis for the pathogenesis of
References Alzheimer’s disease. FASEB J. 31, 1792–1795.
Goetzl, E.J., Mustapic, M., Kapogiannis, D., Eitan, E., Lobach, I.V., Goetzl, L., Schwartz, J.
B., Miller, B.L., 2016. Cargo proteins of plasma astrocyte-derived exosomes in
Arimoto, T., Choi, D.Y., Lu, X., Liu, M., Nguyen, X.V., Zheng, N., Stewart, C.A., Kim, H.C.,
Alzheimer’s disease. FASEB J. 30, 3853–3859.
Bing, G., 2007. Interleukin-10 protects against inflammation-mediated degeneration
Goetzl, E.J., Schwartz, J.B., Abner, E.L., Jicha, G.A., Kapogiannis, D., 2018. High
of dopaminergic neurons in substantia nigra. Neurobiol. Aging 28, 894–906.
complement levels in astrocyte-derived exosomes of Alzheimer disease. Ann. Neurol.
Arteaga-Henriquez, G., Simon, M.S., Burger, B., Weidinger, E., Wijkhuijs, A., Arolt, V.,
83, 544–552.
Birkenhager, T.K., Musil, R., Muller, N., Drexhage, H.A., 2019. Low-grade
Goetzl, E.J., Yaffe, K., Peltz, C.B., Ledreux, A., Gorgens, K., Davidson, B., Granholm, A.C.,
inflammation as a predictor of antidepressant and anti-inflammatory therapy
Mustapic, M., Kapogiannis, D., Tweedie, D., Greig, N.H., 2020. Traumatic brain
response in MDD patients: a systematic review of the literature in combination with
injury increases plasma astrocyte-derived exosome levels of neurotoxic complement
an analysis of experimental data collected in the EU-MOODINFLAME Consortium.
proteins. FASEB J. 34, 3359–3366.
Front. Psych. 10, 458.
Goetzl, E.J., Wolkowitz, O.M., Srihari, V.H., Reus, V.I., Goetzl, L., Kapogiannis, D.,
Barberio, B., Zamani, M., Black, C.J., Savarino, E.V., Ford, A.C., 2021. Prevalence of
Heninger, G.R., Mellon, S.H., 2021. Abnormal levels of mitochondrial proteins in
symptoms of anxiety and depression in patients with inflammatory bowel disease: a
plasma neuronal extracellular vesicles in major depressive disorder. Mol. Psychiatry
systematic review and meta-analysis. Lancet Gastroenterol. Hepatol. 6, 359–370.
26, 7355–7362.
Bay-Richter, C., Janelidze, S., Hallberg, L., Brundin, L., 2011. Changes in behaviour and
Goldsmith, D.R., Rapaport, M.H., Miller, B.J., 2016. A meta-analysis of blood cytokine
cytokine expression upon a peripheral immune challenge. Behav. Brain Res. 222,
network alterations in psychiatric patients: comparisons between schizophrenia,
193–199.
bipolar disorder and depression. Mol. Psychiatry 21, 1696–1709.
Belmaker, R.H., Agam, G., 2008. Major depressive disorder. N. Engl. J. Med. 358, 55–68.

60
X.-h. Xie et al. Brain Behavior and Immunity 109 (2023) 51–62

Gruol, D.L., Nelson, T.E., 1997. Physiological and pathological roles of interleukin-6 in major depressive disorder: a meta-analysis and meta-regression. J. Affect. Disord.
the central nervous system. Mol. Neurobiol. 15, 307–339. 139, 230–239.
Hamilton, M., 1960. A rating scale for depression. J. Neurol. Neurosurg. Psychiatry 23, Losito, I., Patruno, R., Conte, E., Cataldi, T.R., Megli, F.M., Palmisano, F., 2013.
56–62. Phospholipidomics of human blood microparticles. Anal. Chem. 85, 6405–6413.
Hannestad, J., DellaGioia, N., Gallezot, J.D., Lim, K., Nabulsi, N., Esterlis, I., Pittman, B., Ma, X., Reynolds, S.L., Baker, B.J., Li, X., Benveniste, E.N., Qin, H., 2010. IL-17
Lee, J.Y., O’Connor, K.C., Pelletier, D., Carson, R.E., 2013. The neuroinflammation enhancement of the IL-6 signaling cascade in astrocytes. J. Immunol. 184,
marker translocator protein is not elevated in individuals with mild-to-moderate 4898–4906.
depression: a [(1)(1)C]PBR28 PET study. Brain Behav. Immun. 33, 131–138. Matcham, F., Rayner, L., Steer, S., Hotopf, M., 2013. The prevalence of depression in
Herberth, G., Weber, A., Roder, S., Elvers, H.D., Kramer, U., Schins, R.P., Diez, U., rheumatoid arthritis: a systematic review and meta-analysis. Rheumatology (Oxford)
Borte, M., Heinrich, J., Schafer, T., Herbarth, O., Lehmann, I., Group, L.I.s., 2008. 52, 2136–2148.
Relation between stressful life events, neuropeptides and cytokines: results from the Mayo, L., Cunha, A.P., Madi, A., Beynon, V., Yang, Z., Alvarez, J.I., Prat, A., Sobel, R.A.,
LISA birth cohort study. Pediatr. Allergy Immunol. 19, 722–729. Kobzik, L., Lassmann, H., Quintana, F.J., Weiner, H.L., 2016. IL-10-dependent Tr1
Herculano-Houzel, S., 2014. The glia/neuron ratio: how it varies uniformly across brain cells attenuate astrocyte activation and ameliorate chronic central nervous system
structures and species and what that means for brain physiology and evolution. Glia inflammation. Brain 139, 1939–1957.
62, 1377–1391. McColl, A., Thomson, C.A., Nerurkar, L., Graham, G.J., Cavanagh, J., 2016. TLR7-
Hestad, K.A., Engedal, K., Whist, J.E., Aukrust, P., Farup, P.G., Mollnes, T.E., Ueland, T., mediated skin inflammation remotely triggers chemokine expression and leukocyte
2016. Patients with depression display cytokine levels in serum and cerebrospinal accumulation in the brain. J. Neuroinflammation 13, 102.
fluid similar to patients with diffuse neurological symptoms without a defined Medina, A., Watson, S.J., Bunney Jr., W., Myers, R.M., Schatzberg, A., Barchas, J.,
diagnosis. Neuropsychiatr. Dis. Treat. 12, 817–822. Akil, H., Thompson, R.C., 2016. Evidence for alterations of the glial syncytial
Hiles, S.A., Baker, A.L., de Malmanche, T., Attia, J., 2012. A meta-analysis of differences function in major depressive disorder. J. Psychiatr. Res. 72, 15–21.
in IL-6 and IL-10 between people with and without depression: exploring the causes Meldolesi, J., 2018. Exosomes and ectosomes in intercellular communication. Curr. Biol.
of heterogeneity. Brain Behav. Immun. 26, 1180–1188. 28, R435–R444.
Holmes, S.E., Hinz, R., Conen, S., Gregory, C.J., Matthews, J.C., Anton-Rodriguez, J.M., Mesquita, A.R., Correia-Neves, M., Roque, S., Castro, A.G., Vieira, P., Pedrosa, J.,
Gerhard, A., Talbot, P.S., 2018. Elevated translocator protein in anterior cingulate in Palha, J.A., Sousa, N., 2008. IL-10 modulates depressive-like behavior. J. Psychiatr.
major depression and a role for inflammation in suicidal thinking: a positron Res. 43, 89–97.
emission tomography study. Biol. Psychiatry 83, 61–69. Milaneschi, Y., Simmons, W.K., van Rossum, E.F.C., Penninx, B.W., 2019. Depression and
Jia, L., Qiu, Q., Zhang, H., Chu, L., Du, Y., Zhang, J., Zhou, C., Liang, F., Shi, S., Wang, S., obesity: evidence of shared biological mechanisms. Mol. Psychiatry 24, 18–33.
Qin, W., Wang, Q., Li, F., Wang, Q., Li, Y., Shen, L., Wei, Y., Jia, J., 2019. Miller, A.H., Raison, C.L., 2016. The role of inflammation in depression: from
Concordance between the assessment of Abeta42, T-tau, and P-T181-tau in evolutionary imperative to modern treatment target. Nat. Rev. Immunol. 16, 22–34.
peripheral blood neuronal-derived exosomes and cerebrospinal fluid. Alzheimers Miller, A.H., Haroon, E., Felger, J.C., 2017. Therapeutic implications of brain-immune
Dement. 15, 1071–1080. interactions: treatment in translation. Neuropsychopharmacology 42, 334–359.
John, G.R., Lee, S.C., Brosnan, C.F., 2003. Cytokines: powerful regulators of glial cell Moustafa, A.T., Moazzami, M., Engel, L., Bangert, E., Hassanein, M., Marzouk, S.,
activation. Neuroscientist 9, 10–22. Kravtsenyuk, M., Fung, W., Eder, L., Su, J., Wither, J.E., Touma, Z., 2020. Prevalence
Kalluri, R., LeBleu, V.S., 2020. The biology, function, and biomedical applications of and metric of depression and anxiety in systemic lupus erythematosus: A systematic
exosomes. Science (New York N.Y.) 367. review and meta-analysis. Semin. Arthritis Rheum. 50, 84–94.
Kenison, J.E., Jhaveri, A., Li, Z., Khadse, N., Tjon, E., Tezza, S., Nowakowska, D., Mustapic, M., Eitan, E., Werner Jr., J.K., Berkowitz, S.T., Lazaropoulos, M.P., Tran, J.,
Plasencia, A., Stanton Jr., V.P., Sherr, D.H., Quintana, F.J., 2020. Tolerogenic Goetzl, E.J., Kapogiannis, D., 2017. Plasma extracellular vesicles enriched for
nanoparticles suppress central nervous system inflammation. PNAS 117, neuronal origin: a potential window into brain pathologic processes. Front. Neurosci.
32017–32028. 11, 278.
Khakh, B.S., Deneen, B., 2019. The emerging nature of astrocyte diversity. Annu. Rev. Nagy, C., Suderman, M., Yang, J., Szyf, M., Mechawar, N., Ernst, C., Turecki, G., 2015.
Neurosci. 42, 187–207. Astrocytic abnormalities and global DNA methylation patterns in depression and
Khandaker, G.M., Pearson, R.M., Zammit, S., Lewis, G., Jones, P.B., 2014. Association of suicide. Mol. Psychiatry 20, 320–328.
serum interleukin 6 and C-reactive protein in childhood with depression and Najjar, S., Pearlman, D.M., Alper, K., Najjar, A., Devinsky, O., 2013. Neuroinflammation
psychosis in young adult life: a population-based longitudinal study. JAMA Psychiat. and psychiatric illness. J. Neuroinflammation 10, 43.
71, 1121–1128. Ongur, D., Drevets, W.C., Price, J.L., 1998. Glial reduction in the subgenual prefrontal
Kohler, C.A., Freitas, T.H., Maes, M., de Andrade, N.Q., Liu, C.S., Fernandes, B.S., cortex in mood disorders. PNAS 95, 13290–13295.
Stubbs, B., Solmi, M., Veronese, N., Herrmann, N., Raison, C.L., Miller, B.J., Organization, W.H., 1992. The ICD-10 Classification of Mental and Behavioural
Lanctot, K.L., Carvalho, A.F., 2017. Peripheral cytokine and chemokine alterations in Disorders: Clinical Descriptions and Diagnostic Guidelines. World Health
depression: a meta-analysis of 82 studies. Acta Psychiatr. Scand. 135, 373–387. Organization.
Kohno, M., Link, J., Dennis, L.E., McCready, H., Huckans, M., Hoffman, W.F., Loftis, J.M., Osimo, E.F., Pillinger, T., Rodriguez, I.M., Khandaker, G.M., Pariante, C.M., Howes, O.D.,
2019. Neuroinflammation in addiction: A review of neuroimaging studies and 2020. Inflammatory markers in depression: A meta-analysis of mean differences and
potential immunotherapies. Pharmacol. Biochem. Behav 179, 34–42. variability in 5,166 patients and 5,083 controls. Brain Behav. Immun. 87, 901–909.
Krumbholz, M., Theil, D., Derfuss, T., Rosenwald, A., Schrader, F., Monoranu, C.M., Park, K.W., Lee, H.G., Jin, B.K., Lee, Y.B., 2007. Interleukin-10 endogenously expressed
Kalled, S.L., Hess, D.M., Serafini, B., Aloisi, F., Wekerle, H., Hohlfeld, R., Meinl, E., in microglia prevents lipopolysaccharide-induced neurodegeneration in the rat
2005. BAFF is produced by astrocytes and up-regulated in multiple sclerosis lesions cerebral cortex in vivo. Exp. Mol. Med. 39, 812–819.
and primary central nervous system lymphoma. J. Exp. Med. 201, 195–200. Peng, L., Verkhratsky, A., Gu, L., Li, B., 2015. Targeting astrocytes in major depression.
Lau, L.T., Yu, A.C., 2001. Astrocytes produce and release interleukin-1, interleukin-6, Expert Rev. Neurother. 15, 1299–1306.
tumor necrosis factor alpha and interferon-gamma following traumatic and Perneger, T.V., 1998. What’s wrong with Bonferroni adjustments. BMJ 316, 1236–1238.
metabolic injury. J. Neurotrauma 18, 351–359. Pitharouli, M.C., Hagenaars, S.P., Glanville, K.P., Coleman, J.R.I., Hotopf, M., Lewis, C.
Leng, L., Zhuang, K., Liu, Z., Huang, C., Gao, Y., Chen, G., Lin, H., Hu, Y., Wu, D., Shi, M., M., Pariante, C.M., 2021. Elevated C-reactive protein in patients with depression,
Xie, W., Sun, H., Shao, Z., Li, H., Zhang, K., Mo, W., Huang, T.Y., Xue, M., Yuan, Z., independent of genetic, health, and psychosocial factors: results from the UK
Zhang, X., Bu, G., Xu, H., Xu, Q., Zhang, J., 2018. Menin deficiency leads to biobank. Am. J. Psychiatry 178, 522–529.
depressive-like behaviors in mice by modulating astrocyte-mediated Qin, L., Wu, X., Block, M.L., Liu, Y., Breese, G.R., Hong, J.S., Knapp, D.J., Crews, F.T.,
neuroinflammation. Neuron 100 (551–563), e557. 2007. Systemic LPS causes chronic neuroinflammation and progressive
Levine, J., Barak, Y., Chengappa, K.N., Rapoport, A., Rebey, M., Barak, V., 1999. neurodegeneration. Glia 55, 453–462.
Cerebrospinal cytokine levels in patients with acute depression. Neuropsychobiology Rajkowska, G., Stockmeier, C.A., 2013. Astrocyte pathology in major depressive
40, 171–176. disorder: insights from human postmortem brain tissue. Curr. Drug Targets 14,
Liddelow, S.A., Barres, B.A., 2017. Reactive astrocytes: production, function, and 1225–1236.
therapeutic potential. Immunity 46, 957–967. Rajkowska, G., Miguel-Hidalgo, J.J., Wei, J., Dilley, G., Pittman, S.D., Meltzer, H.Y.,
Lindqvist, D., Janelidze, S., Hagell, P., Erhardt, S., Samuelsson, M., Minthon, L., Overholser, J.C., Roth, B.L., Stockmeier, C.A., 1999. Morphometric evidence for
Hansson, O., Bjorkqvist, M., Traskman-Bendz, L., Brundin, L., 2009. Interleukin-6 is neuronal and glial prefrontal cell pathology in major depression. Biol. Psychiatry 45,
elevated in the cerebrospinal fluid of suicide attempters and related to symptom 1085–1098.
severity. Biol. Psychiatry 66, 287–292. Richards, E.M., Zanotti-Fregonara, P., Fujita, M., Newman, L., Farmer, C., Ballard, E.D.,
Lindqvist, D., Hall, S., Surova, Y., Nielsen, H.M., Janelidze, S., Brundin, L., Hansson, O., Machado-Vieira, R., Yuan, P., Niciu, M.J., Lyoo, C.H., Henter, I.D., Salvadore, G.,
2013. Cerebrospinal fluid inflammatory markers in Parkinson’s disease–associations Drevets, W.C., Kolb, H., Innis, R.B., Zarate Jr., C.A., 2018. PET radioligand binding
with depression, fatigue, and cognitive impairment. Brain Behav. Immun. 33, to translocator protein (TSPO) is increased in unmedicated depressed subjects.
183–189. EJNMMI Res. 8, 57.
Lindqvist, D., Dhabhar, F.S., James, S.J., Hough, C.M., Jain, F.A., Bersani, F.S., Reus, V.I., Rothhammer, V., Quintana, F.J., 2015. Control of autoimmune CNS inflammation by
Verhoeven, J.E., Epel, E.S., Mahan, L., Rosser, R., Wolkowitz, O.M., Mellon, S.H., astrocytes. Semin. Immunopathol. 37, 625–638.
2017. Oxidative stress, inflammation and treatment response in major depression. Rowitch, D.H., Kriegstein, A.R., 2010. Developmental genetics of vertebrate glial-cell
Psychoneuroendocrinology 76, 197–205. specification. Nature 468, 214–222.
Linnerbauer, M., Wheeler, M.A., Quintana, F.J., 2020. Astrocyte Crosstalk in CNS Saint-Pol, J., Gosselet, F., Duban-Deweer, S., Pottiez, G., Karamanos, Y., 2020. Targeting
Inflammation. Neuron 108, 608–622. and crossing the blood-brain barrier with extracellular vesicles. Cells 9.
Liu, Y., Ho, R.C., Mak, A., 2012. Interleukin (IL)-6, tumour necrosis factor alpha (TNF- Sasayama, D., Hattori, K., Wakabayashi, C., Teraishi, T., Hori, H., Ota, M., Yoshida, S.,
alpha) and soluble interleukin-2 receptors (sIL-2R) are elevated in patients with Arima, K., Higuchi, T., Amano, N., Kunugi, H., 2013. Increased cerebrospinal fluid

61
X.-h. Xie et al. Brain Behavior and Immunity 109 (2023) 51–62

interleukin-6 levels in patients with schizophrenia and those with major depressive von Elm, E., Altman, D.G., Egger, M., Pocock, S.J., Gotzsche, P.C., Vandenbroucke, J.P.,
disorder. J. Psychiatr. Res. 47, 401–406. Initiative, S., 2007. The Strengthening the Reporting of Observational Studies in
Sheehan, D.V., Lecrubier, Y., Sheehan, K.H., Amorim, P., Janavs, J., Weiller, E., Epidemiology (STROBE) statement: guidelines for reporting observational studies.
Hergueta, T., Baker, R., Dunbar, G.C., 1998. The Mini-International PLoS Med. 4, e296.
Neuropsychiatric Interview (M.I.N.I.): the development and validation of a Wang, Q., Jie, W., Liu, J.H., Yang, J.M., Gao, T.M., 2017. An astroglial basis of major
structured diagnostic psychiatric interview for DSM-IV and ICD-10. J. Clin. depressive disorder? An overview. Glia 65, 1227–1250.
Psychiatry 59 Suppl 20, 22-33;quiz 34-57. Winston, C.N., Goetzl, E.J., Schwartz, J.B., Elahi, F.M., Rissman, R.A., 2019a.
Shi, M., Sheng, L., Stewart, T., Zabetian, C.P., Zhang, J., 2019. New windows into the Complement protein levels in plasma astrocyte-derived exosomes are abnormal in
brain: central nervous system-derived extracellular vesicles in blood. Prog. conversion from mild cognitive impairment to Alzheimer’s disease dementia.
Neurobiol. 175, 96–106. Alzheimers Dement. (Amst) 11, 61–66.
Sofroniew, M.V., 2014. Multiple roles for astrocytes as effectors of cytokines and Winston, C.N., Romero, H.K., Ellisman, M., Nauss, S., Julovich, D.A., Conger, T., Hall, J.
inflammatory mediators. Neuroscientist 20, 160–172. R., Campana, W., O’Bryant, S.E., Nievergelt, C.M., Baker, D.G., Risbrough, V.B.,
Terstappen, G.C., Meyer, A.H., Bell, R.D., Zhang, W., 2021. Strategies for delivering Rissman, R.A., 2019b. Assessing neuronal and astrocyte derived exosomes from
therapeutics across the blood-brain barrier. Nat. Rev. Drug Discov. 20, 362–383. individuals with mild traumatic brain injury for markers of neurodegeneration and
Thomson, C.A., McColl, A., Cavanagh, J., Graham, G.J., 2014. Peripheral inflammation is cytotoxic activity. Front. Neurosci. 13, 1005.
associated with remote global gene expression changes in the brain. Woelfer, M., Kasties, V., Kahlfuss, S., Walter, M., 2019. The role of depressive subtypes
J. Neuroinflammation 11, 73. within the neuroinflammation hypothesis of major depressive disorder.
Torres-Platas, S.G., Hercher, C., Davoli, M.A., Maussion, G., Labonte, B., Turecki, G., Neuroscience 403, 93–110.
Mechawar, N., 2011. Astrocytic hypertrophy in anterior cingulate white matter of Worthen, R.J., Garzon Zighelboim, S.S., Torres Jaramillo, C.S., Beurel, E., 2020. Anti-
depressed suicides. Neuropsychopharmacology 36, 2650–2658. inflammatory IL-10 administration rescues depression-associated learning and
Troubat, R., Barone, P., Leman, S., Desmidt, T., Cressant, A., Atanasova, B., Brizard, B., El memory deficits in mice. J. Neuroinflammation 17, 246.
Hage, W., Surget, A., Belzung, C., Camus, V., 2021. Neuroinflammation and Xin, J., Wainwright, D.A., Mesnard, N.A., Serpe, C.J., Sanders, V.M., Jones, K.J., 2011.
depression: A review. Eur. J. Neurosci. 53, 151–171. IL-10 within the CNS is necessary for CD4+ T cells to mediate neuroprotection. Brain
Valkanova, V., Ebmeier, K.P., Allan, C.L., 2013. CRP, IL-6 and depression: a systematic Behav. Immun. 25, 820–829.
review and meta-analysis of longitudinal studies. J. Affect. Disord. 150, 736–744. Zamanian, J.L., Xu, L., Foo, L.C., Nouri, N., Zhou, L., Giffard, R.G., Barres, B.A., 2012.
Van Wagoner, N.J., Oh, J.W., Repovic, P., Benveniste, E.N., 1999. Interleukin-6 (IL-6) Genomic analysis of reactive astrogliosis. J. Neurosci. 32, 6391–6410.
production by astrocytes: autocrine regulation by IL-6 and the soluble IL-6 receptor. Zhang, H.Y., Wang, Y., He, Y., Wang, T., Huang, X.H., Zhao, C.M., Zhang, L., Li, S.W.,
J. Neurosci. 19, 5236–5244. Wang, C., Qu, Y.N., Jiang, X.X., 2020. A1 astrocytes contribute to murine depression-
Vidal, M., Sainte-Marie, J., Philippot, J.R., Bienvenue, A., 1989. Asymmetric distribution like behavior and cognitive dysfunction, which can be alleviated by IL-10 or
of phospholipids in the membrane of vesicles released during in vitro maturation of fluorocitrate treatment. J. Neuroinflamm. 17, 200.
guinea pig reticulocytes: evidence precluding a role for “aminophospholipid
translocase”. J. Cell. Physiol. 140, 455–462.

62

You might also like