Hostname: page-component-7bb8b95d7b-dtkg6 Total loading time: 0 Render date: 2024-09-26T17:12:38.591Z Has data issue: false hasContentIssue false

The protective effect of vitamin D supplementation as adjunctive therapy to antidepressants on brain structural and functional connectivity of patients with major depressive disorder: a randomized controlled trial

Published online by Cambridge University Press:  14 March 2024

Wenming Zhao
Affiliation:
Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China Research Center of Clinical Medical Imaging, Anhui Province, Hefei 230032, China Anhui Provincial Institute of Translational Medicine, Hefei 230032, China Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, China
Dao-min Zhu
Affiliation:
Department of Sleep Disorders, Affiliated Psychological Hospital of Anhui Medical University, Hefei 230022, China Hefei Fourth People's Hospital, Hefei 230022, China Anhui Mental Health Center, Hefei 230022, China
Yuhao Shen
Affiliation:
Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China Research Center of Clinical Medical Imaging, Anhui Province, Hefei 230032, China Anhui Provincial Institute of Translational Medicine, Hefei 230032, China Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, China
Yu Zhang
Affiliation:
Department of Sleep Disorders, Affiliated Psychological Hospital of Anhui Medical University, Hefei 230022, China Hefei Fourth People's Hospital, Hefei 230022, China Anhui Mental Health Center, Hefei 230022, China
Tao Chen
Affiliation:
Department of Sleep Disorders, Affiliated Psychological Hospital of Anhui Medical University, Hefei 230022, China Hefei Fourth People's Hospital, Hefei 230022, China Anhui Mental Health Center, Hefei 230022, China
Huanhuan Cai
Affiliation:
Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China Research Center of Clinical Medical Imaging, Anhui Province, Hefei 230032, China Anhui Provincial Institute of Translational Medicine, Hefei 230032, China Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, China
Jiajia Zhu*
Affiliation:
Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China Research Center of Clinical Medical Imaging, Anhui Province, Hefei 230032, China Anhui Provincial Institute of Translational Medicine, Hefei 230032, China Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, China
Yongqiang Yu*
Affiliation:
Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China Research Center of Clinical Medical Imaging, Anhui Province, Hefei 230032, China Anhui Provincial Institute of Translational Medicine, Hefei 230032, China Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, China
*
Corresponding author: Yongqiang Yu; Email: cjr.yuyongqiang@vip.163.com; Jiajia Zhu; Email: zhujiajiagraduate@163.com
Corresponding author: Yongqiang Yu; Email: cjr.yuyongqiang@vip.163.com; Jiajia Zhu; Email: zhujiajiagraduate@163.com
Rights & Permissions [Opens in a new window]

Abstract

Background

Growing evidence points to the pivotal role of vitamin D in the pathophysiology and treatment of major depressive disorder (MDD). However, there is a paucity of longitudinal research investigating the effects of vitamin D supplementation on the brain of MDD patients.

Methods

We conducted a double-blind randomized controlled trial in 46 MDD patients, who were randomly allocated into either VD (antidepressant medication + vitamin D supplementation) or NVD (antidepressant medication + placebos) groups. Data from diffusion tensor imaging, resting-state functional MRI, serum vitamin D concentration, and clinical symptoms were obtained at baseline and after an average of 7 months of intervention.

Results

Both VD and NVD groups showed significant improvement in depression and anxiety symptoms but with no significant differences between the two groups. However, a greater increase in serum vitamin D concentration was found to be associated with greater improvement in depression and anxiety symptoms in VD group. More importantly, neuroimaging data demonstrated disrupted white matter integrity of right inferior fronto-occipital fasciculus along with decreased functional connectivity between right frontoparietal and medial visual networks after intervention in NVD group, but no changes in VD group.

Conclusions

These findings suggest that vitamin D supplementation as adjunctive therapy to antidepressants may not only contribute to improvement in clinical symptoms but also help preserve brain structural and functional connectivity in MDD patients.

Type
Original Article
Copyright
Copyright © The Author(s), 2024. Published by Cambridge University Press

Introduction

Major depressive disorder (MDD) is the leading cause of disability in the world and is associated with personal, family, and social burdens (WHO, 2017). In psychiatry, treatment is prescribed on an empirical basis based on clinical profile. Pharmacological and non-pharmacological treatments are available; however, antidepressants are used more frequently than psychological interventions because of inadequate resources (Cipriani et al., Reference Cipriani, Furukawa, Salanti, Chaimani, Atkinson, Ogawa and Geddes2018). When considering the risk-benefit profile of antidepressants in the acute treatment of MDD, these drugs do not seem to offer a clear advantage for young populations (Cipriani et al., Reference Cipriani, Zhou, Del Giovane, Hetrick, Qin, Whittington and Xie2016). Moreover, antidepressants entail side effects and complications, highlighting the importance of alternative therapeutic strategies. Thus, emerging evidence encourages a more integrative approach for MDD and suggests that lifestyle factors such as diet, exercise, and sleep also play a significant mediating role in the development, progression, and treatment of MDD (Jacka et al., Reference Jacka, Kremer, Leslie, Berk, Patton, Toumbourou and Williams2010; Jacka et al., Reference Jacka, Pasco, Williams, Leslie, Dodd, Nicholson and Berk2011; Lopresti, Hood, & Drummond, Reference Lopresti, Hood and Drummond2013; Opie et al., Reference Opie, Itsiopoulos, Parletta, Sanchez-Villegas, Akbaraly, Ruusunen and Jacka2017). Accordingly, lifestyle modification should serve as a routine part of treatment and preventative efforts (Sarris, O'Neil, Coulson, Schweitzer, & Berk, Reference Sarris, O'Neil, Coulson, Schweitzer and Berk2014). In this context, there is increasing recognition of the importance of some nutritional components in MDD (Libuda, Antel, Hebebrand, & Focker, Reference Libuda, Antel, Hebebrand and Focker2017). For example, it is evident that vitamin D might influence risk, pathogenesis, persistence, and treatment of MDD (Bersani et al., Reference Bersani, Ghezzi, Maraone, Vicinanza, Cavaggioni, Biondi and Pasquini2019).

Vitamin D has long been recognized as important for bone health. In recent years, there is also growing interest in its role as a risk factor for mental health. It is quite apparent that vitamin D acts as hormone and fat-soluble vitamin, regulating the expression of more than 900 genes involved in a wide array of physiological functions (Kongsbak, Levring, Geisler, & von Essen, Reference Kongsbak, Levring, Geisler and von Essen2013; Pike et al., Reference Pike, Meyer, Martowicz, Bishop, Lee, Nerenz and Goetsch2010). In addition, vitamin D is engaged in various neural processes, such as the growth and development of neurons as well as the synthesis, release, and regulation of neurotransmitters (Kesby et al., Reference Kesby, Turner, Alexander, Eyles, McGrath and Burne2017; Patrick & Ames, Reference Patrick and Ames2015). Moreover, metabolites of vitamin D can cross the blood-brain barrier and bind to vitamin D receptors (Ryan, Anderson, & Morris, Reference Ryan, Anderson and Morris2015), which are broadly distributed in the brain (e.g. the prefrontal cortex and limbic system) (Eyles, Smith, Kinobe, Hewison, & McGrath, Reference Eyles, Smith, Kinobe, Hewison and McGrath2005), providing a mechanistic account for the role of vitamin D in emotion, cognition, and neuropsychiatric disorders (Di Somma et al., Reference Di Somma, Scarano, Barrea, Zhukouskaya, Savastano, Mele and Marzullo2017; Eyles, Burne, & McGrath, Reference Eyles, Burne and McGrath2013; Schlogl & Holick, Reference Schlogl and Holick2014). Crucially, a large number of clinical studies have provided strong evidence for an association between low vitamin D and MDD (Aghajafari, Letourneau, Mahinpey, Cosic, & Giesbrecht, Reference Aghajafari, Letourneau, Mahinpey, Cosic and Giesbrecht2018; Anglin, Samaan, Walter, & McDonald, Reference Anglin, Samaan, Walter and McDonald2013; Briggs et al., Reference Briggs, McCarroll, O'Halloran, Healy, Kenny and Laird2019; Ju, Lee, & Jeong, Reference Ju, Lee and Jeong2013; Milaneschi et al., Reference Milaneschi, Hoogendijk, Lips, Heijboer, Schoevers, van Hemert and Penninx2014; Parker, Brotchie, & Graham, Reference Parker, Brotchie and Graham2017; Wong, Chin, & Ima-Nirwana, Reference Wong, Chin and Ima-Nirwana2018), suggesting that hypovitaminosis D may represent an underlying vulnerability for depression. Several possible mechanisms that may be responsible for the link between low vitamin D and MDD have been proposed: (1) elevation in both Ca2+ and reactive oxygen species levels in neuronal cells (Berridge, Reference Berridge2015, Reference Berridge2017; Kalueff, Eremin, & Tuohimaa, Reference Kalueff, Eremin and Tuohimaa2004), (2) epigenetic alterations including the hypermethylation of gene promotors (Berridge, Reference Berridge2017; Guidotti et al., Reference Guidotti, Auta, Chen, Davis, Dong, Gavin and Tueting2011; Saavedra, Molina-Marquez, Saavedra, Zambrano, & Salazar, Reference Saavedra, Molina-Marquez, Saavedra, Zambrano and Salazar2016), (3) abnormal serotonin synthesis (Geng et al., Reference Geng, Shaikh, Han, Chen, Guo and Jiang2019), (4) excessive activation of the immune system (Geng et al., Reference Geng, Shaikh, Han, Chen, Guo and Jiang2019), and (5) dysfunctional inflammatory pathways (Alroy, Towers, & Freedman, Reference Alroy, Towers and Freedman1995; Beurel, Toups, & Nemeroff, Reference Beurel, Toups and Nemeroff2020; McCann & Ames, Reference McCann and Ames2008; Sun et al., Reference Sun, Kong, Duan, Szeto, Liao, Madara and Li2006). Moreover, a previous review concludes that vitamin D deficiency is associated with an increased risk of depression and vitamin D supplementation is of benefit for depressed individuals who are vitamin D deficient (Parker et al., Reference Parker, Brotchie and Graham2017). Another recent systematic review and meta-analysis of randomized controlled trial (RCT) suggests that vitamin D supplementation can reduce negative emotions, with patients with MDD and individuals with vitamin D deficiency most likely to benefit from supplementation (Cheng, Huang, & Huang, Reference Cheng, Huang and Huang2020). However, there are prior RCT indicating no effect of vitamin D supplementation on depression (Gowda, Mutowo, Smith, Wluka, & Renzaho, Reference Gowda, Mutowo, Smith, Wluka and Renzaho2015), or no differences in depression outcomes between vitamin D supplementation and placebo (Okereke et al., Reference Okereke, Reynolds, Mischoulon, Chang, Vyas, Cook and Manson2020; Okereke & Singh, Reference Okereke and Singh2016). The heterogeneity in these findings may be due to differences in the samples or the dosage and duration of vitamin D supplementation. Although several attempts have been made to identify the neural substrates underlying the association between low vitamin D and depression (Zhao et al., Reference Zhao, Zhu, Li, Xu, Zhang, Zhang and Yu2023; Zhao et al., Reference Zhao, Zhu, Li, Cui, Jiang, Wang and Yu2022; Zhu et al., Reference Zhu, Zhao, Zhang, Zhang, Yang, Zhang and Yu2019; Zhu et al., Reference Zhu, Zhao, Cui, Jiang, Zhang, Zhang and Yu2022), there is a paucity of longitudinal research investigating the effects of vitamin D supplementation on the brain of MDD patients.

The notion that specific cognitive and emotional processes arise from functionally distinct brain regions has lately shifted toward a connectivity-based approach that emphasizes the role of network-mediated integration across regions. Likewise, the clinical neuroscience has shifted from predominantly lesion-based approach to a connectomic paradigm, framing mental disorders including MDD as dysconnectivity syndromes. Magnetic resonance imaging (MRI) has provided a safe, non-invasive, and easily repeated neuroimaging tool to investigate the underlying neuropathology of MDD (Luo, You, DelBello, Gong, & Li, Reference Luo, You, DelBello, Gong and Li2022). Structural and functional connections of brain networks can be characterized by white matter integrity measured by diffusion tensor imaging (DTI) (Alexander, Lee, Lazar, & Field, Reference Alexander, Lee, Lazar and Field2007) and functional network connectivity measured by resting-state functional MRI (fMRI) (Fox & Raichle, Reference Fox and Raichle2007), respectively. These approaches have been widely applied to examine impairments in white matter integrity (Chen et al., Reference Chen, Guo, Zhu, Kuang, Bi, Ai and Gong2017; Chen et al., Reference Chen, Hu, Li, Huang, Lui, Kuang and Gong2016; Jiang et al., Reference Jiang, Zhao, Hu, Du, Chen, Wu and Gong2017; Yang et al., Reference Yang, Zhu, Zhang, Zhang, Wang, Zhang and Yu2020; Zhang et al., Reference Zhang, Yang, Zhu, Zhao, Zhang, Zhang and Yu2020) and functional network connectivity (Albert, Potter, Boyd, Kang, & Taylor, Reference Albert, Potter, Boyd, Kang and Taylor2019; Chen et al., Reference Chen, Liu, Zhang, Zhang, Xue, Lin and Deng2019; Jiao et al., Reference Jiao, Xu, Teng, Song, Xiao, Fox and Zhong2020; Liu et al., Reference Liu, Jiao, Zhong, Hao, Wang, Xu and Zhang2021; Liu et al., Reference Liu, Xu, Zhang, Jiang, Li and Luo2019; Liu et al., Reference Liu, Chen, Liang, Li, Zheng, Zhang and Qiu2020; Sacchet et al., Reference Sacchet, Ho, Connolly, Tymofiyeva, Lewinn, Han and Yang2016; Wu et al., Reference Wu, Zeng, Shen, Yuan, Qin, Zhang and Hu2017; Yu et al., Reference Yu, Linn, Shinohara, Oathes, Cook, Duprat and Sheline2019; Zhu et al., Reference Zhu, Zhao, Cui, Jiang, Zhang, Zhang and Yu2022) in MDD. Remarkably, using a combination of DTI and resting-state fMRI techniques, investigators have found abnormal brain structural and functional connectivity in MDD (de Kwaasteniet et al., Reference de Kwaasteniet, Ruhe, Caan, Rive, Olabarriaga, Groefsema and Denys2013; Li et al., Reference Li, Lin, Liu, Su, Zhu, Zheng and Sun2020; Yin et al., Reference Yin, He, Xu, Hou, Song, Sui and Yuan2016), pointing to the potential utility of a combined analysis of multimodal imaging data in achieving a more complete investigation of disease mechanisms.

In this study, we conducted a double-blind RCT to investigate the effects of vitamin D supplementation on brain structural and functional connectivity in MDD patients using a combination of DTI and resting-state fMRI. We hypothesized that adjuvant vitamin D supplementation therapy would help to preserve white matter integrity and functional network connectivity in addition to better ameliorating clinical symptoms.

Materials and methods

Participants

Patients were recruited consecutively from Affiliated Psychological Hospital of Anhui Medical University. Healthy controls (HC) were enrolled from the local community via poster advertisements. A total of 98 right-handed subjects were enrolled, including 72 MDD patients and 26 matched HC. Two well-trained clinical psychiatrists confirmed the diagnosis of depression using the MINI-International Neuropsychiatric Interview (M.I.N.I.) following the International Classification of Diseases (ICD-10) criteria. HC were carefully screened to confirm the absence of any psychiatric illness using the M.I.N.I. Right handedness was determined by the Edinburgh handedness inventory (Oldfield, Reference Oldfield1971). The exclusion criteria for all participants included: (1) the presence of other psychiatric disorders such as substance-induced (e.g. drug and alcohol) mood disorder, bipolar disorders, anxiety disorders, schizophrenia, substance abuse, or addiction; (2) a history of significant physical or neurological disease; (3) a history of head injury with loss of consciousness; and (4) contraindications for MRI such as pregnancy. Additional exclusion criterion for HC was a family history of major neurological or psychiatric illnesses among their first-degree relatives. For all patients, we used the 24-item Hamilton Rating Scale for Depression (HAMD) (Williams, Reference Williams1988) and the 14-item Hamilton Rating Scale for Anxiety (HAMA) (Thompson, Reference Thompson2015) to assess the severity of depression and anxiety symptoms. Notably, we used the Chinese versions of HAMD and HAMA, which have been documented to show great reliability and validity (Zheng et al., Reference Zheng, Zhao, Phillips, Liu, Cai, Sun and Huang1988) and have been widely applied in the Chinese population (Guo et al., Reference Guo, Xiang, Xiao, Hu, Chiu, Ungvari and Wang2015; Lai et al., Reference Lai, Deng, Xu, Zhao, Xu, Liu and Rong2021; Shen et al., Reference Shen, Wei, Yang, Zhang, Du, Jia and Zhang2020; Tong et al., Reference Tong, Bo, Shi, Dong, Sun, Gao and Yang2021). This study was approved by the ethics committee of The First Affiliated Hospital of Anhui Medical University. Written informed consent was obtained from all participants after being given a complete description of the study. This study was registered in the Chinese Clinical Trial Registry (ChiCTR) (Registration No: ChiCTR2100054570) at http://www.chictr.org.cn.

Study design

Following successful completion of screening, MDD patients entered a double-blind randomized controlled phase II trial. The 72 patients were randomly allocated into either VD (antidepressant medication + vitamin D supplementation; n = 36) or NVD (antidepressant medication + placebos; n = 36) groups. Both groups of patients received their regular antidepressant medication as prescribed by the attending psychiatrists, including selective serotonin reuptake inhibitors (SSRIs), serotonin-norepinephrine reuptake inhibitors (SNRIs), or noradrenergic and specific serotonergic antidepressants (NaSSA). VD group received vitamin D3 supplements (1600 IU/day), while NVD group received placebos of similar appearance and packaging. The adherence assessment was done based on supplement/placebo count and patient self-report. Notably, the dose of vitamin D used in this study was safe considering the upper tolerable intake level of vitamin D for adults (4000 IU/day) (Patrick & Ames, Reference Patrick and Ames2015). Both groups of patients completed two study visits: baseline (before intervention) and follow-up (after an average of 7 months of intervention). This period was selected as the follow-up visit since it is the conventional time frame for determining recovery from a depressive episode and is thus appropriate for evaluating clinical and psychosocial outcomes. MDD patients underwent serum concentration of vitamin D (SCVD) measurement, MRI examination, and clinical assessment at each visit. HC underwent SCVD measurement, MRI examination, and clinical assessment at baseline.

Serum concentration of vitamin D measurement

After an overnight fasting period, peripheral venous blood samples (2 ml) were collected from all participants in the morning of MRI scanning. Samples were sent to the Department of Clinical Laboratory, Affiliated Psychological Hospital of Anhui Medical University immediately for centrifugation and serum was separated. Vitamin D [25(OH)D] was measured in serum using a chemiluminescence immunoassay (CLIA) technique in a fully automated Maglumi 1000 analyzer (SNIBE Co., Ltd., China). Internal quality control provided by the manufacturer was used to assure quality. SCVD was stratified as follows: 30–100 ng/ml (75–250 nmol/L) as sufficiency, 20–30 ng/ml (50–75 nmol/L) as insufficiency, and <20 ng/ml (50 nmol/L) as deficiency (Ringe & Kipshoven, Reference Ringe and Kipshoven2012).

Image acquisition

MRI data were obtained on a 3.0-Tesla MR system (Discovery MR750w, General Electric, Milwaukee, WI, USA). Details regarding the image acquisition are presented in the Supplemental Materials.

White matter integrity analysis

For DTI data, standard processing steps were performed by using the FMRIB Software Library (FSL, www.fmrib.ox.ac.uk/fsl). Eddy currents in the gradient coils induce stretches and shears in the diffusion weighted images. These distortions are different for different gradient directions. Eddy current correction implemented in the FSL was adopted to correct for these distortions and simple head motions by registering the diffusion weighted images to a reference volume (i.e. the first b0 image) using affine transformations. Correspondingly, the diffusion gradient direction of each diffusion weighted image was rotated according to the resultant affine transformation information (Leemans & Jones, Reference Leemans and Jones2009). Brain tissues were extracted using FSL's brain extraction tool (http://www.fmrib.ox.ac.uk/fsl/bet2). Next, the three-dimensional maps of the diffusion tensor and fractional anisotropy (FA) were calculated by using the DTIFIT toolbox. Then, the tract-based spatial statistics (TBSS) pipeline was conducted (Smith et al., Reference Smith, Jenkinson, Johansen-Berg, Rueckert, Nichols, Mackay and Behrens2006). Briefly, individual FA images were initially non-linearly registered to the Montreal Neurological Institute (MNI) space, with a mean FA image created and thinned to generate a mean FA skeleton. Next, each subject's FA image was projected onto the skeleton via filling the mean FA skeleton with FA values from the nearest relevant tract center by searching perpendicular to the local skeleton structure for maximum FA value. Finally, the Johns Hopkins University (JHU) probabilistic white matter atlas was used to define 50 white matter tracts in the whole brain (Hua et al., Reference Hua, Zhang, Wakana, Jiang, Li, Reich and Mori2008). Mean FA within each white matter tract were extracted from the skeletonized FA images for subsequent analysis.

fMRI data preprocessing

Resting-state fMRI data were preprocessed using Statistical Parametric Mapping software (SPM12, http://www.fil.ion.ucl.ac.uk/spm) and Data Processing & Analysis for Brain Imaging (DPABI, http://rfmri.org/dpabi) (Yan, Wang, Zuo, & Zang, Reference Yan, Wang, Zuo and Zang2016). The first 10 volumes for each subject were discarded and the remaining volumes were corrected for the acquisition time delay between slices. Realignment was then performed to correct the motion between time points. Head motion parameters were computed by estimating the translation in each direction and the angular rotation on each axis for each volume. All participants' BOLD data were within the defined motion thresholds (i.e. maximum translational or rotational motion parameters less than 2.5 mm or 2.5°). We also calculated frame-wise displacement (FD), which indexes the volume-to-volume changes in head position. In the normalization step, individual structural images were firstly co-registered with the mean functional images; the transformed structural images were then segmented and normalized to the MNI space using a high-level nonlinear warping algorithm, i.e. the diffeomorphic anatomical registration through exponentiated Lie algebra (DARTEL) technique (Ashburner, Reference Ashburner2007). Finally, each functional volume was spatially normalized to the MNI space using the deformation parameters estimated during the above step and resampled into a 3-mm isotropic voxel. After spatial normalization, all datasets were smoothed with a 6 mm full-width at half-maximum (FWHM) Gaussian kernel.

Independent component analysis

ICA was conducted to parcellate the preprocessed fMRI data with the GIFT toolbox (mialab.mrn.org/software/gift/) and the number of independent components (N = 24) was estimated automatically by the software using the minimum description length criteria. Spatial ICA decomposes the participant data into linear mixtures of spatially independent components that exhibit a unique time course profile. This was achieved by using two data reduction steps. First, principal component analysis was applied to reduce the subject-specific data into 36 principle components. Next, reduced data of all subjects were concatenated across time and decomposed into 24 independent components using the infomax algorithm. To ensure estimation stability, the infomax algorithm was repeated 20 times in ICASSO (Luo et al., Reference Luo, Li, Wang, He, Wang, You and Li2023b) (http://research.ics.tkk.fi/ica/icasso/), and the most central run was selected and analyzed further. Finally, participant specific spatial maps and time courses were obtained using the GICA back reconstruction approach.

We identified as functional networks several independent components that had peak activations in gray matter, showed low spatial overlap with known vascular, ventricular, motion, and susceptibility artifacts, and exhibited primarily low-frequency power. This selection procedure resulted in 11 functional networks out of the 24 independent components obtained (online Supplementary Figure S1): anterior and posterior default mode networks (aDMN and pDMN), dorsal and ventral attention networks (DAN and VAN), posterior and medial visual networks (pVN and mVN), left and right frontoparietal networks (lFPN and rFPN), sensorimotor network (SMN), and salience network (SN), and auditory network (AN).

Before internetwork functional connectivity calculation, the following additional postprocessing steps were performed on the time courses of the selected functional networks: (1) detrending linear, quadratic, and cubic trends; (2) despiking detected outliers; and (3) low-pass filtering with a cut-off frequency of 0.15 Hz. Then, internetwork functional connectivity was estimated as the Pearson's correlation coefficients between pairs of time courses of the functional networks, resulting in a symmetric 11 × 11 correlation matrix for each subject. Finally, correlations were transformed into Fisher's Z-scores to improve normality.

Statistical analysis

The statistical analyses of demographic and clinical data were performed using the SPSS 23.0 software package (SPSS, Chicago, IL, USA). For cross-sectional analyses of baseline data, we compared age, education, body mass index (BMI), SCVD and FD among VD, NVD, and control groups using one-way analyses of variance (ANOVA). Pearson Chi-square test was used to examine group difference in gender. Two-sample t tests were utilized to compare HAMD, HAMA, illness duration, and intervention duration between VD and NVD groups. For longitudinal analyses of SCVD, HAMD and HAMA, we adopted two-way repeated-measures ANOVA that included the between-subject factor group (VD v. NVD) and the within-subject factor time (baseline v. follow-up). Potential main effects of group and time as well as group-by-time interactions were followed by appropriate two-sample t-tests for comparing groups and paired t-tests for comparing time points. In addition, we calculated longitudinal changes (follow-up - baseline) in SCVD and clinical symptoms (HAMD and HAMA), followed by Pearson's correlation analyses to test their associations within each group. A threshold of p < 0.05 was considered to indicate statistical significance.

For neuroimaging data, we initially tested differences in baseline FA of 50 white matter tracts across VD, NVD, and control groups using one-way ANOVA. Then, we used the above-described two-way repeated-measures ANOVA to assess the main effects and interactions on FA, followed by post hoc two-sample and paired t-tests. Notably, in case of a significant group-by-time interaction on FA of a white matter tract, we further adopted the same analytic strategy (i.e. a combination of two-way repeated-measures ANOVA and post hoc t-tests) to examine functional connectivity between the functional networks connected by that white matter tract. For completeness, we also carried out an exploratory investigation of other functional network connectivity. Moreover, we conducted within-group correlations between changes in neuroimaging measures (FA and functional connectivity) and clinical variables (SCVD, HAMD, and HAMA). For these analyses, multiple comparison correction was performed using the false discovery rate (FDR) method with a corrected significance level of p < 0.05.

Sensitivity analysis

To test the possible effects of antidepressants and illness duration on our results, we included antidepressant types (SSRIs, SNRIs, and NaSSA) and illness duration as nuisance covariates in our analyses.

Results

Demographic and clinical characteristics at baseline

Eligibility, examinations, randomization, and follow-up of the participants are illustrated in Fig. 1. 20 participants were lost to follow-up and six were excluded due to poor MRI quality. Ultimately, a total of 72 participants completed the study, including 20 patients in VD group, 26 patients in NVD group, and 26 HC. Out of the 26 patients in NVD group, 2 were excluded from fMRI analysis due to excessive head motion during scanning. Demographic and clinical characteristics of the participants at baseline are shown in Table 1. The three groups did not diverge on demographic variables including gender, age, education, BMI, and FD. VD and NVD groups did not differ in clinical variables including HAMD, HAMA, illness duration, and intervention duration. With respect to baseline SCVD, there was a significant difference among the three groups, with post hoc t-tests demonstrating lower SCVD in VD and NVD groups relative to HC, but no difference between VD and NVD groups (Fig. 2).

Figure 1. Flowchart of the study design. Abbreviations: HC, healthy controls; MDD, major depressive disorder; MRI, magnetic resonance imaging; SCVD, serum concentration of vitamin D; VD, vitamin D supplementation; NVD, no vitamin D supplementation; DTI, diffusion tensor imaging; fMRI, functional magnetic resonance imaging.

Table 1. Demographic and clinical characteristics of the participants at baseline

Except for gender designation, data are expressed as mean ± standard deviation.

Abbreviations: VD, vitamin D supplementation; NVD, no vitamin D supplementation; HC, healthy controls; BMI, body mass index; SCVD, serum concentration of vitamin D; FD, frame-wise displacement; HAMD, Hamilton Rating Scale for Depression; HAMA, Hamilton Rating Scale for Anxiety; SSRIs, selective serotonin reuptake inhibitors; SNRIs, serotonin norepinephrine reuptake inhibitors; NaSSA, noradrenergic and specific serotonergic antidepressant.

a The data are available for 24 of 26 patients in NVD group.

Figure 2. SCVD at baseline and follow-up. Abbreviations: HC, healthy controls; VD, vitamin D supplementation; NVD, no vitamin D supplementation; SCVD, serum concentration of vitamin D.

Serum concentration of vitamin D and clinical symptoms

Two-way repeated-measures ANOVA revealed a significant group-by-time interaction effect on SCVD (F = 44.137, p < 0.001) (Fig. 2). Post hoc analyses demonstrated increased SCVD from baseline to follow-up in VD group, but no change in NVD group. At follow-up, there was no difference in SCVD between VD and control groups, but NVD group still showed lower SCVD than HC. In terms of clinical symptoms, both VD and NVD groups exhibited significant reductions in HAMD (Fig. 3a) and HAMA (Fig. 3c) from baseline to follow-up, indicating improvement in depression and anxiety symptoms. However, no significant group-by-time interactions were found for HAMD (F = 1.119, p = 0.296) or HAMA (F = 0.124, p = 0.726). Moreover, we observed significant negative correlations of SCVD change with changes in HAMD (r = −0.539, p = 0.017) (Fig. 3b) and HAMA (r = −0.675, p = 0.002) (Fig. 3d) in VD group but not NVD group (HAMD, r = −0.099, p = 0.637; HAMD, r = −0.055, p = 0.795). None of the patients complained about adverse drug reactions.

Figure 3. Longitudinal changes in clinical symptoms and their associations with SCVD change. Changes in HAMD (a) and their associations with SCVD change in VD group (b). Changes in HAMA (c) and their associations with SCVD change in VD group (d). Abbreviations: HAMD, Hamilton Rating Scale for Depression; HAMA, Hamilton Rating Scale for Anxiety; SCVD, serum concentration of vitamin D; VD, vitamin D supplementation; NVD, no vitamin D supplementation.

White matter integrity and functional network connectivity

At baseline, FA of 50 white matter tracts did not differ among VD, NVD, and control groups (online Supplementary Table S1 in the Supplementary Materials). Two-way repeated-measures ANOVA showed a significant group-by-time interaction effect on FA of right inferior fronto-occipital fasciculus (IFOF) (F = 14.677, p < 0.001) that survived FDR correction (Fig. 4 and online Supplementary Table S2 in the Supplementary Materials). Post hoc analyses demonstrated decreased FA of right IFOF from baseline to follow-up in NVD group, but no change in VD group. At follow-up, NVD group showed lower FA of right IFOF than HC, but there was still no difference between VD group and HC.

Figure 4. FA of right IFOF at baseline and follow-up. Abbreviations: FA, fractional anisotropy; IFOF, inferior fronto-occipital fasciculus; HC, healthy controls; VD, vitamin D supplementation; NVD, no vitamin D supplementation; R, right.

Since right IFOF connects right frontal and occipital cortex, we focused our functional connectivity analyses on functional networks involving right frontal cortex (rFPN, aDMN, and VAN) and right occipital cortex (mVN and pVN). Two-way repeated-measures ANOVA revealed a significant group-by-time interaction effect on functional connectivity between rFPN and mVN (F = 7.875, p = 0.008) that survived FDR correction (Fig. 5). Post hoc analyses demonstrated decreased rFPN-mVN connectivity from baseline to follow-up in NVD group, but no change in VD group. The exploratory investigation did not reveal any significant group-by-time interaction effects on other functional networks connectivity (p > 0.05, FDR corrected) (online Supplementary Table S3 in the Supplementary Materials).

Figure 5. Functional connectivity between rFPN and mVN at baseline and follow-up. Abbreviations: HC, healthy controls; VD, vitamin D supplementation; NVD, no vitamin D supplementation; rFPN, right frontoparietal network; mVN, medial visual network.

No significant correlations between changes in neuroimaging measures (FA and functional connectivity) and clinical variables (SCVD, HAMD, and HAMA) were observed in either group (online Supplementary Table S4 in the Supplementary Materials).

Sensitivity analysis

After controlling for antidepressant types and illness duration, the group-by-time interaction effects on SCVD (F = 45.402, p < 0.001), FA of right IFOF (F = 13.744, p < 0.001), and rFPN-mVN connectivity (F = 7.657, p = 0.009) remained significant. The group-by-time interaction effects on HAMD (F = 1.342, p = 0.253) and HAMA (F = 0.088, p = 0.768) were still not significant. The correlation between changes in SCVD and HAMA remained significant (partial correlation coefficient [pr] = −0.512, p = 0.036), but the correlation between changes in SCVD and HAMD was not significant (pr = −0.357, p = 0.159).

Discussion

This is the first RCT to investigate the effects of vitamin D supplementation on brain structural and functional connectivity in MDD patients using a combination of DTI and resting-state fMRI. After an average of 7 months of intervention, both VD and NVD groups showed significant improvement in depression and anxiety symptoms, but with no significant differences between the two groups. However, a greater SCVD increase was found to be associated with greater improvement in depression and anxiety symptoms in VD group. More importantly, neuroimaging data demonstrated decreased FA of right IFOF along with decreased rFPN-mVN functional connectivity after intervention in NVD group, but no changes in VD group. The novel contribution of these findings is the demonstration that vitamin D supplementation as adjunctive therapy to antidepressants may not only contribute to improvement in clinical symptoms but also help to preserve white matter integrity and functional network connectivity in MDD patients, supporting the neuroprotective and anti-inflammatory role of vitamin D.

Previous studies examining the effects of vitamin D supplementation on MDD have produced mixed findings, with some demonstrating that vitamin D supplementation is beneficial for depression treatment and prevention (Alavi, Khademalhoseini, Vakili, & Assarian, Reference Alavi, Khademalhoseini, Vakili and Assarian2019; Alghamdi et al., Reference Alghamdi, Alsulami, Khoja, Alsufiani, Tayeb and Tarazi2020; Casseb, Kaster, & Rodrigues, Reference Casseb, Kaster and Rodrigues2019; Cheng et al., Reference Cheng, Huang and Huang2020; de Koning et al., Reference de Koning, van Schoor, Penninx, Elders, Heijboer, Smit and Lips2015; Kaviani, Nikooyeh, Zand, Yaghmaei, & Neyestani, Reference Kaviani, Nikooyeh, Zand, Yaghmaei and Neyestani2020; Khoraminya, Tehrani-Doost, Jazayeri, Hosseini, & Djazayery, Reference Khoraminya, Tehrani-Doost, Jazayeri, Hosseini and Djazayery2013; Parker et al., Reference Parker, Brotchie and Graham2017) and others showing no effects (Gowda et al., Reference Gowda, Mutowo, Smith, Wluka and Renzaho2015; Okereke et al., Reference Okereke, Reynolds, Mischoulon, Chang, Vyas, Cook and Manson2020; Okereke & Singh, Reference Okereke and Singh2016). These heterogeneous results may be attributed to differences in the samples or the dosage and duration of vitamin D supplementation. In our RCT, the antidepressants plus vitamin D supplements combination was not superior to the antidepressants plus placebos combination in controlling depression and anxiety symptoms after an average of 7 months of intervention, which may be due to the small sample size. However, we found that a greater SCVD increase was associated with greater improvement in depression and anxiety symptoms in VD group, giving indirect support for the view that adjuvant vitamin D supplementation therapy could contribute to better clinical improvement in MDD patients. The underlying biological mechanisms are not completely understood but there are several hypotheses. First, the active form of vitamin D can be produced by the brain and vitamin D receptors are widely distributed across distinct brain systems (e.g. the prefrontal cortex and limbic system) (Eyles et al., Reference Eyles, Smith, Kinobe, Hewison and McGrath2005). Vitamin D in the brain can regulate production of serotonin, which is one of the key neurotransmitters involved in mood regulation (Kesby et al., Reference Kesby, Turner, Alexander, Eyles, McGrath and Burne2017; Patrick & Ames, Reference Patrick and Ames2015). Second, converging evidence indicates that an imbalance between the glutamatergic excitatory and GABAergic inhibitory pathways is implicated in the pathophysiology of MDD (Croarkin, Levinson, & Daskalakis, Reference Croarkin, Levinson and Daskalakis2011; Hasler et al., Reference Hasler, van der Veen, Tumonis, Meyers, Shen and Drevets2007). This imbalance can lead to increased neuronal levels of Ca2+ that may contribute to depression; vitamin D can reduce Ca2+ levels via its function to maintain the expression of the Ca2+ pumps and buffers, which may explain how it acts to ameliorate depressive symptoms (Berridge, Reference Berridge2017). Third, vitamin D may play a neuroimmunological role by regulating the activity and expression of P2X7 receptors, thus preventing the excessive activation of the immune system that is caused by long-term stress, protecting nerve cells, and producing antidepressant effects (Geng et al., Reference Geng, Shaikh, Han, Chen, Guo and Jiang2019).

Multimodal brain imaging data demonstrated that MDD patients, after antidepressants + placebos intervention, showed decreased FA of right IFOF and decreased functional connectivity between rFPN and mVN connected by right IFOF. The present observation of concurrent structural and functional connectivity alterations is consistent with the hypothesis that brain network function is shaped and constrained by underlying structure (Honey, Kotter, Breakspear, & Sporns, Reference Honey, Kotter, Breakspear and Sporns2007; Honey et al., Reference Honey, Sporns, Cammoun, Gigandet, Thiran, Meuli and Hagmann2009). The FPN is responsible for implementing cognitive control and consists of flexible hubs that regulate distributed systems (e.g. visual, limbic, and motor), thereby playing a central role in mental health (Cole, Repovs, & Anticevic, Reference Cole, Repovs and Anticevic2014). The mVN is implicated in detecting and processing visual stimuli. Extensive research has established the presence of abnormal functional connectivity of FPN and mVN in MDD patients (Kaiser, Andrews-Hanna, Wager, & Pizzagalli, Reference Kaiser, Andrews-Hanna, Wager and Pizzagalli2015; Li et al., Reference Li, Lin, Liu, Su, Zhu, Zheng and Sun2020; Luo et al., Reference Luo, Wu, Xu, Chen, Wu, Wang and Wang2021; Yu et al., Reference Yu, Linn, Shinohara, Oathes, Cook, Duprat and Sheline2019; Zhong et al., Reference Zhong, Shi, Ming, Dong, Zhang, Zeng and Yao2017; Zhu et al., Reference Zhu, Yuan, Zhou, Nie, Wang, Hu and Liao2021). The observed brain structural and functional connectivity changes in NVD group may be trait brain abnormalities of MDD, a detrimental consequence of antidepressant medication, or a combination of both. Of note, we did not find such brain changes in VD group, that is, white matter integrity and functional network connectivity were preserved in MDD patients receiving both antidepressant medication and vitamin D supplementation. This finding suggests that adjuvant vitamin D supplementation therapy might have a protective effect on the brain of MDD patients.

There has been increasing recognition of the neuroprotective and neurotrophic actions of vitamin D. FA represents white matter integrity that is related to many factors including axonal count and density, degree of myelination, and fiber organization (Winston, Reference Winston2012). Changes in FA may reflect multiple white matter micro-structural alterations, such as altered axonal density or diameter, abnormal myelination, or altered coherence of fiber tracts. Vitamin D is a potent neuromodulatory compound that can potentiate axon regeneration; in addition, vitamin D could play a protective role by restricting Wallerian degeneration, i.e. degeneration of the distal part to the injury (Chabas et al., Reference Chabas, Alluin, Rao, Garcia, Lavaut, Risso and Feron2008). There is also empirical evidence that vitamin D can enhance neural stem cell proliferation and oligodendrocyte differentiation (Shirazi, Rasouli, Ciric, Rostami, & Zhang, Reference Shirazi, Rasouli, Ciric, Rostami and Zhang2015). Neurotransmitter dysfunction has been suggested to be involved in the pathophysiology of MDD. For instance, past studies have reported reduced histamine H1 receptor binding (Yanai & Tashiro, Reference Yanai and Tashiro2007), reduced GABA levels (Croarkin et al., Reference Croarkin, Levinson and Daskalakis2011), and reduced muscarinic acetylcholine receptor binding (Nikolaus, Hautzel, Heinzel, & Muller, Reference Nikolaus, Hautzel, Heinzel and Muller2012) in the frontal and occipital cortex in MDD patients. It is generally accepted that vitamin D implicates the synthesis, release, and regulation of neurotransmitters (Kesby et al., Reference Kesby, Turner, Alexander, Eyles, McGrath and Burne2017; Patrick & Ames, Reference Patrick and Ames2015). This has led to some speculation that vitamin D supplements help preserve structural and functional connectivity through their beneficial effects on neurotransmitter systems. In addition, depression is associated with elevated biomarkers of inflammation (e.g. cytokines and C-reactive protein), which have been linked to decreased brain connectivity (Felger et al., Reference Felger, Li, Haroon, Woolwine, Jung, Hu and Miller2016). Intense research indicates that vitamin D, a regulator of key components of the immune system, can inhibit abnormal activation of the immune system and thereby has anti-inflammatory activity (Wei & Christakos, Reference Wei and Christakos2015). Despite this growing evidence, the exact mechanisms underlying the neuroprotective effect of vitamin D in MDD are rather complex and need further investigation.

Some limitations of our work should be noted. First, although our small sample size is common for longitudinal neuroimaging studies, it limits the statistical power and the generalizability of our findings. In the future, a larger sample is needed to validate these preliminary findings. Second, most of the MDD patients were antidepressant-medicated and chronic, which may introduce confounds of antidepressant medication and illness duration. Although our results held even after controlling for antidepressant types and illness duration, future studies in drug-naïve first-episode patients with MDD are warranted to further eliminate these confounding factors. Third, the neuroprotective effect of vitamin D in MDD patients is a preliminary finding that needs to be validated in future animal studies. Fourth, it should be noted that patients with anxiety disorders were excluded. Since anxiety is frequently comorbid with MDD, this reduces the generalizability of the findings to the general population with MDD. Fifth, we focused our analysis on static functional network connectivity rather than dynamic connectivity. Since the brain connectome is time-varying and dynamic in ways that are themselves clinically relevant (Luo et al., Reference Luo, Chen, Wang, Li, He, Li and Li2023a; You et al., Reference You, Luo, Yao, Zhao, Li, Wang and Li2022), examination of dynamic functional network connectivity alterations will be part of our future investigations. Finally, we failed to collect more relevant information about participants' lifestyle profiles. Further analysis of these data may facilitate the interpretation of our findings.

In conclusion, our RCT demonstrated that a greater SCVD increase was associated with greater improvement in depression and anxiety symptoms in MDD patients who received a combined intervention of antidepressants and vitamin D. More importantly, multimodal neuroimaging data showed that vitamin D supplementation was able to preserve structural and functional connectivity between the frontal and occipital cortex in MDD patients. Our results suggest that vitamin D supplementation as adjunctive therapy to antidepressants may not only contribute to improvement in clinical symptoms but also have a protective effect on the brain of MDD patients. More broadly, these findings might hold high translational value in informing future MDD intervention in clinical settings.

Supplementary material

The supplementary material for this article can be found at https://doi.org/10.1017/S0033291724000539.

Data availability statement

Data that support the findings of this study are publicly available in the study's Open Science Framework repository (https://osf.io/j9fc2/).

Acknowledgements

The authors are grateful to study participants for volunteering their time. We greatly appreciate the Chinese psychiatrists and psychologists who have developed the Chinese versions of the HAMD and HAMA.

Funding statement

The study was supported by the STI2030-Major Projects (grant number: 2022ZD0205200), the National Natural Science Foundation of China (grant numbers: 82371928 and 82071905), the Anhui Provincial Natural Science Foundation (grant number: 2308085MH277), the Outstanding Youth Support Project of Anhui Province Universities (grant number: gxyqZD2022026), the Scientific Research Key Project of Anhui Province Universities (grant number: 2022AH051135), the Scientific Research Foundation of Anhui Medical University (grant number: 2022xkj143), the Anhui University Collaborative Innovation Project (grant number: GXXT-2021-065), and the Natural Science Research Project of Anhui Province Universities (grant number: KJ2021ZD0037).

Competing interests

All the authors declare that they have no conflict of interest.

Footnotes

*

These authors contributed equally to this work.

References

Aghajafari, F., Letourneau, N., Mahinpey, N., Cosic, N., & Giesbrecht, G. (2018). Vitamin D deficiency and antenatal and postpartum depression: A systematic review. Nutrients, 10(4), 478. 10.3390/nu10040478.CrossRefGoogle ScholarPubMed
Alavi, N. M., Khademalhoseini, S., Vakili, Z., & Assarian, F. (2019). Effect of vitamin D supplementation on depression in elderly patients: A randomized clinical trial. Clinical Nutrition, 38(5), 20652070. doi:10.1016/j.clnu.2018.09.011CrossRefGoogle ScholarPubMed
Albert, K. M., Potter, G. G., Boyd, B. D., Kang, H., & Taylor, W. D. (2019). Brain network functional connectivity and cognitive performance in major depressive disorder. Journal of Psychiatric Research, 110, 5156. doi:10.1016/j.jpsychires.2018.11.020CrossRefGoogle ScholarPubMed
Alexander, A. L., Lee, J. E., Lazar, M., & Field, A. S. (2007). Diffusion tensor imaging of the brain. Neurotherapeutics: The Journal of the American Society for Experimental NeuroTherapeutics, 4(3), 316329. doi:10.1016/j.nurt.2007.05.011CrossRefGoogle ScholarPubMed
Alghamdi, S., Alsulami, N., Khoja, S., Alsufiani, H., Tayeb, H. O., & Tarazi, F. I. (2020). Vitamin D supplementation ameliorates severity of major depressive disorder. Journal of Molecular Neuroscience: MN, 70(2), 230235. doi:10.1007/s12031-019-01461-2CrossRefGoogle ScholarPubMed
Alroy, I., Towers, T. L., & Freedman, L. P. (1995). Transcriptional repression of the interleukin-2 gene by vitamin D3: Direct inhibition of NFATp/AP-1 complex formation by a nuclear hormone receptor. Molecular and Cellular Biology, 15(10), 57895799. doi:10.1128/mcb.15.10.5789CrossRefGoogle ScholarPubMed
Anglin, R. E., Samaan, Z., Walter, S. D., & McDonald, S. D. (2013). Vitamin D deficiency and depression in adults: Systematic review and meta-analysis. The British Journal of Psychiatry: the Journal of Mental Science, 202, 100107. doi:10.1192/bjp.bp.111.106666CrossRefGoogle ScholarPubMed
Ashburner, J. (2007). A fast diffeomorphic image registration algorithm. NeuroImage, 38(1), 95113. doi:10.1016/j.neuroimage.2007.07.007CrossRefGoogle ScholarPubMed
Berridge, M. J. (2015). Vitamin D cell signalling in health and disease. Biochemical and Biophysical Research Communications, 460(1), 5371. doi:10.1016/j.bbrc.2015.01.008CrossRefGoogle ScholarPubMed
Berridge, M. J. (2017). Vitamin D and depression: Cellular and regulatory mechanisms. Pharmacological reviews, 69(2), 8092. doi:10.1124/pr.116.013227CrossRefGoogle ScholarPubMed
Bersani, F. S., Ghezzi, F., Maraone, A., Vicinanza, R., Cavaggioni, G., Biondi, M., & Pasquini, M. (2019). The relationship between Vitamin D and depressive disorders. Rivista di psichiatria, 54(6), 229234. doi:10.1708/3281.32541Google ScholarPubMed
Beurel, E., Toups, M., & Nemeroff, C. B. (2020). The bidirectional relationship of depression and inflammation: Double trouble. Neuron, 107(2), 234256. doi:10.1016/j.neuron.2020.06.002CrossRefGoogle ScholarPubMed
Briggs, R., McCarroll, K., O'Halloran, A., Healy, M., Kenny, R. A., & Laird, E. (2019). Vitamin D deficiency is associated with an increased likelihood of incident depression in Community-Dwelling older adults. Journal of the American Medical Directors Association, 20(5), 517523. doi:10.1016/j.jamda.2018.10.006CrossRefGoogle ScholarPubMed
Casseb, G. A. S., Kaster, M. P., & Rodrigues, A. L. S. (2019). Potential role of vitamin D for the management of depression and anxiety. CNS drugs, 33(7), 619637. doi:10.1007/s40263-019-00640-4CrossRefGoogle ScholarPubMed
Chabas, J. F., Alluin, O., Rao, G., Garcia, S., Lavaut, M. N., Risso, J. J., … Feron, F. (2008). Vitamin D2 potentiates axon regeneration. Journal of Neurotrauma, 25(10), 12471256. doi:10.1089/neu.2008.0593CrossRefGoogle ScholarPubMed
Chen, G., Guo, Y., Zhu, H., Kuang, W., Bi, F., Ai, H., … Gong, Q. (2017). Intrinsic disruption of white matter microarchitecture in first-episode, drug-naive major depressive disorder: A voxel-based meta-analysis of diffusion tensor imaging. Progress in Neuro-Psychopharmacology & Biological Psychiatry, 76, 179187. doi:10.1016/j.pnpbp.2017.03.011CrossRefGoogle ScholarPubMed
Chen, G., Hu, X., Li, L., Huang, X., Lui, S., Kuang, W., … Gong, Q. (2016). Disorganization of white matter architecture in major depressive disorder: A meta-analysis of diffusion tensor imaging with tract-based spatial statistics. Scientific Reports, 6, 21825. doi:10.1038/srep21825CrossRefGoogle ScholarPubMed
Chen, H., Liu, K., Zhang, B., Zhang, J., Xue, X., Lin, Y., … Deng, Y. (2019). More optimal but less regulated dorsal and ventral visual networks in patients with major depressive disorder. Journal of Psychiatric Research, 110, 172178. doi:10.1016/j.jpsychires.2019.01.005CrossRefGoogle ScholarPubMed
Cheng, Y. C., Huang, Y. C., & Huang, W. L. (2020). The effect of vitamin D supplement on negative emotions: A systematic review and meta-analysis. Depression and Anxiety, 37(6), 549564. doi:10.1002/da.23025CrossRefGoogle ScholarPubMed
Cipriani, A., Furukawa, T. A., Salanti, G., Chaimani, A., Atkinson, L. Z., Ogawa, Y., … Geddes, J. R. (2018). Comparative efficacy and acceptability of 21 antidepressant drugs for the acute treatment of adults with major depressive disorder: A systematic review and network meta-analysis. The Lancet, 391(10128), 13571366. doi:10.1016/s0140-6736(17)32802-7CrossRefGoogle ScholarPubMed
Cipriani, A., Zhou, X., Del Giovane, C., Hetrick, S. E., Qin, B., Whittington, C., … Xie, P. (2016). Comparative efficacy and tolerability of antidepressants for major depressive disorder in children and adolescents: A network meta-analysis. Lancet (London, England), 388(10047), 881890. doi:10.1016/S0140-6736(16)30385-3CrossRefGoogle ScholarPubMed
Cole, M. W., Repovs, G., & Anticevic, A. (2014). The frontoparietal control system: A central role in mental health. The Neuroscientist: a Review Journal Bringing Neurobiology, Neurology and Psychiatry, 20(6), 652664. doi:10.1177/1073858414525995CrossRefGoogle ScholarPubMed
Croarkin, P. E., Levinson, A. J., & Daskalakis, Z. J. (2011). Evidence for GABAergic inhibitory deficits in major depressive disorder. Neuroscience and Biobehavioral Reviews, 35(3), 818825. doi:10.1016/j.neubiorev.2010.10.002CrossRefGoogle ScholarPubMed
de Koning, E. J., van Schoor, N. M., Penninx, B. W., Elders, P. J., Heijboer, A. C., Smit, J. H., … Lips, P. (2015). Vitamin D supplementation to prevent depression and poor physical function in older adults: Study protocol of the D-Vitaal study, a randomized placebo-controlled clinical trial. BMC Geriatrics, 15, 151. doi:10.1186/s12877-015-0148-3CrossRefGoogle ScholarPubMed
de Kwaasteniet, B., Ruhe, E., Caan, M., Rive, M., Olabarriaga, S., Groefsema, M., … Denys, D. (2013). Relation between structural and functional connectivity in major depressive disorder. Biological Psychiatry, 74(1), 4047. doi:10.1016/j.biopsych.2012.12.024CrossRefGoogle ScholarPubMed
Di Somma, C., Scarano, E., Barrea, L., Zhukouskaya, V. V., Savastano, S., Mele, C., … Marzullo, P. (2017). Vitamin D and neurological diseases: An endocrine view. International Journal of Molecular Sciences, 18(11), 2482. 10.3390/ijms18112482.CrossRefGoogle ScholarPubMed
Eyles, D. W., Burne, T. H., & McGrath, J. J. (2013). Vitamin D, effects on brain development, adult brain function and the links between low levels of vitamin D and neuropsychiatric disease. Frontiers in Neuroendocrinology, 34(1), 4764. doi:10.1016/j.yfrne.2012.07.001CrossRefGoogle ScholarPubMed
Eyles, D. W., Smith, S., Kinobe, R., Hewison, M., & McGrath, J. J. (2005). Distribution of the vitamin D receptor and 1 alpha-hydroxylase in human brain. Journal of Chemical Neuroanatomy, 29(1), 2130. doi:10.1016/j.jchemneu.2004.08.006CrossRefGoogle ScholarPubMed
Felger, J. C., Li, Z., Haroon, E., Woolwine, B. J., Jung, M. Y., Hu, X., & Miller, A. H. (2016). Inflammation is associated with decreased functional connectivity within corticostriatal reward circuitry in depression. Molecular Psychiatry, 21(10), 13581365. doi:10.1038/mp.2015.168CrossRefGoogle ScholarPubMed
Fox, M. D., & Raichle, M. E. (2007). Spontaneous fluctuations in brain activity observed with functional magnetic resonance imaging. Nature Reviews. Neuroscience, 8(9), 700711. doi:10.1038/nrn2201CrossRefGoogle ScholarPubMed
Geng, C., Shaikh, A. S., Han, W., Chen, D., Guo, Y., & Jiang, P. (2019). Vitamin D and depression: Mechanisms, determination and application. Asia Pacific Journal of Clinical Nutrition, 28(4), 689694. doi:10.6133/apjcn.201912_28(4).0003Google ScholarPubMed
Gowda, U., Mutowo, M. P., Smith, B. J., Wluka, A. E., & Renzaho, A. M. (2015). Vitamin D supplementation to reduce depression in adults: Meta-analysis of randomized controlled trials. Nutrition (Burbank, Los Angeles County, Calif.), 31(3), 421429. doi:10.1016/j.nut.2014.06.017CrossRefGoogle ScholarPubMed
Guidotti, A., Auta, J., Chen, Y., Davis, J. M., Dong, E., Gavin, D. P., … Tueting, P. (2011). Epigenetic GABAergic targets in schizophrenia and bipolar disorder. Neuropharmacology, 60(7-8), 10071016. doi:10.1016/j.neuropharm.2010.10.021CrossRefGoogle ScholarPubMed
Guo, T., Xiang, Y. T., Xiao, L., Hu, C. Q., Chiu, H. F., Ungvari, G. S., … Wang, G. (2015). Measurement-Based care versus standard care for major depression: A randomized controlled trial with blind raters. The American Journal of Psychiatry, 172(10), 10041013. doi:10.1176/appi.ajp.2015.14050652CrossRefGoogle ScholarPubMed
Hasler, G., van der Veen, J. W., Tumonis, T., Meyers, N., Shen, J., & Drevets, W. C. (2007). Reduced prefrontal glutamate/glutamine and gamma-aminobutyric acid levels in major depression determined using proton magnetic resonance spectroscopy. Archives of General Psychiatry, 64(2), 193200. doi:10.1001/archpsyc.64.2.193CrossRefGoogle ScholarPubMed
Honey, C. J., Kotter, R., Breakspear, M., & Sporns, O. (2007). Network structure of cerebral cortex shapes functional connectivity on multiple time scales. Proceedings of the National Academy of Sciences of the United States of America, 104(24), 1024010245. doi:10.1073/pnas.0701519104CrossRefGoogle ScholarPubMed
Honey, C. J., Sporns, O., Cammoun, L., Gigandet, X., Thiran, J. P., Meuli, R., & Hagmann, P. (2009). Predicting human resting-state functional connectivity from structural connectivity. Proceedings of the National Academy of Sciences of the United States of America, 106(6), 20352040. doi:10.1073/pnas.0811168106CrossRefGoogle ScholarPubMed
Hua, K., Zhang, J., Wakana, S., Jiang, H., Li, X., Reich, D. S., … Mori, S. (2008). Tract probability maps in stereotaxic spaces: Analyses of white matter anatomy and tract-specific quantification. NeuroImage, 39(1), 336347. doi:10.1016/j.neuroimage.2007.07.053CrossRefGoogle ScholarPubMed
Jacka, F. N., Kremer, P. J., Leslie, E. R., Berk, M., Patton, G. C., Toumbourou, J. W., & Williams, J. W. (2010). Associations between diet quality and depressed mood in adolescents: Results from the Australian Healthy Neighbourhoods Study. The Australian and New Zealand Journal of Psychiatry, 44(5), 435442. doi:10.3109/00048670903571598CrossRefGoogle ScholarPubMed
Jacka, F. N., Pasco, J. A., Williams, L. J., Leslie, E. R., Dodd, S., Nicholson, G. C., … Berk, M. (2011). Lower levels of physical activity in childhood associated with adult depression. Journal of Science and Medicine in Sport, 14(3), 222226. doi:10.1016/j.jsams.2010.10.458CrossRefGoogle ScholarPubMed
Jiang, J., Zhao, Y. J., Hu, X. Y., Du, M. Y., Chen, Z. Q., Wu, M., … Gong, Q. Y. (2017). Microstructural brain abnormalities in medication-free patients with major depressive disorder: A systematic review and meta-analysis of diffusion tensor imaging. Journal of Psychiatry & Neuroscience: JPN, 42(3), 150163.CrossRefGoogle ScholarPubMed
Jiao, K., Xu, H., Teng, C., Song, X., Xiao, C., Fox, P. T., … Zhong, Y. (2020). Connectivity patterns of cognitive control network in first episode medication-naive depression and remitted depression. Behavioural Brain Research, 379, 112381. doi:10.1016/j.bbr.2019.112381CrossRefGoogle ScholarPubMed
Ju, S. Y., Lee, Y. J., & Jeong, S. N. (2013). Serum 25-hydroxyvitamin D levels and the risk of depression: A systematic review and meta-analysis. The Journal of Nutrition, Health & Aging, 17(5), 447455. doi:10.1007/s12603-012-0418-0CrossRefGoogle ScholarPubMed
Kaiser, R. H., Andrews-Hanna, J. R., Wager, T. D., & Pizzagalli, D. A. (2015). Large-scale network dysfunction in major depressive disorder: A meta-analysis of resting-state functional connectivity. JAMA Psychiatry, 72(6), 603611. doi:10.1001/jamapsychiatry.2015.0071CrossRefGoogle ScholarPubMed
Kalueff, A. V., Eremin, K. O., & Tuohimaa, P. (2004). Mechanisms of neuroprotective action of vitamin D(3). Biochemistry. Biokhimiia, 69(7), 738741. doi:10.1023/b:biry.0000040196.65686.2fCrossRefGoogle ScholarPubMed
Kaviani, M., Nikooyeh, B., Zand, H., Yaghmaei, P., & Neyestani, T. R. (2020). Effects of vitamin D supplementation on depression and some involved neurotransmitters. Journal of Affective Disorders, 269, 2835. doi:10.1016/j.jad.2020.03.029CrossRefGoogle ScholarPubMed
Kesby, J. P., Turner, K. M., Alexander, S., Eyles, D. W., McGrath, J. J., & Burne, T. H. J. (2017). Developmental vitamin D deficiency alters multiple neurotransmitter systems in the neonatal rat brain. International Journal of Developmental Neuroscience, 62, 17. doi:10.1016/j.ijdevneu.2017.07.002CrossRefGoogle ScholarPubMed
Khoraminya, N., Tehrani-Doost, M., Jazayeri, S., Hosseini, A., & Djazayery, A. (2013). Therapeutic effects of vitamin D as adjunctive therapy to fluoxetine in patients with major depressive disorder. The Australian and New Zealand Journal of Psychiatry, 47(3), 271275. doi:10.1177/0004867412465022CrossRefGoogle ScholarPubMed
Kongsbak, M., Levring, T. B., Geisler, C., & von Essen, M. R. (2013). The vitamin D receptor and T cell function. Frontiers in Immunology, 4, 148. doi:10.3389/fimmu.2013.00148CrossRefGoogle ScholarPubMed
Lai, W. T., Deng, W. F., Xu, S. X., Zhao, J., Xu, D., Liu, Y. H., … Rong, H. (2021). Shotgun metagenomics reveals both taxonomic and tryptophan pathway differences of gut microbiota in major depressive disorder patients. Psychological Medicine, 51(1), 90101. doi:10.1017/S0033291719003027CrossRefGoogle ScholarPubMed
Leemans, A., & Jones, D. K. (2009). The B-matrix must be rotated when correcting for subject motion in DTI data. Magnetic Resonance in Medicine, 61(6), 13361349. doi:10.1002/mrm.21890CrossRefGoogle ScholarPubMed
Li, H., Lin, X., Liu, L., Su, S., Zhu, X., Zheng, Y., … Sun, X. (2020). Disruption of the structural and functional connectivity of the frontoparietal network underlies symptomatic anxiety in late-life depression. NeuroImage: Clinical, 28, 102398. doi:10.1016/j.nicl.2020.102398CrossRefGoogle ScholarPubMed
Libuda, L., Antel, J., Hebebrand, J., & Focker, M. (2017). [Nutrition and mental diseases: Focus depressive disorders]. Der Nervenarzt, 88(1), 87101. doi:10.1007/s00115-016-0262-2CrossRefGoogle ScholarPubMed
Liu, G., Jiao, K., Zhong, Y., Hao, Z., Wang, C., Xu, H., … Zhang, N. (2021). The alteration of cognitive function networks in remitted patients with major depressive disorder: An independent component analysis. Behavioural Brain Research, 400, 113018. doi:10.1016/j.bbr.2020.113018CrossRefGoogle ScholarPubMed
Liu, J., Xu, P., Zhang, J., Jiang, N., Li, X., & Luo, Y. (2019). Ventral attention-network effective connectivity predicts individual differences in adolescent depression. Journal of Affective Disorders, 252, 5559. doi:10.1016/j.jad.2019.04.033CrossRefGoogle ScholarPubMed
Liu, Y., Chen, Y., Liang, X., Li, D., Zheng, Y., Zhang, H., … Qiu, S. (2020). Altered resting-state functional connectivity of multiple networks and disrupted correlation with executive function in major depressive disorder. Frontiers in Neurology, 11, 272. doi:10.3389/fneur.2020.00272CrossRefGoogle ScholarPubMed
Lopresti, A. L., Hood, S. D., & Drummond, P. D. (2013). A review of lifestyle factors that contribute to important pathways associated with major depression: Diet, sleep and exercise. Journal of Affective Disorders, 148(1), 1227. doi:10.1016/j.jad.2013.01.014iCrossRefGoogle ScholarPubMed
Luo, L., Chen, L., Wang, Y., Li, Q., He, N., Li, Y., … Li, F. (2023a). Patterns of brain dynamic functional connectivity are linked with attention-deficit/hyperactivity disorder-related behavioral and cognitive dimensions. Psychological Medicine, 53(14), 112. doi:10.1017/S0033291723000089CrossRefGoogle ScholarPubMed
Luo, L., Li, Q., Wang, Y., He, N., Wang, Y., You, W., … Li, F. (2023b). Shared and disorder-specific alterations of brain temporal dynamics in obsessive-compulsive disorder and schizophrenia. Schizophrenia Bulletin, 49(5), 13871398. doi:10.1093/schbul/sbad042CrossRefGoogle Scholar
Luo, L., Wu, H., Xu, J., Chen, F., Wu, F., Wang, C., & Wang, J. (2021). Abnormal large-scale resting-state functional networks in drug-free major depressive disorder. Brain Imaging and Behavior, 15(1), 96106. doi:10.1007/s11682-019-00236-yCrossRefGoogle ScholarPubMed
Luo, L., You, W., DelBello, M. P., Gong, Q., & Li, F. (2022). Recent advances in psychoradiology. Physics in Medicine and Biology, 67(23). doi:10.1088/1361-6560/ac9d1eCrossRefGoogle ScholarPubMed
McCann, J. C., & Ames, B. N. (2008). Is there convincing biological or behavioral evidence linking vitamin D deficiency to brain dysfunction? FASEB Journal: Official Publication of the Federation of American Societies for Experimental Biology, 22(4), 9821001. doi:10.1096/fj.07-9326revCrossRefGoogle ScholarPubMed
Milaneschi, Y., Hoogendijk, W., Lips, P., Heijboer, A. C., Schoevers, R., van Hemert, A. M., … Penninx, B. W. (2014). The association between low vitamin D and depressive disorders. Molecular Psychiatry, 19(4), 444451. doi:10.1038/mp.2013.36CrossRefGoogle ScholarPubMed
Nikolaus, S., Hautzel, H., Heinzel, A., & Muller, H. W. (2012). Key players in major and bipolar depression – A retrospective analysis of in vivo imaging studies. Behavioural Brain Research, 232(2), 358390. doi:10.1016/j.bbr.2012.03.021CrossRefGoogle ScholarPubMed
Okereke, O. I., Reynolds, C. F. 3rd, Mischoulon, D., Chang, G., Vyas, C. M., Cook, N. R., … Manson, J. E. (2020). Effect of long-term vitamin D3 supplementation vs placebo on risk of depression or clinically relevant depressive symptoms and on change in mood scores: A randomized clinical trial. Jama, 324(5), 471480. doi:10.1001/jama.2020.10224CrossRefGoogle ScholarPubMed
Okereke, O. I., & Singh, A. (2016). The role of vitamin D in the prevention of late-life depression. Journal of Affective Disorders, 198, 114. doi:10.1016/j.jad.2016.03.022CrossRefGoogle ScholarPubMed
Oldfield, R. C. (1971). The assessment and analysis of handedness: The Edinburgh inventory. Neuropsychologia, 9(1), 97113. doi:10.1016/0028-3932(71)90067-4CrossRefGoogle ScholarPubMed
Opie, R. S., Itsiopoulos, C., Parletta, N., Sanchez-Villegas, A., Akbaraly, T. N., Ruusunen, A., & Jacka, F. N. (2017). Dietary recommendations for the prevention of depression. Nutritional Neuroscience, 20(3), 161171. doi:10.1179/1476830515Y.0000000043CrossRefGoogle ScholarPubMed
Parker, G. B., Brotchie, H., & Graham, R. K. (2017). Vitamin D and depression. Journal of Affective Disorders, 208, 5661. doi:10.1016/j.jad.2016.08.082CrossRefGoogle ScholarPubMed
Patrick, R. P., & Ames, B. N. (2015). Vitamin D and the omega-3 fatty acids control serotonin synthesis and action, part 2: Relevance for ADHD, bipolar disorder, schizophrenia, and impulsive behavior. FASEB Journal: Official Publication of the Federation of American Societies for Experimental Biology, 29(6), 22072222. doi:10.1096/fj.14-268342CrossRefGoogle ScholarPubMed
Pike, J. W., Meyer, M. B., Martowicz, M. L., Bishop, K. A., Lee, S. M., Nerenz, R. D., & Goetsch, P. D. (2010). Emerging regulatory paradigms for control of gene expression by 1,25-dihydroxyvitamin D3. The Journal of Steroid Biochemistry and Molecular Biology, 121(1-2), 130135. doi:10.1016/j.jsbmb.2010.02.036CrossRefGoogle Scholar
Ringe, J. D., & Kipshoven, C. (2012). Vitamin D-insufficiency: An estimate of the situation in Germany. Dermato-endocrinology, 4(1), 7280. doi:10.4161/derm.19829CrossRefGoogle ScholarPubMed
Ryan, J. W., Anderson, P. H., & Morris, H. A. (2015). Pleiotropic activities of vitamin D receptors – adequate activation for multiple health outcomes. The Clinical Biochemist. Reviews, 36(2), 5361.Google ScholarPubMed
Saavedra, K., Molina-Marquez, A. M., Saavedra, N., Zambrano, T., & Salazar, L. A. (2016). Epigenetic modifications of major depressive disorder. International Journal of Molecular Sciences, 17(8), 1279. 10.3390/ijms17081279.CrossRefGoogle ScholarPubMed
Sacchet, M. D., Ho, T. C., Connolly, C. G., Tymofiyeva, O., Lewinn, K. Z., Han, L. K., … Yang, T. T. (2016). Large-scale hypoconnectivity between resting-state functional networks in unmedicated adolescent major depressive disorder. Neuropsychopharmacology: Official Publication of the American College of Neuropsychopharmacology, 41(12), 29512960. doi:10.1038/npp.2016.76CrossRefGoogle ScholarPubMed
Sarris, J., O'Neil, A., Coulson, C. E., Schweitzer, I., & Berk, M. (2014). Lifestyle medicine for depression. BMC Psychiatry, 14, 107. doi:10.1186/1471-244X-14-107CrossRefGoogle ScholarPubMed
Schlogl, M., & Holick, M. F. (2014). Vitamin D and neurocognitive function. Clinical Interventions in Aging, 9, 559568. doi:10.2147/CIA.S51785Google ScholarPubMed
Shen, Y., Wei, Y., Yang, X. N., Zhang, G., Du, X., Jia, Q., … Zhang, X. Y. (2020). Psychotic symptoms in first-episode and drug naive patients with major depressive disorder: Prevalence and related clinical factors. Depression and Anxiety, 37(8), 793800. doi:10.1002/da.23026CrossRefGoogle ScholarPubMed
Shirazi, H. A., Rasouli, J., Ciric, B., Rostami, A., & Zhang, G. X. (2015). 1,25-Dihydroxyvitamin D3 enhances neural stem cell proliferation and oligodendrocyte differentiation. Experimental and Molecular Pathology, 98(2), 240245. doi:10.1016/j.yexmp.2015.02.004CrossRefGoogle ScholarPubMed
Smith, S. M., Jenkinson, M., Johansen-Berg, H., Rueckert, D., Nichols, T. E., Mackay, C. E., … Behrens, T. E. (2006). Tract-based spatial statistics: Voxelwise analysis of multi-subject diffusion data. NeuroImage, 31(4), 14871505. doi:10.1016/j.neuroimage.2006.02.024CrossRefGoogle ScholarPubMed
Sun, J., Kong, J., Duan, Y., Szeto, F. L., Liao, A., Madara, J. L., & Li, Y. C. (2006). Increased NF-kappaB activity in fibroblasts lacking the vitamin D receptor. American Journal of Physiology. Endocrinology and Metabolism, 291(2), E315E322. doi:10.1152/ajpendo.00590.2005CrossRefGoogle ScholarPubMed
Thompson, E. (2015). Hamilton rating scale for anxiety (HAM-A). Occupational Medicine (Lond), 65(7), 601. doi:10.1093/occmed/kqv054CrossRefGoogle ScholarPubMed
Tong, P., Bo, P., Shi, Y., Dong, L., Sun, T., Gao, X., & Yang, Y. (2021). Clinical traits of patients with major depressive disorder with comorbid borderline personality disorder based on propensity score matching. Depression and Anxiety, 38(1), 100106. doi:10.1002/da.23122CrossRefGoogle ScholarPubMed
Wei, R., & Christakos, S. (2015). Mechanisms underlying the regulation of innate and adaptive immunity by vitamin D. Nutrients, 7(10), 82518260. doi:10.3390/nu7105392CrossRefGoogle ScholarPubMed
WHO (2017). Depression and other common mental disorders: Global health estimates. Geneva: World Health Organization.Google Scholar
Williams, J. B. (1988). A structured interview guide for the Hamilton Depression Rating Scale. Archives of General Psychiatry, 45(8), 742747. doi:10.1001/archpsyc.1988.01800320058007.CrossRefGoogle ScholarPubMed
Winston, G. P. (2012). The physical and biological basis of quantitative parameters derived from diffusion MRI. Quantitative Imaging in Medicine and Surgery, 2(4), 254265. doi:10.3978/j.issn.2223-4292.2012.12.05Google ScholarPubMed
Wong, S. K., Chin, K. Y., & Ima-Nirwana, S. (2018). Vitamin D and depression: The evidence from an indirect clue to treatment strategy. Current Drug Targets, 19(8), 888897. doi:10.2174/1389450118666170913161030CrossRefGoogle ScholarPubMed
Wu, X. J., Zeng, L. L., Shen, H., Yuan, L., Qin, J., Zhang, P., & Hu, D. (2017). Functional network connectivity alterations in schizophrenia and depression. Psychiatry Res Neuroimaging, 263, 113120. doi:10.1016/j.pscychresns.2017.03.012CrossRefGoogle ScholarPubMed
Yan, C. G., Wang, X. D., Zuo, X. N., & Zang, Y. F. (2016). DPABI: Data processing & analysis for (Resting-State) brain imaging. Neuroinformatics, 14(3), 339351. doi:10.1007/s12021-016-9299-4CrossRefGoogle ScholarPubMed
Yanai, K., & Tashiro, M. (2007). The physiological and pathophysiological roles of neuronal histamine: An insight from human positron emission tomography studies. Pharmacology & Therapeutics, 113(1), 115. doi:10.1016/j.pharmthera.2006.06.008CrossRefGoogle ScholarPubMed
Yang, Y., Zhu, D. M., Zhang, C., Zhang, Y., Wang, C., Zhang, B., … Yu, Y. (2020). Brain structural and functional alterations specific to low sleep efficiency in major depressive disorder. Frontiers in Neuroscience, 14, 50. doi:10.3389/fnins.2020.00050CrossRefGoogle ScholarPubMed
Yin, Y., He, X., Xu, M., Hou, Z., Song, X., Sui, Y., … Yuan, Y. (2016). Structural and functional connectivity of default mode network underlying the cognitive impairment in late-onset depression. Scientific Reports, 6, 37617. doi:10.1038/srep37617CrossRefGoogle ScholarPubMed
You, W., Luo, L., Yao, L., Zhao, Y., Li, Q., Wang, Y., … Li, F. (2022). Impaired dynamic functional brain properties and their relationship to symptoms in never treated first-episode patients with schizophrenia. Schizophrenia (Heidelb), 8(1), 90. doi:10.1038/s41537-022-00299-9CrossRefGoogle ScholarPubMed
Yu, M., Linn, K. A., Shinohara, R. T., Oathes, D. J., Cook, P. A., Duprat, R., … Sheline, Y. I. (2019). Childhood trauma history is linked to abnormal brain connectivity in major depression. Proceedings of the National Academy of Sciences of the United States of America, 116(17), 85828590. doi:10.1073/pnas.1900801116CrossRefGoogle ScholarPubMed
Zhang, C., Yang, Y., Zhu, D. M., Zhao, W., Zhang, Y., Zhang, B., … Yu, Y. (2020). Neural correlates of the association between depression and high density lipoprotein cholesterol change. Journal of Psychiatric Research, 130, 918. doi:10.1016/j.jpsychires.2020.07.012CrossRefGoogle ScholarPubMed
Zhao, W., Zhu, D. M., Li, Q., Xu, X., Zhang, Y., Zhang, C., … Yu, Y. (2023). Brain function mediates the association between low vitamin D and neurocognitive status in female patients with major depressive disorder. Psychological Medicine, 53(9), 40324045. doi:10.1017/S0033291722000708CrossRefGoogle ScholarPubMed
Zhao, W., Zhu, D. M., Li, S., Cui, S., Jiang, P., Wang, R., … Yu, Y. (2022). The reduction of vitamin D in females with major depressive disorder is associated with worse cognition mediated by abnormal brain functional connectivity. Progress in Neuro-Psychopharmacology & Biological Psychiatry, 118, 110577. doi:10.1016/j.pnpbp.2022.110577CrossRefGoogle ScholarPubMed
Zheng, Y. P., Zhao, J. P., Phillips, M., Liu, J. B., Cai, M. F., Sun, S. Q., & Huang, M. F. (1988). Validity and reliability of the Chinese Hamilton depression rating scale. The British Journal of Psychiatry: the Journal of Mental Science, 152, 660664. doi:10.1192/bjp.152.5.660CrossRefGoogle ScholarPubMed
Zhong, X., Shi, H., Ming, Q., Dong, D., Zhang, X., Zeng, L. L., & Yao, S. (2017). Whole-brain resting-state functional connectivity identified major depressive disorder: A multivariate pattern analysis in two independent samples. Journal of Affective Disorders, 218, 346352. doi:10.1016/j.jad.2017.04.040CrossRefGoogle ScholarPubMed
Zhu, D. M., Zhao, W., Cui, S., Jiang, P., Zhang, Y., Zhang, C., … Yu, Y. (2022). The relationship between vitamin D, clinical manifestations, and functional network connectivity in female patients with major depressive disorder. Frontiers in Aging Neuroscience, 14, 817607. doi:10.3389/fnagi.2022.817607CrossRefGoogle ScholarPubMed
Zhu, D.-m., Zhao, W., Zhang, B., Zhang, Y., Yang, Y., Zhang, C., … Yu, Y. (2019). The relationship between serum concentration of vitamin D, total intracranial volume, and severity of depressive symptoms in patients with major depressive disorder. Frontiers in Psychiatry, 10, 322. 10.3389/fpsyt.2019.00322.CrossRefGoogle ScholarPubMed
Zhu, X., Yuan, F., Zhou, G., Nie, J., Wang, D., Hu, P., … Liao, W. (2021). Cross-network interaction for diagnosis of major depressive disorder based on resting state functional connectivity. Brain Imaging and Behavior, 15(3), 12791289. doi:10.1007/s11682-020-00326-2CrossRefGoogle ScholarPubMed
Figure 0

Figure 1. Flowchart of the study design. Abbreviations: HC, healthy controls; MDD, major depressive disorder; MRI, magnetic resonance imaging; SCVD, serum concentration of vitamin D; VD, vitamin D supplementation; NVD, no vitamin D supplementation; DTI, diffusion tensor imaging; fMRI, functional magnetic resonance imaging.

Figure 1

Table 1. Demographic and clinical characteristics of the participants at baseline

Figure 2

Figure 2. SCVD at baseline and follow-up. Abbreviations: HC, healthy controls; VD, vitamin D supplementation; NVD, no vitamin D supplementation; SCVD, serum concentration of vitamin D.

Figure 3

Figure 3. Longitudinal changes in clinical symptoms and their associations with SCVD change. Changes in HAMD (a) and their associations with SCVD change in VD group (b). Changes in HAMA (c) and their associations with SCVD change in VD group (d). Abbreviations: HAMD, Hamilton Rating Scale for Depression; HAMA, Hamilton Rating Scale for Anxiety; SCVD, serum concentration of vitamin D; VD, vitamin D supplementation; NVD, no vitamin D supplementation.

Figure 4

Figure 4. FA of right IFOF at baseline and follow-up. Abbreviations: FA, fractional anisotropy; IFOF, inferior fronto-occipital fasciculus; HC, healthy controls; VD, vitamin D supplementation; NVD, no vitamin D supplementation; R, right.

Figure 5

Figure 5. Functional connectivity between rFPN and mVN at baseline and follow-up. Abbreviations: HC, healthy controls; VD, vitamin D supplementation; NVD, no vitamin D supplementation; rFPN, right frontoparietal network; mVN, medial visual network.

Supplementary material: File

Zhao et al. supplementary material

Zhao et al. supplementary material
Download Zhao et al. supplementary material(File)
File 1.3 MB