Elsevier

Neuroscience

Volume 371, 10 February 2018, Pages 207-220
Neuroscience

Integrated Metabolomics and Proteomics Analysis of Hippocampus in a Rat Model of Depression

https://doi.org/10.1016/j.neuroscience.2017.12.001Get rights and content

Highlights

  • Differential analysis identified 30 metabolites and 170 proteins in the hippocampus of CUMS rats.

  • Impairment in amino acid metabolism and protein synthesis/degradation were involved in hippocampal response to CUMS.

  • Dysregulation of glutamate and glycine metabolism and their transport/catabolism related proteins were implicated.

  • Disturbances in fatty acid and glycerophospholipid metabolism linked to the alterations in related enzymes were found.

  • Differentially expressed synapse-associated proteins were identified.

Abstract

Major depressive disorder (MDD) is a prevalent and serious mental disorder with high rates of suicide and disability. However, the underlying pathogenesis of MDD is complicated and remains largely unclear. An integrated analysis of multiple types of omics data may improve comprehensive understanding of the entire molecular mechanism of MDD. In this study, we applied an integrated analysis of gas chromatography/mass spectrometry (GC–MS)-based metabolomics and isobaric tags for relative and absolute quantitation (iTRAQ)-based proteomics to investigate changes in the hippocampus in the chronic unpredictable mild stress (CUMS) rat model of depression. Only the stress-susceptible rats in the CUMS group were selected for profiling against controls. Differential analysis identified 30 metabolites and 170 proteins between the two groups. The integrated analyses revealed four major changes in the hippocampus of CUMS rats: (1) impairment in amino acid metabolism and protein synthesis/degradation; (2) dysregulation of glutamate and glycine metabolism and their transport/catabolism related proteins; (3) disturbances in fatty acid and glycerophospholipid metabolism accompanied by alterations in the corresponding metabolic enzymes; (4) abnormal expression of synapse-associated proteins. These results provide further important insights into the pathophysiology of depression and may help identify potential targets for antidepressant drugs.

Introduction

Major depressive disorder (MDD) is one of the most common mental disorders, with a lifetime prevalence of 16.2% (Kupfer et al., 2012). MDD can be long-lasting or recurrent, substantially impairing quality of life and functioning (Saarni et al., 2007). However, MDD is a multifactorial disorder with substantial molecular alterations and pathway dysregulations involved, which makes the pathophysiology of MDD complicated and largely unknown. Animal models are essential tools for molecular studies of depression. Among these, the chronic unpredictable mild stress (CUMS) rat model, which mimics the variable and unpredictable stressors encountered in human daily life, is probably the most valid and commonly used model of depression (Yan et al., 2010).

As a critical brain region for memory and mood regulation, the hippocampus plays important roles in the pathogenesis of MDD (Campbell and Macqueen, 2004). A reduction of hippocampal volume, indicated by magnetic resonance imaging, has been widely reported in MDD patients (Campbell et al., 2004). Neuropathological studies have shown decreased neuronal and glial size, decreased expression of synaptic markers, loss of dendrites, reduced neurogenesis and increased apoptosis in the hippocampus of animals in models of depression (Harrison, 2002, Pittenger and Duman, 2008, Lucassen et al., 2006). In addition, because of its regulatory effects on the hypothalamus–pituitary–adrenal (HPA) axis, the hippocampus plays an important role in stress responses, which makes it susceptible in stress-related psychiatric disorders (Liu et al., 2017). Thus, this region of brain is the ideal material to investigate the molecular mechanisms of MDD.

In the past decade, omics technologies have been widely applied as useful tools for molecular profiling, identification of biomarkers, characterization of complex biochemical systems, and for examination of pathophysiological processes in various diseases. Our team has previously completed a series of research studies on depression in MDD patients (Zheng et al., 2013a, Zheng et al., 2013b, Zheng et al., 2012, Xu et al., 2012) and animal models (Mu et al., 2007, Yang et al., 2013, Chen et al., 2015, Liu et al., 2016, Zhou et al., 2017) using omics technologies. With respect to the hippocampus, dysfunction of energy metabolism, neurogenesis, synaptic plasticity, and neurotransmission has been found in previous proteomic studies of postmortem MDD patients and in rodent models of depression (Martins-de-Souza et al., 2012a, Martins-de-Souza et al., 2012b, Mu et al., 2007, Henningsen et al., 2012, Han et al., 2015).

However, the biological interpretation of data from a single type of omics study can be a great challenge due to complex biochemical regulation at multiple levels. Thus, integrated analysis by combining multiple types of omics data is promising and may help to identify potential biological relationships and improve understanding of entire biological mechanisms (Cavill et al., 2016). Among these, metabolomics–proteomics is a powerful combination and frequently applied in pathophysiological research, including in psychiatric disorders such as anxiety (Filiou et al., 2011, Zhang et al., 2011) and schizophrenia (Wesseling et al., 2013, Wesseling et al., 2015). In research on depression, our previous study implemented integrated analysis of metabolomic and proteomic profiling, which found significantly perturbed energy metabolism at the level of both the metabolome and the proteome in the cerebellum of chronically stressed rats (Shao et al., 2015).

In this study, we aim to gain further insights into the molecular mechanisms of depression in rat hippocampus. A well-established rat depression model, CUMS, was applied. Isobaric tags for relative and absolute quantitation (iTRAQ)-based proteomic and gas chromatography–mass spectrometry (GC–MS)-based metabolomic approaches were employed to obtain unbiased profiling data. Moreover, Ingenuity Pathway Analysis (IPA) and Integrated Molecular Pathway Level Analysis (IMPaLA) were used for integrated molecular pathway and network analyses. The results will help to enhance comprehensive understanding of the pathogenesis of MDD.

Section snippets

Animals and ethics statement

Thirty-five healthy male Sprague–Dawley rats with initial weights of 200–250 g were selected in the animal facility at Chongqing Medical University (Chongqing, China). All rats were housed in individual cages under a reversed 12-h light/12-h dark cycle (lights on at 19:00 h) and standard laboratory conditions (21 ± 1 °C, 55 ± 5% relative humidity). Food and water were provided ad libitum. This study was approved by the Ethics Committee of Chongqing Medical University. All animal treatments were

Evaluation of the CUMS rat model

To eliminate extremes in innate sensitivity to sucrose water and activity or inactivity in the open field, we excluded rats with baseline values beyond the 95% reference interval in the LAT and the 95% percentile interval in the SPT (Liu et al., 2016). Five rats were excluded from the LAT and two from the SPT, leaving 28 rats (20 CUMS versus 8 CON) for the subsequent experiments. After stress exposure, 9 out of 20 rats from the CUMS group were selected as a susceptible subgroup because their

Discussion

In this study, we established a CUMS rat model to investigate the pathophysiological mechanisms underlying MDD. The baseline LAT and SPT results were used to screen out rats with a congenital abnormality in sucrose preference or activity. After the CUMS paradigm, only the stress-susceptible rats were selected for subsequent analyses. This approach helps reduce the heterogeneity of rats and enhance the specificity of identified biomarkers. Consequently, significant depression-like and

Conclusion

In this study, we applied an integrated analysis of GC–MS-based metabolomics and iTRAQ-based proteomics to obtain a comprehensive picture of the rat hippocampal response to CUMS. Our results found some consistent alterations among metabolomics and proteomics in CUMS rats, including the dysregulated amino acid metabolism, lipid metabolism, and synaptic transmission. These findings provide further insights into the pathogenesis of depression and may help identify potential targets for

Acknowledgements

This work was supported by the National Key Research and Development Programm of China (Grant no. 2017YFA0505700), the National Basic Research Program of China (973 Program, Grant no. 2009CB918300), and the National Natural Science Foundation of China (Grant no. 81701342). P.X. and Y.Z. conceived the study; Y.Z., S.Y., J.P. and L.Y. conducted the experiments; Y.Z., X.Z. and L.L. analyzed and interpreted the data; Y.Z. drafted the manuscript; P.X. and X.Z. revised the manuscript. X.J., H.Z.,

Conflicts of interest

None.

References (64)

  • L. Liu et al.

    The identification of metabolic disturbances in the prefrontal cortex of the chronic restraint stress rat model of depression

    Behav Brain Res

    (2016)
  • Z. Lu et al.

    Calsyntenin-3 molecular architecture and interaction with neurexin 1α

    J Biol Chem

    (2014)
  • A. Medina et al.

    Glutamate transporters: a key piece in the glutamate puzzle of major depressive disorder

    J Psychiatr Res

    (2013)
  • J. Mu et al.

    Neurogenesis and major depression: implications from proteomic analyses of hippocampal proteins in a rat depression model

    Neurosci Lett

    (2007)
  • Y. Ni et al.

    Metabolic profiling reveals disorder of amino acid metabolism in four brain regions from a rat model of chronic unpredictable mild stress

    FEBS Lett

    (2008)
  • A. Pantzar et al.

    Interactive effects of KIBRA and CLSTN2 polymorphisms on episodic memory in old-age unipolar depression

    Neuropsychologia

    (2014)
  • S.V. Ranneva et al.

    Features of emotional and social behavioral phenotypes of calsyntenin2 knockout mice

    Behav Brain Res

    (2017)
  • Y. Ren et al.

    Hypoxia modulates A431 cellular pathways association to tumor radioresistance and enhanced migration revealed by comprehensive proteomic and functional studies

    Mol Cell Proteomics

    (2013)
  • M. Rima et al.

    The β4 subunit of the voltage-gated calcium channel (Cacnb4) regulates the rate of cell proliferation in Chinese Hamster Ovary cells

    Int J Biochem Cell Biol

    (2017)
  • G. Sanacora et al.

    Towards a glutamate hypothesis of depression: an emerging frontier of neuropsychopharmacology for mood disorders

    Neuropharmacology

    (2012)
  • R.M. Tordera et al.

    Chronic stress and impaired glutamate function elicit a depressive-like phenotype and common changes in gene expression in the mouse frontal cortex

    Eur Neuropsychopharmacol

    (2011)
  • A. Vluggens et al.

    Functional significance of the two ACOX1 isoforms and their crosstalks with PPARα and RXRα

    Lab Invest

    (2010)
  • C. Wersinger et al.

    Partial regulation of serotonin transporter function by gamma-synuclein

    Neurosci Lett

    (2009)
  • Y. Yang et al.

    Proteomics reveals energy and glutathione metabolic dysregulation in the prefrontal cortex of a rat model of depression

    Neuroscience

    (2013)
  • P. Zheng et al.

    Identification and validation of urinary metabolite biomarkers for major depressive disorder

    Mol Cell Proteomics

    (2013)
  • X. Zhou et al.

    Metabolomics identifies perturbations in amino acid metabolism in the prefrontal cortex of the learned helplessness rat model of depression

    Neuroscience

    (2017)
  • X. Zhu et al.

    Sub-anesthetic doses of ketamine exert antidepressant-like effects and upregulate the expression of glutamate transporters in the hippocampus of rats

    Neurosci Lett

    (2017)
  • M. Zink et al.

    Reduced expression of glutamate transporters vGluT1, EAAT2 and EAAT4 in learned helpless rats, an animal model of depression

    Neuropharmacology

    (2010)
  • S. Campbell et al.

    The role of the hippocampus in the pathophysiology of major depression

    J Psychiatry Neurosci

    (2004)
  • S. Campbell et al.

    Lower hippocampal volume in patients suffering from depression: a meta-analysis

    Am J Psychiatry

    (2004)
  • R. Cavill et al.

    Transcriptomic and metabolomic data integration

    Brief Bioinform

    (2016)
  • J.D. Clark et al.

    The 1996 guide for the care and use of laboratory animals

    ILAR J

    (1997)
  • Cited by (118)

    • Progress of depression mechanism based on Omics method

      2024, Journal of Pharmaceutical and Biomedical Analysis
    View all citing articles on Scopus

    These authors contributed equally to this work.

    View full text