Activation of dopaminergic VTA inputs to the mPFC ameliorates chronic stress-induced breast tumor progression

CNS Neurosci Ther. 2021 Feb;27(2):206-219. doi: 10.1111/cns.13465. Epub 2020 Oct 28.

Abstract

Aims: Chronic stress plays an important role in promoting the progression and migration of cancers. However, little is known of any direct impact on tumor progression related to the regulation of emotion-related circuitry. The aim of this study was to explore the neural-circuit mechanisms underlying stress-induced progression of cancers and the impact of emotion-related regulation of circuitry on tumor growth.

Methods: Optogenetic manipulation was applied to unpredictable chronic mild stress (UCMS)-treated mice bearing breast tumor cell. The stress-related hormones, tumor-related cytokines, the tyrosine hydroxylase (TH)-positive neurons and their fibers, dopamine receptor-positive cells, and anxiety level were measured using ELISA, immunohistochemical staining, fluorescence in situ hybridization, and behavioral test, respectively.

Results: By investigating breast cancer mouse models with a chronic mild stress model, optogenetic stimulation, and behavioral analysis, we show that chronic stress induced anxiety-like behavior in mice and increased serum concentration of norepinephrine and corticosterone, hormones closely related to stress and anxiety. Optogenetic activation of VTA TH terminals in the mPFC rescued anxiety-like behavior induced by chronic stress. Chronic stress resulted in marked progression of breast tumors, and repetitive optogenetic activation of VTA TH terminals in the mPFC significantly attenuated stress-induced progression of breast cancers and reduced serum concentration of norepinephrine and corticosterone. Furthermore, there was a positive correlation between serum norepinephrine or corticosterone concentration and tumor size.

Conclusions: These findings indicate a positive role of an emotion regulation circuit on the progression of breast cancer and reveal a link between stress, emotion regulation, and the progression of breast cancers. Our findings provide new insights pertinent to therapeutic interventions in the treatment of breast cancers.

Keywords: cancer; emotion; neuromodulation; neurotransmitter; optogenetic; unpredictable mild stress.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Breast Neoplasms / metabolism*
  • Breast Neoplasms / pathology
  • Breast Neoplasms / psychology
  • Chronic Disease
  • Disease Progression*
  • Dopaminergic Neurons / chemistry
  • Dopaminergic Neurons / metabolism*
  • Female
  • Humans
  • MCF-7 Cells
  • Male
  • Mice
  • Mice, Inbred BALB C
  • Mice, Inbred C57BL
  • Mice, Nude
  • Optogenetics / methods
  • Prefrontal Cortex / chemistry
  • Prefrontal Cortex / metabolism*
  • Stress, Psychological / metabolism*
  • Stress, Psychological / pathology
  • Stress, Psychological / psychology
  • Ventral Tegmental Area / chemistry
  • Ventral Tegmental Area / metabolism*
  • Xenograft Model Antitumor Assays / methods