Unknown

Dataset Information

0

Regulation of morphine-induced synaptic alterations: Role of oxidative stress, ER stress, and autophagy.


ABSTRACT: Our findings suggest that morphine dysregulates synaptic balance in the hippocampus, a key center for learning and memory, via a novel signaling pathway involving reactive oxygen species (ROS), endoplasmic reticulum (ER) stress, and autophagy. We demonstrate in this study that exposure of morphine to hippocampal neurons leads to a reduction in excitatory synapse densities with a concomitant enhancement of inhibitory synapse densities via activation of the ? opioid receptor. Furthermore, these effects of morphine are mediated by up-regulation of intracellular ROS from NADPH oxidase, leading, in turn, to sequential induction of ER stress and autophagy. The detrimental effects of morphine on synaptic densities were shown to be reversed by platelet-derived growth factor (PDGF), a pleiotropic growth factor that has been implicated in neuroprotection. These results identify a novel cellular mechanism involved in morphine-mediated synaptic alterations with implications for therapeutic interventions by PDGF.

SUBMITTER: Cai Y 

PROVIDER: S-EPMC5084649 | biostudies-literature | 2016 Oct

REPOSITORIES: biostudies-literature

altmetric image

Publications

Regulation of morphine-induced synaptic alterations: Role of oxidative stress, ER stress, and autophagy.

Cai Yu Y   Yang Lu L   Hu Guoku G   Chen Xufeng X   Niu Fang F   Yuan Li L   Liu Han H   Xiong Huangui H   Arikkath Jyothi J   Buch Shilpa S  

The Journal of cell biology 20161017 2


Our findings suggest that morphine dysregulates synaptic balance in the hippocampus, a key center for learning and memory, via a novel signaling pathway involving reactive oxygen species (ROS), endoplasmic reticulum (ER) stress, and autophagy. We demonstrate in this study that exposure of morphine to hippocampal neurons leads to a reduction in excitatory synapse densities with a concomitant enhancement of inhibitory synapse densities via activation of the μ opioid receptor. Furthermore, these ef  ...[more]

Similar Datasets

| S-EPMC6532427 | biostudies-literature
| S-EPMC6875203 | biostudies-literature
| S-EPMC3716763 | biostudies-literature
| S-EPMC3131882 | biostudies-other
| S-EPMC4201572 | biostudies-literature
| S-EPMC3590254 | biostudies-other
| S-EPMC3934165 | biostudies-literature
| S-EPMC5493624 | biostudies-literature
| S-EPMC8559314 | biostudies-literature
| S-EPMC6194813 | biostudies-literature