Skip to main content

Advertisement

Log in

β-Hydroxybutyrate Alleviates Low Glucose–Induced Apoptosis via Modulation of ROS-Mediated p38 MAPK Signaling

  • Published:
Journal of Molecular Neuroscience Aims and scope Submit manuscript

Abstract

Hypoglycemia has emerged as a prominent complication in anti-diabetic drug therapy or negative energy balance of animals, which causes brain damage, cognitive impairment, and even death. Brain injury induced by hypoglycemia is closely related to oxidative stress and the production of reactive oxygen species (ROS). The intracellular accumulation of ROS leads to neuronal damage, even death. Ketone body β-hydroxybutyrate (BHBA) not only serves as alternative energy source for glucose in extrahepatic tissues, but is also involved in cellular signaling transduction. Previous studies showed that BHBA reduces apoptosis by inhibiting the excessive production of ROS and activation of caspase-3. However, the effects of BHBA on apoptosis induced by glucose deprivation and its related molecular mechanisms have been seldom reported. In the present study, PC12 cells and primary cortical neurons were used to establish a low glucose injury model. The effects of BHBA on the survival and apoptosis in a glucose deficient condition and related molecular mechanisms were investigated by using flow cytometry, immunofluorescence, and western blotting. PC12 cells were incubated with 1 mM glucose for 24 h as a low glucose cell model, in which ROS accumulation and cell mortality were significantly increased. After 24 h and 48 h treatment with different concentrations of BHBA (0 mM, 0.05 mM, 0.5 mM, 1 mM, 2 mM), ROS production was significantly inhibited. Moreover, cell apoptosis rate was decreased and survival rate was significantly increased in 1 mM and 2 mM BHBA groups. In primary cortical neurons, at 24 h after treatment with 2 mM BHBA, the injured length and branch of neurites were significantly improved. Meanwhile, the intracellular ROS level, the proportion of c-Fos+ cells, apoptosis rate, and nuclear translocation of NF-κB protein after treatment with BHBA were significantly decreased when compared with that in low glucose cells. Importantly, the expression of p38, p-p38, NF-κB, and caspase-3 were significantly decreased, while the expression of p-ERK was significantly increased in both PC12 cells and primary cortical neurons. Our results demonstrate that BHBA decreased the accumulation of intracellular ROS, and further inhibited cell apoptosis by mediating the p38 MAPK signaling pathway and caspase-3 apoptosis cascade during glucose deprivation. In addition, BHBA inhibited apoptosis by activating ERK phosphorylation and alleviated the damage of low glucose to PC12 cells and primary cortical neurons. These results provide new insight into the anti-apoptotic effect of BHBA in a glucose deficient condition and the related signaling cascade.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8

Similar content being viewed by others

Data Availability

The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.

Code Availability

Not applicable.

Abbreviations

LG-CM:

Low glucose complete medium

MG-CM:

5 MM medium glucose complete medium

HG-CM:

25 MM high glucose complete medium

BHBA:

β-Hydroxybutyrate

ROS:

Reactive oxygen species

CNS:

Central nervous system

AD:

Alzheimer’s disease

DMEM:

Dulbecco’s modified Eagle’s medium

FBS:

Fetal bovine serum

PI:

Propidium iodide

MTT:

Methylthiazolyldiphenyl-tetrazolium bromide.

References

Download references

Funding

This work was financially supported by the National Natural Science Foundation of China (No. 31802154), National Key Research and Development Program of China (2018YFE0127000), the China Postdoctoral Science Foundation funded project (No. 2019T120957), Shaanxi Provincial Regional Innovation Capability Guiding Plan Project (No. 2020QFY10-04), and General Project of Basic Research of Shaanxi Province (No. 2021JM-492).

Author information

Authors and Affiliations

Authors

Contributions

Cixia Li: writing — original draft, methodology, investigation, data curation; Xuejun Chai: writing — review and editing, validation, investigation; Jiarong Pan: methodology, visualization, investigation, data curation, Jian Huang: validation, investigation; Yongji Wu: methodology, investigation; Yuhuan Xue: investigation; Wentai Zhou: data curation; Jiping Yang: methodology; Xiaoyan Zhu: conceptualization, writing — review and editing, funding acquisition, supervision; Shanting Zhao: writing — review and editing, funding acquisition, resources, project administration.

Corresponding authors

Correspondence to Xiaoyan Zhu or Shanting Zhao.

Ethics declarations

Ethics Approval

No human studies were carried out by the authors for this article. This article does not contain any studies with human participants performed by any of the authors.

Consent to Participate

Informed consent was obtained for each participant.

Consent for Publication

All participants consented to publication.

Conflict of Interest

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, C., Chai, X., Pan, J. et al. β-Hydroxybutyrate Alleviates Low Glucose–Induced Apoptosis via Modulation of ROS-Mediated p38 MAPK Signaling. J Mol Neurosci 72, 923–938 (2022). https://doi.org/10.1007/s12031-022-01974-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12031-022-01974-3

Keywords

Navigation