Aging-US: PTEN nuclear translocation enhances neuronal injury after hypoxia-ischemia

07-25-2021

Aging-US published "PTEN nuclear translocation enhances neuronal injury after hypoxia-ischemia via modulation of the nuclear factor-κB signaling pathway" which reported that this study was designed to investigate phosphatase and tensin homolog nuclear translocation and its possible role in rat cortical neuronal damage following oxygen-glucose deprivation in vitro.

In addition, PTEN expression was increased and the phosphorylation of extracellular signal-regulated kinase 1/2 and activation of nuclear factor kappa B were decreased following OGD.

PTENK13R transfection prevented PTEN nuclear translocation; attenuated the effect of OGD on nuclear p-ERK1/2 and NF-κB, apoptosis, and LDH release; and increased the expression of several anti-apoptotic proteins.

They conclude that PTEN nuclear translocation plays an essential role in neuronal injury following OGD via modulation of the p-ERK1/2 and NF-κB pathways.

Prevention of PTEN nuclear translocation might be a candidate strategy for preventing brain injury following HI.

Dr. Jing Zhao from The Affiliated Hospital of North Sichuan Medical College said, "Hypoxia-ischemia (HI) during fetal and neonatal development can cause damage to neurons, leading to neurological defects and even death."

Dr. Jing Zhao from The Affiliated Hospital of North Sichuan Medical College said, "Hypoxia-ischemia (HI) during fetal and neonatal development can cause damage to neurons, leading to neurological defects and even death."

PTEN possesses both protein phosphatase and lipid phosphatase activity and PTEN nuclear translocation induced by PTEN phosphatase activity has been linked to DNA damage in cancer cells.

Additionally, PTEN has been reported to localize to both the cytoplasm and nucleus of neurons, and an imbalance in its nucleocytoplasmic distribution has been linked to neuronal injury following excitotoxicity or ischemia.

These author’s previous studies showed that the inhibition of PTEN prevents neuronal injury after HI by mediating the activity of glycogen synthase kinase 3β and AKT.

However, the roles of PTEN in the regulation of ERK phosphorylation and NF-κB activity and their effects on the underlying mechanisms of neuronal injury remain unclear.

Figure 6. Inhibition of PTEN nuclear translocation reduces neural injury after OGD. (A) TUNEL-positive cells increased after OGD. This enhanced apoptosis was blocked in GFP-PTENK13R neurons. Scale bars = 50 μm. (B) Extracellular levels of LDH decreased in GFP-PTENK13R neurons after OGD. (C) Bcl-2 and Bcl-xL protein expression was evaluated by western blot which showed that their expression increased in GFP-PTENK13R neurons after OGD. (D) Quantification of Bcl-2 and Bcl-xL protein expression normalized against GFP-PTENWT neurons. n = 5 in each column and ** p < 0.01 vs. WT. ##p < 0.01, vs. WT. OGD: oxygen and glucose deprivation, GFP: green fluorescent protein, TUNEL: terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick-end labeling WT: GFP-PTENWT neurons, K13R: GFP-PTENK13R neurons; P: PDTC.

They anticipate that this study could provide new perspectives into their understanding of the role of PTEN in neuronal injury and therapy development.

The Zhao Research Team concluded in their Aging-US Research Output, "our study provides insight into the molecular mechanisms through which PTEN nuclear translocation contributes to neuronal injury following HI. We found that PTEN nuclear translocation is an essential step in the activation of ERK1/2 and NF-κB in neurons subjected to OGD and that inhibition of PTEN nuclear translocation can provide protection against ischemic brain damage. PTENK13R mutants transfected into neurons effectively inhibited PTEN nuclear translocation, without affecting the cytoplasmic functions of PTEN. Taken together, these results suggest that the inhibition of PTEN nuclear translocation could be a potent novel strategy for protecting neurons following HI in vivo."

Sign up for free Altmetric alerts about this article

DOI - https://doi.org/10.18632/aging.203141

Full Text - https://www.aging-us.com/article/203141/text

Correspondence to: Jing Zhao email: Jenny_ZJ@yeah.net

Keywords: brain damage, hypoxia-ischemia, oxygen and glucose deprivation, phosphatase and tensin homolog deletion from chromosome 10, ERK1/2

About Aging-US:

Aging publishes research papers in all fields of aging research including but not limited, aging from yeast to mammals, cellular senescence, age-related diseases such as cancer and Alzheimer’s diseases and their prevention and treatment, anti-aging strategies and drug development and especially the role of signal transduction pathways such as mTOR in aging and potential approaches to modulate these signaling pathways to extend lifespan. The journal aims to promote treatment of age-related diseases by slowing down aging, validation of anti-aging drugs by treating age-related diseases, prevention of cancer by inhibiting aging. Cancer and COVID-19 are age-related diseases.

Aging is indexed by PubMed/Medline (abbreviated as “Aging (Albany NY)”), PubMed CentralWeb of Science: Science Citation Index Expanded (abbreviated as “Aging‐US” and listed in the Cell Biology and Geriatrics & Gerontology categories), Scopus (abbreviated as “Aging” and listed in the Cell Biology and Aging categories), Biological Abstracts, BIOSIS Previews, EMBASE, META (Chan Zuckerberg Initiative) (2018-2022), and Dimensions (Digital Science).

Please visit our website at www.Aging-US.com and connect with us:

For media inquiries, please contact media@impactjournals.com.