4.5 Article

Protein tyrosine phosphatase 1B inhibition improves endoplasmic reticulum stress-impaired endothelial cell angiogenic response: A critical role for cell survival

Journal

MOLECULAR MEDICINE REPORTS
Volume 24, Issue 3, Pages -

Publisher

SPANDIDOS PUBL LTD
DOI: 10.3892/mmr.2021.12304

Keywords

protein tyrosine phosphatase 1B; tyrosine phosphatases; endoplasmic reticulum stress; endothelial dysfunction; angiogenesis; apoptosis

Funding

  1. Qatar University [QUCG-CPH-20/21-3]
  2. Qatar National Research Fund (Qatar Foundation) [NPRP-8-1750-3-360, UREP24-016-3-004]
  3. office of graduate studies (Qatar University)

Ask authors/readers for more resources

Inhibiting PTP1B can prevent ER stress-induced endothelial dysfunction and impaired angiogenesis. PTP1B blockade reduced ER stress-induced apoptosis and cell cycle arrest, possibly through decreased expression levels of caspase-12 and PARP1.
Endoplasmic reticulum (ER) stress contributes to endothelial dysfunction, which is the initial step in atherogenesis. Blockade of protein tyrosine phosphatase (PTP)1B, a negative regulator of insulin receptors that is critically located on the surface of ER membrane, has been found to improve endothelial dysfunction. However, the role of ER stress and its related apoptotic sub-pathways in PTP1B-mediated endothelial dysfunction, particularly its angiogenic capacity, have not yet been fully elucidated. Thus, the present study aimed to investigate the impact of PTP1B suppression on ER stress-mediated impaired angiogenesis and examined the contribution of apoptotic signals in this process. Endothelial cells were exposed to pharmacological ER stressors, including thapsigargin (TG) or 1,4-dithiothreitol (DTT), in the presence or absence of a PTP1B inhibitor or small interfering (si)RNA duplexes. Then, ER stress, angiogenic capacity, cell cycle, apoptosis and the activation of key apoptotic signals were assessed. It was identified that the inhibition of PTP1B prevented ER stress caused by DTT and TG. Moreover, ER stress induction impaired the activation of endothelial nitric oxide synthase (eNOS) and the angiogenic capacity of endothelial cells, while PTP1B inhibition exerted a protective effect. The results demonstrated that blockade or knockdown of PTP1B prevented ER stress-induced apoptosis and cell cycle arrest. This effect was associated with reduced expression levels of caspase-12 and poly (ADP-Ribose) polymerase 1. PTP1B blockade also suppressed autophagy activated by TG. The current data support the critical role of PTP1B in ER stress-mediated endothelial dysfunction, characterized by reduced angiogenic capacity, with an underlying mechanism involving reduced eNOS activation and cell survival. These findings provide evidence of the therapeutic potential of targeting PTP1B in cardiovascular ischemic conditions.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.5
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available