Abstract

NOD-like receptor family, pyrin domain-containing protein 3 (NLRP3) inflammasome-mediated pyroptosis is a crucial event in the preeclamptic pathogenesis, tightly linked with the uteroplacental TLR4/NF-κB signaling. Trophoblastic glycometabolism reprogramming has now been noticed in the preeclampsia pathogenesis, plausibly modulated by the TLR4/NF-κB signaling as well. Intriguingly, cellular pyroptosis and metabolic phenotypes may be inextricably linked and interacted. Metformin (MET), a widely accepted NF-κB signaling inhibitor, may have therapeutic potential in preeclampsia while the underlying mechanisms remain unclear. Herein, we investigated the role of MET on trophoblastic pyroptosis and its relevant metabolism reprogramming. The safety of pharmacologic MET concentration to trophoblasts was verified at first, which had no adverse effects on trophoblastic viability. Pharmacological MET concentration suppressed NLRP3 inflammasome-induced pyroptosis partly through inhibiting the TLR4/NF-κB signaling in preeclamptic trophoblast models induced via low-dose lipopolysaccharide. Besides, MET corrected the glycometabolic reprogramming and oxidative stress partly via suppressing the TLR4/NF-κB signaling and blocking transcription factor NF-κB1 binding on the promoter PFKFB3, a potent glycolytic accelerator. Furthermore, PFKFB3 can also enhance the NF-κB signaling, reduce NLRP3 ubiquitination, and aggravate pyroptosis. However, MET suppressed pyroptosis partly via inhibiting PFKFB3 as well. These results provided that the TLR4/NF-κB/PFKFB3 pathway may be a novel link between metabolism reprogramming and NLRP3 inflammasome-induced pyroptosis in trophoblasts. Further, MET alleviates the NLRP3 inflammasome-induced pyroptosis, which partly relies on the regulation of TLR4/NF-κB/PFKFB3-dependent glycometabolism reprogramming and redox disorders. Hence, our results provide novel insights into the pathogenesis of preeclampsia and propose MET as a potential therapy.

Highlights

  • Preeclampsia (PE), characterized by new-onset hypertension typically after 20 weeks of gestation together with multisystem involvement, complicates 2%–8% of pregnancies globally with huge associated healthcare burdens to the whole of society [1]

  • With the increased expression of NLRP3 and related mediators found in the placentas [13, 14], trophoblastic pyroptosis has been suggested to be one of the PE pathogenesis [2, 3]. It remains to be verified whether the overactive Toll-like receptor 4 (TLR4)/nuclear factor-κB (NF-κB) signaling in preeclamptic trophoblasts triggers the NLRP3 inflammasome-induced pyroptosis

  • We found that (1) pharmacological concentration of MET alleviated NLRP3 inflammasome-induced pyroptosis partly through inhibiting the TLR4/NF-κB signaling; (2) pharmacological MET concentration restored the metabolic and redox homeostasis in trophoblasts partly via the suppression of the TLR4/NF-κB signaling; (3) MET decreased the transcription of PFKFB3 via blocking transcription factor NF-κB1 binding on PFKFB3 promoter; (4) PFKFB3 augmented the NF-κB signaling as well as the NLRP3 inflammasome-induced pyroptosis while MET suppressed pyroptosis partly via inhibiting PFKFB3

Read more

Summary

Introduction

Preeclampsia (PE), characterized by new-onset hypertension typically after 20 weeks of gestation together with multisystem involvement, complicates 2%–8% of pregnancies globally with huge associated healthcare burdens to the whole of society [1]. Extensive research has reported the vital role of highly expressed and overactivated TLR4/NFκB signaling in the preeclamptic placentas via causing trophoblastic inflammation, oxidative stress, and dysfunction [11, 12]. With the increased expression of NLRP3 and related mediators (caspase 1 and IL-1β) found in the placentas [13, 14], trophoblastic pyroptosis has been suggested to be one of the PE pathogenesis [2, 3]. It remains to be verified whether the overactive TLR4/NF-κB signaling in preeclamptic trophoblasts triggers the NLRP3 inflammasome-induced pyroptosis

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call