Abstract

Intervertebral disc degeneration (IVDD) remains a clinical challenge and requires more effective therapeutic targets. Long noncoding RNAs (lncRNAs) have emerged as critical modulators of multiple biological processes, such as cell proliferation and extracellular matrix (ECM) remodeling. Accordingly, the current study sets out to explore the influence of the lncRNA growth arrest-specific 5 (GAS5) on IVDD and investigate the possible involvement of microRNA-17-3p (miR-17-3p)/Angiopoietin-2 (Ang-2) axis. Firstly, the expression patterns of GAS5, miR-17-3p, and Ang-2 were characterized by RNA quantification from the isolated human degenerative nucleus pulposus (NP) tissues. miR-17-3p was found to express at an abnormal low level while GAS5 and Ang-2 expressed at aberrant high level in the human degenerative NP tissues. Utilizing dual-luciferase reporter, RNA immunoprecipitation, and pull-down assays, GAS5 was found to competitively bound to miR-17-3p and further upregulate the expression of Ang-2, a target gene of miR-17-3p. Employing gain- and loss-of-function approaches, their expressions were altered in human degenerative nucleus pulposus cells (NPCs), followed by IL-1β treatment, in order to identify their roles in NP cell proliferation, apoptosis, and ECM metabolism. Silencing of GAS5 expression restrained the levels of cleaved caspase-3, cleaved caspase-7, cleaved caspase-9, MMP3, MMP13, ADAMTS4, and ADAMTS5 and increased collagen II and aggrecan levels. In vitro experiments also revealed that GAS5 depletion inhibited apoptosis and ECM degradation in HDNPCs, while elevating the proliferation through downregulation of Ang-2 by increasing miR-17-3p. Furthermore, in vivo data further validated that either GAS5 silencing or miR-17-3p reexpression alleviated IVDD degree with the help of IVDD mouse models. Altogether, our findings substantiated that downregulation of GAS5 reduced NPC apoptosis and promoted ECM remodeling, ultimately ameliorating the IVDD via miR-17-3p-dependent inhibition of Ang-2. We hope our discoveries offer a fresh molecular insight that can aid the development of novel therapies against IVDD.

Highlights

  • A multifactorial degenerative disease of the spinal cord, intervertebral disc degeneration (IVDD) represents a tremendous clinical challenge [1]

  • The aforementioned findings suggested that growth arrest-specific 5 (GAS5) might downregulate miR-17-3p in nucleus pulposus cells (NPCs); we focused on elucidating the potential effects of the GAS5/miR-17-3p axis on the functions of NPCs

  • And 6(h), cell viability was reduced, while cell apoptosis was promoted following Ang-2 overexpression, whereas Ang-2 reexpression could negate the above effects of si-GAS5 or miR-17-3p mimic on viability and apoptosis. These findings provided evidence highlighting the involvement of the GAS5/miR-17-3p/Ang-2 axis in modulating the proliferation, apoptosis, and extracellular matrix (ECM) remodeling of NPCs

Read more

Summary

Introduction

A multifactorial degenerative disease of the spinal cord, intervertebral disc degeneration (IVDD) represents a tremendous clinical challenge [1]. IVDD is characterized by the presence of proinflammatory factors, loss of active nucleus pulposus (NP) or annulus fibrosus (AF) cells, extracellular matrix (ECM) degradation, and alterations in disc cell phenotypes [2]. Risk factors responsible for the occurrence of IVDD include a variety of factors, such as genetic polymorphisms, age, smoking habit, and weight gain [3, 4]. The cytokine inflammatory mediator interleukin-1 beta (IL-1β) is known to be produced as a result of pyroptosis, one of cell death mechanisms, after the activation of the inflammasome [5, 6]. Existing evidence suggests that overproduction of IL-1β can Oxidative Medicine and Cellular Longevity facilitate inflammatory process and further precipitate nucleus pulposus cell (NPC) apoptosis and ECM degradation, leading to IVDD and lower back pain [7]. Numerous studies have been conducted in a bid to illuminate potential therapeutic strategies, identification of more prognostic biomarkers or biological targets may contribute to the development of individualized therapeutics for this disease [8]

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