Abstract

Microtubules play critical roles in regulating the activation of NLRP3 inflammasome and microtubule-destabilizing agents such as colchicine have been shown to suppress the activation of this inflammasome. However, it remains largely unknown whether paclitaxel, a microtubule-stabilizing agent being used in cancer therapy, has any influences on NLRP3 inflammasome activation. Here we showed that paclitaxel pre-treatment greatly enhanced ATP- or nigericin-induced NLRP3 inflammasome activation as indicated by increased release of cleaved caspase-1 and mature IL-1β, enhanced formation of ASC speck, and increased gasdermin D cleavage and pyroptosis. Paclitaxel time- and dose-dependently induced α-tubulin acetylation in LPS-primed murine and human macrophages and further increased ATP- or nigericin-induced α-tubulin acetylation. Such increased α-tubulin acetylation was significantly suppressed either by resveratrol or NAD+ (coenzyme required for deacetylase activity of SIRT2), or by genetic knockdown of MEC-17 (gene encoding α-tubulin acetyltransferase 1). Concurrently, the paclitaxel-mediated enhancement of NLRP3 inflammasome activation was significantly suppressed by resveratrol, NAD+, or MEC-17 knockdown, indicating the involvement of paclitaxel-induced α-tubulin acetylation in the augmentation of NLRP3 inflammasome activation. Similar to paclitaxel, epothilone B that is another microtubule-stabilizing agent also induced α-tubulin acetylation and increased NLRP3 inflammasome activation in macrophages in response to ATP treatment. Consistent with the in vitro results, intraperitoneal administration of paclitaxel significantly increased serum IL-1β levels, reduced bacterial burden, dampened infiltration of inflammatory cells in the liver, and improved animal survival in a mouse model of bacterial infection. Collectively, our data indicate that paclitaxel potentiated NLRP3 inflammasome activation by inducing α-tubulin acetylation and thereby conferred enhanced antibacterial innate responses, suggesting its potential application against pathogenic infections beyond its use as a chemotherapeutic agent.

Highlights

  • As colchicine acting as a microtubule-destabilizing agent can suppress NLRP3 inflammasome activation [17, 20], we asked whether paclitaxel, a microtubule-stabilizing agent, could influence NLRP3 inflammasome activation

  • We assessed the effects of paclitaxel on NLRP3 inflammasome activation in LPS-primed murine J774A.1 or bone marrow-derived macrophages (BMDMs) stimulated with extracellular ATP or nigericin, two canonical NLRP3 inflammasome activators [30]

  • Paclitaxel dose-dependently increased the release of cleaved caspase-1p10 and mature IL-1β into the culture supernatants of J774A.1 cells stimulated with ATP (Figures 1A–C) and of Bone Marrow-Derived Macrophages (BMDMs) stimulated with ATP (Figures 1D–F) or nigericin (Figures 1G–I), indicating that this chemical enhanced NLRP3 inflammasome activation in murine macrophages

Read more

Summary

Introduction

In clinic, it is used for the treatment of a broad spectrum of cancers, including breast cancer, lung cancer, as well as ovarian, cervical and pancreatic cancers [1,2,3,4]. It is used for the treatment of a broad spectrum of cancers, including breast cancer, lung cancer, as well as ovarian, cervical and pancreatic cancers [1,2,3,4] It is clinically used intravenously, and its distribution throughout the body is rapid, with large volumes of distribution [5]. It has been known that paclitaxel is a microtubule-stabilizing agent. It has been demonstrated that paclitaxel affects microtubule dynamics at concentrations much lower than those inhibiting mitosis and cell division [11]

Methods
Results
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