Abstract

Although Ikaros (IKZF1) is a well-established transcriptional regulator in leukocyte lymphopoiesis and differentiation, its role in myeloid innate immune responses remains unclear. Sirtuin 1 (SIRT1) is a histone/protein deacetylase involved in cellular senescence, inflammation, and stress resistance. Whether SIRT1 signaling is essential in myeloid cell activation remains uncertain, while the molecular communication between Ikaros and SIRT1, two major transcriptional regulators, has not been studied. We undertook molecular and functional studies to interrogate the significance of the myeloid Ikaros-SIRT1 axis in innate immune activation and whether it may serve as a homeostatic sentinel in human liver transplant recipients (hepatic biopsies) and murine models of sterile hepatic inflammation (liver warm ischemia-reperfusion injury in wild-type, myeloid-specific Sirt1-knockout, and CD11b-DTR mice) as well as primary bone marrow-derived macrophage (BMM) cultures (Ikaros silencing vs. overexpression). In our clinical study, we identified increased post-reperfusion hepatic Ikaros levels, accompanied by augmented inflammasome signaling yet depressed SIRT1, as a mechanism of hepatocellular damage in liver transplant recipients. In our experimental studies, we identified infiltrating macrophages as the major source of Ikaros in IR-stressed mouse livers. Then, we demonstrated that Ikaros-regulated pyroptosis - induced by canonical inflammasome signaling in BMM cultures - was SIRT1 dependent. Consistent with the latter, myeloid-specific Ikaros signaling augmented hepatic pyroptosis to aggravate pro-inflammatory responses invivo by negatively regulating SIRT1 in an AMPK-dependent manner. Finally, myeloid-specific SIRT1 was required to suppress pyroptosis, pro-inflammatory phenotype, and ultimately mitigate hepatocellular injury in ischemia-stressed murine livers. These findings identify the Ikaros-SIRT1 axis as a novel mechanistic biomarker of pyroptosis and a putative checkpoint regulator of homeostasis in response to acute hepatic stress/injury in mouse and human livers. This report describes how crosstalk between Ikaros and SIRT1, two major transcriptional regulators, influence acute hepatic inflammation in murine models of liver ischemia-reperfusion injury and liver transplant recipients. Weshow that the myeloid Ikaros-SIRT1 axis regulates inflammasome-pyroptotic cell death and hepatocellular damage in stressed livers. Thus, the Ikaros-SIRT1 axis may serve as a novel checkpoint regulator that is required for homeostasis in response to acute liver injury in mice and humans.

Highlights

  • Orthotopic liver transplantation (OLT) has become the standard of care for end-stage liver disease and hepatic malignancies[1]

  • We have reported on TLR-primed inflammasome as a critical signaling platform that detects stressors (e.g., DAMPs) in hepatic IRI7,8 while others targeted Gasdermin D (GSDMD) to focus on pyroptosis in innate immune cell[9] and Kupffer cell[10,11] death

  • -p we found no correlation between IKZF1 grouping and donor/graft variables, including sex, height, re BMI, or warm/cold graft ischemia times (Table S2)

Read more

Summary

Introduction

Orthotopic liver transplantation (OLT) has become the standard of care for end-stage liver disease and hepatic malignancies[1]. Liver IRI represents an innate-immune continuum of pro-inflammatory cytokine release ro and hepatocyte death. 4 (TLR4) signaling triggers sterile hepatic inflammatory cascade in OLT recipients[3]. TLR4mediated canonical activation of the inflammasome complex was associated with a more recently re identified inflammatory cell death termed pyroptosis[4]. Ikaros (IKZF1) is a well-established transcriptional regulator in leukocyte lymphopoiesis and differentiation, its role in myeloid innate immune responses remains unclear. Whether SIRT1 signaling is essential in myeloid cell activation remains uncertain. We undertook molecular and functional studies to of interrogate the significance of the myeloid Ikaros–SIRT1 axis in the innate immune activation and ro to determine whether it may serve as a homeostatic sentinel in hepatic rejuvenation in mouse and re -p human liver

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