Abstract

Seasonal influenza epidemics represent a significant global health threat. The exacerbated immune response triggered by respiratory influenza virus infection causes severe pulmonary damage and contributes to substantial morbidity and mortality. Regulator of G-protein signaling 10 (RGS10) belongs to the RGS protein family that act as GTPase activating proteins for heterotrimeric G proteins to terminate signaling pathways downstream of G protein-coupled receptors. While RGS10 is highly expressed in immune cells, in particular monocytes and macrophages, where it has strong anti-inflammatory effects, its physiological role in the respiratory immune system has not been explored yet. Here, we show that Rgs10 negatively modulates lung immune and inflammatory responses associated with severe influenza H1N1 virus respiratory infection in a mouse model. In response to influenza A virus challenge, mice lacking RGS10 experience enhanced weight loss and lung viral titers, higher mortality and significantly faster disease onset. Deficiency of Rgs10 upregulates the levels of several proinflammatory cytokines and chemokines and increases myeloid leukocyte accumulation in the infected lung, markedly neutrophils, monocytes, and inflammatory monocytes, which is associated with more pronounced lung damage. Consistent with this, influenza-infected Rgs10-deficent lungs contain more neutrophil extracellular traps and exhibit higher neutrophil elastase activities than wild-type lungs. Overall, these findings propose a novel, in vivo role for RGS10 in the respiratory immune system controlling myeloid leukocyte infiltration, viral clearance and associated clinical symptoms following lethal influenza challenge. RGS10 also holds promise as a new, potential therapeutic target for respiratory infections.

Highlights

  • Seasonal influenza epidemics, with influenza A virus (IAV) being the most prevalent species, are estimated to result in 3-5 million severe illnesses and 300,000-650,000 deaths globally each year [1]

  • While most people infected with influenza do not die [its mortality rate is around 0.1% [1]], we decided to study the role of Regulator of G-protein signaling 10 (RGS10) in a murine model of lethal influenza infection because severe infections are associated with worse clinical outcomes requiring hospitalization and expensive treatments, mainly in immunocompromised patients

  • These results indicate that RGS10 delays the onset of the clinical consequences of lethal respiratory IAV infection

Read more

Summary

Introduction

With influenza A virus (IAV) being the most prevalent species, are estimated to result in 3-5 million severe illnesses and 300,000-650,000 deaths globally each year [1]. Despite the COVID-19 pandemic, these influenza epidemics continue to represent a significant financial burden and a major global health threat. In response to IAV infection and subsequent replication, infected airway epithelial cells and activated lung-resident immune cells release inflammatory cytokines and chemokines to initiate a robust influx of innate immune cells, such as neutrophils, monocytes and macrophages into the lung [6]. While the initial responses of neutrophils and macrophages are essential for IAV clearance, dysregulated and persistent inflammatory cell recruitment mediating uncontrolled inflammation leads to pulmonary damage and increased morbidity and mortality [7, 8]. Identifying key regulators controlling excessive immune cell recruitment and the subsequent proinflammatory cytokine storm bears significance in the development of strategies for the treatment of influenza infections

Objectives
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