Abstract

Interleukin-10 (IL-10) and tumor necrosis factor-α (TNF-α) are key anti- and pro-inflammatory mediators elicited during the host immune response to Mycobacterium tuberculosis (Mtb). Understanding the opposing effects of these mediators is difficult due to the complexity of processes acting across different spatial (molecular, cellular, and tissue) and temporal (seconds to years) scales. We take an in silico approach and use multi-scale agent based modeling of the immune response to Mtb, including molecular scale details for both TNF-α and IL-10. Our model predicts that IL-10 is necessary to modulate macrophage activation levels and to prevent host-induced tissue damage in a granuloma, an aggregate of cells that forms in response to Mtb. We show that TNF-α and IL-10 parameters related to synthesis, signaling, and spatial distribution processes control concentrations of TNF-α and IL-10 in a granuloma and determine infection outcome in the long-term. We devise an overall measure of granuloma function based on three metrics – total bacterial load, macrophage activation levels, and apoptosis of resting macrophages – and use this metric to demonstrate a balance of TNF-α and IL-10 concentrations is essential to Mtb infection control, within a single granuloma, with minimal host-induced tissue damage. Our findings suggest that a balance of TNF-α and IL-10 defines a granuloma environment that may be beneficial for both host and pathogen, but perturbing the balance could be used as a novel therapeutic strategy to modulate infection outcomes.

Highlights

  • Tuberculosis (TB) is an infectious disease caused by the pathogen Mycobacterium tuberculosis (Mtb)

  • The molecular scale model is composed of three sub-models, each described by systems of differential equations, and is linked to the tissue and cellular scale [83,84]: (1) Single-cell level tumor necrosis factor-a (TNF-a) and IL-10 secretion and receptor-ligand dynamics are described by ordinary differential equations (ODEs), (2) diffusion of TNF-a, IL-10, and chemokines are described by partial differential equations (PDEs), and (3) degradation of TNFa, IL-10, and chemokines are described by ODEs

  • We extend our previous multi-scale hybrid agent-based model of Mtb infection by integrating TNF-a and IL-10 single-cell level receptor-ligand dynamics

Read more

Summary

Introduction

Tuberculosis (TB) is an infectious disease caused by the pathogen Mycobacterium tuberculosis (Mtb). Upon infection with Mtb, 5–10% of individuals develop active pulmonary TB, while about 90% develop a state of chronic infection, known as latent pulmonary TB, showing no clinical signs of disease [3,4,5]. Granulomas are structures which form in the lungs as a result of the immune response to inhaled Mtb. Granulomas serve as the central site of host-pathogen interaction during Mtb infection, with a host typically developing several granulomas based on the number of inhaled bacteria [4,6]. During latent pulmonary TB, granulomas are able to control Mtb but not completely eradicate the bacteria, while during active pulmonary TB Mtb growth is unrestrained in a portion of granulomas. The host factors that control the outcome of infection, in particular the formation and function of a granuloma, are not well understood and are difficult to use as therapeutic targets

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