Dynamic balance of pro- and anti-inflammatory signals controls disease and limits pathology

Joseph M Cicchese, Stephanie Evans, Caitlin Hult, Louis R Joslyn, Timothy Wessler, Jess A Millar, Simeone Marino, Nicholas A Cilfone, Joshua T Mattila, Jennifer J Linderman, Denise E Kirschner, Joseph M Cicchese, Stephanie Evans, Caitlin Hult, Louis R Joslyn, Timothy Wessler, Jess A Millar, Simeone Marino, Nicholas A Cilfone, Joshua T Mattila, Jennifer J Linderman, Denise E Kirschner

Abstract

Immune responses to pathogens are complex and not well understood in many diseases, and this is especially true for infections by persistent pathogens. One mechanism that allows for long-term control of infection while also preventing an over-zealous inflammatory response from causing extensive tissue damage is for the immune system to balance pro- and anti-inflammatory cells and signals. This balance is dynamic and the immune system responds to cues from both host and pathogen, maintaining a steady state across multiple scales through continuous feedback. Identifying the signals, cells, cytokines, and other immune response factors that mediate this balance over time has been difficult using traditional research strategies. Computational modeling studies based on data from traditional systems can identify how this balance contributes to immunity. Here we provide evidence from both experimental and mathematical/computational studies to support the concept of a dynamic balance operating during persistent and other infection scenarios. We focus mainly on tuberculosis, currently the leading cause of death due to infectious disease in the world, and also provide evidence for other infections. A better understanding of the dynamically balanced immune response can help shape treatment strategies that utilize both drugs and host-directed therapies.

Keywords: cytokines infectious disease; granuloma; immune response; mathematical modelling; mycobacterium tuberculosis.

Conflict of interest statement

CONFLICT OF INTEREST

The authors declare no conflict of interest.

© 2018 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.

Figures

FIGURE 1
FIGURE 1
Dynamics of pro- and anti-inflammatory immune responses steer disease progression along various trajectories. Schematic of relative pro- and anti-inflammatory responses to a pathogen over time corresponding to various host outcomes (A). Schematic of disease trajectories corresponding to qualitative ratios of pro- and anti-inflammatory responses (B). The outcomes are (i) high pathogen burden (blue); (ii) severe tissue damage (red); (iii) high pathogen burden along with a large amount of tissue damage (gray); (iv) a cleared infection and return to base levels of inflammation (white); (v) a dynamically balanced pro- and anti-inflammatory response to control pathogen growth and limit host pathology (purple)
FIGURE 2
FIGURE 2
Multi-organ events following infection with Mycobacterium tuberculosis. Mtb is inhaled into lungs and engulfed by macrophages where intracellular replication occurs. Additionally, DCs take up Mtb and traffic to lung-draining LNs via afferent lymphatics, where they prime T cells that have been recruited from high endothelial venules (HEV). These primed T cells migrate to lungs via efferent lymphatics and participate in granuloma formation and function by activating macrophages, secreting cytokines, and participating in adaptive immune responses
FIGURE 3
FIGURE 3
Computational and experimental visualizations of various granulomas. A zoomed in 2-D GranSim snapshot (A), which shows macrophage substates (green-resting, blue-activated, orange-infected, red-chronically infected), T-cell phenotypes (pink IFN-producing, purple-cytotoxic, light blue-Tregs), extracellular bacteria (yellow), necrosis (white). (B) is a GranSim snapshot of a different granuloma that shows the concentration gradient of TNF (units on colorbar are molecules of TNF per grid compartment). (C) is a macaque granuloma image stained for macrophages (Red CD68), T cells (Green CD3), and neutrophils (Blue calprotectin). (D) is a time plot of CFU/lesion NHP data (red, median with error bars indicating min/max) along with GranSim simulations with model predictions (median–solid line, min/max–dashed lines). Simulations plotted as in with a newer generation of GranSim and data derived from 32 NHP in previously published works ,,,,
FIGURE 4
FIGURE 4
Balanced immune responses lead to contained Mtb granulomas. The top row (A, B, C) displays conceptual images of granuloma formation across a spectrum of immune responses. During a primarily anti-inflammatory response (A), a granuloma shows a lack of proper formation, and large amounts of caseum develop as the host struggles to contain the pathogen. A balanced response (B) displays proper granuloma formation and containment of the pathogen. A primarily pro-inflammatory response (C) clears the pathogen, but at the cost of widespread cellular activation, death, and inflammation. The bottom row (D, E, F) displays GranSim simulation snapshots of these scenarios at day 150 post infection. A containment simulation from baseline parameter ranges is demonstrated in (E), whereas a simulation from a TNF-depletion parameter set is shown in (D), and a simulation from an IL-10 knockout parameter set that also exhibits more pro-inflammatory behavior (F). (D), (E), and (F) correspond to the conceptual immune responses displayed in (A), (B), and (C), respectively
FIGURE 5
FIGURE 5
Cytokine distribution across in silico and bead model granulomas. (A) GranSim snapshot of a representative containment granuloma scenario at 50 days post infection. The TNF (red) and IL-10 (blue) gradients of this representative granuloma are shown in (B). In GranSim granulomas, the gradient of TNF is contained within the granuloma (see diameter of granuloma on x-axis), and the gradient of IL-10 spreads along the diameter of the granuloma. This particular set of gradients is representative of containment granulomas and is consistent across time. If the spatial range of TNF is too large, bystander cellular death increases despite little increase in bacterial killing. If TNF spatial distribution is too narrow, simulations show increased bacterial load. Similarly, if IL-10 has a spatial distribution that is too wide, IL-10 decreases bystander cell death, but causes increased bacterial loads in the granuloma. Yet, too narrow of an IL-10 distribution yields too powerful of a pro-inflammatory response. (C) The well-established PPD bead model of granuloma formation in mice (protocol in ,–) can determine whether there is a gradient of soluble TNF within a single granuloma and detect soluble TNF gradients in ex vivo granuloma tissue. Immunofluorescence techniques identify the spatial organization of cells in granulomas, and then this known organization is combined with flow cytometry data, a simple receptor-ligand model, and fluorescent microscopy of unbound TNF receptors to reconstruct the relative soluble TNF gradient. A molecular probe from biotinylation of recombinant TNF was constructed to target unbound TNF receptors.– In the representative PPD bead granuloma shown, stains are for free TNFR (yellow) using the biotinylated TNF probe and cell nuclei (blue) using DAPI. The dashed white circle indicates approximate PPD bead location. Entire image was used. (D) Intensity of biotinylated TNF in image from (C) as a function of radial distance from the center of the PPD bead, binned into 25 pixel lengths. Unbound TNFR is roughly inversely proportional to gradient of soluble TNF (derived from the simple receptor-ligand model). Bars indicate standard deviation
FIGURE 6
FIGURE 6
Simulated antibiotic distributions in granulomas using GranSim. Heat maps of isoniazid (INH) concentration for the 3 representative granulomas shown in Figure 4: one that is highly anti-inflammatory (A), one that is contained (B), and one that is highly pro-inflammatory (C). Each type of granuloma has lower concentrations of antibiotic compared to the surrounding tissue. The heat maps show INH concentration in granulomas after 150 days of infection and 2 hours after a single, oral dose of 5 mg/kg. PK/PD modeled as in,
FIGURE 7
FIGURE 7
Effects of host-directed therapies on treatment outcome. (A) An in silico biorepository of 232 granulomas was created using GranSim and an infection period of 200 days. Starting at day 200, different treatments were simulated for 180 days: antibiotics alone (gray), antibiotics with varying doses of etanercept (500 μg/kg in red, 250 μg/kg in orange, 100 μg/kg in yellow), antibiotics with a 20% TNF-depletion (green), and antibiotics with 20% TNF-depletion and 10% IL-10 promotion (blue). Percent depletions and promotions were simulated by reducing or increasing the amount of cytokine secreted from immune cells compared to a baseline simulation. (B) The percent of granulomas that contain bacteria over the course of treatment shows that antibiotics alone sterilize more granulomas and sterilize faster than any of the host-directed therapies. (C) Host-directed therapies tend to have higher median CFU compared to antibiotics alone from 10-20 days post-treatment and later. (D) Simulations involving host-directed therapies result in granulomas that gradually decrease in size, compared to those involving only antibiotics (only medians shown)

References

    1. Jasenosky LD, Scriba TJ, Hanekom WA, Goldfeld AE. T cells and adaptive immunity to Mycobacterium tuberculosis in humans. Immunol Rev. 2015;264:74–87.
    1. Nussing S, Sant S, Koutsakos M, Subbarao K, Nguyen THO, Kedzierska K. Innate and adaptive T cells in influenza disease. Front Med. 2018;12:34–47.
    1. Zuniga EI, Macal M, Lewis GM, Harker JA. Innate and adaptive immune regulation during chronic viral infections. Annu Rev Virol. 2015;2:573–597.
    1. Murphy K, Weaver C. Immunobiology, 9th edn. New York, NY: Current Biology Ltd./Garland Publishing Inc.; 2016.
    1. Domingo-Gonzalez R, Prince O, Cooper A, Khader SA. Cytokines and chemokines in Mycobacterium tuberculosis infection. Microbiol Spectr. 2016;4:TBTB2-0018-2016.
    1. Lin PL, Rodgers M, Smith L, et al. Quantitative comparison of active and latent tuberculosis in the cynomolgus macaque model. Infect Immun. 2009;77:4631–4642.
    1. Lin PL, Flynn JL. Understanding latent tuberculosis: a moving target. J Immunol. 2010;185:15–22.
    1. Ramakrishnan L Revisiting the role of the granuloma in tuberculosis. Nat Rev Immunol. 2012;12:352–366.
    1. Herrera V, Perry S, Parsonnet J, Banaei N. Clinical application and limitations of interferon-gamma release assays for the diagnosis of latent tuberculosis infection. Clin Infect Dis. 2011;52:1031–1037.
    1. Al-Orainey IO. Diagnosis of latent tuberculosis: Can we do better? Ann Thor Med. 2009;4:5–9.
    1. Flynn JL, Chan J. Tuberculosis: latency and reactivation. Infect Immun. 2001;69:4195–4201.
    1. Zumla A, Raviglione M, Hafner R, von Reyn CF. Tuberculosis. N Engl J Med. 2013;368:745–755.
    1. Diedrich CR, Flynn JL. HIV-1/Mycobacterium tuberculosis coinfection immunology: how does HIV-1 exacerbate tuberculosis? Infect Immun. 2011;79:1407–1417.
    1. Marino S, Sud D, Plessner H, et al. Differences in reactivation of tuberculosis induced from anti-TNF treatments are based on bioavailability in granulomatous tissue. PLoS Comput Biol. 2007;3:1909–1924.
    1. Barry CE 3rd, Boshoff HI, Dartois V, et al. The spectrum of latent tuberculosis: Rethinking the biology and intervention strategies. Nat Rev Microbiol. 2009;7:845–855.
    1. Flynn JL, Klein E. Pulmonary tuberculosis in monkeys In: Leong J, Dick T, eds. A Color Atlas of Comparative Pulmonary Tuberculosis Histopathology. Boca Raton, FL: CRC Press, Taylor & Francis; 2011:83–106.
    1. Lin PL, Ford CB, Coleman MT, et al. Sterilization of granulomas is common in active and latent tuberculosis despite within-host variability in bacterial killing. Nat Med. 2014;20:75–79.
    1. Martin CJ, Cadena AM, Leung VW, et al. Digitally barcoding Mycobacterium tuberculosis reveals in vivo infection dynamics in the macaque model of tuberculosis. MBio. 2017;8:e00312–e00317.
    1. Cadena AM, Fortune SM, Flynn JL. Heterogeneity in tuberculosis. Nat Rev Immunol. 2017;17:691–702.
    1. Cadena AM, Flynn JL, Fortune SM. The importance of first impressions: Early events in Mycobacterium tuberculosis infection influence outcome. MBio. 2016;7:e00342–00316.
    1. Lyadova I Inflammation and immunopathogenesis of tuberculosis progression, understanding tuberculosis – Analyzing the origin of Mycobacterium tuberculosis pathogenicity. InTech; 2012.
    1. Leem AY, Song JH, Lee EH, et al. Changes in cytokine responses to TB antigens ESAT-6, CFP-10 and TB 7.7 and inflammatory markers in peripheral blood during therapy. Sci Rep. 2018;8:1159.
    1. Badell E, Nicolle F, Clark S, et al. Protection against tuberculosis induced by oral prime with Mycobacterium bovis BCG and intranasal subunit boost based on the vaccine candidate Ag85B-ESAT-6 does not correlate with circulating IFN-γ gamma producing T-cells. Vaccine. 2009;27:28–37.
    1. Clark S, Cross ML, Smith A, et al. Assessment of different formulations of oral Mycobacterium bovis Bacille Calmette-Guerin (BCG) vaccine in rodent models for immunogenicity and protection against aerosol challenge with M. bovis. Vaccine. 2008;26:5791–5797.
    1. Dey B, Jain R, Khera A, et al. Boosting with a DNA vaccine expressing ESAT-6 (DNAE6) obliterates the protection imparted by recombinant BCG (rBCGE6) against aerosol Mycobacterium tuberculosis infection in guinea pigs. Vaccine. 2009;28:63–70.
    1. Freches D, Romano M, Korf H, et al. Increased pulmonary tumor necrosis factor alpha, interleukin-6 (IL-6), and IL-17A responses compensate for decreased gamma interferon production in anti-IL-12 autovaccine-treated, Mycobacterium bovis BCG-vaccinated mice. Clin Vaccine Immunol. 2011;18:95–104.
    1. Lawn SD, Wilkinson RJ, Lipman MC, Wood R. Immune reconstitution and “unmasking” of tuberculosis during antiretroviral therapy. Am J Respir Crit Care Med. 2008;177:680–685.
    1. Green AM, Difazio R, Flynn JL. IFN-γ from CD4 T cells is essential for host survival and enhances CD8 T cell function during Mycobacterium tuberculosis infection. J Immunol. 2013;190:270–277.
    1. Einarsdottir T, Lockhart E, Flynn JL. Cytotoxicity and secretion of gamma interferon are carried out by distinct CD8 T cells during Mycobacterium tuberculosis infection. Infect Immun. 2009;77:4621–4630.
    1. Lazarevic V, Flynn J. CD8+ T cells in tuberculosis. Am J Respir Crit Care Med. 2002;166:1116–1121.
    1. Chen CY, Huang D, Wang RC, et al. A critical role for CD8 T cells in a nonhuman primate model of tuberculosis. PLoS Pathog. 2009;5:e1000392.
    1. Saunders BM, Frank AA, Orme IM, Cooper AM. CD4 is required for the development of a protective granulomatous response to pulmonary tuberculosis. Cell Immunol. 2002;216:65–72.
    1. Lin PL, Rutledge T, Green AM, et al. CD4 T cell depletion exacerbates acute Mycobacterium tuberculosis while reactivation of latent infection is dependent on severity of tissue depletion in cynomolgus macaques. AIDS Res Hum Retroviruses. 2012;28:1693–1702.
    1. Scanga CA, Mohan VP, Yu K, et al. Depletion of CD4(+) T cells causes reactivation of murine persistent tuberculosis despite continued expression of interferon gamma and nitric oxide synthase 2. J Exp Med. 2000;192:347–358.
    1. Yao S, Huang D, Chen CY, Halliday L, Wang RC, Chen ZW. CD4+ T cells contain early extrapulmonary tuberculosis (TB) dissemination and rapid TB progression and sustain multieffector functions of CD8+ T and CD3-lymphocytes: Mechanisms of CD4+ T cell immunity. J Immunol. 2014;192:2120–2132.
    1. Bates M, Marais BJ, Zumla A. Tuberculosis comorbidity with communicable and noncommunicable diseases. Cold Spring Harb Perspect Med. 2015;5:a017889.
    1. Diedrich CR, Mattila JT, Klein E, et al. Reactivation of latent tuberculosis in cynomolgus macaques infected with SIV is associated with early peripheral T cell depletion and not virus load. PLoS ONE. 2010;5:e9611.
    1. Barber DL, Mayer-Barber KD, Feng CG, Sharpe AH, Sher A. CD4 T cells promote rather than control tuberculosis in the absence of PD-1-mediated inhibition. J Immunol. 2011;186:1598–1607.
    1. Green AM, Mattila JT, Bigbee CL, Bongers KS, Lin PL, Flynn JL. CD4(+) regulatory T cells in a cynomolgus macaque model of Mycobacterium tuberculosis infection. J Infect Dis. 2010;202:533–541.
    1. Lazar-Molnar E, Chen B, Sweeney KA, et al. Programmed death-1 (PD-1)-deficient mice are extraordinarily sensitive to tuberculosis. Proc Natl Acad Sci USA. 2010;107:13402–13407.
    1. Windish HP, Lin PL, Mattila JT, et al. Aberrant TGF-beta signaling reduces T regulatory cells in ICAM-1-deficient mice, increasing the inflammatory response to Mycobacterium tuberculosis. J Leukoc Biol. 2009;86:713–725.
    1. Gideon HP, Phuah J, Myers AJ, et al. Variability in tuberculosis granuloma T cell responses exists, but a balance of pro- and anti-inflammatory cytokines is associated with sterilization. PLoS Pathog. 2015;11:e1004603.
    1. Orme IM, Robinson RT, Cooper AM. The balance between protective and pathogenic immune responses in the TB-infected lung. Nat Immunol. 2015;16:57–63.
    1. Flynn JL, Chan J, Lin PL. Macrophages and control of granulomatous inflammation in tuberculosis. Mucosal Immunol. 2011;4:271–278.
    1. Gordon S, Martinez FO. Alternative activation of macrophages: Mechanism and functions. Immunity. 2010;32:593–604.
    1. Chan J, Tanaka K, Carroll D, Flynn J, Bloom BR. Effects of nitric oxide synthase inhibitors on murine infection with Mycobacterium tuberculosis. Infect Immun. 1995;63:736–740.
    1. Flynn JL, Scanga CA, Tanaka KE, Chan J. Effects of aminoguanidine on latent murine tuberculosis. J Immunol. 1998;160:1796–1803.
    1. MacMicking JD, North RJ, LaCourse R, Mudgett JS, Shah SK, Nathan CF. Identification of nitric oxide synthase as a protective locus against tuberculosis. Proc Natl Acad Sci USA. 1997;94:5243–5248.
    1. Mattila JT, Ojo OO, Kepka-Lenhart D, et al. Microenvironments in tuberculous granulomas are delineated by distinct populations of macrophage subsets and expression of nitric oxide synthase and arginase isoforms. J Immunol. 2013;191:773–784.
    1. de Cassan SC, Pathan AA, Sander CR, et al. Investigating the induction of vaccine-induced Th17 and regulatory T cells in healthy, Mycobacterium bovis BCG-immunized adults vaccinated with a new tuberculosis vaccine, MVA85A. Clin Vaccine Immunol. 2010;17:1066–1073.
    1. Garcia Jacobo RE, Serrano CJ, Enciso Moreno JA, et al. Analysis of Th1, Th17 and regulatory T cells in tuberculosis case contacts. Cell Immunol. 2014;289:167–173.
    1. Torrado E, Cooper AM. IL-17 and Th17 cells in tuberculosis. Cytokine Growth Factor Rev. 2010;21:455–462.
    1. Ye ZJ, Zhou Q, Du RH, Li X, Huang B, Shi HZ. Imbalance of Th17 cells and regulatory T cells in tuberculous pleural effusion. Clin Vaccine Immunol. 2011;18:1608–1615.
    1. He XY, Xiao L, Chen HB, et al. T regulatory cells and Th1/Th2 cytokines in peripheral blood from tuberculosis patients. Eur J Clin Microbiol Infect Dis. 2010;29:643–650.
    1. Hougardy JM, Place S, Hildebrand M, et al. Regulatory T cells depress immune responses to protective antigens in active tuberculosis. Am J Respir Crit Care Med. 2007;176:409–416.
    1. Lim HJ, Park JS, Cho YJ, et al. CD4(+)FoxP3(+) T regulatory cells in drug-susceptible and multidrug-resistant tuberculosis. Tuberculosis. 2013;93:523–528.
    1. Lowe DM, Redford PS, Wilkinson RJ, O’Garra A, Martineau AR. Neutrophils in tuberculosis: Friend or foe? Trends Immunol. 2012;33:14–25.
    1. Martineau AR, Newton SM, Wilkinson KA, et al. Neutrophil-mediated innate immune resistance to mycobacteria. J Clin Invest. 2007;117:1988–1994.
    1. Christensen D, Mortensen R, Rosenkrands I, Dietrich J, Andersen P. Vaccine-induced Th17 cells are established as resident memory cells in the lung and promote local IgA responses. Mucosal Immunol. 2017;10:260–270.
    1. Khader SA, Bell GK, Pearl JE, et al. IL-23 and IL-17 in the establishment of protective pulmonary CD4+ T cell responses after vaccination and during Mycobacterium tuberculosis challenge. Nat Immunol. 2007;8:369–377.
    1. Khader SA, Cooper AM. IL-23 and IL-17 in tuberculosis. Cytokine. 2008;41:79–83.
    1. Khader SA, Gopal R. IL-17 in protective immunity to intracellular pathogens. Virulence. 2010;1:423–427.
    1. Scott-Browne JP, Shafiani S, Tucker-Heard G, et al. Expansion and function of Foxp3-expressing T regulatory cells during tuberculosis. J Exp Med. 2007;204:2159–2169.
    1. Shafiani S, Tucker-Heard G, Kariyone A, Takatsu K, Urdahl KB. Pathogen-specific regulatory T cells delay the arrival of effector T cells in the lung during early tuberculosis. J Exp Med. 2010;207:1409–1420.
    1. Darrah PA, Patel DT, De Luca PM, et al. Multifunctional TH1 cells define a correlate of vaccine-mediated protection against Leishmania major. Nat Med. 2007;13:843–850.
    1. Macedo AB, Sanchez-Arcila JC, Schubach AO, et al. Multifunctional CD4(+) T cells in patients with American cutaneous leishmaniasis. Clin Exp Immunol. 2012;167:505–513.
    1. Califano D, Sweeney KJ, Le H, et al. Diverting T helper cell trafficking through increased plasticity attenuates autoimmune encephalomyelitis. J Clin Invest. 2014;124:174–187.
    1. Carbo A, Hontecillas R, Kronsteiner B, et al. Systems modeling of molecular mechanisms controlling cytokine-driven CD4+ T cell differentiation and phenotype plasticity. PLoS Comput Biol. 2013;9:e1003027.
    1. Voo KS, Wang YH, Santori FR, et al. Identification of IL-17-producing FOXP3+ regulatory T cells in humans. Proc Natl Acad Sci USA. 2009;106:4793–4798.
    1. Hovhannisyan Z, Treatman J, Littman DR, Mayer L. Characterization of interleukin-17-producing regulatory T cells in inflamed intestinal mucosa from patients with inflammatory bowel diseases. Gastroenterology. 2011;140:957–965.
    1. Morrison PJ, Bending D, Fouser LA, et al. Th17-cell plasticity in Helicobacter hepaticus-induced intestinal inflammation. Mucosal Immunol. 2013;6:1143–1156.
    1. Annunziato F, Cosmi L, Santarlasci V, et al. Phenotypic and functional features of human Th17 cells. J Exp Med. 2007;204:1849–1861.
    1. Wilkinson KA, Wilkinson RJ. Polyfunctional T cells in human tuberculosis. Eur J Immunol. 2010;40:2139–2142.
    1. Mattila JT, Diedrich CR, Lin PL, Phuah J, Flynn JL. Simian immunodeficiency virus-induced changes in T cell cytokine responses in cynomolgus macaques with latent Mycobacterium tuberculosis infection are associated with timing of reactivation. J Immunol. 2011;186:3527–3537.
    1. Qiu Z, Zhang M, Zhu Y, et al. Multifunctional CD4 T cell responses in patients with active tuberculosis. Sci Rep. 2012;2:216.
    1. Harari A, Rozot V, Bellutti Enders F, et al. Dominant TNF-alpha+ Mycobacterium tuberculosis-specific CD4+ T cell responses discriminate between latent infection and active disease. Nat Med. 2011;17:372–376.
    1. Beveridge NE, Price DA, Casazza JP, et al. Immunisation with BCG and recombinant MVA85A induces long-lasting, polyfunctional Mycobacterium tuberculosis-specific CD4+ memory T lymphocyte populations. Eur J Immunol. 2007;37:3089–3100.
    1. Burke MA, Morel BF, Oriss TB, Bray J, McCarthy SA, Morel PA. Modeling the proliferative response of T cells to IL-2 and IL-4. Cell Immunol. 1997;178:42–52.
    1. Segel LA, Bar-Or RL. On the role of feedback in promoting conflicting goals of the adaptive immune system. J Immunol. 1999;163:1342–1349.
    1. Yates A, Callard R, Stark J. Combining cytokine signalling with T-bet and GATA-3 regulation in Th1 and Th2 differentiation: A model for cellular decision-making. J Theor Biol. 2004;231:181–196.
    1. Fishman MA, Perelson AS. Th1/Th2 cross regulation. J Theor Biol. 1994;170:25–56.
    1. Perelson AS. Modelling viral and immune system dynamics. Nat Rev Immunol. 2002;2:28–36.
    1. Kirschner D, Pienaar E, Marino S, Linderman JJ. A review of computational and mathematical modeling contributions to our understanding of Mycobacterium tuberculosis within-host infection and treatment. Curr Opin Syst Biol. 2017;3:170–185.
    1. Wigginton JE, Kirschner D. A model to predict cell-mediated immune regulatory mechanisms during human infection with Mycobacterium tuberculosis. J Immunol. 2001;166:1951–1967.
    1. Murray PJ, Young RA. Increased antimycobacterial immunity in interleukin-10-deficient mice. Infect Immun. 1999;67:3087–3095.
    1. Marino S, Kirschner DE. The human immune response to Mycobacterium tuberculosis in lung and lymph node. J Theor Biol. 2004;227:463–486.
    1. Marino S, Pawar S, Fuller CL, Reinhart TA, Flynn JL, Kirschner DE. Dendritic cell trafficking and antigen presentation in the human immune response to Mycobacterium tuberculosis. J Immunol. 2004;173:494–506.
    1. Marino S, Gideon HP, Gong C, et al. Computational and empirical studies predict Mycobacterium tuberculosis-specific T cells as a biomarker for infection outcome. PLoS Comput Biol. 2016;12:e1004804.
    1. Marino S, Cilfone NA, Mattila JT, Linderman JJ, Flynn JL, Kirschner DE. Macrophage polarization drives granuloma outcome during Mycobacterium tuberculosis infection. Infect Immun. 2015;83:324–338.
    1. Marino S, Myers A, Flynn JL, Kirschner DE. TNF and IL-10 are major factors in modulation of the phagocytic cell environment in lung and lymph node in tuberculosis: A next-generation two-compartmental model. J Theor Biol. 2010;265:586–598.
    1. Myers AJ, Marino S, Kirschner DE, Flynn JL. Inoculation dose of Mycobacterium tuberculosis does not influence priming of T cell responses in lymph nodes. J Immunol. 2013;190:4707–4716.
    1. Marino S, El-Kebir M, Kirschner D. A hybrid multi-compartment model of granuloma formation and T cell priming in Tuberculosis. J Theor Biol. 2011;280:50–62.
    1. Day J, Friedman A, Schlesinger LS. Modeling the immune rheostat of macrophages in the lung in response to infection. Proc Natl Acad Sci USA. 2009;106:11246–11251.
    1. Matzinger P The danger model: A renewed sense of self. Science. 2002;296:301–305.
    1. Lin PL, Coleman T, Carney JP, et al. Radiologic responses in cynomolgous macaques for assessing tuberculosis chemotherapy regimens. Antimicrob Agents Chemother. 2013;57:4237–4244.
    1. Coleman MT, Chen RY, Lee M, et al. PET/CT imaging reveals a therapeutic response to oxazolidinones in macaques and humans with tuberculosis. Sci Transl Med. 2014;6:265–167.
    1. Cilfone NA, Perry CR, Kirschner DE, Linderman JJ. Multi-scale modeling predicts a balance of tumor necrosis factor-alpha and interleukin-10 controls the granuloma environment during Mycobacterium tuberculosis infection. PLoS ONE. 2013;8:e68680.
    1. Fallahi-Sichani M, El-Kebir M, Marino S, Kirschner DE, Linderman JJ. Multiscale computational modeling reveals a critical role for TNF-alpha receptor 1 dynamics in tuberculosis granuloma formation. J Immunol. 2011;186:3472–3483.
    1. Ray JC, Flynn JL, Kirschner DE. Synergy between individual TNF-dependent functions determines granuloma performance for controlling Mycobacterium tuberculosis infection. J Immunol. 2009;182:3706–3717.
    1. Segovia-Juarez JL, Ganguli S, Kirschner D. Identifying control mechanisms of granuloma formation during M. tuberculosis infection using an agent-based model. J Theor Biol. 2004;231:357–376.
    1. Fallahi-Sichani M, Kirschner DE, Linderman JJ. NF-kappaB signaling dynamics play a key role in infection control in tuberculosis. Front Physiol. 2012;3:170.
    1. Pienaar E, Matern WM, Linderman JJ, Bader JS, Kirschner DE. Multiscale model of Mycobacterium tuberculosis infection maps metabolite and gene perturbations to granuloma sterilization predictions. Infect Immun. 2016;84:1650–1669.
    1. Cilfone NA, Ford CB, Marino S, et al. Computational modeling predicts IL-10 control of lesion sterilization by balancing early host immunity-mediated antimicrobial responses with caseation during Mycobacterium tuberculosis infection. J Immunol. 2015;194:664–677.
    1. Kirschner DE, Hunt CA, Marino S, Fallahi-Sichani M, Linderman JJ. Tuneable resolution as a systems biology approach for multi-scale, multi-compartment computational models. Wiley Interdiscip Rev Syst Biol Med. 2014;6:289–309.
    1. Fallahi-Sichani M, Flynn JL, Linderman JJ, Kirschner DE. Differential risk of tuberculosis reactivation among anti-TNF therapies is due to drug binding kinetics and permeability. J Immunol. 2012;188:3169–3178.
    1. Fallahi-Sichani M, Schaller MA, Kirschner DE, Kunkel SL, Linderman JJ. Identification of key processes that control tumor necrosis factor availability in a tuberculosis granuloma. PLoS Comput Biol. 2010;6:e1000778.
    1. Warsinske HC, Pienaar E, Linderman JJ, Mattila JT, Kirschner DE. Deletion of TGF-beta1 increases bacterial clearance by cytotoxic T cells in a tuberculosis granuloma model. Front Immunol. 2017;8:1843.
    1. Fuller CL, Flynn JL, Reinhart TA. In situ study of abundant expression of proinflammatory chemokines and cytokines in pulmonary granulomas that develop in cynomolgus macaques experimentally infected with Mycobacterium tuberculosis. Infect Immun. 2003;71:7023–7034.
    1. Clay H, Volkman HE, Ramakrishnan L. Tumor necrosis factor signaling mediates resistance to mycobacteria by inhibiting bacterial growth and macrophage death. Immunity. 2008;29:283–294.
    1. Algood HM, Lin PL, Flynn JL. Tumor necrosis factor and chemokine interactions in the formation and maintenance of granulomas in tuberculosis. Clin Infect Dis. 2005;41(Suppl 3):S189–S193.
    1. Algood HM, Lin PL, Yankura D, Jones A, Chan J, Flynn JL. TNF influences chemokine expression of macrophages in vitro and that of CD11b+ cells in vivo during Mycobacterium tuberculosis infection. J Immunol. 2004;172:6846–6857.
    1. Lee J, Remold HG, Ieong MH, Kornfeld H. Macrophage apoptosis in response to high intracellular burden of Mycobacterium tuberculosis is mediated by a novel caspase-independent pathway. J Immunol. 2006;176:4267–4274.
    1. Ito T, Schaller M, Hogaboam CM, et al. TLR9 regulates the mycobacteria-elicited pulmonary granulomatous immune response in mice through DC-derived Notch ligand delta-like 4. J Clin Invest. 2009;119:33–46.
    1. Chensue SW, Warmington K, Ruth JH, Lukacs N, Kunkel SL. Mycobacterial and schistosomal antigen-elicited granuloma formation in IFN-γ and IL-4 knockout mice: Analysis of local and regional cytokine and chemokine networks. J Immunol. 1997;159:3565–3573.
    1. Chensue SW, Warmington K, Ruth J, Lincoln P, Kuo MC, Kunkel SL. Cytokine responses during mycobacterial and schistosomal antigen-induced pulmonary granuloma formation. Production of Th1 and Th2 cytokines and relative contribution of tumor necrosis factor. Am J Pathol. 1994;145:1105–1113.
    1. Chensue SW, Ruth JH, Warmington K, Lincoln P, Kunkel SL. In vivo regulation of macrophage IL-12 production during type 1 and type 2 cytokine-mediated granuloma formation. J Immunol. 1995;155:3546–3551.
    1. Warsinske HC, Ashley SL, Linderman JJ, Moore BB, Kirschner DE. Identifying mechanisms of homeostatic signaling in fibroblast differentiation. Bull Math Biol. 2015;77:1556–1582.
    1. Warsinske HC, Wheaton AK, Kim KK, Linderman JJ, Moore BB, Kirschner DE. Computational modeling predicts simultaneous targeting of fibroblasts and epithelial cells is necessary for treatment of pulmonary fibrosis. Front Pharmacol. 2016;7:183.
    1. Warsinske HC, DiFazio RM, Linderman JJ, Flynn JL, Kirschner DE. Identifying mechanisms driving formation of granuloma-associated fibrosis during Mycobacterium tuberculosis infection. J Theor Biol. 2017;429:1–17.
    1. Mayadas TN, Cullere X, Lowell CA. The multifaceted functions of neutrophils. Annu Rev Pathol. 2014;9:181–218.
    1. Lyadova IV. Neutrophils in tuberculosis: Heterogeneity shapes the way? Mediators Inflamm. 2017;2017:8619307.
    1. McCracken JM, Allen LA. Regulation of human neutrophil apoptosis and lifespan in health and disease. J Cell Death. 2014;7:15–23.
    1. Witko-Sarsat V, Rieu P, Descamps-Latscha B, Lesavre P, Halbwachs-Mecarelli L. Neutrophils: Molecules, functions and pathophysiological aspects. Lab Invest. 2000;80:617–653.
    1. Lin PL, Pawar S, Myers A, et al. Early events in Mycobacterium tuberculosis infection in cynomolgus macaques. Infect Immun. 2006;74:3790–3803.
    1. Mattila JT, Maiello P, Sun T, Via LE, Flynn JL. Granzyme B-expressing neutrophils correlate with bacterial load in granulomas from Mycobacterium tuberculosis-infected cynomolgus macaques. Cell Microbiol. 2015;17:1085–1097.
    1. Bru A, Cardona PJ. Mathematical modeling of tuberculosis bacillary counts and cellular populations in the organs of infected mice. PLoS ONE. 2010;5:e12985.
    1. Bieberly J, Ali J. Treatment adherence of the latently infected tuberculosis population (post-Katrina) at Wetmore TB Clinic, New Orleans, USA. Int J Tuberc Lung Dis. 2008;12:1134–1138.
    1. Dartois V The path of anti-tuberculosis drugs: From blood to lesions to mycobacterial cells. Nat Rev Microbiol. 2014;12:159–167.
    1. Kjellsson MC, Via LE, Goh A, et al. Pharmacokinetic evaluation of the penetration of antituberculosis agents in rabbit pulmonary lesions. Antimicrob Agents Chemother. 2012;56:446–457.
    1. Sarathy JP, Zuccotto F, Hsinpin H, et al. Prediction of drug penetration in tuberculosis lesions. ACS Infect Dis. 2016;2:552–563.
    1. Pienaar E, Cilfone NA, Lin PL, et al. A computational tool integrating host immunity with antibiotic dynamics to study tuberculosis treatment. J Theor Biol. 2015;367:166–179.
    1. Prideaux B, Dartois V, Staab D, et al. High-sensitivity MALDI-MRM-MS imaging of moxifloxacin distribution in tuberculosis-infected rabbit lungs and granulomatous lesions. Anal Chem. 2011;83:2112–2118.
    1. Zimmerman M, Lestner J, Prideaux B, et al. Ethambutol partitioning in tuberculous pulmonary lesions explains its clinical efficacy. Antimicrob Agents Chemother. 2017;61:e00924–17.
    1. Pienaar E, Sarathy J, Prideaux B, et al. Comparing efficacies of moxifloxacin, levofloxacin and gatifloxacin in tuberculosis granulomas using a multi-scale systems pharmacology approach. PLoS Comput Biol. 2017;13:e1005650.
    1. Wallis RS, Hafner R. Advancing host-directed therapy for tuberculosis. Nat Rev Immunol. 2015;15:255–263.
    1. Flynn JL, Goldstein MM, Chan J, et al. Tumor necrosis factor-alpha is required in the protective immune response against Mycobacterium tuberculosis in mice. Immunity. 1995;2:561–572.
    1. Wallis RS, Broder M, Wong J, Lee A, Hoq L. Reactivation of latent granulomatous infections by infliximab. Clin Infect Dis. 2005;41(Suppl 3):S194–S198.
    1. Lin PL, Myers A, Smith L, et al. Tumor necrosis factor neutralization results in disseminated disease in acute and latent Mycobacterium tuberculosis infection with normal granuloma structure in a cynomolgus macaque model. Arthritis Rheum. 2010;62:340–350.
    1. Wallis RS, Kyambadde P, Johnson JL, et al. A study of the safety, immunology, virology, and microbiology of adjunctive etanercept in HIV-1-associated tuberculosis. AIDS. 2004;18:257–264.
    1. Mayanja-Kizza H, Jones-Lopez E, Okwera A, et al. Immunoadjuvant prednisolone therapy for HIV-associated tuberculosis: A phase 2 clinical trial in Uganda. J Infect Dis. 2005;191:856–865.
    1. Subbian S, Tsenova L, Holloway J, et al. Adjunctive phosphodiesterase-4 inhibitor therapy improves antibiotic response to pulmonary tuberculosis in a rabbit model. EBioMedicine. 2016;4:104–114.
    1. Subbian S, Tsenova L, O’Brien P, et al. Phosphodiesterase-4 inhibition alters gene expression and improves isoniazid-mediated clearance of Mycobacterium tuberculosis in rabbit lungs. PLoS Pathog. 2011;7:e1002262.
    1. Koo MS, Manca C, Yang G, et al. Phosphodiesterase 4 inhibition reduces innate immunity and improves isoniazid clearance of Mycobacterium tuberculosis in the lungs of infected mice. PLoS ONE. 2011;6:e17091.
    1. Kaye P, Scott P. Leishmaniasis: Complexity at the host-pathogen interface. Nat Rev Microbiol. 2011;9:604–615.
    1. Contreras I, Estrada JA, Guak H, et al. Impact of Leishmania Mexicana infection on dendritic cell signaling and functions. PLoS Negl Trop Dis. 2014;8:e3202.
    1. Maioli TU, Takane E, Arantes RM, Fietto JL, Afonso LC. Immune response induced by New World Leishmania species in C57BL/6 mice. Parasitol Res. 2004;94:207–212.
    1. Mattner F, Magram J, Ferrante J, et al. Genetically resistant mice lacking interleukin-12 are susceptible to infection with Leishmania major and mount a polarized Th2 cell response. Eur J Immunol. 1996;26:1553–1559.
    1. Swihart K, Fruth U, Messmer N, et al. Mice from a genetically resistant background lacking the interferon gamma receptor are susceptible to infection with Leishmania major but mount a polarized T helper cell 1-type CD4+ T cell response. J Exp Med. 1995;181:961–971.
    1. Kopf M, Brombacher F, Kohler G, et al. IL-4-deficient Balb/c mice resist infection with Leishmania major. J Exp Med. 1996;184:1127–1136.
    1. Kaye PM, Beattie L. Lessons from other diseases: Granulomatous inflammation in leishmaniasis. Semin Immunopathol. 2016;38:249–260.
    1. Aoun J, Habib R, Charaffeddine K, Taraif S, Loya A, Khalifeh I. Caseating granulomas in cutaneous leishmaniasis. PLoS Negl Trop Dis. 2014;8:e3255.
    1. De Almeida MC, Moreira HN. A mathematical model of immune response in cutaneous leishmaniasis. J Biol Syst. 2007;15:313–354.
    1. Siewe N, Yakubu AA, Satoskar AR, Friedman A. Immune response to infection by Leishmania: A mathematical model. Math Biosci. 2016;276:28–43.
    1. Albergante L, Timmis J, Beattie L, Kaye PM. A Petri net model of granulomatous inflammation: Implications for IL-10 mediated control of Leishmania donovani infection. PLoS Comput Biol. 2013;9:e1003334.
    1. Ribeiro HAL, Maioli TU, de Freitas LM, Tieri P, Castiglione F. Modeling immune response to leishmania species indicates adenosine as an important inhibitor of Th-cell activation. Front Cell Infect Microbiol. 2017;7:309.
    1. Brown LM. Helicobacter pylori: Epidemiology and routes of transmission. Epidemiol Rev. 2000;22:283–297.
    1. Perry S, de Jong BC, Solnick JV, et al. Infection with Helicobacter pylori is associated with protection against tuberculosis. PLoS ONE. 2010;5:e8804.
    1. Falk GW. The possible role of Helicobacter pylori in GERD. Semin Gastrointest Dis. 2001;12:186–195.
    1. Lord AR, Simms LA, Hanigan K, Sullivan R, Hobson P, Radford-Smith GL. Protective effects of Helicobacter pylori for IBD are related to the cagA-positive strain. Gut. 2018;67:393–394.
    1. Blaser MJ, Chen Y, Reibman J. Does Helicobacter pylori protect against asthma and allergy? Gut. 2008;57:561–567.
    1. Matsushima K, Nagai S. Unraveling the mystery of the hygiene hypothesis through Helicobacter pylori infection. J Clin Invest. 2012;122:801–804.
    1. Wroblewski LE, Peek RM Jr, Wilson KT. Helicobacter pylori and gastric cancer: Factors that modulate disease risk. Clin Microbiol Rev. 2010;23:713–739.
    1. Ferreira AC, Isomoto H, Moriyama M, Fujioka T, Machado JC, Yamaoka Y. Helicobacter and gastric malignancies. Helicobacter. 2008;13(Suppl 1):28–34.
    1. Kusters JG, van Vliet AH, Kuipers EJ. Pathogenesis of Helicobacter pylori infection. Clin Microbiol Rev. 2006;19:449–490.
    1. Dooley CP, Cohen H, Fitzgibbons PL, et al. Prevalence of Helicobacter pylori infection and histologic gastritis in asymptomatic persons. N Engl J Med. 1989;321:1562–1566.
    1. Lindholm C, Quiding-Jarbrink M, Lonroth H, Hamlet A, Svennerholm AM. Local cytokine response in Helicobacter pylori-infected subjects. Infect Immun. 1998;66:5964–5971.
    1. Bodger K, Crabtree JE. Helicobacter pylori and gastric inflammation. Br Med Bull. 1998;54:139–150.
    1. Kivrak Salim D, Sahin M, Koksoy S, Adanir H, Suleymanlar I. Local immune response in Helicobacter pylori infection. Medicine (Baltimore). 2016;95:e3713.
    1. Kirschner DE, Blaser MJ. The dynamics of Helicobacter pylori infection of the human stomach. J Theor Biol. 1995;176:281–290.
    1. Blaser MJ, Kirschner D. Dynamics of Helicobacter pylori colonization in relation to the host response. Proc Natl Acad Sci USA. 1999;96:8359–8364.
    1. Parsonnet J, Friedman GD, Orentreich N, Vogelman H. Risk for gastric cancer in people with CagA positive or CagA negative Helicobacter pylori infection. Gut. 1997;40:297–301.
    1. Bagheri N, Shirzad H, Elahi S, et al. Downregulated regulatory T cell function is associated with increased peptic ulcer in Helicobacter pylori-infection. Microb Pathog. 2017;110:165–175.
    1. Kern KB, Nelson JR, Norman SA, Milander MM, Hilwig RW. Oxygenation and ventilation during cardiopulmonary resuscitation utilizing continuous oxygen delivery via a modified pharyngeal-tracheal lumened airway. Chest. 1992;101:522–529.
    1. Larussa T, Leone I, Suraci E, Imeneo M, Luzza F. Helicobacter pylori and T helper cells: Mechanisms of immune escape and tolerance. J Immunol Res. 2015;2015:981328.
    1. Chen MG. Progress and problems in schistosomiasis control in China. Trop Med Parasitol. 1989;40:174–176.
    1. Carbo A, Bassaganya-Riera J, Pedragosa M, et al. Predictive computational modeling of the mucosal immune responses during Helicobacter pylori infection. PLoS ONE. 2013;8:e73365.
    1. Sriskandan S, Altmann DM. The immunology of sepsis. J Pathol. 2008;214:211–223.
    1. Bone RC, Balk RA, Cerra FB, et al. Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. The ACCP/SCCM Consensus Conference Committee. American College of Chest Physicians/Society of Critical Care Medicine. Chest. 1992;101:1644–1655.
    1. Cohen J The immunopathogenesis of sepsis. Nature. 2002;420: 885–891.
    1. Schulte W, Bernhagen J, Bucala R. Cytokines in sepsis: Potent immunoregulators and potential therapeutic targets–an updated view. Mediators Inflamm. 2013;2013:165974.
    1. Vodovotz Y, Billiar TR. In silico modeling: Methods and applications to trauma and sepsis. Crit Care Med. 2013;41:2008–2014.
    1. Yang H, Lee EK, Wu CJ, Shi Z, Ben-Arieh D, Simpson SQ. Mathematical modeling of innate immunity responses of sepsis: Modeling and computational studies In: Yang H, Lee EK, eds. Healthcare Analytics: From Data to Knowledge to Healthcare Improvement. 1st ed. Hoboken, NJ: John Wiley & Sons, Inc; 2016:221–259.
    1. An G In silico experiments of existing and hypothetical cytokine-directed clinical trials using agent-based modeling. Crit Care Med. 2004;32:2050–2060.
    1. Delano MJ, Ward PA. The immune system’s role in sepsis progression, resolution, and long-term outcome. Immunol Rev. 2016;274:330–353.
    1. Hotchkiss RS, Monneret G, Payen D. Sepsis-induced immunosuppression: From cellular dysfunctions to immunotherapy. Nat Rev Immunol. 2013;13:862–874.
    1. Delano MJ, Moldawer LL. Magic bullets and surrogate biomarkers circa 2009. Crit Care Med. 2009;37:1796–1798.
    1. Bosmann M, Ward PA. The inflammatory response in sepsis. Trends Immunol. 2013;34:129–136.
    1. Hutchins NA, Unsinger J, Hotchkiss RS, Ayala A. The new normal: Immunomodulatory agents against sepsis immune suppression. Trends Mol Med. 2014;20:224–233.
    1. Luan YY, Dong N, Xie M, Xiao XZ, Yao YM. The significance and regulatory mechanisms of innate immune cells in the development of sepsis. J Interferon Cytokine Res. 2014;34:2–15.
    1. Rimmele T, Payen D, Cantaluppi V, et al. Immune cell phenotype and function in sepsis. Shock. 2016;45:282–291.
    1. Averhoff FM, Glass N, Holtzman D. Global burden of hepatitis C: Considerations for healthcare providers in the United States. Clin Infect Dis. 2012;55(Suppl 1):S10–S15.
    1. Kim AY, Onofrey S, Church DR. An epidemiologic update on hepatitis C infection in persons living with or at risk of HIV infection. J Infect Dis. 2013;207(Suppl 1):S1–S6.
    1. Boltjes A, Movita D, Boonstra A, Woltman AM. The role of Kupffer cells in hepatitis B and hepatitis C virus infections. J Hepatol. 2014;61:660–671.
    1. Saha B, Szabo G. Innate immune cell networking in hepatitis C virus infection. J Leukoc Biol. 2014;96:757–766.
    1. Yoon JC, Yang CM, Song Y, Lee JM. Natural killer cells in hepatitis C: Current progress. World J Gastroenterol. 2016;22:1449–1460.
    1. Tian Z, Chen Y, Gao B. Natural killer cells in liver disease. Hepatology. 2013;57:1654–1662.
    1. Nguyen-Lefebvre AT, Horuzsko A. Kupffer cell metabolism and function. J Enzymol Metabol. 2015;1:101.
    1. Mengshol JA, Golden-Mason L, Arikawa T, et al. A crucial role for Kupffer cell-derived galectin-9 in regulation of T cell immunity in hepatitis C infection. PLoS ONE. 2010;5:e9504.
    1. Hartling HJ, Gaardbo JC, Ronit A, et al. CD4(+) and CD8(+) regulatory T cells (Tregs) are elevated and display an active phenotype in patients with chronic HCV mono-infection and HIV/HCV co-infection. Scand J Immunol. 2012;76:294–305.
    1. Barjon C, Dahlqvist G, Calmus Y, Conti F. Role of regulatory T-cells during hepatitis C infection: From the acute phase to post-transplantation recurrence. Digest Liver Dis. 2015;47:913–917.
    1. Terilli RR, Cox AL. Immunity and hepatitis C: A review. Curr HIV/AIDS Rep. 2013;10:51–58.
    1. Gremion C, Cerny A. Hepatitis C virus and the immune system: A concise review. Rev Med Virol. 2005;15:235–268.
    1. Burg D, Rong L, Neumann AU, Dahari H. Mathematical modeling of viral kinetics under immune control during primary HIV-1 infection. J Theor Biol. 2009;259:751–759.
    1. Obaid A, Ahmad J, Naz A, et al. Modeling and analysis of innate immune responses induced by the host cells against hepatitis C virus infection. Integr Biol (Camb). 2015;7:544–559.
    1. Osburn WO, Levine JS, Chattergoon MA, Thomas DL, Cox AL. Anti-inflammatory cytokines, pro-fibrogenic chemokines and persistence of acute HCV infection. J Viral Hepatitis. 2013;20:404–413.
    1. Wiesner DL, Klein BS. The balance between immunity and inflammation. Science. 2017;357:973–974.
    1. Martin SJ. Oncogene-induced autophagy and the Goldilocks principle. Autophagy. 2011;7:922–923.
    1. Monin L, Khader SA. Chemokines in tuberculosis: The good, the bad and the ugly. Semin Immunol. 2014;26:552–558.
    1. Slight SR, Khader SA. Chemokines shape the immune responses to tuberculosis. Cytokine Growth Factor Rev. 2013;24:105–113.
    1. Eizenberg-Magar I, Rimer J, Zaretsky I, Lara-Astiaso D, Reich-Zeliger S, Friedman N. Diverse continuum of CD4(+) T-cell states is determined by hierarchical additive integration of cytokine signals. Proc Natl Acad Sci USA. 2017;114:E6447–E6456.
    1. Fang M, Xie H, Dougan SK, Ploegh H, van Oudenaarden A. Stochastic cytokine expression induces mixed T helper cell States. PLoS Biol. 2013;11:e1001618.
    1. Lin PL, Dartois V, Johnston PJ, et al. Metronidazole prevents reactivation of latent Mycobacterium tuberculosis infection in macaques. Proc Natl Acad Sci USA. 2012;109:14188–14193.
    1. Via LE, Weiner DM, Schimel D, et al. Differential virulence and disease progression following Mycobacterium tuberculosis complex infection of the common marmoset (Callithrix jacchus). Infect Immun. 2013;81:2909–2919.
    1. Stanton P, Richards S, Reeves J, et al. Epidermal growth factor receptor expression by human squamous cell carcinomas of the head and neck, cell lines and xenografts. Br J Cancer. 1994;70:427–433.
    1. Corti A, Gasparri A, Sacchi A, et al. Tumor targeting with biotinylated tumor necrosis factor alpha: Structure-activity relationships and mechanism of action on avidin pretargeted tumor cells. Cancer Res. 1998;58:3866–3872.
    1. Zimmermann VS, Bondanza A, Monno A, Rovere-Querini P, Corti A, Manfredi AA. TNF-alpha coupled to membrane of apoptotic cells favors the cross-priming to melanoma antigens. J Immunol. 2004;172:2643–2650.
    1. Desnoyers L, Simonette RA, Vandlen RL, Fendly BM. Novel non-isotopic method for the localization of receptors in tissue sections. J Histochem Cytochem. 2001;49:1509–1518.
    1. Wiley HS, Cunningham DD. The endocytotic rate constant. A cellular parameter for quantitating receptor-mediated endocytosis. J Biol Chem. 1982;257:4222–4229.
    1. Cilfone NA. Understanding and Treating Mycobacterium tuberculosis Infection: A Multi-Scale Modeling Approach: Chemical Engineering. Ann Arbor, MI: University of Michigan; 2014.
    1. Pienaar E, Dartois V, Linderman JJ, Kirschner DE. In silico evaluation and exploration of antibiotic tuberculosis treatment regimens. BMC Syst Biol. 2015;9:79.

Source: PubMed

3
订阅