The pathophysiology of tuberculosis (TB) is linked to the ability of the causative organism, Mycobacterium tuberculosis (M.tb), to grow within human macrophages. Lung myeloid dendritic cells are a newly recognized reservoir of M.tb during lung infection. M.tb metabolism in macrophages and dendritic cells may differ. Iron (Fe) acquisition is critical for M.tb growth. In vivo, most extracellular Fe is chelated to transferrin (TF) and lactoferrin (LF), decreasing microbial access to Fe. We find that M.tb in human macrophages can acquire Fe from TF and LF, as well as the macrophage cytoplasm. Fe uptake from extracellular TF and LF may involve Fe moving into the cytoplasm. Some (e.g. hereditary hemochromatosis) conditions that lower intracellular Fe in macrophages, decrease M.tb Fe uptake. Whether other factors that regulate macrophage Fe metabolism (e.g. ferroportin, heme oxygenase 1 (HO-1), hepcidin) impact M.tb Fe acquisition is unknown. Similarly unknown is whether Fe acquisition by M.tb growing in dendritic cells is different from that occurring in macrophages. We hypothesize that: 1) M.tb acquires Fe from an intracellular Fe pool(s) in macrophages and dendritic cells; 2) acquisition of extracellular Fe by M.tb involves initial Fe movement into this pool; 3) the kinetics/route of M.tb Fe uptake varies with the extracellular Fe chelate and host cell type (macrophage vs. dendritic cell) involved; 4) genetic or physiological factors that modulate macrophage and/or dendritic cell cytoplasmic Fe alter Fe available for use by M.tb; and 4) administration of Ga, which disrupts M.tb Fe-dependent metabolism could prove to be a novel therapy for TB. In order to test these hypotheses we will pursue the following specific aims: 1. Delineate and compare the ability of M.tb residing within human macrophages and myeloid dendritic cells to: A) acquire Fe from physiologic extracellular chelates (e.g. TF and LF) and from cytoplasmic sites in the macrophage and dendritic cell; and B) to what extent Fe acquisition from extracellular chelates involves initial Fe movement into these same cytoplasmic sites. 2. Determine: A) if M.tb modulates macrophage Fe metabolism, which in turn alters Fe acquisition and growth by the bacteria; and B) the role of key components of macrophage Fe metabolism (e.g. ferroportin, HFE, HO-1, hepcidin) on M.tb Fe acquisition and growth. 3. Determine the efficacy of Ga as a therapeutic agent against M.tb using a murine model of M.tb infection. If positive results are seen, further ascertain the mechanism(s) of Ga's anti-tuberculous activity. The proposed studies use human macrophages and dendritic cells along with fully virulent M.tb strains to accomplish these aims. Experimental methods employed include: measuring uptake of 59Fe, siRNA, confocal microscopy, immunoblot analysis, RT-PCR, and ELISA and use of a murine model of pulmonary TB. Insight into M.tb Fe metabolism could lead to novel treatments that would benefit veterans.

Public Health Relevance

Project Narrative - Relevance to VA Mission: Veterans are at high risk for pulmonary TB. There is a growing problem in treating tuberculosis in that the causative organism, Myocobacterium tuberculosis, is becoming increasingly resistant to available antibiotics. The ability to acquire iron (Fe) from the infected host is required for M. tuberculosis to be able to grow and therefore cause infection. Insight into M. tuberculosis Fe metabolism could lead to development of novel approaches to interfere with the organism's ability to acquire Fe treatments that could lead to the development of new kinds of antibiotics. This application will explore one potential mechanism of doing this using gallium. Thus, increased understanding of how M. tuberculosis acquires and uses Fe during infection could lead to the development of new antibiotics to be used in treating TB, something that would be of great benefit to veterans.

Agency
National Institute of Health (NIH)
Institute
Veterans Affairs (VA)
Type
Non-HHS Research Projects (I01)
Project #
5I01CX000102-02
Application #
7784541
Study Section
Infectious Diseases B (INFB)
Project Start
2009-04-01
Project End
2013-09-30
Budget Start
2010-04-01
Budget End
2011-09-30
Support Year
2
Fiscal Year
2011
Total Cost
Indirect Cost
Name
Cincinnati VA Medical Center Research
Department
Type
DUNS #
827658092
City
Cincinnati
State
OH
Country
United States
Zip Code
45220
Choi, Seoung-Ryoung; Britigan, Bradley E; Switzer, Barbara et al. (2018) In Vitro Efficacy of Free and Nanoparticle Formulations of Gallium(III) meso-Tetraphenylporphyrine against Mycobacterium avium and Mycobacterium abscessus and Gallium Biodistribution in Mice. Mol Pharm 15:1215-1225
Choi, Seoung-Ryoung; Britigan, Bradley E; Moran, David M et al. (2017) Gallium nanoparticles facilitate phagosome maturation and inhibit growth of virulent Mycobacterium tuberculosis in macrophages. PLoS One 12:e0177987
Pasula, Rajamouli; Martin 2nd, William J; Kesavalu, Banu Rekha et al. (2017) Passive transfer of interferon-? over-expressing macrophages enhances resistance of SCID mice to Mycobacterium tuberculosis infection. Cytokine 95:70-79
Abdalla, Maher Y; Hoke, Traci; Seravalli, Javier et al. (2017) Pseudomonas Quinolone Signal Induces Oxidative Stress and Inhibits Heme Oxygenase-1 Expression in Lung Epithelial Cells. Infect Immun 85:
Choi, Seoung-Ryoung; Britigan, Bradley E; Narayanasamy, Prabagaran (2017) Ga(III) Nanoparticles Inhibit Growth of both Mycobacterium tuberculosis and HIV and Release of Interleukin-6 (IL-6) and IL-8 in Coinfected Macrophages. Antimicrob Agents Chemother 61:
Reingle Gonzalez, Jennifer M; Cannell, M Brad; Jetelina, Katelyn K et al. (2016) Barriers in detecting elder abuse among emergency medical technicians. BMC Emerg Med 16:36
Cannell, M Brad; Jetelina, Katelyn K; Zavadsky, Matt et al. (2016) Towards the development of a screening tool to enhance the detection of elder abuse and neglect by emergency medical technicians (EMTs): a qualitative study. BMC Emerg Med 16:19