101
|
Kearney S, Delgado C, Lenz LL. Differential effects of type I and II interferons on myeloid cells and resistance to intracellular bacterial infections. Immunol Res 2013; 55:187-200. [PMID: 22983898 DOI: 10.1007/s12026-012-8362-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The type I and II interferons (IFNs) play important roles in regulating immune responses during viral and bacterial infections and in the context of autoimmune and neoplastic diseases. These two IFN types bind to distinct cell surface receptors that are expressed by nearly all cells to trigger signal transduction events and elicit diverse cellular responses. In some cases, type I and II IFNs trigger similar cellular responses, while in other cases, the IFNs have unique or antagonistic effects on host cells. Negative regulators of IFN signaling also modulate cellular responses to the IFNs and play important roles in maintaining immunological homeostasis. In this review, we provide an overview of how IFNs stimulate cellular responses. We discuss the disparate effects of type I and II IFNs on host resistance to certain intracellular bacterial infections and provide an overview of models that have been proposed to account for these disparate effects. Mechanisms of antagonistic cross talk between type I and II IFNs are also introduced.
Collapse
Affiliation(s)
- Staci Kearney
- Integrated Department of Immunology, University of Colorado School of Medicine, Aurora, CO, USA
| | | | | |
Collapse
|
102
|
Ghorpade DS, Holla S, Sinha AY, Alagesan SK, Balaji KN. Nitric oxide and KLF4 protein epigenetically modify class II transactivator to repress major histocompatibility complex II expression during Mycobacterium bovis bacillus Calmette-Guerin infection. J Biol Chem 2013; 288:20592-606. [PMID: 23733190 DOI: 10.1074/jbc.m113.472183] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Pathogenic mycobacteria employ several immune evasion strategies such as inhibition of class II transactivator (CIITA) and MHC-II expression, to survive and persist in host macrophages. However, precise roles for specific signaling components executing down-regulation of CIITA/MHC-II have not been adequately addressed. Here, we demonstrate that Mycobacterium bovis bacillus Calmette-Guérin (BCG)-mediated TLR2 signaling-induced iNOS/NO expression is obligatory for the suppression of IFN-γ-induced CIITA/MHC-II functions. Significantly, NOTCH/PKC/MAPK-triggered signaling cross-talk was found critical for iNOS/NO production. NO responsive recruitment of a bifunctional transcription factor, KLF4, to the promoter of CIITA during M. bovis BCG infection of macrophages was essential to orchestrate the epigenetic modifications mediated by histone methyltransferase EZH2 or miR-150 and thus calibrate CIITA/MHC-II expression. NO-dependent KLF4 regulated the processing and presentation of ovalbumin by infected macrophages to reactive T cells. Altogether, our study delineates a novel role for iNOS/NO/KLF4 in dictating the mycobacterial capacity to inhibit CIITA/MHC-II-mediated antigen presentation by infected macrophages and thereby elude immune surveillance.
Collapse
Affiliation(s)
- Devram Sampat Ghorpade
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, India
| | | | | | | | | |
Collapse
|
103
|
Alteration of human macrophages microRNA expression profile upon infection with Mycobacterium tuberculosis. Int J Mycobacteriol 2013; 2:128-34. [PMID: 26785980 DOI: 10.1016/j.ijmyco.2013.04.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 04/20/2013] [Accepted: 04/22/2013] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Mycobacterium tuberculosis (Mtb) has evolved multiple mechanisms to manipulate its cellular niche for its own advantage. Many efforts have been made to understand basal mechanisms of mycobacterial infections. However, the underlying molecular regulation is not fully understood. Recently, a new class of non-coding, small RNAs, called microRNAs (miRNAs), has emerged as important regulators in biological processes, and their involvement in mycobacterial infection has been identified, thus opening a new field of research. METHODS This study aimed to determine by TaqMan Low Density Array the host genome-wide miRNA expression profile of primary human monocyte-derived macrophages (MDM) infected with two members of the Mtb complex: virulent Mtb H37Rv and the non-virulent vaccine strain Mycobacterium bovis Bacillus Calmette-Guerin (BCG) in comparison with chemically-inactivated Mtb bacilli. RESULTS The findings of this study showed that infection of MDM with H37Rv or BCG results in a signature of miRNA expression mostly overlapping between the two mycobacteria. A substantially different signature emerged from infection with killed virulent bacilli, suggesting an active influence of live intracellular bacteria on cellular miRNA metabolism. Specifically, Mtb induced miRNA signature is composed of miRNAs well established in immune regulation, miR-155 and miR-146a, as well as a set of miRNAs newly associated with Mtb infection: miR-145, miR-222(∗), miR-27a and miR-27b. All of these miRNAs are predicted to target important immune-related genes. CONCLUSIONS This study signifies the miRNA host response upon intracellular mycobacterial infection in macrophages, providing new aspects of regulation in host-pathogen interactions, at post-transcriptional levels.
Collapse
|
104
|
Bell C, English L, Boulais J, Chemali M, Caron-Lizotte O, Desjardins M, Thibault P. Quantitative proteomics reveals the induction of mitophagy in tumor necrosis factor-α-activated (TNFα) macrophages. Mol Cell Proteomics 2013; 12:2394-407. [PMID: 23674617 DOI: 10.1074/mcp.m112.025775] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Macrophages play an important role in innate and adaptive immunity as professional phagocytes capable of internalizing and degrading pathogens to derive antigens for presentation to T cells. They also produce pro-inflammatory cytokines such as tumor necrosis factor alpha (TNF-α) that mediate local and systemic responses and direct the development of adaptive immunity. The present work describes the use of label-free quantitative proteomics to profile the dynamic changes of proteins from resting and TNF-α-activated mouse macrophages. These analyses revealed that TNF-α activation of macrophages led to the down-regulation of mitochondrial proteins and the differential regulation of several proteins involved in vesicle trafficking and immune response. Importantly, we found that the down-regulation of mitochondria proteins occurred through mitophagy and was specific to TNF-α, as other cytokines such as IL-1β and IFN-γ had no effect on mitochondria degradation. Furthermore, using a novel antigen presentation system, we observed that the induction of mitophagy by TNF-α enabled the processing and presentation of mitochondrial antigens at the cell surface by MHC class I molecules. These findings highlight an unsuspected role of TNF-α in mitophagy and expanded our understanding of the mechanisms responsible for MHC presentation of self-antigens.
Collapse
Affiliation(s)
- Christina Bell
- Institute for Research in Immunology and Cancer, Université de Montréal, P.O. Box 6128, Station Centre-ville, Montréal, Québec, Canada H3C 3J7
| | | | | | | | | | | | | |
Collapse
|
105
|
Spindle-shaped CD163+ rosetting macrophages replace CD4+ T-cells in HIV-related classical Hodgkin lymphoma. Mod Pathol 2013; 26:648-57. [PMID: 23307058 DOI: 10.1038/modpathol.2012.217] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Combination antiretroviral therapy is highly effective in HIV infection, leading to decreased incidences of AIDS-defining neoplasms. However, HIV patients still have a 10-fold increased risk of developing classical Hodgkin lymphoma compared with the general population. As Hodgkin- and Reed-Sternberg cells represent only a minority in the tumor infiltrate, the aim of the present study was to characterize the microenvironment of HIV-related classical Hodgkin lymphoma and compare it with classical Hodgkin lymphoma cases of immunocompetent individuals. The major morphologic differences were the presence of necrotic foci and the absence of epithelioid cell formation in HIV-related Hodgkin lymphoma. We observed a significantly decreased number of CD4+ T-cells and a significantly increased number of CD163+ macrophages in HIV-related Hodgkin lymphoma. Cases exhibiting a 'sarcomatoid' pattern of the reactive infiltrate exhibited significantly greater numbers of macrophages, associating the 'sarcomatoid' pattern to the presence of spindle-shaped macrophages. Whereas, rosetting of CD4+ T-cells around Hodgkin- and Reed-Sternberg cells was frequently observed in classical Hodgkin lymphoma in immunocompetent persons; rosetting in a subset of HIV-related Hodgkin lymphoma cases appeared to involve cytoplasmic protrusions of spindle-shaped macrophages. HIV-related Hodgkin lymphoma, therefore, is characterized by unique morphologic features, which should be recognized by pathologists.
Collapse
|
106
|
González-Perilli L, Álvarez MN, Prolo C, Radi R, Rubbo H, Trostchansky A. Nitroarachidonic acid prevents NADPH oxidase assembly and superoxide radical production in activated macrophages. Free Radic Biol Med 2013; 58:126-33. [PMID: 23318789 PMCID: PMC3622795 DOI: 10.1016/j.freeradbiomed.2012.12.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Revised: 12/03/2012] [Accepted: 12/21/2012] [Indexed: 11/29/2022]
Abstract
Nitration of arachidonic acid (AA) to nitroarachidonic acid (AANO2) leads to anti-inflammatory intracellular activities during macrophage activation. However, less is known about the capacity of AANO2 to regulate the production of reactive oxygen species under proinflammatory conditions. One of the immediate responses upon macrophage activation involves the production of superoxide radical (O2(•-)) due to the NADPH-dependent univalent reduction of oxygen to O2(•-) by the phagocytic NADPH oxidase isoform (NOX2), the activity of NOX2 being the main source of O2(•-) in monocytes/macrophages. Because the NOX2 and AA pathways are connected, we propose that AANO2 can modulate macrophage activation by inhibiting O2(•-) formation by NOX2. When macrophages were activated in the presence of AANO2, a significant inhibition of NOX2 activity was observed as evaluated by cytochrome c reduction, luminol chemiluminescence, Amplex red fluorescence, and flow cytometry; this process also occurs under physiological mimic conditions within the phagosomes. AANO2 decreased O2(•-) production in a dose- (IC50=4.1±1.8 μM AANO2) and time-dependent manner. The observed inhibition was not due to a decreased phosphorylation of the cytosolic subunits (e.g., p40(phox) and p47(phox)), as analyzed by immunoprecipitation and Western blot. However, a reduction in the migration to the membrane of p47(phox) was obtained, suggesting that the protective actions involve the prevention of the correct assembly of the active enzyme in the membrane. Finally, the observed in vitro effects were confirmed in an in vivo inflammatory model, in which subcutaneous injection of AANO2 was able to decrease NOX2 activity in macrophages from thioglycolate-treated mice.
Collapse
Affiliation(s)
| | | | | | | | | | - Andrés Trostchansky
- Address correspondence to: Andrés Trostchansky, Ph.D., Departamento de Bioquímica, Facultad de Medicina, Avda. Gral. Flores 2125, C.P. 11800, Montevideo, Uruguay; Phone: (598)-2924 9562; Fax: (598)-2924 9563;
| |
Collapse
|
107
|
HE ZONGLIN, DU FAWANG, DU XIANZHI. The viable Mycobacterium tuberculosis H37Ra strain induces a stronger mouse macrophage response compared to the heat-inactivated H37Rv strain. Mol Med Rep 2013; 7:1597-602. [PMID: 23483126 DOI: 10.3892/mmr.2013.1363] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2012] [Accepted: 02/28/2013] [Indexed: 11/05/2022] Open
|
108
|
Salamon H, Qiao Y, Cheng JC, Yamaguchi KD, Soteropoulos P, Weiden M, Gennaro ML, Pine R. Evidence for postinitiation regulation of mRNA biogenesis in tuberculosis. THE JOURNAL OF IMMUNOLOGY 2013; 190:2747-55. [PMID: 23378427 DOI: 10.4049/jimmunol.1202185] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Mycobacterium tuberculosis infection alters macrophage gene expression and macrophage response to IFN-γ, a critical host defense cytokine. However, regulation of these changes is poorly understood. We report discordance of changes in nascent transcript and total nuclear RNA abundance for the transcription factors STAT1 and IRF1, together with lack of effect on their RNA half-lives, in human THP-1 cells infected with M. tuberculosis and stimulated with IFN-γ. The results indicate that negative postinitiation regulation of mRNA biogenesis limits the expression of these factors, which mediate host defense against M. tuberculosis through the cellular response to IFN-γ. Consistent with the results for STAT1 and IRF1, transcriptome analysis reveals downregulation of postinitiation mRNA biogenesis processes and pathways by infection, with and without IFN-γ stimulation. Clinical relevance for regulation of postinitiation mRNA biogenesis is demonstrated by studies of donor samples showing that postinitiation mRNA biogenesis pathways are repressed in latent tuberculosis infection compared with cured disease and in active tuberculosis compared with ongoing treatment or with latent tuberculosis. For active disease and latent infection donors from two populations (London, U.K., and The Gambia), each analyzed using a different platform, pathway-related gene expression differences were highly correlated, demonstrating substantial specificity in the effect. Collectively, the molecular and bioinformatic analyses point toward downregulation of postinitiation mRNA biogenesis pathways as a means by which M. tuberculosis infection limits expression of immunologically essential transcription factors. Thus, negative regulation of postinitiation mRNA biogenesis can constrain the macrophage response to infection and overall host defense against tuberculosis.
Collapse
Affiliation(s)
- Hugh Salamon
- Knowledge Synthesis, Inc., Berkeley, CA 94716, USA
| | | | | | | | | | | | | | | |
Collapse
|
109
|
Schwartz YS, Svistelnik AV. Functional phenotypes of macrophages and the M1-M2 polarization concept. Part I. Proinflammatory phenotype. BIOCHEMISTRY (MOSCOW) 2013; 77:246-60. [PMID: 22803942 DOI: 10.1134/s0006297912030030] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Current concepts concerning the main functional phenotypes of mononuclear phagocytes are systematized, molecular mechanisms of their formation are considered, and the functional polarization concept of macrophages is critically analyzed. Mechanisms of macrophage priming activation mediated by pattern recognition receptors TLR, NLR, RLR, and CLR are described, and the features of each phenotype acquired via various pattern recognition receptors are emphasized. It is concluded that there is a huge variety of proinflammatory phenotypes from highly to poorly polarized ones. Thus the widespread notion of "classical activation" of macrophage concerns just a particular case of proinflammatory phenotype formation.
Collapse
Affiliation(s)
- Y Sh Schwartz
- Research Institute of Internal Medicine, Siberian Branch of the Russian Academy of Medical Sciences, Novosibirsk, 630089, Russia.
| | | |
Collapse
|
110
|
Lugo-Villarino G, Hudrisier D, Benard A, Neyrolles O. Emerging trends in the formation and function of tuberculosis granulomas. Front Immunol 2013; 3:405. [PMID: 23308074 PMCID: PMC3538282 DOI: 10.3389/fimmu.2012.00405] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Accepted: 12/15/2012] [Indexed: 11/13/2022] Open
Abstract
The granuloma is an elaborated aggregate of immune cells found in non-infectious as well as infectious diseases. It is a hallmark of tuberculosis (TB). Predominantly thought as a host-driven strategy to constrain the bacilli and prevent dissemination, recent discoveries indicate the granuloma can also be modulated into an efficient tool to promote microbial pathogenesis. The aim of future studies will certainly focus on better characterization of the mechanisms driving the modulation of the granuloma functions. Here, we provide unique perspectives from both the innate and adaptive immune system in the formation and the role of the TB granuloma. As macrophages (Mϕs) comprise the bulk of granulomas, we highlight the emerging concept of Mϕ polarization and its potential impact in the microbicide response, and other activities, that may ultimately shape the fate of granulomas. Alternatively, we shed light on the ability of B-cells to influence inflammatory status within the granuloma.
Collapse
Affiliation(s)
- Geanncarlo Lugo-Villarino
- CNRS, Institut de Pharmacologie et de Biologie Structurale Toulouse, France ; Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, Université Paul Sabatier Toulouse, France
| | | | | | | |
Collapse
|
111
|
Shepelkova G, Pommerenke C, Alberts R, Geffers R, Evstifeev V, Apt A, Schughart K, Wilk E. Analysis of the lung transcriptome in Mycobacterium tuberculosis-infected mice reveals major differences in immune response pathways between TB-susceptible and resistant hosts. Tuberculosis (Edinb) 2012; 93:263-9. [PMID: 23276693 DOI: 10.1016/j.tube.2012.11.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Revised: 11/07/2012] [Accepted: 11/28/2012] [Indexed: 10/27/2022]
Abstract
Using whole genome microarrays, we compared changes in gene expression patterns in the lungs of TB-resistant A/Sn and TB-susceptible I/St mice at day 14 following infection with Mycobacterium tuberculosis H37Rv. Analyses of differentially expressed genes for representation of gene ontology terms and activation of regulatory pathways revealed interstrain differences in antigen presentation, NK, T and B cell activation pathways. In general, resistant A/Sn mice exhibited a more complex pattern and stronger activation of host defense pathways compared to the TB-susceptible I/St mouse strain. In addition, in I/St mice elevated activation of genes involved in neutrophil response was observed and confirmed by quantitative RT-PCR and histopathology. Furthermore, a specific post infection upregulation of cysteine protease inhibitors was found in susceptible I/St mice.
Collapse
Affiliation(s)
- Galina Shepelkova
- Laboratory for Immunogenetics, Central Institute for Tuberculosis, Moscow, Russia.
| | | | | | | | | | | | | | | |
Collapse
|
112
|
Opländer C, Suschek CV. The role of photolabile dermal nitric oxide derivates in ultraviolet radiation (UVR)-induced cell death. Int J Mol Sci 2012; 14:191-204. [PMID: 23344028 PMCID: PMC3565258 DOI: 10.3390/ijms14010191] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Revised: 12/11/2012] [Accepted: 12/12/2012] [Indexed: 01/22/2023] Open
Abstract
Human skin is exposed to solar ultraviolet radiation comprising UVB (280–315 nm) and UVA (315–400 nm) on a daily basis. Within the last two decades, the molecular and cellular response to UVA/UVB and the possible effects on human health have been investigated extensively. It is generally accepted that the mutagenic and carcinogenic properties of UVB is due to the direct interaction with DNA. On the other hand, by interaction with non-DNA chromophores as endogenous photosensitizers, UVA induces formation of reactive oxygen species (ROS), which play a pivotal role as mediators of UVA-induced injuries in human skin. This review gives a short overview about relevant findings concerning the molecular mechanisms underlying UVA/UVB-induced cell death. Furthermore, we will highlight the potential role of cutaneous antioxidants and photolabile nitric oxide derivates (NODs) in skin physiology. UVA-induced decomposition of the NODs, like nitrite, leads not only to non-enzymatic formation of nitric oxide (NO), but also to toxic reactive nitrogen species (RNS), like peroxynitrite. Whereas under antioxidative conditions the generation of protective amounts of NO is favored, under oxidative conditions, less injurious reactive nitrogen species are generated, which may enhance UVA-induced cell death.
Collapse
Affiliation(s)
- Christian Opländer
- Department of Plastic and Reconstructive Surgery, Hand Surgery, and Burn Center, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, D-52074 Aachen, Germany
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +49-241-80-35271; Fax: +49-241-80-82448
| | - Christoph V. Suschek
- Department of Trauma and Hand Surgery, Medical Faculty of the Heinrich-Heine-University, 40225 Düsseldorf, Germany; E-Mail:
| |
Collapse
|
113
|
Human macrophage response to L. (Viannia) panamensis: microarray evidence for an early inflammatory response. PLoS Negl Trop Dis 2012; 6:e1866. [PMID: 23145196 PMCID: PMC3493378 DOI: 10.1371/journal.pntd.0001866] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2011] [Accepted: 09/01/2012] [Indexed: 11/19/2022] Open
Abstract
Background Previous findings indicate that susceptibility to Leishmania (Viannia) panamensis infection of monocyte-derived macrophages from patients and asymptomatically infected individuals were associated with the adaptive immune response and clinical outcome. Methodology/Principal Findings To understand the basis for this difference we examined differential gene expression of human monocyte-derived macrophages following exposure to L. (V.) panamensis. Gene activation profiles were determined using macrophages from healthy volunteers cultured with or without stationary phase promastigotes of L. (V.) panamensis. Significant changes in expression (>1.5-fold change; p<0.05; up- or down-regulated) were identified at 0.5, 4 and 24 hours. mRNA abundance profiles varied over time, with the highest level of activation occurring at earlier time points (0.5 and 4 hrs). In contrast to observations for other Leishmania species, most significantly changed mRNAs were up- rather than down-regulated, especially at early time points. Up-regulated transcripts over the first 24 hours belonged to pathways involving eicosanoid metabolism, oxidative stress, activation of PKC through G protein coupled receptors, or mechanism of gene regulation by peroxisome proliferators via PPARα. Additionally, a marked activation of Toll-receptor mediated pathways was observed. Comparison with published microarray data from macrophages infected with L. (Leishmania) chagasi indicate differences in the regulation of genes involved in signaling, motility and the immune response. Conclusions Results show that the early (0.5 to 24 hours) human monocyte-derived macrophage response to L. (Viannia) panamensis is not quiescent, in contrast to published reports examining later response times (48–96 hours). Early macrophage responses are important for the developing cellular response at the site of infection. The kinetics and the mRNA abundance profiles induced by L. (Viannia) panamensis illustrate the dynamics of these interactions and the distinct biologic responses to different Leishmania species from the outset of infection within their primary host cell. Leishmania parasites cause a spectrum of diseases (cutaneous, visceral and the deforming forms—chronic cutaneous and mucocutaneous) known as leishmaniasis. The macrophage, a key cell in the immune system, is the cellular target of Leishmania parasites in the mammalian host. Previous studies showed the responses of monocytederived macrophages from naturally infected humans to infection with Leishmania (Viannia) panamensis were key to adaptive immune responses and clinical outcome. Consequently, an mRNA microarray approach was employed to assess the changes in macrophage gene expression over time (0.5 to 24 hours) induced by L. panamensis. The highest level of gene expression induction occurred early (0.5–4 hours); the early pathways (groups of genes) activated included those involved in the innate immune response (signaling, phagocytosis, TLR activation, and inflammatory). Early gene activation is presumed to be important for the developing cellular milieu at the site of infection. By 24 hours post-infection the dominant pathways involved metabolic functions. However, a comparison of the macrophage response to L. (V.) panamensis to that of L. (L.) chagasi (causative agent of visceral leishmaniasis) at 24 hours revealed a differential up-regulation of genes (cell adhesion, signaling, and inflammation) in response to these species. These observations underscore the distinct biology of different Leishmania species from the outset of infection.
Collapse
|
114
|
Prantner D, Perkins DJ, Lai W, Williams MS, Sharma S, Fitzgerald KA, Vogel SN. 5,6-Dimethylxanthenone-4-acetic acid (DMXAA) activates stimulator of interferon gene (STING)-dependent innate immune pathways and is regulated by mitochondrial membrane potential. J Biol Chem 2012; 287:39776-88. [PMID: 23027866 DOI: 10.1074/jbc.m112.382986] [Citation(s) in RCA: 177] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The chemotherapeutic agent 5,6-dimethylxanthenone-4-acetic acid (DMXAA) is a potent inducer of type I IFNs and other cytokines. This ability is essential for its chemotherapeutic benefit in a mouse cancer model and suggests that it might also be useful as an antiviral agent. However, the mechanism underlying DMXAA-induced type I IFNs, including the host proteins involved, remains unclear. Recently, it was reported that the antioxidant N-acetylcysteine (NAC) decreased DMXAA-induced TNF-α and IL-6, suggesting that oxidative stress may play a role. The goal of this study was to identify host proteins involved in DMXAA-dependent signaling and determine how antioxidants modulate this response. We found that expression of IFN-β in response to DMXAA in mouse macrophages requires the mitochondrial and endoplasmic reticulum resident protein STING. Addition of the antioxidant diphenylene iodonium (DPI) diminished DMXAA-induced IFN-β, but this decrease was independent of both the NADPH oxidase, Nox2, and de novo generation of reactive oxygen species. Additionally, IFN-β up-regulation by DMXAA was inhibited by agents that target the mitochondrial electron transport chain and, conversely, loss of mitochondrial membrane potential correlated with diminished innate immune signaling in response to DMXAA. Up-regulation of Ifnb1 gene expression mediated by cyclic dinucleotides was also impaired by DPI, whereas up-regulation of Ifnb1 mRNA due to cytosolic double-stranded DNA was not. Although both stimuli signal through STING, cyclic dinucleotides interact directly with STING, suggesting that recognition of DMXAA by STING may also be mediated by direct interaction.
Collapse
Affiliation(s)
- Daniel Prantner
- Department of Microbiology and Immunology, University of Maryland, School of Medicine (UMB), Baltimore, Maryland 21201, USA
| | | | | | | | | | | | | |
Collapse
|
115
|
Thakur D, Saxena R, Singh V, Haq W, Katti SB, Singh BN, Tripathi RK. Human beta casein fragment (54-59) modulates M. bovis BCG survival and basic transcription factor 3 (BTF3) expression in THP-1 cell line. PLoS One 2012; 7:e45905. [PMID: 23029305 PMCID: PMC3461027 DOI: 10.1371/journal.pone.0045905] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2012] [Accepted: 08/27/2012] [Indexed: 12/15/2022] Open
Abstract
Immunostimulatory peptides potentiate the immune system of the host and are being used as a viable adjunct to established therapeutic modalities in treatment of cancer and microbial infections. Several peptides derived from milk protein have been reported to induce immunostimulatory activity. Human β -casein fragment (54–59), natural sequence peptide (NS) carrying the Val-Glu-Pro-Ile-Pro-Tyr amino acid residues, was reported to activate the macrophages and impart potent immunostimulatory activity. In present study, we found that this peptide increases the clearance of M. bovis BCG from THP-1 cell line in vitro. The key biomolecules, involved in the clearance of BCG from macrophage like, nitric oxide, pro-inflammatory cytokines and chemokines, were not found to be significantly altered after peptide treatment in comparison to the untreated control. Using proteomic approach we found that BTF3a, an isoform of the Basic Transcription Factor, BTF3, was down regulated in THP-1 cell line after peptide treatment. This was reconfirmed by real time RT-PCR and western blotting. We report the BTF3a as a novel target of this hexapeptide. Based on the earlier findings and the results from the present studies, we suggest that the down regulation of BTF3a following the peptide treatment may augment the M. bovis BCG mediated apoptosis resulting in enhanced clearance of M. bovis BCG from THP-1 cell line.
Collapse
Affiliation(s)
| | - Reshu Saxena
- Division of Toxicology, Central Drug Research Institute, Lucknow, India
| | - Vandana Singh
- Division of Microbiology, Central Drug Research Institute, Lucknow, India
| | - Wahajul Haq
- Division of Medicinal and Process Chemistry, Central Drug Research Institute, Lucknow, India
| | - S. B. Katti
- Division of Medicinal and Process Chemistry, Central Drug Research Institute, Lucknow, India
| | - Bhupendra Narain Singh
- Division of Microbiology, Central Drug Research Institute, Lucknow, India
- * E-mail: (BNS); (RKT)
| | - Raj Kamal Tripathi
- Division of Toxicology, Central Drug Research Institute, Lucknow, India
- * E-mail: (BNS); (RKT)
| |
Collapse
|
116
|
Parikka M, Hammarén MM, Harjula SKE, Halfpenny NJA, Oksanen KE, Lahtinen MJ, Pajula ET, Iivanainen A, Pesu M, Rämet M. Mycobacterium marinum causes a latent infection that can be reactivated by gamma irradiation in adult zebrafish. PLoS Pathog 2012; 8:e1002944. [PMID: 23028333 PMCID: PMC3459992 DOI: 10.1371/journal.ppat.1002944] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 08/18/2012] [Indexed: 01/04/2023] Open
Abstract
The mechanisms leading to latency and reactivation of human tuberculosis are still unclear, mainly due to the lack of standardized animal models for latent mycobacterial infection. In this longitudinal study of the progression of a mycobacterial disease in adult zebrafish, we show that an experimental intraperitoneal infection with a low dose (≈ 35 bacteria) of Mycobacterium marinum, results in the development of a latent disease in most individuals. The infection is characterized by limited mortality (25%), stable bacterial loads 4 weeks following infection and constant numbers of highly organized granulomas in few target organs. The majority of bacteria are dormant during a latent mycobacterial infection in zebrafish, and can be activated by resuscitation promoting factor ex vivo. In 5-10% of tuberculosis cases in humans, the disease is reactivated usually as a consequence of immune suppression. In our model, we are able to show that reactivation can be efficiently induced in infected zebrafish by γ-irradiation that transiently depletes granulo/monocyte and lymphocyte pools, as determined by flow cytometry. This immunosuppression causes reactivation of the dormant mycobacterial population and a rapid outgrowth of bacteria, leading to 88% mortality in four weeks. In this study, the adult zebrafish presents itself as a unique non-mammalian vertebrate model for studying the development of latency, regulation of mycobacterial dormancy, as well as reactivation of latent or subclinical tuberculosis. The possibilities for screening for host and pathogen factors affecting the disease progression, and identifying novel therapeutic agents and vaccine targets make this established model especially attractive.
Collapse
|
117
|
Pathogenic Mycobacterium bovis strains differ in their ability to modulate the proinflammatory activation phenotype of macrophages. BMC Microbiol 2012; 12:166. [PMID: 22863292 PMCID: PMC3478980 DOI: 10.1186/1471-2180-12-166] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 07/18/2012] [Indexed: 01/05/2023] Open
Abstract
Background Tuberculosis, caused by Mycobacterium tuberculosis or Mycobacterium bovis, remains one of the leading infectious diseases worldwide. The ability of mycobacteria to rapidly grow in host macrophages is a factor contributing to enhanced virulence of the bacteria and disease progression. Bactericidal functions of phagocytes are strictly dependent on activation status of these cells, regulated by the infecting agent and cytokines. Pathogenic mycobacteria can survive the hostile environment of the phagosome through interference with activation of bactericidal responses. To study the mechanisms employed by highly virulent mycobacteria to promote their intracellular survival, we investigated modulating effects of two pathogenic M. bovis isolates and a reference M. tuberculosis H37Rv strain, differing in their ability to multiply in macrophages, on activation phenotypes of the cells primed with major cytokines regulating proinflammatory macrophage activity. Results Bone marrow- derived macrophages obtained from C57BL/6 mice were infected by mycobacteria after a period of cell incubation with or without treatment with IFN-γ, inducing proinflammatory type-1 macrophages (M1), or IL-10, inducing anti-inflammatory type-2 cells (M2). Phenotypic profiling of M1 and M2 was then evaluated. The M. bovis strain MP287/03 was able to grow more efficiently in the untreated macrophages, compared with the strains B2 or H37Rv. This strain induced weaker secretion of proinflammatory cytokines, coinciding with higher expression of M2 cell markers, mannose receptor (MR) and arginase-1 (Arg-1). Treatment of macrophages with IFN-γ and infection by the strains B2 and H37Rv synergistically induced M1 polarization, leading to high levels of inducible nitric oxide synthase (iNOS) expression, and reduced expression of the Arg-1. In contrast, the cells infected with the strain MP287/03 expressed high levels of Arg-1 which competed with iNOS for the common substrate arginine, leading to lower levels of NO production. Conclusions The data obtained demonstrated that the strain, characterized by increased growth in macrophages, down- modulated classical macrophage activation, through induction of an atypical mixed M1/M2 phenotype.
Collapse
|
118
|
Oliveira RAS, Correia-Oliveira J, Tang LJ, Garcia RC. A proteomic insight into the effects of the immunomodulatory hydroxynaphthoquinone lapachol on activated macrophages. Int Immunopharmacol 2012; 14:54-65. [PMID: 22705049 DOI: 10.1016/j.intimp.2012.05.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2012] [Revised: 05/29/2012] [Accepted: 05/29/2012] [Indexed: 12/31/2022]
Abstract
We report the effect of an immunomodulatory and anti-mycobacterial naphthoquinone, lapachol, on the bi-dimensional patterns of protein expression of toll-like receptor 2 (TLR2)-agonised and IFN-γ-treated THP-1 macrophages. This non-hypothesis driven proteomic analysis intends to shed light on the cellular functions lapachol may be affecting. Proteins of both cytosol and membrane fractions were analysed. After quantification of the protein spots, the protein levels corresponding to macrophages activated in the absence or presence of lapachol were compared. A number of proteins were identified, the levels of which were appreciably and significantly increased or decreased as a result of the action of lapachol on the activated macrophages: cofilin-1, fascin, plastin-2, glucose-6-P-dehydrogenase, adenylyl cyclase-associated protein 1, pyruvate kinase, sentrin-specific protease 6, cathepsin B, cathepsin D, cytosolic aminopeptidase, proteasome β type-4 protease, tryptophan-tRNA ligase, DnaJ homolog and protein disulphide isomerase. Altogether, the comparative analysis performed indicates that lapachol could be hypothetically causing an impairment of cell migration and/or phagocytic capacity, an increase in NADPH availability, a decrease in pyruvate concentration, protection from proteosomal protein degradation, a decrease in lysosomal protein degradation, an impairment of cytosolic peptide generation, and an interference with NOS2 activation and grp78 function. The present proteomic results suggest issues that should be experimentally addressed ex- and in-vivo, to establish more accurately the potential of lapachol as an anti-infective drug. This study also constitutes a model for the pre-in-vivo evaluation of drug actions.
Collapse
Affiliation(s)
- Renato A S Oliveira
- Leukocyte Biology Group, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy.
| | | | | | | |
Collapse
|
119
|
Wu K, Dong D, Fang H, Levillain F, Jin W, Mei J, Gicquel B, Du Y, Wang K, Gao Q, Neyrolles O, Zhang J. An interferon-related signature in the transcriptional core response of human macrophages to Mycobacterium tuberculosis infection. PLoS One 2012; 7:e38367. [PMID: 22675550 PMCID: PMC3366933 DOI: 10.1371/journal.pone.0038367] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Accepted: 05/03/2012] [Indexed: 11/23/2022] Open
Abstract
The W-Beijing family of Mycobacterium tuberculosis (Mtb) strains is known for its high-prevalence and -virulence, as well as for its genetic diversity, as recently reported by our laboratories and others. However, little is known about how the immune system responds to these strains. To explore this issue, here we used reverse engineering and genome-wide expression profiling of human macrophage-like THP-1 cells infected by different Mtb strains of the W-Beijing family, as well as by the reference laboratory strain H37Rv. Detailed data mining revealed that host cell transcriptome responses to H37Rv and to different strains of the W-Beijing family are similar and overwhelmingly induced during Mtb infections, collectively typifying a robust gene expression signature (“THP1r2Mtb-induced signature”). Analysis of the putative transcription factor binding sites in promoter regions of genes in this signature identified several key regulators, namely STATs, IRF-1, IRF-7, and Oct-1, commonly involved in interferon-related immune responses. The THP1r2Mtb-induced signature appeared to be highly relevant to the interferon-inducible signature recently reported in active pulmonary tuberculosis patients, as revealed by cross-signature and cross-module comparisons. Further analysis of the publicly available transcriptome data from human patients showed that the signature appears to be relevant to active pulmonary tuberculosis patients and their clinical therapy, and be tuberculosis specific. Thus, our results provide an additional layer of information at the transcriptome level on mechanisms involved in host macrophage response to Mtb, which may also implicate the robustness of the cellular defense system that can effectively fight against genetic heterogeneity in this pathogen.
Collapse
Affiliation(s)
- Kang Wu
- State Key Laboratory of Medical Genomics and Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dandan Dong
- Key Laboratory of Medical Molecular Virology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hai Fang
- State Key Laboratory of Medical Genomics and Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Florence Levillain
- Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France
- Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France
| | - Wen Jin
- State Key Laboratory of Medical Genomics and Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Mei
- Department of Tuberculosis Control, Shanghai Municipal CDC, Shanghai, China
| | - Brigitte Gicquel
- Unité de Génétique Mycobactérienne, Institut Pasteur, Paris, France
| | - Yanzhi Du
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kankan Wang
- State Key Laboratory of Medical Genomics and Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Gao
- Key Laboratory of Medical Molecular Virology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Olivier Neyrolles
- Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France
- Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France
- * E-mail: (ON); (JZ)
| | - Ji Zhang
- State Key Laboratory of Medical Genomics and Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, Shanghai, China
- * E-mail: (ON); (JZ)
| |
Collapse
|
120
|
Blumenthal A, Nagalingam G, Huch JH, Walker L, Guillemin GJ, Smythe GA, Ehrt S, Britton WJ, Saunders BM. M. tuberculosis induces potent activation of IDO-1, but this is not essential for the immunological control of infection. PLoS One 2012; 7:e37314. [PMID: 22649518 PMCID: PMC3359358 DOI: 10.1371/journal.pone.0037314] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2012] [Accepted: 04/18/2012] [Indexed: 11/18/2022] Open
Abstract
Indoleamine 2,3-dioxygenesae-1 (IDO-1) catalyses the initial, rate-limiting step in tryptophan metabolism, thereby regulating tryptophan availability and the formation of downstream metabolites, including picolinic and quinolinic acid. We found that Mycobacterium tuberculosis infection induced marked upregulation of IDO-1 expression in both human and murine macrophages in vitro and in the lungs of mice following aerosol challenge with M. tuberculosis. The absence of IDO-1 in dendritic cells enhanced the activation of mycobacteria-specific T cells in vitro. Interestingly, IDO-1-deficiency during M. tuberculosis infection in mice was not associated with altered mycobacteria-specific T cell responses in vivo. The bacterial burden of infected organs, pulmonary inflammatory responses, and survival were also comparable in M. tuberculosis-infected IDO-1 deficient and wild type animals. Tryptophan is metabolised into either picolinic acid or quinolinic acid, but only picolinic acid inhibited the growth of M. tuberculosis in vitro. By contrast macrophages infected with pathogenic mycobacteria, produced quinolinic, rather than picolinic acid, which did not reduce M. tuberculosis growth in vitro. Therefore, although M. tuberculosis induces robust expression of IDO-1 and activation of tryptophan metabolism, IDO-1-deficiency fails to impact on the immune control and the outcome of the infection in the mouse model of tuberculosis.
Collapse
Affiliation(s)
- Antje Blumenthal
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, United States of America
- The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
- * E-mail: (AB); (BMS)
| | - Gayathri Nagalingam
- Mycobacterial Research Program, Centenary Institute Sydney, New South Wales, Australia
| | - Jennifer H. Huch
- Mycobacterial Research Program, Centenary Institute Sydney, New South Wales, Australia
| | - Lara Walker
- Mycobacterial Research Program, Centenary Institute Sydney, New South Wales, Australia
| | - Gilles J. Guillemin
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - George A. Smythe
- Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Sabine Ehrt
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, United States of America
- Program in Immunology and Microbial Pathogenesis, Weill Graduate School of Medical Sciences of Cornell University, New York, New York, United States of America
| | - Warwick J. Britton
- The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
- Department of Medicine, University of Sydney, Sydney, New South Wales, Australia
| | - Bernadette M. Saunders
- The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
- Department of Medicine, University of Sydney, Sydney, New South Wales, Australia
- * E-mail: (AB); (BMS)
| |
Collapse
|
121
|
Richter-Dahlfors A, Rhen M, Udekwu K. Tissue microbiology provides a coherent picture of infection. Curr Opin Microbiol 2012; 15:15-22. [DOI: 10.1016/j.mib.2011.10.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Revised: 10/14/2011] [Accepted: 10/17/2011] [Indexed: 12/26/2022]
|
122
|
Koo MS, Subbian S, Kaplan G. Strain specific transcriptional response in Mycobacterium tuberculosis infected macrophages. Cell Commun Signal 2012; 10:2. [PMID: 22280836 PMCID: PMC3317440 DOI: 10.1186/1478-811x-10-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Accepted: 01/26/2012] [Indexed: 11/10/2022] Open
Abstract
Background Tuberculosis (TB), a bacterial infection caused by Mycobacterium tuberculosis (Mtb) remains a significant health problem worldwide with a third of the world population infected and nearly nine million new cases claiming 1.1 million deaths every year. The outcome following infection by Mtb is determined by a complex and dynamic host-pathogen interaction in which the phenotype of the pathogen and the immune status of the host play a role. However, the molecular mechanism by which Mtb strains induce different responses during intracellular infection of the host macrophage is not fully understood. To explore the early molecular events triggered upon Mtb infection of macrophages, we studied the transcriptional responses of murine bone marrow-derived macrophages (BMM) to infection with two clinical Mtb strains, CDC1551 and HN878. These strains have previously been shown to differ in their virulence/immunogenicity in the mouse and rabbit models of pulmonary TB. Results In spite of similar intracellular growth rates, we observed that compared to HN878, infection by CDC1551 of BMM was associated with an increased global transcriptome, up-regulation of a specific early (6 hours) immune response network and significantly elevated nitric oxide production. In contrast, at 24 hours post-infection of BMM by HN878, more host genes involved in lipid metabolism, including cholesterol metabolism and prostaglandin synthesis were up-regulated, compared to infection with CDC1551. In association with the differences in the macrophage responses to infection with the 2 Mtb strains, intracellular CDC1551 expressed higher levels of stress response genes than did HN878. Conclusions In association with the early and more robust macrophage activation, intracellular CDC1551 cells were exposed to a higher level of stress leading to increased up-regulation of the bacterial stress response genes. In contrast, sub-optimal activation of macrophages and induction of a dysregulated host cell lipid metabolism favored a less stressful intracellular environment for HN878. Our findings suggest that the ability of CDC1551 and HN878 to differentially activate macrophages during infection probably determines their ability to either resist host cell immunity and progress to active disease or to succumb to the host protective responses and be driven into a non-replicating latent state in rabbit lungs.
Collapse
Affiliation(s)
- Mi-Sun Koo
- Laboratory of Mycobacterial Immunity and Pathogenesis, The Public Health Research Institute (PHRI) at the University of Medicine and Dentistry of New Jersey (UNDNJ), 225 Warren Street, Newark, New Jersey 07103, USA.
| | | | | |
Collapse
|
123
|
Machado FS, Tyler KM, Brant F, Esper L, Teixeira MM, Tanowitz HB. Pathogenesis of Chagas disease: time to move on. Front Biosci (Elite Ed) 2012; 4:1743-58. [PMID: 22201990 DOI: 10.2741/495] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Trypanosoma cruzi is the etiologic agent of Chagas disease. The contributions of parasite and immune system for disease pathogenesis remain unresolved and controversial. The possibility that Chagas disease was an autoimmune progression triggered by T. cruzi infection led some to question the benefit of treating chronically T. cruzi-infected persons with drugs. Furthermore, it provided the rationale for not investing in research aimed at a vaccine which might carry a risk of inducing autoimmunity or exacerbating inflammation. This viewpoint was adopted by cash-strapped health systems in the developing economies where the disease is endemic and has been repeatedly challenged by researchers and clinicians in recent years and there is now a considerable body of evidence and broad consensus that parasite persistence is requisite for pathogenesis and that antiparasitic immunity can be protective against T. cruzi pathogenesis without eliciting autoimmune pathology. Thus, treatment of chronically infected patients is likely to yield positive outcomes and efforts to understand immunity and vaccine development should be recognized as a priority area of research for Chagas disease.
Collapse
Affiliation(s)
- Fabiana S Machado
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Belo Horizonte, Brazil
| | | | | | | | | | | |
Collapse
|
124
|
Onoprienko LV. [Molecular mechanisms regulating the activity of macrophages]. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2011; 37:437-51. [PMID: 22096986 DOI: 10.1134/s1068162011040091] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This article reviews modern concepts of the most common types of macrophage activation: classical, alternative, and type II. Molecular mechanisms of induction and regulation of these three types of activation are discussed. Any population of macrophages was shown to change its properties depending on its microenvironment and concrete biological situation (the "functional plasticity of macrophages"). Many intermediate states of macrophages were described along with the most pronounced and well-known activation types (classical activation, alternative activation, and type II activation). These intermediate states are characterized by a variety of combinations of their biological properties, including elements of the three afore mentioned types of activation. Macrophage activity is regulated by a complex network of interrelated cascade mechanisms.
Collapse
|
125
|
He XN, Su F, Lou ZZ, Jia WZ, Song YL, Chang HY, Wu YH, Lan J, He XY, Zhang Y. Ipr1 gene mediates RAW 264.7 macrophage cell line resistance to Mycobacterium bovis. Scand J Immunol 2011; 74:438-44. [PMID: 21790702 DOI: 10.1111/j.1365-3083.2011.02596.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Tuberculosis caused by Mycobacterium bovis (M. bovis) seriously affects efficiency of animal production with impacts on public health as well. Effective programmes of prevention and eradication of M. bovis infection therefore are urgently needed. Intracellular pathogen resistance gene 1 (Ipr1) is well known to mediate innate immunity to Mycobacterium tuberculosis (MTB), but there are no reports as to whether Ipr1 can enhance the phagocytic ability of macrophage against M. bovis. In this investigation, RAW 264.7 macrophage was transduced with lentiviral vector carrying Ipr1 (named Lenti-Ipr1); transgenic cells were identified by RT-PCR and western blotting. Transgenic positive cells (R-Ipr1) were then infected with an M. bovis virulent strain, with non-transduced cells used as control. When cell proliferation, viability and apoptosis of the two groups were investigated, it was found that infected RAW 264.7 died by necrosis whereas R-Ipr1 underwent apoptosis. Furthermore, the numbers of intracellular bacteria in R-Ipr1 were lower than those in control cells (P < 0.05). To identify the role of Ipr1, we measured the genes of Casp3, Mcl-1 and NOS2A which associated with macrophage activation and apoptosis by real-time quantitative PCR. The results demonstrated that Ipr1 gene expression can enhance anti-M. bovis infection of macrophage. This establishes a basis for the future production of Ipr1-transgenic cattle to strengthen the tuberculosis resistance.
Collapse
Affiliation(s)
- X N He
- Key Laboratory of Animal Reproductive Physiology & Embryo Technology, Ministry of Agriculture, College of Veterinary Medicine, Northwest A & F University, Yangling, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
126
|
Nandi B, Behar SM. Regulation of neutrophils by interferon-γ limits lung inflammation during tuberculosis infection. ACTA ACUST UNITED AC 2011; 208:2251-62. [PMID: 21967766 PMCID: PMC3201199 DOI: 10.1084/jem.20110919] [Citation(s) in RCA: 278] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
IFN-γ functions to suppress neutrophil accumulation in the lungs of mice infected with M. tuberculosis, in part by suppressing IL-17 production from CD4+ T cells. Resistance to Mycobacterium tuberculosis requires the host to restrict bacterial replication while preventing an over-exuberant inflammatory response. Interferon (IFN) γ is crucial for activating macrophages and also regulates tissue inflammation. We dissociate these two functions and show that IFN-γ−/− memory CD4+ T cells retain their antimicrobial activity but are unable to suppress inflammation. IFN-γ inhibits CD4+ T cell production of IL-17, which regulates neutrophil recruitment. In addition, IFN-γ directly inhibits pathogenic neutrophil accumulation in the infected lung and impairs neutrophil survival. Regulation of neutrophils is important because their accumulation is detrimental to the host. We suggest that neutrophilia during tuberculosis indicates failed Th1 immunity or loss of IFN-γ responsiveness. These results establish an important antiinflammatory role for IFN-γ in host protection against tuberculosis.
Collapse
Affiliation(s)
- Bisweswar Nandi
- Division of Rheumatology, Immunology, and Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
127
|
Lugo-Villarino G, Vérollet C, Maridonneau-Parini I, Neyrolles O. Macrophage polarization: convergence point targeted by mycobacterium tuberculosis and HIV. Front Immunol 2011; 2:43. [PMID: 22566833 PMCID: PMC3342390 DOI: 10.3389/fimmu.2011.00043] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Accepted: 08/25/2011] [Indexed: 01/30/2023] Open
Abstract
In the arms race of host–microbe co-evolution, macrophages (Mɸs) have been endowed with strategies to neutralize pathogenic challenge while preserving host integrity. During steady-states conditions, Mɸs perform multiple house-keeping functions governed by their differentiation state, tissue distribution, and signals from the microenvironment. In response to pathogenic challenge and host mediators, however, Mɸs undergo different programs of activation rendering them either pro-inflammatory and microbicidal (M1), or immunosuppressants and tissue repairers (M2). An excessive or prolonged polarization of either program may be detrimental to the host due to potential tissue injury or contribution to pathogenesis. Conversely, intracellular microbes that cause chronic diseases such as tuberculosis and acquired immunodeficiency syndrome exemplify strategies for survival in the host. Indeed, both Mycobacterium tuberculosis (Mtb) and human immunodeficiency virus (HIV-1) are successful intracellular microbes that thrive in Mɸs. Given these microbes not only co-circulate throughout the developing world but each has contributed to prevalence and mortality caused by the other, substantial insights into microbe physiology and host defenses then rest in the attempt to fully understand their influence on Mɸ polarization. This review addresses the role of Mɸ polarization in the immune response to, and pathogenesis of, Mtb and HIV.
Collapse
|
128
|
Virreira Winter S, Niedelman W, Jensen KD, Rosowski EE, Julien L, Spooner E, Caradonna K, Burleigh BA, Saeij JPJ, Ploegh HL, Frickel EM. Determinants of GBP recruitment to Toxoplasma gondii vacuoles and the parasitic factors that control it. PLoS One 2011; 6:e24434. [PMID: 21931713 PMCID: PMC3169597 DOI: 10.1371/journal.pone.0024434] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 08/09/2011] [Indexed: 01/20/2023] Open
Abstract
IFN-γ is a major cytokine that mediates resistance against the intracellular parasite Toxoplasma gondii. The p65 guanylate-binding proteins (GBPs) are strongly induced by IFN-γ. We studied the behavior of murine GBP1 (mGBP1) upon infection with T. gondii in vitro and confirmed that IFN-γ-dependent re-localization of mGBP1 to the parasitophorous vacuole (PV) correlates with the virulence type of the parasite. We identified three parasitic factors, ROP16, ROP18, and GRA15 that determine strain-specific accumulation of mGBP1 on the PV. These highly polymorphic proteins are held responsible for a large part of the strain-specific differences in virulence. Therefore, our data suggest that virulence of T. gondii in animals may rely in part on recognition by GBPs. However, phagosomes or vacuoles containing Trypanosoma cruzi did not recruit mGBP1. Co-immunoprecipitation revealed mGBP2, mGBP4, and mGBP5 as binding partners of mGBP1. Indeed, mGBP2 and mGBP5 co-localize with mGBP1 in T. gondii-infected cells. T. gondii thus elicits a cell-autonomous immune response in mice with GBPs involved. Three parasitic virulence factors and unknown IFN-γ-dependent host factors regulate this complex process. Depending on the virulence of the strains involved, numerous GBPs are brought to the PV as part of a large, multimeric structure to combat T. gondii.
Collapse
Affiliation(s)
| | - Wendy Niedelman
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Kirk D. Jensen
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Emily E. Rosowski
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Lindsay Julien
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Eric Spooner
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, United States of America
| | - Kacey Caradonna
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Barbara A. Burleigh
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Jeroen P. J. Saeij
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Hidde L. Ploegh
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, United States of America
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- * E-mail: (HLP); (E-MF)
| | - Eva-Maria Frickel
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, United States of America
- * E-mail: (HLP); (E-MF)
| |
Collapse
|
129
|
Lamichhane G. Mycobacterium tuberculosis response to stress from reactive oxygen and nitrogen species. Front Microbiol 2011; 2:176. [PMID: 21904537 PMCID: PMC3163289 DOI: 10.3389/fmicb.2011.00176] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2011] [Accepted: 08/09/2011] [Indexed: 11/25/2022] Open
Affiliation(s)
- Gyanu Lamichhane
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University Baltimore, MD, USA
| |
Collapse
|
130
|
Traver MK, Henry SC, Cantillana V, Oliver T, Hunn JP, Howard JC, Beer S, Pfeffer K, Coers J, Taylor GA. Immunity-related GTPase M (IRGM) proteins influence the localization of guanylate-binding protein 2 (GBP2) by modulating macroautophagy. J Biol Chem 2011; 286:30471-30480. [PMID: 21757726 DOI: 10.1074/jbc.m111.251967] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The immunity-related GTPases (IRGs) are a family of proteins induced by interferon-γ that play a crucial role in innate resistance to intracellular pathogens. The M subfamily of IRG proteins (IRGM) plays a profound role in this context, in part because of the ability of its members to regulate the localization and expression of other IRG proteins. We present here evidence that IRGM proteins affect the localization of the guanylate-binding proteins (GBPs), a second family of interferon-induced GTP-binding proteins that also function in innate immunity. Absence of Irgm1 or Irgm3 led to accumulation of Gbp2 in intracellular compartments that were positive for both the macroautophagy (hereafter referred to as autophagy) marker LC3 and the autophagic adapter molecule p62/Sqstm1. Gbp2 was similarly relocalized in cells in which autophagy was impaired because of the absence of Atg5. Both in Atg5- and IRGM-deficient cells, the IRG protein Irga6 relocalized to the same compartments as Gbp2, raising the possibility of a common regulatory mechanism. However, other data indicated that Irga6, but not Gbp2, was ubiquitinated in IRGM-deficient cells. Similarly, coimmunoprecipitation studies indicated that although Irgm3 did interact directly with Irgb6, it did not interact with Gbp2. Collectively, these data suggest that IRGM proteins indirectly modulate the localization of GBPs through a distinct mechanism from that through which they regulate IRG protein localization. Further, these results suggest that a core function of IRGM proteins is to regulate autophagic flux, which influences the localization of GBPs and possibly other factors that instruct cell-autonomous immune resistance.
Collapse
Affiliation(s)
- Maria K Traver
- Department of Molecular Genetics and Microbiology, Duke University, Medical Center, Durham, North Carolina 27710
| | - Stanley C Henry
- Geriatric Research, Education, and Clinical Center, VA Medical Center, Durham, North Carolina 27705
| | - Viviana Cantillana
- Center for the Study of Aging and Human Development, Duke University, Medical Center, Durham, North Carolina 27710
| | - Tim Oliver
- Cell Biology, and Medicine, Division of Geriatrics, Duke University, Medical Center, Durham, North Carolina 27710
| | - Julia P Hunn
- Institute for Genetics, Department of Cell Genetics, University of Cologne, Cologne 50674, Germany
| | - Jonathan C Howard
- Institute for Genetics, Department of Cell Genetics, University of Cologne, Cologne 50674, Germany
| | - Sandra Beer
- Institute of Medical Microbiology and Hospital Hygiene, Heinrich Heine University of Düsseldorf, Düsseldorf 40225, Germany
| | - Klaus Pfeffer
- Institute of Medical Microbiology and Hospital Hygiene, Heinrich Heine University of Düsseldorf, Düsseldorf 40225, Germany
| | - Jörn Coers
- Department of Molecular Genetics and Microbiology, Duke University, Medical Center, Durham, North Carolina 27710
| | - Gregory A Taylor
- Department of Molecular Genetics and Microbiology, Duke University, Medical Center, Durham, North Carolina 27710; Geriatric Research, Education, and Clinical Center, VA Medical Center, Durham, North Carolina 27705; Center for the Study of Aging and Human Development, Duke University, Medical Center, Durham, North Carolina 27710; Immunology, Duke University, Medical Center, Durham, North Carolina 27710.
| |
Collapse
|
131
|
Abstract
Sarcoidosis is an uncommon systemic inflammatory disorder characterized by noncaseating granulomatous inflammation that most commonly affects the lungs, intrathoracic lymph nodes, eyes and skin. One-third or more of patients with sarcoidosis have chronic, unremitting inflammation with progressive organ impairment. Findings of family and genetic studies indicate a genetic susceptibility to sarcoidosis, with genes in the MHC region having a dominant role. Immunologic hallmarks of the disease include highly polarized expression of cytokines produced by type 1 T helper cells and tumor necrosis factor (TNF) at sites of inflammation. Increasing evidence obtained within the past decade suggests the etiology of sarcoidosis predominantly involves microbial triggers, with the most convincing data implicating mycobacterial or propionibacterial organisms. Innate immune mechanisms, possibly involving misfolding and aggregation of serum amyloid A, might have a critical role in the pathobiology of sarcoidosis. Despite these advances, there are no clinically useful biomarkers that can assist the clinician in diagnosis, prognosis or assessment of treatment effects. Corticosteroids remain the cornerstone of therapy when organ function is threatened or progressively impaired. The role of immunosuppressive drugs and anti-TNF agents in the treatment of sarcoidosis remains uncertain, and there are no FDA-approved therapies. Meaningful progress in developing clinically useful tools and new therapies will depend on further advances in understanding the pathogenesis of sarcoidosis and its disease-specific pathways.
Collapse
Affiliation(s)
- Edward S Chen
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The Johns Hopkins University, 5501 Hopkins Bayview Circle, Room 4B63, Baltimore, MD 21224, USA
| | | |
Collapse
|
132
|
Kirksey MA, Tischler AD, Siméone R, Hisert KB, Uplekar S, Guilhot C, McKinney JD. Spontaneous phthiocerol dimycocerosate-deficient variants of Mycobacterium tuberculosis are susceptible to gamma interferon-mediated immunity. Infect Immun 2011; 79:2829-38. [PMID: 21576344 PMCID: PMC3191967 DOI: 10.1128/iai.00097-11] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Accepted: 05/03/2011] [Indexed: 11/20/2022] Open
Abstract
Onset of the adaptive immune response in mice infected with Mycobacterium tuberculosis is accompanied by slowing of bacterial replication and establishment of a chronic infection. Stabilization of bacterial numbers during the chronic phase of infection is dependent on the activity of the gamma interferon (IFN-γ)-inducible nitric oxide synthase (NOS2). Previously, we described a differential signature-tagged mutagenesis screen designed to identify M. tuberculosis "counterimmune" mechanisms and reported the isolation of three mutants in the H37Rv strain background containing transposon insertions in the rv0072, rv0405, and rv2958c genes. These mutants were impaired for replication and virulence in NOS2(-/-) mice but were growth-proficient and virulent in IFN-γ(-/-) mice, suggesting that the disrupted genes were required for bacterial resistance to an IFN-γ-dependent immune mechanism other than NOS2. Here, we report that the attenuation of these strains is attributable to an underlying transposon-independent deficiency in biosynthesis of phthiocerol dimycocerosate (PDIM), a cell wall lipid that is required for full virulence in mice. We performed whole-genome resequencing of a PDIM-deficient clone and identified a spontaneous point mutation in the putative polyketide synthase PpsD that results in a G44C amino acid substitution. We demonstrate by complementation with the wild-type ppsD gene and reversion of the ppsD gene to the wild-type sequence that the ppsD(G44C) point mutation is responsible for PDIM deficiency, virulence attenuation in NOS2(-/-) and wild-type C57BL/6 mice, and a growth advantage in vitro in liquid culture. We conclude that PDIM biosynthesis is required for M. tuberculosis resistance to an IFN-γ-mediated immune response that is independent of NOS2.
Collapse
Affiliation(s)
- Meghan A. Kirksey
- Laboratory of Infection Biology, The Rockefeller University, New York, New York 10021
| | - Anna D. Tischler
- Global Health Institute, Swiss Federal Institute of Technology Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Roxane Siméone
- Institut de Pharmacologie et de Biologie Structurale, Centre National de la Recherche Scientifique and Université P. Sabatier (Unité Mixte de Recherche 5089), 31077 Toulouse Cedex, France
| | - Katherine B. Hisert
- Laboratory of Infection Biology, The Rockefeller University, New York, New York 10021
| | - Swapna Uplekar
- Global Health Institute, Swiss Federal Institute of Technology Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Christophe Guilhot
- Institut de Pharmacologie et de Biologie Structurale, Centre National de la Recherche Scientifique and Université P. Sabatier (Unité Mixte de Recherche 5089), 31077 Toulouse Cedex, France
| | - John D. McKinney
- Laboratory of Infection Biology, The Rockefeller University, New York, New York 10021
- Global Health Institute, Swiss Federal Institute of Technology Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| |
Collapse
|
133
|
Exosomes/microvesicles: mediators of cancer-associated immunosuppressive microenvironments. Semin Immunopathol 2011; 33:441-54. [PMID: 21688197 DOI: 10.1007/s00281-010-0234-8] [Citation(s) in RCA: 305] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2010] [Accepted: 11/28/2010] [Indexed: 12/14/2022]
Abstract
Cancer cells, both in vivo and in vitro, have been demonstrated to release membranous structures, defined as microvesicles or exosomes, consisting of an array of macromolecules derived from the originating cells, including proteins, lipids, and nucleic acids. While only recently have the roles of these vesicular components in intercellular communication become elucidated, significant evidence has demonstrated that tumor exosomes can exert a broad array of detrimental effects on the immune system-ranging from apoptosis of activated cytotoxic T cells to impairment of monocyte differentiation into dendritic cells, to induction of myeloid-suppressive cells and T regulatory cells. Immunosuppressive exosomes of tumor origin can be found within neoplastic lesions and in biologic fluids from cancer patients, implying a potential role of these pathways in in vivo tumor progression and systemic paraneoplastic syndromes. Through the expression of molecules involved in angiogenesis promotion, stromal remodeling, signaling pathway activation through growth factor/receptor transfer, chemoresistance, and genetic intercellular exchange, tumor exosomes could represent a central mediator of the tumor microenvironment. By understanding the nature of these tumor-derived exosomes/microvesicles and their roles in mediating cancer progression and modulating the host immune response will significantly impact therapeutic approaches targeting exosomes.
Collapse
|
134
|
Inhaled therapies for tuberculosis and the relevance of activation of lung macrophages by particulate drug-delivery systems. Ther Deliv 2011; 2:753-68. [DOI: 10.4155/tde.11.34] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Pathogenic strains of Mycobacterium tuberculosis (Mtb) induce ‘alternative activation’ of lung macrophages that they colonize, in order to create conditions that promote the establishment and progression of infection. There is some evidence to indicate that such macrophages may be rescued from alternative activation by inhalable microparticles containing a variety of drugs. This review summarizes the experience of various groups of researchers, relating to observations of induction of a number of classical macrophage activation pathways. Restoration of a ‘respiratory burst’ and upregulation of reactive oxygen species and nitrogen intermediates through the phagocyte oxidase and nitric oxide synthetase enzyme systems; induction of proinflammatory macrophage cytokines; and finally induction of apoptosis rather than necrosis of the infected macrophage are discussed. It is suggested that there is scope to co-opt host responses in the management of tuberculosis, through the route of pulmonary drug delivery.
Collapse
|
135
|
Carow B, Ye XQ, Gavier-Widén D, Bhuju S, Oehlmann W, Singh M, Sköld M, Ignatowicz L, Yoshimura A, Wigzell H, Rottenberg ME. Silencing suppressor of cytokine signaling-1 (SOCS1) in macrophages improves Mycobacterium tuberculosis control in an interferon-gamma (IFN-gamma)-dependent manner. J Biol Chem 2011; 286:26873-87. [PMID: 21622562 DOI: 10.1074/jbc.m111.238287] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Protection against infection with Mycobacterium tuberculosis demands IFN-γ. SOCS1 has been shown to inhibit responses to IFN-γ and might thereby play a central role in the outcome of infection. We found that M. tuberculosis is a highly efficient stimulator of SOCS1 expression in murine and human macrophages and in tissues from infected mice. Surprisingly, SOCS1 reduced responses to IL-12, resulting in an impaired IFN-γ secretion by macrophages that in turn accounted for a deteriorated intracellular mycobacterial control. Despite SOCS1 expression, mycobacteria-infected macrophages responded to exogenously added IFN-γ. SOCS1 attenuated the expression of the majority of genes modulated by M. tuberculosis infection of macrophages. Using a conditional knockdown strategy in mice, we found that SOCS1 expression by macrophages hampered M. tuberculosis clearance early after infection in vivo in an IFN-γ-dependent manner. On the other hand, at later time points, SOCS1 expression by non-macrophage cells protected the host from infection-induced detrimental inflammation.
Collapse
Affiliation(s)
- Berit Carow
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm 17177, Sweden
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
136
|
Spivak AY, Khalitova RR, Bel’skii YP, Ivanova AN, Shakurova ER, Bel’skaya NV, Odinokov VN, Danilets MG, Ligacheva AA. Synthesis of conjugates of lupane triterpenoids with chromane antioxidants and in vitro study of their influence on the production of nitrogen monoxide and on the arginase activity in activated macrophages. Russ Chem Bull 2011. [DOI: 10.1007/s11172-010-0382-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
137
|
Gallegos AM, van Heijst JWJ, Samstein M, Su X, Pamer EG, Glickman MS. A gamma interferon independent mechanism of CD4 T cell mediated control of M. tuberculosis infection in vivo. PLoS Pathog 2011; 7:e1002052. [PMID: 21625591 PMCID: PMC3098235 DOI: 10.1371/journal.ppat.1002052] [Citation(s) in RCA: 164] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Accepted: 03/19/2011] [Indexed: 12/12/2022] Open
Abstract
CD4 T cell deficiency or defective IFNγ signaling render humans and mice highly susceptible to Mycobacterium tuberculosis (Mtb) infection. The prevailing model is that Th1 CD4 T cells produce IFNγ to activate bactericidal effector mechanisms of infected macrophages. Here we test this model by directly interrogating the effector functions of Th1 CD4 T cells required to control Mtb in vivo. While Th1 CD4 T cells specific for the Mtb antigen ESAT-6 restrict in vivo Mtb growth, this inhibition is independent of IFNγ or TNF and does not require the perforin or FAS effector pathways. Adoptive transfer of Th17 CD4 T cells specific for ESAT-6 partially inhibited Mtb growth while Th2 CD4 T cells were largely ineffective. These results imply a previously unrecognized IFNγ/TNF independent pathway that efficiently controls Mtb and suggest that optimization of this alternative effector function may provide new therapeutic avenues to combat Mtb through vaccination.
Collapse
Affiliation(s)
- Alena M. Gallegos
- NIH/NIAID Laboratory of Parasitic Diseases,
Bethesda, Maryland, United States of America
- Immunology Program, Infectious Disease
Service, Memorial Sloan-Kettering Cancer Center, New York, New York, United
States of America
| | - Jeroen W. J. van Heijst
- Immunology Program, Infectious Disease
Service, Memorial Sloan-Kettering Cancer Center, New York, New York, United
States of America
| | - Miriam Samstein
- Program in Immunology and Microbial
Pathogenesis, Weill Graduate School of Medical Sciences, New York, New York,
United States of America
| | - Xiaodi Su
- Program in Immunology and Microbial
Pathogenesis, Weill Graduate School of Medical Sciences, New York, New York,
United States of America
| | - Eric G. Pamer
- Immunology Program, Infectious Disease
Service, Memorial Sloan-Kettering Cancer Center, New York, New York, United
States of America
- * E-mail: (EGP); (MSG)
| | - Michael S. Glickman
- Immunology Program, Infectious Disease
Service, Memorial Sloan-Kettering Cancer Center, New York, New York, United
States of America
- * E-mail: (EGP); (MSG)
| |
Collapse
|
138
|
Central carbon metabolism in Mycobacterium tuberculosis: an unexpected frontier. Trends Microbiol 2011; 19:307-14. [PMID: 21561773 DOI: 10.1016/j.tim.2011.03.008] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Revised: 03/29/2011] [Accepted: 03/31/2011] [Indexed: 11/23/2022]
Abstract
Recent advances in liquid chromatography and mass spectrometry have enabled the highly parallel, quantitative measurement of metabolites within a cell and the ability to trace their biochemical fates. In Mycobacterium tuberculosis (Mtb), these advances have highlighted major gaps in our understanding of central carbon metabolism (CCM) that have prompted fresh interpretations of the composition and structure of its metabolic pathways and the phenotypes of Mtb strains in which CCM genes have been deleted. High-throughput screens have demonstrated that small chemical compounds can selectively inhibit some enzymes of Mtb's CCM while sparing homologs in the host. Mtb's CCM has thus emerged as a frontier for both fundamental and translational research.
Collapse
|
139
|
Widdison S, Watson M, Coffey TJ. Early response of bovine alveolar macrophages to infection with live and heat-killed Mycobacterium bovis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2011; 35:580-91. [PMID: 21232552 DOI: 10.1016/j.dci.2011.01.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2010] [Revised: 01/04/2011] [Accepted: 01/05/2011] [Indexed: 05/30/2023]
Abstract
Bovine tuberculosis (TB) is a disease of economic importance and a significant animal health and welfare issue. The alveolar macrophage (AlvMϕ) plays a vital role in the immune response to TB and recent studies provide insights into the interactions between Mϕ and Mycobacterium bovis. Here we reveal the early transcriptional response of bovine AlvMϕ to M. bovis infection. We demonstrate up-regulation of immune response genes, including chemokines, members of the NF-κB pathway which may be involved in their transcription and also pro- and anti-apoptotic genes. M. bovis may therefore induce multiple mechanisms to manipulate the host immune response. We compared the response of AlvMϕ to infection with live and heat-killed M. bovis to determine transcriptional differences dependent on the viable pathogen. Several chemokines up-regulated following live M. bovis infection were not up-regulated after heat-killed M. bovis stimulation; hence the Mϕ seems to differentiate between the two stimuli.
Collapse
Affiliation(s)
- Stephanie Widdison
- Livestock Infectious Diseases Programme, Institute for Animal Health, Compton, Berkshire, RG20 7NN, UK
| | | | | |
Collapse
|
140
|
He G, Ma Y, Chou SY, Li H, Yang C, Chuang JZ, Sung CH, Ding A. Role of CLIC4 in the host innate responses to bacterial lipopolysaccharide. Eur J Immunol 2011; 41:1221-30. [PMID: 21469130 PMCID: PMC3099427 DOI: 10.1002/eji.201041266] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Revised: 01/11/2011] [Accepted: 02/15/2011] [Indexed: 11/08/2022]
Abstract
Chloride intracellular channel (CLIC) 4 has diverse functions in membrane trafficking, apoptosis, angiogenesis and cell differentiation. CLIC4 is abundantly expressed in macrophages, but its role in innate immune functions is unclear. Here, we show that primary murine macrophages express increased amounts of CLIC4 after exposure to bacterial lipopolysaccharide (LPS). Endogenous CLIC4 level was significantly elevated in the brain, heart, lung, kidney, liver and spleen after LPS injection of mice. Stable macrophage lines overexpressing CLIC4 produced more TNF, IL-6, IL-12 and CCL5 than mock transfectants when exposed to LPS. To explore the role of CLIC4 in vivo, we generated CLIC4-null mice. These mice were protected from LPS-induced death, and had reduced serum levels of inflammatory cytokines. Upon infection with Listeria monocytogenes, CLIC4-deficient mice were impaired in their ability to clear infection, and their macrophages responded to Listeria by producing less inflammatory cytokines and chemokines than the WT controls. When challenged with LPS in vitro, deletion of clic4 gene had little effect on MAPK and NF-κB activation, but led to a reduced accumulation of phosphorylated interferon response factor 3 (IRF3) within macrophages. Conversely, overexpression of CLIC4 enhanced LPS-mediated IRF3. Thus, these findings suggest that CLIC4 is an LPS-induced product that can serve as a positive regulator of LPS signaling.
Collapse
Affiliation(s)
- Guoan He
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10065
| | - Yao Ma
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10065
- Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, China
| | - Szu-Yi Chou
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065
- Department of Ophthalmology, Weill Cornell Medical College, New York, NY 10065
| | - Huihong Li
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10065
| | - Chingwen Yang
- Gene Targeting Resource Center, Rockefeller University
| | - Jen-Zen Chuang
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065
- Department of Ophthalmology, Weill Cornell Medical College, New York, NY 10065
| | - Ching-Hwa Sung
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065
- Department of Ophthalmology, Weill Cornell Medical College, New York, NY 10065
| | - Aihao Ding
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10065
| |
Collapse
|
141
|
Atay S, Gercel-Taylor C, Taylor DD. Human trophoblast-derived exosomal fibronectin induces pro-inflammatory IL-1β production by macrophages. Am J Reprod Immunol 2011; 66:259-69. [PMID: 21410811 DOI: 10.1111/j.1600-0897.2011.00995.x] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
PROBLEM Our previous studies demonstrated that trophoblast-derived exosomes induced synthesis and release of pro-inflammatory cytokines, including interleukin-1β (IL-1β) by macrophages. The objective of this study was to characterize the mechanism and receptors associated with this induction. METHOD OF STUDY Exosomes were isolated from Sw71 trophoblast-conditioned media by ultrafiltration and ultracentrifugation. Using macrophages isolated from normal donors, cytochalasin D was used to block exosome uptake. Induction of IL-1β mRNA was investigated by qRT-PCR, pro-IL-1β protein by western immunoblotting, and mature IL-1β release by ELISA. RGD peptides were used to block fibronectin binding by macrophage α5β1 integrin. RESULTS Uptake of exosomes by macrophages was completely blocked by pre-treatment with cytochalasin D. Although induction of some cytokines (such as C4A and CCL11) requires uptake, induction of IL-1β occurred without exosome internalization. Cytochalasin D treatment did not inhibit exosome-mediated induction of IL-1β mRNA, production of the pro-protein, or release of mature IL-1β. Blocking of fibronectin binding using RGD peptides demonstrated the abrogation of exosome-mediated IL-1β production. CONCLUSION Although trophoblast-derived exosomes have been demonstrated to induce IL-1β, this is the first demonstration of IL-1β induction by exosome-associated fibronectin. Based on this pro-inflammatory role of exosome-associated fibronectin, it may represent an important general immunoregulatory mechanism.
Collapse
Affiliation(s)
- Safinur Atay
- Department of Microbiology & Immunology, University of Louisville School of Medicine, KY, USA
| | | | | |
Collapse
|
142
|
Nitric oxide-mediated intracellular growth restriction of pathogenic Rhodococcus equi can be prevented by iron. Infect Immun 2011; 79:2098-111. [PMID: 21383050 DOI: 10.1128/iai.00983-10] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Rhodococcus equi is an intracellular pathogen which causes pneumonia in young horses and in immunocompromised humans. R. equi arrests phagosome maturation in macrophages at a prephagolysosome stage and grows inside a privileged compartment. Here, we show that, in murine macrophages activated with gamma interferon and lipopolysaccharide, R. equi does not multiply but stays viable for at least 24 h. Whereas infection control of other intracellular pathogens by activated macrophages is executed by enhanced phagosome acidification or phagolysosome formation, by autophagy or by the interferon-inducible GTPase Irgm1, none of these mechanisms seems to control R. equi infection. Growth control by macrophage activation is fully mimicked by treatment of resting macrophages with nitric oxide donors, and inhibition of bacterial multiplication by either activation or nitric oxide donors is annihilated by cotreatment of infected macrophages with ferrous sulfate. Transcriptional analysis of the R. equi iron-regulated gene iupT demonstrates that intracellular R. equi encounters iron stress in activated, but not in resting, macrophages and that this stress is relieved by extracellular addition of ferrous sulfate. Our results suggest that nitric oxide is central to the restriction of bacterial access to iron in activated macrophages.
Collapse
|
143
|
Pedersen ER, Midttun Ø, Ueland PM, Schartum-Hansen H, Seifert R, Igland J, Nordrehaug JE, Ebbing M, Svingen G, Bleie Ø, Berge R, Nygård O. Systemic Markers of Interferon-γ–Mediated Immune Activation and Long-Term Prognosis in Patients With Stable Coronary Artery Disease. Arterioscler Thromb Vasc Biol 2011; 31:698-704. [DOI: 10.1161/atvbaha.110.219329] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Eva Ringdal Pedersen
- From the Section for Cardiology, Institute of Medicine (E.R.P., H.S.-H., J.E.N., R.B., O.N.), Section for Pharmacology, Institute of Medicine (P.M.U.), Institute of Public Health and Primary Health Care (J.I.), and Nordic Centre of Excellence in Human Nutrition–MitoHealth (H.S.-H., R.B., O.N.), University of Bergen, Bergen, Norway; Bevital A/S, Bergen, Norway (Ø.M.); Laboratory of Clinical Biochemistry (P.M.U.) and Department of Heart Disease (R.S., J.E.N., M.E., G.S., Ø.B., R.B., O.N.), Haukeland
| | - Øivind Midttun
- From the Section for Cardiology, Institute of Medicine (E.R.P., H.S.-H., J.E.N., R.B., O.N.), Section for Pharmacology, Institute of Medicine (P.M.U.), Institute of Public Health and Primary Health Care (J.I.), and Nordic Centre of Excellence in Human Nutrition–MitoHealth (H.S.-H., R.B., O.N.), University of Bergen, Bergen, Norway; Bevital A/S, Bergen, Norway (Ø.M.); Laboratory of Clinical Biochemistry (P.M.U.) and Department of Heart Disease (R.S., J.E.N., M.E., G.S., Ø.B., R.B., O.N.), Haukeland
| | - Per Magne Ueland
- From the Section for Cardiology, Institute of Medicine (E.R.P., H.S.-H., J.E.N., R.B., O.N.), Section for Pharmacology, Institute of Medicine (P.M.U.), Institute of Public Health and Primary Health Care (J.I.), and Nordic Centre of Excellence in Human Nutrition–MitoHealth (H.S.-H., R.B., O.N.), University of Bergen, Bergen, Norway; Bevital A/S, Bergen, Norway (Ø.M.); Laboratory of Clinical Biochemistry (P.M.U.) and Department of Heart Disease (R.S., J.E.N., M.E., G.S., Ø.B., R.B., O.N.), Haukeland
| | - Hall Schartum-Hansen
- From the Section for Cardiology, Institute of Medicine (E.R.P., H.S.-H., J.E.N., R.B., O.N.), Section for Pharmacology, Institute of Medicine (P.M.U.), Institute of Public Health and Primary Health Care (J.I.), and Nordic Centre of Excellence in Human Nutrition–MitoHealth (H.S.-H., R.B., O.N.), University of Bergen, Bergen, Norway; Bevital A/S, Bergen, Norway (Ø.M.); Laboratory of Clinical Biochemistry (P.M.U.) and Department of Heart Disease (R.S., J.E.N., M.E., G.S., Ø.B., R.B., O.N.), Haukeland
| | - Reinhard Seifert
- From the Section for Cardiology, Institute of Medicine (E.R.P., H.S.-H., J.E.N., R.B., O.N.), Section for Pharmacology, Institute of Medicine (P.M.U.), Institute of Public Health and Primary Health Care (J.I.), and Nordic Centre of Excellence in Human Nutrition–MitoHealth (H.S.-H., R.B., O.N.), University of Bergen, Bergen, Norway; Bevital A/S, Bergen, Norway (Ø.M.); Laboratory of Clinical Biochemistry (P.M.U.) and Department of Heart Disease (R.S., J.E.N., M.E., G.S., Ø.B., R.B., O.N.), Haukeland
| | - Jannicke Igland
- From the Section for Cardiology, Institute of Medicine (E.R.P., H.S.-H., J.E.N., R.B., O.N.), Section for Pharmacology, Institute of Medicine (P.M.U.), Institute of Public Health and Primary Health Care (J.I.), and Nordic Centre of Excellence in Human Nutrition–MitoHealth (H.S.-H., R.B., O.N.), University of Bergen, Bergen, Norway; Bevital A/S, Bergen, Norway (Ø.M.); Laboratory of Clinical Biochemistry (P.M.U.) and Department of Heart Disease (R.S., J.E.N., M.E., G.S., Ø.B., R.B., O.N.), Haukeland
| | - Jan Erik Nordrehaug
- From the Section for Cardiology, Institute of Medicine (E.R.P., H.S.-H., J.E.N., R.B., O.N.), Section for Pharmacology, Institute of Medicine (P.M.U.), Institute of Public Health and Primary Health Care (J.I.), and Nordic Centre of Excellence in Human Nutrition–MitoHealth (H.S.-H., R.B., O.N.), University of Bergen, Bergen, Norway; Bevital A/S, Bergen, Norway (Ø.M.); Laboratory of Clinical Biochemistry (P.M.U.) and Department of Heart Disease (R.S., J.E.N., M.E., G.S., Ø.B., R.B., O.N.), Haukeland
| | - Marta Ebbing
- From the Section for Cardiology, Institute of Medicine (E.R.P., H.S.-H., J.E.N., R.B., O.N.), Section for Pharmacology, Institute of Medicine (P.M.U.), Institute of Public Health and Primary Health Care (J.I.), and Nordic Centre of Excellence in Human Nutrition–MitoHealth (H.S.-H., R.B., O.N.), University of Bergen, Bergen, Norway; Bevital A/S, Bergen, Norway (Ø.M.); Laboratory of Clinical Biochemistry (P.M.U.) and Department of Heart Disease (R.S., J.E.N., M.E., G.S., Ø.B., R.B., O.N.), Haukeland
| | - Gard Svingen
- From the Section for Cardiology, Institute of Medicine (E.R.P., H.S.-H., J.E.N., R.B., O.N.), Section for Pharmacology, Institute of Medicine (P.M.U.), Institute of Public Health and Primary Health Care (J.I.), and Nordic Centre of Excellence in Human Nutrition–MitoHealth (H.S.-H., R.B., O.N.), University of Bergen, Bergen, Norway; Bevital A/S, Bergen, Norway (Ø.M.); Laboratory of Clinical Biochemistry (P.M.U.) and Department of Heart Disease (R.S., J.E.N., M.E., G.S., Ø.B., R.B., O.N.), Haukeland
| | - Øyvind Bleie
- From the Section for Cardiology, Institute of Medicine (E.R.P., H.S.-H., J.E.N., R.B., O.N.), Section for Pharmacology, Institute of Medicine (P.M.U.), Institute of Public Health and Primary Health Care (J.I.), and Nordic Centre of Excellence in Human Nutrition–MitoHealth (H.S.-H., R.B., O.N.), University of Bergen, Bergen, Norway; Bevital A/S, Bergen, Norway (Ø.M.); Laboratory of Clinical Biochemistry (P.M.U.) and Department of Heart Disease (R.S., J.E.N., M.E., G.S., Ø.B., R.B., O.N.), Haukeland
| | - Rolf Berge
- From the Section for Cardiology, Institute of Medicine (E.R.P., H.S.-H., J.E.N., R.B., O.N.), Section for Pharmacology, Institute of Medicine (P.M.U.), Institute of Public Health and Primary Health Care (J.I.), and Nordic Centre of Excellence in Human Nutrition–MitoHealth (H.S.-H., R.B., O.N.), University of Bergen, Bergen, Norway; Bevital A/S, Bergen, Norway (Ø.M.); Laboratory of Clinical Biochemistry (P.M.U.) and Department of Heart Disease (R.S., J.E.N., M.E., G.S., Ø.B., R.B., O.N.), Haukeland
| | - Ottar Nygård
- From the Section for Cardiology, Institute of Medicine (E.R.P., H.S.-H., J.E.N., R.B., O.N.), Section for Pharmacology, Institute of Medicine (P.M.U.), Institute of Public Health and Primary Health Care (J.I.), and Nordic Centre of Excellence in Human Nutrition–MitoHealth (H.S.-H., R.B., O.N.), University of Bergen, Bergen, Norway; Bevital A/S, Bergen, Norway (Ø.M.); Laboratory of Clinical Biochemistry (P.M.U.) and Department of Heart Disease (R.S., J.E.N., M.E., G.S., Ø.B., R.B., O.N.), Haukeland
| |
Collapse
|
144
|
de Carvalho LPS, Fischer SM, Marrero J, Nathan C, Ehrt S, Rhee KY. Metabolomics of Mycobacterium tuberculosis reveals compartmentalized co-catabolism of carbon substrates. ACTA ACUST UNITED AC 2011; 17:1122-31. [PMID: 21035735 DOI: 10.1016/j.chembiol.2010.08.009] [Citation(s) in RCA: 268] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2010] [Revised: 07/26/2010] [Accepted: 08/20/2010] [Indexed: 10/18/2022]
Abstract
Metabolic adaptation to the host environment is a defining feature of the pathogenicity of Mycobacterium tuberculosis (Mtb), but we lack biochemical knowledge of its metabolic networks. Many bacteria use catabolite repression as a regulatory mechanism to maximize growth by consuming individual carbon substrates in a preferred sequence and growing with diauxic kinetics. Surprisingly, untargeted metabolite profiling of Mtb growing on ¹³C-labeled carbon substrates revealed that Mtb could catabolize multiple carbon sources simultaneously to achieve enhanced monophasic growth. Moreover, when co-catabolizing multiple carbon sources, Mtb differentially catabolized each carbon source through the glycolytic, pentose phosphate, and/or tricarboxylic acid pathways to distinct metabolic fates. This unusual topologic organization of bacterial intermediary metabolism has not been previously observed and may subserve the pathogenicity of Mtb.
Collapse
Affiliation(s)
- Luiz Pedro S de Carvalho
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10065, USA
| | | | | | | | | | | |
Collapse
|
145
|
Venugopal A, Bryk R, Shi S, Rhee K, Rath P, Schnappinger D, Ehrt S, Nathan C. Virulence of Mycobacterium tuberculosis depends on lipoamide dehydrogenase, a member of three multienzyme complexes. Cell Host Microbe 2011; 9:21-31. [PMID: 21238944 PMCID: PMC3040420 DOI: 10.1016/j.chom.2010.12.004] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Revised: 10/18/2010] [Accepted: 12/01/2010] [Indexed: 01/24/2023]
Abstract
Mycobacterium tuberculosis (Mtb) adapts to persist in a nutritionally limited macrophage compartment. Lipoamide dehydrogenase (Lpd), the third enzyme (E3) in Mtb's pyruvate dehydrogenase complex (PDH), also serves as E1 of peroxynitrite reductase/peroxidase (PNR/P), which helps Mtb resist host-reactive nitrogen intermediates. In contrast to Mtb lacking dihydrolipoamide acyltransferase (DlaT), the E2 of PDH and PNR/P, Lpd-deficient Mtb is severely attenuated in wild-type and immunodeficient mice. This suggests that Lpd has a function that DlaT does not share. When DlaT is absent, Mtb upregulates an Lpd-dependent branched-chain keto acid dehydrogenase (BCKADH) encoded by pdhA, pdhB, pdhC, and lpdC. Without Lpd, Mtb cannot metabolize branched-chain amino acids and potentially toxic branched-chain intermediates accumulate. Mtb deficient in both DlaT and PdhC phenocopies Lpd-deficient Mtb. Thus, Mtb critically requires BCKADH along with PDH and PNR/P for pathogenesis. These findings position Lpd as a potential target for anti-infectives against Mtb.
Collapse
Affiliation(s)
- Aditya Venugopal
- Department of Microbiology and Immunology, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065
- Program in Immunology and Microbial Pathogenesis, Weill Graduate School of Medical Sciences of Cornell University
| | - Ruslana Bryk
- Department of Microbiology and Immunology, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065
| | - Shuangping Shi
- Department of Microbiology and Immunology, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065
- Program in Immunology and Microbial Pathogenesis, Weill Graduate School of Medical Sciences of Cornell University
| | - Kyu Rhee
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065
| | - Poonam Rath
- Department of Microbiology and Immunology, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065
- Program in Immunology and Microbial Pathogenesis, Weill Graduate School of Medical Sciences of Cornell University
| | - Dirk Schnappinger
- Department of Microbiology and Immunology, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065
| | - Sabine Ehrt
- Department of Microbiology and Immunology, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065
- Program in Immunology and Microbial Pathogenesis, Weill Graduate School of Medical Sciences of Cornell University
| | - Carl Nathan
- Department of Microbiology and Immunology, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065
- Program in Immunology and Microbial Pathogenesis, Weill Graduate School of Medical Sciences of Cornell University
| |
Collapse
|
146
|
Kang DD, Lin Y, Moreno JR, Randall TD, Khader SA. Profiling early lung immune responses in the mouse model of tuberculosis. PLoS One 2011; 6:e16161. [PMID: 21249199 PMCID: PMC3020951 DOI: 10.1371/journal.pone.0016161] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Accepted: 12/08/2010] [Indexed: 01/01/2023] Open
Abstract
Tuberculosis (TB) is caused by the intracellular bacteria Mycobacterium tuberculosis, and kills more than 1.5 million people every year worldwide. Immunity to TB is associated with the accumulation of IFNγ-producing T helper cell type 1 (Th1) in the lungs, activation of M.tuberculosis-infected macrophages and control of bacterial growth. However, very little is known regarding the early immune responses that mediate accumulation of activated Th1 cells in the M.tuberculosis-infected lungs. To define the induction of early immune mediators in the M.tuberculosis-infected lung, we performed mRNA profiling studies and characterized immune cells in M.tuberculosis-infected lungs at early stages of infection in the mouse model. Our data show that induction of mRNAs involved in the recognition of pathogens, expression of inflammatory cytokines, activation of APCs and generation of Th1 responses occurs between day 15 and day 21 post infection. The induction of these mRNAs coincides with cellular accumulation of Th1 cells and activation of myeloid cells in M.tuberculosis-infected lungs. Strikingly, we show the induction of mRNAs associated with Gr1+ cells, namely neutrophils and inflammatory monocytes, takes place on day 12 and coincides with cellular accumulation of Gr1+ cells in M.tuberculosis-infected lungs. Interestingly, in vivo depletion of Gr1+ neutrophils between days 10-15 results in decreased accumulation of Th1 cells on day 21 in M.tuberculosis-infected lungs without impacting overall protective outcomes. These data suggest that the recruitment of Gr1+ neutrophils is an early event that leads to production of chemokines that regulate the accumulation of Th1 cells in the M.tuberculosis-infected lungs.
Collapse
Affiliation(s)
- Dongwan D. Kang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Division of Infectious Diseases, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Yinyao Lin
- Division of Infectious Diseases, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Javier-Rangel Moreno
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Troy D. Randall
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Shabaana A. Khader
- Division of Infectious Diseases, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
147
|
Beaulieu AM, Rath P, Imhof M, Siddall ME, Roberts J, Schnappinger D, Nathan CF. Genome-wide screen for Mycobacterium tuberculosis genes that regulate host immunity. PLoS One 2010; 5:e15120. [PMID: 21170273 PMCID: PMC3000826 DOI: 10.1371/journal.pone.0015120] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Accepted: 10/22/2010] [Indexed: 12/17/2022] Open
Abstract
In spite of its highly immunogenic properties, Mycobacterium tuberculosis (Mtb) establishes persistent infection in otherwise healthy individuals, making it one of the most widespread and deadly human pathogens. Mtb's prolonged survival may reflect production of microbial factors that prevent even more vigorous immunity (quantitative effect) or that divert the immune response to a non-sterilizing mode (qualitative effect). Disruption of Mtb genes has produced a list of several dozen candidate immunomodulatory factors. Here we used robotic fluorescence microscopy to screen 10,100 loss-of-function transposon mutants of Mtb for their impact on the expression of promoter-reporter constructs for 12 host immune response genes in a mouse macrophage cell line. The screen identified 364 candidate immunoregulatory genes. To illustrate the utility of the candidate list, we confirmed the impact of 35 Mtb mutant strains on expression of endogenous immune response genes in primary macrophages. Detailed analysis focused on a strain of Mtb in which a transposon disrupts Rv0431, a gene encoding a conserved protein of unknown function. This mutant elicited much more macrophage TNFα, IL-12p40 and IL-6 in vitro than wild type Mtb, and was attenuated in the mouse. The mutant list provides a platform for exploring the immunobiology of tuberculosis, for example, by combining immunoregulatory mutations in a candidate vaccine strain.
Collapse
Affiliation(s)
- Aimee M. Beaulieu
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, United States of America
| | - Poonam Rath
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, United States of America
| | - Marianne Imhof
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, United States of America
| | - Mark E. Siddall
- Division of Invertebrate Zoology, American Museum of Natural History, New York, New York, United States of America
| | - Julia Roberts
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, United States of America
| | - Dirk Schnappinger
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, United States of America
| | - Carl F. Nathan
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
148
|
Slaney CY, Toker A, La Flamme A, Bäckström BT, Harper JL. Naïve blood monocytes suppress T-cell function. A possible mechanism for protection from autoimmunity. Immunol Cell Biol 2010; 89:7-13. [PMID: 21060323 DOI: 10.1038/icb.2010.110] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In certain disease context, cells of the monocyte/macrophage lineage are known to exhibit T-cell suppressor function. However, whether naïve monocytes are also able to suppress T-cell responses has not been previously investigated. In this study, we have discovered that CD11b(+)Ly6G(-) mononuclear cells in the blood of naïve mice are potent suppressors of T-cell proliferation in vitro. The suppression of T-cell proliferation requires cell-cell contact and is partially dependent on nitric oxide production. Following the induction of experimental autoimmune encephalomyelitis in mice, the suppressor function of this blood CD11b(+)Ly6G(-) cell population is impaired. Therefore, blood CD11b(+)Ly6G(-) cells appear to be intrinsically suppressive and may have a key role in maintaining immune homoeostasis. Loss of this suppressive function may contribute to development of autoimmunity.
Collapse
Affiliation(s)
- Clare Y Slaney
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | | | | | | | | |
Collapse
|
149
|
Bowden SD, Ramachandran VK, Knudsen GM, Hinton JCD, Thompson A. An incomplete TCA cycle increases survival of Salmonella Typhimurium during infection of resting and activated murine macrophages. PLoS One 2010; 5:e13871. [PMID: 21079785 PMCID: PMC2975626 DOI: 10.1371/journal.pone.0013871] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Accepted: 10/14/2010] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND In comparison to the comprehensive analyses performed on virulence gene expression, regulation and action, the intracellular metabolism of Salmonella during infection is a relatively under-studied area. We investigated the role of the tricarboxylic acid (TCA) cycle in the intracellular replication of Salmonella Typhimurium in resting and activated macrophages, epithelial cells, and during infection of mice. METHODOLOGY/PRINCIPAL FINDINGS We constructed deletion mutations of 5 TCA cycle genes in S. Typhimurium including gltA, mdh, sdhCDAB, sucAB, and sucCD. We found that the mutants exhibited increased net intracellular replication in resting and activated murine macrophages compared to the wild-type. In contrast, an epithelial cell infection model showed that the S. Typhimurium ΔsucCD and ΔgltA strains had reduced net intracellular replication compared to the wild-type. The glyoxylate shunt was not responsible for the net increased replication of the TCA cycle mutants within resting macrophages. We also confirmed that, in a murine infection model, the S. Typhimurium ΔsucAB and ΔsucCD strains are attenuated for virulence. CONCLUSIONS/SIGNIFICANCE Our results suggest that disruption of the TCA cycle increases the ability of S. Typhimurium to survive within resting and activated murine macrophages. In contrast, epithelial cells are non-phagocytic cells and unlike macrophages cannot mount an oxidative and nitrosative defence response against pathogens; our results show that in HeLa cells the S. Typhimurium TCA cycle mutant strains show reduced or no change in intracellular levels compared to the wild-type. The attenuation of the S. Typhimurium ΔsucAB and ΔsucCD mutants in mice, compared to their increased net intracellular replication in resting and activated macrophages suggest that Salmonella may encounter environments within the host where a complete TCA cycle is advantageous.
Collapse
Affiliation(s)
| | | | | | - Jay C. D. Hinton
- Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College, Dublin, Ireland
| | | |
Collapse
|
150
|
Gibbings S, Elkins ND, Fitzgerald H, Tiao J, Weyman ME, Shibao G, Fini MA, Wright RM. Xanthine oxidoreductase promotes the inflammatory state of mononuclear phagocytes through effects on chemokine expression, peroxisome proliferator-activated receptor-{gamma} sumoylation, and HIF-1{alpha}. J Biol Chem 2010; 286:961-75. [PMID: 21059659 DOI: 10.1074/jbc.m110.150847] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The protective effects of pharmacological inhibitors of xanthine oxidoreductase (XOR) have implicated XOR in many inflammatory diseases. Nonetheless, the role played by XOR during inflammation is poorly understood. We previously observed that inhibition of XOR within the inflammatory mononuclear phagocytes (MNP) prevented neutrophil recruitment during adoptive transfer demonstrating the role of XOR in MNP-mediated neutrophil recruitment. To further explore the role of XOR in the inflammatory state of MNP, we studied MNP isolated from inflammatory lungs combined with analyses of MNP cell lines. We demonstrated that XOR activity was increased in inflammatory MNP following insufflation of Th-1 cytokines in vivo and that activity was specifically increased by MNP differentiation. Inhibition of XOR reduced levels of CINC-1 secreted by MNP. Expression of peroxisome proliferator-activated receptor γ (PPARγ) in purified rat lung MNP and MNP cell lines reflected both the presence of PPARγ isoforms and PPARγ SUMOylation, and XOR inhibitors increased levels of SUMO-PPARγ in MNP cell lines. Both ectopic overexpression of XOR cDNA and uric acid supplementation reduced SUMO-PPARγ in MNP cells. Levels of the M2 markers CD36, CD206, and arginase-1 were modulated by uric acid and oxonic acid, whereas siRNA to SUMO-1 or PIAS-1 also reduced arginase-1 in RAW264.7 cells. We also observed that HIF-1α was increased by XOR inhibitors in inflammatory MNP and in MNP cell lines. These data demonstrate that XOR promotes the inflammatory state of MNP through effects on chemokine expression, PPARγ SUMOylation, and HIF-1α and suggest that strategies for inhibiting XOR may be valuable in modulating lung inflammatory disorders.
Collapse
Affiliation(s)
- Sophie Gibbings
- Division of Pulmonary Sciences, Division of Pulmonary Sciences, University of Colorado Denver, Aurora, Colorado 80045, USA
| | | | | | | | | | | | | | | |
Collapse
|