Skip to main content
Advertisement
  • Loading metrics

CD4+ T Cell-derived IL-10 Promotes Brucella abortus Persistence via Modulation of Macrophage Function

  • Mariana N. Xavier,

    Affiliations Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, California, United States of America, Departamento de Clínica e Cirurgia Veterinárias, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil

  • Maria G. Winter,

    Affiliation Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, California, United States of America

  • Alanna M. Spees,

    Affiliation Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, California, United States of America

  • Kim Nguyen,

    Affiliation Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, California, United States of America

  • Vidya L. Atluri,

    Affiliation Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, California, United States of America

  • Teane M. A. Silva,

    Affiliations Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, California, United States of America, Departamento de Clínica e Cirurgia Veterinárias, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil

  • Andreas J. Bäumler,

    Affiliation Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, California, United States of America

  • Werner Müller,

    Affiliation Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom

  • Renato L. Santos,

    Affiliation Departamento de Clínica e Cirurgia Veterinárias, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil

  • Renée M. Tsolis

    rmtsolis@ucdavis.edu

    Affiliation Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, California, United States of America

Abstract

Evasion of host immune responses is a prerequisite for chronic bacterial diseases; however, the underlying mechanisms are not fully understood. Here, we show that the persistent intracellular pathogen Brucella abortus prevents immune activation of macrophages by inducing CD4+CD25+ T cells to produce the anti-inflammatory cytokine interleukin-10 (IL-10) early during infection. IL-10 receptor (IL-10R) blockage in macrophages resulted in significantly higher NF-kB activation as well as decreased bacterial intracellular survival associated with an inability of B. abortus to escape the late endosome compartment in vitro. Moreover, either a lack of IL-10 production by T cells or a lack of macrophage responsiveness to this cytokine resulted in an increased ability of mice to control B. abortus infection, while inducing elevated production of pro-inflammatory cytokines, which led to severe pathology in liver and spleen of infected mice. Collectively, our results suggest that early IL-10 production by CD25+CD4+ T cells modulates macrophage function and contributes to an initial balance between pro-inflammatory and anti-inflammatory cytokines that is beneficial to the pathogen, thereby promoting enhanced bacterial survival and persistent infection.

Author Summary

Brucella spp. are pathogens causing chronic intracellular infections that evade detection by pattern recognition receptors of the innate immune system. In this work, we tested the hypothesis that, in addition to eliciting a weak proinflammatory response during infection, induction of an immunoregulatory environment early during infection could promote persistent infection. Our results show that IL-10 produced at early time points is important for blunting inflammatory responses to B. abortus in infected tissues. CD4+ T cells are an important source of this cytokine, since mice lacking T cell-derived IL-10 exhibited increased inflammatory pathology and also were better able to control B. abortus infection. A target of this CD4 T cell-derived IL-10 is macrophages, since treatment of these cells with IL-10 in vitro supported intracellular replication of B. abortus, while blocking IL-10 restricted the ability of B. abortus to exit the phagolysosomal compartment and replicate intracellularly. Further, mice conditionally deficient for IL-10 receptor on macrophages were better able to control infection with B. abortus. Taken together, our results support a model in which IL-10 production by T cells promotes chronic infection by rendering macrophages more permissive for intracellular replication of B. abortus.

Introduction

Persistent bacterial infections have a significant impact on public health [1]. While evasion of host immune responses is a prerequisite for these chronic infections, the underlying mechanisms are not fully understood. Human brucellosis, caused by the intracellular gram-negative coccobacilli Brucella spp., is considered one of the most important zoonotic diseases worldwide, with more than 500,000 new human cases reported annually [2]. The disease is characterized by a long incubation period that leads to a chronic, sometimes lifelong, debilitating infection with serious clinical manifestations such as fever, arthritis, hepatomegaly, and splenomegaly [3], [4]. Human and animal brucellosis share many similarities, such as persistence in tissues of the mononuclear phagocyte system, including spleen, liver, lymph nodes, and bone marrow [4]. Therefore, the use of animal models such as mice has been an important tool to better characterize the immune response to Brucella infection that leads to long-term bacterial persistence and chronic disease.

There is general agreement that the initial interferon gamma (IFN-γ) mediated Th1 immune response is crucial for the control of Brucella infection, since absence of IFN-γ results in decreased control of bacterial growth [5], [6] and IFN-γ-deficient C57BL/6 mice succumb to overwhelming disease [7]. However, the inflammatory response induced by Brucella spp. in vivo is much milder than that observed with pyogenic infections such as salmonellosis, suggesting the stealth of Brucella as a possible reason for the absence of early proinflammatory responses [8], [9]. Recent studies have shown that Brucella spp. use both passive and active mechanisms to evade initial innate immune recognition through toll-like receptors (TLRs) [10]. Although avoidance of TLR recognition is a key factor in the lack of initial inflammation during Brucella infection, how subsequent interactions of Brucella with the host immune system result in chronic disease is poorly understood.

Interleukin-10 (IL-10) is an immunoregulatory cytokine produced by most T cell subsets, B cells, neutrophils, macrophages, and some dendritic cell subsets [11]. It is suggested that by acting on antigen-presenting cells such as macrophages, IL-10 can inhibit the development of Th1 type responses [12]. In the context of infectious diseases, it is believed that the host uses IL-10 to control over-exuberant immune responses to pathogenic microorganisms in order to limit tissue damage [11]. Interestingly, studies using chronic pathogens such as Leishmania major [13], human cytomegalovirus [14], or Mycobacterium tuberculosis (reviewed in [15]) have demonstrated that the absence of IL-10 leads to a better clearance of these pathogens, with variable degrees of immunopathology. These studies suggest that pathogens have developed mechanisms to take advantage of the host immune-regulation in order to persist for longer periods and establish chronic infection.

Similar to other chronic pathogens, B. abortus infection induces IL-10 production [5], [6], [16]. Moreover, IL-10 gene polymorphisms have been associated with increased susceptibility to human brucellosis [17]. However, questions regarding the impact of IL-10 in B. abortus persistence and establishment of chronic infection, as well as the cell types responsible for this cytokine production remain to be answered. Therefore, we used IL-10 deficient mice to determine the role of IL-10 in modulating the initial immune response to Brucella infection. Furthermore, using cell-specific knock-out mice, we elucidated the immunological mechanisms underlying IL-10 induced immune-regulation during Brucellosis.

Results

Lack of IL-10 production during early B. abortus infection results in lower bacterial survival and increased pathology in vivo

IL-10 has an important role in controlling the immune response induced by different inflammatory processes [15]. Moreover, Brucella infection has been shown to induce IL-10 production by splenocytes in vitro [5] and during intravenous in vivo infection [6], [16]. To determine the time-course of IL-10 production during B. abortus infection, C57BL/6 mice were infected intraperitoneally (IP) with 5×105 CFU of the virulent B. abortus strain 2308 and IL-10 production was determined at 3, 9, 15, and 21 days post-infection (d.p.i.). Infected mice exhibited significantly higher levels of IL-10 in the serum (Fig. 1A), which was associated with increased IL-10 transcript levels in the spleen (Fig. 1B) and liver (Fig. 1C) as early as 3 d.p.i. Importantly, significantly increased levels of IL-10 in serum and infected organs was only detected until 15 d.p.i., suggesting a possible regulatory function for this cytokine during acute Brucella infection.

thumbnail
Figure 1. Brucella abortus induces IL-10 production by infected organs during early in vivo infection.

(A) IL-10 levels in serum of C57BL/6 infected with B. abortus 2308 for 3, 9, 15, and 21 days compared to uninfected mice (inoculated with sterile PBS). (B, C) qRT-PCR analysis of IL-10 gene expression in spleen (B) and liver (C) of C57BL/6 mice infected with B. abortus 2308 for 3, 9, 15, and 21 days compared to uninfected controls. n = 5. (*) represents P<0.05 when compared to control. (**) represents P<0.05 when compared to days 15 and day 21 for (C) using unpaired t- test statistical analysis.

https://doi.org/10.1371/journal.ppat.1003454.g001

To further investigate if IL-10 plays a role in modulating the inflammatory response during acute brucellosis, C57BL/6 wild-type and Il10-deficient mice (IL-10−/−) were infected IP with 5×105 CFU of B. abortus 2308 and responses were evaluated at 9 d.p.i. Interestingly, IL-10−/− mice had significantly lower bacterial survival in both the spleen (Fig. 2A) and the liver (Fig. 2B). IL-10−/− mice also exhibited increased induction of pro-inflammatory cytokines such as IFN-γ, interleukin-6 (IL-6) and tumor necrosis factor alpha (TNF-α) in infected organs (Fig. 2C) and in serum from infected mice (Fig. 2D).

thumbnail
Figure 2. Lack of IL-10 results in lower bacterial survival and increased pathological changes during early Brucella abortus infection in vivo.

(A,B) Colonization of spleen (A) and liver (B) of control or IL-10−/− mice by B. abortus at 9 d.p.i. (C) qRT-PCR analysis of IFN-γ, IL-6 and TNF-α gene expression in spleens of C57BL/6 and IL-10−/− mice infected with B. abortus 2308 for 9 days (similar results for liver). (D) IL-6 and TNF-α levels measured by ELISA in serum of C57BL/6 and IL-10−/− mice infected with B. abortus 2308 for 9 days. n = 5. Values represent mean ± SEM. *P<0.05 using unpaired t-test statistical analysis.( E) Histopathology score of spleen and liver from C57BL/6 and IL-10−/− mice infected with B. abortus 2308 for 9 days. (F) Representative pictures from (E). Black arrow shows microgranulomas, white arrowheads shows neutrophilic infiltrate and upper case N shows areas of coagulative necrosis (×20). n = 5. Values represent individual mice (black circles) and geometric mean (black dash). *P<0.05 using Mann-Whitney statistical analysis.

https://doi.org/10.1371/journal.ppat.1003454.g002

The typical tissue response to Brucella infection is granulomatous inflammation [4]. However, in the spleen and liver of IL-10−/− mice B. abortus infection resulted in development of an acute inflammatory response characterized by vasculitis and thrombosis, necrosis, and influx of neutrophils (Fig. 2E and 2F). Collectively, these data demonstrate a critical role for IL-10 in modulating the initial inflammation and pathology in response to B. abortus infection, which in turn benefits the pathogen due to enhanced bacterial survival.

CD4+CD25+ T cells are the main IL-10 producers during early B. abortus infection in vivo

IL-10 can be produced by different T cell subsets, as well as by B cells, neutrophils, macrophages, and some DC subsets [18]. To determine the cell types responsible for IL-10 production during early B. abortus infection, Il10-GFP reporter mice [19] were infected IP with 5×105 CFU of B. abortus and IL-10 producing cells were identified at 3 and 9 d.p.i. by flow cytometry. A significant increase in the number of IL-10 producing T cells was observed in infected mice at 3 and 9 days post infection, whereas macrophages presented increased production of IL-10 only at 3 d.p.i. Moreover, the number of IL-10 producing B cells, neutrophils, and dendritic cells did not change significantly when compared to uninfected mice (Fig. 3A and 3B, and data not shown). Importantly, even though a significant increase in the number of IL-10 producing CD8+ T cells was observed (Fig. 3C), a tenfold higher number of IL-10 producing T cells was observed in the CD4+ T cell population (Fig. 3C). No IL-10 production by γδ T cells was observed (data not shown).

thumbnail
Figure 3. CD4+CD25+ T cells are the main producers of IL-10 during early Brucella abortus in vivo infection.

(A) Flow cytometry measurement of IL-10 expression in splenic T cells, B cells, and macrophages from C57BL/6 IL-10 GFP-reporter mice infected with B. abortus 2308 for 3 and 9 days. (B) Representative data plot of IL-10 expression in splenic T cells (CD3), B cells (B220), and macrophages (CD11b+F4/80+ cells) from C57BL/6 IL-10 GFP-reporter mice. (C) Flow cytometry measurement of IL-10 expression in splenic CD8+ T cells and CD4+ T cells from C57BL/6 IL-10 GFP-reporter mice infected with B. abortus 2308 for 9 days. (D) Flow cytometry measurement of IL-10 expression in splenic CD4+CD25+ T cells and CD4+CD25 T cells from C57BL/6 IL-10 GFP-reporter mice infected with B. abortus 2308 for 9 days. (E) Representative data plot of IL-10 expression in splenic CD4+CD25+ (upper panel) and CD4+CD25 (lower panel) T cells from C57BL/6 IL-10 GFP-reporter mice. Values represent mean ± SEM. n = 4. (*) represents P<0.05 relative to uninfected control (day 0), (**) represent P<0.05 relative to day 3 infection for (A), relative to CD8+ day 9 infected for (C) and relative to CD4+CD25 for (D) using unpaired t-test statistical analysis. FMO = fluorescence minus one.

https://doi.org/10.1371/journal.ppat.1003454.g003

Various IL-10 producing CD4+ T cells have been described, including the CD4+CD25+ subset [20]. Interestingly, an expansion of the CD4+CD25+ T cell population was observed in the spleen of B. abortus infected mice at 9 d.p.i. (Fig. S1). Moreover, a 5-fold greater number of IL-10 producing CD4+CD25+ T cells was detected at 9 d.p.i., compared to IL-10 producing CD4+CD25 T cells (Fig. 3D and 3E), suggesting that the CD4+CD25+ T cell subset is the major population responsible for IL-10 production during acute brucellosis in the mouse.

IL-10 production by T cells is required for B. abortus persistence and for pathology regulation in vivo

Our previous data suggested that T cells and possibly macrophages would be the main cell types producing IL-10 during early Brucella infection. Therefore, to further investigate the importance of macrophage derived IL-10 during Brucella infection, we generated Il-10flox/floxLysMCre+/− (IL-10flox/LysMCre) mice, which have macrophages and neutrophils that are unable to produce IL-10. IL-10flox/LysMCre mice and littermate Il-10flox/floxLysMCre−/− controls were infected IP with 5×105 CFU of B. abortus 2308, and at 3, 9 and 21 d.p.i. disease progression was evaluated. Interestingly, IL-10flox/LysMCre infected mice exhibited decreased levels of IL-10 in the serum (Fig. 4A) when compared to control animals only at 3 d.p.i., Moreover, IL-10flox/LysMCre mice exhibited increased ability to control B. abortus infection in both spleen (Fig. 4B) and liver (Fig. 4C), only at initial stages of infection. This increased host resistance was accompanied by significantly higher levels of the pro-inflammatory cytokines IL-6, TNF-α and IFN-γ in serum (Fig. 4D), spleen (Fig. 4E) and liver (data not shown) as well as increased histopathological lesions in infected organs (Fig. S2) of IL-10flox/LysMCre mice when compared to littermate controls (Fig. 4D, 4E and data not shown). Collectively, this data suggests that macrophage derived IL-10 plays a limited role in development of the chronic disease caused by B. abortus and raises the possibility that T-cells could, indeed, be the main cell type responsible for IL-10 production during early Brucellosis.

thumbnail
Figure 4. IL-10 production by macrophages and/or neutrophils is not required for Brucella abortus long-term persistence in vivo.

(A) ELISA essay for IL-10 production in serum from littermate control mice (grey bars) compared with IL10flox/LysMCre mice (white bars) at 3, 9 and 21 d.p.i. (B, C) B. abortus 2308 CFU counts in spleen (B) and liver (C) from littermate control mice (grey bars) compared with IL10flox/LysMCre mice (white bars) at 3, 9 and 21 d.p.i.). (D) ELISA essay for IL-6 and TNF-α production in serum from littermate control mice (grey bars) compared with IL10flox/LysMCre mice (white bars) at 3, 9 and 21 d.p.i. (E) RT-PCR analysis of pro-inflammatory cytokines in spleens from littermate control mice (grey bars) compared with IL10flox/LysMCre mice (white bars) at 3, 9 and 21 d.p.i.

https://doi.org/10.1371/journal.ppat.1003454.g004

Therefore, to further investigate the importance of T-cell derived IL-10 during Brucella infection, we used Il-10flox/floxCd4Cre+/−(IL-10flox/CD4Cre) mice, which have T cells that are unable to produce IL-10 [21]. IL-10flox/CD4Cre and littermate Il-10flox/floxCd4Cre/− control mice were infected IP with 5×105 CFU of B. abortus 2308, and at 3, 9 ,21 and 42 d.p.i., disease progression was evaluated. Remarkably, IL-10flox/CD4Cre infected mice exhibited lower levels of IL-10 in serum (Fig. S3A), and spleen (Fig. S3B) when compared to control animals at 9 d.p.i., providing compelling support for the hypothesis that T-cells are a major source of IL-10 production during early B. abortus infection.

Previous results using IL-10−/− and IL-10flox/LysMCre mice (Fig. 2 and 4) suggested that IL-10 was important for initial B. abortus persistence in the host. Therefore, we investigated whether the lack of T cell-derived IL-10 would affect bacterial persistence. Indeed, IL-10flox/CD4Cre mice exhibited significantly improved control of B. abortus infection in the spleen and liver at 9, 21 and 42 d.p.i. (Fig. 5A and 5B).

thumbnail
Figure 5. IL-10 production by T cells is required for Brucella abortus persistence and for control of pathology in vivo.

(A, B) B. abortus 2308 CFU counts in spleen (A) and liver (B) from littermate control (grey bars) compared with IL10flox/CD4Cre mice (white bars) at 3, 9, 21 and 42 d.p.i. (C) Measurement of mouse weight over time in grams of littermate control mice (grey dots) compared with IL10flox/CD4Cre mice (white dots). (D) ELISA essay for IL-6 and TNF-α production in serum from littermate control mice (grey bars) compared with IL10flox/CD4Cre mice (white bars) at 3, 9, 21 and 42 d.p.i. (E) qRT-PCR analysis of pro-inflammatory cytokines genes (Ifnγ, Il6 and Tnfα) in spleen from littermate control mice (grey bars) compared with IL10flox/CD4Cre mice (white bars) at 3, 9, 21 and 42 d.p.i. n = 5. Values represent mean ± SEM. (*) represents P<0.05 using unpaired t-test statistical analysis.

https://doi.org/10.1371/journal.ppat.1003454.g005

Controlling B. abortus replication could be beneficial to the host, since the ability to survive for longer periods is a key mechanism for chronic pathogens to thrive. However, loss of IL-10 driven immune modulation has been shown to cause severe and sometimes lethal inflammatory responses in different infectious disease models [15]. To determine the disease progression, weight changes in IL-10flox/CD4Cre and control mice were followed at 3, 9, 15, 21, 28, 35 and 42 d.p.i. (Fig. 5C). To ensure that any detectable change in weight was the result of B. abortus infection, uninfected IL-10flox/CD4Cre and littermate control mice were also used (data not shown). IL-10flox/CD4Ce mice exhibited significantly decreased weight gain during early infection and, by 21 days post-infection, they started to behave like control animals. IL-10flox/CD4Cre uninfected mice did not exhibit any slower weight gain and behaved like uninfected control mice (data not shown). Moreover, increased levels of the pro-inflammatory cytokines IFN-γ, IL-6, and TNF-α were observed in serum (Fig. 5D) spleen (Fig. 5E) and liver (Fig. S3C) of IL-10flox/CD4Cre at 3, 9 days and, to a lesser extent, at 21 d.p.i.

To determine if the significantly reduced weight gain and higher induction of pro-inflammatory cytokines were associated with a detrimental inflammatory response, spleen and liver sections from IL-10flox/CD4Cre and control mice were blindly evaluated by veterinary pathologists (MNX and TMS). Interestingly, IL-10flox/CD4Cre mice exhibited increased pathology, characterized by marked influx of neutrophils and histiocytes in the spleen (Fig. 6A and 6C), as well as tissue necrosis and multifocal neutrophilic vasculitis and thrombosis in the liver (Fig. 6B and 6C) at 3 and 9 d.p.i., suggesting an acute inflammatory response. Importantly, a hallmark of B. abortus infection is a mild initial pro-inflammatory response, which leads to a chronic infection characterized by formation of granulomas in infected organs [4], [22]. However, by 21 d.p.i., IL-10flox/CD4Cre mice exhibited decreased granuloma formation in the spleen (Fig. 6A and 6C) when compared to littermate control mice. Taken together, these data demonstrate that T cell-derived IL-10 production during early B. abortus infection is crucial for the development of the chronic disease and morbidity caused by B. abortus, and limits the production of the pro-inflammatory response necessary to control the infection. However, the cell types affected by the IL-10 production during Brucella infection remained unclear.

thumbnail
Figure 6. IL-10 production by T cells is required for control of Brucella abortus induced pathology in vivo.

(A, B) Histopathology score of spleen (A) and liver (B) from littermate mice (Cre−/−) compared with IL10flox/CD4Cre mice (Cre+/−) at 3, 9, 21 and 42 d.p.i. (C) Representative histopathology figures from (A,B) - Black arrows indicate microgranulomas, white arrowheads show neutrophilic infiltrate, and white upper case WP indicates white pulp (×20). n = 5. (*) represents P<0.05 using Mann-Whitney statistical analysis.

https://doi.org/10.1371/journal.ppat.1003454.g006

Lack of IL-10 results in lower B. abortus survival in infected macrophages due to bacterial inability to escape late endosome

Since Brucella spp. are known to invade and survive inside phagocytic cells such as macrophages [23], we hypothesized that macrophages could be the cell type affected by IL-10 during Brucella infection. To determine the effect of IL-10 on B. abortus survival during macrophage infection in vitro, bone marrow derived macrophages (BMDM) from C57BL/6 and IL-10−/− mice were infected with B. abortus 2308 (MOI = 100) and the bacterial survival was measured at 1, 8, and 24 h post-infection (h.p.i.) (Fig. 7A). B. abortus infected wild-type BMDM produced significant amounts of IL-10 (Fig. S4A). B. abortus exhibited a significantly decreased ability to survive inside BMDM from IL-10−/− mice at 8 and 24 h.p.i. when compared to BMDM from wild-type mice. Importantly, B. abortus infected IL-10−/− BMDM did not exhibit increased cell death, determined by LDH assay (data not shown). Moreover, to ensure that the observed effect resulted from an absence of IL-10 production, recombinant IL-10 (rIL-10) was added to the IL-10−/− BMDM media during infection (Fig. 7A). As expected, the addition of rIL-10 restored B. abortus survival inside macrophages. Results similar to those described above (Fig. 7A) were also observed in IFNγ-activated BMDM (data not shown).

thumbnail
Figure 7. Lack of endogenous IL-10 results in lower Brucella abortus survival inside macrophages due to bacterial inability to escape the late endosome.

(A) Survival of B. abortus over time in BMDM from C57BL/6 WT control mice and IL-10 deficient mice with (IL-10−/−+rIL-10) or without (IL-10−/−) rIL-10 added to the culture. (B) Confocal microscopy of BMDM from C57BL/6 WT control mice and IL-10 deficient mice with (IL-10−/−+rIL-10) or without (IL-10−/−) added rIL-10, infected with mCherry-expressing B. abortus 2308 for 24 h. Colocalization of the bacteria (red) with the late endosomal marker LAMP-1 (green) is shown by arrowheads, while arrows indicate bacteria that were able to escape the endosomal compartment. Intracellular survival of B. abortus in each treatment group is shown in Panel (A). (C) Quantification of % of colocalization of mCherry-expressing B. abortus and LAMP1 in infected macrophages shown in (B). Representative results of two independent experiments are shown. n = 4. Values represent mean ± SEM. *P<0.05 using unpaired t-test statistical analysis.

https://doi.org/10.1371/journal.ppat.1003454.g007

The ability of Brucella spp. to persist and replicate within macrophages involves a temporary fusion of Brucella-containing vacuole with the late endosome/lysosome during the initial hours post-infection, and subsequent exclusion of the endosomal/lysosomal proteins from the Brucella-containing vacuole [24]. To determine if B. abortus inability to persist inside macrophages was due to changes in the pathogen's intracellular trafficking, we infected wild-type and IL-10−/− BMDM with B. abortus expressing mCherry (MOI = 100) and bacterial co-localization with the late endosome marker LAMP1 was determined at 24 h.p.i. by confocal microscopy. Interestingly, while the majority of B. abortus was found to lack LAMP1 in wild type BMDM, the opposite was found in BMDM derived from IL-10−/− mice, in which over 80% of bacteria were co-localized with LAMP1 (Fig. 7B and 7C). This phenotype was IL-10-dependent, since addition of rIL-10 to the IL-10−/− BMDM restored the ability of the pathogen to escape the late endosome (Fig. 7B and 7C). Results similar to those described (Fig. 7B and Fig. 7C) were also observed in IFNγ activated BMDM (data not shown).

Lack of IL-10 results in higher levels of NF-κB activation and pro-inflammatory cytokine production in B. abortus infected macrophages

It has been demonstrated that IL-10 can inhibit a protective immune response, possibly by blocking NF-κB activation [25] and downstream production of pro-inflammatory cytokines by antigen-presenting cells such as macrophages [15], [26]. To investigate if IL-10 production would have an effect on NF-κB activation in Brucella infected macrophages, we used an NF-κB reporter RAW murine macrophage cell line (RAW-Blue cells). RAW-Blue cells were infected with B. abortus 2308 (MOI = 100) in the presence of IL-10 receptor blocking antibody (IL-10R Ab) or rIL-10, and NF-κB activation was measured at 8 and 24 h.p.i. Brucella infected RAW-Blue macrophages produced significant amounts of IL-10 (Fig. S4B). Blockage of IL-10R resulted in decreased B. abortus intracellular survival inside RAW-Blue macrophages when compared to untreated controls at 24 h.p.i., as previously observed in IL-10−/− BMDM (data not shown). B. abortus infection did not result in significant NF-κB activation by any of the treatment groups at 8 h.p.i. (Fig. 8A). The use of IL-10R Ab to block the response of B. abortus infected RAW-Blue macrophages to IL-10 resulted in a significant increase in NF-κB activation at 24 h.p.i. when compared to untreated infected macrophages. Conversely, addition of exogenous rIL-10 resulted in a significant inhibition of NF-κB activation in B. abortus infected cells (Fig. 8A). The results above described were also observed in IFNγ activated RAW blue cells (data not shown).

thumbnail
Figure 8. Lack of endogenous IL-10 results in higher NF-κB activation and production of pro-inflammatory cytokines by macrophages infected with B. abortus.

(A) NF-κB activation measured in RAW-Blue macrophages infected with B. abortus 2308 for 8 h and 24 h in the presence of IL-10 receptor blocking antibody (IL-10R Ab), isotype control (IgG Ab) or exogenous IL-10 (rIL-10). (B) qRT-PCR analysis of IL-6, and TNF-α expression by BMDM from wild type mice (C57BL/6) and IL-10 deficient mice (IL-10−/−) in the presence or absence of recombinant IL-10. (C) ELISA essay for IL-6 and TNF-α production in supernatant from wild type mice (C57BL/6) and IL-10 deficient mice (IL-10−/−) in the presence or absence of recombinant IL-10. Results shown are representative of two independent experiments. n = 5. Values represent mean ± SEM. *P<0.05 using unpaired t-test statistical analysis.

https://doi.org/10.1371/journal.ppat.1003454.g008

To confirm that IL-10 affects pro-inflammatory cytokine production by infected macrophages, bone marrow-derived macrophages (BMDM) from C57BL/6 and IL-10−/− mice were infected with B. abortus 2308 (MOI = 100) and cytokine expression was measured at 24 h.p.i. by ELISA and quantitative real-time PCR. Significantly, an absence of IL-10 resulted in higher expression levels of the pro-inflammatory cytokines IL-6 and TNFα by infected macrophages (Fig. 8B and 8C). Moreover, the phenotype observed was shown to be IL-10 dependent, since the addition of rIL-10 to IL-10−/− infected BMDM restored IL-6 and TNF-α expression to wild-type levels. Results similar to those described in Fig. 8B and Fig. 8C were also observed in IFN-γ activated BMDM (data not shown). Our results demonstrate that IL-10 production during B. abortus infection in vitro affects macrophage function by modulating NF-κB activation and the production of pro-inflammatory cytokines by infected cells.

Inability of macrophages to respond to IL-10 results in severe acute pathology and decreased B. abortus survival in vivo

To further investigate the possibility that macrophages are the cell type responding to the IL-10 produced during B. abortus infection, we used Il-10Rflox/floxLysMCre+/− (IL-10Rflox/LysMCre) mice, which are unable to express the IL-10 receptor 1 chain (IL-10R1) specifically in monocytes/macrophages and/or neutrophils [27]. IL-10Rflox/LysMCre and Il-10Rflox/floxLysMCre−/− control mice were infected intraperitoneally with 5×105 CFU of B. abortus 2308 for 3, 9, 21 and 42 days and bacterial survival in infected organs was evaluated. Our results from in vitro infection suggested that IL-10 affects the ability of B. abortus to survive inside macrophages. Remarkably, IL-10Rflox/LysMCre mice showed lower CFU counts in both spleen (Fig. 9A) and liver (Fig. 9B) at 9, 21 and 42 d.p.i. when compared to littermate control mice. This data provided strong support for the idea that macrophage responsiveness to IL-10 is necessary for optimal initial B. abortus colonization of the host as well as long-term persistence.

thumbnail
Figure 9. Inability of macrophages to respond to IL-10 results in decreased persistence of B. abortus in vivo.

(A,B) B. abortus 2308 CFU counts in spleen (A) and liver (B) from littermate control mice (grey bars) compared with IL10Rflox/LysMCre (white bars) at 3, 9, 21 and 42 d.p.i. (C) Measurement of mouse weight over time in grams of littermate control mice (grey losangle) compared with IL10Rflox/LysMCre (white dots). (D) ELISA essay for IL-6 and TNF-α production in serum from littermate control mice (grey bars) compared with IL10Rflox/LysMCre mice (white bars) at 3, 9, 21 and 42 d.p.i. (E) qRT-PCR analysis of pro-inflammatory cytokines genes (Ifnγ, Il6 and Tnfα) in spleen from littermate control mice (grey bars) compared with IL10Rflox/LysMCre mice (white bars) at 3, 9, 21 and 42 d.p.i. n = 5. Values represent mean ± SEM. (*) represents P<0.05 using unpaired t-test statistical analysis.

https://doi.org/10.1371/journal.ppat.1003454.g009

The results shown above (Fig. 5) demonstrated that a lack of IL-10 production by T cells during B. abortus in vivo infection resulted in increased pro-inflammatory responses and evident clinical signs of disease in mice. Moreover, our in vitro results suggested that blockage of IL-10R played a role in the control of NF-κB activation and pro-inflammatory cytokine production by B. abortus-infected macrophages. Therefore, we sought to determine the effect of macrophage responsiveness to IL-10 in the early host response to B. abortus infection. B. abortus-infected IL-10R/LysMCre mice exhibited decreased weight gain at 9, 15, and 21 d.p.i. when compared to wild-type infected mice (Fig. 9C). Furthermore, levels of IFN-γ, IL-6 and TNF-α were significantly increased in serum (Fig. 9D) spleens (Fig. 9E) and livers (Fig. S5) of IL-10R/LysMCre at 3, 9 d.p.i. and, to a lesser extent, at 21 d.p.i.

To determine if macrophage non-responsiveness to IL-10 would result in detrimental pathologic changes, spleen and liver sections from infected IL-10Rflox/LysMCre and control mice were blindly evaluated by veterinary pathologists (MNX and TMS). As expected, IL-10Rflox/LysMCre showed severe acute lesions characterized by influx of neutrophils and histiocytes, as well as tissue necrosis and multifocal neutrophilic vasculitis and thrombosis in the spleen at 9 d.p.i. (Fig. 10A and 10C) and in the liver (Fig. 10B and 10C) at 9 and 21 d.p.i. However, at 21 and 42 days post-infection, IL-10Rflox/LysMCre mice exhibited decreased granuloma formation in spleen (Fig. 10A and 10C) when compared to littermate control mice, suggesting that macrophage responsiveness to IL-10 is important for development of chronic pathological lesions in spleens of infected animals. These data provide the direct support for the idea that induction of IL-10 during B. abortus in vivo infection plays a key role in modulation of macrophage function, which, in turn, provides the ideal initial immunological environment for bacterial colonization and development of chronic infection.

thumbnail
Figure 10. Inability of macrophages to respond to IL-10 results in severe acute B. abortus induced pathology in vivo.

(A, B) Histopathology score of spleen (A) and liver (B) from littermate mice (Cre−/−) compared with IL10Rflox/LysMCre mice (Cre+/−) at 3, 9, 21 and 42 d.p.i. (C) Representative histopathology figures from (A,B) - Black arrows indicate microgranulomas, white arrowheads show neutrophilic infiltrate and white upper case WP indicates white pulp (×20). n = 5. (*) represents P<0.05 using Mann-Whitney statistical analysis.

https://doi.org/10.1371/journal.ppat.1003454.g010

Discussion

The balance between pro-inflammatory and anti-inflammatory cytokine production appears to be crucial for the ability of the host to eradicate an infection, as well as for the clinical presentation and/or pathology resulting from the infection. This balance appears to shift in the case of persistent pathogens such as Brucella spp., which are able to evade TLR signaling during the early stages of infection, thereby preventing development of an immune response that is appropriate to clear the infection [28]. It is known that during the acute phase of B. abortus infection in mice, neutralization of IL-10 reduces bacterial colonization [5]. Here, we provide support to the idea that during this phase of B. abortus infection, early production of IL-10 by T cells is key to promoting persistent intracellular infection.

In a number of infectious disease models, several cell types, including T cell subsets, B cells, neutrophils, macrophages and some DC subsets have been shown to be able to produce IL-10 [18]. Even though B. abortus infected macrophages are capable of producing IL-10 in vitro, only B cells have been implicated as a potential source of IL-10 during in vivo infection [29]. In this study, however, we have demonstrated that macrophages play a limited role in IL10 production during early acute B. abortus infection. Additionally, we identified T cells, more specifically CD4+CD25+ T cells, as the major source of this cytokine during acute brucellosis. Indeed, Svetić and collaborators [1] have suggested a possible role for CD4+ T cells in IL-10 production during acute murine Brucellosis. Interestingly, previous studies have demonstrated elevated numbers of CD4+CD25+ T cells in PBMCs from human patients with acute brucellosis [30] as well as in draining lymph nodes from B. melitensis infected sheep [28]. Moreover, Pasquali and collaborators demonstrated that depletion of CD4+CD25+ T cells resulted in increased control of B. abortus infection due to elevated activation of effector T cells and higher production of pro-inflammatory cytokines such as IFN-γ by infected mice [31]. Most likely, the effects seen in this latter study were an indirect effect of the decreased IL-10 production by T cells, resulting in elevated macrophage activation in CD25-depleted mice upon B. abortus infection. Taken together, these results point to CD4+CD25+ T cells as important players in modulating the early immune response to B. abortus in vivo.

There is a general agreement that macrophages represent a critical niche for Brucella persistence in the host [4]. Importantly, they have been described as one of the cell types responding to IL-10 production in other infection models [11], [27]. Here we have demonstrated that macrophages are a main cell type responding to the immunomodulatory functions of IL-10 during both in vitro and in vivo Brucella infection. Moreover, IL-10 signaling had a significant impact on the ability of B. abortus-infected macrophages to produce pro-inflammatory cytokines and to permit intracellular growth of B. abortus. Interestingly, O'Leary and collaborators have demonstrated that IL-10 production by immune cells can affect the ability of M. tuberculosis to escape the LAMP1+ late endosomal compartment and to establish infection in human macrophages in vitro [32]. In agreement with this previous study, we have demonstrated that the capacity of macrophages to respond to IL-10 impacts intracellular survival of Brucella by decreasing the pathogen's ability to escape the LAMP1+ late endosome, a prerequisite for replication in an endoplasmic reticulum-associated compartment.

It is important to note that the LysM promoter used to drive Cre expression in IL10Rflox mice is expressed in both neutrophils and macrophages [27]. Although our understanding of the role of neutrophils during brucellosis is still evolving [8], [33], we and others have described recruitment of these cells to both spleens and livers of B. abortus-infected mice during the acute infection phase ([33], Fig. 6 and Fig. 9). Moreover, the neutrophil recruitment was more evident in the absence of IL-10 (Fig. 2, Fig. 6 and Fig. 9). Therefore, although Brucella resistance to neutrophil killing has been well described [34], it is possible that the increased cytokine expression and pathology observed in IL10Rflox/LysMCre mice could also be due in part to a failure of neutrophils recruited to the site of infection to respond to IL-10.

It should be pointed out that, although IL-10 contributes to persistence of B. abortus in vivo, abrogation of IL-10 production (Fig. 2) or neutralization of IL-10 in vivo [5] did not result in eradication of B. abortus from tissues, contrary to what has been shown for Leishmania [13]. Therefore, factors in addition to IL-10 production must also contribute to chronic persistence of B. abortus. Although TGF-β has been shown to be produced by B cells and macrophages in BALB/c mice [29], consistent with this report, we did not observe any increase in circulating TGF-β1, TGF-β2, or TGF-β3 at 21 or 42 days post infection of C57BL/6 mice (data not shown). However, these results do not rule out a role for local activation of TGF-β in promoting chronic infection. An important factor in persistence of Brucella is the transient nature of IFNγ production in infected mice, which subsides by 21 d post infection in mice [35], therefore our observed lack of a role of IL-10 later in infection could suggest that its role is to antagonize the activity of IFNγ at earlier stages of infection. Finally, the possibility should be considered that during chronic infection, B. abortus may reside in a cell type that has inherently low microbicidal activity, as has been found for M. tuberculosis [36] and B. melitensis [37].

At first glance, our data suggest that inhibition of IL-10 signaling would be beneficial to the host, since IL-10−/−, IL-10flox/CD4Cre and IL-10Rflox/LysMcre showed increased ability to control B. abortus infection at both acute and chronic stages of infection. Moreover, both IL-10flox/CD4Cre and IL-10Rflox/LysMcre exhibited reduced formation of granulomas, a potential niche for B. abortus persistence [4], during the chronic stage of infection. However, in spite of the increased bacterial clearance, we demonstrated that lack of IL-10 during Brucella infection could potentially be detrimental to the host, since B. abortus infected IL-10 deficient mice presented evident signs of acute disease, characterized by changes in weight gain and marked histopathological lesions in both spleen and liver. Indeed, studies on other chronic pathogens such as Leishmania major [38], human cytomegalovirus [14], and M. tuberculosis (reviewed in [15]) have demonstrated that even though absence of IL-10 leads to better clearance of these pathogens, it can also result in severe and sometimes lethal pathologic changes. Therefore, although modulation of the IL-10 signaling pathway could be a potential target to avoid the establishment of chronic infection, more studies are needed to elucidate the optimal activation of the immune system necessary to improve clearance of chronic pathogens without a great cost to the host.

Materials and Methods

Bacterial strains, media and culture conditions

Bacterial strains used in this study were the virulent strain Brucella abortus 2308 and its isogenic mutant strain MX2 which has an insertion of pKSoriT-bla-kan-PsojA-mCherry plasmid [37]. For strain MX2, positive clones were kanamycin resistant and fluorescent, as previously described [37]. Strains were cultured on tryptic soy agar (Difco/Becton-Dickinson, Sparks, MD) or tryptic soy broth at 37°C on a rotary shaker. Bacterial inocula for mouse infection were cultured on tryptic soy agar plus 5% blood for 3 days [39]. For cultures of strain MX2, kanamycin (Km) was added to the culture medium at 100 µg/mL. All work with B. abortus cells was performed at biosafety level 3.

Bone marrow derived macrophage infection

Bone marrow-derived macrophages were differentiated from bone marrow precursors from femora and tibiae of female, 6 to 8 weeks old, C57BL/6J and IL-10−/− mice obtained from The Jackson Laboratory (Bar Harbor) following a previously published procedure [40]. For BMDM experiments, 24-well microtiter plates were seeded with macrophages at concentration of 5×105 cells/well in 0.5 mL of RPMI media (Invitrogen, Grand Island, NY) supplemented with 10% FBS and 10 mM L-glutamine (RPMI supl) incubated for 48 h at 37°C in 5% CO2. Preparation of the inoculum and BMDM infection was performed as previously described [40]. Briefly, for inoculum preparation, B. abortus 2308 was grown for 24 h and then diluted in RPMI supl, and about 5×107 bacteria in 0.5 mL of RPMI supl were added to each well of BMDM, reaching multiplicity of infection (MOI) of 100. Microtiter plates were centrifuged at 210× g for 5 min at room temperature in order to synchronize infection. Cells were incubated for 20 min at 37°C in 5% CO2, free bacteria were removed by three washes with phosphate-buffered saline (PBS), and the zero-time-point sample was taken as described below. After the PBS wash, RMPI supl plus 50 mg gentamicin per mL was added to the wells, and the cells were incubated at 37°C in 5% CO2. For cytokine production assays, supernatants from each well were sampled at 0, 8, 24, or 48 h after infection, depending on the experiment performed. In order to determine bacterial survival, the medium was aspirated at the time points described above, and the BMDM were lysed with 0.5 mL of 0.5% Tween 20, followed by rinsing of each well with 0.5 mL of PBS. Viable bacteria were quantified by serial dilution in sterile PBS and plating on TSA. For gene expression assays, BMDM were resuspended in 0.5 mL of TRI-reagent (Molecular Research Center, Cincinnati) at the time-points described above and kept at −80°C until further use. When necessary, 1 ng/mL of mouse rIL-10 (eBioscience, San Diego, CA) or 1 ng/mL of mouse rIFN-γ (BD Bioscience, San Jose, CA) was added to the wells and kept throughout the experiments. All experiments were performed independently in triplicate at least three times and the standard error for each time point calculated.

RAW-Blue macrophages experiments

RAW-Blue cells (Invivogen, San Diego, CA) were derived from RAW-264.7 macrophages with chromosomal integration of a SEAP reporter construct inducible by NF-κB and AP-1. RAW-Blue cells were maintained in Zeocin (Invivogen, San Diego, CA) selective medium. For RAW-Blue experiments, 24-well microtiter plates were seeded with macrophages at concentration of 2×105 cells/well in 0.5 mL of DMEM media (Invitrogen, Grand Island, NY) supplemented with 10% FBS and 10 mM L-glutamine (DMEM supl). Preparation of the inoculum and RAW-Blue infection was performed as previously described [40], using MOI = 100. For NF-κB activation assays, supernatant from each well was sampled at 8 h and 24 h after infection and secretion of the substrate SEAP was detected and measured in a spectrophotometer at 650 nm with QUANTI-Blue (Invivogen, San Diego, CA) according to manufacturer's instructions. When necessary, 1 ng/mL of mouse rIL-10 (eBioscience, San Diego, CA),1 ng/mL of mouse rIFN-γ (BD Bioscience, San Jose, CA),1 µg/mL of anti-mouse IL-10R antibody (R&D Systems, Minneapolis, MN) or anti-mouse IgG isotype antibody control (R&D Systems, Minneapolis, MN) were added to the wells and kept throughout the experiments. All experiments were performed independently in triplicate at least three times and the standard error for each time point calculated.

Ethics statement

Experiments with mice were carried out in strict accordance with the recommendations in the Guide for Care and Use of Laboratory Animals of the National Institute of Health and were approved by the Institutional Animal Care and Use Committees at the University of California at Davis (protocol number: 16468).

Animal experiments

Female C57BL/6J wild-type mice, B6.129P2-Il10tm1Cgn/J; (IL-10−/−) mice [29] and Il-10 GFP reporter mice [19], aged 6–8 weeks, were obtained from The Jackson Laboratory (Bar Harbor). Female and male Il10flox/floxCd4cre+/− (IL-10flox/CD4Cre), and Il10Rflox/floxLysmcre+/− (IL-10Rflox/LysMCre) aged 6–8 weeks, were reported previously [27], [31]. Female and male IL10flox/floxLysMCre+/− (IL-10flox/LysMCre) were generated at UC Davis. For the strains IL-10 flox/CD4Cre, IL-10Rflox/LysMCre and IL-10flox/LysMCre, littermate Il10flox/floxCd4cre−/−, Il10Rflox/floxLysmcre−/−, and IL10flox/floxLysMCre−/− mice were used as control, respectively. Mice were held in microisolator cages with sterile bedding and irradiated feed in a biosafety level 3 laboratory. Groups of 3 to 5 mice were inoculated intraperitoneally (i.p.) with 0.2 mL of phosphate-buffered saline (PBS) containing 5×105 CFU of B. abortus 2308 as previously described [41]. At 3, 9, 15, 21 and/or 42 days after infection, depending on the experiment performed, the mice were euthanized by CO2 asphyxiation and their serum, livers and spleens were collected aseptically at necropsy. The livers and spleens were homogenized in 2 mL of PBS, and serial dilutions of the homogenate were plated on TSA for enumeration of CFU. Samples of liver and spleen tissue were also collected for gene expression and histopathology analysis as described below.

ELISA

The presence of IL-10, IL-6 and TNF-α in BMDM supernatant and in serum samples from C57BL/6, IL-10flox/CD4Cre, IL-10flox/LysMCre and littermate control mice infected with B. abortus 2308 was determined by indirect enzyme-linked immunosorbent assay (ELISA) (eBioscience, San Diego, CA) according to the manufacturer's instructions. The ELISA test was read at 450 nm with an ELISA microplate reader (MR5000; Dynatech). The sensitivity of the ELISA used was 7.8 pg/mL. Data points are the averages of duplicate dilutions, with each measurement being performed twice.

RT-PCR and real time PCR analysis

Eukaryotic gene expression was determined by real-time PCR as previously described [40]. Briefly, eukaryotic RNA was isolated using TRI reagent (Molecular Research Center, Cincinnati) according to the manufacturer's instructions. A Reverse transcriptase reaction was performed to prepare complementary DNA (cDNA) using TaqMan reverse transcription reagents (Applied Biosystems, Carlsbad). A volume of 4 µL of cDNA was used as template for each real-time PCR reaction in a total reaction volume of 25 µL. Real-time PCR was performed using SYBR-Green (Applied Biosystems) along with the primers listed in Table S1 in Text S1. Data were analyzed using the comparative Ct method (Applied Biosystems, Carlsbad). Transcript levels of Il10, Il6, Ifng and Tnfa were normalized to mRNA levels of the housekeeping gene βactin.

Histopathology

Formalin fixed spleen and liver tissue sections were stained with hematoxylin and eosin, and two veterinary pathologists (MX and TS) performed a blinded evaluation using criteria described in Table S2 in Text S1. Representative images were obtained using an Olympus BX41 microscope and the brightness adjusted (Adobe Photoshop CS2).

Flow cytometry

Flow cytometric analysis of IL-10 producing cells was performed in splenocytes from IL-10 GFP reporter mice infected for 3 and 9 days with B. abortus 2308. Briefly, after passing the spleen cells through a 100-µm cell strainer and treating the samples with ACK buffer (0.15 M NH4Cl, 1.0 mM KHCO3, 0.1 mM Na2EDTA [pH 7.2]) to lyse red blood cells, splenocytes were washed with PBS (Gibco) containing 1% bovine serum albumin (fluorescence-activated cell sorter [FACS] buffer). After cell counting, 4×106 cells/mouse were re-suspended in PBS and stained with Aqua Live/Dead cell discriminator (Invitrogen, Grand Island, NY) according to the manufacturer's protocol. After Live/Dead staining, splenocytes were resuspended in 50 µL of FACS buffer and cells were stained with a cocktail of anti-B220 Brilliant Violet 421 (Biolegend, San Diego, CA), anti-CD3 PE (BD Pharmingen, San Jose, CA), anti-CD11b APC.Cy7 (Biolegend, San Diego, CA), anti-F4/80 Pe.Cy7 (Biolegend, San Diego, CA), anti-Cd11c APC (Biolegend, San Diego, CA). To determine the T cell subset responsible for IL-10 production, cells were stained with a cocktail of anti-CD3 APC.Cy7 (eBioscience, San Diego, CA), anti-CD8 AF700 (BD Pharmigen, San Jose, CA), anti-TCRγδ PE (BD Pharmigen, San Jose, CA), anti-CD4 eFluor 450 (eBioscience, San Diego, CA), anti-CD25 Pe.Cy7 (eBioscience, San Diego, CA). The cells were washed with FACS buffer and fixed with 4% formaldehyde for 30 min at 4°C, and resuspended in FACS buffer prior to analysis. Flow cytometry analysis was performed using an LSRII apparatus (Becton Dickinson, San Diego, CA), and data were collected for 5×105 cells/mouse. Resulting data were analyzed using Flowjo software (Treestar, inc. Ashland, OR). Gates were based on Fluorescence-Minus-One (FMO) controls.

Immunofluorescence microscopy

Immunofluorescence of Brucella infected BMDM was performed as previously described [24]. Briefly, B. abortus MX2 infected BMDM were grown on 12-mm glass coverslips in 24-well plates were washed three times with PBS, fixed with 3% paraformaldehyde, pH 7.4, at 37°C for 20 min, washed three times with PBS and then incubated for 10 min in 50 mm NH4Cl in PBS in order to quench free aldehyde groups. Samples were blocked and permeabilized in 10% goat serum and 0.1% saponin in PBS for 30 min at room temperature. Cells were labeled by inverting coverslips onto drops of primary antibodies diluted in 10% horse serum and 0.1% saponin in PBS and incubating for 45 min at room temperature. The primary antibody used was rat anti-mouse LAMP-1 (BD Pharmigen, San Jose, CA). Bound antibodies were detected by incubation with 1∶500 dilution of Alexa Fluor 488 donkey anti-rat (Invitrogen, Grand Island, NY) for 45 min at room temperature. Cells were washed twice with 0.1% saponin in PBS, once in PBS, once in H2O and then mounted in Mowiol 4-88 mounting medium (Calbiochem). Samples were observed on a Carl Zeiss LSM 510 confocal laser scanning microscope for image acquisition (Carl Zeiss Micro Imaging). Confocal images of 1024×1024 pixels were acquired as projections of three consecutive slices with a 0.38-µm step and assembled using Adobe Photoshop CS2 (Adobe Systems). For quantification of Brucella MX2 and Lamp1+ compartment colocalization, at least 100 bacteria/sample were counted. All experiments were performed independently in quadruplicate at least two times.

Statistical analysis

Fold changes of ratios (bacterial numbers or mRNA levels) and percentages (flow cytometry and fluorescent microscopy) were transformed logarithmically prior to statistical analysis. An unpaired Student's t-test was performed on the transformed data to determine whether differences in fold changes between groups were statistically significant (P<0.05). Significance of differences in histopathology scores was determined by a one-tailed non-parametric test (Mann-Whitney).

Supporting Information

Figure S1.

Expansion of CD4+CD25+ T cells during Brucella infection. (A) Flow cytometry quantification of CD4+CD25+ splenic T cells from C57BL/6 IL-10 GFP-reporter mice infected with B. abortus 2308 for 9 days. (B) Representative data plot of CD4 and CD25 expression in splenic T cells from C57BL/6 IL-10 GFP-reporter mice infected with B. abortus 2308 for 9 days. Values represent mean ± SEM. *P<0.05. n = 4. Values represent mean ± SEM. (*) represents P<0.05 relative to uninfected control using unpaired t-test statistical analysis. FMO = fluorescence minus one.

https://doi.org/10.1371/journal.ppat.1003454.s001

(TIF)

Figure S2.

(A, B) Histopathology score of spleen (A) and liver (B) from littermate mice (Cre−/−) compared with IL10flox/LysMCre mice (Cre+/−) at 3, 9 and 21 d.p.i. (C) Representative histopathology figures from (A,B) - Black arrows indicate microgranulomas, white arrowheads show neutrophilic infiltrate, and white upper case WP indicates white pulp (×20). n = 5. (*) represents P<0.05 using Mann-Whitney statistical analysis.

https://doi.org/10.1371/journal.ppat.1003454.s002

(TIF)

Figure S3.

(A) ELISA essay for IL-10 production in serum from littermate control mice (grey bars) compared with IL10flox/CD4Cre mice (white bars) at 0, 3, 9, 21 and 42 d.p.i. (B) qRT-PCR analysis of IL-10 expression in spleen from littermate control (grey bars) compared with IL10flox/CD4Cre mice (white bars) at 3, 9, 21 and 42 d.p.i. (C) qRT-PCR analysis of pro-inflammatory cytokines genes (Ifnγ, Il6 and Tnfα) in liver from littermate control (grey bars) compared with IL10flox/CD4Cre mice (white bars) at 3, 9, 21 and 42 d.p.i. n = 5. Values represent mean ± SEM. (*) represents P<0.05 relative to uninfected control using unpaired t-test statistical analysis.

https://doi.org/10.1371/journal.ppat.1003454.s003

(TIF)

Figure S4.

(A) ELISA essay for IL-10 production in supernatant from C57BL/6 wild type BMDM infected with B. abortus 2308 for 24 h. (B) ELISA essay for IL-10 production in supernatant from RAW-Blue macrophages infected with B. abortus 2308 for 8 h and 24 h in the presence of IL-10 receptor blocking antibody (IL-10R Ab), isotype control (IgG Ab) or exogenous IL-10 (rIL-10).

https://doi.org/10.1371/journal.ppat.1003454.s004

(TIF)

Figure S5.

qRT-PCR analysis of pro-inflammatory cytokines genes (Ifnγ, Il6 and Tnfα) in liver from littermate control (grey bars) compared with IL10Rflox/LysMCre mice (white bars) at 3, 9, 21 and 42 d.p.i. n = 5. Values represent mean ± SEM. (*) represents P<0.05 relative to uninfected control using unpaired t-test statistical analysis.

https://doi.org/10.1371/journal.ppat.1003454.s005

(TIF)

Text S1.

Supplementary Methods for real-time RT-PCR and histopathology scoring. Table S1: Real-time RT-PCR primers used in this study. Table S2: Histopathology scoring used in this study.

https://doi.org/10.1371/journal.ppat.1003454.s006

(DOCX)

Acknowledgments

The authors thank Xavier DeBolle for providing pKSoriT-bla-kan-PsojA-mCherry, and Tish Ashley for assistance with mouse breeding.

Author Contributions

Conceived and designed the experiments: MNX RMT RLS. Performed the experiments: MNX MGW KN AMS VLA TMAS. Analyzed the data: MNX AMS TMAS RLS RMT. Contributed reagents/materials/analysis tools: WM. Wrote the paper: MNX MGW TMAS AJB RLS RMT.

References

  1. 1. Svetic A, Jian YC, Lu P, Finkeiman FD, Gause WC (1993) Brucella abortus induces a novel cytokine gene expression pattern characterized by elevated IL-10 and IFN-g in CD4+ T cells. International Immunology 5: 877–883.
  2. 2. Pappas G, Papadimitriou P, Akritidis N, Christou L, Tsianos EV (2006) The new global map of human brucellosis. Lancet Infect Dis 6: 91–99.
  3. 3. Corbel MJ (1997) Brucellosis: an overview. Emerg Infect Dis 3: 213–221.
  4. 4. Atluri VL, Xavier MN, de Jong MF, den Hartigh AB, Tsolis RM (2011) Interactions of the Human Pathogenic Brucella Species with Their Hosts. Ann Rev Microbiol 65: 523–541.
  5. 5. Fernandes DM, Baldwin CL (1995) Interleukin-10 downregulates protective immunity to Brucella abortus. Infection and Immunity 63: 1130–1133.
  6. 6. Fernandes DM, Jiang X, Jung JH, Baldwin CL (1996) Comparison of T cell cytokines in resistant and susceptible mice infected with virulent Brucella abortus strain 2308. FEMS Immunology & Medical Microbiology 16: 193–203.
  7. 7. Murphy EA, Sathiyaseelan J, Parent MA, Zou B, Baldwin CL (2001) Interferon-γ is crucial for surviving a Brucella abortus infection in both resistant C57BL/6 and susceptible BALB/c mice. Immunology 103: 511–518.
  8. 8. Barquero-Calvo E, Chaves-Olarte E, Weiss DS, Guzman-Verri C, Chacon-Diaz C, et al. (2007) Brucella abortus uses a stealthy strategy to avoid activation of the innate immune system during the onset of infection. PLoS ONE 2: e631.
  9. 9. Martirosyan A, Moreno E, Gorvel J-P (2011) An evolutionary strategy for a stealthy intracellular Brucella pathogen. Immunological Reviews 240: 211–234.
  10. 10. Andersen-Nissen E, Smith KD, Strobe KL, Barrett SL, Cookson BT, et al. (2005) Evasion of Toll-like receptor 5 by flagellated bacteria. Proc Natl Acad Sci U S A 102: 9247–9252.
  11. 11. Saraiva M, O'Garra A (2010) The regulation of IL-10 production by immune cells. Nat Rev Immunol 10: 170–181.
  12. 12. Sabat R, Grütz G, Warszawska K, Kirsch S, Witte E, et al. (2010) Biology of interleukin-10. Cytokine & Growth Factor Reviews 21: 331–344.
  13. 13. Belkaid Y, Hoffmann KF, Mendez S, Kamhawi S, Udey MC, et al. (2001) The role of interleukin (IL)-10 in the persistence of Leishmania major in the skin after healing and the therapeutic potential of anti-IL-10 receptor antibody for sterile cure. J Exp Med 194: 1497–1506.
  14. 14. Chang WLW, Barry PA (2010) Attenuation of innate immunity by cytomegalovirus IL-10 establishes a long-term deficit of adaptive antiviral immunity. Proceedings of the National Academy of Sciences 107: 22647–22652.
  15. 15. Redford PS, Murray PJ, O'Garra A (2011) The role of IL-10 in immune regulation during M. tuberculosis infection. Mucosal Immunol 4: 261–270.
  16. 16. Fernández-Lago L, Monte M, Chordi A (1996) Endogenous gamma interferon and interleukin-10 in Brucella abortus 2308 infection in mice. FEMS Immunology & Medical Microbiology 15: 109–114.
  17. 17. Budak F, Göral G, Heper Y, Yilmaz E, Aymak F, et al. (2007) IL-10 and IL-6 gene polymorphisms as potential host susceptibility factors in Brucellosis. Cytokine 38: 32–36.
  18. 18. Moore KW, de Waal Malefyt R, Coffman RL, O'Garra A (2001) Interleukin-10 and the interleukin-10 receptor. Annual Review of Immunology 19: 683–765.
  19. 19. Kamanaka M, Kim ST, Wan YY, Sutterwala FS, Lara-Tejero M, et al. (2006) Expression of Interleukin-10 in Intestinal Lymphocytes Detected by an Interleukin-10 Reporter Knockin tiger Mouse. Immunity 25: 941–952.
  20. 20. Shevach EM (2002) CD4+CD25+ suppressor T cells: more questions than answers. Nat Rev Immunol 2: 389–400.
  21. 21. Roers A, Siewe L, Strittmatter E, Deckert M, Schlüter D, et al. (2004) T Cell-specific Inactivation of the Interleukin 10 Gene in Mice Results in Enhanced T Cell Responses but Normal Innate Responses to Lipopolysaccharide or Skin Irritation. The Journal of Experimental Medicine 200: 1289–1297.
  22. 22. Hunt AC, Bothwell PW (1967) Histological findings in human brucellosis. J Clin Pathol 20: 267–272.
  23. 23. Celli J (2006) Surviving inside a macrophage: the many ways of Brucella. Res Microbiol 157: 93–98.
  24. 24. Starr T, Ng TW, Wehrly TD, Knodler LA, Celli J (2008) Brucella intracellular replication requires trafficking through the late endosomal/lysosomal compartment. Traffic 9: 678–694.
  25. 25. Wang P, Wu P, Siegel MI, Egan RW, Billah MM (1995) Interleukin (IL)-10 Inhibits Nuclear Factor B (NFB) Activation in Human Monocytes. Journal of Biological Chemistry 270: 9558–9563.
  26. 26. Fiorentino DF, Zlotnik A, Mosmann TR, Howard M, O'Garra A (1991) IL-10 inhibits cytokine production by activated macrophages. The Journal of Immunology 147: 3815–3822.
  27. 27. Pils MC, Pisano F, Fasnacht N, Heinrich J-M, Groebe L, et al. (2010) Monocytes/macrophages and/or neutrophils are the target of IL-10 in the LPS endotoxemia model. European Journal of Immunology 40: 443–448.
  28. 28. Suraud V, Olivier M, Bodier CC, Guilloteau LA (2007) Differential expression of homing receptors and vascular addressins in tonsils and draining lymph nodes: Effect of Brucella infection in sheep. Veterinary Immunology and Immunopathology 115: 239–250.
  29. 29. Goenka R, Parent MA, Elzer PH, Baldwin CL (2011) B Cell-deficient Mice Display Markedly Enhanced Resistance to the Intracellular Bacterium Brucella abortus. J Infect Dis 203: 1136–1146.
  30. 30. Skendros P, Boura P, Chrisagis D, Raptopoulou-Gigi M (2007) Diminished percentage of CD4+ T-lymphocytes expressing interleukine-2 receptor alpha in chronic brucellosis. Journal of Infection 54: 192–197.
  31. 31. Pasquali P, Thornton AM, Vendetti S, Pistoia C, Petrucci P, et al. (2010) CD4+CD25+ T regulatory cells limit effector T cells and favor the progression of brucellosis in BALB/c mice. Microbes and Infection 12: 3–10.
  32. 32. O'Leary S, O'Sullivan MP, Keane J (2011) IL-10 Blocks Phagosome Maturation in Mycobacterium tuberculosis-infected Human Macrophages. American Journal of Respiratory Cell and Molecular Biology 45: 172–180.
  33. 33. Barquero-Calvo E, Martirosyan A, Ordoñez-Rueda D, Arce-Gorvel V, Alfaro-Alarcón A, et al. (2013) Neutrophils Exert a Suppressive Effect on Th1 Responses to Intracellular Pathogen Brucella abortus. PLoS Pathog 9: e1003167.
  34. 34. Kreutzer DL, Dreyfus LA, Robertson DC (1979) Interaction of polymorphonuclear leukocytes with smooth and rough strains of Brucella abortus. Infect Immun 23: 737–742.
  35. 35. Copin R, De Baetselier P, Carlier Y, Letesson JJ, Muraille E (2007) MyD88-Dependent Activation of B220-CD11b+LY-6C+ Dendritic Cells during Brucella melitensis Infection. J Immunol 178: 5182–5191.
  36. 36. Peyron P, Vaubourgeix J, Poquet Y, Levillain F, Botanch C, et al. (2008) Foamy macrophages from tuberculous patients' granulomas constitute a nutrient-rich reservoir for M. tuberculosis persistence. PLoS Pathog 4: e1000204.
  37. 37. Copin R, Vitry M-A, Hanot Mambres D, Machelart A, De Trez C, et al. (2012) In Situ Microscopy Analysis Reveals Local Innate Immune Response Developed around Brucella Infected Cells in Resistant and Susceptible Mice. PLoS Pathog 8: e1002575.
  38. 38. Belkaid Y, Hoffmann KF, Mendez S, Kamhawi S, Udey MC, et al. (2001) The Role of Interleukin (IL)-10 in the Persistence of Leishmania major in the Skin after Healing and the Therapeutic Potential of Anti‚ÄìIL-10 Receptor Antibody for Sterile Cure. The Journal of Experimental Medicine 194: 1497–1506.
  39. 39. Alton GG, Jones LM, Pietz DE (1975) Laboratory Techniques in Brucellosis. 2nd edition. Geneva: World Health Organization.
  40. 40. Rolan HG, Tsolis RM (2007) Mice lacking components of adaptive immunity show increased Brucella abortus virB mutant colonization. Infect Immun 75: 2965–2973.
  41. 41. Rolán HG, Tsolis RM (2008) Inactivation of the Type IV Secretion System Reduces the Th1 Polarization of the Immune Response to Brucella abortus Infection. Infection and Immunity 76: 3207–3213.