INTRODUCTION
Tuberculosis (TB) remains a leading cause of death by an infectious agent. Indeed, TB, caused by
Nonribosomal peptide synthases (NRPSs) are large multifunctional proteins involved in the biosynthesis of short peptides in a manner that is distinct from the ribosome-mediated translation of mRNA (5). The genes encoding NRPSs are predominantly found in bacteria and fungi that produce secondary metabolites (6). The short peptides produced by NRPSs usually form part of a larger more complex molecule. The genomes of mycobacteria, including the TB-causing
Importantly, several in vitro and in vivo infection studies have highlighted a potential role for nrp in the pathogenesis of TB. Specifically, nrp was reported as a nonessential gene for in vitro growth by transposon mutagenesis (15) but was otherwise required for growth in the spleens of C57BL/6J mice (16). Additionally, the gene cluster spanning Rv0096-Rv0101 was shown to be critical for survival in mice (17). Furthermore, Nrp was reported to be the most abundant
In this study, we sought to query the role of the nrp gene in the pathogenesis of TB in the mouse model of infection. Our strategy consisted of the generation of
RESULTS
nrp cluster in mycobacteria.
The
FIG 1
The nrp cluster is highly conserved among the MTBC and slow-growing species of the genus Mycobacterium. (A) Sequence identity scores of the nrp cluster were obtained by querying the
Loss of the nrp gene results in attenuation of
To investigate the role of nrp in the virulence of slow-growing mycobacteria, we first generated a null mutant of
To assess the role of nrp in the pathogenesis of
FIG 2
Loss of the nrp gene results in attenuation of
To further explore the growth kinetics of the Δnrp strain in the early stages of infection, we repeated the infection of C57BL/6 mice to monitor CFU levels in lungs from day 14 to up to 90 days postinfection. The difference in CFU between Δnrp-strain- and nrp-comp-strain-infected mice was readily visible on day 14 postinfection, corresponding to a 48-fold lower bacterial burden (1.683 log10 average difference) in the case of mice infected with the Δnrp strain on day 20. From day 30 postinfection and up to day 90, the Δnrp strain reached lung bacillary counts comparable to those of the nrp-comp strain (Fig. 2D), as observed before (Fig. 2A).
Δnrp-strain-infected mice develop less lung pathology.
In addition to the differences observed in the progression of bacterial burdens, a distinct phenotype was also observed in lung histopathological features during infection with the Δnrp strain versus the nrp-comp strain (Fig. 2E and F). Specifically, by day 20 postinfection, no discernible lesions were observed in the lungs of Δnrp-strain-infected animals, whereas multiple small inflammatory infiltrates were already present in the lungs of animals infected with the nrp-comp strain (Fig. 2E and F). This was likely due to the differences in bacterial burdens observed at this time point (Fig. 2D). By day 30 postinfection, the lesions in the lungs of mice infected with the nrp-comp strain were larger than those in mice infected with the Δnrp strain (Fig. 2E and F), even though both presented the same bacterial burden (Fig. 2D). Remarkably, the histological differences were maintained throughout the chronic stage of infection, as on day 90 postinfection, the animals infected with the nrp-comp strain still presented a greater degree of pathology than those infected with the Δnrp strain (Fig. 2E and F), despite the fact that the bacterial burdens had been similar in both cases for 60 days (Fig. 2D). This subdued pathology may explain the longer survival of mice infected with the Δnrp strain (Fig. 2C).
Kinetics of the immune response is altered in Δnrp-strain-infected mice.
So far, our data indicated that in the absence of nrp, mice had a better ability to control the infection at initial time points and showed a striking lesser pathology even at late time points of infection. These findings suggest that a differential immune response may be occurring in the absence of Nrp function. To test this hypothesis, we next investigated the dynamics of immune cell populations elicited during infection with Δnrp or nrp-comp strains. We focused on myeloid (alveolar macrophages, inflammatory monocytes, and neutrophils) (Fig. 3A to C) and lymphoid (CD4+ T cells, CD8+ T cells, and B cells) (Fig. 3D to F) cells known to be relevant to the pathogenesis of TB. Whereas no substantial differences were found throughout the infection with either strain in alveolar macrophages (Fig. 3A), neutrophils (Fig. 3C), CD8+ T cells (Fig. 3E), and B cells (Fig. 3F), significantly fewer inflammatory monocytes (Fig. 3B) and CD4+ T cells (Fig. 3D) were detected at day 20 postinfection in the lungs of mice infected with the Δnrp strain. These differences were attenuated by day 30 postinfection and were no longer present by day 90 postinfection (Fig. 3B and D). In addition to the decreased numbers of inflammatory monocytes and CD4+ T cells observed 20 days postinfection in the absence of nrp expression, the number of activated cells within these populations, as measured by the expression of surface markers major histocompatibility complex (MHC) class II and CD44, was also decreased (Fig. 3G and H). The gating strategy using in the definition of these populations is presented in Fig. S2.
FIG 3
Lack of nrp alters the dynamics of the lung immune response to infection. (A to H) At the indicated time points postinfection, the lungs of C57BL/6 mice infected via aerosol exposure to Δnrp or nrp-comp strains were harvested, a cellular suspension was prepared, and the indicated immune cell populations were determined by flow cytometry. The gating strategy is shown in Fig. S2 in the supplemental material. (I) On day 20 postinfection, the expression of CCL2, CCL7, IFN-γ, and TNF was determined by real-time PCR, as described in Materials and Methods. Data are shown as the means ± SEMs from 5 independent animals in at least 2 independent experiments. The initial bacterial burdens were log10 1.86 ± log10 0.04460 and log10 1.931 ± log10 0.0822 for Δnrp and nrp-comp strains, respectively. Statistical analysis was performed with a two-way ANOVA using Sidak’s test for multiple comparisons. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001.
At the molecular level, the diminished recruitment of inflammatory monocytes seen in the Δnrp strain may be explained by a lower expression of CCL2 and CCL7, chemokines that play a role in the recruitment of myeloid cells to the site of infection (21), in the lungs of these animals (Fig. 3I). In line with a lower frequency of activated CD4+ T cells in Δnrp-strain-infected animals, a decreased expression of gamma interferon (IFN-γ) was observed in the lungs of these animals compared to that in nrp-comp-strain-infected mice (Fig. 3I). Tumor necrosis factor (TNF), another inflammatory mediator during TB, is also expressed at a lower level upon infection with the Δnrp strain (Fig. 3I).
In all, the absence of nrp function in
Δnrp mutant is attenuated for growth in immune-deficient mice.
Our findings point to a role for nrp during the early stages of
FIG 4
Growth of Δnrp mutant is attenuated in immune-deficient mice. C57BL/6 RAG2−/− (A, C, and E) and IFN-γ−/− (B, D, and F) mice were infected by aerosol exposure to a low dose of the Δnrp or nrp-comp strain. (A and B) The weights of the animals were monitored weekly up to day 30 and every 2 days thereafter to determine the survival curves. At the indicated time points (C) or at the time of death (D), the lungs of infected mice were collected and the bacterial burden was determined by CFU enumeration. The lung pathology was determined on day 40 postinfection for RAG2−/− (E) or at the time of death for IFN-γ−/− (F) animals. Shown are pictures from H&E staining for one animal representative of the experimental group; ×40 magnification. The initial bacterial burdens were log10 1.92 ± log10 0.069 and log10 1.701 ± log10 0.0707 (RAG2−/− experiment) and log10 1.92 ± log10 0.069 and log10 1.748 ± log10 0.0707 (IFN-γ−/− experiment) for Δnrp and nrp-comp strains, respectively. BMDM (G and H) or pMac (G) were infected with Δnrp or nrp-comp strains at an MOI of 1 in the absence (G) or presence (H) of exogenous IFN-γ. On day 4 postinfection, the intracellular bacterial load was determined by CFU enumeration. Data are shown as the means ± SEMs from 8 independent animals (A to F) or from 6 replicate wells in 2 independent experiments (G and H). Statistical analysis was performed with log-rank (Mantel-Cox) tests for the Kaplan-Meier curves (A and B) or two-way ANOVAs using Sidak’s tests for multiple comparisons (C to H). *, P < 0.05; ***, P < 0.001; ****, P < 0.0001.
Primary macrophages are more efficient at controlling the growth of the Δnrp strain.
Altogether, our findings suggest a more effective control of the Δnrp strain by the host during the innate immune response. Since macrophages are one of the first types of cells to be infected by
DISCUSSION
TB remains a devastating disease to mankind, with high human and economic tolls (25, 26). To efficiently tackle TB, an improvement of several current tools is needed, notably with regard to prevention, diagnosis, and treatment. Owing to the alarming drug resistance rates, which caused 480,000 cases of complicated TB in 2016 (1), there is a pressing need for the development of novel treatment strategies. In this context, a clearer understanding of the key virulence factors in
Previous studies reporting that the nrp gene harbored by
A previous screen aimed at finding
Our findings collectively support a role for nrp during the early phases of the immune response and demonstrate that
The most notable and interesting finding on the role of nrp in TB pathogenesis came from the observation that the lack of this gene highly attenuates
In all, our findings highlight a role for the nrp cluster in evading the basic microbicidal mechanisms of the macrophage, which is then overcome by the adaptive immune response. It is tempting to speculate that the mechanisms that enable this result from host-pathogen coevolution events. Several outstanding questions arise from this study: which is the mechanism hijacked by Nrp at the level of innate immunity? How does the acquired immune response abrogate the impact of Nrp in the macrophage? As an answer to these questions, the infection of mice deficient for Toll-like receptor 2 (TLR2), TLR4, IL-10, arachidonate 5-lipoxygenase (ALOX5), or arachidonate 15-lipoxygenase (ALOX15) with the
MATERIALS AND METHODS
Comparative genomics.
Sequence identity scores were obtained by using the BLAST+ 2.6.0 (30) application to search a local database of 72 genomic sequences from species selected to be representative of the phylogeny of the genus Mycobacterium (19) (NZ_LQOM00000000, NZ_LQOQ00000000, NZ_LQOR00000000, NZ_LQOS00000000, NZ_LQOT00000000, NZ_LQOY00000000, NZ_ARBU00000000, NZ_LQPB00000000, NZ_LQPI00000000, NZ_ADNV00000000, NZ_LQPP00000000, NZ_CCAU000000000, NZ_MIHD00000000, NZ_LQQA00000000, CP000480, GCA_000243415.3_ASM24341v3, GCA_002102065.1_ASM210206v1, NC_000962, NC_002677, NC_002945, NC_008595, NC_008611, NC_008726, NC_010397, NC_010612, NC_014814, NC_015564, NC_015576, NC_015758, NC_015848, NC_016604, NC_016946, NC_018027, NC_018612, NC_019966, NC_020133, NC_021715, NC_022663, NZ_AGVE00000000, NZ_AJFI00000000, NZ_APKD00000000, NZ_CCBF000000000, NZ_CP007220, NZ_CP011269, NZ_CP011491, NZ_CP011530, NZ_CP014475, NZ_CP015278, NZ_CP020821, NZ_CSTD00000000, NZ_LQOJ00000000, NZ_LQOL00000000, NZ_LQOU00000000, NZ_LQOV00000000, NZ_LQOW00000000, NZ_LQOX00000000, NZ_LQOZ00000000, NZ_LQPC00000000, NZ_LQPD00000000, NZ_LQPE00000000, NZ_LQPF00000000, NZ_LQPG00000000, NZ_LQPH00000000, NZ_LQPJ00000000, NZ_LQPN00000000, NZ_LQPO00000000, NZ_LQPQ00000000, NZ_LQPR00000000, NZ_LQPS00000000, NZ_LQPT00000000, NZ_LQPU00000000, NZ_LQPW00000000, and NZ_LQPX00000000). The database was queried using the sequence of the nrp cluster (Rv00097-Rv0101) from H37Rv (NC_000962.3). The sequence identity score was calculated by combining the query coverage, E value, and identity values for each hit with the weights 0.5, 0.25, and 0.25, respectively. A multiple-genome alignment of the sequences NC_002677 (
Construction of a
A null mutant of
Generation of an nrp-complemented strain.
A 28-kb
Bacterial growth and stocks.
Bacteria were grown in Middlebrook 7H9 liquid medium (BD Biosciences) supplemented with 0.05% Tween 80, 0.2% glycerol, and 10% oleic acid-albumin-dextrose-catalase (OADC) for 7 to 10 days and then subcultured in Proskauer Beck (PB) medium supplemented with 0.05% Tween 80 and 2% glycerol to the mid-log phase. Bacterial stocks were aliquoted and frozen at −80°C. To determine the concentration of
Ethics statement.
All animal experiments were performed in strict accordance with the recommendations of the European Union Directive 2010/63/EU. All experimental procedures were approved by the local animal ethics committee, and licensed by the Portuguese National Authority for Animal Health (DGAV, Portugal) with reference number 014811/2016-07-13. Animal procedures were conducted by laboratory personnel with accreditation for animal research given by DGAV. The mice were euthanized by CO2 inhalation, with efforts to minimize suffering.
Mouse strains.
C57BL/6 mice were purchased from Charles River Laboratory (Barcelona, Spain). RAG2-deficient mice were purchased from Instituto Gulbenkian de Ciência (IGC) or generously provided by Margarida Correia-Neves (ICVS), and IFN-γ deficient mice were generously provided Susana Roque (ICVS). All transgenic mice were of a C57BL/6 genetic background. SCID mice were obtained from Charles River Laboratory (USA).
Experimental infection.
Mice were infected with
Survival.
The weights of the infected mice were determined every week after infection for immunocompetent mice and every week after infection for the first 30 days and every 48 h from day 30 postinfection onwards for immunodeficient hosts. Mice were humanely euthanized if they lost 20% of their maximum weight or upon losing responsiveness to physical stimulation. Whenever possible, the lungs of moribund animals were harvested for histology and bacterial burden assessment.
Organ processing.
At the indicated time points postinfection, the mice were euthanized by CO2 asphyxiation, and the organs were aseptically excised and processed as described previously (35, 36). Briefly, the lungs were perfused by applying 10 ml phosphate-buffered saline (PBS) through the right ventricle of the heart to flush blood cells, and single-cell suspensions were prepared in complete Dulbecco’s modified Eagle medium (cDMEM; DMEM supplemented with 10% fetal bovine serum [FBS], glutamine, HEPES, and sodium pyruvate, all from Gibco). Single-cell suspensions were used for bacterial burden determination, flow cytometry analysis, and RNA extraction. The number of cells was counted using a Countess automated cell counter (Life Technologies).
Bacterial burden determination.
The assessment of initial bacterial burden was performed 3 days postinfection by growing viable bacteria from whole-lung homogenates. For the other time points, lung and spleen single-cell suspensions were incubated with 0.1% saponin (Sigma-Aldrich) for 10 min to release intracellular bacteria. The CFU was determined by plating 10-fold serial dilutions of saponin-treated cell suspensions on Middlebrook 7H11 agar plates supplemented as described in “Bacterial growth and stocks.” BBL MGIT PANTA antibiotic mixture (BD Bioscience) was added to prevent the contamination of lung samples. Viable
Flow cytometry.
Lung cells (1 × 106 to 2 × 106) were stained for surface antigens for 30 min at 4°C. Stained cells were washed and then fixed overnight in PBS containing 2% paraformaldehyde. The following antibodies were used: CD3-PE (clone 145-2C11; eBioscience), CD4-PB (clone RM4-5; eBioscience), CD8-FITC (clone 5H10-1; BioLegend), CD44-PerCPCy5.5 (clone 1M7; eBioscience), CD62L-PECy7 (clone MEL-14; BioLegend), CD19-APC (clone eBio1D3; eBioscience), Ly6G-APC (clone 1A8; BioLegend), Ly6C-PerCPCy5.5 (clone AL-21; BD Pharmingen), MHC II-FITC (clone AMS-32.1; BD Pharmingen), CD11b-PE (clone M1/70; BioLegend), and CD11c-PB (clone N418; BioLegend). Samples were run on an LSRII flow cytometer with Diva software, and the data were analyzed using FlowJo version 10.1.r7 software. The total number of cells in each gate was calculated using the total number of cells determined by the Countess automated cell counter. The gating strategy used is shown in Fig. S2 in the supplemental material.
RNA extraction, reverse transcription, and real-time PCR.
Total RNA from infected lungs was extracted with TRIzol reagent (Invitrogen), and cDNA was synthesized and analyzed by real-time PCR, as described previously (35, 36). Target IFN-γ and TNF mRNA expression was quantified using SYBR green (GrisP) and specific oligonucleotides and normalized to the ubiquitin mRNA levels. The expression of CCL2 and CCL7 was quantified using specific primer-probe sets (Applied Biosystems) and normalized to the expression of hypoxanthine phosphoribosyltransferase (HPRT).
Histology analysis.
The right upper lobes of the lungs were fixed in PBS containing 3.7% formaldehyde, embedded in paraffin, sectioned in 2- to 3 -μm-thick slices, and stained with hematoxylin-eosin (35). The lung surface area of inflammation was measured using ImageJ software (version 1.50e; NIH). The percentage of total lung area involved with inflammation was calculated by dividing the cumulative area of inflammation by the total lung surface area for each sample.
Infection of bone marrow-derived macrophages and peritoneal macrophages.
BMDM were differentiated from bone marrow cells obtained from the femurs and tibiae of C57BL/6 mice and cultured in the presence of L-cell conditioned medium, as described previously (37). pMac were obtained by intraperitoneally injecting 1 ml of thioglycolate into WT C57BL/6 mice. After 4 days, the mice were euthanized and the peritoneums were washed with PBS. In both cases, 1 × 106 cells were infected with Δnrp or nrp-comp strains at a multiplicity of infection (MOI) of 1 bacteria to 1 macrophage. Four hours after infection, the cells were washed 4 times with PBS (Gibco) to remove extracellular bacteria. The adherent cells were cultured at 37°C in 1 ml of cDMEM in the presence or absence of 100 U/ml of IFN-γ for 96 h. Four or ninety-six hours postinfection, 0.1% saponin (Sigma-Aldrich) in PBS was added to the wells, and the cells were incubated at room temperature (RT) for 10 min to release intracellular bacteria. The number of viable bacteria was determined by plating 10-fold serial dilutions of the saponin-treated cell suspensions on supplemented Middlebrook 7H1 agar, as described in “Bacterial growth and stocks.”
Cytokine determination by ELISA.
TNF, IL-1β, and IL-10 concentrations in the supernatants of infected macrophages were determined 24 h postinfection by enzyme-linked immunosorbent assays (ELISAs) with commercially available kits (eBioscience), according to the manufacturer’s instructions. Cytokine levels from uninfected cells were below the assay level of detection (not shown).
Cell viability assay.
Cell viability was determined in noninfected or infected macrophages 2 or 4 days postinfection. Macrophages were gently detached from low-adherence tissue culture plates using a cell scraper. Cell viability was assessed by using trypan blue exclusion.
Statistical analysis.
Data were analyzed using GraphPad Prism 6. The differences between groups were analyzed with two-way analyses of variance (ANOVAs) using Sidak’s tests for multiple comparisons. Kaplan-Meier survival curves were analyzed with the log-rank (Mantel-Cox) test. Differences were considered significant at P values of ≤0.05.
b ICVS, University of Minho, Braga, Portugal
c ICVS/3B’s–PT Government Associate Laboratory, Braga, Portugal
d i3S–Instituto de Investigação e Inovação em Saúde, Porto, Portugal
e IBMC–Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
f Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, New York, USA
g School of Biosciences and Institute of Microbiology and Infection, University of Birmingham, Birmingham, United Kingdom
h Howard Hughes Medical Institute, Albert Einstein College of Medicine, New York, New York, USA
Washington University in St. Louis School of Medicine
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Copyright © 2018 Bhatt et al. This work is published under https://creativecommons.org/licenses/by/4.0/ (the “License”). Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
Abstract
ABSTRACT
Nonribosomal peptide synthases produce short peptides in a manner that is distinct from classical mRNA-dependent ribosome-mediated translation. The
IMPORTANCE Over 10 million people developed tuberculosis (TB) in 2016, and over 1.8 million individuals succumbed to the disease. These numbers make TB the ninth leading cause of death worldwide and the leading cause from a single infectious agent. Therefore, finding novel therapeutic targets in
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer