Persistent p55tnfr expression impairs t cell responses during chronic tuberculosis and promotes reactivation

Nature

Persistent p55tnfr expression impairs t cell responses during chronic tuberculosis and promotes reactivation"


Play all audios:

Loading...

ABSTRACT The pleiotropic activities of TNF are mediated by two structurally related but functionally distinct type I transmembrane receptors, p55TNFR and p75TNFR expressed in most cell


types, that can be cleaved and act as TNF scavengers. Here, we investigated the effect of persistent p55TNFR cell surface expression during aerosol inhalation challenge with virulent _M.


tuberculosis_ H37Rv. We demonstrated that persistency of p55TNFR in macrophage cultures increased the synthesis of soluble TNF, p75TNFR and NO, however, had no effects on bacteria killing


ability. Furthermore, it did not facilitate enhanced protection to primary acute _M. tuberculosis_ infection in p55∆NS mice. Without exacerbated lung inflammation, we found a compensatory


increase in p75TNFR shedding and decrease in bioactive TNF in BAL of p55∆NS mice after _M. tuberculosis_ challenge. Defective expressions of CD44 and INFγ attributed to an impaired T cell


response during persistent p55TNFR expression that caused marginal transient susceptibility during chronic infection. Moreover, persistent p55TNFR expression induced early reactivation


during latent tuberculosis infection. These data indicate a prominent role of p55TNFR shedding in Th1 mediated protection against chronic and latent tuberculosis infection. SIMILAR CONTENT


BEING VIEWED BY OTHERS DIFFERENTIAL REQUIREMENT OF FORMYL PEPTIDE RECEPTOR 1 IN MACROPHAGES AND NEUTROPHILS IN THE HOST DEFENSE AGAINST _MYCOBACTERIUM TUBERCULOSIS_ INFECTION Article Open


access 09 October 2024 HUMAN M1 MACROPHAGES EXPRESS UNIQUE INNATE IMMUNE RESPONSE GENES AFTER MYCOBACTERIAL INFECTION TO DEFEND AGAINST TUBERCULOSIS Article Open access 19 May 2022 AUTOPHAGY


PROMOTES EFFICIENT T CELL RESPONSES TO RESTRICT HIGH-DOSE _MYCOBACTERIUM TUBERCULOSIS_ INFECTION IN MICE Article 27 February 2024 INTRODUCTION Tumor Necrosis Factor (TNF) is a member of the


TNF superfamily (TNFSF), a class of structurally related cytokines that are involved in diverse immunological and developmental pathways1,2. Murine TNF is initially synthesized as a 26 kDa


glycosylated type II transmembrane molecule (Tm-TNF) that can be released from the cell surface by the metalloprotease TNF-α-converting enzyme (TACE) to generate a 17 kDa protein, and


subsequent to homotrimerisation, a 51 kDa soluble TNF (solTNF) molecule is formed3,4,5,6. Both TNF forms are biologically active and their extensive biological effects are mediated by two


receptors: p55TNFR (TNFRSF1A, CD120a, TNFR1) and p75TNFR (TNFRSF1B, CD120b, TNFR2) with Tm-TNF preferentially signaling through p75TNFR and solTNF binding strongly to p55TNFR7. Both


receptors are type I transmembrane proteins sharing approximately 25% homology in their extracellular domains but have divergent cytoplasmic domains. Their extracellular domains can be


released from the cell surface through proteolysis mediated by metalloproteases of the ADAM family, thereby generating soluble TNFRs capable of binding TNF8,9. Soluble TNFRs are thought to


serve as (a) TNF antagonists, (b) TNF carrier proteins (c) a slow release reservoir for TNF, and as (d) stabilizers of TNF-bioactivity10 and have been detected during various disease states


including typhoid fever11, malaria12, sub-acute bacterial endocarditis13 and visceral leishmaniasis14, and mycobacterial infections including tuberculosis15,16,17. Numerous clinical studies


have demonstrated increased serum TNF receptors concentrations, associated with severity of M. tuberculosis disease18,19. TNF shares its receptors with Lymphotoxin (LT); in its homotrimeric


form LTα interacts with TNFRp55 and p75 while the LTαLTβ heterotrimers, interact with LTβ-R20. TNF gene deletion21,22,23,24, p55TNFR gene deletion16,25,26, and TNF neutralization


studies27,28 demonstrated TNF-TNFR signaling as critical for protective immunity against mycobacterial infections. In the absence of TNF-p55TNFR signaling, challenge of mutant mice with


either attenuated _M. bovis_ BCG29 or virulent _M. tuberculosis_21,24,25 resulted in elevated bacterial burdens, impaired granuloma structure formation and early death. Owing to its strong


pro-inflammatory and immunostimulatory activities, the duration and amplitude of TNF functions are tightly regulated. One mechanism, which controls bioavailability of TNF, is p55TNFR and


p75TNFR shedding. Both soluble p55TNFR and p75TNFR are found in the lung of _M. tuberculosis_ infected patients and mice15,16,30,31. However, virulent pathogens such as _M. tuberculosis_


H37Rv and _M. avium_ can actively manipulate this system to evade the immune response15,16,32. Indeed, we showed recently that a major role of p75TNFR in mycobacterial infection control


seems to be through its shed soluble form, which effectively regulate the level of bioactive TNF that contributes to the disease outcome16. Patients treated with TNF inhibitors including


Etanercept, a p75TNFR IgG fusion protein show a marked increased risk for TB infection33. Here we address the specific role of shed, soluble p55TNFR in host control of M. tuberculosis


infection. The generation of a nonsheddable p55TNFR (p55ΔNS) mouse strain has provided insight into the roles of both the soluble and the membrane-bound forms of p55TNFR in pathogenesis of


infectious, inflammatory and autoimmune diseases. Xanthoulea and colleagues indicated that mice expressing a mutated nonsheddable form of p55TNFR (p55∆NS) developed a Toll-like receptor


dependent innate immune hyper responsiveness34. In this mouse model p55TNFR is rendered nonsheddable on all cell types which express p55TNFR. We thus investigated the role of signaling and


regulatory effects of nonsheddable p55TNFR in host immunity against _M. tuberculosis_. We report here after _M. tuberculosis_ aerosol exposure that p55∆NS mice expressing nonsheddable


p55TNFR controlled acute infection similar to their WT counterparts while they exhibited a transient loss of control during chronic infection and an increased susceptibility to reactivation


of latent tuberculosis. RESULTS SUSTAINED P55TNFR SURFACE EXPRESSION IN P55∆NS PERITONEAL ELICITED CELLS AFTER _M. TUBERCULOSIS_ INFECTION Numerous studies have demonstrated release of TNFRs


after infection or immunization _in vitro_ and _in vivo_15,16. TNFR shedding from cell surfaces coincides with the generation of TNF16,17; its functional significance being associated with


the regulation of TNF mediated effects35,36,37,38. Xanthoulea and colleagues showed that upon activation with PMA, peritoneal exudate cells derived from p55∆NS mice expressed a nonsheddable


p55TNFR34. Here, we investigated the relationship between p55TNFR expression and _M. tuberculosis_ infection. Thioglycollate elicited macrophages from WT mice or p55∆NS mice were exposed to


_M. tuberculosis_ H37Rv for 20 minutes and p55TNFR cell surface expression determined by flow cytometry. p55TNFR expression occurred in both WT and p55∆NS unstimulated macrophages (Fig. 1a).


However, exposure to _M. tuberculosis_ H37Rv caused a loss of p55TNFR surface expression on WT macrophages (reflected by reduced Mean Fluorescent Intensity) compared to the elevated and


sustained expression noted in p55∆NS macrophages (Fig. 1a). Quantification of the cleaved p55TNFR revealed detectable levels of soluble p55TNFR present in unstimulated culture supernatants


of thioglycollate elicited macrophages from WT mice, which increased significantly at all the time points (p < 0.01) after activation with LPS (Fig. 1b) and increased significantly at 90


and 150 minutes after viable _M. tuberculosis_ H37Rv (Fig. 1c) infection (p < 0.01). No soluble p55TNFR was detected in supernatants of p55∆NS macrophages before or after stimulation. The


data confirms sustained expression of p55TNFR in p55∆NS mice and demonstrate that p55TNFR shedding is induced by _M. tuberculosis_. ENHANCED MACROPHAGE BACTERICIDAL POTENTIAL DOES NOT


CORRELATE WITH INCREASED BACILLI KILLING IN THE PRESENCE OF SUSTAINED P55TNFR EXPRESSION We hypothesized that sustained p55TNFR expression may allow for continued or amplification of TNF


signalling yielding improved control of infection. To determine how sustained membrane p55TNFR expression influences restriction of bacterial replication, we investigated the bactericidal


potential and bacterial killing in bone marrow derived macrophages (BMDM). Unlike the thioglycolate elicited macrophages, BMDM primary cultures represent a more naïve homogeneous macrophage


population which can be stimulated and activated under different experimental conditions. These BMDM’s from WT mice and p55∆NS mice were pre-activated with (equimolar) 100 U/mL IFNγ


concentrations for 24 hours to achieve optimal macrophage activities similar to those during inflammation, in particular the NO production by active macrophages. This requirement for IFNγ


pre-activation was confirmed by initial optimization studies in which nitrite production were below the baseline without IFNγ pre-stimulation (Supplementary Fig. 1). TNF and nitric oxide


(NO) levels were quantified following stimulation with LPS or _M. tuberculosis_ H37Rv. Consistent with published data34, stimulating p55∆NS derived macrophages with 100 ng/ml LPS increased


TNF synthesis in a time-dependent manner with a significant increase (_p_ < 0.05) observed 300 minutes post stimulation compared to WT derived macrophages (Fig. 2a). Similarly,


significantly higher TNF production was observed in p55∆NS derived macrophages when challenged with _M. tuberculosis_ H37Rv, however TNF concentrations were found to be approximately 10 fold


less compared to LPS stimulation (Fig. 2d). TNF and IFNγ synergize for optimum macrophage activation resulting in production of the effector molecule NO39,40, which can be measured


relatively by the Griess Reagent System to determine the nitrite (NO2−) concentration as one of two primary stable breakdown products of NO. Therefore, we quantified nitrite concentrations


in supernatants of activated macrophages and found levels significantly enhanced (_p_ < 0.05) in p55∆NS derived macrophages relative to WT controls after stimulation with LPS for 48 hours


(Fig. 2c). Similar results were attained when macrophages were stimulated with _M. tuberculosis_ H37Rv, with a significant increase in nitrite concentration (_p_ < 0.001) as early as 24 


hours after stimulation and a further increase in NO synthesis (_p_ < 0.05) observed 48 hours later in p55∆NS mice compared to WT mice (Fig. 2f). Therefore, the increased synthesis of TNF


and NO in p55∆NS macrophages points to a heightened state of macrophage activation and bactericidal potential. On the contrary, _M. tuberculosis_-GFP bacilli killing assays revealed no


significant enhancement of bactericidal ability in p55∆NS macrophages compared to WT macrophages (Fig. 2g). Both TNFRs are proteolytically cleaved from the cell surface in the presence of


inflammatory stimuli and the levels of soluble TNFRs are thought to modulate TNF responsiveness. We and others have shown that increased shedding implied a more dominant role for p75TNFR in


regulating the bioactivity of TNF16,41. To determine whether there was a difference in p75TNFR shedding between WT and p55ΔNS derived macrophages, the levels of soluble p75TNFR were also


quantified in the supernatants after stimulation with LPS or _M. tuberculosis_ H37Rv. Results showed that p55ΔNS derived macrophages produced significantly higher levels (_p_ < 0.01) of


soluble p75TNFR when stimulated with either LPS (Fig. 2b) or _M. tuberculosis_ H37Rv (Fig. 2e) for 300 minutes. To ascertain whether p75TNFR shedding affects the bioactivity of TNF in p55ΔNS


mice, we challenged the mice with M. tuberculosis H37Rv by aerosol inhalation. Here we found higher levels of p75TNFR and lower levels of bioactive TNF in the bronchoalveolar lavage (BAL)


fluid of p55ΔNS mice compared to WT mice at both day 3 and day 6 post-infection (Fig. 2h,i) that corroborates our previous findings16. These data suggest that defective p55TNFR shedding


results in significant increased release of soluble p75TNFR as a compensatory mechanism with potential to modulate TNF activity. SUSTAINED EXPRESSION OF SURFACE P55TNFR DOES NOT ENHANCE


MYCOBACTERICIDAL RESPONSES DURING ACUTE _IN VIVO M. TUBERCULOSIS_ INFECTION The abrogation of TNF-p55TNFR signaling, either genetically or with neutralizing antibodies, results in


susceptibility to mycobacterial and other intracellular infections21,25,28,42. Specific functions attributable to soluble and membrane TNFRs in various infections and disease states are


still not fully understood. In both mice and humans, defective p55TNFR shedding resulted in enhanced sensitivity to TNF mediated immunopathology43,44. Xanthoulea and colleagues reported


enhanced innate immune activation in p55∆NS mice, which mediated increased resistance to _L. monocytogenes_ infection _in vivo_ and suggested that persistently expressed membrane p55TNFR


conferred enhanced protective antibacterial immunity34. In consideration of these results, we addressed the _in vivo_ functional role of membrane bound p55TNFR and receptor shedding during


_M. tuberculosis_ H37Rv infection. WT mice and p55∆NS mice were exposed to a low dose aerosol inhalation infection of 50–100 CFUs/mouse _M. tuberculosis_ H37Rv and pulmonary bacilli burdens


were assessed over 8 weeks. Results showed that p55ΔNS mice controlled acute infection with comparable kinetics to WT mice reflected by equivalent bacilli numbers in the lungs (Fig. 3a) at


all the acute time points investigated. We also observed similar degrees of pulmonary inflammation in terms of comparable lung weights (Fig. 3b) and total cell counts of the lung (Fig. 3c).


Further, pulmonary pathology was qualitatively equivalent with no differences in cellular infiltration or granuloma formation (Fig. 3d). Thus sustained p55TNFR expression does not enhance


the bactericidal capacity of the host during acute _M. tuberculosis_ infection. MACROPHAGE FUNCTION IS NOT ENHANCED IN THE PRESENCE OF SUSTAINED P55TNFR EXPRESSION _IN VIVO_ DURING ACUTE _M.


TUBERCULOSIS_ INFECTION To understand how persistent p55TNFR expression affects macrophage function _in vivo_ we measured and compared recruitment of CD11b+ cells to the lungs and their


activation state in _M. tuberculosis_-infected WT mice and p55ΔNS mice. Flow cytometric analyses of pulmonary cells revealed equivalent frequencies of CD11b+ cell recruitment in p55∆NS mice


compared to WT mice at day 21 post-infection (Fig. 4a,b; Gating strategy in Supplementary Fig. 2). To determine macrophage activation under conditions of sustained p55TNFR presence, we


analyzed the frequencies of CD11b+MHC-II+ cells as well as the MFIs of cell surface MHC-II, CD80 and CD86 expression on CD11b+ cells in the lungs at 21 days post-infection. p55ΔNS mice


displayed comparable CD11b+MHC-II+ cells compared to WT mice 21 days post infection (Fig. 4c). The number of CD11b+CD80+ and CD11+CD86+ cells as well as the MFIs of MHC-II, CD80 and CD86


expressed on CD11b+ cells was also found to be equivalent in both strains (Data not shown). The data indicate that persistent membrane p55TNFR expression does not enhance the expression of


costimulatory molecules on CD11b+ cells. To determine the influence of persistent p55TNFR expression on macrophage function _in vivo_, IL-12p70, NO and TNF concentrations were measured in


the lungs of _M. tuberculosis_-infected WT mice and p55∆NS mice. IL-12p70 and TNF are critical mediators of anti-mycobacterial immunity and primarily synthesized by


macrophages45,46,47,48,49. TNF levels in p55∆NS mice were significantly higher (_p_ < 0.05) at day 21 compared to WT mice (Fig. 4d), while IL-12p70 concentrations showed a non-significant


trend towards enhancement in p55∆NS mice compared to WT mice at 21 days post-infection but was significantly reduced si at day 28 (Fig. 4e). NO levels in lung homogenates of p55∆NS mice


were significantly lower (p < 0.001) compared to WT mice at 28 and 49 days post-infection (Fig. 4f). Collectively the data indicates that sustained p55TNFR expression does not enhance


macrophage activation. TH1 IMMUNE ACTIVATION IS IMPAIRED IN P55∆NS MICE As a key determinant of disease outcome we investigated whether sustained p55TNFR expression influences Th1 immune


function. Here, we first evaluated the influx and activation state of CD4+ T-cells in the lungs of infected WT mice and p55∆NS mice by flow cytometry. CD44 and IFNγ were used as markers of T


cell activation. CD44 is a cell adhesion receptor and wildly distributed. Ligation of CD44 with hyaluronanis results in a variety of downstream responses that includes cell adhesion, cell


migration, and hematopoietic process. In mature lymphocytes, CD44 is upregulated in response to antigenic stimuli and represents the effector stage of immunological responses50,51,52. The


use of CD44 as marker for effector T cells is generally accepted and demonstrated in various fields of studies16,53,54. Comparable frequencies of CD4+ T cells were present between the two


strains at both day 14 and day 21 post-infection (Fig. 5a,b; Gating strategy in Supplementary Fig. 2), Interestingly while equivalent frequencies of pulmonary CD4+CD44+ T cells were measured


in both strains 14 days and 21 days post-infection (Fig. 5c), the MFI of cell surface CD44 expression gated on CD4+ T cells was lower in p55∆NS mice compared to WT mice, significantly so


(_p_ < 0.05) at day 14 post-infection (Fig. 5d,e). The data illustrate that despite the comparable CD4+ T cell recruitment to lungs of WT mice and p55∆NS mice, persistent p55TNFR


expression might play a suppressive role in T cell activation during early infection. We next compared the kinetics of IFNγ production in the lungs of infected WT mice and p55∆NS mice as a


further measurement of _in vivo_ T cell functionality. We observed lower IFNγ concentrations with significant differences at day 21 (_p_ < 0.05) and day 28 (_p_ < 0.01) post-infection


in p55∆NS mice compared to WT mice (Fig. 5f). The data suggest that persistent p55TNFR expression may suppress pulmonary T cell activation which associates with reduced IFNγ production.


SUSTAINED EXPRESSION OF SURFACE P55TNFR RESULTS IN MARGINALLY IMPAIRED BACTERICIDAL RESPONSES DURING CHRONIC _IN VIVO M. TUBERCULOSIS_ INFECTION We postulated that a dysregulated Th1 immune


response in p55∆NS mice may compromise long term host protective immunity during a primary infection. Therefore, to determine the effect of sustained p55TNFR expression on disease outcome


during chronic stages of a primary infection, _M. tuberculosis_ infected p55∆NS mice and WT mice were monitored for >500 days. The bacilli burdens of p55∆NS mice were higher than those of


WT mice at 180 days post-infection and, although moderate, was nonetheless significantly lower at 300 days post-infection (p < 0.05) (Fig. 6a) which correlated with a non-significant


trend towards more rapid death in p55∆NS mice as compared to WT controls, where WT mice had an increased medium survival of 476 days compared to 395 days in p55∆NS mice (Fig. 6b). Although


the analysis of total lung cell counts (Fig. 6d) and free alveolar space (Fig. 6e) showed no statistical difference, we observed a transient increase in lung histopathology (Fig. 6f) at 180


days, which was quantitatively confirmed by an increase in lung weights (Fig. 6c) at this time point in p55∆NS mice. The data therefore indicates that persistent p55TNFR surface expression


compromises protection against chronic _M. tuberculosis_ infection. SUSTAINED EXPRESSION OF SURFACE P55TNFR RESULTS IN LOSS OF CONTROL AGAINST REACTIVATION OF LATENT MYCOBACTERIAL INFECTION


Our data demonstrated that persistent p55TNFR expression might play a suppressive role in T cell activation, we next assessed the quality of the T cell response generated in a reactivation


challenge model. The rationale was that defects in the development of a fully functional T cell response during the primary infection might compromise the ability of _M. tuberculosis_


specific T cells to protect against reactivation of a latent infection. Infected p55∆NS mice and WT mice were treated with rifampicin and isoniazid for 6 weeks, beginning at day 28


post-infection to induce a state of latency and were subsequently assessed at 90, 180, 300 days for reactivation of tuberculosis. Similar to our earlier finding, the acute phase of latent


infection showed no differences in CFU burdens (Fig. 7a). Low bacilli burdens measured at 90 days post-infection confirmed successful chemotherapy in both strains. However p55∆NS mice showed


enhanced reactivation of disease with significantly higher bacilli burdens by 300 days (Fig. 7a) accompanied by significantly higher lung weights (Fig. 7b) compared to WT control mice.


Despite the equivalent numbers of total lung cells (Fig. 7c) and percentages of free alveolar space (Fig. 7d), we observed microscopically higher degree of inflammation in the lung of p55∆NS


mice than in WT mice (Fig. 7e). To further assess the effects of persistent p55TNFR expression on p75TNFR levels and the cytokine productions during reactivation of latent M. tuberculosis


infection, we compared the levels of TNF, p75TNFR, NO and IL-12p70 in the lungs of WT mice and p55∆NS mice at 90 and 300 days post-infection. Both TNF and p75TNFR levels in p55∆NS mice were


similar to WT mice at day 90 but significantly increased at day 300 (p < 0.05) (Fig. 8a,b). Nitrite concentrations in p55∆NS infected lungs were significantly lower at day 90 (p < 


0.05) but higher at day 300 (p < 0.05) than the WT lungs (Fig. 8c), Pulmonary IL-12p70 concentrations were reduced significantly (p < 0.01) in p55∆NS mice at both day 90 and 300 (Fig.


8d). We next assessed the quality of the T cell response generated during latent infection by analyzing the activation state of CD4+ T-cells in infected lungs after 6 weeks of rifampicin and


isoniazid treatment, and a further 3 weeks of infection reactivation. While comparable frequencies of pulmonary CD4+ T cells were present between the two strains at 90 days post-infection


(Fig. 9a,b), the frequencies of pulmonary CD4+CD44+ T cells were reduced in p55∆NS mice (Fig. 9a,c). The MFI of cell surface CD44 gated on CD4+ T cells was also significantly lower in p55∆NS


mice compared to WT mice (Fig. 9d,e). Moreover, we observed significantly lower IFNγ concentrations in p55∆NS mice compared to WT mice (Fig. 9f). Together the data indicates an inability of


p55∆NS mice to control reactivation of latent tuberculosis, which may be linked to the fact that persistent p55TNFR expression suppresses effector T cell activation during latent infection.


DISCUSSION TNF-p55TNFR mediated signaling is a critical requirement for control and resolution of mycobacterial infection21,22,28,29. The widespread expression of TNFRs on different cell


types and tissues55,56,57 indicates the spectrum breath of target cells with which TNF can interact and necessitates tight regulation around the TNF-TNFR signaling axis. TNF mediated


activities are controlled by multiple regulatory mechanisms including proteolytic shedding of TNFRs from cell surfaces. The resultant soluble p55TNFR and p75 bind TNF, competing with cell


surface receptors and limit TNF function in target cells35,36,37,38. We and others have shown TNFRp55 mediated TNF signaling to be essential to generate host immune protection, while shed,


soluble p75TNFR has a critical role in regulating TNF bioavailability15,16. Clinically, increased soluble p55TNFR and p75TNFR has been associated with exacerbated _M. tuberculosis_ disease


severity18,19,30. In humans, specific missense mutations in the p55TNFR gene that cause defective receptor shedding strongly associate with autosomal dominant periodic fever syndromes known


as TRAPS44, and are thought to be primarily caused by irregular innate immune function in these patients. The effects of persistent membrane TNFR expression, in the absence of the shed form,


within disease settings are unknown. In this study, we investigated the functional relevance of persistent p55TNFR cell surface expression and impaired p55TNFR shedding during infection


with _M. tuberculosis_ H37Rv using a mutant mouse strain (p55∆NS) with p55TNFR wild type receptor functionality but defective shedding capability34. We confirmed that p55TNFR and p75TNFR are


released from the cell surface prior to and after macrophage activation with either LPS or viable _M. tuberculosis_ H37Rv bacilli, in agreement with previous reports demonstrating that


soluble TNFRs are generated after cell activation58,59,60. Moreover, our results show that both constitutive and _M. tuberculosis_ induced shedding are inhibited in p55∆NS derived cells,


correlating with persistency of p55TNFR cell surface expression. Interference of the TNF-p55TNFR signaling pathway either by gene deletion of ligand or receptor, or by neutralizing


antibodies leads to susceptibility to mycobacterial and other intracellular pathogens21,29,61,62,63. We hypothesized that persistent cell surface p55TNFR expression would result in enhanced


TNF-p55TNFR interaction leading to improved disease outcome. In contrast, our data demonstrate that persistent cell surface p55TNFR expression does not confer a protective advantage against


_M. tuberculosis_ H37Rv infection in either acute or chronic infection. We have previously reported that treatment of WT dendritic cells with recombinant p75TNFR reduced bioactive TNF levels


which resulted in impaired dendritic cell activation. In contrast treatment with anti-p75TNFR had the opposite effect with increased bioactive TNF translating to enhanced dendritic cell


activation16. We would therefore postulate an enhanced protection in these p55∆NS mice by blocking its p75TNFR to increase macrophage activation and killing ability. Although the p55∆NS


macrophages from our _in vitro_ experiments exhibited better bactericidal potential, the p55∆NS mice failed to demonstrate improved bacterial control, which was expected considering the


similar _in vitro_ bacilli killing abilities in WT and p55∆NS macrophages. However caution must be exercised when directly comparing _in vitro_ and _in vivo_ data; the antigen presenting


cells (APC’s) in the lungs are comprised of a more complex array of cells which include inflammatory monocytes, alveolar macrophages and dendritic cells. The alveolar macrophages are the key


cells to first line defense against _M. tuberculosis_ in the lung; however they are not the ideal cell type for our _in vitro_ experiments to study bacilli killing. We used IFNγ activated


BMDM’s resembling inflammatory monocytes, whose primary immune function is bacilli killing during infections. We found that mice expressing the mutant persistent cell surface receptor


succumbed to _M. tuberculosis_ chronic infection earlier than the WT group with a significant increase in bacilli burdens. The p55∆NS mice also demonstrated a loss of bacilli control


resulting during latent infection. These data demonstrate that the host protection against tuberculosis infection was not improved during persistent p55TNR expression as initially


hypothesized. These findings contrasted earlier studies which reported that challenging p55∆NS mice with increasing doses of _L. monocytogenes_ results in improved host resistance compared


to control animals. Under such conditions sustained p55TNFR expression enhanced antibacterial host mechanisms and suggested that _L. monocytogenes_ might evoke p55TNFR shedding as an immune


escape mechanism34. This contrast highlights the differences in the pathogenesis and immunity of these two pathogens. For example, the functions of membrane TNF has been studied in both


listeria and tuberculosis disease models. Using the mice with noncleavable membrane TNF (mem-TNF) Torres _et al_. showed partial protection against Listeria in a dosage dependent manner64.


On the other hand, Fremond _et al_. reported that the mem-TNF mice were protected from the Mycobacterium tuberculosis acute infection but not the chronic infection65. We investigated host


immune responses in p55∆NS mice to gain deeper understanding of immune function during _M. tuberculosis_ infection. We found that pulmonary cellular recruitment did not increase over the


course of infection in p55∆NS mice relative to control animals. This outcome was interesting, as TNF is known to be a master orchestrator of inflammation. Closer inspection of the lung cell


populations during early infection revealed a transient increase in the frequency of CD11b+/MHC-II+ expressing cells in mice persistently expressing p55TNFR, however these cells displayed a


state of activation equivalent to WT control mice. Moreover, we found persistent p55TNFR expression associated with defective pulmonary IFNγ synthesis correlating with a decrease in CD4+ T


cell activation despite normal pulmonary CD4+ T cell recruitment. Collectively, our findings suggest that persistent p55TNFR expression induces hypo-immune response during _M. tuberculosis_.


This is in line with the fact that I T cell hybridoma clones exposed to chronic TNF stimulation down regulated TCRζ and TCR/CD3 complex cell surface expression, leading to T cell


hyporesponsiveness reflected by a decrease in antigen-specific proliferation and suppression of cytokine responses66. The authors further demonstrated that the observed T cell


hyporesponsiveness was mediated via p55TNFR66. Persistent p55TNFR signaling also led to down modulation of T cell responses in a model of chronic inflammation involving TCR transgenic


mice67. Collectively the data supports a mechanism by which T cell activation is regulated in a p55TNFR dependent manner during chronic _M. tuberculosis_ infection. This is likely indirect,


as we showed recently that the p55TNFR pathway in T cells is dispensable for controlling _M. tuberculosis_ infection26. However, given our previous published data in which we demonstrated an


increased p75TNFR shedding in the absence of p55TNFR resulting in reduced bioactive TNF16, we hypothesized that a similar compensationary increase in p75TNFR in the non-sheddable p55TNFR


mouse strain and subsequent reduced bioactive TNF levels may be the underlying mechanism driving the observed T cell hyporesponsiveness in the p55∆NS mice. Indeed an analysis of soluble


p75TNFR both _in vitro_ in bone marrow derived macrophages as well as _in vivo_ in BAL fluid samples, demonstrated an increased level of soluble p75TNFR in p55∆NS mice compared to WT mice


which we confirmed _in vivo_ to induce impaired bioactive TNF concentrations. We have also previously shown that bioactive TNF signaling through p55TNFR in dendritic cells plays an important


role in IL-12 dependent DC migration to the draining lymph node and subsequent _M. tuberculosis-_specific T cell activation16. It is therefore possible that disruption of p55TNFR shedding


leads to a compensationary increase in soluble p75TNFR which reduces bioavailable TNF and impairs dendritic cell migration, all of which results in defective Th1 responses. Our hypothesis of


a potential defect in T cell responses during persistent p55TNFR expression was indeed validated when p55∆NS mice displayed enhanced sensitivity to tuberculosis reactivation. The mechanism


associated with defective T cell responses during chronic tuberculosis is unknown. Given the similar frequencies of T cell recruitment, a reduction in T cell proliferation is unlikely to be


the cause of defective T cell responses. This opens the possibility to a potential role for regulatory T cells (Tregs) in the suppressed CD4+CD44+ T cell activation during persistent p55TNFR


expression. Previous studies have indicated that neutralization of TNF during rheumatoid arthritis treatment elicited a significant population of Tregs. And the ablation of p55TNF in


reactive arthritis mice resulted in a reduced Tregs number and activity indicate that TNF signaling through p55TNFR and p75TNFR is important to the regulation of Treg cell function68,69.


Another possible mechanism associated with defective T cell responses might be that sustained p55TNFR signaling induces T cell exhaustion. Recently, Beyer _et al_., reported reversal of T


cell exhaustion through TNF neutralization which reconstituted specific immunity against chronic LCMV infection70. Interestingly, selective TNF receptor expression on T cells was shown to be


responsible for T cell exhaustion. In conclusion, our results demonstrate that while sustained p55TNFR expression mediated normal acute control of acute _M. tuberculosis_ infection, it was


associated with suboptimal T cell responses during chronic infection and promoted tuberculosis reactivation. METHODS MICE C57BL/6 wild type (WT) and p55∆NS mice34 were maintained and housed


in individually ventilated cages under specific pathogen free conditions in the Research Animal Facility at the University of Cape Town (South Africa). For all the experiments, age matched


mice were used. ETHICS STATEMENT All experiments and protocols were approved and performed in accordance with the guidelines of the Research Ethics Committee of the University of Cape Town,


South Africa and complied with South African regulations as stipulated in _The South African National Standard 10386-The care and use of animals for scientific purposes_. MYCOBACTERIA AND


INFECTION _M. tuberculosis_ H37Rv was grown in Middlebrook 7H9 broth (Becton, Dickinson and Company, Le Pont de Claix, France) supplemented with 10% Middlebrook OADC enrichment medium (Life


Technologies, Gaitherburg, MD), 0.5% glycerol and 0.05% Tween 80 at 37 °C until log phase. H37Rv-GFP (kindly provided by Joel Ernst, University of San Fransciso, California) was grown


similarly with 25 mg/L kanamycin in the broth. Prior to use, mycobacterial aliquots were passed 30× through a 29.5G needle to minimize bacterial clumping. Pulmonary infection at a dose of


50–100 live _M. tuberculosis_ H37Rv bacilli was performed using a Glas-Col Inhalation Exposure System, Model A4224. Inoculum dosage was confirmed 24 h post-infection by determining the


bacilli burden in the lungs of infected mice. To establish a latent infection, mice were treated with 25 mg/Kg INH-RIF for 6 weeks commencing at day 28 post- infection, which resulted in


undetectable levels of bacilli. For disease reactivation, chemotherapy was withdrawn and bacilli burdens determined at day 90, 180 and 300 days post infection. For colony enumeration organs


were weighed and homogenized in 0.04% Tween 80/PBS. Tenfold serial dilutions of organ homogenates were plated in duplicates on Middlebrook 7H10 (Becton, Dickinson and Company) agar plates


containing 10% OADC (Life Technologies, Gaitherburg, MD) and incubated at 37 °C for 19–21 days. LUNG MORPHOLOGY Lung weights were measured for use as a surrogate marker of inflammation, and


thereafter fixed in 10% formalin and paraffin-embedded. Tissues were sectioned at 2–3 μm and stained with haematoxylin and eosin (H&E), then analysed using a Nikon 90i Eclipse


microscope. The percentage of free alveolar space was calculated based on the area measurement using the microscope imaging software NIS-Elements, Nikon to analyze the open space in the


whole lung section. CYTOKINE ELISA Whole lungs were homogenized in 1 ml 0.04% Tween 80 saline containing protease inhibitor (Sigma) and the supernatants, aliquoted and frozen at −80 °C.


Cytokine concentrations were measured in organ homogenates or supernatants from cultured cells using commercially available ELISA reagents for TNF, p55TNFR, p75TNFR, IFNγ, and IL-12p70,


(R&D Systems, Germany and BD Pharmingen, San Diego), according to the manufacturer’s instructions. LUNG SINGLE CELL PREPARATION AND FLOW CYTOMETRY Lungs from infected mice were perfused


by injecting 5 ml cold PBS containing 20 U/ml heparin (Bodene (PTY) Limited, RSA) in the right ventricle of the heart. Lungs were removed, sectioned and incubated in PBS containing 50 U/ml


collagenase 1 (Worthington Biomedical Coorporation, Lakewood, NJ) and 13 μg/ml DNAse 1 (Boeringer-Mannheim, Germany) at 37 °C for 90 min with rotation. For single cell suspension, lung


tissue was passed through a 70 μm nylon cell strainer (Beckton and Dickinson), washed 2× with PBS and viable cell numbers were determined by counting in the presence of trypan blue. Isolated


lung cells from infected mice were stained with the following antibodies: anti-CD3 BV421 (145-2C11), anti-CD4-FITC (H129.19), anti-CD4 Alexo-fluor 700 (RM4-5), anti-CD44-PE (IM7),


anti-CD86-PE (GL1), anti-CD80-PE (16-10A1), anti-I-A/I-E-PE (M5/14.15.2), anti-CD11b-FITC. All the antibodies were purchased from Pharmingen and used at 2 μg/ml/106 cells. Cells were fixed


in 4% paraformaldehyde and were acquired using a FACS Calibur or BD LSR Fortessa (Beckton and Dickinson) and analyzed using FlowJo 7.5 software (Tree star, Ashland, OR, USA). PERITONEAL


MACROPHAGES Mice were injected with 3% thioglycollate (Difco, St. Louis, USA). Five days later, peritoneal exudate cells were isolated from the peritoneal cavity by washing with ice-cold


RPMI (Sigma, Germany) supplemented with 10% FCS (Gibco, Invitrogen Corporation, Germany). Cells were cultured overnight at 37 °C and 5% CO2 incubator. Adherent monolayer cells were used as


peritoneal macrophages and were cultured at 5 × 105 cells/ml in RPMI supplemented with 10% FCS and stimulated for 20 min with either 100 ng/ml LPS (_E. coli_, serotype O111:B4, Sigma) or _M.


tuberculosis_ H37Rv (MOI 2:1). PRIMARY MACROPHAGE CULTURES Bone marrow cells were isolated from femurs of 6 to 8 weeks old naive mice and were cultivated on 90 mm Sterilin plates (Bibby


Sterilin, UK) at 2 × 106 cells/ml for 7 days in RPMI (Sigma, Germany) supplemented with 2 mM L-glutamine (Gibco, Invitrogen Corporation, Germany), 0.2 μM 2-ME (Sigma, St. Louis, USA), 20%


horse serum (Gibco, Invitrogen Corporation, Germany) and 30% L929 cell-conditioned medium at 37 °C and 5% CO2. Confluent cells were harvested and seeded at 5 × 105 cells/ml in 96 well tissue


culture plates (Nunclon, Denmark) and incubated for 24 h at 37 °C and 5% CO2 to allow for cell adherence. Cells were then stimulated with either 100 ng/ml LPS (_E. coli_, serotype O111:B4,


Sigma) or live _M. tuberculosis_ H37Rv bacteria (MOI 2:1) for 90 min, 150 min, 300 min, 24 h, and 48 h. Supernatants were collected and analysed for cytokine content using ELISA and nitrite


concentration using Griess reagent (3% phosphoric acid, 1% p-aminobenzene-sulphonamide, 1% n-naphthylenediamide) as described71. To determine bactericidal capacity to kill _M. tuberculosis_


bacilli, BMDMs were infected with H37Rv-GFP at 2:1 MOI for 4 to 120 hours. The amount of intracellular bacilli was determined by lysing the macrophages and measuring the relative fluorescent


unit (RFU) using Modulus microplate fluorometer (Turner Biosystems Inc.). BAL COLLECTION Mice were euthanized to reveal the trachea. BAL fluid was collected by 2 lavages in which an


18-gauge cannula was inserted into the trachea and aspirated 10 times with 300 μl sterile PBS. The BAL fluid was aliquoted and stored at −80 °C for cytokine analysis. BAL cells were


collected by 3 lavages with 700 μl sterile PBS each. Cells were kept on ice for flow cytometric analysis. BIOACTIVE TNF ASSAY Bioactive TNF was determined as previously described16, using


TNF sensitive fibroblast cell line WEHI 164, clone 13 (Walter and Eliza Hall Institute). WEHI cells were cultured in RPMI (Sigma, Germany) containing 10% FCS (Gibco, Invitrogen Corporation,


Germany), 10 U/ml penicillin (Gibco, Germany), 10 μg/ml streptomycin (Gibco, Invitrogen Corporation, Germany) and 0.5× amino acid supplement (MEM Amino acids without L-glutamine, Gibco,


Germany) at 37 °C with 5% CO2. Confluent cells were reseeded at 2 × 105 cells/ml in 96 well tissue culture plates (Nunclon, Denmark). TNF standards (recombinant mouse TNF, BD PharMingen, San


Diego) in 2 fold serial dilutions or samples were added to WEHI cells and incubated for 18 h at 37 °C with 5% CO2. MTT solution (2 mg/ml, Sigma, Germany) was then added and incubated for a


further 2 h, after which, the supernatants were aspirated and 50 μl DMSO was added and samples were read at 570 nm using VERSAmax Tunable Microplate Reader (Molecular devices Coorporation,


California, USA). Data was analyzed using SoftMax Pro (Molecular devices Coorporation, California, USA). STATISTICAL ANALYSIS Statistical analysis was performed using Graphpad Prism 6 by


ANOVA. For mortality studies, analysis was performed using the logrank test. For all tests, a p value of <0.05 was considered significant. ADDITIONAL INFORMATION HOW TO CITE THIS ARTICLE:


Dambuza, I. M. _et al_. Persistent p55TNFR expression impairs T cell responses during chronic tuberculosis and promotes reactivation. _Sci. Rep._ 6, 39499; doi: 10.1038/srep39499 (2016).


PUBLISHER'S NOTE: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. REFERENCES * Bodmer, J. L., Schneider, P. &


Tschopp, J. The molecular architecture of the TNF superfamily. Trends Biochem Sci 27, 19–26 (2002). CAS  PubMed  Google Scholar  * Ware, C. F. The TNF superfamily. Cytokine Growth Factor Rev


14, 181–184 (2003). CAS  PubMed  Google Scholar  * Black, R. A. et al. A metalloproteinase disintegrin that releases tumour-necrosis factor-alpha from cells. Nature 385, 729–733, doi:


10.1038/385729a0 (1997). Article  ADS  CAS  PubMed  Google Scholar  * Kriegler, M., Perez, C., DeFay, K., Albert, I. & Lu, S. D. A novel form of TNF/cachectin is a cell surface cytotoxic


transmembrane protein: ramifications for the complex physiology of TNF. Cell 53, 45–53 (1988). CAS  PubMed  Google Scholar  * Mueller, C. et al. Noncleavable transmembrane mouse tumor


necrosis factor-alpha (TNFalpha) mediates effects distinct from those of wild-type TNFalpha _in vitro_ and _in vivo_. J Biol Chem 274, 38112–38118 (1999). CAS  PubMed  Google Scholar  *


Perez, C. et al. A nonsecretable cell surface mutant of tumor necrosis factor (TNF) kills by cell-to-cell contact. Cell 63, 251–258 (1990). CAS  PubMed  Google Scholar  * Grell, M., Wajant,


H., Zimmermann, G. & Scheurich, P. The type 1 receptor (CD120a) is the high-affinity receptor for soluble tumor necrosis factor. Proc Natl Acad Sci USA 95, 570–575 (1998). ADS  CAS 


PubMed  PubMed Central  Google Scholar  * Mohan, M. J. et al. The tumor necrosis factor-alpha converting enzyme (TACE): a unique metalloproteinase with highly defined substrate selectivity.


Biochemistry 41, 9462–9469 (2002). CAS  PubMed  Google Scholar  * Horiuchi, K. et al. Cutting edge: TNF-alpha-converting enzyme (TACE/ADAM17) inactivation in mouse myeloid cells prevents


lethality from endotoxin shock. J Immunol 179, 2686–2689 (2007). CAS  PubMed  Google Scholar  * Aderka, D. The potential biological and clinical significance of the soluble tumor necrosis


factor receptors. Cytokine Growth Factor Rev 7, 231–240 (1996). CAS  PubMed  Google Scholar  * Keuter, M. et al. Patterns of proinflammatory cytokines and inhibitors during typhoid fever. J


Infect Dis 169, 1306–1311 (1994). CAS  PubMed  Google Scholar  * Kern, P. et al. Soluble tumor necrosis factor receptors correlate with parasitemia and disease severity in human malaria. J


Infect Dis 166, 930–934 (1992). CAS  PubMed  Google Scholar  * Kern, W. V., Engel, A., Schieffer, S., Prummer, O. & Kern, P. Circulating tumor necrosis factor alpha (TNF), soluble TNF


receptors, and interleukin-6 in human subacute bacterial endocarditis. Infect Immun 61, 5413–5416 (1993). CAS  PubMed  PubMed Central  Google Scholar  * Zijlstra, E. E., van der Poll, T.


& Mevissen, M. Soluble receptors for tumor necrosis factor as markers of disease activity in visceral leishmaniasis. J Infect Dis 171, 498–501 (1995). CAS  PubMed  Google Scholar  *


Balcewicz-Sablinska, M. K., Keane, J., Kornfeld, H. & Remold, H. G. Pathogenic Mycobacterium tuberculosis evades apoptosis of host macrophages by release of TNF-R2, resulting in


inactivation of TNF-alpha. J Immunol 161, 2636–2641 (1998). CAS  PubMed  Google Scholar  * Keeton, R. et al. Soluble TNFRp75 regulates host protective immunity against Mycobacterium


tuberculosis. J Clin Invest 124, 1537–1551, doi: 10.1172/JCI45005 (2014). Article  CAS  PubMed  PubMed Central  Google Scholar  * Olleros, M. L. et al. Membrane-bound TNF induces protective


immune responses to M. bovis BCG infection: regulation of memTNF and TNF receptors comparing two memTNF molecules. PLoS One 7, e31469, doi: 10.1371/journal.pone.0031469 (2012). Article  ADS


  CAS  PubMed  PubMed Central  Google Scholar  * Juffermans, N. P. et al. Tumor necrosis factor and interleukin-1 inhibitors as markers of disease activity of tuberculosis. Am J Respir Crit


Care Med 157, 1328–1331, doi: 10.1164/ajrccm.157.4.9709126 (1998). Article  CAS  PubMed  Google Scholar  * Brahmbhatt, S. et al. Immune markers measured before treatment predict outcome of


intensive phase tuberculosis therapy. Clin Exp Immunol 146, 243–252, doi: 10.1111/j.1365-2249.2006.03211.x (2006). Article  CAS  PubMed  PubMed Central  Google Scholar  * Crowe, P. D. et al.


A lymphotoxin-beta-specific receptor. Science 264, 707–710 (1994). ADS  CAS  PubMed  Google Scholar  * Bean, A. G. et al. Structural deficiencies in granuloma formation in TNF gene-targeted


mice underlie the heightened susceptibility to aerosol Mycobacterium tuberculosis infection, which is not compensated for by lymphotoxin. J Immunol 162, 3504–3511 (1999). CAS  PubMed 


Google Scholar  * Roach, D. R. et al. TNF regulates chemokine induction essential for cell recruitment, granuloma formation, and clearance of mycobacterial infection. J Immunol 168,


4620–4627 (2002). CAS  PubMed  Google Scholar  * Allie, N. et al. Prominent role for T cell-derived tumour necrosis factor for sustained control of Mycobacterium tuberculosis infection. Sci


Rep 3, 1809, doi: 10.1038/srep01809 (2013). Article  CAS  PubMed  PubMed Central  Google Scholar  * Dambuza, I. et al. Efficacy of membrane TNF mediated host resistance is dependent on


mycobacterial virulence. Tuberculosis (Edinb) 88, 221–234, doi: 10.1016/j.tube.2007.08.011 (2008). Article  CAS  Google Scholar  * Flynn, J. L. et al. Tumor necrosis factor-alpha is required


in the protective immune response against Mycobacterium tuberculosis in mice. Immunity 2, 561–572 (1995). CAS  PubMed  Google Scholar  * Segueni, N. et al. Innate myeloid cell TNFR1


mediates first line defence against primary Mycobacterium tuberculosis infection. Sci Rep 6, 22454, doi: 10.1038/srep22454 (2016). Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 


* Eriks, I. S. & Emerson, C. L. Temporal effect of tumor necrosis factor alpha on murine macrophages infected with Mycobacterium avium. Infect Immun 65, 2100–2106 (1997). CAS  PubMed 


PubMed Central  Google Scholar  * Kindler, V., Sappino, A. P., Grau, G. E., Piguet, P. F. & Vassalli, P. The inducing role of tumor necrosis factor in the development of bactericidal


granulomas during BCG infection. Cell 56, 731–740 (1989). CAS  PubMed  Google Scholar  * Jacobs, M. et al. Correction of defective host response to Mycobacterium bovis BCG infection in


TNF-deficient mice by bone marrow transplantation. Lab Invest 80, 901–914 (2000). CAS  PubMed  Google Scholar  * Tsao, T. C. et al. Imbalances between tumor necrosis factor-alpha and its


soluble receptor forms, and interleukin-1beta and interleukin-1 receptor antagonist in BAL fluid of cavitary pulmonary tuberculosis. Chest 117, 103–109 (2000). CAS  PubMed  Google Scholar  *


Tsao, T. C., Li, L., Hsieh, M., Liao, S. & Chang, K. S. Soluble TNF-alpha receptor and IL-1 receptor antagonist elevation in BAL in active pulmonary TB. Eur Respir J 14, 490–495 (1999).


CAS  PubMed  Google Scholar  * Corti, A. et al. Upregulation of p75 tumor necrosis factor alpha receptor in Mycobacterium avium-infected mice: evidence for a functional role. Infect Immun


67, 5762–5767 (1999). CAS  PubMed  PubMed Central  Google Scholar  * Navarra, S. V. et al. Risk of tuberculosis with anti-tumor necrosis factor-alpha therapy: substantially higher number of


patients at risk in Asia. Int J Rheum Dis 17, 291–298, doi: 10.1111/1756-185X.12188 (2014). Article  CAS  PubMed  Google Scholar  * Xanthoulea, S. et al. Tumor necrosis factor (TNF) receptor


shedding controls thresholds of innate immune activation that balance opposing TNF functions in infectious and inflammatory diseases. J Exp Med 200, 367–376, doi: 10.1084/jem.20040435


(2004). Article  CAS  PubMed  PubMed Central  Google Scholar  * Engelmann, H., Aderka, D., Rubinstein, M., Rotman, D. & Wallach, D. A tumor necrosis factor-binding protein purified to


homogeneity from human urine protects cells from tumor necrosis factor toxicity. J Biol Chem 264, 11974–11980 (1989). CAS  PubMed  Google Scholar  * Engelmann, H. et al. Antibodies to a


soluble form of a tumor necrosis factor (TNF) receptor have TNF-like activity. J Biol Chem 265, 14497–14504 (1990). CAS  PubMed  Google Scholar  * Engelmann, H., Novick, D. & Wallach, D.


Two tumor necrosis factor-binding proteins purified from human urine. Evidence for immunological cross-reactivity with cell surface tumor necrosis factor receptors. J Biol Chem 265,


1531–1536 (1990). CAS  PubMed  Google Scholar  * Olsson, I. et al. Isolation and characterization of a tumor necrosis factor binding protein from urine. Eur J Haematol 42, 270–275 (1989).


CAS  PubMed  Google Scholar  * Ding, A. H., Nathan, C. F. & Stuehr, D. J. Release of reactive nitrogen intermediates and reactive oxygen intermediates from mouse peritoneal macrophages.


Comparison of activating cytokines and evidence for independent production. J Immunol 141, 2407–2412 (1988). CAS  PubMed  Google Scholar  * Flesch, I. E. & Kaufmann, S. H. Activation of


tuberculostatic macrophage functions by gamma interferon, interleukin-4, and tumor necrosis factor. Infect Immun 58, 2675–2677 (1990). CAS  PubMed  PubMed Central  Google Scholar  * Peschon,


J. J. et al. TNF receptor-deficient mice reveal divergent roles for p55 and p75 in several models of inflammation. J Immunol 160, 943–952 (1998). CAS  PubMed  Google Scholar  * Bekker, L.


G., Freeman, S., Murray, P. J., Ryffel, B. & Kaplan, G. TNF-alpha controls intracellular mycobacterial growth by both inducible nitric oxide synthase-dependent and inducible nitric oxide


synthase-independent pathways. J Immunol 166, 6728–6734 (2001). CAS  PubMed  Google Scholar  * Jesus, A. A. et al. TNF receptor-associated periodic syndrome (TRAPS): description of a novel


TNFRSF1A mutation and response to etanercept. Eur J Pediatr 167, 1421–1425, doi: 10.1007/s00431-008-0685-2 (2008). Article  PubMed  Google Scholar  * McDermott, M. F. et al. Germline


mutations in the extracellular domains of the 55 kDa TNF receptor, TNFR1, define a family of dominantly inherited autoinflammatory syndromes. Cell 97, 133–144 (1999). CAS  PubMed  Google


Scholar  * Cooper, A. M. et al. Disseminated tuberculosis in interferon gamma gene-disrupted mice. J Exp Med 178, 2243–2247 (1993). CAS  PubMed  Google Scholar  * Cooper, A. M., Magram, J.,


Ferrante, J. & Orme, I. M. Interleukin 12 (IL-12) is crucial to the development of protective immunity in mice intravenously infected with mycobacterium tuberculosis. J Exp Med 186,


39–45 (1997). CAS  PubMed  PubMed Central  Google Scholar  * Feng, C. G. et al. Maintenance of pulmonary Th1 effector function in chronic tuberculosis requires persistent IL-12 production. J


Immunol 174, 4185–4192 (2005). CAS  PubMed  Google Scholar  * Flynn, J. L. et al. An essential role for interferon gamma in resistance to Mycobacterium tuberculosis infection. J Exp Med


178, 2249–2254 (1993). CAS  PubMed  Google Scholar  * Frankova, D. & Zidek, Z. IFN-gamma-induced TNF-alpha is a prerequisite for _in vitro_ production of nitric oxide generated in murine


peritoneal macrophages by IFN-gamma. Eur J Immunol 28, 838–843 (1998). CAS  PubMed  Google Scholar  * Blass, S. L., Pure, E. & Hunter, C. A. A role for CD44 in the production of


IFN-gamma and immunopathology during infection with Toxoplasma gondii. J Immunol 166, 5726–5732 (2001). CAS  PubMed  Google Scholar  * DeGrendele, H. C., Estess, P. & Siegelman, M. H.


Requirement for CD44 in activated T cell extravasation into an inflammatory site. Science 278, 672–675 (1997). ADS  CAS  PubMed  Google Scholar  * Graham, V. A., Marzo, A. L. & Tough, D.


F. A role for CD44 in T cell development and function during direct competition between CD44+ and CD44− cells. Eur J Immunol 37, 925–934, doi: 10.1002/eji.200635882 (2007). Article  CAS 


PubMed  Google Scholar  * Geddes, K. et al. Identification of an innate T helper type 17 response to intestinal bacterial pathogens. Nat Med 17, 837–844, doi: 10.1038/nm.2391 (2011). Article


  CAS  PubMed  Google Scholar  * Vandanmagsar, B. et al. The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat Med 17, 179–188, doi: 10.1038/nm.2279


(2011). Article  CAS  PubMed  PubMed Central  Google Scholar  * Aggarwal, B. B. et al. Human tumor necrosis factor. Production, purification, and characterization. J Biol Chem 260, 2345–2354


(1985). CAS  PubMed  Google Scholar  * Smith, R. A. & Baglioni, C. Multimeric structure of the tumor necrosis factor receptor of HeLa cells. J Biol Chem 264, 14646–14652 (1989). CAS 


PubMed  Google Scholar  * Tsujimoto, M., Yip, Y. K. & Vilcek, J. Tumor necrosis factor: specific binding and internalization in sensitive and resistant cells. Proc Natl Acad Sci USA 82,


7626–7630 (1985). ADS  CAS  PubMed  PubMed Central  Google Scholar  * Lantz, M., Malik, S., Slevin, M. L. & Olsson, I. Infusion of tumor necrosis factor (TNF) causes an increase in


circulating TNF-binding protein in humans. Cytokine 2, 402–406 (1990). CAS  PubMed  Google Scholar  * Leeuwenberg, J. F., Dentener, M. A. & Buurman, W. A. Lipopolysaccharide LPS-mediated


soluble TNF receptor release and TNF receptor expression by monocytes. Role of CD14, LPS binding protein, and bactericidal/permeability-increasing protein. J Immunol 152, 5070–5076 (1994).


CAS  PubMed  Google Scholar  * Porteu, F. & Nathan, C. Shedding of tumor necrosis factor receptors by activated human neutrophils. J Exp Med 172, 599–607 (1990). CAS  PubMed  Google


Scholar  * Kaneko, H. et al. Role of tumor necrosis factor-alpha in Mycobacterium-induced granuloma formation in tumor necrosis factor-alpha-deficient mice. Lab Invest 79, 379–386 (1999).


CAS  PubMed  Google Scholar  * Pfeffer, K. et al. Mice deficient for the 55 kd tumor necrosis factor receptor are resistant to endotoxic shock, yet succumb to L. monocytogenes infection.


Cell 73, 457–467 (1993). CAS  PubMed  Google Scholar  * Rothe, J. et al. Mice lacking the tumour necrosis factor receptor 1 are resistant to TNF-mediated toxicity but highly susceptible to


infection by Listeria monocytogenes. Nature 364, 798–802, doi: 10.1038/364798a0 (1993). Article  ADS  CAS  PubMed  Google Scholar  * Torres, D. et al. Membrane tumor necrosis factor confers


partial protection to Listeria infection. Am J Pathol 167, 1677–1687, doi: 10.1016/S0002-9440(10)61250-3 (2005). Article  CAS  PubMed  PubMed Central  Google Scholar  * Fremond, C. et al.


Membrane TNF confers protection to acute mycobacterial infection. Respir Res 6, 136, doi: 10.1186/1465-9921-6-136 (2005). Article  CAS  PubMed  PubMed Central  Google Scholar  * Isomaki, P.


et al. Prolonged exposure of T cells to TNF down-regulates TCR zeta and expression of the TCR/CD3 complex at the cell surface. J Immunol 166, 5495–5507 (2001). CAS  PubMed  Google Scholar  *


Cope, A. P. et al. Chronic tumor necrosis factor alters T cell responses by attenuating T cell receptor signaling. J Exp Med 185, 1573–1584 (1997). CAS  PubMed  PubMed Central  Google


Scholar  * Biton, J., Boissier, M. C. & Bessis, N. TNFalpha: activator or inhibitor of regulatory T cells? Joint Bone Spine 79, 119–123, doi: 10.1016/j.jbspin.2011.09.017 (2012). Article


  CAS  PubMed  Google Scholar  * Cargnelutti, E., Arias, J. L., Valdez, S. R., Rabinovich, G. A. & Di Genaro, M. S. TNFRp55 controls regulatory T cell responses in Yersinia-induced


reactive arthritis. Immunol Cell Biol 91, 159–166, doi: 10.1038/icb.2012.65 (2013). Article  CAS  PubMed  Google Scholar  * Beyer, M. et al. Tumor-necrosis factor impairs CD4(+) T


cell-mediated immunological control in chronic viral infection. Nat Immunol 17, 593–603, doi: 10.1038/ni.3399 (2016). Article  CAS  PubMed  Google Scholar  * Stuehr, D. J. & Nathan, C.


F. Nitric oxide. A macrophage product responsible for cytostasis and respiratory inhibition in tumor target cells. J Exp Med 169, 1543–1555 (1989). CAS  PubMed  Google Scholar  Download


references ACKNOWLEDGEMENTS We thank Lizette Fick for her contribution to histopathology. We thank Faried Abbass for technical support. We thank the support staff of the Division of


Immunology and the Research Animal Facility at the University of Cape Town for their contribution to animal care and technical support. The study was supported by the University of Cape


Town, National Research Foundation (South Africa), South African Medical Research Council (SAMRC) National Health Laboratory Service (South Africa), The European Union (contract number:


028190), FP6 NEST project N°028190 “TB REACT”. Research carried out within the scope of the Franco/South African Laboratory “TB Immunity” (Associated International Laboratory ‘AIL’). AUTHOR


INFORMATION Author notes * Present address: Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK. * Present address: MRC Centre for Tuberculosis


Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, University of Stellenbosch, South Africa. * Dambuza Ivy M. and Keeton Roanne contributed


equally to this work. AUTHORS AND AFFILIATIONS * Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences,


University of Cape Town, South Africa Ivy M. Dambuza, Roanne Keeton, Nai-Jen Hsu, Nasiema Allie & Muazzam Jacobs * CNRS UMR7355, Experimental and Molecular Immunology and Neurogenetics,


Orleans, 45071, France Valérie F. J. Quesniaux & Bernhard Ryffel * National Health Laboratory Service, South Africa Muazzam Jacobs * South African Medical Research Council,, South Africa


Muazzam Jacobs Authors * Ivy M. Dambuza View author publications You can also search for this author inPubMed Google Scholar * Roanne Keeton View author publications You can also search for


this author inPubMed Google Scholar * Nai-Jen Hsu View author publications You can also search for this author inPubMed Google Scholar * Nasiema Allie View author publications You can also


search for this author inPubMed Google Scholar * Valérie F. J. Quesniaux View author publications You can also search for this author inPubMed Google Scholar * Bernhard Ryffel View author


publications You can also search for this author inPubMed Google Scholar * Muazzam Jacobs View author publications You can also search for this author inPubMed Google Scholar CONTRIBUTIONS


I.M.D., R.K. and N.J.H. performed the experiments and subsequent analysis. N.A. performed the aerosol infection. V.Q. and B.R. contributed to the experimental design. M.J. oversaw the


project. I.M.D., R.K., N.J.H. and M.J. prepared the figures and wrote the manuscript. All authors read and approved the final manuscript. ETHICS DECLARATIONS COMPETING INTERESTS The authors


declare no competing financial interests. ELECTRONIC SUPPLEMENTARY MATERIAL SUPPLEMENTARY INFORMATION RIGHTS AND PERMISSIONS This work is licensed under a Creative Commons Attribution 4.0


International License. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in the credit line; if the


material is not included under the Creative Commons license, users will need to obtain permission from the license holder to reproduce the material. To view a copy of this license, visit


http://creativecommons.org/licenses/by/4.0/ Reprints and permissions ABOUT THIS ARTICLE CITE THIS ARTICLE Dambuza, I., Keeton, R., Hsu, NJ. _et al._ Persistent p55TNFR expression impairs T


cell responses during chronic tuberculosis and promotes reactivation. _Sci Rep_ 6, 39499 (2016). https://doi.org/10.1038/srep39499 Download citation * Received: 06 July 2016 * Accepted: 23


November 2016 * Published: 20 December 2016 * DOI: https://doi.org/10.1038/srep39499 SHARE THIS ARTICLE Anyone you share the following link with will be able to read this content: Get


shareable link Sorry, a shareable link is not currently available for this article. Copy to clipboard Provided by the Springer Nature SharedIt content-sharing initiative


Trending News

Jalisco officials say this indigenous leader died in a car crash. His community isn't buying it

The Indigenous Wixárika community in northern Jalisco is raising doubts about the official explanation that a vehicle ac...

High stakes get build-to-rent star claire solon going at greystar

A business trip to London about 10 years ago gave Claire Solon a glimpse of a future that she believes will fail. At the...

Targeted modulation of immune cells and tissues using engineered biomaterials

ABSTRACT Therapies modulating the immune system offer the prospect of treating a wide range of conditions including infe...

At an average of 1,200 passengers per day, maya train numbers far from eventual target

Will the Maya Train railroad eventually become a popular mode of travel between beach resorts, colonial cities and archa...

What we’ve learnt from building africa’s biggest genome library

The human genome was first sequenced in 2003 by multiple research centres across the world. The breakthrough was hailed ...

Latests News

Persistent p55tnfr expression impairs t cell responses during chronic tuberculosis and promotes reactivation

ABSTRACT The pleiotropic activities of TNF are mediated by two structurally related but functionally distinct type I tra...

Statsguru | Searchable Cricket Statistics database | ESPNcricinfo.com

ESPN CricinfoLive ScoresLive Scores HomeScheduleResultsMonth viewSeason viewInternational calendarDesktop ScoreboardSeri...

Meghan markle ‘empowered by divorce’ after abrupt break-up decision

MEGHAN MARKLE ‘MAILED RINGS BACK TO FIRST HUSBAND’ SAYS BROTHER The Duchess of Sussex was in a relationship with the bra...

Premier league verdict as var decision sees liverpool wrap up another victory

The Premier League have confirmed why Liverpool were awarded a second penalty in their 3-1 win against Southampton. The ...

High-energy cosmic rays traced to source

Supermassive black-hole galaxies spin out super-energetic particles. Access through your institution Buy or subscribe Th...

Top