Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Neuroinflammation ; 12: 125, 2015 Jun 26.
Article in English | MEDLINE | ID: mdl-26112704

ABSTRACT

BACKGROUND: Tuberculosis (TB) affects one third of the global population, and TB of the central nervous system (CNS-TB) is the most severe form of tuberculosis which often associates with high mortality. The pro-inflammatory cytokine tumour necrosis factor (TNF) plays a critical role in the initial and long-term host immune protection against Mycobacterium tuberculosis (M. tuberculosis) which involves the activation of innate immune cells and structure maintenance of granulomas. However, the contribution of TNF, in particular neuron-derived TNF, in the control of cerebral M. tuberculosis infection and its protective immune responses in the CNS were not clear. METHODS: We generated neuron-specific TNF-deficient (NsTNF(-/-)) mice and compared outcomes of disease against TNF(f/f) control and global TNF(-/-) mice. Mycobacterial burden in brains, lungs and spleens were compared, and cerebral pathology and cellular contributions analysed by microscopy and flow cytometry after M. tuberculosis infection. Activation of innate immune cells was measured by flow cytometry and cell function assessed by cytokine and chemokine quantification using enzyme-linked immunosorbent assay (ELISA). RESULTS: Intracerebral M. tuberculosis infection of TNF(-/-) mice rendered animals highly susceptible, accompanied by uncontrolled bacilli replication and eventual mortality. In contrast, NsTNF(-/-) mice were resistant to infection and presented with a phenotype similar to that in TNF(f/f) control mice. Impaired immunity in TNF(-/-) mice was associated with altered cytokine and chemokine synthesis in the brain and characterised by a reduced number of activated innate immune cells. Brain pathology reflected enhanced inflammation dominated by neutrophil influx. CONCLUSION: Our data show that neuron-derived TNF has a limited role in immune responses, but overall TNF production is necessary for protective immunity against CNS-TB.


Subject(s)
Host-Pathogen Interactions/physiology , Immunity, Innate/physiology , Mycobacterium tuberculosis/physiology , Neurons/microbiology , Neurons/pathology , Tuberculosis, Central Nervous System/immunology , Tumor Necrosis Factor-alpha/physiology , Virus Replication/physiology , Animals , Brain/metabolism , Brain/microbiology , Brain/pathology , Cell Proliferation/physiology , Chemokines/metabolism , Cytokines/metabolism , Dendritic Cells/microbiology , Dendritic Cells/pathology , Disease Models, Animal , Disease Resistance/immunology , Host-Pathogen Interactions/immunology , Immunity, Innate/immunology , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Knockout , Microglia/microbiology , Microglia/pathology , Tuberculosis, Central Nervous System/pathology , Tuberculosis, Central Nervous System/physiopathology , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics
2.
Infect Immun ; 82(5): 1880-90, 2014 May.
Article in English | MEDLINE | ID: mdl-24566619

ABSTRACT

Mycobacterium tuberculosis infection of the central nervous system is thought to be initiated once the bacilli have breached the blood brain barrier and are phagocytosed, primarily by microglial cells. In this study, the interactions of M. tuberculosis with neurons in vitro and in vivo were investigated. The data obtained demonstrate that neurons can act as host cells for M. tuberculosis. M. tuberculosis bacilli were internalized by murine neuronal cultured cells in a time-dependent manner after exposure, with superior uptake by HT22 cells compared to Neuro-2a cells (17.7% versus 9.8%). Internalization of M. tuberculosis bacilli by human SK-N-SH cultured neurons suggested the clinical relevance of the findings. Moreover, primary murine hippocampus-derived neuronal cultures could similarly internalize M. tuberculosis. Internalized M. tuberculosis bacilli represented a productive infection with retention of bacterial viability and replicative potential, increasing 2- to 4-fold within 48 h. M. tuberculosis bacillus infection of neurons was confirmed in vivo in the brains of C57BL/6 mice after intracerebral challenge. This study, therefore, demonstrates neurons as potential new target cells for M. tuberculosis within the central nervous system.


Subject(s)
Mycobacterium tuberculosis/physiology , Neurons/microbiology , Tuberculosis, Central Nervous System/microbiology , Animals , Cell Line , Female , Humans , Mice , Mice, Inbred C57BL , Tuberculosis, Central Nervous System/immunology
3.
Infect Immun ; 81(4): 1234-44, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23381991

ABSTRACT

The contraceptive depot medroxyprogesterone acetate (DMPA), with progestin as the single active compound, possesses selective glucocorticoid activity and can alter the expression of glucocorticoid receptor-regulated genes. We therefore propose that pharmacological doses of DMPA used for endocrine therapy could have significant immune modulatory effects and impact on susceptibility to, as well as clinical manifestation and outcome of, infectious diseases. We investigated the effect of contraceptive doses of DMPA in two different murine Mycobacterium tuberculosis models. Multiplex bead array analysis revealed that DMPA altered serum cytokine levels of tumor necrosis factor alpha (TNF-α), granulocyte colony-stimulating factor (G-CSF), and interleukin 10 (IL-10) in C57BL/6 mice and gamma interferon (IFN-γ) in BALB/c mice. DMPA also suppressed antigen-specific production of TNF-α, G-CSF, IL-10, and IL-6 and induced the production of IP-10 in C57BL/6 mice. In BALB/c mice, DMPA altered the antigen-specific secretion of IFN-γ, IL-17, granulocyte-macrophage colony-stimulating factor (GM-CSF), IL-6, and monocyte chemotactic protein 1 (MCP-1). Furthermore, we show that C57BL/6 mice treated with doses of DMPA, which result in serum concentrations similar to those observed in contraceptive users, have a significantly higher bacterial load in their lungs. Our data show for the first time that DMPA impacts tuberculosis (TB) disease severity in a mouse model and that the effects of this contraceptive are not confined to infections of the genital tract. This could have major implications for the contraceptive policies not only in developing countries like South Africa but also worldwide.


Subject(s)
Contraceptive Agents/administration & dosage , Cytokines/blood , Immunologic Factors/administration & dosage , Immunosuppression Therapy , Medroxyprogesterone Acetate/administration & dosage , Mycobacterium tuberculosis/immunology , Tuberculosis/immunology , Animals , Contraceptive Agents/adverse effects , Cytokines/metabolism , Disease Models, Animal , Female , Immunologic Factors/adverse effects , Medroxyprogesterone Acetate/adverse effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
4.
J Immunol ; 185(7): 4292-301, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20817877

ABSTRACT

The contribution of lymphotoxin (LT)α in the host immune response to virulent Mycobacterium tuberculosis and Mycobacterium bovis bacillus Calmette-Guérin infections was investigated. Despite their ability to induce Th1 cytokine, IFN-γ, and IL-12 pulmonary response, "conventional" LTα(-/-) mice succumb rapidly to virulent M. tuberculosis aerosol infection, with uncontrolled bacilli growth, defective granuloma formation, necrosis, and reduced pulmonary inducible NO synthase expression, similar to TNF(-/-) mice. Contributions from developmental lymphoid abnormalities in LTα(-/-) mice were excluded because hematopoietic reconstitution with conventional LTα(-/-) bone marrow conferred enhanced susceptibility to wild-type mice, comparable to conventional LTα(-/-) control mice. However, conventional LTα(-/-) mice produced reduced levels of TNF after M. bovis bacillus Calmette-Guérin infection, and their lack of control of mycobacterial infection could be due to a defective contribution of either LTα or TNF, or both, to the host immune response. To address this point, the response of "neo-free" LTα(-/-) mice with unperturbed intrinsic TNF expression to M. tuberculosis infection was investigated in a direct comparative study with conventional LTα(-/-) mice. Strikingly, although conventional LTα(-/-) mice were highly sensitive, similar to TNF(-/-) mice, neo-free LTα(-/-) mice controlled acute M. tuberculosis infection essentially as wild-type mice. Pulmonary bacterial burden and inflammation was, however, slightly increased in neo-free LTα(-/-) mice 4-5 mo postinfection, but importantly, they did not succumb to infection. Our findings revise the notion that LTα might have a critical role in host defense to acute mycobacterial infection, independent of TNF, but suggest a contribution of LTα in the control of chronic M. tuberculosis infection.


Subject(s)
Lymphotoxin-alpha/immunology , Tuberculosis/immunology , Animals , Cytokines/biosynthesis , Cytokines/immunology , Enzyme-Linked Immunosorbent Assay , Lymphotoxin-alpha/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium bovis/immunology , Mycobacterium tuberculosis/immunology , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/immunology , Tuberculosis/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
5.
Heliyon ; 8(12): e12406, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36582695

ABSTRACT

The number and diversity of drugs in the tuberculosis (TB) drug development process has increased over the years, yet the attrition rate remains very high, signaling the need for continued research in drug discovery. In this study, crude secondary metabolites from marine fungi associated with ascidians collected from Saldanha and False Bays (South Africa) were investigated for antimycobacterial activity. Isolation of fungi was performed by sectioning thin inner-tissues of ascidians and spreading them over potato dextrose agar (PDA). Solid state fermentation of fungal isolates on PDA was then performed for 28 days to allow production of secondary metabolites. Afterwards, PDA cultures were dried and solid-liquid extraction using methanol was performed to extract fungal metabolites. Profiling of metabolites was performed using untargeted liquid chromatography quadrupole time-of-flight tandem mass spectrometry (LC-QTOF-MS/MS). The broth microdilution method was used to determine antimycobacterial activity against Mycobacterium smegmatis mc2155 and Mycobacterium tuberculosis H37Rv, while in silico flexible docking was performed on selected target proteins from M. tuberculosis. A total of 16 ascidians were sampled and 46 fungi were isolated. Only 32 fungal isolates were sequenced, and their sequences submitted to GenBank to obtain accession numbers. Metabolite profiling of 6 selected fungal extracts resulted in the identification of 65 metabolites. The most interesting extract was that of Clonostachys rogersoniana MGK33 which inhibited Mycobacterium smegmatis mc2155 and Mycobacterium tuberculosis H37Rv growth with minimum inhibitory concentrations (MICs) of 0.125 and 0.2 mg/mL, respectively. These results were in accordance with those from in silico molecular docking studies which showed that bionectin F produced by C. rogersoniana MGK33 is a potential inhibitor of M. tuberculosis ß-ketoacyl-acyl carrier protein reductase (MabA, PDB ID = 1UZN), with the docking score observed as -11.17 kcal/mol. These findings provided evidence to conclude that metabolites from marine-derived fungi are potential sources of bioactive metabolites with antimycobacterial activity. Even though in silico studies showed that bionectin F is a potent inhibitor of an essential enzyme, MabA, the results should be validated by performing purification of bionectin F from C. rogersoniana MGK33 and in vitro assays against MabA and whole cells (M. tuberculosis).

6.
Curr Dir Autoimmun ; 11: 157-79, 2010.
Article in English | MEDLINE | ID: mdl-20173394

ABSTRACT

TNF is essential to control Mycobacterium tuberculosis infection and cannot be replaced by other proinflammatory cytokines. Overproduction of TNF may cause immunopathology, while defective TNF production results in uncontrolled infection. The critical role of TNF in the control of tuberculosis has been illustrated recently by primary and reactivation of latent infection in some patients under pharmacological anti-TNF therapy for rheumatoid arthritis or Crohn's disease. In this review, we discuss results of recent studies aimed at better understanding of molecular, cellular and kinetic aspects of TNF-mediated regulation of host-mycobacteria interactions. In particular, recent data using either mutant mice expressing solely membrane TNF or specific inhibitor sparing membrane TNF demonstrated that membrane TNF is sufficient to control acute M. tuberculosis infection. This is opening the way to selective TNF neutralization that might retain the desired anti-inflammatory effect but reduce the infectious risk.


Subject(s)
Host-Pathogen Interactions/immunology , Tuberculosis/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Antibodies, Neutralizing/adverse effects , Humans , Inflammation/immunology , Lymphocyte Activation , Macrophage Activation , Mice , Mice, Knockout , Mice, Transgenic , Models, Immunological , Mycobacterium tuberculosis/immunology , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics
7.
Iran J Basic Med Sci ; 23(5): 680-690, 2020 May.
Article in English | MEDLINE | ID: mdl-32742607

ABSTRACT

OBJECTIVES: This study aimed to explore the contribution of tumor necrosis factor (TNF) in the recruitment of B-cell and secretion of immunoglobulins (Igs) during cerebral tuberculosis (TB). MATERIALS AND METHODS: In this work, the contributing role of TNF in regulating Ig secretions was investigated by comparing wild type TNF (TNFf/f), B-cell-derived TNF (BTNF-/-), and complete TNF ablation (TNF-/-) in a mouse cerebral Mycobacterium tuberculosis infection. Using flow cytometry and ELISA, we were able to examine the recruitment of B-cell subsets, and the production of Igs; also assessed the expression of surface markers on B cell subsets. RESULTS: Here, we found that TNF-/- mice showed defective expression of IgA, IgG, and IgM antibodies compared with TNFf/f and BTNF-/- mice, which was significantly decreased in the expression of surface markers and co-stimulatory molecules. Moreover, mice that produced low antibody levels were not able to control infection, therefore progressed to disease; providing direct evidence for the TNF gene-regulating humoral immunity during central nervous system (CNS) M. tuberculosis infection. In contrast, BTNF-/- mice controlled the infection and had levels of IgA, IgG, and IgM comparable to TNFf/f mice. CONCLUSION: Together, our results demonstrate that TNF may serve as an essential regulator of antibody-mediated immune responses in CNS TB. However, the protective level exhibited by TNF-producing B cells could be defined as baseline protection that could be used in the development of new therapeutic targets or designing new vaccines.

8.
Immunology ; 125(4): 522-34, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18544042

ABSTRACT

Tumour necrosis factor-alpha (TNF-alpha) plays a critical role in the recruitment and activation of mononuclear cells in mycobacterial infection. The role of membrane TNF, in host resistance against Mycobacterium bovis bacille Calmette-Guérin (BCG), was tested in knock-in mice in which the endogenous TNF was replaced by a non-cleavable and regulated allele (Delta1-12, TNF(tm/tm)). While 100% of mice with complete TNF deficiency (TNF(-/-)) succumbed to infection, 50% of TNF(tm/tm) mice were able to control M. bovis BCG infection and survived the experimental period. Membrane expressed TNF allowed a substantial recruitment of activated T cells and macrophages with granuloma formation and expression of bactericidal inducible nitric oxide synthase (iNOS). Using virulent Mycobacterium tuberculosis infection we confirm that membrane TNF conferred partial protection. Infection in TNF(tm/tm) double transgenic mice with TNF-R1 or TNF-R2 suggest protection is mediated through TNF-R2 signalling. Therefore, the data suggest that membrane-expressed TNF plays a critical role in host defence to mycobacterial infection and may partially substitute for soluble TNF.


Subject(s)
Mycobacterium Infections/immunology , Mycobacterium bovis , Tumor Necrosis Factor-alpha/immunology , Animals , BCG Vaccine/pharmacology , Bronchoalveolar Lavage Fluid/immunology , Cell Count , Cytokines/immunology , Female , Flow Cytometry , Immunohistochemistry , Lipopolysaccharides , Lymphocyte Activation , Lymphocytes/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Neutrophils/immunology , Shock, Septic/immunology , Tumor Necrosis Factor-alpha/genetics
9.
Microbes Infect ; 9(5): 623-8, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17409008

ABSTRACT

Tumor necrosis factor (TNF) is critical and non-redundant to control Mycobacterium tuberculosis infection and cannot be replaced by other proinflammatory cytokines. Overproduction of TNF may cause immunopathology, while TNF neutralization reactivates latent and chronic, controlled infection, which is relevant for the use of neutralizing TNF therapies in patients with rheumatoid arthritis.


Subject(s)
Mycobacterium tuberculosis/physiology , Tuberculosis/immunology , Tumor Necrosis Factors/physiology , Animals , Humans , Mycobacterium tuberculosis/immunology , Tuberculosis/physiopathology , Tumor Necrosis Factors/deficiency , Tumor Necrosis Factors/immunology
10.
Eur Cytokine Netw ; 18(1): 5-13, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17400533

ABSTRACT

Tumor necrosis factor (TNF) is required in the control of infection with Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis. TNF is essential and non-redundant for forming microbiocidal granulomas, and cannot be replaced by other members of the TNF family. We established a model of latent Mtb infection in mice, allowing investigation of the reactivation of latent Mtb as observed in patients receiving TNF-neutralizing therapy used in rheumatoid arthritis and Crohn's disease. Antibody neutralization of TNF is able to reactivate clinically silent Mtb infection. Using mutant mice expressing solely membrane, but not soluble TNF, we demonstrated that membrane TNF is sufficient to control acute Mtb infection. Therefore, we hypothesize that TNF-neutralizing therapy, sparing membrane TNF, may have an advantage as compared to complete neutralization. In conclusion, endogenous TNF is critical for the control of tuberculosis infection. Genetic absence or pharmacological neutralization of TNF results in uncontrolled infection, while selective neutralization might retain the desired anti-inflammatory effect but reduce the infectious risk.


Subject(s)
Tuberculosis/immunology , Tumor Necrosis Factor-alpha/deficiency , Animals , Cell Membrane/immunology , Cell Membrane/metabolism , Cell Membrane/microbiology , Humans , Mice , Models, Biological , Mycobacterium tuberculosis/immunology , Tuberculosis/metabolism , Tuberculosis/microbiology , Tumor Necrosis Factor-alpha/immunology
11.
Front Immunol ; 8: 180, 2017.
Article in English | MEDLINE | ID: mdl-28280495

ABSTRACT

Tuberculosis of the central nervous system (CNS-TB) is a devastating complication of tuberculosis, and tumor necrosis factor (TNF) is crucial for innate immunity and controlling the infection. TNF is produced by many cell types upon activation, in particularly the myeloid and T cells during neuroinflammation. Here we used mice with TNF ablation targeted to myeloid and T cell (MT-TNF-/-) to assess the contribution of myeloid and T cell-derived TNF in immune responses during CNS-TB. These mice exhibited impaired innate immunity and high susceptibility to cerebral Mycobacterium tuberculosis infection, a similar phenotype to complete TNF-deficient mice. Further, MT-TNF-/- mice were not able to control T cell responses and cytokine/chemokine production. Thus, our data suggested that collective TNF production by both myeloid and T cells are required to provide overall protective immunity against CNS-TB infection.

13.
Sci Rep ; 6: 39499, 2016 12 20.
Article in English | MEDLINE | ID: mdl-27995986

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.


Subject(s)
Latent Tuberculosis/immunology , Receptors, Tumor Necrosis Factor, Type I/metabolism , T-Lymphocytes/cytology , Animals , Chronic Disease , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Hyaluronan Receptors/metabolism , Interferon-gamma/metabolism , Lung/pathology , Macrophages, Peritoneal/microbiology , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis , Nitric Oxide/chemistry , T-Lymphocytes/metabolism
14.
Respir Res ; 6: 136, 2005 Nov 14.
Article in English | MEDLINE | ID: mdl-16285886

ABSTRACT

BACKGROUND: Tumour necrosis factor (TNF) is crucial for the control of mycobacterial infection as TNF deficient (KO) die rapidly of uncontrolled infection with necrotic pneumonia. Here we investigated the role of membrane TNF for host resistance in knock-in mice with a non-cleavable and regulated allele (mem-TNF). METHODS: C57BL/6, TNF KO and mem-TNF mice were infected with M. tuberculosis H37Rv (Mtb at 100 CFU by intranasal administration) and the survival, bacterial load, lung pathology and immunological parameters were investigated. Bone marrow and lymphocytes transfers were used to test the role of membrane TNF to confer resistance to TNF KO mice. RESULTS: While TNF-KO mice succumbed to infection within 4-5 weeks, mem-TNF mice recruited normally T cells and macrophages, developed mature granuloma in the lung and controlled acute Mtb infection. However, during the chronic phase of infection mem-TNF mice succumbed to disseminated infection with necrotic pneumonia at about 150 days. Reconstitution of irradiated TNF-KO mice with mem-TNF derived bone marrow cells, but not with lymphocytes, conferred host resistance to Mtb infection in TNF-KO mice. CONCLUSION: Membrane expressed TNF is sufficient to allow cell-cell signalling and control of acute Mtb infection. Bone marrow cells, but not lymphocytes from mem-TNF mice confer resistance to infection in TNF-KO mice. Long-term infection control with chronic inflammation likely disrupting TNF mediated cell-cell signalling, additionally requires soluble TNF.


Subject(s)
Cell Membrane/immunology , Lung/immunology , Lung/pathology , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/pathology , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/immunology , Acute Disease , Animals , Cells, Cultured , Cytoprotection/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout
15.
J Clin Invest ; 124(4): 1537-51, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24569452

ABSTRACT

Development of host protective immunity against Mycobacterium tuberculosis infection is critically dependent on the inflammatory cytokine TNF. TNF signals through 2 receptors, TNFRp55 and TNFRp75; however, the role of TNFRp75-dependent signaling in immune regulation is poorly defined. Here we found that mice lacking TNFRp75 exhibit greater control of M. tuberculosis infection compared with WT mice. TNFRp75-/- mice developed effective bactericidal granulomas and demonstrated increased pulmonary recruitment of activated DCs. Moreover, IL-12p40-dependent migration of DCs to lung draining LNs of infected TNFRp75-/- mice was substantially higher than that observed in WT M. tuberculosis-infected animals and was associated with enhanced frequencies of activated M. tuberculosis-specific IFN-γ-expressing CD4+ T cells. In WT mice, TNFRp75 shedding correlated with markedly reduced bioactive TNF levels and IL-12p40 expression. Neutralization of TNFRp75 in M. tuberculosis-infected WT BM-derived DCs (BMDCs) increased production of bioactive TNF and IL-12p40 to a level equivalent to that produced by TNFRp75-/- BMDCs. Addition of exogenous TNFRp75 to TNFRp75-/- BMDCs infected with M. tuberculosis decreased IL-12p40 synthesis, demonstrating that TNFRp75 shedding regulates DC activation. These data indicate that TNFRp75 shedding downmodulates protective immune function and reduces host resistance and survival; therefore, targeting TNFRp75 may be beneficial for improving disease outcome.


Subject(s)
Mycobacterium tuberculosis/immunology , Receptors, Tumor Necrosis Factor, Type II/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Female , Granuloma/pathology , Host-Pathogen Interactions/immunology , Interleukin-12 Subunit p40/metabolism , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium tuberculosis/pathogenicity , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/immunology , Receptors, Tumor Necrosis Factor, Type II/deficiency , Receptors, Tumor Necrosis Factor, Type II/genetics , Signal Transduction/immunology , Solubility , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/microbiology , Tuberculosis, Pulmonary/pathology , Tumor Necrosis Factor Decoy Receptors/deficiency , Tumor Necrosis Factor Decoy Receptors/genetics , Tumor Necrosis Factor Decoy Receptors/immunology , Tumor Necrosis Factor-alpha/metabolism
16.
Sci Rep ; 3: 1809, 2013.
Article in English | MEDLINE | ID: mdl-23657146

ABSTRACT

Tumour Necrosis Factor (TNF) is critical for host control of M. tuberculosis, but the relative contribution of TNF from innate and adaptive immune responses during tuberculosis infection is unclear. Myeloid versus T-cell-derived TNF function in tuberculosis was investigated using cell type-specific TNF deletion. Mice deficient for TNF expression in macrophages/neutrophils displayed early, transient susceptibility to M. tuberculosis but recruited activated, TNF-producing CD4(+) and CD8(+) T-cells and controlled chronic infection. Strikingly, deficient TNF expression in T-cells resulted in early control but susceptibility and eventual mortality during chronic infection with increased pulmonary pathology. TNF inactivation in both myeloid and T-cells rendered mice critically susceptible to infection with a phenotype resembling complete TNF deficient mice, indicating that myeloid and T-cells are the primary TNF sources collaborating for host control of tuberculosis. Thus, while TNF from myeloid cells mediates early immune function, T-cell derived TNF is essential to sustain protection during chronic tuberculosis infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lung/immunology , Mycobacterium tuberculosis/immunology , Myeloid Cells/immunology , Pneumonia/etiology , Tuberculosis/prevention & control , Tumor Necrosis Factor-alpha/physiology , Animals , Blotting, Western , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Flow Cytometry , Integrases/metabolism , Lung/cytology , Lung/microbiology , Macrophages/cytology , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Knockout , Myeloid Cells/cytology , Myeloid Cells/microbiology , Neutrophils/cytology , Neutrophils/immunology , Neutrophils/microbiology , Pneumonia/metabolism , Pneumonia/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tuberculosis/immunology , Tuberculosis/microbiology
17.
PLoS One ; 6(11): e25121, 2011.
Article in English | MEDLINE | ID: mdl-22132068

ABSTRACT

Of those individuals who are infected with M. tuberculosis, 90% do not develop active disease and represents a large reservoir of M. tuberculosis with the potential for reactivation of infection. Sustained TNF expression is required for containment of persistent infection and TNF neutralization leads to tuberculosis reactivation. In this study, we investigated the contribution of soluble TNF (solTNF) and transmembrane TNF (Tm-TNF) in immune responses generated against reactivating tuberculosis. In a chemotherapy induced tuberculosis reactivation model, mice were challenged by aerosol inhalation infection with low dose M. tuberculosis for three weeks to establish infection followed chemotherapeutic treatment for six weeks, after which therapy was terminated and tuberculosis reactivation investigated. We demonstrate that complete absence of TNF results in host susceptibility to M. tuberculosis reactivation in the presence of established mycobacteria-specific adaptive immunity with mice displaying unrestricted bacilli growth and diffused granuloma structures compared to WT control mice. Interestingly, bacterial re-emergence is contained in Tm-TNF mice during the initial phases of tuberculosis reactivation, indicating that Tm-TNF sustains immune pressure as in WT mice. However, Tm-TNF mice show susceptibility to long term M. tuberculosis reactivation associated with uncontrolled influx of leukocytes in the lungs and reduced IL-12p70, IFNγ and IL-10, enlarged granuloma structures, and failure to contain mycobacterial replication relative to WT mice. In conclusion, we demonstrate that both solTNF and Tm-TNF are required for maintaining immune pressure to contain reactivating M. tuberculosis bacilli even after mycobacteria-specific immunity has been established.


Subject(s)
Cell Membrane/metabolism , Mycobacterium tuberculosis/physiology , Tuberculosis/microbiology , Tuberculosis/pathology , Tumor Necrosis Factor-alpha/metabolism , Animals , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Granuloma/complications , Granuloma/pathology , Inflammation/pathology , Lung/drug effects , Lung/enzymology , Lung/microbiology , Lung/pathology , Lymphocytes/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/growth & development , Nitric Oxide Synthase Type II/metabolism , Tuberculosis/complications , Tuberculosis/prevention & control
18.
Tuberculosis (Edinb) ; 89 Suppl 1: S33-40, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20006302

ABSTRACT

Artemisia afra [Jacq] (Asteraceae) phytotherapy is widely used for its medicinal properties in traditional practices. In this study we investigated whether extracts of A. afra are capable of controlling mycobacterial replication. For Mycobacterium aurum cultured in the presence of aqueous-, methanol- and dichloromethane (DCM) extracts of A. afra we found that bacterial replication was inhibited by the dichloromethane extract only. Activity of the DCM extract was confirmed in dose-dependent studies against both M. aurum and M. tuberculosis with an IC(50) =270 microg/ml and IC(50) = 290microg/ml, respectively. Fractionation of the DCM extract and evaluation of its efficacy in vitro found that most of the antimycobacterial activity was associated with isolate fraction C8 that contained several sesquiterpene lactones, the most prominent of which are Artemin and Arsubin. Evaluation of the bactericidal efficacy in vitro showed that isolate fraction C8 reduced replication of M. aurum and M. tuberculosis in a dose-dependent manner with IC(50) =1.9 microg/ml and IC(50) = 2.0 microg/ml, respectively, and an MIC = 10 microg/ml. Further, isolate fraction C8 and the DCM extract was administered to M. tuberculosis-infected mice at a tolerated dose of 1000 microg/kg for up to 26 weeks and mycobacterial burdens compared to untreated-, INH/RIF treated- and aqueous-extract-treated animals to assess its bactericidal activity in vivo. Bacterial replication remained unaffected during treatment with either isolate fraction C8 or the DCM extract resulting in pulmonary and splenic bacilli burdens comparable to that of untreated mice. In contrast, INH/RIF treatment cleared M. tuberculosis infection after only 8 weeks to undetectable levels. Interestingly, treatment of M. tuberculosis-infected mice with aqueous extract of A. afra regulated pulmonary inflammation during early infection notwithstanding its inability to inhibit mycobacterial growth. This study clearly demonstrates that A. afra contains in vitro anti-mycobacterial activity, modulates pulmonary inflammation in early mycobacterial infection, and that the mouse experimental tuberculosis model may serve as a useful assay for evaluating the utility of phytotherapy.


Subject(s)
Artemisia , Methylene Chloride/pharmacology , Plant Extracts/pharmacology , Tuberculosis/drug therapy , Animals , Female , Mice , Mice, Inbred C57BL , Models, Animal , Phytotherapy , Tuberculosis/pathology
19.
J Infect Dis ; 196(6): 954-62, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17703428

ABSTRACT

Control of Trypanosoma congolense infections requires an early cell-mediated immune response. To unravel the role of tumor necrosis factor (TNF) in this process, 6 different T. congolense strains were used in 6 different gene-deficient mouse models that included TNF(-/-), TNF receptor-1 (TNFp55)(-/-), and TNF receptor-2 (TNFp75)(-/-) mice, 2 cell type-specific TNF(-/-) mice, as well as TNF-knock-in mice that expressed only membrane-bound TNF. Our results indicate that soluble TNF produced by macrophages/neutrophils and TNFp55 signaling are essential and sufficient to control parasitemia. The downstream mechanism in the control of T. congolense infection depends on inducible nitric oxide synthase activation in the liver. Such a role for nitric oxide is corroborated ex vivo, because the inhibitor N(G)-monomethyl-l-arginine blocks the trypanolytic activity of the adherent liver cell population, whereas exogenous interferon- gamma that stimulates nitric oxide production enhances parasite killing. In conclusion, the control of T. congolense infection depends on macrophage/neutrophil-derived soluble TNF and intact TNFp55 signaling, which induces trypanolytic nitric oxide.


Subject(s)
Macrophages/immunology , Nitric Oxide/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Trypanosoma congolense/immunology , Trypanosomiasis, African/immunology , Tumor Necrosis Factor-alpha/metabolism , Animals , Cells, Cultured , Liver/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Parasitemia/immunology , Trypanosomiasis, African/metabolism , Trypanosomiasis, African/parasitology
20.
Clin Chem Lab Med ; 40(9): 893-902, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12435106

ABSTRACT

The role of the endogenous interleukin-10 (IL-10) in the control of Mycobacterium bovis Bacille Calmette Guerin (BCG) infection was assessed using IL-10-deficient (IL-10-/-) mice. Similar to wild-type (WT) mice, IL-10-/- mice were resistant to intravenous challenge with Mycobacterium bovis BCG. Significantly higher plasma concentrations of IL-12 and tumour necrosis factor (TNF) indicated an elevated protective immune response of IL-10-/- mice. Determination of bacilli burden in IL-10-/- mice showed accelerated clearance in the lungs, spleen and the liver in comparison to WT mice. Enhanced inflammation and a vigorous granulomatous response accompanied accelerated mycobacterial clearance. Immunohistochemical analysis of hepatic granulomas from IL-10-/- mice revealed augmented lymphocyte recruitment and macrophage activation, such as increased major histocompatibility complex (MHC) class II and inducible nitric oxide synthase (iNOS) expression. Further, it was found that enlarged granulomas persisted subsequent to mycobacterial clearance and failed to resolve in the absence of IL-10. In conclusion, endogenous IL-10 dampens the cell-mediated immune response to mycobacterial infection.


Subject(s)
Interleukin-10/deficiency , Interleukin-10/immunology , Mycobacterium bovis/immunology , Tuberculosis/immunology , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Granuloma/physiopathology , Immunohistochemistry/methods , Interleukin-10/genetics , Interleukin-12/biosynthesis , Interleukin-12/blood , Macrophage Activation/physiology , Mice , Mycobacterium bovis/genetics , Organ Size , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Tuberculosis/microbiology , Tumor Necrosis Factor-alpha/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL