Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
J Exp Med ; 220(8)2023 08 07.
Article in English | MEDLINE | ID: mdl-37097292

ABSTRACT

Control of Mycobacterium tuberculosis (Mtb) infection requires generation of T cells that migrate to granulomas, complex immune structures surrounding sites of bacterial replication. Here we compared the gene expression profiles of T cells in pulmonary granulomas, bronchoalveolar lavage, and blood of Mtb-infected rhesus macaques to identify granuloma-enriched T cell genes. TNFRSF8/CD30 was among the top genes upregulated in both CD4 and CD8 T cells from granulomas. In mice, CD30 expression on CD4 T cells is required for survival of Mtb infection, and there is no major role for CD30 in protection by other cell types. Transcriptomic comparison of WT and CD30-/- CD4 T cells from the lungs of Mtb-infected mixed bone marrow chimeric mice showed that CD30 directly promotes CD4 T cell differentiation and the expression of multiple effector molecules. These results demonstrate that the CD30 co-stimulatory axis is highly upregulated on granuloma T cells and is critical for protective T cell responses against Mtb infection.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Animals , Mice , CD4-Positive T-Lymphocytes , Cell Differentiation , Granuloma/metabolism , Macaca mulatta , Tuberculosis/microbiology , Ki-1 Antigen/immunology
2.
Nat Immunol ; 24(5): 855-868, 2023 05.
Article in English | MEDLINE | ID: mdl-37012543

ABSTRACT

Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), is a global cause of death. Granuloma-associated lymphoid tissue (GrALT) correlates with protection during TB, but the mechanisms of protection are not understood. During TB, the transcription factor IRF4 in T cells but not B cells is required for the generation of the TH1 and TH17 subsets of helper T cells and follicular helper T (TFH)-like cellular responses. A population of IRF4+ T cells coexpress the transcription factor BCL6 during Mtb infection, and deletion of Bcl6 (Bcl6fl/fl) in CD4+ T cells (CD4cre) resulted in reduction of TFH-like cells, impaired localization within GrALT and increased Mtb burden. In contrast, the absence of germinal center B cells, MHC class II expression on B cells, antibody-producing plasma cells or interleukin-10-expressing B cells, did not increase Mtb susceptibility. Indeed, antigen-specific B cells enhance cytokine production and strategically localize TFH-like cells within GrALT via interactions between programmed cell death 1 (PD-1) and its ligand PD-L1 and mediate Mtb control in both mice and macaques.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Mice , Animals , T-Lymphocytes, Helper-Inducer , B-Lymphocytes , Lymphoid Tissue , Germinal Center , Transcription Factors
3.
J Exp Med ; 219(11)2022 11 07.
Article in English | MEDLINE | ID: mdl-36069923

ABSTRACT

Cellular necrosis during Mycobacterium tuberculosis (Mtb) infection promotes both immunopathology and bacterial dissemination. Glutathione peroxidase-4 (Gpx4) is an enzyme that plays a critical role in preventing iron-dependent lipid peroxidation-mediated cell death (ferroptosis), a process previously implicated in the necrotic pathology seen in Mtb-infected mice. Here, we document altered GPX4 expression, glutathione levels, and lipid peroxidation in patients with active tuberculosis and assess the role of this pathway in mice genetically deficient in or overexpressing Gpx4. We found that Gpx4-deficient mice infected with Mtb display substantially increased lung necrosis and bacterial burdens, while transgenic mice overexpressing the enzyme show decreased bacterial loads and necrosis. Moreover, Gpx4-deficient macrophages exhibited enhanced necrosis upon Mtb infection in vitro, an outcome suppressed by the lipid peroxidation inhibitor, ferrostatin-1. These findings provide support for the role of ferroptosis in Mtb-induced necrosis and implicate the Gpx4/GSH axis as a target for host-directed therapy of tuberculosis.


Subject(s)
Ferroptosis , Glutathione Peroxidase/metabolism , Tuberculosis , Animals , Glutathione/metabolism , Lipid Peroxidation , Mice , Mice, Transgenic , Necrosis , Phospholipid Hydroperoxide Glutathione Peroxidase , Tuberculosis/immunology , Tuberculosis/metabolism
4.
bioRxiv ; 2022 Sep 15.
Article in English | MEDLINE | ID: mdl-36172119

ABSTRACT

The pro- and anti-inflammatory pathways that determine the balance of inflammation and viral control during SARS-CoV-2 infection are not well understood. Here we examine the roles of IFNγ and IL-10 in regulating inflammation, immune cell responses and viral replication during SARS-CoV-2 infection of rhesus macaques. IFNγ blockade tended to decrease lung inflammation based on 18 FDG-PET/CT imaging but had no major impact on innate lymphocytes, neutralizing antibodies, or antigen-specific T cells. In contrast, IL-10 blockade transiently increased lung inflammation and enhanced accumulation of virus-specific T cells in the lower airways. However, IL-10 blockade also inhibited the differentiation of virus-specific T cells into airway CD69 + CD103 + T RM cells. While virus-specific T cells were undetectable in the nasal mucosa of all groups, IL-10 blockade similarly reduced the frequency of total T RM cells in the nasal mucosa. Neither cytokine blockade substantially affected viral load and infection ultimately resolved. Thus, in the macaque model of mild COVID-19, the pro- and anti-inflammatory effects of IFNγ and IL-10 have no major role in control of viral replication. However, IL-10 has a key role in suppressing the accumulation of SARS-CoV-2-specific T cells in the lower airways, while also promoting T RM at respiratory mucosal surfaces.

5.
J Clin Invest ; 132(18)2022 09 15.
Article in English | MEDLINE | ID: mdl-35862216

ABSTRACT

A once-weekly oral dose of isoniazid and rifapentine for 3 months (3HP) is recommended by the CDC for treatment of latent tuberculosis infection (LTBI). The aim of this study is to assess 3HP-mediated clearance of M. tuberculosis bacteria in macaques with asymptomatic LTBI. Twelve Indian-origin rhesus macaques were infected with a low dose (~10 CFU) of M. tuberculosis CDC1551 via aerosol. Six animals were treated with 3HP and 6 were left untreated. The animals were imaged via PET/CT at frequent intervals. Upon treatment completion, all animals except 1 were coinfected with SIV to assess reactivation of LTBI to active tuberculosis (ATB). Four of 6 treated macaques showed no evidence of persistent bacilli or extrapulmonary spread until the study end point. PET/CT demonstrated the presence of significantly more granulomas in untreated animals relative to the treated group. The untreated animals harbored persistent bacilli and demonstrated tuberculosis (TB) reactivation following SIV coinfection, while none of the treated animals reactivated to ATB. 3HP treatment effectively reduced persistent infection with M. tuberculosis and prevented reactivation of TB in latently infected macaques.


Subject(s)
Latent Tuberculosis , Mycobacterium tuberculosis , Tuberculosis , Animals , Antitubercular Agents/pharmacology , Isoniazid/pharmacology , Latent Tuberculosis/drug therapy , Latent Tuberculosis/microbiology , Lung , Macaca mulatta , Positron Emission Tomography Computed Tomography , Rifampin/analogs & derivatives
6.
Cell Rep ; 39(9): 110896, 2022 05 31.
Article in English | MEDLINE | ID: mdl-35649361

ABSTRACT

HIV/Mycobacterium tuberculosis (Mtb) co-infected individuals have an increased risk of tuberculosis prior to loss of peripheral CD4 T cells, raising the possibility that HIV co-infection leads to CD4 T cell depletion in lung tissue before it is evident in blood. Here, we use rhesus macaques to study the early effects of simian immunodeficiency virus (SIV) co-infection on pulmonary granulomas. Two weeks after SIV inoculation of Mtb-infected macaques, Mtb-specific CD4 T cells are dramatically depleted from granulomas, before CD4 T cell loss in blood, airways, and lymph nodes, or increases in bacterial loads or radiographic evidence of disease. Spatially, CD4 T cells are preferentially depleted from the granuloma core and cuff relative to B cell-rich regions. Moreover, live imaging of granuloma explants show that intralesional CD4 T cell motility is reduced after SIV co-infection. Thus, granuloma CD4 T cells may be decimated before many co-infected individuals experience the first symptoms of acute HIV infection.


Subject(s)
Coinfection , HIV Infections , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus , Tuberculosis , Animals , CD4-Positive T-Lymphocytes , Coinfection/pathology , Granuloma/pathology , HIV Infections/complications , HIV Infections/pathology , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/complications , Simian Acquired Immunodeficiency Syndrome/pathology , Tuberculosis/pathology
7.
Sci Immunol ; : eabo0535, 2022 03 10.
Article in English | MEDLINE | ID: mdl-35271298

ABSTRACT

SARS-CoV-2 primarily replicates in mucosal sites, and more information is needed about immune responses in infected tissues. Here, we used rhesus macaques to model protective primary immune responses in tissues during mild COVID-19. Viral RNA levels were highest on days 1-2 post-infection and fell precipitously thereafter. 18F-fluorodeoxyglucose (FDG)-avid lung abnormalities and interferon (IFN)-activated monocytes and macrophages in the bronchoalveolar lavage (BAL) were found on days 3-4 post-infection. Virus-specific effector CD8+ and CD4+ T cells became detectable in the BAL and lung tissue on days 7-10, after viral RNA, radiologic evidence of lung inflammation, and IFN-activated myeloid cells had substantially declined. Notably, SARS-CoV-2-specific T cells were not detectable in the nasal turbinates, salivary glands, and tonsils on day 10 post-infection. Thus, SARS-CoV-2 replication wanes in the lungs of rhesus macaques prior to T cell responses, and in the nasal and oral mucosa despite the apparent lack of antigen-specific T cells, suggesting that innate immunity efficiently restricts viral replication during mild COVID-19.

8.
Sci Immunol ; 6(55)2021 01 15.
Article in English | MEDLINE | ID: mdl-33452107

ABSTRACT

Boosting immune cell function by targeting the coinhibitory receptor PD-1 may have applications in the treatment of chronic infections. Here, we examine the role of PD-1 during Mycobacterium tuberculosis (Mtb) infection of rhesus macaques. Animals treated with anti-PD-1 monoclonal antibody developed worse disease and higher granuloma bacterial loads compared with isotype control-treated monkeys. PD-1 blockade increased the number and functionality of granuloma Mtb-specific CD8 T cells. In contrast, Mtb-specific CD4 T cells in anti-PD-1-treated macaques were not increased in number or function in granulomas, expressed increased levels of CTLA-4, and exhibited reduced intralesional trafficking in live imaging studies. In granulomas of anti-PD-1-treated animals, multiple proinflammatory cytokines were elevated, and more cytokines correlated with bacterial loads, leading to the identification of a role for caspase 1 in the exacerbation of tuberculosis after PD-1 blockade. Last, increased Mtb bacterial loads after PD-1 blockade were found to associate with the composition of the intestinal microbiota before infection in individual macaques. Therefore, PD-1-mediated coinhibition is required for control of Mtb infection in macaques, perhaps because of its role in dampening detrimental inflammation and allowing for normal CD4 T cell responses.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Immune Checkpoint Inhibitors/adverse effects , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Tuberculosis/drug therapy , Animals , Bacterial Load/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CTLA-4 Antigen/metabolism , Disease Models, Animal , Humans , Immune Checkpoint Inhibitors/administration & dosage , Macaca mulatta , Male , Mice , Mice, Knockout , Mycobacterium tuberculosis/immunology , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Severity of Illness Index , Symptom Flare Up , Tuberculosis/diagnosis , Tuberculosis/immunology , Tuberculosis/microbiology
9.
Am J Respir Crit Care Med ; 201(4): 469-477, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31647877

ABSTRACT

Rationale: Direct evidence for persistence of Mycobacterium tuberculosis (Mtb) during asymptomatic latent tuberculosis infection (LTBI) in humans is currently lacking. Moreover, although a 12-week regimen of once-weekly isoniazid and rifapentine (3HP) is currently recommended by the CDC as treatment for LTBI, experimental evidence for 3HP-mediated clearance of persistent Mtb infection in human lungs has not been established.Objectives: Using a nonhuman primate (NHP) model of TB, we sought to assess 3HP treatment-mediated clearance of Mtb infection in latently infected macaques.Methods: Sixteen NHPs were infected via inhalation with ∼10 cfu of Mtb CDC1551, after which asymptomatic animals were either treated with 3HP or left untreated. Pharmacokinetics of the 3HP regimen were measured. Following treatment, animals were coinfected with simian immunodeficiency virus to assess reactivation of LTBI and development of active TB disease.Measurements and Main Results: Fourteen NHPs remained free of clinical signs or microbiological evidence of active TB following infection with Mtb and were subsequently either treated with 3HP (n = 7) or left untreated (n = 7). Untreated NHPs were asymptomatic for 7 months but harbored persistent Mtb infection, as shown by reactivation of latent infection following simian immunodeficiency virus coinfection. However, none of the treated animals developed TB reactivation disease, and they remained without clinical or microbiological evidence of persistent bacilli, suggesting treatment-mediated clearance of bacteria.Conclusions:Mtb can persist in asymptomatic macaques for at least 7 months. Furthermore, 3HP treatment effectively cleared bacteria and prevented reactivation of TB in latently infected macaques.


Subject(s)
Antibiotics, Antitubercular/therapeutic use , Antitubercular Agents/therapeutic use , Isoniazid/therapeutic use , Latent Tuberculosis/drug therapy , Mycobacterium tuberculosis/drug effects , Rifampin/analogs & derivatives , Tuberculosis/drug therapy , Animals , Drug Therapy, Combination , Macaca , Models, Animal , Rifampin/therapeutic use , Treatment Outcome
11.
J Clin Invest ; 129(12): 5254-5260, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31479428

ABSTRACT

HIV is a major driver of tuberculosis (TB) reactivation. Depletion of CD4+ T cells is assumed to be the basis behind TB reactivation in individuals with latent tuberculosis infection (LTBI) coinfected with HIV. Nonhuman primates (NHPs) coinfected with a mutant simian immunodeficiency virus (SIVΔGY) that does not cause depletion of tissue CD4+ T cells during infection failed to reactivate TB. To investigate the contribution of CD4+ T cell depletion relative to other mechanisms of SIV-induced reactivation of LTBI, we used CD4R1 antibody to deplete CD4+ T cells in animals with LTBI without lentiviral infection. The mere depletion of CD4+ T cells during LTBI was insufficient in generating reactivation of LTBI. Instead, direct cytopathic effects of SIV resulting in chronic immune activation, along with the altered effector T cell phenotypes and dysregulated T cell homeostasis, were likely mediators of reactivation of LTBI. These results revealed important implications for TB control in HIV-coinfected individuals.


Subject(s)
Coinfection/microbiology , Coinfection/virology , Latent Tuberculosis/complications , Simian Acquired Immunodeficiency Syndrome/complications , Animals , CD4-Positive T-Lymphocytes/microbiology , CD4-Positive T-Lymphocytes/virology , Homeostasis , Latent Tuberculosis/virology , Lentivirus , Lymphocyte Depletion , Macaca mulatta , Mutation , Mycobacterium tuberculosis , Phenotype , Simian Acquired Immunodeficiency Syndrome/microbiology , Simian Immunodeficiency Virus
12.
Tuberculosis (Edinb) ; 116S: S11-S18, 2019 05.
Article in English | MEDLINE | ID: mdl-31072689

ABSTRACT

TB is a catastrophic infectious disease, affecting roughly one third of the world's population. Mucosal-associated invariant T (MAIT) cells are innate-like T cells that recognize vitamin B metabolites produced by bacteria, possess effector memory phenotype, and express tissue-homing markers driving migration to sites of infection. Previous research in both Mtb and HIV infections has shown that MAIT cells are depleted in the human periphery, possibly migrating to the tissue sites of infection. We investigated this hypothesis using rhesus macaques (RMs) with active TB, latent TB (LTBI), and SIV-coinfection to explore the effects of different disease states on the MAIT cell populations in vivo. Early in infection, we observed that MAIT cells increased in the blood and bronchoalveolar lavage fluid (BAL) of all infected RMs, irrespective of clinical outcome. However, the frequency of MAIT cells rapidly normalized such that they had returned to baseline levels prior to endpoint. Furthermore, following infection, the chemokines expressed on MAIT cells reflected a strong shift towards a Th1 phenotype from a shared Th1/Th17 phenotype. In conclusion, MAIT cells with enhanced Th1 functions migrating to the site of Mtb-infection. The anti-mycobacterial effector functions of MAIT cells, particularly during the early stages of Mtb infection, had been of interest in promoting protective long-term TB immunity. Our research shows, however, that they have relatively short-acting responses in the host.


Subject(s)
Cell Movement , Cell Proliferation , Immunity, Mucosal , Latent Tuberculosis/microbiology , Lung/microbiology , Lymphocyte Activation , Mucosal-Associated Invariant T Cells/microbiology , Mycobacterium tuberculosis/pathogenicity , Tuberculosis, Pulmonary/microbiology , Animals , Coinfection , Disease Models, Animal , Host-Pathogen Interactions , Latent Tuberculosis/immunology , Lung/immunology , Macaca mulatta , Mucosal-Associated Invariant T Cells/immunology , Mucosal-Associated Invariant T Cells/virology , Mycobacterium tuberculosis/immunology , Phenotype , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian virus 40/immunology , Simian virus 40/pathogenicity , Th1 Cells/immunology , Th1 Cells/microbiology , Th2 Cells/immunology , Th2 Cells/microbiology , Time Factors , Tuberculosis, Pulmonary/immunology
13.
Nat Microbiol ; 3(11): 1198-1205, 2018 11.
Article in English | MEDLINE | ID: mdl-30202016

ABSTRACT

Mycobacterium tuberculosis infection (Mtb) is the leading cause of death due to a single infectious agent and is among the top ten causes of all human deaths worldwide1. CD4 T cells are essential for resistance to Mtb infection, and for decades it has been thought that IFNγ production is the primary mechanism of CD4 T-cell-mediated protection2,3. However, IFNγ responses do not correlate with host protection, and several reports demonstrate that additional anti-tuberculosis CD4 T-cell effector functions remain unaccounted for4-8. Here we show that the tumour-necrosis factor (TNF) superfamily molecule CD153 (encoded by the gene Tnfsf8) is required for control of pulmonary Mtb infection by CD4 T cells. In Mtb-infected mice, CD153 expression is highest on Mtb-specific T helper 1 (TH1) cells in the lung tissue parenchyma, but its induction does not require TH1 cell polarization. CD153-deficient mice develop high pulmonary bacterial loads and succumb early to Mtb infection. Reconstitution of T-cell-deficient hosts with either Tnfsf8-/- or Ifng-/- CD4 T cells alone fails to rescue mice from early mortality, but reconstitution with a mixture of Tnfsf8-/- and Ifng-/- CD4 T cells provides similar protection as wild-type T cells. In Mtb-infected non-human primates, CD153 expression is much higher on Ag-specific CD4 T cells in the airways compared to blood, and the frequency of Mtb-specific CD153-expressing CD4 T cells inversely correlates with bacterial loads in granulomas. In Mtb-infected humans, CD153 defines a subset of highly polyfunctional Mtb-specific CD4 T cells that are much more abundant in individuals with controlled latent Mtb infection compared to those with active tuberculosis. In all three species, Mtb-specific CD8 T cells did not upregulate CD153 following peptide stimulation. Thus, CD153 is a major immune mediator of host protection against pulmonary Mtb infection and CD4 T cells are one important source of this molecule.


Subject(s)
CD30 Ligand/genetics , Disease Resistance/genetics , Gene Expression , Mycobacterium tuberculosis/immunology , Tuberculosis/immunology , Animals , Bacterial Load , CD30 Ligand/deficiency , CD30 Ligand/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Disease Resistance/immunology , Host-Pathogen Interactions/immunology , Humans , Latent Tuberculosis/immunology , Latent Tuberculosis/microbiology , Lung/immunology , Lung/microbiology , Mice , Mycobacterium tuberculosis/physiology , Primates , Th1 Cells/immunology , Th1 Cells/metabolism , Tuberculosis/microbiology
14.
Proc Natl Acad Sci U S A ; 115(1): E62-E71, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29255022

ABSTRACT

Mycobacterium tuberculosis continues to cause devastating levels of mortality due to tuberculosis (TB). The failure to control TB stems from an incomplete understanding of the highly specialized strategies that M. tuberculosis utilizes to modulate host immunity and thereby persist in host lungs. Here, we show that M. tuberculosis induced the expression of indoleamine 2,3-dioxygenase (IDO), an enzyme involved in tryptophan catabolism, in macrophages and in the lungs of animals (mice and macaque) with active disease. In a macaque model of inhalation TB, suppression of IDO activity reduced bacterial burden, pathology, and clinical signs of TB disease, leading to increased host survival. This increased protection was accompanied by increased lung T cell proliferation, induction of inducible bronchus-associated lymphoid tissue and correlates of bacterial killing, reduced checkpoint signaling, and the relocation of effector T cells to the center of the granulomata. The enhanced killing of M. tuberculosis in macrophages in vivo by CD4+ T cells was also replicated in vitro, in cocultures of macaque macrophages and CD4+ T cells. Collectively, these results suggest that there exists a potential for using IDO inhibition as an effective and clinically relevant host-directed therapy for TB.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Lung/immunology , Mycobacterium tuberculosis/immunology , Tryptophan/immunology , Tuberculoma/immunology , Tuberculosis, Pulmonary/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Granuloma/immunology , Granuloma/pathology , Lung/pathology , Macaca mulatta , Macrophages/immunology , Macrophages/pathology , Mycobacterium tuberculosis/pathogenicity , Tuberculoma/pathology , Tuberculosis, Pulmonary/pathology
15.
Am J Pathol ; 187(12): 2811-2820, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28935575

ABSTRACT

Failure to replace Bacille Calmette-Guerin vaccines with efficacious anti-tuberculosis (TB) vaccines have prompted outside-the-box thinking, including pulmonary vaccination to elicit local immunity. Inhalational MtbΔsigH, a stress-response-attenuated strain, protected against lethal TB in macaques. While live mycobacterial vaccines show promising efficacy, HIV co-infection and the resulting immunodeficiency prompts safety concerns about their use. We assessed the persistence and safety of MtbΔsigH, delivered directly to the lungs, in the setting of HIV co-infection. Macaques were aerosol-vaccinated with ΔsigH and subsequently challenged with SIVmac239. Bronchoalveolar lavage and tissues were sampled for mycobacterial persistence, pathology, and immune correlates. Only 35% and 3.5% of lung samples were positive for live bacilli and granulomas, respectively. Our results therefore suggest that the nonpathologic infection of macaque lungs by ΔsigH was not reactivated by simian immunodeficiency virus, despite high viral levels and massive ablation of pulmonary CD4+ T cells. Protective pulmonary responses were retained, including vaccine-induced bronchus-associated lymphoid tissue and CD8+ effector memory T cells. Despite acute simian immunodeficiency virus infection, all animals remained asymptomatic of pulmonary TB. These findings highlight the efficacy of mucosal vaccination via this attenuated strain and will guide its further development to potentially combat TB in HIV-endemic areas. Our results also suggest that a lack of pulmonary pathology is a key correlate of the safety of live mycobacterial vaccines.


Subject(s)
Simian Acquired Immunodeficiency Syndrome/complications , Tuberculosis Vaccines/pharmacology , Tuberculosis/prevention & control , Virus Activation/drug effects , Administration, Inhalation , Animals , Coinfection , HIV , Macaca mulatta , Mycobacterium tuberculosis , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/physiology , Tuberculosis/complications , Vaccines, Attenuated/pharmacology
16.
PLoS One ; 12(9): e0180413, 2017.
Article in English | MEDLINE | ID: mdl-28880895

ABSTRACT

CD4+ T-cell mediated Th1 immune responses are critical for immunity to TB. The immunomodulatory protein, lymphocyte activation gene-3 (LAG-3) decreases Th1-type immune responses in T-cells. LAG-3 expression is significantly induced in the lungs of macaques with active TB and correlates with increased bacterial burden. Overproduction of LAG-3 can greatly diminish responses and could lead to uncontrolled Mtb replication. To assess the effect of LAG-3 on the progression of Mtb infection, we developed a co-culture system wherein blood-derived macrophages are infected with Mtb and supplemented with macaque blood or lung derived CD4+ T-cells. Silencing LAG-3 signaling in macaque lung CD4+ T-cells enhanced killing of Mtb in co-cultures, accompanied by reduced mitochondrial electron transport and increased IFN-γ expression. Thus, LAG-3 may modulate adaptive immunity to Mtb infection by interfering with the mitochondrial apoptosis pathway. Better understanding this pathway could allow us to circumvent immune features that promote disease.


Subject(s)
Antigens, CD/metabolism , Granuloma/metabolism , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Phagocytes/metabolism , Phagocytes/microbiology , Adaptive Immunity/genetics , Adaptive Immunity/physiology , Animals , Antigens, CD/genetics , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/physiology , Cells, Cultured , Coculture Techniques , Flow Cytometry , Granuloma/immunology , Granuloma/microbiology , Macaca mulatta , Microscopy, Confocal , Mitochondria/immunology , Mitochondria/metabolism , Mitochondria/microbiology , Phagocytes/immunology , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Transcriptome/genetics , Lymphocyte Activation Gene 3 Protein
17.
Sci Immunol ; 2(9)2017 Mar.
Article in English | MEDLINE | ID: mdl-28707004

ABSTRACT

Mycobacterium tuberculosis (Mtb) executes a plethora of immune-evasive mechanisms, which contribute to its pathogenesis, limited efficacy of current therapy, and the emergence of drug-resistant strains. This has led to resurgence in attempts to develop new therapeutic strategies/targets against tuberculosis (TB). We show that Mtb down-regulates sirtuin 1 (SIRT1), a nicotinamide adenine dinucleotide (NAD+)-dependent deacetylase, in monocytes/macrophages, TB animal models, and TB patients with active disease. Activation of SIRT1 reduced intracellular growth of drug-susceptible and drug-resistant strains of Mtb and induced phagosome-lysosome fusion and autophagy in a SIRT1-dependent manner. SIRT1 activation dampened Mtb-mediated persistent inflammatory responses via deacetylation of RelA/p65, leading to impaired binding of RelA/p65 on the promoter of inflammatory genes. In Mtb-infected mice, the use of SIRT1 activators ameliorated lung pathology, reduced chronic inflammation, and enhanced efficacy of anti-TB drug. Mass cytometry-based high-dimensional analysis revealed that SIRT1 activation mediated modulation of lung myeloid cells in Mtb-infected mice. Myeloid cell-specific SIRT1 knockout mice display increased inflammatory responses and susceptibility to Mtb infection. Collectively, these results provide a link between SIRT1 activation and TB pathogenesis and indicate a potential of SIRT1 activators in designing an effective and clinically relevant host-directed therapies for TB.

18.
ILAR J ; 58(2): 151-159, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28575319

ABSTRACT

Infection with Mycobacterium tuberculosis predominantly establishes subclinical latent infection over the lifetime of an individual, with a fraction of infected individuals rapidly progressing to active disease. The immune control in latent infection can be perturbed by comorbidities such as diabetes mellitus, obesity, smoking, and coinfection with helminthes or HIV. Modeling the varying aspects of natural infection remains incomplete when using zebrafish and mice. However, the nonhuman primate model of tuberculosis offers a unique and accurate model to investigate host responses to infection, test novel therapeutics, and thoroughly assess preclinical vaccine candidates. Rhesus macaques and cynomolgus macaques manifest the full gamut of clinical and pathological findings in human Mycobacterium tuberculosis infection, including the ability to co-infect macaques with Simian Immunodeficiency Virus to model HIV co-infection. Here we discuss advanced techniques to assay various clinical outcomes of the natural progression of infection as well as therapeutics in development and novel preclinical vaccines. Finally, we survey the translational aspects of nonhuman primate research and argue the urgent need to thoroughly examine preclinical therapeutics and vaccines using this model prior to clinical implementation.


Subject(s)
Translational Research, Biomedical/methods , Animals , Coinfection , HIV Infections/immunology , Macaca mulatta , Mice , Mycobacterium tuberculosis/pathogenicity , Simian Immunodeficiency Virus/pathogenicity , Tuberculosis/immunology , Tuberculosis/microbiology , Zebrafish
19.
Am J Respir Cell Mol Biol ; 56(5): 637-647, 2017 05.
Article in English | MEDLINE | ID: mdl-28135421

ABSTRACT

Although it is accepted that the environment within the granuloma profoundly affects Mycobacterium tuberculosis (Mtb) and infection outcome, our ability to understand Mtb gene expression in these niches has been limited. We determined intragranulomatous gene expression in human-like lung lesions derived from nonhuman primates with both active tuberculosis (ATB) and latent TB infection (LTBI). We employed a non-laser-based approach to microdissect individual lung lesions and interrogate the global transcriptome of Mtb within granulomas. Mtb genes expressed in classical granulomas with central, caseous necrosis, as well as within the caseum itself, were identified and compared with other Mtb lesions in animals with ATB (n = 7) or LTBI (n = 7). Results were validated using both an oligonucleotide approach and RT-PCR on macaque samples and by using human TB samples. We detected approximately 2,900 and 1,850 statistically significant genes in ATB and LTBI lesions, respectively (linear models for microarray analysis, Bonferroni corrected, P < 0.05). Of these genes, the expression of approximately 1,300 (ATB) and 900 (LTBI) was positively induced. We identified the induction of key regulons and compared our results to genes previously determined to be required for Mtb growth. Our results indicate pathways that Mtb uses to ensure its survival in a highly stressful environment in vivo. A large number of genes is commonly expressed in granulomas with ATB and LTBI. In addition, the enhanced expression of the dormancy survival regulon was a key feature of lesions in animals with LTBI, stressing its importance in the persistence of Mtb during the chronic phase of infection.


Subject(s)
Gene Expression Regulation, Bacterial , Genes, Bacterial , Granuloma/microbiology , Microbial Viability/genetics , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/physiology , Anaerobiosis , Animals , Gene Expression Profiling , Granuloma/pathology , Lung/microbiology , Lung/pathology , Macaca , Real-Time Polymerase Chain Reaction , Regulon/genetics , Reproducibility of Results , Transcriptome/genetics , Tuberculosis/genetics , Tuberculosis/microbiology , Tuberculosis/pathology
20.
Proc Natl Acad Sci U S A ; 113(38): E5636-44, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27601645

ABSTRACT

The synergy between Mycobacterium tuberculosis (Mtb) and HIV in coinfected patients has profoundly impacted global mortality because of tuberculosis (TB) and AIDS. HIV significantly increases rates of reactivation of latent TB infection (LTBI) to active disease, with the decline in CD4(+) T cells believed to be the major causality. In this study, nonhuman primates were coinfected with Mtb and simian immunodeficiency virus (SIV), recapitulating human coinfection. A majority of animals exhibited rapid reactivation of Mtb replication, progressing to disseminated TB and increased SIV-associated pathology. Although a severe loss of pulmonary CD4(+) T cells was observed in all coinfected macaques, a subpopulation of the animals was still able to prevent reactivation and maintain LTBI. Investigation of pulmonary immune responses and pathology in this cohort demonstrated that increased CD8(+) memory T-cell proliferation, higher granzyme B production, and expanded B-cell follicles correlated with protection from reactivation. Our findings reveal mechanisms that control SIV- and TB-associated pathology. These CD4-independent protective immune responses warrant further studies in HIV coinfected humans able to control their TB infection. Moreover, these findings will provide insight into natural immunity to Mtb and will guide development of novel vaccine strategies and immunotherapies.


Subject(s)
HIV Infections/immunology , Latent Tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Simian Immunodeficiency Virus/pathogenicity , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/microbiology , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , Cell Proliferation/genetics , Coinfection/virology , HIV/immunology , HIV/pathogenicity , HIV Infections/physiopathology , HIV Infections/virology , Humans , Immunologic Memory/genetics , Latent Tuberculosis/microbiology , Latent Tuberculosis/pathology , Latent Tuberculosis/virology , Lymphocyte Activation/immunology , Macaca mulatta/immunology , Macaca mulatta/microbiology , Macaca mulatta/virology , Mycobacterium tuberculosis/immunology , Simian Immunodeficiency Virus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...