Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Pathogens ; 12(9)2023 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-37764928

RESUMEN

HIV and TB are the cause of significant worldwide mortality and pose a grave danger to the global public health. TB is the leading cause of death in HIV-infected persons, with one in four deaths attributable to TB. While the majority of healthy individuals infected with M. tuberculosis (Mtb) are able to control the infection, co-infection with HIV increases the risk of TB infection progressing to TB disease by over 20-fold. While antiretroviral therapy (ART), the cornerstone of HIV care, decreases the incidence of TB in HIV-uninfected people, this remains 4- to 7-fold higher after ART in HIV-co-infected individuals in TB-endemic settings, regardless of the duration of therapy. Thus, the immune control of Mtb infection in Mtb/HIV-co-infected individuals is not fully restored by ART. We do not fully understand the reasons why Mtb/HIV-co-infected individuals maintain a high susceptibility to the reactivation of LTBI, despite an effective viral control by ART. A deep understanding of the molecular mechanisms that govern HIV-induced reactivation of TB is essential to develop improved treatments and vaccines for the Mtb/HIV-co-infected population. We discuss potential strategies for the mitigation of the observed chronic immune activation in combination with both anti-TB and anti-retroviral approaches.

2.
Nat Immunol ; 24(5): 855-868, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37012543

RESUMEN

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.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Ratones , Animales , Linfocitos T Colaboradores-Inductores , Linfocitos B , Tejido Linfoide , Centro Germinal , Factores de Transcripción
3.
JCI Insight ; 8(2)2023 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-36692017

RESUMEN

The expression of indoleamine 2,3-dioxygenase (IDO), a robust immunosuppressant, is significantly induced in macaque tuberculosis (TB) granulomas, where it is expressed on IFN-responsive macrophages and myeloid-derived suppressor cells. IDO expression is also highly induced in human TB granulomas, and products of its activity are detected in patients with TB. In vivo blockade of IDO activity resulted in the reorganization of the granuloma with substantially greater T cells being recruited to the core of the lesions. This correlated with better immune control of TB and reduced lung M. tuberculosis burdens. To study if the IDO blockade strategy can be translated to a bona fide host-directed therapy in the clinical setting of TB, we studied the effect of IDO inhibitor 1-methyl-d-tryptophan adjunctive to suboptimal anti-TB chemotherapy. While two-thirds of controls and one-third of chemotherapy-treated animals progressed to active TB, inhibition of IDO adjunctive to the same therapy protected macaques from TB, as measured by clinical, radiological, and microbiological attributes. Although chemotherapy improved proliferative T cell responses, adjunctive inhibition of IDO further enhanced the recruitment of effector T cells to the lung. These results strongly suggest the possibility that IDO inhibition can be attempted adjunctive to anti-TB chemotherapy in clinical trials.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis Pulmonar , Tuberculosis , Animales , Humanos , Granuloma , Indolamina-Pirrol 2,3,-Dioxigenasa , Macrófagos/metabolismo , Mycobacterium tuberculosis/metabolismo
4.
Front Immunol ; 13: 1001727, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36389795

RESUMEN

Despite advances in combination antiretroviral therapy (cART), people living with HIV (PLWH) continue to experience gastrointestinal dysfunction. Infusions of anti-α4ß7 monoclonal antibodies (mAbs) have been proposed to increase virologic control during simian immunodeficiency virus (SIV) infection in macaques with mixed results. Recent evidences suggested that therapeutic efficacy of vedolizumab (a humanized anti-α4ß7 mAb), during inflammatory bowel diseases depends on microbiome composition, myeloid cell differentiation, and macrophage phenotype. We tested this hypothesis in SIV-infected, anti-α4ß7 mAb-treated macaques and provide flow cytometric and microscopic evidence that anti-α4ß7 administered to SIV-infected macaques increases the maturity of macrophage phenotypes typically lost in the small intestines during SIV disease progression. Further, this increase in mature macrophage phenotype was associated with tissue viral loads. These phenotypes were also associated with dysbiosis markers in the gut previously identified as predictors of HIV replication and immune activation in PLWH. These findings provide a novel model of anti-α4ß7 efficacy offering new avenues for targeting pathogenic mucosal immune response during HIV/SIV infection.


Asunto(s)
Infecciones por VIH , Síndrome de Inmunodeficiencia Adquirida del Simio , Virus de la Inmunodeficiencia de los Simios , Animales , Macaca mulatta , Anticuerpos/uso terapéutico , Macrófagos , Infecciones por VIH/tratamiento farmacológico
5.
Front Immunol ; 13: 777733, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36275677

RESUMEN

Mycobacterium tuberculosis (Mtb) has developed specialized mechanisms to parasitize its host cell, the macrophage. These mechanisms allow it to overcome killing by oxidative burst and persist in the wake of an inflammatory response. Mtb infection in the majority of those exposed is controlled in an asymptomatic form referred to as latent tuberculosis infection (LTBI). HIV is a well-known catalyst of reactivation of LTBI to active TB infection (ATB). Through the use of nonhuman primates (NHPs) co-infected with Mtb and Simian Immunodeficiency Virus (Mtb/SIV), we are able to simulate human progression of TB/AIDS comorbidity. The advantage of NHP models is that they recapitulate the breadth of human TB outcomes, including immune control of infection, and loss of this control due to SIV co-infection. Identifying correlates of immune control of infection is important for both vaccine and therapeutics development. Using macaques infected with Mtb or Mtb/SIV and with different clinical outcomes we attempted to identify signatures between those that progress to active infection after SIV challenge (reactivators) and those that control the infection (non-reactivators). We particularly focused on pathways relevant to myeloid origin cells such as macrophages, as these innate immunocytes have an important contribution to the initial control or the lack thereof, following Mtb infection. Using bacterial burden, C-reactive protein (CRP), and other clinical indicators of disease severity as a guide, we were able to establish gene signatures of host disease state and progression. In addition to gene signatures, clustering algorithms were used to differentiate between host disease states and identify relationships between genes. This allowed us to identify clusters of genes which exhibited differential expression profiles between the three groups of macaques: ATB, LTBI and Mtb/SIV. The gene signatures were associated with pathways relevant to apoptosis, ATP production, phagocytosis, cell migration, and Type I interferon (IFN), which are related to macrophage function. Our results suggest novel macrophage functions that may play roles in the control of Mtb infection with and without co-infection with SIV. These results particularly point towards an interplay between Type I IFN signaling and IFN-γ signaling, and the resulting impact on lung macrophages as an important determinant of progression to TB.


Asunto(s)
Coinfección , Infecciones por VIH , Interferón Tipo I , Tuberculosis Latente , Infecciones por Lentivirus , Síndrome de Inmunodeficiencia Adquirida del Simio , Virus de la Inmunodeficiencia de los Simios , Animales , Humanos , Macaca , Proteína C-Reactiva , Biomarcadores , Infecciones por VIH/complicaciones , Adenosina Trifosfato
6.
J Clin Invest ; 132(18)2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35862216

RESUMEN

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.


Asunto(s)
Tuberculosis Latente , Mycobacterium tuberculosis , Tuberculosis , Animales , Antituberculosos/farmacología , Isoniazida/farmacología , Tuberculosis Latente/tratamiento farmacológico , Tuberculosis Latente/microbiología , Pulmón , Macaca mulatta , Tomografía Computarizada por Tomografía de Emisión de Positrones , Rifampin/análogos & derivados
7.
Am J Respir Crit Care Med ; 206(1): 94-104, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35412961

RESUMEN

Rationale: Different Mycobacterium tuberculosis (Mtb) strains exhibit variable degrees of virulence in humans and animal models. Differing stress response strategies used by different strains of Mtb could influence virulence. Objectives: We compared the virulence of two strains of Mtb with use in animal model research: CDC1551 and Erdman. Methods: Rhesus macaques, which develop human-like tuberculosis attributes and pathology, were infected with a high dose of either strain via aerosol, and virulence was compared by bacterial burden and pathology. Measurements and Main Results: Infection with Erdman resulted in significantly shorter times to euthanasia and higher bacterial burdens and greater systemic inflammation and lung pathology relative to those infected with CDC1551. Macaques infected with Erdman also exhibited significantly higher early inflammatory myeloid cell influx to the lung, greater macrophage and T cell activity, and higher expression of lung remodeling (extracellular matrix) genes, consistent with greater pathology. Expression of NOTCH4 (neurogenic locus notch homolog 4) signaling, which is induced in response to hypoxia and promotes undifferentiated cellular state, was also higher in Erdman-infected lungs. The granulomas generated by Erdman, and not CDC1551, infection appeared to have larger regions of necrosis, which is strongly associated with hypoxia. To better understand the mechanisms of differential hypoxia induction by these strains, we subjected both to hypoxia in vitro. Erdman induced higher concentrations of DosR regulon relative to CDC1551. The DosR regulon is the global regulator of response to hypoxia in Mtb and critical for its persistence in granulomas. Conclusions: Our results show that the response to hypoxia is a critical mediator of virulence determination in Mtb, with potential impacts on bacillary persistence, reactivation, and efficiency of therapeutics.


Asunto(s)
Mycobacterium tuberculosis , Animales , Granuloma , Hipoxia , Inflamación/patología , Pulmón/patología , Macaca mulatta , Mycobacterium tuberculosis/genética , Virulencia
9.
Nat Commun ; 13(1): 679, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35115549

RESUMEN

Emergence of mutant SARS-CoV-2 strains associated with an increased risk of COVID-19-related death necessitates better understanding of the early viral dynamics, host responses and immunopathology. Single cell RNAseq (scRNAseq) allows for the study of individual cells, uncovering heterogeneous and variable responses to environment, infection and inflammation. While studies have reported immune profiling using scRNAseq in terminal human COVID-19 patients, performing longitudinal immune cell dynamics in humans is challenging. Macaques are a suitable model of SARS-CoV-2 infection. Our longitudinal scRNAseq of bronchoalveolar lavage (BAL) cell suspensions from young rhesus macaques infected with SARS-CoV-2 (n = 6) demonstrates dynamic changes in transcriptional landscape 3 days post- SARS-CoV-2-infection (3dpi; peak viremia), relative to 14-17dpi (recovery phase) and pre-infection (baseline) showing accumulation of distinct populations of both macrophages and T-lymphocytes expressing strong interferon-driven inflammatory gene signature at 3dpi. Type I interferon response is induced in the plasmacytoid dendritic cells with appearance of a distinct HLADR+CD68+CD163+SIGLEC1+ macrophage population exhibiting higher angiotensin-converting enzyme 2 (ACE2) expression. These macrophages are significantly enriched in the lungs of macaques at 3dpi and harbor SARS-CoV-2 while expressing a strong interferon-driven innate anti-viral gene signature. The accumulation of these responses correlated with decline in viremia and recovery.


Asunto(s)
COVID-19/inmunología , Interferones/farmacología , Células Mieloides/inmunología , SARS-CoV-2/efectos de los fármacos , Animales , Antivirales , Lavado Broncoalveolar , Modelos Animales de Enfermedad , Humanos , Inmunidad Innata , Inflamación , Interferón Tipo I/genética , Interferón Tipo I/farmacología , Interferones/genética , Pulmón/inmunología , Pulmón/patología , Macaca mulatta , Macrófagos/inmunología , Linfocitos T/inmunología
10.
Nat Immunol ; 23(2): 318-329, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35058616

RESUMEN

Tuberculosis (TB) in humans is characterized by formation of immune-rich granulomas in infected tissues, the architecture and composition of which are thought to affect disease outcome. However, our understanding of the spatial relationships that control human granulomas is limited. Here, we used multiplexed ion beam imaging by time of flight (MIBI-TOF) to image 37 proteins in tissues from patients with active TB. We constructed a comprehensive atlas that maps 19 cell subsets across 8 spatial microenvironments. This atlas shows an IFN-γ-depleted microenvironment enriched for TGF-ß, regulatory T cells and IDO1+ PD-L1+ myeloid cells. In a further transcriptomic meta-analysis of peripheral blood from patients with TB, immunoregulatory trends mirror those identified by granuloma imaging. Notably, PD-L1 expression is associated with progression to active TB and treatment response. These data indicate that in TB granulomas, there are local spatially coordinated immunoregulatory programs with systemic manifestations that define active TB.


Asunto(s)
Granuloma/inmunología , Tuberculosis/inmunología , Antígeno B7-H1/inmunología , Células Cultivadas , Citocinas/inmunología , Perfilación de la Expresión Génica/métodos , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Pulmón/inmunología , Mycobacterium tuberculosis/inmunología , Células Mieloides/inmunología
11.
mBio ; 12(6): e0318921, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34903057

RESUMEN

Myeloid-derived suppressor cells (MDSCs) represent an innate immune cell population comprised of immature myeloid cells and myeloid progenitors with very potent immunosuppressive potential. MDSCs are reported to be abundant in the lungs of active tuberculosis (TB) patients. We sought to perform an in-depth study of MDSCs during latent TB infection (LTBI) and active TB (ATB) using the nonhuman primate (NHP) model of pulmonary TB. We found a higher proportion of granulocytic, polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) in the lungs of ATB animals compared to those with LTBI or naive control animals. Active disease in the lung, but not LTBI, was furthermore associated with higher proliferation, expansion, and immunosuppressive capabilities of PMN-MDSCs, as shown by enhanced expression of Ki67, indoleamine 2,3-dioxygenase (IDO1), interleukin-10 (IL-10), matrix metallopeptidase 9 (MMP-9), inducible nitric oxide synthase (iNOS), and programmed death-ligand 1 (PD-L1). These immunosuppressive PMN-MDSCs specifically localized to the lymphocytic cuff at the periphery of the granulomas in animals with ATB. Conversely, these cells were scarcely distributed in interstitial lung tissue and the inner core of granulomas. This spatial regulation suggests an important immunomodulatory role of PMN-MDSCs by restricting T cell access to the TB granuloma core and can potentially explain dysfunctional anti-TB responses in active granuloma. Our results raise the possibility that the presence of MDSCs can serve as a biomarker for ATB, while their disappearance can indicate successful therapy. Furthermore, MDSCs may serve as a potential target cell for adjunctive TB therapy. IMPORTANCE Myeloid cells are immunocytes of innate origin that orchestrate the first response toward pathogens via immune surveillance (uptake and killing), antigen presentation, and initiation of adaptive immunity by T cell stimulation. However, MDSCs are a subset of innate immunocytes that deviate to an immunoregulatory phenotype. MDSCs possess strong immunosuppressive capabilities that are induced in autoimmune, malignant neoplastic, and chronic inflammatory diseases. Induction of MDSCs has been found in peripheral blood, bronchoalveolar lavage (BAL) fluid, and pleural effusions of active TB patients, but their precise localization in lung tissue and in TB granulomas remains unclear due to challenges associated with sampling lungs and granulomas from active TB patients. Nonhuman primates (NHPs) are an important animal model with TB granulomas that closely mimic those found in humans and can therefore be used for studies that are otherwise challenging with patient material. Herein, we study MDSC localization in the lungs of NHPs exhibiting latent and active TB. Our findings reveal that MDSCs localize and exert their immunosuppressive roles at the periphery rather than in the core of TB granulomas.


Asunto(s)
Granuloma/inmunología , Tuberculosis Latente/inmunología , Células Supresoras de Origen Mieloide/inmunología , Linfocitos T/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Modelos Animales de Enfermedad , Femenino , Granuloma/microbiología , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Interleucina-10/genética , Interleucina-10/inmunología , Tuberculosis Latente/genética , Tuberculosis Latente/microbiología , Macaca mulatta , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/inmunología , Mycobacterium tuberculosis/fisiología , Tuberculosis Pulmonar/genética , Tuberculosis Pulmonar/microbiología
12.
Front Microbiol ; 12: 744800, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34721344

RESUMEN

Smoking is known to be an added risk factor for tuberculosis (TB), with nearly a quarter of the TB cases attributed to cigarette smokers in the 22 countries with the highest TB burden. Many studies have indicated a link between risk of active TB and cigarette smoke. Smoking is also known to significantly decrease TB cure and treatment completion rate and increase mortality rates. Cigarette smoke contains thousands of volatile compounds including carcinogens, toxins, reactive solids, and oxidants in both particulate and gaseous phase. Yet, to date, limited studies have analyzed the impact of cigarette smoke components on Mycobacterium tuberculosis (Mtb), the causative agent of TB. Here we report the impact of cigarette smoke condensate (CSC) on survival, mutation frequency, and gene expression of Mtb in vitro. We show that exposure of virulent Mtb to cigarette smoke increases the mutation frequency of the pathogen and strongly induces the expression of the regulon controlled by SigH-a global transcriptional regulator of oxidative stress. SigH has previously been shown to be required for Mtb to respond to oxidative stress, survival, and granuloma formation in vivo. A high-SigH expression phenotype is known to be associated with greater virulence of Mtb. In patients with pulmonary TB who smoke, these changes may therefore play an important, yet unexplored, role in the treatment efficacy by potentially enhancing the virulence of tubercle bacilli.

13.
Cell ; 184(7): 1757-1774.e14, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33761328

RESUMEN

The central pathogen-immune interface in tuberculosis is the granuloma, a complex host immune structure that dictates infection trajectory and physiology. Granuloma macrophages undergo a dramatic transition in which entire epithelial modules are induced and define granuloma architecture. In tuberculosis, relatively little is known about the host signals that trigger this transition. Using the zebrafish-Mycobacterium marinum model, we identify the basis of granuloma macrophage transformation. Single-cell RNA-sequencing analysis of zebrafish granulomas and analysis of Mycobacterium tuberculosis-infected macaques reveal that, even in the presence of robust type 1 immune responses, countervailing type 2 signals associate with macrophage epithelialization. We find that type 2 immune signaling, mediated via stat6, is absolutely required for epithelialization and granuloma formation. In mixed chimeras, stat6 acts cell autonomously within macrophages, where it is required for epithelioid transformation and incorporation into necrotic granulomas. These findings establish the signaling pathway that produces the hallmark structure of mycobacterial infection.


Asunto(s)
Granuloma/patología , Inmunidad/fisiología , Infecciones por Mycobacterium no Tuberculosas/patología , Animales , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Diferenciación Celular , Modelos Animales de Enfermedad , Células Epitelioides/citología , Células Epitelioides/inmunología , Células Epitelioides/metabolismo , Granuloma/inmunología , Granuloma/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Interferón gamma/metabolismo , Interleucina-12/metabolismo , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/metabolismo , Infecciones por Mycobacterium no Tuberculosas/inmunología , Mycobacterium marinum/aislamiento & purificación , Mycobacterium marinum/fisiología , Necrosis , ARN Guía de Kinetoplastida/metabolismo , Receptores de Interleucina-4/antagonistas & inhibidores , Receptores de Interleucina-4/genética , Receptores de Interleucina-4/metabolismo , Factor de Transcripción STAT6/antagonistas & inhibidores , Factor de Transcripción STAT6/genética , Factor de Transcripción STAT6/metabolismo , Transducción de Señal , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo
14.
Cell Host Microbe ; 29(2): 165-178.e8, 2021 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-33340449

RESUMEN

Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb) latently infects approximately one-fourth of the world's population. The immune mechanisms that govern progression from latent (LTBI) to active pulmonary TB (PTB) remain poorly defined. Experimentally Mtb-infected non-human primates (NHP) mirror the disease observed in humans and recapitulate both PTB and LTBI. We characterized the lung immune landscape in NHPs with LTBI and PTB using high-throughput technologies. Three defining features of PTB in macaque lungs include the influx of plasmacytoid dendritic cells (pDCs), an Interferon (IFN)-responsive macrophage population, and activated T cell responses. In contrast, a CD27+ Natural killer (NK) cell subset accumulated in the lungs of LTBI macaques. This NK cell population was also detected in the circulation of LTBI individuals. This comprehensive analysis of the lung immune landscape will improve the understanding of TB immunopathogenesis, providing potential targets for therapies and vaccines for TB control.


Asunto(s)
Células Dendríticas/inmunología , Células Asesinas Naturales/inmunología , Tuberculosis Latente/inmunología , Macrófagos/inmunología , Mycobacterium tuberculosis/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Humanos , Pulmón/citología , Pulmón/inmunología , Macaca mulatta , Tuberculosis Pulmonar/patología
15.
J Clin Invest ; 130(6): 3098-3112, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32134742

RESUMEN

Neutrophil accumulation is associated with lung pathology during active tuberculosis (ATB). However, the molecular mechanism or mechanisms by which neutrophils accumulate in the lung and contribute to TB immunopathology are not fully delineated. Using the well-established mouse model of TB, our new data provide evidence that the alarmin S100A8/A9 mediates neutrophil accumulation during progression to chronic TB. Depletion of neutrophils or S100A8/A9 deficiency resulted in improved Mycobacterium tuberculosis (Mtb) control during chronic but not acute TB. Mechanistically, we demonstrate that, following Mtb infection, S100A8/A9 expression is required for upregulation of the integrin molecule CD11b specifically on neutrophils, mediating their accumulation during chronic TB disease. These findings are further substantiated by increased expression of S100A8 and S100A9 mRNA in whole blood in human TB progressors when compared with nonprogressors and rapidly decreased S100A8/A9 protein levels in the serum upon TB treatment. Furthermore, we demonstrate that S100A8/A9 serum levels along with chemokines are useful in distinguishing between ATB and asymptomatic Mtb-infected latent individuals. Thus, our results support targeting S100A8/A9 pathways as host-directed therapy for TB.


Asunto(s)
Antígeno CD11b/inmunología , Calgranulina A/inmunología , Calgranulina B/inmunología , Mycobacterium tuberculosis/inmunología , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Tuberculosis/inmunología , Animales , Antígeno CD11b/genética , Calgranulina A/genética , Calgranulina B/genética , Ratones , Ratones Noqueados , Neutrófilos/patología , Tuberculosis/genética , Tuberculosis/patología , Tuberculosis/terapia
16.
J Infect Dis ; 221(10): 1636-1646, 2020 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-31832640

RESUMEN

Specific spatial organization of granulomas within the lungs is crucial for protective anti-tuberculosis (TB) immune responses. However, only large animal models such as macaques are thought to reproduce the morphological hallmarks of human TB granulomas. In this study, we show that infection of mice with clinical "hypervirulent" Mycobacterium tuberculosis (Mtb) HN878 induces human-like granulomas composed of bacilli-loaded macrophages surrounded by lymphocytes and organized localization of germinal centers and B-cell follicles. Infection with laboratory-adapted Mtb H37Rv resulted in granulomas that are characterized by unorganized clusters of macrophages scattered between lymphocytes. An in-depth exploration of the functions of B cells within these follicles suggested diverse roles and the activation of signaling pathways associated with antigen presentation and immune cell recruitment. These findings support the use of clinical Mtb HN878 strain for infection in mice as an appropriate model to study immune parameters associated with human TB granulomas.


Asunto(s)
Linfocitos B/fisiología , Granuloma/microbiología , Mycobacterium tuberculosis/clasificación , Mycobacterium tuberculosis/patogenicidad , Tuberculosis Pulmonar/microbiología , Animales , Granuloma/patología , Cadenas mu de Inmunoglobulina/genética , Cadenas mu de Inmunoglobulina/metabolismo , Pulmón/microbiología , Pulmón/patología , Linfocitos/fisiología , Macaca mulatta , Macrófagos/fisiología , Ratones Noqueados , Tuberculosis Pulmonar/patología , Virulencia
17.
Commun Biol ; 2: 349, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31552302

RESUMEN

Dormancy is a key characteristic of the intracellular life-cycle of Mtb. The importance of sensor kinase DosS in mycobacteria are attributed in part to our current findings that DosS is required for both persistence and full virulence of Mtb. Here we show that DosS is also required for optimal replication in macrophages and involved in the suppression of TNF-α and autophagy pathways. Silencing of these pathways during the infection process restored full virulence in MtbΔdosS mutant. Notably, a mutant of the response regulator DosR did not exhibit the attenuation in macrophages, suggesting that DosS can function independently of DosR. We identified four DosS targets in Mtb genome; Rv0440, Rv2859c, Rv0994, and Rv0260c. These genes encode functions related to hypoxia adaptation, which are not directly controlled by DosR, e.g., protein recycling and chaperoning, biosynthesis of molybdenum cofactor and nitrogen metabolism. Our results strongly suggest a DosR-independent role for DosS in Mtb.


Asunto(s)
Autofagosomas/metabolismo , Autofagosomas/microbiología , Proteínas Bacterianas/metabolismo , Mycobacterium tuberculosis/fisiología , Protamina Quinasa/metabolismo , Proteínas Quinasas/metabolismo , Tuberculosis/metabolismo , Tuberculosis/microbiología , Autofagosomas/inmunología , Autofagia , Proteínas Bacterianas/genética , Proteínas de Unión al ADN , Perfilación de la Expresión Génica , Silenciador del Gen , Interacciones Huésped-Patógeno/inmunología , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Mutación , Mycobacterium tuberculosis/enzimología , Fagocitos/inmunología , Fagocitos/metabolismo , Fagocitos/microbiología , Fosforilación , Protamina Quinasa/genética , Proteínas Quinasas/genética , Tuberculosis/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Virulencia
18.
J Infect Dis ; 220(12): 1989-1998, 2019 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-31412123

RESUMEN

BACKGROUND: Tuberculosis is caused by Mycobacterium tuberculosis. Recent emergence of multidrug-resistant (MDR) tuberculosis strains seriously threatens tuberculosis control and prevention. However, the role of macrophage multidrug resistance gene MDR1 on intracellular M. tuberculosis survival during antituberculosis drug treatment is not known. METHODS: We used the human monocyte-derived macrophages to study the role of M. tuberculosis in regulation of MDR1 and drug resistance. RESULTS: We discovered that M. tuberculosis infection increases the expression of macrophage MDR1 to extrude various chemical substances, including tuberculosis drugs, resulting in enhanced survival of intracellular M. tuberculosis. The pathway of regulation involves M. tuberculosis infection of macrophages and suppression of heat shock factor 1, a transcriptional regulator of MDR1 through the up-regulation of miR-431. Notably, nonpathogenic Mycobacterium smegmatis did not increase MDR1 expression, indicating active secretion of virulence factors in pathogenic M. tuberculosis contributing to this phenotype. Finally, inhibition of MDR1 improves antibiotic-mediated killing of M. tuberculosis. CONCLUSION: We report a novel finding that M. tuberculosis up-regulates MDR1 during infection, which limits the exposure of M. tuberculosis to sublethal concentrations of antimicrobials. This condition promotes M. tuberculosis survival and potentially enhances the emergence of resistant variants.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Regulación de la Expresión Génica , Macrófagos/metabolismo , Macrófagos/microbiología , Mycobacterium tuberculosis/fisiología , Tuberculosis/genética , Tuberculosis/microbiología , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Animales , Antituberculosos/farmacología , Antituberculosos/uso terapéutico , Modelos Animales de Enfermedad , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Macrófagos/inmunología , Ratones , MicroARNs/genética , Viabilidad Microbiana/efectos de los fármacos , Mycobacterium tuberculosis/efectos de los fármacos , Tuberculosis/tratamiento farmacológico , Tuberculosis/metabolismo , Tuberculosis Pulmonar/genética , Tuberculosis Pulmonar/metabolismo , Tuberculosis Pulmonar/microbiología , Tuberculosis Pulmonar/patología , Factores de Virulencia
19.
Proc Natl Acad Sci U S A ; 115(1): E62-E71, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29255022

RESUMEN

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.


Asunto(s)
Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Pulmón/inmunología , Mycobacterium tuberculosis/inmunología , Triptófano/inmunología , Tuberculoma/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Proliferación Celular , Granuloma/inmunología , Granuloma/patología , Pulmón/patología , Macaca mulatta , Macrófagos/inmunología , Macrófagos/patología , Mycobacterium tuberculosis/patogenicidad , Tuberculoma/patología , Tuberculosis Pulmonar/patología
20.
Am J Pathol ; 187(12): 2811-2820, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28935575

RESUMEN

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.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida del Simio/complicaciones , Vacunas contra la Tuberculosis/farmacología , Tuberculosis/prevención & control , Activación Viral/efectos de los fármacos , Administración por Inhalación , Animales , Coinfección , VIH , Macaca mulatta , Mycobacterium tuberculosis , Virus de la Inmunodeficiencia de los Simios/efectos de los fármacos , Virus de la Inmunodeficiencia de los Simios/fisiología , Tuberculosis/complicaciones , Vacunas Atenuadas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA