Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Front Microbiol ; 12: 757134, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34925266

RESUMEN

Mycobacterium tuberculosis (Mtb) bacilli readily aggregate. We previously reported that Mtb aggregates lead to phagocyte death and subsequent efficient replication in the dead infected cells. Here, we examined the transcriptional response of human monocyte derived macrophages to phagocytosis of aggregated Mtb relative to phagocytosis of non-aggregated single or multiple bacilli. Infection with aggregated Mtb led to an early upregulation of pro-inflammatory associated genes and enhanced TNFα signaling via the NFκB pathway. These pathways were significantly more upregulated relative to infection with single or multiple non-aggregated bacilli per cell. Phagocytosis of aggregates led to a decreased phagosome acidification on a per bacillus basis and increased phagocyte cell death, which was not observed when Mtb aggregates were heat killed prior to phagocytosis. Mtb aggregates, observed in a granuloma from a patient, were found surrounding a lesion cavity. These observations suggest that TB aggregation may be a mechanism for pathogenesis. They raise the possibility that aggregated Mtb, if spread from individual to individual, could facilitate increased inflammation, Mtb growth, and macrophage cell death, potentially leading to active disease, cell necrosis, and additional cycles of transmission.

2.
Commun Biol ; 4(1): 1256, 2021 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-34732811

RESUMEN

In vitro phagocytosis of Mycobacterium tuberculosis (Mtb) aggregates (Mtb-AG), rather than similar numbers of single bacilli (Mtb-SC), induces host macrophage death and favors bacterial growth. Here, we examined whether aggregation contributes to enhanced Mtb pathogenicity in vivo in rabbit lungs. Rabbits were exposed to infectious aerosols containing mainly Mtb-AG or Mtb-SC. The lung bacterial load, systemic immune response, histology, and immune cell composition were investigated over time. Genome-wide transcriptome analysis, cellular and tissue-level assays, and immunofluorescent imaging were performed on lung tissue to define and compare immune activation and pathogenesis between Mtb-AG and Mtb-SC infection. Lung bacillary loads, disease scores, lesion size, and structure were significantly higher in Mtb-AG than Mtb-SC infected animals. Differences in immune cell distribution and activation were noted in the lungs of the two groups of infected animals. Consistently larger lung granulomas with large aggregates of Mtb, extensive necrotic foci, and elevated matrix metalloproteases expression were observed in Mtb-AG infected rabbits. Our findings suggest that bacillary aggregation increases Mtb fitness for improved growth and accelerates lung inflammation and infected host cell death, thereby exacerbating disease pathology in the lungs.


Asunto(s)
Inmunidad Adaptativa , Interacciones Huésped-Patógeno , Inmunidad Innata , Enfermedades Pulmonares/inmunología , Mycobacterium tuberculosis/fisiología , Fagocitosis , Animales , Femenino , Enfermedades Pulmonares/microbiología , Conejos
3.
Prog Biophys Mol Biol ; 152: 6-9, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31626823

RESUMEN

Tuberculosis (TB) incidence is projected to decline at too slow a rate to meet the targets set at the UN high level meeting on ending TB, convened in New York in 2018. To understand the causes of the slow progress in TB control, the Bill and Melinda Gates Foundation supported a patient-pathway analysis that identified significant gaps with patients being "lost" at all points along the care cascade. Although each country differed in the reasons for failure, some commonalities were identified. Most striking was the failure to diagnose and report TB patients to a national registry making finding the missing patients a priority for TB control globally. The analysis also found that optimal use of existing tools will not accelerate the rate of decline in TB incidence sufficiently, necessitating the development of new and improved tools for TB control. Finally, it was recognized that gaps in our understanding of TB pathogenesis and protective immunity as well as limited knowledge about the host pathogen interactions in granulomas of infected tissues, hinder the progress needed to develop the new tools for improved TB control. A commitment of all governments to support research, develop new tools such as new diagnostics, better drug regimens and efficacious vaccines, and improving service delivery by TB control programs will be needed to accelerate the reduction in incidence and reduce disease burden globally.


Asunto(s)
Brotes de Enfermedades/prevención & control , Control de Infecciones/métodos , Tuberculosis/epidemiología , Tuberculosis/prevención & control , Antituberculosos/farmacología , Salud Global , Humanos , Incidencia , Mycobacterium tuberculosis/efectos de los fármacos , Tuberculosis/diagnóstico , Tuberculosis/terapia
4.
Artículo en Inglés | MEDLINE | ID: mdl-32010638

RESUMEN

Tuberculous meningitis (TBM) is the most devastating form of extrapulmonary Mycobacterium tuberculosis infection in humans. Severe inflammation and extensive tissue damage drive the morbidity and mortality of this manifestation of tuberculosis (TB). Antibiotic treatment is ineffective at curing TBM due to variable and incomplete drug penetration across the blood-brain barrier (BBB) and blood-cerebrospinal fluid (CSF) barriers. Adjunctive corticosteroid therapy, used to dampen the inflammation, and the pathologic manifestation of TBM, improves overall survival but does not entirely prevent the morbidity of the disease and has significant toxicities, including immune-suppression. The rabbit has served as a fit for purpose experimental model of human TBM since the early 1900s due to the similarity in the developmental processes of the brain, including neuronal development, myelination, and microglial functions between humans and rabbits. Consistent with the observations made in humans, proinflammatory cytokines, including TNF-α, play a critical role in the pathogenesis of TBM in rabbits focusing the attention on the utility of TNF-α inhibitors in treating the disease. Thalidomide, an inhibitor of monocyte-derived TNF-α, was evaluated in the rabbit model of TBM and shown to improve survival and reduce inflammation of the brain and the meninges. Clinical studies in humans have also shown a beneficial response to thalidomide. However, the teratogenicity and T-cell activation function of the drug limit the use of thalidomide in the clinic. Thus, new drugs with more selective anti-inflammatory properties and a better safety profile are being developed. Some of these candidate drugs, such as phosphodiesterase-4 inhibitors, have been shown to reduce the morbidity and increase the survival of rabbits with TBM. Future studies are needed to assess the beneficial effects of these drugs for their potential to improve the current treatment strategy for TBM in humans.


Asunto(s)
Antituberculosos/uso terapéutico , Inhibidores de Fosfodiesterasa 4/uso terapéutico , Talidomida/uso terapéutico , Tuberculosis Meníngea/tratamiento farmacológico , Animales , Barrera Hematoencefálica/patología , Encéfalo/patología , Citocinas , Modelos Animales de Enfermedad , Humanos , Inflamación/patología , Activación de Linfocitos/efectos de los fármacos , Morbilidad , Conejos , Tuberculosis Meníngea/líquido cefalorraquídeo , Tuberculosis Meníngea/fisiopatología , Factor de Necrosis Tumoral alfa
5.
Mucosal Immunol ; 11(6): 1727-1742, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30115997

RESUMEN

C-C motif chemokine receptor 2 (CCR2) is a major chemokine axis that recruits myeloid cells including monocytes and macrophages. Thus far, CCR2-/- mice have not been found to be susceptible to infection with Mycobacterium tuberculosis (Mtb). Here, using a prototype W-Beijing family lineage 2 Mtb strain, HN878, we show that CCR2-/- mice exhibit increased susceptibility to tuberculosis (TB). Following exposure to Mtb HN878, alveolar macrophages (AMs) are amongst the earliest cells infected. We show that AMs accumulate early in the airways following infection and express CCR2. During disease progression, CCR2-expressing AMs exit the airways and localize within the TB granulomas. RNA-sequencing of sorted airway and non-airway AMs from infected mice show distinct gene expression profiles, suggesting that upon exit from airways and localization within granulomas, AMs become classically activated. The absence of CCR2+ cells specifically at the time of AM egress from the airways resulted in enhanced susceptibility to Mtb infection. Furthermore, infection with an Mtb HN878 mutant lacking phenolic glycolipid (PGL) expression still resulted in increased susceptibility in CCR2-/- mice. Together, these data show a novel role for CCR2 in protective immunity against clinically relevant Mtb infections.


Asunto(s)
Granuloma/inmunología , Macrófagos/inmunología , Mycobacterium tuberculosis/fisiología , Receptores CCR2/metabolismo , Tuberculosis/inmunología , Animales , Movimiento Celular , Quimiocina CCL2/metabolismo , Predisposición Genética a la Enfermedad , Humanos , Pulmón/patología , Activación de Macrófagos/genética , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mycobacterium tuberculosis/patogenicidad , Receptores CCR2/genética , Transcriptoma , Virulencia
6.
Exp Mol Med ; 50(5): 1-10, 2018 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-29789565

RESUMEN

Studies using the European rabbit Oryctolagus cuniculus contributed to elucidating numerous fundamental aspects of antibody structure and diversification mechanisms and continue to be valuable for the development and testing of therapeutic humanized polyclonal and monoclonal antibodies. Additionally, during the last two decades, the use of the European rabbit as an animal model has been increasingly extended to many human diseases. This review documents the continuing wide utility of the rabbit as a reliable disease model for development of therapeutics and vaccines and studies of the cellular and molecular mechanisms underlying many human diseases. Examples include syphilis, tuberculosis, HIV-AIDS, acute hepatic failure and diseases caused by noroviruses, ocular herpes, and papillomaviruses. The use of rabbits for vaccine development studies, which began with Louis Pasteur's rabies vaccine in 1881, continues today with targets that include the potentially blinding HSV-1 virus infection and HIV-AIDS. Additionally, two highly fatal viral diseases, rabbit hemorrhagic disease and myxomatosis, affect the European rabbit and provide unique models to understand co-evolution between a vertebrate host and viral pathogens.


Asunto(s)
Modelos Animales de Enfermedad , Animales , Evolución Biológica , Humanos , Sistema Inmunológico/fisiología , Inmunidad , Conejos
7.
JCI Insight ; 2(19)2017 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-28978810

RESUMEN

Mycobacterium tuberculosis (Mtb) is a global health threat, compounded by the emergence of drug-resistant strains. A hallmark of pulmonary tuberculosis (TB) is the formation of hypoxic necrotic granulomas, which upon disintegration, release infectious Mtb. Furthermore, hypoxic necrotic granulomas are associated with increased disease severity and provide a niche for drug-resistant Mtb. However, the host immune responses that promote the development of hypoxic TB granulomas are not well described. Using a necrotic Mtb mouse model, we show that loss of Mtb virulence factors, such as phenolic glycolipids, decreases the production of the proinflammatory cytokine IL-17 (also referred to as IL-17A). IL-17 production negatively regulates the development of hypoxic TB granulomas by limiting the expression of the transcription factor hypoxia-inducible factor 1α (HIF1α). In human TB patients, HIF1α mRNA expression is increased. Through genotyping and association analyses in human samples, we identified a link between the single nucleotide polymorphism rs2275913 in the IL-17 promoter (-197G/G), which is associated with decreased IL-17 production upon stimulation with Mtb cell wall. Together, our data highlight a potentially novel role for IL-17 in limiting the development of hypoxic necrotic granulomas and reducing disease severity in TB.


Asunto(s)
Granuloma/inmunología , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Interleucina-17/inmunología , Tuberculosis Pulmonar/inmunología , Adulto , Anciano , Animales , Hipoxia de la Célula/inmunología , Femenino , Regulación de la Expresión Génica/inmunología , Granuloma/microbiología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Mediadores de Inflamación/metabolismo , Interleucina-17/biosíntesis , Masculino , Ratones Endogámicos , Persona de Mediana Edad , ARN Mensajero/genética , Tuberculosis Pulmonar/complicaciones , Adulto Joven
8.
PLoS One ; 12(5): e0176660, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28464011

RESUMEN

OBJECTIVE: To identify plasma markers predictive of therapeutic response in patients with multidrug resistant tuberculosis (MDR-TB). METHODS: Fifty HIV-negative patients with active pulmonary MDR-TB were analysed for six soluble analytes in plasma at the time of initiating treatment (baseline) and over six months thereafter. Patients were identified as sputum culture positive or negative at baseline. Culture positive patients were further stratified by the median time to sputum culture conversion (SCC) as fast responders (< 76 days) or slow responders (≥ 76 days). Chest X-ray scores, body mass index, and sputum smear microscopy results were obtained at baseline. RESULTS: Unsupervised hierarchical clustering revealed that baseline plasma levels of IP-10/CXCL10, VEGF-A, SAA and CRP could distinguish sputum culture and cavitation status of patients. Among patients who were culture positive at baseline, there were significant positive correlations between plasma levels of CRP, SAA, VEGF-A, sIL-2Rα/CD40, and IP-10 and delayed SCC. Using linear discriminant analysis (LDA) and Receiver Operating Curves (ROC), we showed that a combination of MCP-1/CCL2, IP-10, sIL-2Rα, SAA, CRP and AFB smear could distinguish fast from slow responders and were predictive of delayed SCC with high sensitivity and specificity. CONCLUSION: Plasma levels of specific chemokines and inflammatory markers measured before MDR-TB treatment are candidate predictive markers of delayed SCC. These findings require validation in a larger study.


Asunto(s)
Antituberculosos/uso terapéutico , Tuberculosis Resistente a Múltiples Medicamentos/tratamiento farmacológico , Adulto , Biomarcadores/sangre , Proteína C-Reactiva/análisis , Quimiocina CXCL10/sangre , Femenino , Humanos , Masculino , Pruebas de Sensibilidad Microbiana , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Proteína Amiloide A Sérica/análisis , Tuberculosis Resistente a Múltiples Medicamentos/sangre , Tuberculosis Resistente a Múltiples Medicamentos/inmunología , Factor A de Crecimiento Endotelial Vascular/sangre
9.
EBioMedicine ; 4: 104-14, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26981575

RESUMEN

OBJECTIVES: Adjunctive host-directed therapy is emerging as a new potential approach to improve the outcome of conventional antimicrobial treatment for tuberculosis (TB). We tested the ability of a phosphodiesterase-4 inhibitor (PDE4i) CC-11050, co-administered with the first-line anti-TB drug isoniazid (INH), to accelerate bacillary killing and reduce chronic inflammation in the lungs of rabbits with experimental Mycobacterium tuberculosis (Mtb) infection. METHODS: A rabbit model of pulmonary TB that recapitulates the pathologic manifestations seen in humans was used. Rabbits were infected with virulent Mtb by aerosol exposure and treated for eight weeks with INH with or without CC-11050, starting at four weeks post infection. The effect of CC-11050 treatment on disease severity, pathology, bacillary load, T cell proliferation and global lung transcriptome profiles were analyzed. RESULTS: Significant improvement in bacillary clearance and reduced lung pathology and fibrosis were noted in the rabbits treated for eight weeks with INH + CC-11050, compared to those treated with INH or CC-11050 only. In addition, expression of host genes associated with tissue remodeling, tumor necrosis factor alpha (TNF-α) regulation, macrophage activation and lung inflammation networks was dampened in CC-11050-treated, compared to the untreated rabbits. CONCLUSIONS: Adjunctive CC-11050 therapy significantly improves the response of rabbits with experimental pulmonary TB to INH treatment. We propose that CC-11050 may be a promising candidate for host directed therapy of patients with pulmonary TB, reducing the duration and improving clinical outcome of antibiotic treatment.


Asunto(s)
Antituberculosos/uso terapéutico , Isoniazida/uso terapéutico , Inhibidores de Fosfodiesterasa 4/uso terapéutico , Tuberculosis Pulmonar/tratamiento farmacológico , Animales , Antituberculosos/administración & dosificación , Combinación de Medicamentos , Sinergismo Farmacológico , Femenino , Isoniazida/administración & dosificación , Inhibidores de Fosfodiesterasa 4/administración & dosificación , Conejos
10.
PLoS One ; 10(7): e0132249, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26133981

RESUMEN

The formation and maintenance of granulomas is central to the host response to Mycobacterium tuberculosis (Mtb) infection. It is widely accepted that the lungs of patients with tuberculosis (TB) usually contain multiple infection foci, and that the granulomas evolve and differentiate independently, resulting in considerable heterogeneity. Although gene expression profiles of human blood cells have been proposed as biomarkers of Mtb infection and/or active disease, the immune profiles of discrete lesion types has not been studied extensively. Using histology, immunopathology and genome-wide transcriptome analysis, we explored the immunological profile of human lung TB granulomas. We show that although the different granulomas share core similarities in their immunological/inflammatory characteristics, they also exhibit significant divergence. Despite similar numbers of CD68+ macrophages in the different lesions, the extent of immune reactivity, as determined by the density of CD3+ T cells in the macrophage rich areas, and the extent of fibrosis, shows considerable variation. Both quantitative and qualitative differences among significantly differentially expressed genes (SDEG) were noted in each of the lesion types studied. Further, network/pathway analysis of SDEG revealed differential regulation of inflammatory response, immune cell trafficking, and cell mediated immune response in the different lesions. Our data highlight the formidable challenges facing ongoing efforts to identify peripheral blood biomarkers due to the diversity of lesion types and complexity of local immune responses in the lung.


Asunto(s)
Granuloma del Sistema Respiratorio/patología , Pulmón/patología , Tuberculosis Pulmonar/patología , Microambiente Celular , Fibrosis , Perfilación de la Expresión Génica , Granuloma del Sistema Respiratorio/genética , Granuloma del Sistema Respiratorio/inmunología , Humanos , Inflamación , Interleucina-7/fisiología , Activación de Linfocitos , Macrófagos/inmunología , Necrosis , Proyectos Piloto , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Mensajero/aislamiento & purificación , Receptores de Calcitriol/fisiología , Factor de Transcripción STAT1/fisiología , Transducción de Señal , Subgrupos de Linfocitos T/inmunología , Transcriptoma , Tuberculosis Resistente a Múltiples Medicamentos/genética , Tuberculosis Resistente a Múltiples Medicamentos/inmunología , Tuberculosis Resistente a Múltiples Medicamentos/patología , Tuberculosis Pulmonar/genética , Tuberculosis Pulmonar/inmunología
11.
J Immunol Methods ; 417: 22-33, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25523923

RESUMEN

BACKGROUND: Qualified or validated assays are essential in clinical trials. Short-term stimulation of whole blood and intracellular cytokine staining assay is commonly used to measure immunogenicity in tuberculosis vaccine clinical trials. Previously, the short-term stimulation process of whole blood with BCG was optimized. We aimed to qualify the intracellular cytokine staining process and assess the effects of long-term cryopreservation. Our hypotheses were that the assay is robust in the measurement of the mycobacteria-specific T cells, and long-term cryopreservation of fixed cells from stimulated whole blood would not compromise reliable measurement of mycobacteria induced CD4 T cell immunity. METHODS: Whole blood from healthy adults was collected in sodium heparinized tubes. The blood was left unstimulated or stimulated with mycobacterial antigens or mitogens for 12h. Cells were harvested, fixed and multiple aliquots from each participant cryopreserved. Later, mycobacteria-specific CD4 and CD8 T cells expressing IFN-γ, TNF-α, IL-2 and IL-17 were quantitated by flow cytometry. Assay performance characteristics evaluated included limit of quantification and detection, reproducibility, precision, robustness, specificity and sensitivity. To assess the effects of long-term cryopreservation, fixed cells from the stimulated bloods were analysed one week post-cryopreservation and at 3-month intervals over a 3-year period. RESULTS: The limit of quantification for the different cytokines was variable: 0.04% for frequencies of IFN-γ- and IL-2-expressing T cells and less than 0.01% for TNF-α- and IL-17-expressing T cells. When measurement of the mycobacteria-specific T cells was assessed at levels above the detection limit, the whole blood intracellular cytokine assay showed high precision that was operator-independent. The assay was also robust: variation in staining conditions including temperature (4 °C or 20-23 °C) and time (45, 60 or 90 min) did not markedly affect quantification of specific T cells. Finally, prolonged periods of cryopreservation also did not significantly influence quantification of mycobacteria-specific CD4 T cells. CONCLUSIONS: The whole blood intracellular cytokine assay is robust and reliable in quantification of the mycobacteria-specific T cells and is not significantly affected by cryopreservation of fixed cells.


Asunto(s)
Vacuna BCG/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Citocinas/sangre , Mycobacterium bovis/inmunología , Tuberculosis Pulmonar/inmunología , Adulto , Antígenos Bacterianos/inmunología , Criopreservación , Citocinas/inmunología , Citometría de Flujo , Humanos , Interferón gamma/inmunología , Interleucina-17/inmunología , Interleucina-2/inmunología , Límite de Detección , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Coloración y Etiquetado/métodos , Tuberculosis Pulmonar/diagnóstico , Factor de Necrosis Tumoral alfa/inmunología
12.
Sci Transl Med ; 6(263): 263ra159, 2014 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-25411472

RESUMEN

The global burden of tuberculosis (TB) morbidity and mortality remains immense. A potential new approach to TB therapy is to augment protective host immune responses. We report that the antidiabetic drug metformin (MET) reduces the intracellular growth of Mycobacterium tuberculosis (Mtb) in an AMPK (adenosine monophosphate-activated protein kinase)-dependent manner. MET controls the growth of drug-resistant Mtb strains, increases production of mitochondrial reactive oxygen species, and facilitates phagosome-lysosome fusion. In Mtb-infected mice, use of MET ameliorated lung pathology, reduced chronic inflammation, and enhanced the specific immune response and the efficacy of conventional TB drugs. Moreover, in two separate human cohorts, MET treatment was associated with improved control of Mtb infection and decreased disease severity. Collectively, these data indicate that MET is a promising candidate host-adjunctive therapy for improving the effective treatment of TB.


Asunto(s)
Metformina/uso terapéutico , Mycobacterium tuberculosis/efectos de los fármacos , Tuberculosis/tratamiento farmacológico , Humanos , Metformina/farmacología , Pruebas de Sensibilidad Microbiana , Mycobacterium tuberculosis/crecimiento & desarrollo , Especies Reactivas de Oxígeno/metabolismo , Tuberculosis/inmunología
13.
PLoS One ; 9(7): e102178, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25048802

RESUMEN

We investigated 18 HIV-negative patients with MDR-TB for M. tuberculosis (Mtb)- and PPD-specific CD4 T cell responses and followed them over 6 months of drug therapy. Twelve of these patients were sputum culture (SC) positive and six patients were SC negative upon enrollment. Our aim was to identify a subset of mycobacteria-specific CD4 T cells that would predict time to culture conversion. The total frequency of mycobacteria-specific CD4 T cells at baseline could not distinguish patients showing positive or negative SC. However, a greater proportion of late-differentiated (LD) Mtb- and PPD-specific memory CD4 T cells was found in SC positive patients than in those who were SC negative (p = 0.004 and p = 0.0012, respectively). Similarly, a higher co-expression of HLA-DR+ Ki67+ on Mtb- and PPD-specific CD4 T cells could also discriminate between sputum SC positive versus SC negative (p = 0.004 and p = 0.001, respectively). Receiver operating characteristic (ROC) analysis revealed that baseline levels of Ki67+ HLA-DR+ Mtb- and PPD-specific CD4 T cells were predictive of the time to sputum culture conversion, with area-under-the-curve of 0.8 (p = 0.027). Upon treatment, there was a significant decline of these Ki67+ HLA-DR+ T cell populations in the first 2 months, with a progressive increase in mycobacteria-specific polyfunctional IFNγ+ IL2+ TNFα+ CD4 T cells over 6 months. Thus, a subset of activated and proliferating mycobacterial-specific CD4 T cells (Ki67+ HLA-DR+) may provide a valuable marker in peripheral blood that predicts time to sputum culture conversion in TB patients at the start of treatment.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Mycobacterium tuberculosis/inmunología , Esputo/microbiología , Tuberculosis/inmunología , Adulto , Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos/efectos de los fármacos , Estudios de Cohortes , Femenino , VIH/aislamiento & purificación , Infecciones por VIH/diagnóstico , Humanos , Masculino , Persona de Mediana Edad , Mycobacterium tuberculosis/efectos de los fármacos , Esputo/efectos de los fármacos , Esputo/inmunología , Tuberculosis/tratamiento farmacológico
14.
J Interferon Cytokine Res ; 34(9): 716-26, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24831609

RESUMEN

Treatment of chronic inflammatory diseases with tumor necrosis factor alpha (TNF-α) antagonists has been associated with increased risk of tuberculosis (TB). We examined the usefulness of the rabbit model of active pulmonary TB for studying the impact of the human immune modulatory reagent etanercept on the host immune response. Control of Mycobacterium tuberculosis (Mtb) infection, disease pathology, and the global transcriptional response in Mtb-infected lungs of rabbits were studied. Etanercept treatment exacerbated disease pathology and reduced bacillary control in the lungs, compared with infected untreated animals. Reduced collagen and fibrin deposition in the granulomas was associated with significant downregulation of the collagen metabolism and fibrosis network genes and upregulation of genes in the inflammatory response and cell recruitment networks in the lungs of etanercept treated, compared with untreated rabbits. Our results suggest that targeting the TNF-α signaling pathway disrupts the tissue remodeling process, which is required for the formation and maintenance of well-differentiated granulomas and for control of Mtb growth in the lungs. These results validate the use of the rabbit model for investigating the impact of selected human immune modulatory drugs, such as a TNF-α antagonist, on the host immune response and pathogenesis in TB.


Asunto(s)
Etanercept/inmunología , Etanercept/farmacología , Inflamación/patología , Tuberculosis Pulmonar/patología , Animales , Colágeno/inmunología , Modelos Animales de Enfermedad , Regulación hacia Abajo/inmunología , Etanercept/efectos adversos , Fibrina/inmunología , Granuloma/inmunología , Granuloma/microbiología , Inflamación/inmunología , Inflamación/microbiología , Pulmón/inmunología , Pulmón/microbiología , Mycobacterium tuberculosis/inmunología , Conejos , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/microbiología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/inmunología , Regulación hacia Arriba/inmunología
15.
BMC Infect Dis ; 13: 484, 2013 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-24134504

RESUMEN

BACKGROUND: Previous studies have shown that Mycobacterium tuberculosis (MTB) Uganda family, a sub-lineage of the MTB Lineage 4, is the main cause of tuberculosis (TB) in Uganda. Using a well characterized patient population, this study sought to determine whether there are clinical and patient characteristics associated with the success of the MTB Uganda family in Kampala. METHODS: A total of 1,746 MTB clinical isolates collected from 1992-2009 in a household contact study were genotyped. Genotyping was performed using Single Nucleotide Polymorphic (SNP) markers specific for the MTB Uganda family, other Lineage 4 strains, and Lineage 3, respectively. Out of 1,746 isolates, 1,213 were from patients with detailed clinical data. These data were used to seek associations between MTB lineage/sub-lineage and patient phenotypes. RESULTS: Three MTB lineages were found to dominate the MTB population in Kampala during the last two decades. Overall, MTB Uganda accounted for 63% (1,092/1,746) of all cases, followed by other Lineage 4 strains accounting for 22% (394/1,746), and Lineage 3 for 11% (187/1,746) of cases, respectively. Seventy-three (4 %) strains remained unclassified. Our longitudinal data showed that MTB Uganda family occurred at the highest frequency during the whole study period, followed by other Lineage 4 strains and Lineage 3. To explore whether the long-term success of MTB Uganda family was due to increased virulence, we used cavitary disease as a proxy, as this form of TB is the most transmissible. Multivariate analysis revealed that even though cavitary disease was associated with known risk factors such as smoking (adjusted odds ratio (aOR) 4.8, 95% confidence interval (CI) 3.33-6.84) and low income (aOR 2.1, 95% CI 1.47-3.01), no association was found between MTB lineage and cavitary TB. CONCLUSION: The MTB Uganda family has been dominating in Kampala for the last 18 years, but this long-term success is not due to increased virulence as defined by cavitary disease.


Asunto(s)
Mycobacterium tuberculosis/clasificación , Tuberculosis/microbiología , Adulto , Femenino , Humanos , Masculino , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/aislamiento & purificación , Mycobacterium tuberculosis/patogenicidad , Fenotipo , Polimorfismo de Nucleótido Simple , Estudios Prospectivos , Tuberculosis/epidemiología , Uganda/epidemiología
16.
Cell Commun Signal ; 11: 60, 2013 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-23958185

RESUMEN

BACKGROUND: Pulmonary infection of humans by Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), results in active disease in 5-10% of individuals, while asymptomatic latent Mtb infection (LTBI) is established in the remainder. The host immune responses that determine this differential outcome following Mtb infection are not fully understood. Using a rabbit model of pulmonary TB, we have shown that infection with the Mtb clinical isolate HN878 (a hyper-virulent W-Beijing lineage strain) leads to progressive cavitary disease similar to what is seen in humans with active TB. In contrast, infection with Mtb CDC1551 (a hyper-immunogenic clinical isolate) is efficiently controlled in rabbit lungs, with establishment of LTBI, which can be reactivated upon treatment with immune-suppressive drugs. We hypothesize that the initial interaction of Mtb with the cells of the host response in the lungs determine later outcome of infection. RESULTS: To test this hypothesis, we used our rabbit model of pulmonary TB and infected the animals with Mtb HN878 or CDC1551. At 3 hours, with similar lung bacillary loads, HN878 infection caused greater accumulation of mononuclear and polymorphonuclear leukocytes (PMN) in the lungs, compared to animals infected with CDC1551. Using whole-genome microarray gene expression analysis, we delineated the early transcriptional changes in the lungs of HN878- or CDC1551-infected rabbits at this time and compared them to the differential response at 4 weeks of Mtb-infection. Our gene network and pathway analysis showed that the most significantly differentially expressed genes involved in the host response to HN878, compared to CDC1551, at 3 hours of infection, were components of the inflammatory response and STAT1 activation, recruitment and activation of macrophages, PMN, and fMLP (N-formyl-Methionyl-Leucyl-Phenylalanine)-stimulation. At 4 weeks, the CDC1551 bacillary load was significantly lower and the granulomatous response reduced compared to HN878 infection. Moreover, although inflammation was dampened in both Mtb infections at 4 weeks, the majority of the differentially expressed gene networks were similar to those seen at 3 hours. CONCLUSIONS: We propose that differential regulation of the inflammation-associated innate immune response and related gene expression changes seen at 3 hours determine the long term outcome of Mtb infection in rabbit lungs.


Asunto(s)
Inmunidad Innata , Tuberculosis Pulmonar/inmunología , Animales , Inflamación/inmunología , Leucocitos Mononucleares/metabolismo , Macrófagos/metabolismo , Mycobacterium tuberculosis , Neutrófilos/metabolismo , Conejos , Factor de Transcripción STAT1/metabolismo , Factores de Tiempo , Transcriptoma , Tuberculosis Pulmonar/microbiología
17.
Proc Natl Acad Sci U S A ; 110(32): E2997-3006, 2013 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-23882081

RESUMEN

Macrophage migration inhibitory factor (MIF), an innate cytokine encoded in a functionally polymorphic genetic locus, contributes to detrimental inflammation but may be crucial for controlling infection. We explored the role of variant MIF alleles in tuberculosis. In a Ugandan cohort, genetic low expressers of MIF were 2.4-times more frequently identified among patients with Mycobacterium tuberculosis (TB) bacteremia than those without. We also found mycobacteria-stimulated transcription of MIF and serum MIF levels to be correlated with MIF genotype in human macrophages and in a separate cohort of US TB patients, respectively. To determine mechanisms for MIF's protective role, we studied both aerosolized and i.v. models of mycobacterial infection and observed MIF-deficient mice to succumb more quickly with higher organism burden, increased lung pathology, and decreased innate cytokine production (TNF-α, IL-12, IL-10). MIF-deficient animals showed increased pulmonary neutrophil accumulation but preserved adaptive immune response. MIF-deficient macrophages demonstrated decreased cytokine and reactive oxygen production and impaired mycobacterial killing. Transcriptional investigation of MIF-deficient macrophages revealed reduced expression of the pattern recognition receptor dectin-1; restoration of dectin-1 expression recovered innate cytokine production and mycobacterial killing. Our data place MIF in a crucial upstream position in the innate immune response to mycobacteria and suggest that commonly occurring low expression MIF alleles confer an increased risk of TB disease in some populations.


Asunto(s)
Inmunidad Innata/inmunología , Factores Inhibidores de la Migración de Macrófagos/inmunología , Mycobacterium tuberculosis/inmunología , Tuberculosis/inmunología , Adulto , Animales , Línea Celular , Citocinas/inmunología , Citocinas/metabolismo , Femenino , Expresión Génica/inmunología , Genotipo , Humanos , Inmunidad Innata/genética , Lectinas Tipo C/genética , Lectinas Tipo C/inmunología , Lectinas Tipo C/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/microbiología , Factores Inhibidores de la Migración de Macrófagos/sangre , Factores Inhibidores de la Migración de Macrófagos/genética , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/inmunología , Neutrófilos/metabolismo , Polimorfismo Genético , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tasa de Supervivencia , Tuberculosis/genética , Tuberculosis/mortalidad , Uganda , Adulto Joven
18.
Semin Immunopathol ; 35(5): 563-83, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23864058

RESUMEN

Tuberculosis (TB) remains one of the greatest threats to human health. The causative bacterium, Mycobacterium tuberculosis (Mtb), is acquired by the respiratory route. It is exquisitely human adapted and a prototypic intracellular pathogen of macrophages, with alveolar macrophages (AMs) being the primary conduit of infection and disease. The outcome of primary infection is most often a latently infected healthy human host, in whom the bacteria are held in check by the host immune response. Such individuals can develop active TB later in life with impairment in the immune system. In contrast, in a minority of infected individuals, the host immune response fails to control the growth of bacilli, and progressive granulomatous disease develops, facilitating spread of the bacilli via infectious aerosols coughed out into the environment and inhaled by new hosts. The molecular details of the Mtb-macrophage interaction continue to be elucidated. However, it is clear that a number of complex processes are involved at the different stages of infection that may benefit either the bacterium or the host. Macrophages demonstrate tremendous phenotypic heterogeneity and functional plasticity which, depending on the site and stage of infection, facilitate the diverse outcomes. Moreover, host responses vary depending on the specific characteristics of the infecting Mtb strain. In this chapter, we describe a contemporary view of the behavior of AMs and their interaction with various Mtb strains in generating unique immunologic lung-specific responses.


Asunto(s)
Macrófagos/inmunología , Macrófagos/microbiología , Tuberculosis/inmunología , Tuberculosis/transmisión , Células Dendríticas/inmunología , Granuloma/inmunología , Humanos , Pulmón/citología , Pulmón/inmunología , Pulmón/microbiología , Mycobacterium tuberculosis/inmunología
19.
J Infect Dis ; 207(7): 1084-94, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23293360

RESUMEN

BACKGROUND: Improved vaccination strategies against tuberculosis are needed, such as approaches to boost immunity induced by the current vaccine, BCG. Design of these strategies has been hampered by a lack of knowledge of the kinetics of the human host response induced by neonatal BCG vaccination. Furthermore, the functional and phenotypic attributes of BCG-induced long-lived memory T-cell responses remain unclear. METHODS: We assessed the longitudinal CD4 T-cell response following BCG vaccination of human newborns. The kinetics, function, and phenotype of these cells were measured using flow cytometric whole-blood assays. RESULTS: We showed that the BCG-specific CD4 T-cell response peaked 6-10 weeks after vaccination and gradually waned over the first year of life. Highly activated T-helper 1 cells, predominantly expressing interferon γ, tumor necrosis factor α, and/or interleukin 2, were present at the peak response. Following contraction, BCG-specific CD4 T cells expressed high levels of Bcl-2 and displayed a predominant CD45RACCR7 central memory phenotype. However, cytokine and cytotoxic marker expression by these cells was more characteristic of effector memory cells. CONCLUSIONS: Our findings suggest that boosting of BCG-primed CD4 T cells with heterologous tuberculosis vaccines may be best after 14 weeks of age, once an established memory response has developed.


Asunto(s)
Vacuna BCG/inmunología , Linfocitos T CD4-Positivos/inmunología , Memoria Inmunológica , Recién Nacido/inmunología , Vacunación/métodos , Vacuna BCG/administración & dosificación , Biomarcadores/sangre , Estudios Transversales , Femenino , Citometría de Flujo , Humanos , Interferón gamma/sangre , Interleucina-2/sangre , Estudios Longitudinales , Activación de Linfocitos , Masculino , Mycobacterium tuberculosis/inmunología , Fenotipo , Factores de Tiempo , Resultado del Tratamiento , Tuberculosis/inmunología , Tuberculosis/microbiología , Tuberculosis/terapia , Factor de Necrosis Tumoral alfa/sangre
20.
Am J Pathol ; 181(5): 1711-24, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22960076

RESUMEN

Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), is an exquisitely adapted human pathogen capable of surviving for decades in the lungs of immune-competent individuals in the absence of disease. The World Health Organization estimates that 2 billion people have latent TB infection (LTBI), defined by a positive immunological response to Mtb antigens, with no clinical signs of disease. A better understanding of host and pathogen determinants of LTBI and subsequent reactivation would benefit TB control efforts. Animal models of LTBI have been hampered generally by an inability to achieve complete bacillary clearance. Herein, we have characterized a rabbit model of LTBI in which, similar to most humans, complete clearance of pulmonary Mtb infection and pathological characteristics occurs spontaneously. The evidence that Mtb-CDC1551-infected rabbits achieve LTBI, rather than sterilization, is based on the ability of the bacilli to be reactivated after immune suppression. These rabbits showed early activation of T cells and macrophages and an early peak in the TNFα level, which decreased in association with clearance of bacilli from the lungs. In the absence of sustained tumor necrosis factor-α production, no necrosis was seen in the evolving lung granulomas. In addition, bacillary control was associated with down-regulation of several metalloprotease genes and an absence of lung fibrosis. This model will be used to characterize molecular markers of protective immunity and reactivation.


Asunto(s)
Tuberculosis Latente/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Carga Bacteriana/inmunología , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Tuberculosis Latente/genética , Tuberculosis Latente/microbiología , Tuberculosis Latente/patología , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Activación de Macrófagos/genética , Activación de Macrófagos/inmunología , Mycobacterium tuberculosis/crecimiento & desarrollo , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/microbiología , Fibrosis Pulmonar/patología , Conejos , Transducción de Señal/genética , Bazo/inmunología , Bazo/microbiología , Linfocitos T/inmunología , Transcripción Genética , Tuberculosis Pulmonar/genética , Tuberculosis Pulmonar/microbiología , Tuberculosis Pulmonar/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA