Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Infect Dis ; 219(5): 685-694, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30376080

RESUMEN

Latent tuberculosis has been recognized for over a century, but discovery of new niches, where Mycobacterium tuberculosis resides, continues. We evaluated literature on M.tuberculosis locations during latency, highlighting that mesenchymal and hematopoietic stem cells harbor organisms in sensitized asymptomatic individuals.


Asunto(s)
Células Madre Hematopoyéticas/microbiología , Tuberculosis Latente/microbiología , Tuberculosis Latente/patología , Células Madre Mesenquimatosas/microbiología , Mycobacterium tuberculosis/aislamiento & purificación , Fagocitos/microbiología , Humanos , Mycobacterium tuberculosis/crecimiento & desarrollo
2.
Mol Ther ; 26(2): 446-455, 2018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29275847

RESUMEN

New vaccine platforms are needed to address the time gap between pathogen emergence and vaccine licensure. RNA-based vaccines are an attractive candidate for this role: they are safe, are produced cell free, and can be rapidly generated in response to pathogen emergence. Two RNA vaccine platforms are available: synthetic mRNA molecules encoding only the antigen of interest and self-amplifying RNA (sa-RNA). sa-RNA is virally derived and encodes both the antigen of interest and proteins enabling RNA vaccine replication. Both platforms have been shown to induce an immune response, but it is not clear which approach is optimal. In the current studies, we compared synthetic mRNA and sa-RNA expressing influenza virus hemagglutinin. Both platforms were protective, but equivalent levels of protection were achieved using 1.25 µg sa-RNA compared to 80 µg mRNA (64-fold less material). Having determined that sa-RNA was more effective than mRNA, we tested hemagglutinin from three strains of influenza H1N1, H3N2 (X31), and B (Massachusetts) as sa-RNA vaccines, and all protected against challenge infection. When sa-RNA was combined in a trivalent formulation, it protected against sequential H1N1 and H3N2 challenges. From this we conclude that sa-RNA is a promising platform for vaccines against viral diseases.


Asunto(s)
Virus de la Influenza A/inmunología , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/inmunología , Gripe Humana/prevención & control , ARN Viral/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Inmunización , Inmunización Secundaria , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/inmunología , Virus de la Influenza A/genética , Vacunas contra la Influenza/genética , Ratones , ARN Mensajero/genética , ARN Mensajero/inmunología , ARN Viral/genética , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
3.
J Infect Dis ; 217(10): 1667-1671, 2018 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-29471332

RESUMEN

Persistence of Mycobacterium tuberculosis within human bone marrow stem cells has been identified as a potential bacterial niche during latent tuberculosis. Using a murine model of tuberculosis, we show here that bone marrow stem and progenitor cells containing M. tuberculosis propagated tuberculosis when transferred to naive mice, given that both transferred cells and recipient mice were unable to express inducible nitric oxide synthase, which mediates killing of intracellular bacteria via nitric oxide. Our findings suggest that bone marrow stem and progenitor cells containing M. tuberculosis propagate hallmarks of disease if nitric oxide-mediated killing of bacteria is defective.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/microbiología , Mycobacterium tuberculosis/patogenicidad , Óxido Nítrico Sintasa de Tipo II/metabolismo , Células Madre/metabolismo , Células Madre/microbiología , Tuberculosis/metabolismo , Animales , Modelos Animales de Enfermedad , Trasplante de Células Madre Hematopoyéticas/métodos , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico/metabolismo , Tuberculosis/microbiología
4.
Proc Natl Acad Sci U S A ; 111(38): E4024-32, 2014 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-25201986

RESUMEN

Lung granulomas develop upon Mycobacterium tuberculosis (Mtb) infection as a hallmark of human tuberculosis (TB). They are structured aggregates consisting mainly of Mtb-infected and -uninfected macrophages and Mtb-specific T cells. The production of NO by granuloma macrophages expressing nitric oxide synthase-2 (NOS2) via l-arginine and oxygen is a key protective mechanism against mycobacteria. Despite this protection, TB granulomas are often hypoxic, and bacterial killing via NOS2 in these conditions is likely suboptimal. Arginase-1 (Arg1) also metabolizes l-arginine but does not require oxygen as a substrate and has been shown to regulate NOS2 via substrate competition. However, in other infectious diseases in which granulomas occur, such as leishmaniasis and schistosomiasis, Arg1 plays additional roles such as T-cell regulation and tissue repair that are independent of NOS2 suppression. To address whether Arg1 could perform similar functions in hypoxic regions of TB granulomas, we used a TB murine granuloma model in which NOS2 is absent. Abrogation of Arg1 expression in macrophages in this setting resulted in exacerbated lung granuloma pathology and bacterial burden. Arg1 expression in hypoxic granuloma regions correlated with decreased T-cell proliferation, suggesting that Arg1 regulation of T-cell immunity is involved in disease control. Our data argue that Arg1 plays a central role in the control of TB when NOS2 is rendered ineffective by hypoxia.


Asunto(s)
Arginasa/metabolismo , Granuloma/enzimología , Hipoxia/enzimología , Macrófagos/enzimología , Mycobacterium tuberculosis , Tuberculosis Pulmonar/enzimología , Animales , Arginasa/genética , Arginasa/inmunología , Arginina/genética , Arginina/inmunología , Arginina/metabolismo , Proliferación Celular/genética , Modelos Animales de Enfermedad , Granuloma/genética , Granuloma/inmunología , Granuloma/patología , Humanos , Hipoxia/genética , Hipoxia/inmunología , Hipoxia/patología , Pulmón/enzimología , Pulmón/inmunología , Pulmón/patología , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Noqueados , Óxido Nítrico/genética , Óxido Nítrico/inmunología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/inmunología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/patología , Tuberculosis Pulmonar/genética , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/patología
5.
Infect Immun ; 84(10): 2914-21, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27481246

RESUMEN

The neonatal Fc receptor (FcRn) extends the systemic half-life of IgG antibodies by chaperoning bound Fc away from lysosomal degradation inside stromal and hematopoietic cells. FcRn also transports IgG across mucosal barriers into the lumen, and yet little is known about how FcRn modulates immunity in the lung during homeostasis or infection. We infected wild-type (WT) and FcRn-deficient (fcgrt(-/-)) mice with Pseudomonas aeruginosa or Mycobacterium tuberculosis to investigate whether recycling and transport of IgG via FcRn influences innate and adaptive immunity in the lung in response to bacterial infection. We found that FcRn expression maintains homeostatic IgG levels in lung and leads to preferential secretion of low-affinity IgG ligands into the lumen. Fcgrt(-/-) animals exhibited no evidence of developmental impairment of innate immunity in the lung and were able to efficiently recruit neutrophils in a model of acute bacterial pneumonia. Although local humoral immunity in lung increased independently of the presence of FcRn during tuberculosis, there was nonetheless a strong impact of FcRn deficiency on local adaptive immunity. We show that the quantity and quality of IgG in airways, as well as the abundance of dendritic cells in the lung, are maintained by FcRn. FcRn ablation transiently enhanced local T cell immunity and neutrophil recruitment during tuberculosis, leading to a lower bacterial burden in lung. This novel understanding of tissue-specific modulation of mucosal IgG isotypes in the lung by FcRn sheds light on the role of mucosal IgG in immune responses in the lung during homeostasis and bacterial disease.


Asunto(s)
Células Dendríticas/inmunología , Antígenos de Histocompatibilidad Clase I/fisiología , Inmunoglobulina G/metabolismo , Pulmón , Receptores Fc/fisiología , Tuberculosis/inmunología , Inmunidad Adaptativa , Animales , Antígenos CD/metabolismo , Carga Bacteriana , Células Dendríticas/citología , Modelos Animales de Enfermedad , Antígenos de Histocompatibilidad Clase I/metabolismo , Inmunidad Innata , Inmunoglobulina G/inmunología , Cadenas alfa de Integrinas/metabolismo , Pulmón/citología , Pulmón/inmunología , Pulmón/metabolismo , Ratones , Ratones Transgénicos , Membrana Mucosa/metabolismo , Mycobacterium tuberculosis , Células Mieloides/metabolismo , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa , Receptores Fc/metabolismo , Tuberculosis/microbiología
6.
Eur J Immunol ; 44(8): 2380-93, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24782112

RESUMEN

General interest in the biological functions of IFN type I in Mycobacterium tuberculosis (Mtb) infection increased after the recent identification of a distinct IFN gene expression signature in tuberculosis (TB) patients. Here, we demonstrate that TB-susceptible mice lacking the receptor for IFN I (IFNAR1) were protected from death upon aerogenic infection with Mtb. Using this experimental model to mimic primary progressive pulmonary TB, we dissected the immune processes affected by IFN I. IFNAR1 signaling did not affect T-cell responses, but markedly altered migration of inflammatory monocytes and neutrophils to the lung. This process was orchestrated by IFNAR1 expressed on both immune and tissue-resident radioresistant cells. IFNAR1-driven TB susceptibility was initiated by augmented Mtb replication and in situ death events, along with CXCL5/CXCL1-driven accumulation of neutrophils in alveoli, followed by the discrete compartmentalization of Mtb in lung phagocytes. Early depletion of neutrophils rescued TB-susceptible mice to levels observed in mice lacking IFNAR1. We conclude that IFN I alters early innate events at the site of Mtb invasion leading to fatal immunopathology. These data furnish a mechanistic explanation for the detrimental role of IFN I in pulmonary TB and form a basis for understanding the complex roles of IFN I in chronic inflammation.


Asunto(s)
Interferón Tipo I/inmunología , Pulmón/inmunología , Fagocitos/inmunología , Transducción de Señal/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Células Cultivadas , Quimiocina CXCL1/inmunología , Quimiocina CXCL5/inmunología , Inflamación/inmunología , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología , Mycobacterium tuberculosis/inmunología , Neutrófilos/inmunología , Alveolos Pulmonares/inmunología , Receptor de Interferón alfa y beta/inmunología , Linfocitos T/inmunología
7.
Immunol Rev ; 240(1): 235-51, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21349097

RESUMEN

Tuberculosis (TB) is a complex disease, and the success of the bacterium as an intracellular pathogen is the outcome of its close and longstanding coevolution with the mammalian host. The dialogue between Mycobacterium tuberculosis and the host is becoming understandable at the molecular, cellular, and tissue level. This has led to the elucidation of the (i) interaction between pattern recognition receptors and pathogen-associated molecular patterns, (ii) cross-talk between immune cells, and (iii) mechanisms underlying granuloma development. Disease as an eventual but not a necessary consequence of infection results from a sensitive balance between protective immunity and destructive pathology. Early events, governed largely by conserved mechanisms of host recognition, impact not only on type and course of adaptive immunity but also on lung parenchymal function. New interpretations of how these responses shape the lung environment and direct granuloma development emphasize that the disease results from pathologic consequences of non-resolving inflammation. We review recent advances in TB research within the context of this ambitious view of TB.


Asunto(s)
Evolución Biológica , Evasión Inmune , Mycobacterium tuberculosis/inmunología , Tuberculosis/inmunología , Tuberculosis/patología , Humanos
8.
Int J Infect Dis ; 141S: 106988, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38417613

RESUMEN

The World Health Organization's aim to end the global tuberculosis (TB) epidemic by 2050 cannot be achieved without taking measures to identify people with asymptomatic Mycobacterium tuberculosis (Mtb) infection and offer them an intervention to reduce the risk of disease progression, such as preventive antimicrobial therapy. Implementation of this strategy is limited by the fact that existing tests for Mtb infection, which use immunosensitization to Mtb-specific antigens as a proxy for infection, have low positive predictive value for progression to TB. A blood test that detects Mtb deoxyribonucleic acid (DNA) could allow preventive therapy to be targeted at individuals with microbiological evidence of persistent infection. In this review, we summarize recent advances in the development of molecular microbial blood tests for Mtb infection and discuss potential explanations for discordance between their results and those of immunodiagnostic tests in adults with recent exposure to an infectious index case. We also present a roadmap for further development of molecular microbial blood tests for Mtb infection, and highlight the potential for research in this area to provide novel insights into the biology of Mtb infection and yield new tools to support efforts to control the global TB epidemic.


Asunto(s)
Tuberculosis Latente , Mycobacterium tuberculosis , Tuberculosis , Adulto , Humanos , Tuberculosis/microbiología , Tuberculosis Latente/microbiología , Mycobacterium tuberculosis/genética , Valor Predictivo de las Pruebas , Pruebas Hematológicas
9.
Nat Commun ; 15(1): 5074, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38871710

RESUMEN

Antimicrobial resistance (AMR) is a growing public health crisis that requires innovative solutions. Current susceptibility testing approaches limit our ability to rapidly distinguish between antimicrobial-susceptible and -resistant organisms. Salmonella Typhimurium (S. Typhimurium) is an enteric pathogen responsible for severe gastrointestinal illness and invasive disease. Despite widespread resistance, ciprofloxacin remains a common treatment for Salmonella infections, particularly in lower-resource settings, where the drug is given empirically. Here, we exploit high-content imaging to generate deep phenotyping of S. Typhimurium isolates longitudinally exposed to increasing concentrations of ciprofloxacin. We apply machine learning algorithms to the imaging data and demonstrate that individual isolates display distinct growth and morphological characteristics that cluster by time point and susceptibility to ciprofloxacin, which occur independently of ciprofloxacin exposure. Using a further set of S. Typhimurium clinical isolates, we find that machine learning classifiers can accurately predict ciprofloxacin susceptibility without exposure to it or any prior knowledge of resistance phenotype. These results demonstrate the principle of using high-content imaging with machine learning algorithms to predict drug susceptibility of clinical bacterial isolates. This technique may be an important tool in understanding the morphological impact of antimicrobials on the bacterial cell to identify drugs with new modes of action.


Asunto(s)
Antibacterianos , Ciprofloxacina , Farmacorresistencia Bacteriana , Aprendizaje Automático , Pruebas de Sensibilidad Microbiana , Salmonella typhimurium , Ciprofloxacina/farmacología , Salmonella typhimurium/efectos de los fármacos , Salmonella typhimurium/aislamiento & purificación , Antibacterianos/farmacología , Humanos , Infecciones por Salmonella/microbiología , Infecciones por Salmonella/tratamiento farmacológico , Algoritmos
10.
Eur J Immunol ; 42(2): 374-84, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22101787

RESUMEN

As a hallmark of tuberculosis (TB), Mycobacterium tuberculosis (MTB) induces granulomatous lung lesions and systemic inflammatory responses during active disease. Molecular regulation of inflammation is associated with inflammasome assembly. We determined the extent to which MTB triggers inflammasome activation and how this impacts on the severity of TB in a mouse model. MTB stimulated release of mature IL-1ß in macrophages while attenuated M. bovis BCG failed to do so. Tubercle bacilli specifically activated the NLRP3 inflammasome and this propensity was strictly controlled by the virulence-associated RD1 locus of MTB. However, Nlrp3-deficient mice controlled pulmonary TB, a feature correlated with NLRP3-independent production of IL-1ß in infected lungs. Our studies demonstrate that MTB activates the NLRP3 inflammasome in macrophages in an ESX-1-dependent manner. However, during TB, MTB promotes NLRP3- and caspase-1-independent IL-1ß release in myeloid cells recruited to lung parenchyma and thus overcomes NLRP3 deficiency in vivo in experimental models.


Asunto(s)
Proteínas Portadoras/metabolismo , Inflamasomas/inmunología , Macrófagos/metabolismo , Mycobacterium tuberculosis/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Susceptibilidad a Enfermedades , Proteínas de Homeodominio/metabolismo , Humanos , Interleucina-1beta/metabolismo , Pulmón/patología , Macrófagos/inmunología , Macrófagos/microbiología , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mycobacterium tuberculosis/patogenicidad , Proteína con Dominio Pirina 3 de la Familia NLR , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción/metabolismo , Tuberculosis Pulmonar/fisiopatología , Vacunas Atenuadas , Virulencia
11.
J Infect ; 86(6): 563-573, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36858180

RESUMEN

OBJECTIVES: The multidrug-resistant bacteria Acinetobacter baumannii is a major cause of hospital-associated infection; a vaccine could significantly reduce this burden. The aim was to develop a clinically relevant model of A. baumannii respiratory tract infection and to test the impact of different immunization routes on protective immunity provided by an outer membrane vesicle (OMV) vaccine. METHODS: BALB/c mice were intranasally challenged with isolates of oxa23-positive global clone GC2 A. baumannii from the lungs of patients with ventilator-associated pneumonia. Mice were immunized with OMVs by the intramuscular, subcutaneous or intranasal routes; protection was determined by measuring local and systemic bacterial load. RESULTS: Infection with A. baumannii clinical isolates led to a more disseminated infection than the prototype A. baumannii strain ATCC17978; with bacteria detectable in upper and lower airways and the spleen. Intramuscular immunization induced an antibody response but did not protect against bacterial infection. However, intranasal immunization significantly reduced airway colonization and prevented systemic bacterial dissemination. CONCLUSIONS: Use of clinically relevant isolates of A. baumannii provides stringent model for vaccine development. Intranasal immunization with OMVs was an effective route for providing protection, demonstrating that local immunity is important in preventing A. baumannii infection.


Asunto(s)
Acinetobacter baumannii , Sepsis , Animales , Ratones , Inmunización , Vacunación , Pulmón/microbiología , Sepsis/microbiología , Proteínas de la Membrana Bacteriana Externa , Vacunas Bacterianas , Ratones Endogámicos BALB C
12.
PLoS One ; 18(3): e0281559, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36972254

RESUMEN

BACKGROUND: The tuberculin skin test is commonly used to diagnose latent tuberculosis infection (LTBI) in resource-limited settings, but its specificity is limited by factors including cross-reactivity with BCG vaccine and environmental mycobacteria. Interferon-gamma release assays (IGRA) overcome this problem by detecting M. tuberculosis complex-specific responses, but studies to determine risk factors for IGRA-positivity in high TB burden settings are lacking. METHODS: We conducted a cross-sectional study to determine factors associated with a positive IGRA by employing the QuantiFERON-TB® Gold-plus (QFT Plus) assay in a cohort of asymptomatic adult TB contacts in Kampala, Uganda. Multivariate logistic regression analysis with forward stepwise logit function was employed to identify independent correlates of QFT Plus-positivity. RESULTS: Of the 202 participants enrolled, 129/202 (64%) were female, 173/202 (86%) had a BCG scar, and 67/202 (33%) were HIV-infected. Overall, 105/192 (54%, 95% CI 0.48-0.62) participants had a positive QFT Plus result. Increased risk of QFT-Plus positivity was independently associated with casual employment/unemployment vs. non-casual employment (adjusted odds ratio (aOR) 2.18, 95% CI 1.01-4.72), a family vs. non-family relation to the index patient (aOR 2.87, 95% CI 1.33-6.18), living in the same vs. a different house as the index (aOR 3.05, 95% CI 1.28-7.29), a higher body mass index (BMI) (aOR per additional kg/m2 1.09, 95% CI 1.00-1.18) and tobacco smoking vs. not (aOR 2.94, 95% CI 1.00-8.60). HIV infection was not associated with QFT-Plus positivity (aOR 0.91, 95% CI 0.42-1.96). CONCLUSION: Interferon Gamma Release Assay positivity in this study population was lower than previously estimated. Tobacco smoking and BMI were determinants of IGRA positivity that were previously unappreciated.


Asunto(s)
Infecciones por VIH , Tuberculosis Latente , Tuberculosis , Humanos , Adulto , Femenino , Masculino , Tuberculosis Latente/diagnóstico , Tuberculosis Latente/epidemiología , Estudios Transversales , Uganda/epidemiología , Tuberculosis/diagnóstico , Tuberculosis/epidemiología , Ensayos de Liberación de Interferón gamma , Prueba de Tuberculina , Infecciones por VIH/diagnóstico , Infecciones por VIH/epidemiología
13.
Eur J Immunol ; 40(6): 1663-73, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20222088

RESUMEN

Tuberculosis causes 2 million deaths per year, yet in most cases the immune response successfully contains the infection and prevents disease outbreak. Induced lymphoid structures associated with pulmonary granuloma are observed during tuberculosis in both humans and mice and could orchestrate host defense. To investigate whether granuloma perform lymphoid functions, mice lacking secondary lymphoid organs (SLO) were infected with Mycobacterium tuberculosis (MTB). As in WT mice, granuloma developed, exponential growth of MTB was controlled, and antigen-specific T-cell responses including memory T cells were generated in the absence of SLO. Moreover, adoptively transferred T cells were primed locally in lungs in a granuloma-dependent manner. T-cell activation was delayed in the absence of SLO, but resulted in a normal development program including protective subsets and functional recall responses that protected mice against secondary MTB infection. Our data demonstrate that protective immune responses can be generated independently of SLO during MTB infection and implicate local pulmonary T-cell priming as a mechanism contributing to host defense.


Asunto(s)
Granuloma/inmunología , Tejido Linfoide/inmunología , Linfocitos T/inmunología , Tuberculosis Pulmonar/inmunología , Traslado Adoptivo , Adulto , Animales , Separación Celular , Quimiocinas/biosíntesis , Citometría de Flujo , Expresión Génica , Perfilación de la Expresión Génica , Granuloma/microbiología , Humanos , Rayos Láser , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Microdisección , Persona de Mediana Edad , Mycobacterium tuberculosis/inmunología , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores de Quimiocina/biosíntesis
14.
Lancet Microbe ; 2(6): e267-e275, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34100007

RESUMEN

BACKGROUND: Haematopoietic stem cells expressing the CD34 surface marker have been posited as a niche for Mycobacterium tuberculosis complex bacilli during latent tuberculosis infection. Our aim was to determine whether M tuberculosis complex DNA is detectable in CD34-positive peripheral blood mononuclear cells (PBMCs) isolated from asymptomatic adults living in a setting with a high tuberculosis burden. METHODS: We did a cross-sectional study in Ethiopia between Nov 22, 2017, and Jan 10, 2019. Digital PCR (dPCR) was used to determine whether M tuberculosis complex DNA was detectable in PBMCs isolated from 100 mL blood taken from asymptomatic adults with HIV infection or a history of recent household or occupational exposure to an index case of human or bovine tuberculosis. Participants were recruited from HIV clinics, tuberculosis clinics, and cattle farms in and around Addis Ababa. A nested prospective study was done in a subset of HIV-infected individuals to evaluate whether administration of isoniazid preventive therapy was effective in clearing M tuberculosis complex DNA from PBMCs. Follow-up was done between July 20, 2018, and Feb 13, 2019. QuantiFERON-TB Gold assays were also done on all baseline and follow-up samples. FINDINGS: Valid dPCR data (ie, droplet counts >10 000 per well) were available for paired CD34-positive and CD34-negative PBMC fractions from 197 (70%) of 284 participants who contributed data to cross-sectional analyses. M tuberculosis complex DNA was detected in PBMCs of 156 of 197 participants with valid dPCR data (79%, 95% CI 74-85). It was more commonly present in CD34-positive than in CD34-negative fractions (154 [73%] of 197 vs 46 [23%] of 197; p<0·0001). Prevalence of dPCR-detected M tuberculosis complex DNA did not differ between QuantiFERON-negative and QuantiFERON-positive participants (77 [78%] of 99 vs 79 [81%] of 98; p=0·73), but it was higher in HIV-infected than in HIV-uninfected participants (67 [89%] of 75 vs 89 [73%] of 122, p=0·0065). By contrast, the proportion of QuantiFERON-positive participants was lower in HIV-infected than in HIV-uninfected participants (25 [33%] of 75 vs 73 [60%] of 122; p<0·0001). Administration of isoniazid preventive therapy reduced the prevalence of dPCR-detected M tuberculosis complex DNA from 41 (95%) of 43 HIV-infected individuals at baseline to 23 (53%) of 43 after treatment (p<0·0001), but it did not affect the prevalence of QuantiFERON positivity (17 [40%] of 43 at baseline vs 13 [30%] of 43 after treatment; p=0·13). INTERPRETATION: We report a novel molecular microbiological biomarker of latent tuberculosis infection with properties that are distinct from those of a commercial interferon-γ release assay. Our findings implicate the bone marrow as a niche for M tuberculosis in latently infected individuals. Detection of M tuberculosis complex DNA in PBMCs has potential applications in the diagnosis of latent tuberculosis infection, in monitoring response to preventive therapy, and as an outcome measure in clinical trials of interventions to prevent or treat latent tuberculosis infection. FUNDING: UK Medical Research Council.


Asunto(s)
Infecciones por VIH , Tuberculosis Latente , Mycobacterium tuberculosis , Tuberculosis , Estudios Transversales , ADN , Etiopía/epidemiología , Infecciones por VIH/tratamiento farmacológico , Humanos , Isoniazida/farmacología , Tuberculosis Latente/diagnóstico , Leucocitos Mononucleares , Mycobacterium tuberculosis/genética , Estudios Prospectivos , Prueba de Tuberculina , Tuberculosis/diagnóstico
15.
Eur J Immunol ; 39(12): 3369-84, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19795415

RESUMEN

Progression and outcome of tuberculosis is governed by extensive crosstalk between pathogen and host. Analyses of global changes in gene expression during immune response to infection with Mycobacterium tuberculosis (M.tb) can help identify molecular markers of disease state and progression. Global distribution of M.tb strains with different degrees of virulence and drug resistance, especially for the immunocompromised host, make closer analyses of host responses more pressing than ever. Here, we describe global transcriptional responses of inducible nitric oxide synthase-deficient (iNOS(-/-)) and WT mice infected with two related M.tb strains of markedly different virulence, namely the M.tb laboratory strains H37Rv and H37Ra. Both hosts exhibited highly similar resistance to infection with H37Ra. In contrast, iNOS(-/-) mice rapidly succumbed to H37Rv, whereas WT mice developed chronic course of disease. By differential analyses, virulence-specific changes in global host gene expression were analyzed to identify molecular markers characteristic for chronic versus acute infection. We identified several markers unique for different stages of disease progression and not previously associated with virulence-specific host responses in tuberculosis.


Asunto(s)
Perfilación de la Expresión Génica , Mycobacterium tuberculosis/inmunología , Tuberculosis Pulmonar/genética , Tuberculosis Pulmonar/inmunología , Animales , Predisposición Genética a la Enfermedad , Interacciones Huésped-Patógeno , Inmunidad Innata/genética , Pulmón/metabolismo , Pulmón/microbiología , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Mycobacterium tuberculosis/patogenicidad , Mycobacterium tuberculosis/fisiología , Óxido Nítrico Sintasa de Tipo II/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Especificidad de la Especie , Factor de Crecimiento Transformador beta1/genética , Tuberculosis Pulmonar/microbiología , Virulencia
16.
Sci Rep ; 10(1): 12414, 2020 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-32709982

RESUMEN

The increase of antimicrobial resistance (AMR), and lack of new classes of licensed antimicrobials, have made alternative treatment options for AMR pathogens increasingly attractive. Recent studies have demonstrated anti-bacterial efficacy of a humanised monoclonal antibody (mAb) targeting the O25b O-antigen of Escherichia coli ST131. To evaluate the phenotypic effects of antibody binding to diverse clinical E. coli ST131 O25b bacterial isolates in high-throughput, we designed a novel mAb screening method using high-content imaging (HCI) and image-based morphological profiling to screen a mAb targeting the O25b O-antigen. Screening the antibody against a panel of 86 clinical E. coli ST131 O25:H4 isolates revealed 4 binding phenotypes: no binding (18.60%), weak binding (4.65%), strong binding (69.77%) and strong agglutinating binding (6.98%). Impaired antibody binding could be explained by the presence of insertion sequences or mutations in O-antigen or lipopolysaccharide core biosynthesis genes, affecting the amount, structure or chain length of the O-antigen. The agglutinating binding phenotype was linked with lower O-antigen density, enhanced antibody-mediated phagocytosis and increased serum susceptibly. This study highlights the need to screen candidate mAbs against large panels of clinically relevant isolates, and that HCI can be used to evaluate mAb binding affinity and potential functional efficacy against AMR bacteria.


Asunto(s)
Antibacterianos/farmacología , Anticuerpos Monoclonales Humanizados/farmacología , Infecciones por Escherichia coli/tratamiento farmacológico , Escherichia coli/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Antibacterianos/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Farmacorresistencia Bacteriana/genética , Escherichia coli/genética , Escherichia coli/inmunología , Escherichia coli/ultraestructura , Infecciones por Escherichia coli/microbiología , Estudios de Factibilidad , Humanos , Secuencias Repetitivas Esparcidas/genética , Pruebas de Sensibilidad Microbiana , Microscopía Electrónica , Antígenos O/genética , Antígenos O/inmunología , Filogenia , Polimorfismo de Nucleótido Simple , Virulencia/inmunología
17.
Int J Med Microbiol ; 298(1-2): 143-50, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17702652

RESUMEN

Tuberculosis represents a serious problem for public health worldwide, and effective vaccines are urgently required. This represents a significant challenge as the causative bacterial agent, Mycobacterium tuberculosis, has developed strategies to persist in infected hosts despite the presence of potent T-cell-mediated immune responses. New advances in basic immunology are giving us improved understanding of what constitutes a protective immune response and ways this response is manipulated by the bacillus. Such insights should inform us how to design more effective vaccination strategies against intracellular pathogens.


Asunto(s)
Antígenos Bacterianos/inmunología , Mycobacterium tuberculosis/inmunología , Vacunas contra la Tuberculosis/inmunología , Tuberculosis/inmunología , Tuberculosis/prevención & control , Animales , Humanos , Inmunidad Celular/inmunología , Inmunidad Innata/inmunología , Linfocitos T/inmunología , Tuberculosis/microbiología , Vacunas contra la Tuberculosis/uso terapéutico , Vacunación/métodos , Vacunas de Subunidad/inmunología
18.
PLoS One ; 12(1): e0169119, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28046053

RESUMEN

An estimated third of the world's population is latently infected with Mycobacterium tuberculosis (Mtb), with no clinical signs of tuberculosis (TB), but lifelong risk of reactivation to active disease. The niches of persisting bacteria during latent TB infection remain unclear. We detect Mtb DNA in peripheral blood selectively in long-term repopulating pluripotent hematopoietic stem cells (LT-pHSCs) as well as in mesenchymal stem cells from latently infected human donors. In mice infected with low numbers of Mtb, that do not develop active disease we, again, find LT-pHSCs selectively infected with Mtb. In human and mouse LT-pHSCs Mtb are stressed or dormant, non-replicating bacteria. Intratracheal injection of Mtb-infected human and mouse LT-pHSCs into immune-deficient mice resuscitates Mtb to replicating bacteria within the lung, accompanied by signs of active infection. We conclude that LT-pHSCs, together with MSCs of Mtb-infected humans and mice serve as a hitherto unappreciated quiescent cellular depot for Mtb during latent TB infection.


Asunto(s)
Células Madre Hematopoyéticas/microbiología , Tuberculosis Latente/microbiología , Células Madre Mesenquimatosas/microbiología , Mycobacterium tuberculosis , Adulto , Animales , Antígenos CD34/metabolismo , Células de la Médula Ósea/metabolismo , Separación Celular , Femenino , Citometría de Flujo , Humanos , Pulmón/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Adulto Joven
19.
Sci Rep ; 7(1): 8853, 2017 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-28821804

RESUMEN

During active TB in humans a spectrum of pulmonary granulomas with central necrosis and hypoxia exists. BALB/c mice, predominantly used in TB drug development, do not reproduce this complex pathology thereby inaccurately predicting clinical outcome. We found that Nos2 -/- mice incapable of NO-production in immune cells as microbial defence uniformly develop hypoxic necrotizing lung lesions, widely observed in human TB. To study the impact of hypoxic necrosis on the efficacy of antimycobacterials and drug candidates, we subjected Nos2 -/- mice with TB to monotherapy before or after establishment of human-like pathology. Isoniazid induced a drug-tolerant persister population only when necrotic lesions were present. Rifapentine was more potent than rifampin prior to development of human-like pathology and equally potent thereafter, in agreement with recent clinical trials. Pretomanid, delamanid and the pre-clinical candidate BTZ043 were bactericidal independent of pulmonary pathology. Linezolid was bacteriostatic in TB-infected Nos2 -/- mice but significantly improved lung pathology. Hypoxic necrotizing lesions rendered moxifloxacin less active. In conclusion, Nos2 -/- mice are a predictive TB drug development tool owing to their consistent development of human-like pathology.


Asunto(s)
Hipoxia/metabolismo , Necrosis/genética , Necrosis/metabolismo , Óxido Nítrico Sintasa de Tipo II/deficiencia , Tuberculosis Pulmonar/etiología , Tuberculosis Pulmonar/metabolismo , Animales , Antituberculosos/farmacología , Modelos Animales de Enfermedad , Fibrosis , Células Espumosas/inmunología , Células Espumosas/metabolismo , Células Espumosas/patología , Humanos , Hipoxia/patología , Isoniazida/farmacología , Ratones , Ratones Noqueados , Necrosis/patología , Rifampin/análogos & derivados , Rifampin/farmacología , Resultado del Tratamiento , Tuberculosis Pulmonar/tratamiento farmacológico , Tuberculosis Pulmonar/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA