Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Med ; 2(3): 217-232, 2021 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-34693385

RESUMEN

There is hope that host-directed therapy (HDT) for Tuberculosis (TB) can either shorten treatment duration, help cure drug resistant disease or limit the immunopathology. Many candidate HDT drugs have been proposed, however solid evidence only exists for a few select patient groups. The clinical presentation of TB is variable, with differences in severity, tissue pathology, and bacillary burden. TB clinical phenotypes likely determine the potential benefit of HDT. Underlying TB clinical phenotypes, there are TB "endotypes," defined as distinct molecular profiles, with specific metabolic, epigenetic, transcriptional, and immune phenotypes. TB endotypes can be characterized by either immunodeficiency or pathologic excessive inflammation. Additional factors, like comorbidities (HIV, diabetes, helminth infection), structural lung disease or Mycobacterial virulence also drive TB endotypes. Precise disease phenotyping, combined with in-depth immunologic and molecular profiling and multimodal omics integration, can identify TB endotypes, guide endotype-specific HDT, and improve TB outcomes, similar to advances in cancer medicine.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Antituberculosos/uso terapéutico , Humanos , Mycobacterium tuberculosis/genética , Tuberculosis/tratamiento farmacológico
2.
Nat Rev Urol ; 18(10): 611-622, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34131332

RESUMEN

Bacillus Calmette-Guérin (BCG) is the most widely used vaccine worldwide and has been used to prevent tuberculosis for a century. BCG also stimulates an anti-tumour immune response, which urologists have harnessed for the treatment of non-muscle-invasive bladder cancer. A growing body of evidence indicates that BCG offers protection against various non-mycobacterial and viral infections. The non-specific effects of BCG occur via the induction of trained immunity and form the basis for the hypothesis that BCG vaccination could be used to protect against the severity of coronavirus disease 2019 (COVID-19). This Perspective article highlights key milestones in the 100-year history of BCG and projects its potential role in the COVID-19 pandemic.


Asunto(s)
Adyuvantes Inmunológicos/historia , Vacuna BCG/historia , Vacunas contra la COVID-19 , COVID-19/prevención & control , Inmunoterapia/historia , Animales , Bovinos , Historia del Siglo XIX , Historia del Siglo XX , Humanos , Lactante
3.
PLoS Pathog ; 17(5): e1009570, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33989345

RESUMEN

Mycobacterium tuberculosis (Mtb) has complex and dynamic interactions with the human host, and subpopulations of Mtb that emerge during infection can influence disease outcomes. This study implicates zinc ion (Zn2+) availability as a likely driver of bacterial phenotypic heterogeneity in vivo. Zn2+ sequestration is part of "nutritional immunity", where the immune system limits micronutrients to control pathogen growth, but this defense mechanism seems to be ineffective in controlling Mtb infection. Nonetheless, Zn2+-limitation is an environmental cue sensed by Mtb, as calprotectin triggers the zinc uptake regulator (Zur) regulon response in vitro and co-localizes with Zn2+-limited Mtb in vivo. Prolonged Zn2+ limitation leads to numerous physiological changes in vitro, including differential expression of certain antigens, alterations in lipid metabolism and distinct cell surface morphology. Furthermore, Mtb enduring limited Zn2+ employ defensive measures to fight oxidative stress, by increasing expression of proteins involved in DNA repair and antioxidant activity, including well described virulence factors KatG and AhpC, along with altered utilization of redox cofactors. Here, we propose a model in which prolonged Zn2+ limitation defines a population of Mtb with anticipatory adaptations against impending immune attack, based on the evidence that Zn2+-limited Mtb are more resistant to oxidative stress and exhibit increased survival and induce more severe pulmonary granulomas in mice. Considering that extracellular Mtb may transit through the Zn2+-limited caseum before infecting naïve immune cells or upon host-to-host transmission, the resulting phenotypic heterogeneity driven by varied Zn2+ availability likely plays a key role during early interactions with host cells.


Asunto(s)
Granuloma/microbiología , Lipidómica , Mycobacterium tuberculosis/fisiología , Proteoma , Transcriptoma , Zinc/deficiencia , Adaptación Fisiológica , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Granuloma/inmunología , Homeostasis , Interacciones Huésped-Patógeno , Humanos , Pulmón/microbiología , Ratones , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/patogenicidad , Oxidación-Reducción , Estrés Oxidativo , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
4.
J Clin Invest ; 130(6): 3113-3123, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32125282

RESUMEN

Mycobacterium tuberculosis (M. tuberculosis) has coevolved with humans for millennia and developed multiple mechanisms to evade host immunity. Restoring host immunity in order to improve outcomes and potentially shorten existing therapy will require identification of the full complement by which host immunity is inhibited. Perturbation of host DNA methylation is a mechanism induced by chronic infections such as HIV, HPV, lymphocytic choriomeningitis virus (LCMV), and schistosomiasis to evade host immunity. Here, we evaluated the DNA methylation status of patients with tuberculosis (TB) and their asymptomatic household contacts and found that the patients with TB have DNA hypermethylation of the IL-2/STAT5, TNF/NF-κB, and IFN-γ signaling pathways. We performed methylation-sensitive restriction enzyme-quantitative PCR (MSRE-qPCR) and observed that multiple genes of the IL-12/IFN-γ signaling pathway (IL12B, IL12RB2, TYK2, IFNGR1, JAK1, and JAK2) were hypermethylated in patients with TB. The DNA hypermethylation of these pathways was associated with decreased immune responsiveness with decreased mitogen-induced upregulation of IFN-γ, TNF, IL-6, CXCL9, CXCL10, and IL-1ß production. The DNA hypermethylation of the IL-12/IFN-γ pathway was associated with decreased IFN-γ-induced gene expression and decreased IL-12-inducible upregulation of IFN-γ. This study demonstrates that immune cells from patients with TB are characterized by DNA hypermethylation of genes critical to mycobacterial immunity resulting in decreased mycobacteria-specific and nonspecific immune responsiveness.


Asunto(s)
Metilación de ADN/inmunología , Regulación de la Expresión Génica/inmunología , Leucocitos/inmunología , Mycobacterium tuberculosis/inmunología , Transducción de Señal/inmunología , Tuberculosis/inmunología , Humanos , Leucocitos/patología , Tuberculosis/patología
5.
Artículo en Inglés | MEDLINE | ID: mdl-32161724

RESUMEN

Mycobacterium tuberculosis is primarily a respiratory pathogen. However, 15% of infections worldwide occur at extrapulmonary sites causing additional complications for diagnosis and treatment of the disease. In addition, dissemination of M. tuberculosis out of the lungs is thought to be more than just a rare event leading to extrapulmonary tuberculosis, but rather a prerequisite step that occurs during all infections, producing secondary lesions that can become latent or productive. In this review we will cover the clinical range of extrapulmonary infections and the process of dissemination including evidence from both historical medical literature and animal experiments for dissemination and subsequent reseeding of the lungs through the lymphatic and circulatory systems. While the mechanisms of M. tuberculosis dissemination are not fully understood, we will discuss the various models that have been proposed to address how this process may occur and summarize the bacterial virulence factors that facilitate M. tuberculosis dissemination.


Asunto(s)
Mycobacterium tuberculosis/patogenicidad , Tuberculosis/microbiología , Animales , Células Dendríticas/microbiología , Modelos Animales de Enfermedad , Células Epiteliales/microbiología , Humanos , Pulmón/microbiología , Macrófagos Alveolares/microbiología , Tuberculosis/inmunología , Tuberculosis/patología , Tuberculosis Ganglionar/microbiología , Tuberculosis Pleural/microbiología , Factores de Virulencia/fisiología
6.
ACS Appl Mater Interfaces ; 12(12): 13657-13670, 2020 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-32091877

RESUMEN

Light-activated molecular nanomachines (MNMs) can be used to drill holes into prokaryotic (bacterial) cell walls and the membrane of eukaryotic cells, including mammalian cancer cells, by their fast rotational movement, leading to cell death. We examined how these MNMs function in multicellular organisms and investigated their use for treatment and eradication of specific diseases by causing damage to certain tissues and small organisms. Three model eukaryotic species, Caenorhabditis elegans, Daphnia pulex, and Mus musculus (mouse), were evaluated. These organisms were exposed to light-activated fast-rotating MNMs and their physiological and pathological changes were studied in detail. Slow rotating MNMs were used to control for the effects of rotation rate. We demonstrate that fast-rotating MNMs caused depigmentation and 70% mortality in C. elegans while reducing the movement as well as heart rate and causing tissue damage in Daphnia. Topically applied light-activated MNMs on mouse skin caused ulceration and microlesions in the epithelial tissue, allowing MNMs to localize into deeper epidermal tissue. Overall, this study shows that the nanomechanical action of light-activated MNMs is effective against multicellular organisms, disrupting cell membranes and damaging tissue in vivo. Customized MNMs that target specific tissues for therapy combined with spatial and temporal control could have broad clinical applications in a variety of benign and malignant disease states including treatment of cancer, parasites, bacteria, and diseased tissues.


Asunto(s)
Membrana Celular/efectos de los fármacos , Eucariontes/efectos de los fármacos , Nanoestructuras/química , Neoplasias/tratamiento farmacológico , Animales , Bacterias/efectos de los fármacos , Caenorhabditis elegans/efectos de los fármacos , Membrana Celular/química , Humanos , Luz , Ratones , Nanoestructuras/efectos de la radiación , Nanoestructuras/uso terapéutico
7.
ACS Nano ; 13(12): 14377-14387, 2019 12 24.
Artículo en Inglés | MEDLINE | ID: mdl-31815423

RESUMEN

Multidrug resistance in pathogenic bacteria is an increasing problem in patient care and public health. Molecular nanomachines (MNMs) have the ability to open cell membranes using nanomechanical action. We hypothesized that MNMs could be used as antibacterial agents by drilling into bacterial cell walls and increasing susceptibility of drug-resistant bacteria to recently ineffective antibiotics. We exposed extensively drug-resistant Klebsiella pneumoniae to light-activated MNMs and found that MNMs increase the susceptibility to Meropenem. MNMs with Meropenem can effectively kill K. pneumoniae that are considered Meropenem-resistant. We examined the mechanisms of MNM action using permeability assays and transmission electron microscopy, finding that MNMs disrupt the cell wall of extensively drug-resistant K. pneumoniae, exposing the bacteria to Meropenem. These observations suggest that MNMs could be used to make conventional antibiotics more efficacious against multi-drug-resistant pathogens.


Asunto(s)
Pared Celular/efectos de los fármacos , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Klebsiella pneumoniae/efectos de los fármacos , Meropenem/farmacología , Nanopartículas/toxicidad , Animales , Antibacterianos/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular , Klebsiella pneumoniae/efectos de la radiación , Klebsiella pneumoniae/ultraestructura , Luz , Macrófagos/citología , Macrófagos/efectos de los fármacos , Meropenem/química , Ratones , Pruebas de Sensibilidad Microbiana , Viabilidad Microbiana/efectos de los fármacos , Movimiento
8.
Sci Rep ; 9(1): 10084, 2019 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-31300732

RESUMEN

We recently identified inhibitors targeting Mycobacterium marinum MelF (Rv1936) by in silico analysis, which exhibited bacteriostatic/bactericidal activity against M. marinum and M. tuberculosis in vitro. Herein, we evaluated the effect of best four inhibitors (# 5175552, # 6513745, # 5255829, # 9125618) obtained from the ChemBridge compound libraries, on intracellular replication and persistence of bacteria within IFN-γ activated murine RAW264.7 and human THP-1 macrophages infected with M. marinum. Inhibitors # 5175552 and # 6513745 significantly reduced (p < 0.05) the intracellular replication of bacilli during day 7 post-infection (p.i.) within RAW264.7 and THP-1 macrophages infected at multiplicity of infection (MOI) of ~1.0. These observations were substantiated by electron microscopy, which revealed the protective effect of # 5175552 in clearing the bacilli inside murine macrophages. Strikingly, # 6513745 displayed synergism with isoniazid against M. marinum in murine macrophages, whereas # 5175552 significantly suppressed (p < 0.05) the persistent bacilli during day 10-14 p.i. in infected RAW264.7 and THP-1 macrophages (MOI of ~ 0.1). Moreover, # 5175552 and # 6513745 were non-cytotoxic to host macrophages at both 1X and 5X MIC. Further validation of these inhibitors against M. tuberculosis-infected macrophages and animal models has potential for development as novel anti-tubercular agents.


Asunto(s)
Antituberculosos/farmacología , Macrófagos/microbiología , Infecciones por Mycobacterium no Tuberculosas/tratamiento farmacológico , Mycobacterium marinum/efectos de los fármacos , Mycobacterium tuberculosis/efectos de los fármacos , Tuberculosis/tratamiento farmacológico , Animales , Línea Celular , Sinergismo Farmacológico , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Interferón gamma/genética , Interferón gamma/inmunología , Isoniazida/farmacología , Activación de Macrófagos/inmunología , Ratones , Ratones Noqueados , Microscopía Electrónica de Transmisión , Células RAW 264.7 , Especies Reactivas de Oxígeno/metabolismo , Células THP-1
9.
Tuberculosis (Edinb) ; 116S: S28-S33, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31080089

RESUMEN

The human immunodeficiency virus (HIV) pandemic is driving the re-emergence of tuberculosis (TB) as a global health threat, both by increasing the susceptibility of HIV-infected people to infection with Mycobacterium tuberculosis (Mtb), and increasing the rate of emergence of drug-resistant Mtb. There are several other clinical challenges for treatment of co-infected patients including: expense, pill burden, toxicity, and malabsorption that further necessitate the search for new drugs that may be effective against both pathogens simultaneously. The anti-helminthic niclosamide has been shown to have activity against a laboratory strain of Mtb in liquid culture while bacteriostatic activity against non-replicating M. abscessus was also recently described. Here we extend these findings to further demonstrate that niclosamide inhibits mycobacterial growth in infected human macrophages and mediates potent bacteriostatic activity against the virulent Mtb Beijing strain. Importantly, we provide the first evidence that niclosamide inhibits HIV replication in human macrophages and Jurkat T cells through post-integration effects on pro-virus transcription. The dual antiviral and anti-mycobacterial activity was further observed in an in vitro model of HIV and Mtb co-infection using human primary monocyte-derived macrophages. These results support further investigation of niclosamide and derivatives as anti-retroviral/anti-mycobacterial agents that may reduce clinical challenges associated with multi-drug regimens and drug resistance.


Asunto(s)
VIH-1 , Macrófagos , Mycobacterium tuberculosis , Niclosamida , Linfocitos T , Replicación Viral , Humanos , Fármacos Anti-VIH/farmacología , Antituberculosos/farmacología , Coinfección , VIH-1/efectos de los fármacos , VIH-1/crecimiento & desarrollo , Células Jurkat , Macrófagos/efectos de los fármacos , Macrófagos/microbiología , Macrófagos/virología , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/crecimiento & desarrollo , Mycobacterium tuberculosis/patogenicidad , Niclosamida/farmacología , Linfocitos T/efectos de los fármacos , Linfocitos T/virología , Virulencia , Replicación Viral/efectos de los fármacos , Tuberculosis
10.
Methods Mol Biol ; 1790: 75-85, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29858784

RESUMEN

Fluorescence imaging has been applied to various areas of biological research, including studies of physiological, neurological, oncological, cell biological, molecular, developmental, immunological, and infectious processes. In this chapter, we describe methods of fluorescent imaging applied to examination of subcutaneous and pulmonary mycobacterial infections in an animal model. Since slow growth of Mycobacterium tuberculosis (Mtb) hinders development of new diagnostics, therapeutics, and vaccines for tuberculosis (TB), we developed fluorescent protein (FP) expressing mycobacterial strains for in vivo imaging, which can be used to track bacterial location and to quantitate bacterial load directly in living animals. After comparison of imaging data using strains expressing different fluorescent proteins, we found that strains expressing L5-tdTomato display the greatest fluorescence. Here, we describe detailed protocols for tdTomato-labeled M. bovis BCG imaging in real time for subcutaneous and pulmonary infections in living mice. These procedures allow rapid and accurate determination of bacterial numbers in live mice.


Asunto(s)
Proteínas Luminiscentes/metabolismo , Imagen Molecular/métodos , Mycobacterium tuberculosis/aislamiento & purificación , Tuberculosis Pulmonar/patología , Animales , Femenino , Fluorescencia , Ratones , Ratones Endogámicos BALB C , Tuberculosis Pulmonar/metabolismo , Tuberculosis Pulmonar/microbiología
11.
J Control Release ; 266: 238-247, 2017 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-28987879

RESUMEN

Worldwide, tuberculosis (TB) remains one of the most prevalent infectious diseases causing morbidity and death in >1.5 million patients annually. Mycobacterium tuberculosis (Mtb), the etiologic agent of TB, usually resides in the alveolar macrophages. Current tuberculosis treatment methods require more than six months, and low compliance often leads to therapeutic failure and multidrug resistant strain development. Critical to improving TB-therapy is shortening treatment duration and increasing therapeutic efficacy. In this study, we sought to determine if lung hemodynamics and pathological changes in Mtb infected cells can be used for the selective targeting of microparticles to infected tissue(s). Thioaptamers (TA) with CD44 (CD44TA) targeting moiety were conjugated to discoidal silicon mesoporous microparticles (SMP) to enhance accumulation of these agents/carriers in the infected macrophages in the lungs. In vitro, CD44TA-SMP accumulated in macrophages infected with mycobacteria efficiently killing the infected cells and decreasing survival of mycobacteria. In vivo, increased accumulations of CD44TA-SMP were recorded in the lung of M. tuberculosis infected mice as compared to controls. TA-targeted carriers significantly diminished bacterial load in the lungs and caused recruitment of T lymphocytes. Proposed mechanism of action of the designed vector accounts for a combination of increased uptake of particles that leads to infected macrophage death, as well as, activation of cellular immunity by the TA, causing increased T-cell accumulation in the treated lungs. Based on our data with CD44TA-SMP, we anticipate that this drug carrier can open new avenues in TB management.


Asunto(s)
Aptámeros de Nucleótidos/administración & dosificación , Portadores de Fármacos/administración & dosificación , Receptores de Hialuranos/genética , Mycobacterium tuberculosis , Tuberculosis/tratamiento farmacológico , Animales , Células Cultivadas , Femenino , Humanos , Receptores de Hialuranos/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Macrófagos/metabolismo , Ratones Endogámicos BALB C , Silicio/administración & dosificación , Linfocitos T/inmunología , Tuberculosis/inmunología , Tuberculosis/metabolismo
12.
J Infect Dis ; 214(3): 479-88, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27389351

RESUMEN

Helminth-infected individuals possess a higher risk of developing tuberculosis, but the precise immunologic mechanism of Mycobacterium tuberculosis control remains unclear. We hypothesized that a perturbation of the M. tuberculosis-specific CD4(+) T-cell response weakens the ability of macrophages to contain M. tuberculosis We exposed peripheral blood mononuclear cells from M. tuberculosis-infected humans to schistosome soluble egg antigen (SEA) and then profiled M. tuberculosis-specific CD4(+) T cells via multiparametric flow cytometry. SEA decreased the frequency of cells producing interferon γ (6.79% vs 3.20%; P = .017) and tumor necrosis factor α (6.98% vs 2.96%; P = .012), with a concomitant increase in the median fluorescence intensity of interleukin 4 (IL-4; P < .05) and interleukin 10 (IL-10; 1440 vs 1273; P < .05). Macrophages polarized with SEA-exposed, autologous CD4(+) T-cell supernatant had a 2.19-fold decreased colocalization of lysosomes and M. tuberculosis (P < .05). When polarized with IL-4 or IL-10, macrophages had increased expression of CD206 (P < .0001), 1.5-fold and 1.9 fold increased intracellular numbers of M. tuberculosis per macrophage (P < .0005), and 1.4-fold and 1.7-fold decreased colocalization between M. tuberculosis and lysosomes (P < .001). This clarifies a relationship in which helminth-induced CD4(+) T cells disrupt M. tuberculosis control by macrophages, thereby providing a mechanism for the observation that helminth infection advances the progression of tuberculosis among patients with M. tuberculosis infection.


Asunto(s)
Antígenos Helmínticos/metabolismo , Linfocitos T CD4-Positivos/efectos de los fármacos , Factores Inmunológicos/metabolismo , Macrófagos/inmunología , Macrófagos/microbiología , Mycobacterium tuberculosis/inmunología , Schistosoma/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Citocinas/biosíntesis , Citometría de Flujo , Humanos , Tolerancia Inmunológica , Inmunofenotipificación , Leucocitos Mononucleares/inmunología , Lisosomas/metabolismo , Macrófagos/fisiología , Fagosomas/metabolismo
13.
Sci Rep ; 6: 21522, 2016 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-26908312

RESUMEN

Co-infection with HIV increases the morbidity and mortality associated with tuberculosis due to multiple factors including a poorly understood microbial synergy. We developed a novel small animal model of co-infection in the humanized mouse to investigate how HIV infection disrupts pulmonary containment of Mtb. Following dual infection, HIV-infected cells were localized to sites of Mtb-driven inflammation and mycobacterial replication in the lung. Consistent with disease in human subjects, we observed increased mycobacterial burden, loss of granuloma structure, and increased progression of TB disease, due to HIV co-infection. Importantly, we observed an HIV-dependent pro-inflammatory cytokine signature (IL-1ß, IL-6, TNFα, and IL-8), neutrophil accumulation, and greater lung pathology in the Mtb-co-infected lung. These results suggest that in the early stages of acute co-infection in the humanized mouse, infection with HIV exacerbates the pro-inflammatory response to pulmonary Mtb, leading to poorly formed granulomas, more severe lung pathology, and increased mycobacterial burden and dissemination.


Asunto(s)
Coinfección/inmunología , Infecciones por VIH/inmunología , VIH-1/inmunología , Huésped Inmunocomprometido , Tuberculosis Pulmonar/inmunología , Animales , Modelos Animales de Enfermedad , Infecciones por VIH/virología , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Ratones , Infiltración Neutrófila
14.
PLoS One ; 9(9): e108341, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25265287

RESUMEN

Molecular analysis of infectious processes in bacteria normally involves construction of isogenic mutants that can then be compared to wild type in an animal model. Pathogenesis and antimicrobial studies are complicated by variability between animals and the need to sacrifice individual animals at specific time points. Live animal imaging allows real-time analysis of infections without the need to sacrifice animals, allowing quantitative data to be collected at multiple time points in all organs simultaneously. However, imaging has not previously allowed simultaneous imaging of both mutant and wild type strains of mycobacteria in the same animal. We address this problem by using both firefly (Photinus pyralis) and click beetle (Pyrophorus plagiophthalamus) red luciferases, which emit distinct bioluminescent spectra, allowing simultaneous imaging of two different mycobacterial strains during infection. We also demonstrate that these same bioluminescence reporters can be used to evaluate therapeutic efficacy in real-time, greatly facilitating our ability to screen novel antibiotics as they are developed. Due to the slow growth rate of mycobacteria, novel imaging technologies are a pressing need, since they can they can impact the rate of development of new therapeutics as well as improving our understanding of virulence mechanisms and the evaluation of novel vaccine candidates.


Asunto(s)
Coinfección/diagnóstico , Mediciones Luminiscentes/métodos , Macrófagos/microbiología , Mycobacterium tuberculosis/clasificación , Tuberculosis Pulmonar/diagnóstico , Animales , Línea Celular , Coinfección/microbiología , Diagnóstico por Imagen , Femenino , Luciferasas de Luciérnaga , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Mycobacterium tuberculosis/genética , Photorhabdus , Tuberculosis Pulmonar/microbiología
15.
Mol Microbiol ; 89(3): 403-19, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23734719

RESUMEN

Salmonella Typhimurium gene STM2215 (rtn) is conserved among many enterobacteriaceae. Mutants lacking STM2215 poorly colonized the liver and spleen in intraperitoneal infection of mice and poorly colonized the intestine and deeper tissues in oral infection. These phenotypes were complemented by a wild-type copy of STM2215 provided in trans. STM2215 deletion mutants grew normally in J774A.1 murine macrophages but were unable to invade Caco-2 colonic epithelial cells. Consistent with this finding, mutants in STM2215 produced lower levels of effectors of the TTSS-1. STM2215 is a predicted c-di-GMP phosphodiesterase, but lacks identifiable sensor domains. Biochemical analysis of STM2215 determined that it is located in the inner membrane and has c-di-GMP phosphodiesterase activity in vitro dependent on an intact EAL motif. Unlike some previously identified members of this family, STM2215 did not affect motility, was expressed on plates, and in liquid media at late exponential and early stationary phase during growth. Defined mutations in STM2215 revealed that neither the predicted periplasmic domain nor the anchoring of the protein to the inner membrane is necessary for the activity of this protein during infection. However, the EAL domain of STM2215 is required during infection, suggesting that its phosphodiesterase activity is necessary during infection.


Asunto(s)
Proteínas Bacterianas/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Infecciones por Salmonella/microbiología , Salmonella typhimurium/enzimología , Secuencia de Aminoácidos , Animales , Proteínas Bacterianas/genética , Células CACO-2 , Femenino , Eliminación de Gen , Humanos , Macrófagos/microbiología , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Hidrolasas Diéster Fosfóricas/genética , Estructura Terciaria de Proteína , Salmonella typhimurium/genética
16.
PLoS One ; 8(5): e63331, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23691024

RESUMEN

Mycobacterium tuberculosis (M.tb) is the second leading infectious cause of death worldwide and the primary cause of death in people living with HIV/AIDS. There are several excellent animal models employed to study tuberculosis (TB), but many have limitations for reproducing human pathology and none are amenable to the direct study of HIV/M.tb co-infection. The humanized mouse has been increasingly employed to explore HIV infection and other pathogens where animal models are limiting. Our goal was to develop a small animal model of M.tb infection using the bone marrow, liver, thymus (BLT) humanized mouse. NOD-SCID/γc(null) mice were engrafted with human fetal liver and thymus tissue, and supplemented with CD34(+) fetal liver cells. Excellent reconstitution, as measured by expression of the human CD45 pan leukocyte marker by peripheral blood populations, was observed at 12 weeks after engraftment. Human T cells (CD3, CD4, CD8), as well as natural killer cells and monocyte/macrophages were all observed within the human leukocyte (CD45(+)) population. Importantly, human T cells were functionally competent as determined by proliferative capacity and effector molecule (e.g. IFN-γ, granulysin, perforin) expression in response to positive stimuli. Animals infected intranasally with M.tb had progressive bacterial infection in the lung and dissemination to spleen and liver from 2-8 weeks post infection. Sites of infection in the lung were characterized by the formation of organized granulomatous lesions, caseous necrosis, bronchial obstruction, and crystallization of cholesterol deposits. Human T cells were distributed throughout the lung, liver, and spleen at sites of inflammation and bacterial growth and were organized to the periphery of granulomas. These preliminary results demonstrate the potential to use the humanized mouse as a model of experimental TB.


Asunto(s)
Modelos Animales de Enfermedad , Tuberculosis/fisiopatología , Animales , Trasplante de Médula Ósea/métodos , Humanos , Hígado/citología , Hígado/patología , Pulmón/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Linfocitos T/inmunología , Timo/citología
17.
Tuberculosis (Edinb) ; 93 Suppl: S83-7, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24388655

RESUMEN

Using a high throughput genetic strategy, designated Random Inducible Controlled Expression (RICE), we identified the six gene mel2 locus in Mtb and M. marinum. Interestingly, three of the genes present in mel2 have similarities to bioluminescence genes. Similar to other bacterial bioluminescence systems, mel2 facilitates detoxification of reactive oxygen species (ROS). Through the use of thin layer chromatography (TLC) we demonstrate enhanced production of the cell wall virulence lipid, pthiocerol dimycoserosate (PDIM), in a Mtb mel2 mutant relative to the wild type strain in the presence of both H2O2 and diamide oxidative stresses. Furthermore, propionate toxicity assays revealed increased accumulation of triacylglycerol (TAG) in the mel2 mutant relative to wild type. These observations provide the first evidence that mel2 plays a critical role in Mtb lipid biosynthesis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Pared Celular/metabolismo , Lípidos/biosíntesis , Mycobacterium marinum/genética , Mycobacterium tuberculosis/genética , Operón , Proliferación Celular , Cromatografía en Capa Delgada , Prueba de Complementación Genética , Humanos , Proteínas Luminiscentes/metabolismo , Macrófagos/metabolismo , Infecciones por Mycobacterium no Tuberculosas/genética , Infecciones por Mycobacterium no Tuberculosas/microbiología , Estrés Oxidativo/genética , Especies Reactivas de Oxígeno/metabolismo , Virulencia
18.
Curr Protoc Microbiol ; Chapter 2: Unit 2C.3, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21538304

RESUMEN

Optical imaging is emerging as a powerful tool to study physiological, neurological, oncological, cell biological, molecular, developmental, immunological, and infectious processes. This unit describes the use of fluorescent reporters for biological organisms, components, or events. We describe the application of fluorescence imaging to examination of infectious processes, in particular subcutaneous and pulmonary bacterial infections, but the same approaches are applicable to nearly any infectious route. The strategies described use mycobacterial infections as an example, but nearly identical systems can be used for Pseudomonas, Legionella, Salmonella, Escherichia, Borrelia, and Staphylococus, suggesting that the approaches are generally applicable to nearly any infectious agent. Two strategies for fluorescence imaging are described: the first method uses reporter enzyme fluorescence (REF), and the second uses fluorescent proteins for fluorescence imaging. Methods are described in detail to facilitate successful application of these emerging technologies to nearly any experimental system.


Asunto(s)
Infecciones Bacterianas/patología , Fluorescencia , Imagen de Cuerpo Entero/métodos , Animales , Bacterias/patogenicidad , Bronconeumonía/patología , Modelos Animales de Enfermedad , Colorantes Fluorescentes/metabolismo , Genes Reporteros , Enfermedades Cutáneas Bacterianas/patología , Infecciones de los Tejidos Blandos/patología
19.
Antimicrob Agents Chemother ; 55(6): 3058-62, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21464254

RESUMEN

Tuberculosis is a leading cause of death worldwide. Resistance of Mycobacterium to antibiotics can make treatments less effective in some cases. We tested selected oligopeptoids--previously reported as mimics of natural host defense peptides--for activity against Mycobacterium tuberculosis and assessed their cytotoxicity. A tetrameric, alkylated, cationic peptoid (1-C13(4mer)) was most potent against M. tuberculosis and least cytotoxic, whereas an unalkylated analogue, peptoid 1(4mer), was inactive. Peptoid 1-C13(4mer) thus merits further study as a potential antituberculosis drug.


Asunto(s)
Antibacterianos/farmacología , Mycobacterium tuberculosis/efectos de los fármacos , Peptoides/farmacología , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Macrófagos/efectos de los fármacos , Ratones , Mycobacterium bovis/efectos de los fármacos
20.
J Vis Exp ; (48)2011 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-21372790

RESUMEN

Imaging is a valuable technique that can be used to monitor biological processes. In particular, the presence of cancer cells, stem cells, specific immune cell types, viral pathogens, parasites and bacteria can be followed in real-time within living animals. Application of bioluminescence imaging to the study of pathogens has advantages as compared to conventional strategies for analysis of infections in animal models. Infections can be visualized within individual animals over time, without requiring euthanasia to determine the location and quantity of the pathogen. Optical imaging allows comprehensive examination of all tissues and organs, rather than sampling of sites previously known to be infected. In addition, the accuracy of inoculation into specific tissues can be directly determined prior to carrying forward animals that were unsuccessfully inoculated throughout the entire experiment. Variability between animals can be controlled for, since imaging allows each animal to be followed individually. Imaging has the potential to greatly reduce animal numbers needed because of the ability to obtain data from numerous time points without having to sample tissues to determine pathogen load. This protocol describes methods to visualize infections in live animals using bioluminescence imaging for recombinant strains of bacteria expressing luciferase. The click beetle (CBRLuc) and firefly luciferases (FFluc) utilize luciferin as a substrate. The light produced by both CBRluc and FFluc has a broad wavelength from 500 nm to 700 nm, making these luciferases excellent reporters for the optical imaging in living animal models. This is primarily because wavelengths of light greater than 600 nm are required to avoid absorption by hemoglobin and, thus, travel through mammalian tissue efficiently. Luciferase is genetically introduced into the bacteria to produce light signal. Mice are pulmonary inoculated with bioluminescent bacteria intratracheally to allow monitoring of infections in real time. After luciferin injection, images are acquired using the IVIS Imaging System. During imaging, mice are anesthetized with isoflurane using an XGI-8 Gas Anethesia System. Images can be analyzed to localize and quantify the signal source, which represents the bacterial infection site(s) and number, respectively. After imaging, CFU determination is carried out on homogenized tissue to confirm the presence of bacteria. Several doses of bacteria are used to correlate bacterial numbers with luminescence. Imaging can be applied to study of pathogenesis and evaluation of the efficacy of antibacterial compounds and vaccines.


Asunto(s)
Bacterias/enzimología , Infecciones Bacterianas/microbiología , Luciferasas/análisis , Mediciones Luminiscentes/métodos , Animales , Bacterias/genética , Infecciones Bacterianas/diagnóstico , Procesamiento de Imagen Asistido por Computador , Imagenología Tridimensional , Luciferasas/genética , Luciferasas/metabolismo , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA