Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros











Intervalo de año de publicación
1.
Methods Mol Biol ; 2813: 137-144, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38888776

RESUMEN

Air-liquid interface (ALI) airway culture models serve as a powerful tool to emulate the characteristic features of the respiratory tract in vitro. These models are particularly valuable for studying emerging respiratory viral and bacterial infections. Here, we describe an optimized protocol to obtain the ALI airway culture models using normal human bronchial epithelial cells (NHBECs). The protocol outlined below enables the generation of differentiated mucociliary airway epithelial cultures by day 28 following exposure to air.


Asunto(s)
Técnicas de Cultivo de Célula , Células Epiteliales , Humanos , Técnicas de Cultivo de Célula/métodos , Células Epiteliales/microbiología , Células Epiteliales/virología , Células Epiteliales/citología , Bronquios/citología , Mucosa Respiratoria/citología , Mucosa Respiratoria/microbiología , Mucosa Respiratoria/virología , Aire , Células Cultivadas , Enfermedades Transmisibles/microbiología
2.
EMBO J ; 42(9): e113490, 2023 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-36920246

RESUMEN

Mycobacterium tuberculosis (Mtb) infection is initiated by inhalation of bacteria into lung alveoli, where they are phagocytosed by resident macrophages. Intracellular Mtb replication induces the death of the infected macrophages and the release of bacterial aggregates. Here, we show that these aggregates can evade phagocytosis by killing macrophages in a contact-dependent but uptake-independent manner. We use time-lapse fluorescence microscopy to show that contact with extracellular Mtb aggregates triggers macrophage plasma membrane perturbation, cytosolic calcium accumulation, and pyroptotic cell death. These effects depend on the Mtb ESX-1 secretion system, however, this system alone cannot induce calcium accumulation and macrophage death in the absence of the Mtb surface-exposed lipid phthiocerol dimycocerosate. Unexpectedly, we found that blocking ESX-1-mediated secretion of the EsxA/EsxB virulence factors does not eliminate the uptake-independent killing of macrophages and that the 50-kDa isoform of the ESX-1-secreted protein EspB can mediate killing in the absence of EsxA/EsxB secretion. Treatment with an ESX-1 inhibitor reduces uptake-independent killing of macrophages by Mtb aggregates, suggesting that novel therapies targeting this anti-phagocytic mechanism could prevent the propagation of extracellular bacteria within the lung.


Asunto(s)
Mycobacterium tuberculosis , Proteínas Bacterianas/metabolismo , Calcio/metabolismo , Macrófagos/metabolismo , Factores de Virulencia/metabolismo
3.
Elife ; 102021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-34219648

RESUMEN

Uropathogenic Escherichia coli (UPEC) proliferate within superficial bladder umbrella cells to form intracellular bacterial communities (IBCs) during early stages of urinary tract infections. However, the dynamic responses of IBCs to host stresses and antibiotic therapy are difficult to assess in situ. We develop a human bladder-chip model wherein umbrella cells and bladder microvascular endothelial cells are co-cultured under flow in urine and nutritive media respectively, and bladder filling and voiding mimicked mechanically by application and release of linear strain. Using time-lapse microscopy, we show that rapid recruitment of neutrophils from the vascular channel to sites of infection leads to swarm and neutrophil extracellular trap formation but does not prevent IBC formation. Subsequently, we tracked bacterial growth dynamics in individual IBCs through two cycles of antibiotic administration interspersed with recovery periods which revealed that the elimination of bacteria within IBCs by the antibiotic was delayed, and in some instances, did not occur at all. During the recovery period, rapid proliferation in a significant fraction of IBCs reseeded new foci of infection through bacterial shedding and host cell exfoliation. These insights reinforce a dynamic role for IBCs as harbors of bacterial persistence, with significant consequences for non-compliance with antibiotic regimens.


Urinary tract infections are one of the most common reasons people need antibiotics. These bacterial infections are typically caused by uropathogenic Escherichia coli (also known as UPEC), which either float freely in the urine and wash away when the bladder empties, or form communities inside cells that the bladder struggles to clear. It is possible that the bacteria living within cells are also more protected from the immune system and antibiotics. But this is hard to study in animal models. To overcome this, Sharma et al. built a 'bladder-chip' which mimics the interface between the blood vessels and the tissue layers of the human bladder. Similar chip devices have also been made for other organs. However, until now, no such model had been developed for the bladder. On the chip created by Sharma et al. is a layer of bladder cells which sit at the bottom of a channel filled with diluted human urine. These cells were infected with UPEC, and then imaged over time to see how the bacteria moved, interacted with the bladder cells, and aggregated together. Immune cells from human blood were then added to a vascular channel underneath the bladder tissue, which is coated with endothelial cells that normally line blood vessels. The immune cells rapidly crossed the endothelial barrier and entered the bladder tissue, and swarmed around sites of infection. In some instances, they released the contents of their cells to form net-like traps to catch the bacteria. But these traps failed to remove the bacteria living inside bladder cells. Antibiotics were then added to the urine flowing over the bladder cells as well as the vascular channel, similar to how drugs would be delivered in live human tissue. Sharma et al. discovered that the antibiotics killed bacteria residing in bladder cells slower than bacteria floating freely in the urine. Furthermore, they found that bacteria living in tightly packed communities within bladder cells were more likely to survive treatment and go on to re-infect other parts of the tissue. Antibiotic resistance is a pressing global challenge, and recurrent urinary tract infections are a significant contributor. The bladder-chip presented here could further our understanding of how these bacterial infections develop in vivo and how good antibiotics are at removing them. This could help researchers identify the best dosing and treatment strategies, as well as provide a platform for rapidly testing new antibiotic drugs and other therapies.


Asunto(s)
Técnicas Bacteriológicas/instrumentación , Dispositivos Laboratorio en un Chip , Vejiga Urinaria/irrigación sanguínea , Infecciones Urinarias/microbiología , Escherichia coli Uropatógena , Línea Celular Tumoral , Técnicas de Cocultivo , Células Endoteliales/fisiología , Humanos , Neutrófilos/fisiología
4.
ACS Chem Biol ; 13(11): 3184-3192, 2018 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-30289689

RESUMEN

Benzothiazinones (BTZ) are highly potent bactericidal inhibitors of mycobacteria and the lead compound, BTZ043, and the optimized drug candidate, PBTZ169, have potential for the treatment of tuberculosis. Here, we exploited the tractability of the BTZ scaffold by attaching a range of fluorophores to the 2-substituent of the BTZ ring via short linkers. We show by means of fluorescence imaging that the most advanced derivative, JN108, is capable of efficiently labeling its target, the essential flavoenzyme DprE1, both in cell-free extracts and after purification as well as in growing cells of different actinobacterial species. DprE1 displays a polar localization in Mycobacterium tuberculosis, M. marinum, M. smegmatis, and Nocardia farcinica but not in Corynebacterium glutamicum. Finally, mutation of the cysteine residue in DprE1 in these species, to which BTZ covalently binds, abolishes completely the interaction with JN108, thereby highlighting the specificity of this fluorescent probe.


Asunto(s)
Marcadores de Afinidad/farmacología , Oxidorreductasas de Alcohol/antagonistas & inhibidores , Antituberculosos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Tiazinas/farmacología , Actinomycetales/efectos de los fármacos , Actinomycetales/enzimología , Marcadores de Afinidad/síntesis química , Oxidorreductasas de Alcohol/genética , Antituberculosos/síntesis química , Proteínas Bacterianas/genética , Membrana Celular/metabolismo , Diseño de Fármacos , Inhibidores Enzimáticos/síntesis química , Fluoresceínas/síntesis química , Fluoresceínas/farmacología , Fluorescencia , Colorantes Fluorescentes/síntesis química , Colorantes Fluorescentes/farmacología , Células Hep G2 , Humanos , Pruebas de Sensibilidad Microbiana , Microscopía Fluorescente/métodos , Mutación , Tiazinas/síntesis química
5.
IUBMB Life ; 70(9): 836-844, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30092117

RESUMEN

Bacterial persistence, the ability of bacteria to survive high concentrations of antibiotics for extended periods of time, is an important contributing factor to therapy failure and development of chronic and recurrent infections. Several recent studies have suggested that this persistence is mediated primarily by (p)ppGpp, through its interactions with toxin-antitoxin modules and polyphosphates. In this study, we address whether these key players play a role in mycobacterial persistence against antibiotics. We targeted these specific pathways in Mycobacterium smegmatis by constructing deletion strains of (p)ppGpp synthetase/hydrolase (relA), polyphosphate kinases (ppk1 and ppk2), exopolyphosphatases (ppx1 and ppx2), and the lon protease. None of these mutant strains exhibited altered levels of persisters against isoniazid and ciprofloxacin, when compared with wild-type strain. Even under conditions in which the stringent response usually gets activated, these strains displayed wild-type persister levels. Interestingly, we also found that unlike Escherichia coli, maintaining M. smegmatis in exponential phase by repeated passaging does not eliminate persisters suggesting that at least against the antibiotics tested, stationary-phase dependent persisters (type I) are not the major contributors. Thus, our data demonstrate that multiple mechanisms of antibiotic persistence exist and that these vary widely among different bacterial species. © 2018 IUBMB Life, 70(9):836-844, 2018.


Asunto(s)
Antibacterianos/farmacología , Farmacorresistencia Bacteriana , Guanosina Pentafosfato/metabolismo , Mycobacterium smegmatis/efectos de los fármacos , Tuberculosis/tratamiento farmacológico , Humanos , Mycobacterium smegmatis/crecimiento & desarrollo , Mycobacterium smegmatis/metabolismo , Fosfotransferasas (Aceptor del Grupo Fosfato)/metabolismo , Tuberculosis/metabolismo , Tuberculosis/microbiología
6.
Mol Microbiol ; 93(5): 1057-1065, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25039394

RESUMEN

The function of EspI, a 70 kDa protein in Mycobacterium tuberculosis, has remained unclear. Although EspI is encoded by a gene within the esx-1 locus, in this study we clarify previous conflicting results and show that EspI is not essential for ESX-1-mediated secretion or virulence in M. tuberculosis. We also provide evidence that reduction of cellular ATP levels in wild-type M. tuberculosis using the drug bedaquiline completely blocks ESX-1-mediated secretion. Remarkably, M. tuberculosis lacking EspI fails to exhibit this phenotype. Furthermore, mutagenesis of a highly conserved ATP-binding motif in EspI renders M. tuberculosis incapable of shutting down ESX-1-mediated secretion during ATP depletion. Collectively these results show that M. tuberculosis EspI negatively regulates the ESX-1 secretion system in response to low cellular ATP levels and this function requires the ATP-binding motif. In light of our results the potential significance of EspI in ESX-1 function during latent tuberculosis infection and reactivation is also discussed.


Asunto(s)
Adenosina Trifosfato/metabolismo , Proteínas Bacterianas/metabolismo , Sistemas de Secreción Bacterianos , Regulación Bacteriana de la Expresión Génica , Mycobacterium tuberculosis/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Datos de Secuencia Molecular , Mycobacterium tuberculosis/química , Mycobacterium tuberculosis/genética , Alineación de Secuencia , Transducción de Señal
7.
J Bacteriol ; 196(19): 3441-51, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25049093

RESUMEN

The cell envelope of Mycobacterium tuberculosis contains glycans and lipids of peculiar structure that play prominent roles in the biology and pathogenesis of tuberculosis. Consequently, the chemical structure and biosynthesis of the cell wall have been intensively investigated in order to identify novel drug targets. Here, we validate that the function of phosphatidyl-myo-inositol mannosyltransferase PimA is vital for M. tuberculosis in vitro and in vivo. PimA initiates the biosynthesis of phosphatidyl-myo-inositol mannosides by transferring a mannosyl residue from GDP-Man to phosphatidyl-myo-inositol on the cytoplasmic side of the plasma membrane. To prove the essential nature of pimA in M. tuberculosis, we constructed a pimA conditional mutant by using the TetR-Pip off system and showed that downregulation of PimA expression causes bactericidality in batch cultures. Consistent with the biochemical reaction catalyzed by PimA, this phenotype was associated with markedly reduced levels of phosphatidyl-myo-inositol dimannosides, essential structural components of the mycobacterial cell envelope. In addition, the requirement of PimA for viability was clearly demonstrated during macrophage infection and in two different mouse models of infection, where a dramatic decrease in viable counts was observed upon silencing of the gene. Notably, depletion of PimA resulted in complete clearance of the mouse lungs during both the acute and chronic phases of infection. Altogether, the experimental data highlight the importance of the phosphatidyl-myo-inositol mannoside biosynthetic pathway for M. tuberculosis and confirm that PimA is a novel target for future drug discovery programs.


Asunto(s)
Proteínas Bacterianas/metabolismo , Manosiltransferasas/metabolismo , Mycobacterium tuberculosis/enzimología , Mycobacterium tuberculosis/crecimiento & desarrollo , Tuberculosis/microbiología , Animales , Proteínas Bacterianas/genética , Femenino , Humanos , Macrófagos/metabolismo , Macrófagos/microbiología , Manosiltransferasas/genética , Ratones , Ratones Endogámicos C57BL , Mycobacterium tuberculosis/genética , Fosfatidilinositoles/biosíntesis
8.
J Med Chem ; 57(12): 5419-34, 2014 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-24871036

RESUMEN

4-Aminoquinolone piperidine amides (AQs) were identified as a novel scaffold starting from a whole cell screen, with potent cidality on Mycobacterium tuberculosis (Mtb). Evaluation of the minimum inhibitory concentrations, followed by whole genome sequencing of mutants raised against AQs, identified decaprenylphosphoryl-ß-d-ribose 2'-epimerase (DprE1) as the primary target responsible for the antitubercular activity. Mass spectrometry and enzyme kinetic studies indicated that AQs are noncovalent, reversible inhibitors of DprE1 with slow on rates and long residence times of ∼100 min on the enzyme. In general, AQs have excellent leadlike properties and good in vitro secondary pharmacology profile. Although the scaffold started off as a single active compound with moderate potency from the whole cell screen, structure-activity relationship optimization of the scaffold led to compounds with potent DprE1 inhibition (IC50 < 10 nM) along with potent cellular activity (MIC = 60 nM) against Mtb.


Asunto(s)
Amidas/química , Antituberculosos/química , Proteínas Bacterianas/antagonistas & inhibidores , Mycobacterium tuberculosis/efectos de los fármacos , Oxidorreductasas/antagonistas & inhibidores , Piperidinas/química , Quinolonas/química , Oxidorreductasas de Alcohol , Amidas/farmacocinética , Amidas/farmacología , Animales , Antituberculosos/farmacocinética , Antituberculosos/farmacología , Dominio Catalítico , Línea Celular Tumoral , Farmacorresistencia Bacteriana , Genoma Bacteriano , Humanos , Cinética , Pruebas de Sensibilidad Microbiana , Simulación del Acoplamiento Molecular , Mutación , Mycobacterium tuberculosis/enzimología , Mycobacterium tuberculosis/genética , Piperidinas/farmacocinética , Piperidinas/farmacología , Unión Proteica , Quinolonas/farmacocinética , Quinolonas/farmacología , Ratas Wistar , Estereoisomerismo , Relación Estructura-Actividad
9.
Mol Microbiol ; 92(1): 194-211, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24517327

RESUMEN

In Mycobacterium tuberculosis the decaprenyl-phospho-d-arabinofuranose (DPA) pathway is a validated target for the drugs ethambutol and benzothiazinones. To identify other potential drug targets in the pathway, we generated conditional knock-down mutants of each gene involved using the TET-PIP OFF system. dprE1, dprE2, ubiA, prsA, rv2361c, tkt and rpiB were confirmed to be essential under non-permissive conditions, whereas rv3807c was not required for survival. In the most vulnerable group, DprE1-depleted cells died faster in vitro and intracellularly than those lacking UbiA and PrsA. Downregulation of DprE1 and UbiA resulted in similar phenotypes, namely swelling of the bacteria, cell wall damage and lysis as observed at the single cell level, by real time microscopy and electron microscopy. By contrast, depletion of PrsA led to cell elongation and implosion, which was suggestive of a more pleiotropic effect. Drug sensitivity assays with known DPA-inhibitors supported the use of conditional knock-down strains for target-based whole-cell screens. Together, our work provides strong evidence for the vulnerability of all but one of the enzymes in the DPA pathway and generates valuable tools for the identification of lead compounds targeting the different biosynthetic steps. PrsA, phosphoribosyl-pyrophosphate synthetase, appears to be a particularly attractive new target for drug discovery.


Asunto(s)
Arabinosa/análogos & derivados , Genes Bacterianos , Mycobacterium tuberculosis/crecimiento & desarrollo , Transducción de Señal , Antibacterianos/farmacología , Arabinosa/antagonistas & inhibidores , Arabinosa/biosíntesis , Proteínas Bacterianas , Línea Celular Tumoral , Pared Celular/ultraestructura , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Genes Bacterianos/efectos de los fármacos , Genes Esenciales/efectos de los fármacos , Humanos , Lipoproteínas , Macrófagos/microbiología , Proteínas de la Membrana , Microscopía , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/ultraestructura , Transducción de Señal/efectos de los fármacos
10.
Nat Commun ; 5: 3369, 2014 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-24569628

RESUMEN

Bedaquiline (BDQ), an ATP synthase inhibitor, is the first drug to be approved for treatment of multidrug-resistant tuberculosis in decades. Though BDQ has shown excellent efficacy in clinical trials, its early bactericidal activity during the first week of chemotherapy is minimal. Here, using microfluidic devices and time-lapse microscopy of Mycobacterium tuberculosis, we confirm the absence of significant bacteriolytic activity during the first 3-4 days of exposure to BDQ. BDQ-induced inhibition of ATP synthesis leads to bacteriostasis within hours after drug addition. Transcriptional and proteomic analyses reveal that M. tuberculosis responds to BDQ by induction of the dormancy regulon and activation of ATP-generating pathways, thereby maintaining bacterial viability during initial drug exposure. BDQ-induced bacterial killing is significantly enhanced when the mycobacteria are grown on non-fermentable energy sources such as lipids (impeding ATP synthesis via glycolysis). Our results show that BDQ exposure triggers a metabolic remodelling in mycobacteria, thereby enabling transient bacterial survival.


Asunto(s)
Diarilquinolinas/farmacología , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Glucólisis/efectos de los fármacos , Mycobacterium tuberculosis/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Antituberculosos/farmacología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Relación Dosis-Respuesta a Droga , Perfilación de la Expresión Génica/métodos , Viabilidad Microbiana/efectos de los fármacos , Viabilidad Microbiana/genética , Técnicas Analíticas Microfluídicas , Microscopía Fluorescente , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteoma/genética , Proteoma/metabolismo , Proteómica/métodos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de la Célula Individual/métodos , Factores de Tiempo , Imagen de Lapso de Tiempo
11.
J Bacteriol ; 195(24): 5421-30, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24078612

RESUMEN

The EspA protein of Mycobacterium tuberculosis is essential for the type VII ESX-1 protein secretion apparatus, which delivers the principal virulence factors ESAT-6 and CFP-10. In this study, site-directed mutagenesis of EspA was performed to elucidate its influence on the ESX-1 system. Replacing Trp(55) (W55) or Gly(57) (G57) residues in the putative W-X-G motif of EspA with arginines impaired ESAT-6 and CFP-10 secretion in vitro and attenuated M. tuberculosis. Replacing the Phe(50) (F50) and Lys(62) (K62) residues, which flank the W-X-G motif, with arginine and alanine, respectively, destabilized EspA, abolished ESAT-6 and CFP-10 secretion in vitro, and attenuated M. tuberculosis. Likewise, replacing the Phe(5) (F5) and Lys(41) (K41) residues with arginine and alanine, respectively, also destabilized EspA and blocked ESAT-6 and CFP-10 secretion in vitro. However, these two particular mutations did not attenuate M. tuberculosis in cellular models of infection or during acute infection in mice. We have thus identified amino acid residues in EspA that are important for facilitating ESAT-6 and CFP-10 secretion and virulence. However, our data also indicate for the first time that blockage of M. tuberculosis ESAT-6 and CFP-10 secretion in vitro and attenuation are mutually exclusive.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Mycobacterium tuberculosis/patogenicidad , Sustitución de Aminoácidos , Animales , Antígenos Bacterianos , Línea Celular , Modelos Animales de Enfermedad , Humanos , Macrófagos/microbiología , Ratones , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Tuberculosis/microbiología , Tuberculosis/patología , Virulencia
12.
Sci Transl Med ; 4(150): 150ra121, 2012 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-22956199

RESUMEN

The benzothiazinone BTZ043 is a tuberculosis drug candidate with nanomolar whole-cell activity. BTZ043 targets the DprE1 catalytic component of the essential enzyme decaprenylphosphoryl-ß-D-ribofuranose-2'-epimerase, thus blocking biosynthesis of arabinans, vital components of mycobacterial cell walls. Crystal structures of DprE1, in its native form and in a complex with BTZ043, reveal formation of a semimercaptal adduct between the drug and an active-site cysteine, as well as contacts to a neighboring catalytic lysine residue. Kinetic studies confirm that BTZ043 is a mechanism-based, covalent inhibitor. This explains the exquisite potency of BTZ043, which, when fluorescently labeled, localizes DprE1 at the poles of growing bacteria. Menaquinone can reoxidize the flavin adenine dinucleotide cofactor in DprE1 and may be the natural electron acceptor for this reaction in the mycobacterium. Our structural and kinetic analysis provides both insight into a critical epimerization reaction and a platform for structure-based design of improved inhibitors.


Asunto(s)
Antituberculosos/química , Antituberculosos/farmacología , Viabilidad Microbiana/efectos de los fármacos , Mycobacterium tuberculosis/efectos de los fármacos , Tiazinas/química , Tiazinas/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/química , Cristalografía por Rayos X , Cisteína/química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Flavina-Adenina Dinucleótido/análogos & derivados , Flavina-Adenina Dinucleótido/metabolismo , Flavoproteínas/química , Colorantes Fluorescentes/metabolismo , Cinética , Lisina/química , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Mycobacterium smegmatis/efectos de los fármacos , Mycobacterium smegmatis/enzimología , Mycobacterium tuberculosis/enzimología , Oxidación-Reducción/efectos de los fármacos , Oxidorreductasas/antagonistas & inhibidores , Oxidorreductasas/química , Estructura Terciaria de Proteína , Transporte de Proteínas/efectos de los fármacos , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo
13.
Vaccine ; 29(40): 6959-66, 2011 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-21787826

RESUMEN

Vaccines that drive robust T-cell immunity against Mycobacterium tuberculosis (Mtb) are needed both for prophylactic and therapeutic purposes. We have recently developed a synthetic vaccine delivery platform with Pluronic-stabilized polypropylene sulfide nanoparticles (NPs), which target lymphoid tissues by their small size (∼ 30 nm) and which activate the complement cascade by their surface chemistry. Here we conjugated the tuberculosis antigen Ag85B to the NPs (NP-Ag85B) and compared their efficacy in eliciting relevant immune responses in mice after intradermal or pulmonary administration. Pulmonary administration of NP-Ag85B with the adjuvant CpG led to enhanced induction of antigen-specific polyfunctional Th1 responses in the spleen, the lung and lung-draining lymph nodes as compared to soluble Ag85B with CpG and to the intradermally-delivered formulations. Mucosal and systemic Th17 responses were also observed with this adjuvanted NP formulation and vaccination route, especially in the lung. We then evaluated protection induced by the adjuvanted NP formulation following a Mtb aerosol challenge and found that vaccination with NP-Ag85B and CpG via the pulmonary route displayed a substantial reduction of the lung bacterial burden, both compared to soluble Ag85B with CpG and to the corresponding intradermally delivered formulations. These findings highlight the potential of administrating NP-based formulations by the pulmonary route for TB vaccination.


Asunto(s)
Aciltransferasas/inmunología , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Islas de CpG/inmunología , Inmunoconjugados/inmunología , Nanopartículas/administración & dosificación , Vacunas contra la Tuberculosis/administración & dosificación , Vacunas contra la Tuberculosis/inmunología , Tuberculosis/prevención & control , Animales , Antígenos Bacterianos/administración & dosificación , Proteínas del Sistema Complemento/inmunología , Células Dendríticas/inmunología , Inmunoconjugados/administración & dosificación , Inmunoconjugados/química , Pulmón/citología , Pulmón/inmunología , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos C57BL , Mycobacterium tuberculosis/inmunología , Nanopartículas/química , Tamaño de la Partícula , Polipropilenos/química , Bazo/inmunología , Células TH1/inmunología , Células Th17/inmunología , Tuberculosis/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología
14.
Proc Natl Acad Sci U S A ; 107(27): 12275-80, 2010 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-20566858

RESUMEN

Tuberculosis (TB) is notoriously difficult to cure, requiring administration of multiple antibiotics for 6 mo or longer. Conventional anti-TB drugs inhibit biosynthetic processes involved in cell growth and division, such as DNA replication, RNA transcription, protein translation, and cell wall biogenesis. Although highly effective against bacteria cultured in vitro under optimal growth conditions, these antibiotics are less effective against bacteria grown in vivo in the tissues of a mammalian host. The factors that contribute to the antibiotic tolerance of bacteria grown in vivo are unknown, although altered metabolism and sluggish growth are hypothesized to play a role. To address this question, we identified mutations in Mycobacterium tuberculosis that impaired or enhanced persistence in mice treated with isoniazid (INH), a front-line anti-TB drug. Disruption of cydC, encoding a putative ATP-binding cassette transporter subunit, accelerated bacterial clearance in INH-treated mice without affecting growth or survival in untreated mice. Conversely, transposon insertions within the rv0096-rv0101 gene cluster attenuated bacterial growth and survival in untreated mice but paradoxically prevented INH-mediated killing of bacteria in treated mice. These contrasting phenotypes were dependent on the interaction of the bacteria with the tissue environment because both mutants responded normally to INH when grown in macrophages ex vivo or in axenic cultures in vitro. Our findings have important implications because persistence-impairing mutations would be missed by conventional genetic screens to identify candidate drug targets. Conversely, persistence-enhancing mutations would be missed by standard diagnostic methods, which are performed on bacteria grown in vitro, to detect drug resistance.


Asunto(s)
Isoniazida/farmacología , Mutación , Mycobacterium tuberculosis/efectos de los fármacos , Tuberculosis/tratamiento farmacológico , Transportadoras de Casetes de Unión a ATP/genética , Animales , Antituberculosos/farmacología , Proteínas Bacterianas/genética , Células Cultivadas , Farmacorresistencia Bacteriana/genética , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Prueba de Complementación Genética , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pruebas de Sensibilidad Microbiana , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/crecimiento & desarrollo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Tuberculosis/microbiología
15.
PLoS One ; 3(12): e3869, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19052643

RESUMEN

BACKGROUND: The variable efficacy (0-80%) of Mycobacterium bovis Bacille Calmette Guréin (BCG) vaccine against adult tuberculosis (TB) necessitates development of alternative vaccine candidates. Development of recombinant BCG (rBCG) over-expressing promising immunodominant antigens of M. tuberculosis represents one of the potential approaches for the development of vaccines against TB. METHODS/PRINCIPAL FINDINGS: A recombinant strain of BCG - rBCG85C, over expressing the antigen 85C, a secretory immuno-dominant protein of M. tuberculosis, was evaluated for its protective efficacy in guinea pigs against M. tuberculosis challenge by aerosol route. Immunization with rBCG85C resulted in a substantial reduction in the lung (1.87 log(10), p<0.01) and spleen (2.36 log(10), p<0.001) bacillary load with a commensurate reduction in pathological damage, when compared to the animals immunized with the parent BCG strain at 10 weeks post-infection. rBCG85C continued to provide superior protection over BCG even when post-challenge period was prolonged to 16 weeks. The cytokine profile of pulmonary granulomas revealed that the superior protection imparted by rBCG85C was associated with the reduced levels of pro-inflammatory cytokines - interleukin (IL)-12, interferon (IFN)-gamma, tumor necrosis factor (TNF)-alpha, moderate levels of anti-inflammatory cytokine - transforming growth factor (TGF)-beta along with up-regulation of inducible nitric oxide synthase (iNOS). In addition, the rBCG85C vaccine induced modulation of the cytokine levels was found to be associated with reduced fibrosis and antigen load accompanied by the restoration of normal lung architecture. CONCLUSIONS/SIGNIFICANCE: These results clearly indicate the superiority of rBCG85C over BCG as a promising prophylactic vaccine against TB. The enduring protection observed in this study gives enough reason to postulate that if an open-ended study is carried out with low dose of infection, rBCG85C vaccine in all likelihood would show enhanced survival of guinea pigs.


Asunto(s)
Vacuna BCG , Citocinas/metabolismo , Pulmón/inmunología , Tuberculosis/prevención & control , Animales , Vacuna BCG/genética , Vacuna BCG/inmunología , Femenino , Cobayas , Inmunohistoquímica , Pulmón/microbiología , Mycobacterium bovis/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Tuberculosis/inmunología
16.
Mol Microbiol ; 50(3): 751-62, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14617138

RESUMEN

Protein tyrosine kinases and tyrosine phosphatases from several bacterial pathogens have been shown to act as virulence factors by modulating the phosphorylation and dephosphorylation of host proteins. The identification and characterization of two tyrosine phosphatases namely MptpA and MptpB from Mycobacterium tuberculosis has been reported earlier. MptpB is secreted by M. tuberculosis into extracellular mileu and exhibits a pH optimum of 5.6, similar to the pH of the lysosomal compartment of the cell. To determine the role of MptpB in the pathogenesis of M. tuberculosis, we constructed a mptpB mutant strain by homologous recombination and compared the ability of parent and the mutant strain to survive intracellularly. We show that disruption of the mptpB gene impairs the ability of the mutant strain to survive in activated macrophages and guinea pigs but not in resting macrophages suggesting the importance of its role in the host-pathogen interaction. Infection of guinea pigs with the mutant strain resulted in a 70-fold reduction in the bacillary load of spleens in infected animals as compared with the bacillary load in animals infected with the parental strain. Upon reintroduction of the mptpB gene into the mutant strain, the complemented strain was able to establish infection and survive in guinea pigs at rates comparable to the parental strain. These observations demonstrate a role of MptpB in the pathogenesis of M. tuberculosis.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Mycobacterium tuberculosis/patogenicidad , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Animales , Células Cultivadas , Prueba de Complementación Genética , Cobayas , Interacciones Huésped-Parásitos/genética , Macrófagos/microbiología , Ratones , Mutación , Mycobacterium tuberculosis/genética , Recombinación Genética , Bazo/microbiología , Bazo/patología , Tuberculosis/microbiología , Tuberculosis/patología , Virulencia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA