Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
Immunity ; 54(3): 526-541.e7, 2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33515487

RESUMEN

Chronic viral infections increase severity of Mycobacterium tuberculosis (Mtb) coinfection. Here, we examined how chronic viral infections alter the pulmonary microenvironment to foster coinfection and worsen disease severity. We developed a coordinated system of chronic virus and Mtb infection that induced central clinical manifestations of coinfection, including increased Mtb burden, extra-pulmonary dissemination, and heightened mortality. These disease states were not due to chronic virus-induced immunosuppression or exhaustion; rather, increased amounts of the cytokine TNFα initially arrested pulmonary Mtb growth, impeding dendritic cell mediated antigen transportation to the lymph node and subverting immune-surveillance, allowing bacterial sanctuary. The cryptic Mtb replication delayed CD4 T cell priming, redirecting T helper (Th) 1 toward Th17 differentiation and increasing pulmonary neutrophilia, which diminished long-term survival. Temporally restoring CD4 T cell induction overcame these diverse disease sequelae to enhance Mtb control. Thus, Mtb co-opts TNFα from the chronic inflammatory environment to subvert immune-surveillance, avert early immune function, and foster long-term coinfection.


Asunto(s)
Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/fisiología , Mycobacterium tuberculosis/fisiología , Neutrófilos/inmunología , Células TH1/inmunología , Células Th17/inmunología , Tuberculosis/inmunología , Inmunidad Adaptativa , Animales , Enfermedad Crónica , Coinfección , Inmunidad Innata , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fagocitosis , Índice de Severidad de la Enfermedad , Factores de Tiempo
2.
Cell ; 160(5): 940-951, 2015 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-25723168

RESUMEN

Type VI secretion systems (T6SSs) are newly identified contractile nanomachines that translocate effector proteins across bacterial membranes. The Francisella pathogenicity island, required for bacterial phagosome escape, intracellular replication, and virulence, was presumed to encode a T6SS-like apparatus. Here, we experimentally confirm the identity of this T6SS and, by cryo electron microscopy (cryoEM), show the structure of its post-contraction sheath at 3.7 Å resolution. We demonstrate the assembly of this T6SS by IglA/IglB and secretion of its putative effector proteins in response to environmental stimuli. The sheath has a quaternary structure with handedness opposite that of contracted sheath of T4 phage tail and is organized in an interlaced two-dimensional array by means of ß sheet augmentation. By structure-based mutagenesis, we show that this interlacing is essential to secretion, phagosomal escape, and intracellular replication. Our atomic model of the T6SS will facilitate design of drugs targeting this highly prevalent secretion apparatus.


Asunto(s)
Proteínas Bacterianas/química , Sistemas de Secreción Bacterianos , Francisella/ultraestructura , Proteínas Bacterianas/ultraestructura , Bacteriófago T4/química , Bacteriófagos/química , Microscopía por Crioelectrón , Modelos Moleculares , Estructura Secundaria de Proteína
3.
Acc Chem Res ; 52(6): 1531-1542, 2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31082188

RESUMEN

Mesoporous silica nanoparticles (MSNs) are delivery vehicles that can carry cargo molecules and release them on command. The particles used in the applications reported in this Account are around 100 nm in diameter (about the size of a virus) and contain 2.5 nm tubular pores with a total volume of about 1 cm3/g. For the biomedical applications discussed here, the cargo is trapped in the pores until the particles are stimulated to release it. The challenges are to get the particles to the site of a disease and then to deliver the cargo on command. We describe methods to do both, and we illustrate the applicability of the particles to cure cancer and intracellular infectious disease. Our first steps were to design multifunctional nanoparticles with properties that allow them to carry and deliver hydrophobic drugs. Many important pharmaceuticals are hydrophobic and cannot reach the diseased sites by themselves. We describe how we modified MSNs to make them dispersible, imagable, and targetable and discuss in vitro studies. We then present examples of surface modifications that allow them to deliver large molecules such as siRNA. In vivo studies of siRNA delivery to treat triple-negative breast and ovarian cancers are presented. The next steps are to attach nanomachines and other types of caps that trap drug molecules but release them when stimulated. We describe nanomachines that respond autonomously (without human intervention) to stimuli specific to disease sites. A versatile type of machine is a nanovalve that is closed at neutral (blood) pH but opens upon acidification that occurs in endolysosomes of cancer cells. Another type of machine, a snap-top cap, is stimulated by reducing agents such as glutathione in the cytosol of cells. Both of these platforms were studied in vitro to deliver antibiotics to infected macrophages and in vivo to cure and kill the intracellular bacteria M. tuberculosis and F. tularensis. The latter is a tier 1 select agent of bioterrorism. Finally, we describe nanomachines for drug delivery that are controlled by externally administered light and magnetic fields. A futuristic dream for nanotherapy is the ability to control a nano-object everywhere in the body. Magnetic fields penetrate completely and have spatial selectivity governed by the size of the field-producing coil. We describe how to control nanovalves with alternating magnetic fields (AMFs) and superparamagnetic cores inside the MSNs. The AMF heats the cores, and temperature-sensitive caps release the cargo. In vitro studies demonstrate dose control of the therapeutic to cause apoptosis without overheating the cells. Nanocarriers have great promise for therapeutic applications, and MSNs that can carry drugs to the site of a disease to produce a high local concentration without premature release and off-target damage may have the capability of realizing this goal.


Asunto(s)
Portadores de Fármacos/química , Nanopartículas/química , Nanotecnología/métodos , Dióxido de Silicio/química , Animales , Antineoplásicos/farmacología , Antituberculosos/farmacología , Liberación de Fármacos , Calefacción , Humanos , Fenómenos Magnéticos , Ratones , ARN Interferente Pequeño/farmacología
4.
Immunity ; 34(5): 807-19, 2011 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-21596592

RESUMEN

Cell-mediated adaptive immunity is critical for host defense, but little is known about T cell behavior during delivery of effector function. Here we investigate relationships among antigen presentation, T cell motility, and local production of effector cytokines by CD4+ T cells within hepatic granulomas triggered by Bacille Calmette-Guérin or Mycobacterium tuberculosis. At steady-state, only small fractions of mycobacteria-specific T cells showed antigen-induced migration arrest within granulomas, resulting in low-level, polarized secretion of cytokines. However, exogenous antigen elicited rapid arrest and robust cytokine production by the vast majority of effector T cells. These findings suggest that limited antigen presentation and/or recognition within granulomas evoke a muted T cell response drawing on only a fraction of the host's potential effector capacity. Our results provide new insights into the regulation of host-protective functions, especially how antigen availability influences T cell dynamics and, in turn, effector T cell function during chronic infection.


Asunto(s)
Presentación de Antígeno , Granuloma/inmunología , Hepatopatías/inmunología , Mycobacterium bovis/inmunología , Mycobacterium tuberculosis/inmunología , Linfocitos T/inmunología , Animales , Movimiento Celular , Células Cultivadas , Citocinas/biosíntesis , Citocinas/inmunología , Granuloma/microbiología , Hepatopatías/microbiología , Ratones , Ratones Endogámicos C57BL , Linfocitos T/citología
5.
Infect Immun ; 87(2)2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30455201

RESUMEN

Mycobacterium tuberculosis, one of the world's leading causes of death, must acquire nutrients, such as iron, from the host to multiply and cause disease. Iron is an essential metal and M. tuberculosis possesses two different systems to acquire iron from its environment: siderophore-mediated iron acquisition (SMIA) and heme-iron acquisition (HIA), involving uptake and degradation of heme to release ferrous iron. We have discovered that Mycobacterium bovis BCG, the tuberculosis vaccine strain, is severely deficient in HIA, and we exploited this phenotypic difference between BCG and M. tuberculosis to identify genes involved in HIA by complementing BCG's defect with a fosmid library. We identified ppe37, an iron-regulated PPE family gene, as being essential for HIA. BCG complemented with M. tuberculosisppe37 exhibits HIA as efficient as that of M. tuberculosis, achieving robust growth with <0.2 µM hemin. Conversely, deletion of ppe37 from M. tuberculosis results in a strain severely attenuated in HIA, with a phenotype nearly identical to that of BCG, requiring a 200-fold higher concentration of hemin to achieve growth equivalent to that of its parental strain. A nine-amino-acid deletion near the N terminus of BCG PPE37 (amino acids 31 to 39 of the M. tuberculosis PPE37 protein) underlies BCG's profound defect in HIA. Significant genetic variability exists in ppe37 genes across different M. tuberculosis strains, with more than 60% of sequences from completely sequenced M. tuberculosis genomes having mutations that result in altered PPE37 proteins; furthermore, these altered PPE37 proteins are nonfunctional in HIA. Our findings should allow delineation of the relative roles of HIA and SMIA in M. tuberculosis pathogenesis.


Asunto(s)
Proteínas Bacterianas/fisiología , Hemo/metabolismo , Proteínas de Unión a Hierro/inmunología , Proteínas de la Membrana/fisiología , Mycobacterium bovis/inmunología , Mycobacterium tuberculosis/inmunología , Vacuna BCG/inmunología , Proteínas Bacterianas/inmunología , ADN Bacteriano/genética , Proteínas de la Membrana/inmunología , Mycobacterium bovis/genética , Mycobacterium tuberculosis/genética , Análisis de Secuencia de ADN , Eliminación de Secuencia , Tuberculosis/microbiología , Vacunas contra la Tuberculosis , Virulencia/genética
6.
Blood ; 130(3): 245-257, 2017 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-28465342

RESUMEN

The iron-regulatory hormone hepcidin is induced early in infection, causing iron sequestration in macrophages and decreased plasma iron; this is proposed to limit the replication of extracellular microbes, but could also promote infection with macrophage-tropic pathogens. The mechanisms by which hepcidin and hypoferremia modulate host defense, and the spectrum of microbes affected, are poorly understood. Using mouse models, we show that hepcidin was selectively protective against siderophilic extracellular pathogens (Yersinia enterocolitica O9) by controlling non-transferrin-bound iron (NTBI) rather than iron-transferrin concentration. NTBI promoted the rapid growth of siderophilic but not nonsiderophilic bacteria in mice with either genetic or iatrogenic iron overload and in human plasma. Hepcidin or iron loading did not affect other key components of innate immunity, did not indiscriminately promote intracellular infections (Mycobacterium tuberculosis), and had no effect on extracellular nonsiderophilic Y enterocolitica O8 or Staphylococcus aureus Hepcidin analogs may be useful for treatment of siderophilic infections.


Asunto(s)
Infecciones Relacionadas con Catéteres/inmunología , Hemocromatosis/inmunología , Hepcidinas/inmunología , Sobrecarga de Hierro/inmunología , Hierro/metabolismo , Infecciones Estafilocócicas/inmunología , Animales , Unión Competitiva , Infecciones Relacionadas con Catéteres/metabolismo , Infecciones Relacionadas con Catéteres/microbiología , Infecciones Relacionadas con Catéteres/mortalidad , Modelos Animales de Enfermedad , Resistencia a la Enfermedad , Expresión Génica , Hemocromatosis/metabolismo , Hemocromatosis/microbiología , Hemocromatosis/mortalidad , Hepcidinas/agonistas , Hepcidinas/deficiencia , Hepcidinas/genética , Humanos , Hierro/inmunología , Sobrecarga de Hierro/metabolismo , Sobrecarga de Hierro/microbiología , Sobrecarga de Hierro/mortalidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/crecimiento & desarrollo , Mycobacterium tuberculosis/metabolismo , Oligopéptidos/farmacología , Unión Proteica , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/mortalidad , Staphylococcus aureus , Análisis de Supervivencia , Transferrina/genética , Transferrina/metabolismo , Yersinia enterocolitica/efectos de los fármacos , Yersinia enterocolitica/crecimiento & desarrollo , Yersinia enterocolitica/metabolismo
7.
Proc Natl Acad Sci U S A ; 113(15): E2172-9, 2016 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-27035987

RESUMEN

Tuberculosis (TB) remains a major global public health problem, and improved treatments are needed to shorten duration of therapy, decrease disease burden, improve compliance, and combat emergence of drug resistance. Ideally, the most effective regimen would be identified by a systematic and comprehensive combinatorial search of large numbers of TB drugs. However, optimization of regimens by standard methods is challenging, especially as the number of drugs increases, because of the extremely large number of drug-dose combinations requiring testing. Herein, we used an optimization platform, feedback system control (FSC) methodology, to identify improved drug-dose combinations for TB treatment using a fluorescence-based human macrophage cell culture model of TB, in which macrophages are infected with isopropyl ß-D-1-thiogalactopyranoside (IPTG)-inducible green fluorescent protein (GFP)-expressing Mycobacterium tuberculosis (Mtb). On the basis of only a single screening test and three iterations, we identified highly efficacious three- and four-drug combinations. To verify the efficacy of these combinations, we further evaluated them using a methodologically independent assay for intramacrophage killing of Mtb; the optimized combinations showed greater efficacy than the current standard TB drug regimen. Surprisingly, all top three- and four-drug optimized regimens included the third-line drug clofazimine, and none included the first-line drugs isoniazid and rifampin, which had insignificant or antagonistic impacts on efficacy. Because top regimens also did not include a fluoroquinolone or aminoglycoside, they are potentially of use for treating many cases of multidrug- and extensively drug-resistant TB. Our study shows the power of an FSC platform to identify promising previously unidentified drug-dose combinations for treatment of TB.


Asunto(s)
Antituberculosos/farmacología , Mycobacterium tuberculosis/efectos de los fármacos , Antituberculosos/administración & dosificación , Línea Celular , Células Cultivadas , Combinación de Medicamentos , Interacciones Farmacológicas , Retroalimentación , Proteínas Fluorescentes Verdes/genética , Ensayos Analíticos de Alto Rendimiento , Humanos , Isopropil Tiogalactósido/genética , Macrófagos/microbiología , Mycobacterium tuberculosis/genética , Tuberculosis/tratamiento farmacológico
8.
Nat Methods ; 12(5): 439-44, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25849636

RESUMEN

We report a high-throughput platform for delivering large cargo elements into 100,000 cells in 1 min. Our biophotonic laser-assisted surgery tool (BLAST) generates an array of microcavitation bubbles that explode in response to laser pulsing, forming pores in adjacent cell membranes through which cargo is gently driven by pressurized flow. The platform delivers large items including bacteria, enzymes, antibodies and nanoparticles into diverse cell types with high efficiency and cell viability. We used this platform to explore the intracellular lifestyle of Francisella novicida and discovered that the iglC gene is unexpectedly required for intracellular replication even after phagosome escape into the cell cytosol.


Asunto(s)
Francisella/fisiología , Rayos Láser , Microburbujas , Animales , Línea Celular , Regulación Bacteriana de la Expresión Génica/fisiología , Humanos
9.
PLoS Pathog ; 12(1): e1005356, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26808628

RESUMEN

Persistent viral infections are simultaneously associated with chronic inflammation and highly potent immunosuppressive programs mediated by IL-10 and PDL1 that attenuate antiviral T cell responses. Inhibiting these suppressive signals enhances T cell function to control persistent infection; yet, the underlying signals and mechanisms that program immunosuppressive cell fates and functions are not well understood. Herein, we use lymphocytic choriomeningitis virus infection (LCMV) to demonstrate that the induction and functional programming of immunosuppressive dendritic cells (DCs) during viral persistence are separable mechanisms programmed by factors primarily considered pro-inflammatory. IFNγ first induces the de novo development of naive monocytes into DCs with immunosuppressive potential. Type I interferon (IFN-I) then directly targets these newly generated DCs to program their potent T cell immunosuppressive functions while simultaneously inhibiting conventional DCs with T cell stimulating capacity. These mechanisms of monocyte conversion are constant throughout persistent infection, establishing a system to continuously interpret and shape the immunologic environment. MyD88 signaling was required for the differentiation of suppressive DCs, whereas inhibition of stimulatory DCs was dependent on MAVS signaling, demonstrating a bifurcation in the pathogen recognition pathways that promote distinct elements of IFN-I mediated immunosuppression. Further, a similar suppressive DC origin and differentiation was also observed in Mycobacterium tuberculosis infection, HIV infection and cancer. Ultimately, targeting the underlying mechanisms that induce immunosuppression could simultaneously prevent multiple suppressive signals to further restore T cell function and control persistent infections.


Asunto(s)
Células Dendríticas/inmunología , Tolerancia Inmunológica/inmunología , Interferones/inmunología , Virosis/inmunología , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , VIH , Infecciones por VIH/inmunología , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias/inmunología , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa , Linfocitos T/inmunología , Tuberculosis/inmunología
10.
Infect Immun ; 85(9)2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28630063

RESUMEN

A potent vaccine against tuberculosis, one of the world's deadliest diseases, is needed to enhance the immunity of people worldwide, most of whom have been vaccinated with the partially effective Mycobacterium bovis BCG vaccine. Here we investigate novel live attenuated recombinant Listeria monocytogenes (rLm) vaccines expressing the Mycobacterium tuberculosis 30-kDa major secretory protein (r30/antigen 85B [Ag85B]) (rLm30) as heterologous booster vaccines in animals primed with BCG. Using three attenuated L. monocytogenes vectors, L. monocytogenes ΔactA (LmI), L. monocytogenes ΔactA ΔinlB (LmII), and L. monocytogenes ΔactA ΔinlB prfA* (LmIII), we constructed five rLm30 vaccine candidates expressing r30 linked in frame to the L. monocytogenes listeriolysin O signal sequence and driven by the hly promoter (h30) or linked in frame to the ActA N-terminal 100 amino acids and driven by the actA promoter (a30). All five rLm30 vaccines secreted r30 in broth and macrophages; while rLm30 expressing r30 via a constitutively active prfA* regulon (rLmIII/a30) expressed the largest amount of r30 in broth culture, all five rLm30 vaccines expressed equivalent amounts of r30 in infected macrophages. In comparative studies, boosting of BCG-immunized mice with rLmIII/a30 induced the strongest antigen-specific T-cell responses, including splenic and lung polyfunctional CD4+ T cells expressing the three cytokines interferon gamma (IFN-γ), tumor necrosis factor alpha (TNF-α), and interleukin-2 (IL-2) (P < 0.001) and splenic and lung CD8+ T cells expressing IFN-γ (P < 0.0001). In mice and guinea pigs, the rLmIII/a30 and rLmI/h30 vaccines were generally more potent booster vaccines than r30 with an adjuvant and a recombinant adenovirus vaccine expressing r30. In a setting in which BCG alone was highly immunoprotective, boosting of mice with rLmIII/a30, the most potent of the vaccines, significantly enhanced protection against aerosolized M. tuberculosis (P < 0.01).

11.
J Am Chem Soc ; 139(19): 6663-6668, 2017 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-28437093

RESUMEN

We present a synthetic approach to a highly pathogen-selective detection and delivery platform based on the interaction of an antibody nanovalve with a tetrasaccharide from the O-antigen of the lipopolysaccharide (LPS) of Francisella tularensis bacteria, a Tier 1 Select Agent of bioterrorism. Different design considerations are explored, and proof-of-concept for highly pathogen-specific cargo release from mesoporous silica nanoparticles is demonstrated by comparisons of the release of a signal transducer and model drug by LPS from F. tularensis vs Pseudomonas aeruginosa and by F. tularensis live bacteria vs the closely related bacterium Francisella novocida. In addition to the specific response to a biowarfare agent, treatment of infectious diseases in general could benefit tremendously from a delivery platform that releases its antibiotic payload only at the site of infection and only in the presence of the target pathogen, thereby minimizing off-target toxicities.

12.
Small ; 12(27): 3690-702, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27246117

RESUMEN

Effective and rapid treatment of tularemia is needed to reduce morbidity and mortality of this potentially fatal infectious disease. The etiologic agent, Francisella tularensis, is a facultative intracellular bacterial pathogen which infects and multiplies to high numbers in macrophages. Nanotherapeutics are particularly promising for treatment of infectious diseases caused by intracellular pathogens, whose primary host cells are macrophages, because nanoparticles preferentially target and are avidly internalized by macrophages. A mesoporous silica nanoparticle (MSN) has been developed functionalized with disulfide snap-tops that has high drug loading and selectively releases drug intracellularly in response to the redox potential. These nanoparticles, when loaded with Hoechst fluorescent dye, release their cargo exclusively intracellularly and stain the nuclei of macrophages. The MSNs loaded with moxifloxacin kill F. tularensis in macrophages in a dose-dependent fashion. In a mouse model of lethal pneumonic tularemia, MSNs loaded with moxifloxacin prevent weight loss, illness, and death, markedly reduce the burden of F. tularensis in the lung, liver, and spleen, and are significantly more efficacious than an equivalent amount of free drug. An important proof-of-principle for the potential therapeutic use of a novel nanoparticle drug delivery platform for the treatment of infectious diseases is provided.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Fluoroquinolonas/química , Fluoroquinolonas/uso terapéutico , Nanopartículas/química , Dióxido de Silicio/química , Tularemia/tratamiento farmacológico , Animales , Femenino , Fluoroquinolonas/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Moxifloxacino
13.
J Biol Chem ; 289(26): 18279-89, 2014 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-24855650

RESUMEN

Mycobacterium tuberculosis has evolved various mechanisms by which the bacterium can maintain homeostasis under numerous environmental assaults generated by the host immune response. M. tuberculosis harbors enzymes involved in the oxidative stress response that aid in survival during the production of reactive oxygen species in activated macrophages. Previous studies have shown that a dye-decolorizing peroxidase (DyP) is encapsulated by a bacterial nanocompartment, encapsulin (Enc), whereby packaged DyP interacts with Enc via a unique C-terminal extension. M. tuberculosis also harbors an encapsulin homolog (CFP-29, Mt-Enc), within an operon with M. tuberculosis DyP (Mt-DyP), which contains a C-terminal extension. Together these observations suggest that Mt-DyP interacts with Mt-Enc. Furthermore, it has been suggested that DyPs may function as either a heme-dependent peroxidase or a deferrochelatase. Like Mt-DyP, M. tuberculosis iron storage ferritin protein, Mt-BfrB, and an M. tuberculosis protein involved in folate biosynthesis, 7,8-dihydroneopterin aldolase (Mt-FolB), have C-terminal tails that could also interact with Mt-Enc. For the first time, we show by co-purification and electron microscopy that mycobacteria via Mt-Enc can encapsulate Mt-DyP, Mt-BfrB, and Mt-FolB. Functional studies of free or encapsulated proteins demonstrate that they retain their enzymatic activity within the Mt-Enc nanocompartment. Mt-DyP, Mt-FolB, and Mt-BfrB all have antioxidant properties, suggesting that if these proteins are encapsulated by Mt-Enc, then this nanocage may play a role in the M. tuberculosis oxidative stress response. This report provides initial structural and biochemical clues regarding the molecular mechanisms that utilize compartmentalization by which the mycobacterial cell may aid in detoxification of the local environment to ensure long term survival.


Asunto(s)
Aldehído-Liasas/metabolismo , Proteínas Bacterianas/metabolismo , Mycobacterium tuberculosis/enzimología , Orgánulos/metabolismo , Peroxidasa/metabolismo , Aldehído-Liasas/genética , Proteínas Bacterianas/genética , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/metabolismo , Orgánulos/genética , Peroxidasa/genética , Unión Proteica
14.
Small ; 11(38): 5066-78, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26193431

RESUMEN

Tuberculosis is a major global health problem for which improved therapeutics are needed to shorten the course of treatment and combat emergence of drug resistance. Mycobacterium tuberculosis, the etiologic agent of tuberculosis, is an intracellular pathogen of mononuclear phagocytes. As such, it is an ideal pathogen for nanotherapeutics because macrophages avidly ingest nanoparticles even without specific targeting molecules. Hence, a nanoparticle drug delivery system has the potential to target and deliver high concentrations of drug directly into M. tuberculosis-infected cells-greatly enhancing efficacy while avoiding off-target toxicities. Stimulus-responsive mesoporous silica nanoparticles of two different sizes, 100 and 50 nm, are developed as carriers for the major anti-tuberculosis drug isoniazid in a prodrug configuration. The drug is captured by the aldehyde-functionalized nanoparticle via hydrazone bond formation and coated with poly(ethylene imine)-poly(ethylene glycol) (PEI-PEG). The drug is released from the nanoparticles in response to acidic pH at levels that naturally occur within acidified endolysosomes. It is demonstrated that isoniazid-loaded PEI-PEG-coated nanoparticles are avidly ingested by M. tuberculosis-infected human macrophages and kill the intracellular bacteria in a dose-dependent manner. It is further demonstrated in a mouse model of pulmonary tuberculosis that the nanoparticles are well tolerated and much more efficacious than an equivalent amount of free drug.


Asunto(s)
Isoniazida/uso terapéutico , Nanopartículas/química , Tuberculosis/tratamiento farmacológico , Aldehídos/química , Animales , Células CHO , Cricetinae , Cricetulus , Modelos Animales de Enfermedad , Femenino , Humanos , Concentración de Iones de Hidrógeno , Pulmón/efectos de los fármacos , Pulmón/microbiología , Pulmón/patología , Macrófagos/efectos de los fármacos , Macrófagos/microbiología , Ratones Endogámicos BALB C , Viabilidad Microbiana/efectos de los fármacos , Mycobacterium tuberculosis/efectos de los fármacos , Nanopartículas/ultraestructura , Polietilenglicoles/química , Polietileneimina/química , Porosidad , Profármacos/uso terapéutico , Dióxido de Silicio/química , Espectrofotometría Ultravioleta , Tuberculosis/microbiología
15.
Small ; 11(38): 5065, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26450161

RESUMEN

On page 5066, J. I. Zink, M. A. Horwitz, and co-workers use confocal microscopy to demonstrate the avid uptake of RITC-labeled mesoporous silica nanoparticles loaded with the anti-tuberculosis drug isoniazid (shown here in red) by human macrophages (nuclei stained blue with DAPI) infected with GFP-expressing Mycobacterium tuberculosis (shown here in green).


Asunto(s)
Isoniazida/uso terapéutico , Nanopartículas/química , Tuberculosis/tratamiento farmacológico , Animales , Concentración de Iones de Hidrógeno , Ratones
16.
Infect Immun ; 82(9): 3900-9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25001602

RESUMEN

Leprosy remains a major global health problem and typically occurs in regions in which tuberculosis is endemic. Vaccines are needed that protect against both infections and do so better than the suboptimal Mycobacterium bovis BCG vaccine. Here, we evaluated rBCG30, a vaccine previously demonstrated to induce protection superior to that of BCG against Mycobacterium tuberculosis and Mycobacterium bovis challenge in animal models, for efficacy against Mycobacterium leprae challenge in a murine model of leprosy. rBCG30 overexpresses the M. tuberculosis 30-kDa major secretory protein antigen 85B, which is 85% homologous with the M. leprae homolog (r30ML). Mice were sham immunized or immunized intradermally with BCG or rBCG30 and challenged 2.5 months later by injection of viable M. leprae into each hind footpad. After 7 months, vaccine efficacy was assessed by enumerating the M. leprae bacteria per footpad. Both BCG and rBCG30 induced significant protection against M. leprae challenge. In the one experiment in which a comparison between BCG and rBCG30 was feasible, rBCG30 induced significantly greater protection than did BCG. Immunization of mice with purified M. tuberculosis or M. leprae antigen 85B also induced protection against M. leprae challenge but less so than BCG or rBCG30. Notably, boosting rBCG30 with M. tuberculosis antigen 85B significantly enhanced r30ML-specific immune responses, substantially more so than boosting BCG, and significantly augmented protection against M. leprae challenge. Thus, rBCG30, a vaccine that induces improved protection against M. tuberculosis, induces cross-protection against M. leprae that is comparable or potentially superior to that induced by BCG, and boosting rBCG30 with antigen 85B further enhances immune responses and protective efficacy.


Asunto(s)
Aciltransferasas/inmunología , Antígenos Bacterianos/inmunología , Vacuna BCG/inmunología , Proteínas Bacterianas/inmunología , Protección Cruzada/inmunología , Mycobacterium leprae/inmunología , Mycobacterium tuberculosis/inmunología , Vacunas contra la Tuberculosis/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Femenino , Inmunización/métodos , Lepra/inmunología , Lepra/prevención & control , Ratones , Ratones Endogámicos BALB C , Mycobacterium bovis/inmunología , Tuberculosis/inmunología , Tuberculosis/prevención & control , Vacunación/métodos
17.
Proc Natl Acad Sci U S A ; 108(12): 5051-6, 2011 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-21383189

RESUMEN

Mycobacterium tuberculosis must import iron from its host for survival, and its siderophore-dependent iron acquisition pathways are well established. Here we demonstrate a newly characterized pathway, whereby M. tuberculosis can use free heme and heme from hemoglobin as an iron source. Significantly, we identified the genomic region, Rv0202c-Rv0207c, responsible for the passage of heme iron across the mycobacterial membrane. Key players of this heme uptake system were characterized including a secreted protein and two transmembrane proteins, all three specific to mycobacteria. Furthermore, the crystal structure of the key heme carrier protein Rv0203 was found to have a unique fold. The discovery of a unique mycobacterial heme acquisition pathway opens new avenues of exploration into mycobacterial therapeutics.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas Portadoras/metabolismo , Hemo/metabolismo , Hierro/metabolismo , Mycobacterium tuberculosis/metabolismo , Proteínas Bacterianas/genética , Transporte Biológico/fisiología , Proteínas Portadoras/genética , Membrana Celular/genética , Membrana Celular/metabolismo , Hemo/genética , Mycobacterium tuberculosis/genética , Tuberculosis/tratamiento farmacológico , Tuberculosis/genética , Tuberculosis/metabolismo
18.
mBio ; 15(4): e0018624, 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38511933

RESUMEN

Melioidosis, caused by the intracellular bacterial pathogen and Tier 1 select agent Burkholderia pseudomallei (Bp), is a highly fatal disease endemic in tropical areas. No licensed vaccine against melioidosis exists. In preclinical vaccine studies, demonstrating protection against respiratory infection in the highly sensitive BALB/c mouse has been especially challenging. To address this challenge, we have used a safe yet potent live attenuated platform vector, LVS ΔcapB, previously used successfully to develop vaccines against the Tier 1 select agents of tularemia, anthrax, and plague, to develop a melioidosis vaccine. We have engineered melioidosis vaccines (rLVS ΔcapB/Bp) expressing multiple immunoprotective Bp antigens among type VI secretion system proteins Hcp1, Hcp2, and Hcp6, and membrane protein LolC. Administered intradermally, rLVS ΔcapB/Bp vaccines strongly protect highly sensitive BALB/c mice against lethal respiratory Bp challenge, but protection is overwhelmed at very high challenge doses. In contrast, administered intranasally, rLVS ΔcapB/Bp vaccines remain strongly protective against even very high challenge doses. Under some conditions, the LVS ΔcapB vector itself provides significant protection against Bp challenge, and consistent with this, both the vector and vaccines induce humoral immune responses to Bp antigens. Three-antigen vaccines expressing Hcp6-Hcp1-Hcp2 or Hcp6-Hcp1-LolC are among the most potent and provide long-term protection and protection even with a single intranasal immunization. Protection via the intranasal route was either comparable to or statistically significantly better than the single-deletional Bp mutant Bp82, which served as a positive control. Thus, rLVS ΔcapB/Bp vaccines are exceptionally promising safe and potent melioidosis vaccines. IMPORTANCE: Melioidosis, a major neglected disease caused by the intracellular bacterial pathogen Burkholderia pseudomallei, is endemic in many tropical areas of the world and causes an estimated 165,000 cases and 89,000 deaths in humans annually. Moreover, B. pseudomallei is categorized as a Tier 1 select agent of bioterrorism, largely because inhalation of low doses can cause rapidly fatal pneumonia. No licensed vaccine is available to prevent melioidosis. Here, we describe a safe and potent melioidosis vaccine that protects against lethal respiratory challenge with B. pseudomallei in a highly sensitive small animal model-even a single immunization is highly protective, and the vaccine gives long-term protection. The vaccine utilizes a highly attenuated replicating intracellular bacterium as a vector to express multiple key proteins of B. pseudomallei; this vector platform has previously been used successfully to develop potent vaccines against other Tier 1 select agent diseases including tularemia, anthrax, and plague.


Asunto(s)
Carbunco , Burkholderia pseudomallei , Melioidosis , Peste , Tularemia , Humanos , Animales , Ratones , Burkholderia pseudomallei/genética , Melioidosis/prevención & control , Ratones Endogámicos BALB C , Vacunas Bacterianas , Vacunas Atenuadas , Antígenos Bacterianos/genética
19.
Infect Immun ; 81(5): 1550-61, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23439306

RESUMEN

Francisella tularensis, the causative agent of tularemia, is a category A bioterrorism agent. A vaccine that is safer and more effective than the currently available unlicensed F. tularensis live vaccine strain (LVS) is needed to protect against intentional release of aerosolized F. tularensis, the most dangerous type of exposure. In this study, we employed a heterologous prime-boost vaccination strategy comprising intradermally administered LVS ΔcapB (highly attenuated capB-deficient LVS mutant) as the primer vaccine and rLm/iglC (recombinant attenuated Listeria monocytogenes expressing the F. tularensis immunoprotective antigen IglC) as the booster vaccine. Boosting LVS ΔcapB-primed mice with rLm/iglC significantly enhanced T cell immunity; their splenic T cells secreted significantly more gamma interferon (IFN-γ) and had significantly more cytokine (IFN-γ and/or tumor necrosis factor [TNF] and/or interleukin-2 [IL-2])-producing CD4(+) and CD8(+) T cells upon in vitro IglC stimulation. Importantly, mice primed with LVS ΔcapB or rLVS ΔcapB/IglC, boosted with rLm/iglC, and subsequently challenged with 10 50% lethal doses (LD50) of aerosolized highly virulent F. tularensis Schu S4 had a significantly higher survival rate and mean survival time than mice immunized with only LVS ΔcapB (P < 0.0001); moreover, compared with mice immunized once with LVS, primed-boosted mice had a higher survival rate (75% versus 62.5%) and mean survival time during the first 21 days postchallenge (19 and 20 days for mice boosted after being primed with LVS ΔcapB and rLVS ΔcapB/IglC, respectively, versus 17 days for mice immunized with LVS) and maintained their weight significantly better (P < 0.01). Thus, the LVS ΔcapB-rLm/iglC prime-boost vaccination strategy holds substantial promise for a vaccine that is safer and at least as potent as LVS.


Asunto(s)
Vacunas Bacterianas , Francisella tularensis/inmunología , Inmunización Secundaria/métodos , Listeria monocytogenes/inmunología , Tularemia/prevención & control , Vacunación/métodos , Aerosoles , Análisis de Varianza , Animales , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/inmunología , Citocinas/metabolismo , Femenino , Inmunidad Celular , Inmunoglobulina G/sangre , Ratones , Ratones Endogámicos BALB C , Bazo/citología , Tasa de Supervivencia , Linfocitos T/inmunología , Linfocitos T/metabolismo , Tularemia/inmunología , Tularemia/metabolismo , Vacunas Atenuadas , Vacunas Sintéticas
20.
J Neurosci Res ; 91(10): 1292-302, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23907992

RESUMEN

We previously showed that, in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson's disease (PD), vaccination with bacillus Calmette-Guerin (BCG) prior to MPTP exposure limited the loss of striatal dopamine (DA) and dopamine transporter (DAT) and prevented the activation of nigral microglia. Here, we conducted BCG dose studies and investigated the mechanisms underlying BCG vaccination's neuroprotective effects in this model. We found that a dose of 1 × 10(6) cfu BCG led to higher levels of striatal DA and DAT ligand binding (28% and 42%, respectively) in BCG-vaccinated vs. unvaccinated MPTP-treated mice, but without a significant increase in substantia nigra tyrosine hydroxylase-staining neurons. Previous studies showed that BCG can induce regulatory T cells (Tregs) and that Tregs are neuroprotective in models of neurodegenerative diseases. However, MPTP is lymphotoxic, so it was unclear whether Tregs were maintained after MPTP treatment and whether a relationship existed between Tregs and the preservation of striatal DA system integrity. We found that, 21 days post-MPTP treatment, Treg levels in mice that had received BCG prior to MPTP were threefold greater than those in MPTP-only-treated mice and elevated above those in saline-only-treated mice, suggesting that the persistent BCG infection continually promoted Treg responses. Notably, the magnitude of the Treg response correlated positively with both striatal DA levels and DAT ligand binding. Therefore, BCG vaccine-mediated neuroprotection is associated with Treg levels in this mouse model. Our results suggest that BCG-induced Tregs could provide a new adjunctive therapeutic approach to ameliorating pathology associated with PD and other neurodegenerative diseases.


Asunto(s)
Vacuna BCG/inmunología , Fármacos Neuroprotectores/inmunología , Trastornos Parkinsonianos/inmunología , Trastornos Parkinsonianos/prevención & control , Linfocitos T Reguladores/inmunología , Animales , Vacuna BCG/administración & dosificación , Encéfalo/inmunología , Encéfalo/patología , Cromatografía Líquida de Alta Presión , Modelos Animales de Enfermedad , Citometría de Flujo , Masculino , Ratones , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/administración & dosificación
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda