Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Eur J Immunol ; 54(5): e2350515, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38361219

RESUMEN

Caspase-1 location in cells has been studied with fluorochrome-labeled inhibitors of caspase-1 (FLICA reagents). We report that FLICA reagents have limited cell-membrane permeability. This impacts experimental design as cells with intact membranes, including caspase-1 knockout cells, are not appropriate controls for cells with inflammasome-induced gasdermin D membrane pores.


Asunto(s)
Caspasa 1 , Inhibidores de Caspasas , Permeabilidad de la Membrana Celular , Colorantes Fluorescentes , Inflamasomas , Macrófagos , Caspasa 1/metabolismo , Animales , Macrófagos/inmunología , Macrófagos/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Ratones , Inflamasomas/metabolismo , Inhibidores de Caspasas/farmacología , Ratones Noqueados , Proteínas de Unión a Fosfato/metabolismo , Humanos
2.
Methods Mol Biol ; 2696: 199-210, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37578724

RESUMEN

The Nod-like Receptor (NLR) apoptosis inhibitory proteins (NAIPs) are cytosolic receptors that sense cytosolic bacterial proteins. NAIP ligation induces its association with NLRC4, leading to the assembly of the NAIP/NLRC4 inflammasome, which induces the activation of the caspase-1 protease. Caspase-1 then cleaves pro-interleukin (IL)-1ß, pro-IL-18, and gasdermin D and induces a form of pro-inflammatory cell death, pyroptosis. These processes culminate in host defense against bacterial infection. Here we describe methods for activating NAIP/NLRC4 inflammasome signalling in human and murine macrophages and quantifying inflammasome-induced cell death.


Asunto(s)
Proteínas de Unión al Calcio , Inflamasomas , Animales , Ratones , Humanos , Inflamasomas/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Muerte Celular , Caspasas/metabolismo , Caspasa 1/metabolismo , Proteínas Adaptadoras de Señalización CARD/metabolismo
3.
Methods Mol Biol ; 2459: 29-37, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35212951

RESUMEN

Neutrophils are innate immune cells that play critical functions during infections through diverse mechanisms. One such mechanism, the generation of extracellular traps (NETs), enables direct bacterial killing during infections. We recently reported that the activation of the non-canonical inflammasomes in neutrophils allows for the generation of NETs and is an important host defence mechanism in vivo in response to intracellular Gram-negative bacterium. This process is dependent on inflammatory caspases and the cell death effector Gasdermin D. Here, we describe a simple approach to study the functions of the non-canonical inflammasome in murine neutrophils using microscopy and cellular fragmentation assays.


Asunto(s)
Trampas Extracelulares , Inflamasomas , Animales , Caspasas/metabolismo , Trampas Extracelulares/metabolismo , Inflamasomas/metabolismo , Ratones , Neutrófilos/metabolismo , Piroptosis
4.
Immunol Cell Biol ; 99(10): 1040-1052, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34462965

RESUMEN

Group A Streptococcus (GAS) is a Gram-positive bacterial pathogen that causes an array of infectious diseases in humans. Accumulating clinical evidence suggests that proinflammatory interleukin (IL)-1ß signaling plays an important role in GAS disease progression. The host regulates the production and secretion of IL-1ß via the cytosolic inflammasome pathway. Activation of the NLR family pyrin domain-containing 3 (NLRP3) inflammasome complex requires two signals: a priming signal that stimulates increased transcription of genes encoding the components of the inflammasome pathway, and an activating signal that induces assembly of the inflammasome complex. Here we show that GAS-derived lipoteichoic acid can provide a priming signal for NLRP3 inflammasome activation. As only few GAS-derived proteins have been associated with inflammasome-dependent IL-1ß signaling, we investigated novel candidates that might play a role in activating the inflammasome pathway by infecting mouse bone marrow-derived macrophages and human THP-1 macrophage-like cells with a panel of isogenic GAS mutant strains. We found that the cytolysins streptolysin O (SLO) and streptolysin S are the main drivers of IL-1ß release in proliferating logarithmic phase GAS. Using a mutant form of recombinant SLO, we confirmed that bacterial pore formation on host cell membranes is a key mechanism required for inflammasome activation. Our results suggest that streptolysins are major determinants of GAS-induced inflammation and present an attractive target for therapeutic intervention.


Asunto(s)
Inflamasomas , Infecciones Estreptocócicas , Animales , Interleucina-1beta , Macrófagos , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR , Streptococcus pyogenes , Estreptolisinas
5.
J Exp Med ; 218(5)2021 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-33656514

RESUMEN

Vincristine is an important component of many regimens used for pediatric and adult malignancies, but it causes a dose-limiting sensorimotor neuropathy for which there is no effective treatment. This study aimed to delineate the neuro-inflammatory mechanisms contributing to the development of mechanical allodynia and gait disturbances in a murine model of vincristine-induced neuropathy, as well as to identify novel treatment approaches. Here, we show that vincristine-induced peripheral neuropathy is driven by activation of the NLRP3 inflammasome and subsequent release of interleukin-1ß from macrophages, with mechanical allodynia and gait disturbances significantly reduced in knockout mice lacking NLRP3 signaling pathway components, or after treatment with the NLRP3 inhibitor MCC950. Moreover, treatment with the IL-1 receptor antagonist anakinra prevented the development of vincristine-induced neuropathy without adversely affecting chemotherapy efficacy or tumor progression in patient-derived medulloblastoma xenograph models. These results detail the neuro-inflammatory mechanisms leading to vincristine-induced peripheral neuropathy and suggest that repurposing anakinra may be an effective co-treatment strategy to prevent vincristine-induced peripheral neuropathy.


Asunto(s)
Hiperalgesia/genética , Interleucina-1beta/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Enfermedades del Sistema Nervioso Periférico/genética , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Animales , Antineoplásicos/administración & dosificación , Antirreumáticos/administración & dosificación , Cisplatino/administración & dosificación , Furanos/administración & dosificación , Humanos , Hiperalgesia/inducido químicamente , Hiperalgesia/tratamiento farmacológico , Indenos/administración & dosificación , Inflamasomas/efectos de los fármacos , Inflamasomas/genética , Inflamasomas/metabolismo , Proteína Antagonista del Receptor de Interleucina 1/administración & dosificación , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Oxaliplatino/administración & dosificación , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Enfermedades del Sistema Nervioso Periférico/tratamiento farmacológico , Sulfonamidas/administración & dosificación , Vincristina
6.
Mol Microbiol ; 112(4): 1284-1307, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31389636

RESUMEN

Bacterial nutrition is an essential aspect of host-pathogen interaction. For the intracellular pathogen Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis in humans, fatty acids derived from lipid droplets are considered the major carbon source. However, many other soluble nutrients are available inside host cells and may be used as alternative carbon sources. Lactate and pyruvate are abundant in human cells and fluids, particularly during inflammation. In this work, we study Mtb metabolism of lactate and pyruvate combining classic microbial physiology with a 'multi-omics' approach consisting of transposon-directed insertion site sequencing (TraDIS), RNA-seq transcriptomics, proteomics and stable isotopic labelling coupled with mass spectrometry-based metabolomics. We discovered that Mtb is well adapted to use both lactate and pyruvate and that their metabolism requires gluconeogenesis, valine metabolism, the Krebs cycle, the GABA shunt, the glyoxylate shunt and the methylcitrate cycle. The last two pathways are traditionally associated with fatty acid metabolism and, unexpectedly, we found that in Mtb the methylcitrate cycle operates in reverse, to allow optimal metabolism of lactate and pyruvate. Our findings reveal a novel function for the methylcitrate cycle as a direct route for the biosynthesis of propionyl-CoA, the essential precursor for the biosynthesis of the odd-chain fatty acids.


Asunto(s)
Ácido Láctico/metabolismo , Mycobacterium tuberculosis/metabolismo , Ácido Pirúvico/metabolismo , Acilcoenzima A/metabolismo , Proteínas Bacterianas/metabolismo , Citrato (si)-Sintasa/metabolismo , Citratos/metabolismo , Ácidos Grasos/metabolismo , Regulación Bacteriana de la Expresión Génica/genética , Glioxilatos , Tuberculosis/microbiología
7.
J Leukoc Biol ; 105(2): 401-410, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30368901

RESUMEN

Inflammasomes are signaling hubs that activate inflammatory caspases to drive cytokine maturation and cell lysis. Inflammasome activation by Salmonella Typhimurium infection or Salmonella-derived molecules is extensively studied in murine myeloid cells. Salmonella-induced inflammasome signaling in human innate immune cells, is however, poorly characterized. Here, we show that Salmonella mutation to inactivate the Salmonella pathogenicity island-2 type III secretion system (SPI2 T3SS) potentiates S. Typhimurium-induced inflammasome responses from primary human macrophages, resulting in strong IL-1ß production and macrophage death. Inactivation of the SPI1 T3SS diminished human macrophage responses to WT and ΔSPI2 Salmonella. Salmonella ΔSPI2 elicited a mixed inflammasome response from human myeloid cells, in which NLR family CARD-domain containing protein 4 (NLRC4) and NLR family PYRIN-domain containing protein 3 (NLRP3) perform somewhat redundant functions in generating IL-1ß and inducing pyroptosis. Our data suggest that Salmonella employs the SPI2 T3SS to subvert SPI1-induced NLRP3 and NLRC4 inflammasome responses in human primary macrophages, in a species-specific immune evasion mechanism.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/metabolismo , Proteínas de Unión al Calcio/metabolismo , Islas Genómicas , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Salmonella typhimurium/genética , Animales , Muerte Celular , Humanos , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones Endogámicos C57BL , Células Mieloides/metabolismo , Piroptosis
8.
Cell Rep ; 24(6): 1425-1433, 2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-30089254

RESUMEN

IL-1ß requires processing by caspase-1 to generate the active, pro-inflammatory cytokine. Acute IL-1ß secretion from inflammasome-activated macrophages requires caspase-1-dependent GSDMD cleavage, which also induces pyroptosis. Mechanisms of IL-1ß secretion by pyroptotic and non-pyroptotic cells, and the precise functions of caspase-1 and GSDMD therein, are unresolved. Here, we show that, while efficient early secretion of endogenous IL-1ß from primary non-pyroptotic myeloid cells in vitro requires GSDMD, later IL-1ß release in vitro and in vivo proceeds independently of GSDMD. IL-1ß maturation is sufficient for slow, caspase-1/GSDMD-independent secretion of ectopic IL-1ß from resting, non-pyroptotic macrophages, but the speed of IL-1ß release is boosted by inflammasome activation, via caspase-1 and GSDMD. IL-1ß cleavage induces IL-1ß enrichment at PIP2-enriched plasma membrane ruffles, and this is a prerequisite for IL-1ß secretion and is mediated by a polybasic motif within the cytokine. We thus reveal a mechanism in which maturation-induced IL-1ß trafficking facilitates its unconventional secretion.


Asunto(s)
Membrana Celular/metabolismo , Interleucina-1beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Humanos , Transfección
9.
Sci Immunol ; 3(26)2018 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-30143554

RESUMEN

Neutrophil extrusion of neutrophil extracellular traps (NETs) and concomitant cell death (NETosis) provides host defense against extracellular pathogens, whereas macrophage death by pyroptosis enables defense against intracellular pathogens. We report the unexpected discovery that gasdermin D (GSDMD) connects these cell death modalities. We show that neutrophil exposure to cytosolic lipopolysaccharide or cytosolic Gram-negative bacteria (Salmonella ΔsifA and Citrobacter rodentium) activates noncanonical (caspase-4/11) inflammasome signaling and triggers GSDMD-dependent neutrophil death. GSDMD-dependent death induces neutrophils to extrude antimicrobial NETs. Caspase-11 and GSDMD are required for neutrophil plasma membrane rupture during the final stage of NET extrusion. Unexpectedly, caspase-11 and GSDMD are also required for early features of NETosis, including nuclear delobulation and DNA expansion; this is mediated by the coordinate actions of caspase-11 and GSDMD in mediating nuclear membrane permeabilization and histone degradation. In vivo application of deoxyribonuclease I to dissolve NETs during murine Salmonella ΔsifA challenge increases bacterial burden in wild-type but not in Casp11-/- and Gsdmd -/- mice. Our studies reveal that neutrophils use an inflammasome- and GSDMD-dependent mechanism to activate NETosis as a defense response against cytosolic bacteria.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/inmunología , Caspasas/inmunología , Trampas Extracelulares/inmunología , Inflamasomas/inmunología , Neutrófilos/inmunología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Caspasas Iniciadoras , Muerte Celular , Citrobacter rodentium , Citosol/inmunología , Citosol/microbiología , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular , Lipopolisacáridos , Macrófagos/inmunología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Unión a Fosfato , Infecciones por Salmonella/inmunología , Salmonella enterica
10.
J Exp Med ; 215(3): 827-840, 2018 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-29432122

RESUMEN

Host-protective caspase-1 activity must be tightly regulated to prevent pathology, but mechanisms controlling the duration of cellular caspase-1 activity are unknown. Caspase-1 is activated on inflammasomes, signaling platforms that facilitate caspase-1 dimerization and autoprocessing. Previous studies with recombinant protein identified a caspase-1 tetramer composed of two p20 and two p10 subunits (p20/p10) as an active species. In this study, we report that in the cell, the dominant species of active caspase-1 dimers elicited by inflammasomes are in fact full-length p46 and a transient species, p33/p10. Further p33/p10 autoprocessing occurs with kinetics specified by inflammasome size and cell type, and this releases p20/p10 from the inflammasome, whereupon the tetramer becomes unstable in cells and protease activity is terminated. The inflammasome-caspase-1 complex thus functions as a holoenzyme that directs the location of caspase-1 activity but also incorporates an intrinsic self-limiting mechanism that ensures timely caspase-1 deactivation. This intrinsic mechanism of inflammasome signal shutdown offers a molecular basis for the transient nature, and coordinated timing, of inflammasome-dependent inflammatory responses.


Asunto(s)
Caspasa 1/metabolismo , Inflamasomas/metabolismo , Animales , Cinética , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Modelos Biológicos , Nigericina/farmacología , Multimerización de Proteína
11.
FASEB J ; 30(5): 1901-12, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26839376

RESUMEN

We aimed to characterize antimicrobial zinc trafficking within macrophages and to determine whether the professional intramacrophage pathogen Salmonella enterica serovar Typhimurium (S Typhimurium) subverts this pathway. Using both Escherichia coli and S Typhimurium, we show that TLR signaling promotes the accumulation of vesicular zinc within primary human macrophages. Vesicular zinc is delivered to E. coli to promote microbial clearance, whereas S. Typhimurium evades this response via Salmonella pathogenicity island (SPI)-1. Even in the absence of SPI-1 and the zinc exporter ZntA, S Typhimurium resists the innate immune zinc stress response, implying the existence of additional host subversion mechanisms. We also demonstrate the combinatorial antimicrobial effects of zinc and copper, a pathway that S. Typhimurium again evades. Our use of complementary tools and approaches, including confocal microscopy, direct assessment of intramacrophage bacterial zinc stress responses, specific E. coli and S Typhimurium mutants, and inductively coupled plasma mass spectroscopy, has enabled carefully controlled characterization of this novel innate immune antimicrobial pathway. In summary, our study provides new insights at the cellular level into the well-documented effects of zinc in promoting host defense against infectious disease, as well as the complex host subversion strategies employed by S Typhimurium to combat this pathway.-Kapetanovic, R., Bokil, N. J., Achard, M. E. S., Ong, C.-L. Y., Peters, K. M., Stocks, C. J., Phan, M.-D., Monteleone, M., Schroder, K., Irvine, K. M., Saunders, B. M., Walker, M. J., Stacey, K. J., McEwan, A. G., Schembri, M. A., Sweet, M. J. Salmonella employs multiple mechanisms to subvert the TLR-inducible zinc-mediated antimicrobial response of human macrophages.


Asunto(s)
Macrófagos/inmunología , Macrófagos/metabolismo , Salmonella typhimurium/fisiología , Salmonella/fisiología , Receptores Toll-Like/metabolismo , Zinc/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Línea Celular , Células Cultivadas , Cobre , Vesículas Citoplasmáticas/química , Vesículas Citoplasmáticas/metabolismo , Regulación Bacteriana de la Expresión Génica , ARN Bacteriano/genética , ARN Bacteriano/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Toll-Like/genética
12.
Eur J Immunol ; 45(10): 2918-26, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26173988

RESUMEN

Humans encode two inflammatory caspases that detect cytoplasmic LPS, caspase-4 and caspase-5. When activated, these trigger pyroptotic cell death and caspase-1-dependent IL-1ß production; however the mechanism underlying this process is not yet confirmed. We now show that a specific NLRP3 inhibitor, MCC950, prevents caspase-4/5-dependent IL-1ß production elicited by transfected LPS. Given that both caspase-4 and caspase-5 can detect cytoplasmic LPS, it is possible that these proteins exhibit some degree of redundancy. Therefore, we generated human monocytic cell lines in which caspase-4 and caspase-5 were genetically deleted either individually or together. We found that the deletion of caspase-4 suppressed cell death and IL-1ß production following transfection of LPS into the cytoplasm, or in response to infection with Salmonella typhimurium. Although deletion of caspase-5 did not confer protection against transfected LPS, cell death and IL-1ß production were reduced after infection with Salmonella. Furthermore, double deletion of caspase-4 and caspase-5 had a synergistic effect in the context of Salmonella infection. Our results identify the NLRP3 inflammasome as the specific platform for IL-1ß maturation, downstream of cytoplasmic LPS detection by caspase-4/5. We also show that both caspase-4 and caspase-5 are functionally important for appropriate responses to intracellular Gram-negative bacteria.


Asunto(s)
Proteínas Portadoras/inmunología , Caspasas Iniciadoras/inmunología , Caspasas/inmunología , Lipopolisacáridos/inmunología , Monocitos/inmunología , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Línea Celular Tumoral , Humanos , Interleucina-1beta/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR
13.
Cytokine ; 74(2): 213-8, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25922276

RESUMEN

One of the most poorly understood processes in cell biology is the peculiar ability of specific leaderless proteins to be secreted via ER/Golgi-independent mechanisms ('unconventional protein secretion'). One such leaderless protein is the major immune-activating cytokine, interleukin-1ß (IL-1ß). Unusual amongst cytokines, IL-1ß is expressed in the cytosol as an inactive precursor protein. It requires maturation by the caspase-1 protease, which itself requires activation upon immune cell sensing of infection or cell stress. Despite 25 years of intensive research into IL-1ß secretory mechanisms, how it exits the cell is still not well understood. Here we will review the various mechanisms by which macrophages have been proposed to secrete IL-1 family cytokines, and the potential involvement of caspase-1 therein. Since aberrant IL-1ß production drives inherited and acquired human diseases (e.g. autoinflammatory diseases, arthritic diseases, gout, Alzheimer's disease), elucidation of the IL-1ß secretory pathway may offer new therapeutic opportunities for treatment across this wide range of human conditions.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Artritis/inmunología , Caspasa 1/inmunología , Gota/inmunología , Interleucina-1beta/inmunología , Macrófagos/inmunología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Artritis/metabolismo , Artritis/patología , Caspasa 1/metabolismo , Gota/metabolismo , Gota/patología , Humanos , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Macrófagos/patología
14.
J Innate Immun ; 6(2): 240-50, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-23969887

RESUMEN

The globally significant human pathogen group A Streptococcus (GAS) sequesters the host protease plasmin to the cell surface during invasive disease initiation. Recent evidence has shown that localized plasmin activity prevents opsonization of several bacterial species by key components of the innate immune system in vitro. Here we demonstrate that plasmin at the GAS cell surface resulted in degradation of complement factor C3b, and that plasminogen acquisition is associated with a decrease in C3b opsonization and neutrophil-mediated killing in vitro. Furthermore, the ability to acquire cell surface plasmin(ogen) correlates directly with a decrease in C3b opsonization, neutrophil phagocytosis, and increased bacterial survival in a humanized plasminogen mouse model of infection. These findings demonstrate that localized plasmin(ogen) plays an important role in facilitating GAS escape from the host innate immune response and increases bacterial virulence in the early stages of infection.


Asunto(s)
Complemento C3b/inmunología , Neutrófilos/inmunología , Fagocitosis/inmunología , Plasminógeno/inmunología , Streptococcus pyogenes/inmunología , Animales , Western Blotting , Complemento C3b/metabolismo , Femenino , Fibrinolisina/inmunología , Fibrinolisina/metabolismo , Citometría de Flujo , Interacciones Huésped-Patógeno/inmunología , Humanos , Evasión Inmune/inmunología , Masculino , Ratones Transgénicos , Neutrófilos/metabolismo , Neutrófilos/microbiología , Plasminógeno/genética , Plasminógeno/metabolismo , Infecciones Estreptocócicas/genética , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/metabolismo , Streptococcus pyogenes/fisiología , Estreptoquinasa/inmunología , Estreptoquinasa/metabolismo
15.
Proc Natl Acad Sci U S A ; 110(47): 19095-100, 2013 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-24191058

RESUMEN

Antibacterial drug development suffers from a paucity of targets whose inhibition kills replicating and nonreplicating bacteria. The latter include phenotypically dormant cells, known as persisters, which are tolerant to many antibiotics and often contribute to failure in the treatment of chronic infections. This is nowhere more apparent than in tuberculosis caused by Mycobacterium tuberculosis, a pathogen that tolerates many antibiotics once it ceases to replicate. We developed a strategy to identify proteins that Mycobacterium tuberculosis requires to both grow and persist and whose inhibition has the potential to prevent drug tolerance and persister formation. This strategy is based on a tunable dual-control genetic switch that provides a regulatory range spanning three orders of magnitude, quickly depletes proteins in both replicating and nonreplicating mycobacteria, and exhibits increased robustness to phenotypic reversion. Using this switch, we demonstrated that depletion of the nicotinamide adenine dinucleotide synthetase (NadE) rapidly killed Mycobacterium tuberculosis under conditions of standard growth and nonreplicative persistence induced by oxygen and nutrient limitation as well as during the acute and chronic phases of infection in mice. These findings establish the dual-control switch as a robust tool with which to probe the essentiality of Mycobacterium tuberculosis proteins under different conditions, including those that induce antibiotic tolerance, and NadE as a target with the potential to shorten current tuberculosis chemotherapies.


Asunto(s)
Amida Sintasas/antagonistas & inhibidores , Descubrimiento de Drogas/métodos , Tolerancia a Medicamentos/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Mycobacterium tuberculosis/efectos de los fármacos , Tuberculosis/prevención & control , Animales , Proteínas Portadoras , Proteínas de Escherichia coli , Ingeniería Genética/métodos , Luciferasas , Ratones , Mycobacterium tuberculosis/crecimiento & desarrollo
16.
Nat Med ; 13(12): 1515-20, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18059281

RESUMEN

The success of Mycobacterium tuberculosis (Mtb) as a human pathogen relies on its ability to resist eradication by the immune system. The identification of mechanisms that enable Mtb to persist is key for finding ways to limit latent tuberculosis, which affects one-third of the world's population. Here we show that conditional gene silencing can be used to determine whether an Mtb gene required for optimal growth in vitro is also important for virulence and, if so, during which phase of an infection it is required. Application of this approach to the prcBA genes, which encode the core of the mycobacterial proteasome, revealed an unpredicted requirement of the core proteasome for the persistence of Mtb during the chronic phase of infection in mice. Proteasome depletion also attenuated Mtb in interferon-gamma-deficient mice, pointing to a function of the proteasome beyond defense against the adaptive immune response. Genes that are essential for growth in vitro, in vivo or both account for approximately 20% of Mtb's genome. Conditional gene silencing could therefore facilitate the validation of up to 800 potential Mtb drug targets and improve our understanding of host-pathogen dynamics.


Asunto(s)
Silenciador del Gen , Mycobacterium tuberculosis/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Animales , Proteínas Bacterianas/química , Modelos Animales de Enfermedad , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Interferón gamma/metabolismo , Pulmón/inmunología , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Modelos Biológicos , Mutación
17.
J Bacteriol ; 189(13): 4614-23, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17483222

RESUMEN

Many processes that are essential for mycobacterial growth are poorly understood. To facilitate genetic analyses of such processes in mycobacteria, we and others have developed regulated expression systems that are repressed by a tetracycline repressor (TetR) and induced with tetracyclines, permitting the construction of conditional mutants of essential genes. A disadvantage of these systems is that tetracyclines function as transcriptional inducers and have to be removed to initiate gene silencing. Recently, reverse TetR mutants were identified that require tetracyclines as co-repressors. Here, we report that one of these mutants, TetR r1.7, allows efficient repression of lacZ expression in Mycobacterium smegmatis in the presence but not the absence of anhydrotetracycline (atc). TetR and TetR r1.7 also allowed efficient silencing of the essential secA1 gene, as demonstrated by inhibition of the growth of a conditional mutant and dose-dependent depletion of the SecA1 protein after the removal or addition, respectively, of atc. The kinetics of SecA1 depletion were similar with TetR and TetR r1.7. To test whether silencing of secA1 could help identify substrates of the general secretion pathway, we analyzed the main porin of M. smegmatis, MspA. This showed that the amount of cell envelope-associated MspA decreased more than 90-fold after secA1 silencing. We thus demonstrated that TetR r1.7 allows the construction of conditional mycobacterial mutants in which the expression of an essential gene can be efficiently silenced by the addition of atc and that gene silencing permits the identification of candidate substrates of mycobacterial secretion systems.


Asunto(s)
Proteínas Bacterianas/genética , Silenciador del Gen , Mycobacterium smegmatis/genética , Transactivadores/genética , Proteínas Bacterianas/metabolismo , Western Blotting , Relación Dosis-Respuesta a Droga , Electroforesis en Gel de Poliacrilamida , Cinética , Mutación , Mycobacterium smegmatis/efectos de los fármacos , Mycobacterium smegmatis/crecimiento & desarrollo , Regiones Promotoras Genéticas/genética , Tetraciclina/farmacología , Factores de Tiempo , Transactivadores/metabolismo , beta-Galactosidasa/metabolismo
18.
Nucleic Acids Res ; 33(2): e21, 2005 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-15687379

RESUMEN

Gene expression systems that allow the regulation of bacterial genes during an infection are valuable molecular tools but are lacking for mycobacterial pathogens. We report the development of mycobacterial gene regulation systems that allow controlling gene expression in fast and slow-growing mycobacteria, including Mycobacterium tuberculosis, using anhydrotetracycline (ATc) as inducer. The systems are based on the Escherichia coli Tn10-derived tet regulatory system and consist of a strong tet operator (tetO)-containing mycobacterial promoter, expression cassettes for the repressor TetR and the chemical inducer ATc. These systems allow gene regulation over two orders of magnitude in Mycobacterium smegmatis and M.tuberculosis. TetR-controlled gene expression was inducer concentration-dependent and maximal with ATc concentrations at least 10- and 20-fold below the minimal inhibitory concentration for M.smegmatis and M.tuberculosis, respectively. Using the essential mycobacterial gene ftsZ, we showed that these expression systems can be used to construct conditional knockouts and to analyze the function of essential mycobacterial genes. Finally, we demonstrated that these systems allow gene regulation in M.tuberculosis within the macrophage phagosome.


Asunto(s)
Regulación Bacteriana de la Expresión Génica , Mycobacterium/genética , Proteínas Represoras/metabolismo , Tetraciclinas/farmacología , Animales , Proteínas Bacterianas/genética , Proteínas del Citoesqueleto/genética , Relación Dosis-Respuesta a Droga , Macrófagos/microbiología , Ratones , Mycobacterium smegmatis/citología , Mycobacterium smegmatis/genética , Mycobacterium smegmatis/metabolismo , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/metabolismo , Regiones Operadoras Genéticas , Fagosomas/microbiología , Regiones Promotoras Genéticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...