Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
PLoS Pathog ; 18(6): e1010621, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35771771

RESUMEN

Brucellae are facultative intracellular Gram-negative coccobacilli that chronically infect various mammals and cause brucellosis. Human brucellosis is among the most common bacterial zoonoses and the vast majority of cases are attributed to B. melitensis. Using transposon sequencing (Tn-seq) analysis, we showed that among 3369 predicted genes of the B. melitensis genome, 861 are required for optimal growth in rich medium and 186 additional genes appeared necessary for survival of B. melitensis in RAW 264.7 macrophages in vitro. As the mucosal immune system represents the first defense against Brucella infection, we investigated the early phase of pulmonary infection in mice. In situ analysis at the single cell level indicates a succession of killing and growth phases, followed by heterogenous proliferation of B. melitensis in alveolar macrophages during the first 48 hours of infection. Tn-seq analysis identified 94 additional genes that are required for survival in the lung at 48 hours post infection. Among them, 42 genes are common to RAW 264.7 macrophages and the lung conditions, including the T4SS and purine synthesis genes. But 52 genes are not identified in RAW 264.7 macrophages, including genes implicated in lipopolysaccharide (LPS) biosynthesis, methionine transport, tryptophan synthesis as well as fatty acid and carbohydrate metabolism. Interestingly, genes implicated in LPS synthesis and ß oxidation of fatty acids are no longer required in Interleukin (IL)-17RA-/- mice and asthmatic mice, respectively. This demonstrates that the immune status determines which genes are required for optimal survival and growth of B. melitensis in vivo.


Asunto(s)
Brucella melitensis , Brucelosis , Administración Intranasal , Animales , Brucella melitensis/genética , Brucella melitensis/metabolismo , Lipopolisacáridos/metabolismo , Macrófagos , Mamíferos , Ratones
2.
Proc Natl Acad Sci U S A ; 118(32)2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34353909

RESUMEN

Perturbation of the endoplasmic reticulum (ER), a central organelle of the cell, can have critical consequences for cellular homeostasis. An elaborate surveillance system known as ER quality control ensures that cells can respond and adapt to stress via the unfolded protein response (UPR) and that only correctly assembled proteins reach their destination. Interestingly, several bacterial pathogens hijack the ER to establish an infection. However, it remains poorly understood how bacterial pathogens exploit ER quality-control functions to complete their intracellular cycle. Brucella spp. replicate extensively within an ER-derived niche, which evolves into specialized vacuoles suited for exit from infected cells. Here we present Brucella-secreted protein L (BspL), a Brucella abortus effector that interacts with Herp, a central component of the ER-associated degradation (ERAD) machinery. We found that BspL enhances ERAD at the late stages of the infection. BspL targeting of Herp and ERAD allows tight control of the kinetics of autophagic Brucella-containing vacuole formation, delaying the last step of its intracellular cycle and cell-to-cell spread. This study highlights a mechanism by which a bacterial pathogen hijacks ERAD components for fine regulation of its intracellular trafficking.


Asunto(s)
Proteínas Bacterianas/metabolismo , Brucella abortus/patogenicidad , Brucelosis/metabolismo , Animales , Proteínas Bacterianas/genética , Brucella abortus/metabolismo , Brucelosis/microbiología , Retículo Endoplásmico/metabolismo , Degradación Asociada con el Retículo Endoplásmico , Células HeLa , Interacciones Huésped-Patógeno/fisiología , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Factor de Transcripción CHOP/genética , Sistemas de Secreción Tipo IV/metabolismo , Proteína 1 de Unión a la X-Box/genética
3.
PLoS Pathog ; 17(9): e1009887, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34525130

RESUMEN

Brucellosis is one of the most widespread bacterial zoonoses worldwide. Here, our aim was to identify the effector mechanisms controlling the early stages of intranasal infection with Brucella in C57BL/6 mice. During the first 48 hours of infection, alveolar macrophages (AMs) are the main cells infected in the lungs. Using RNA sequencing, we identified the aconitate decarboxylase 1 gene (Acod1; also known as Immune responsive gene 1), as one of the genes most upregulated in murine AMs in response to B. melitensis infection at 24 hours post-infection. Upregulation of Acod1 was confirmed by RT-qPCR in lungs infected with B. melitensis and B. abortus. We observed that Acod1-/- C57BL/6 mice display a higher bacterial load in their lungs than wild-type (wt) mice following B. melitensis or B. abortus infection, demonstrating that Acod1 participates in the control of pulmonary Brucella infection. The ACOD1 enzyme is mostly produced in mitochondria of macrophages, and converts cis-aconitate, a metabolite in the Krebs cycle, into itaconate. Dimethyl itaconate (DMI), a chemically-modified membrane permeable form of itaconate, has a dose-dependent inhibitory effect on Brucella growth in vitro. Interestingly, structural analysis suggests the binding of itaconate into the binding site of B. abortus isocitrate lyase. DMI does not inhibit multiplication of the isocitrate lyase deletion mutant ΔaceA B. abortus in vitro. Finally, we observed that, unlike the wt strain, the ΔaceA B. abortus strain multiplies similarly in wt and Acod1-/- C57BL/6 mice. These data suggest that bacterial isocitrate lyase might be a target of itaconate in AMs.


Asunto(s)
Brucelosis/inmunología , Carboxiliasas/inmunología , Enfermedades Pulmonares/inmunología , Macrófagos Alveolares/inmunología , Animales , Isocitratoliasa/metabolismo , Ratones , Ratones Endogámicos C57BL
4.
Mol Microbiol ; 116(6): 1449-1463, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34662460

RESUMEN

Cyclic-di-GMP plays crucial role in the cell cycle regulation of the α-Proteobacterium Caulobacter crescentus. Here we investigated its role in the α-Proteobacterium Brucella abortus, a zoonotic intracellular pathogen. Surprisingly, deletion of all predicted cyclic-di-GMP synthesizing or degrading enzymes did not drastically impair the growth of B. abortus, nor its ability to grow inside cell lines. As other Rhizobiales, B. abortus displays unipolar growth from the new cell pole generated by cell division. We found that the phosphodiesterase PdeA, the ortholog of the essential polar growth factor RgsP of the Rhizobiale Sinorhizobium meliloti, is required for rod shape integrity but is not essential for B. abortus growth. Indeed, the radius of the pole is increased by 31 ± 1.7% in a ΔpdeA mutant, generating a coccoid morphology. A mutation in the cyclic-di-GMP phosphodiesterase catalytic site of PdeA does not generate the coccoid morphology and the ΔpdeA mutant kept the ability to recruit markers of new and old poles. However, the presence of PdeA is required in an intra-nasal mouse model of infection. In conclusion, we propose that PdeA contributes to bacterial morphology and virulence in B. abortus, but it is not crucial for polarity and asymmetric growth.


Asunto(s)
Proteínas Bacterianas/metabolismo , Brucella abortus/enzimología , Brucella abortus/crecimiento & desarrollo , Brucelosis/microbiología , Hidrolasas Diéster Fosfóricas/metabolismo , Animales , Proteínas Bacterianas/genética , Brucella abortus/genética , Brucella abortus/metabolismo , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , Femenino , Regulación Bacteriana de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Hidrolasas Diéster Fosfóricas/genética
5.
J Immunol ; 196(9): 3780-93, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-27036913

RESUMEN

The mucosal immune system represents the first line of defense against Brucella infection in nature. We used genetically deficient mice to identify the lymphocytes and signaling pathways implicated in the control of primary and secondary intranasal infection with B. melitensis Our analysis of primary infection demonstrated that the effectors implicated differ at the early and late stages and are dependent on the organ. TCR-δ, TAP1, and IL-17RA deficiency specifically affects early control of Brucella in the lungs, whereas MHC class II (MHCII) and IFN-γR deficiency impairs late control in the lungs, spleen, and liver. Interestingly, IL-12p35(-/-) mice display enhanced Brucella growth in the spleen but not in the lungs or liver. Secondary intranasal infections are efficiently contained in the lung. In contrast to an i.p. infectious model, in which IL-12p35, MHCII, and B cells are strictly required for the control of secondary infection, we observed that only TCR-ß deficiency or simultaneous neutralization of IL-12p35- and IL-17A-dependent pathways impairs the memory protective response against a secondary intranasal infection. Protection is not affected by TCR-δ, MHCII, TAP1, B cell, IL-17RA, or IL-12p35 deficiency, suggesting that CD4(+) and CD8(+) α/ß(+) T cells are sufficient to mount a protective immune response and that an IL-17A-mediated response can compensate for the partial deficiency of an IFN-γ-mediated response to control a Brucella challenge. These findings demonstrate that the nature of the protective memory response depends closely on the route of infection and highlights the role of IFN-γ-and IL-17RA-mediated responses in the control of mucosal infection by Brucella.


Asunto(s)
Brucella melitensis/inmunología , Brucelosis/inmunología , Linfocitos T CD8-positivos/inmunología , Interferón gamma/metabolismo , Senos Paranasales/microbiología , Receptores de Interleucina-17/metabolismo , Animales , Células Cultivadas , Inmunidad Mucosa , Memoria Inmunológica , Interferón gamma/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Receptores de Interleucina-17/genética , Transducción de Señal
6.
Infect Immun ; 85(11)2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28808159

RESUMEN

The spleen is known as an important filter for blood-borne pathogens that are trapped by specialized macrophages in the marginal zone (MZ): the CD209+ MZ macrophages (MZMs) and the CD169+ marginal metallophilic macrophages (MMMs). Acute systemic infection strongly impacts MZ populations and the location of T and B lymphocytes. This phenomenon has been linked to reduced chemokine secretion by stromal cells. Brucella spp. are the causative agent of brucellosis, a widespread zoonotic disease. Here, we used Brucella melitensis infection as a model to investigate the impact of chronic stealth infection on splenic MZ macrophage populations. During the late phase of Brucella infection, we observed a loss of both MZMs and MMMs, with a durable disappearance of MZMs, leading to a reduction of the ability of the spleen to take up soluble antigens, beads, and unrelated bacteria. This effect appears to be selective as every other lymphoid and myeloid population analyzed increased during infection, which was also observed following Brucella abortus and Brucella suis infection. Comparison of wild-type and deficient mice suggested that MZ macrophage population loss is dependent on interferon gamma (IFN-γ) receptor but independent of T cells or tumor necrosis factor alpha receptor 1 (TNF-αR1) signaling pathways and is not correlated to an alteration of CCL19, CCL21, and CXCL13 chemokine mRNA expression. Our results suggest that MZ macrophage populations are particularly sensitive to persistent low-level IFN-γ-mediated inflammation and that Brucella infection could reduce the ability of the spleen to perform certain MZM- and MMM-dependent tasks, such as antigen delivery to lymphocytes and control of systemic infection.


Asunto(s)
Brucelosis/inmunología , Interacciones Huésped-Patógeno , Interferón gamma/inmunología , Macrófagos/inmunología , Receptores de Interferón/inmunología , Bazo/inmunología , Animales , Antibacterianos/farmacología , Linfocitos B/inmunología , Linfocitos B/microbiología , Brucella abortus/efectos de los fármacos , Brucella abortus/inmunología , Brucella abortus/patogenicidad , Brucella melitensis/efectos de los fármacos , Brucella melitensis/inmunología , Brucella melitensis/patogenicidad , Brucella suis/efectos de los fármacos , Brucella suis/inmunología , Brucella suis/patogenicidad , Brucelosis/tratamiento farmacológico , Brucelosis/genética , Brucelosis/microbiología , Quimiocina CCL19/genética , Quimiocina CCL19/inmunología , Quimiocina CCL21/genética , Quimiocina CCL21/inmunología , Quimiocina CXCL13/genética , Quimiocina CXCL13/inmunología , Enfermedad Crónica , Regulación de la Expresión Génica , Interferón gamma/genética , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/genética , ARN Mensajero/inmunología , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Receptores Tipo I de Factores de Necrosis Tumoral/deficiencia , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/inmunología , Rifampin/farmacología , Transducción de Señal , Bazo/microbiología , Estreptomicina/farmacología , Linfocitos T/inmunología , Linfocitos T/microbiología , Receptor de Interferón gamma
7.
J Immunol ; 192(8): 3740-52, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24646742

RESUMEN

Brucella spp are intracellular bacteria that cause brucellosis, one of the most common zoonoses in the world. Given the serious medical consequences of this disease, a safe and effective human vaccine is urgently needed. Efforts to develop this vaccine have been hampered by our lack of understanding of what constitutes a protective memory response against Brucella. In this study, we characterize the cells and signaling pathways implicated in the generation of a protective immune memory response following priming by the injection of heat-killed or live Brucella melitensis 16M. Using a panel of gene-deficient mice, we demonstrated that during a secondary recall response, both the Brucella-specific humoral response and CD4+ Th1 cells must act together to confer protective immunity in the spleen to B. melitensis infection. Humoral protective immunity is induced by the inoculation of both heat-killed and live bacteria, and its development does not require T cells, MyD88/IL-12p35 signaling pathways, or an activation-induced deaminase-mediated isotype switch. In striking contrast, the presence of memory IFN-γ-producing CD4+ Th1 cells requires the administration of live bacteria and functional MyD88/IL-12p35 pathways. In summary, our work identifies several immune markers closely associated with protective immune memory and could help to define a rational strategy to obtain an effective human vaccine against brucellosis.


Asunto(s)
Brucella melitensis/inmunología , Brucelosis/inmunología , Inmunidad Humoral , Células TH1/inmunología , Animales , Anticuerpos Antibacterianos/inmunología , Especificidad de Anticuerpos/inmunología , Bacteriemia/inmunología , Bacteriemia/prevención & control , Vacuna contra la Brucelosis/administración & dosificación , Vacuna contra la Brucelosis/inmunología , Brucelosis/metabolismo , Brucelosis/prevención & control , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Antígenos H-2/inmunología , Memoria Inmunológica , Interferón gamma/biosíntesis , Interleucina-12/metabolismo , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/metabolismo , Fenotipo , Transducción de Señal , Bazo/citología , Bazo/inmunología , Bazo/microbiología , Células TH1/metabolismo , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/inmunología , Vacunas Vivas no Atenuadas/administración & dosificación , Vacunas Vivas no Atenuadas/inmunología
8.
Infect Immun ; 82(9): 3927-38, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25001604

RESUMEN

Brucella spp. are facultative intracellular Gram-negative coccobacilli responsible for brucellosis, a worldwide zoonosis. We observed that Brucella melitensis is able to persist for several weeks in the blood of intraperitoneally infected mice and that transferred blood at any time point tested is able to induce infection in naive recipient mice. Bacterial persistence in the blood is dramatically impaired by specific antibodies induced following Brucella vaccination. In contrast to Bartonella, the type IV secretion system and flagellar expression are not critically required for the persistence of Brucella in blood. ImageStream analysis of blood cells showed that following a brief extracellular phase, Brucella is associated mainly with the erythrocytes. Examination by confocal microscopy and transmission electron microscopy formally demonstrated that B. melitensis is able to invade erythrocytes in vivo. The bacteria do not seem to multiply in erythrocytes and are found free in the cytoplasm. Our results open up new areas for investigation and should serve in the development of novel strategies for the treatment or prophylaxis of brucellosis. Invasion of erythrocytes could potentially protect the bacterial cells from the host's immune response and hamper antibiotic treatment and suggests possible Brucella transmission by bloodsucking insects in nature.


Asunto(s)
Brucella melitensis/inmunología , Eritrocitos/inmunología , Animales , Sistemas de Secreción Bacterianos/inmunología , Vacuna contra la Brucelosis/inmunología , Brucelosis/inmunología , Brucelosis/microbiología , Eritrocitos/microbiología , Flagelos/inmunología , Flagelos/microbiología , Ratones , Ratones Endogámicos C57BL
9.
PLoS Pathog ; 8(3): e1002575, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22479178

RESUMEN

Brucella are facultative intracellular bacteria that chronically infect humans and animals causing brucellosis. Brucella are able to invade and replicate in a broad range of cell lines in vitro, however the cells supporting bacterial growth in vivo are largely unknown. In order to identify these, we used a Brucella melitensis strain stably expressing mCherry fluorescent protein to determine the phenotype of infected cells in spleen and liver, two major sites of B. melitensis growth in mice. In both tissues, the majority of primary infected cells expressed the F4/80 myeloid marker. The peak of infection correlated with granuloma development. These structures were mainly composed of CD11b⁺ F4/80⁺ MHC-II⁺ cells expressing iNOS/NOS2 enzyme. A fraction of these cells also expressed CD11c marker and appeared similar to inflammatory dendritic cells (DCs). Analysis of genetically deficient mice revealed that differentiation of iNOS⁺ inflammatory DC, granuloma formation and control of bacterial growth were deeply affected by the absence of MyD88, IL-12p35 and IFN-γ molecules. During chronic phase of infection in susceptible mice, we identified a particular subset of DC expressing both CD11c and CD205, serving as a reservoir for the bacteria. Taken together, our results describe the cellular nature of immune effectors involved during Brucella infection and reveal a previously unappreciated role for DC subsets, both as effectors and reservoir cells, in the pathogenesis of brucellosis.


Asunto(s)
Brucella/inmunología , Brucelosis/inmunología , Células Dendríticas/inmunología , Inmunidad Innata , Enfermedades Pulmonares/inmunología , Animales , Biomarcadores/metabolismo , Brucella/patogenicidad , Brucelosis/microbiología , Brucelosis/patología , Separación Celular , Células Dendríticas/microbiología , Células Dendríticas/patología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Hígado/inmunología , Hígado/microbiología , Hígado/patología , Enfermedades Pulmonares/microbiología , Enfermedades Pulmonares/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo I/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fenotipo , Bazo/inmunología , Bazo/microbiología , Bazo/patología
10.
Infect Immun ; 80(12): 4271-80, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23006848

RESUMEN

Brucella spp. are facultative intracellular bacterial pathogens responsible for brucellosis, a worldwide zoonosis that causes abortion in domestic animals and chronic febrile disease associated with serious complications in humans. There is currently no approved vaccine against human brucellosis, and antibiotic therapy is long and costly. Development of a safe protective vaccine requires a better understanding of the roles played by components of adaptive immunity in the control of Brucella infection. The importance of lymphocyte subsets in the control of Brucella growth has been investigated separately by various research groups and remains unclear or controversial. Here, we used a large panel of genetically deficient mice to compare the importance of B cells, transporter associated with antigen processing (TAP-1), and major histocompatibility complex class II-dependent pathways of antigen presentation as well as T helper 1 (Th1), Th2, and Th17-mediated responses on the immune control of Brucella melitensis 16 M infection. We clearly confirmed the key function played by gamma interferon (IFN-γ)-producing Th1 CD4(+) T cells in the control of B. melitensis infection, whereas IFN-γ-producing CD8(+) T cells or B cell-mediated humoral immunity plays only a modest role in the clearance of bacteria during primary infection. In the presence of a Th1 response, Th2 or Th17 responses do not really develop or play a positive or negative role during the course of B. melitensis infection. On the whole, these results could improve our ability to develop protective vaccines or therapeutic treatments against brucellosis.


Asunto(s)
Brucella melitensis/patogenicidad , Brucelosis/inmunología , Linfocitos T CD4-Positivos/inmunología , Interferón gamma/biosíntesis , Células TH1/inmunología , Animales , Linfocitos B/inmunología , Brucella melitensis/inmunología , Brucelosis/microbiología , Brucelosis/prevención & control , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , Células Th17/inmunología , Células Th2/inmunología
11.
PLoS Pathog ; 6(10): e1001154, 2010 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-20976202

RESUMEN

Protozoa and bacteria infect various types of phagocytic cells including macrophages, monocytes, dendritic cells and eosinophils. However, it is not clear which of these cells process and present microbial antigens in vivo and in which cellular compartments parasite peptides are loaded onto Major Histocompatibility Complex molecules. To address these issues, we have infected susceptible BALB/c (H-2d) mice with a recombinant Leishmania major parasite expressing a fluorescent tracer. To directly visualize the antigen presenting cells that present parasite-derived peptides to CD4+ T cells, we have generated a monoclonal antibody that reacts to an antigenic peptide derived from the parasite LACK antigen bound to I-Ad Major Histocompatibility Complex class II molecule. Immunogold electron microscopic analysis of in vivo infected cells showed that intracellular I-Ad/LACK complexes were present in the membrane of amastigote-containing phagosomes in dendritic cells, eosinophils and macrophages/monocytes. In both dendritic cells and macrophages, these complexes were also present in smaller vesicles that did not contain amastigote. The presence of I-Ad/LACK complexes at the surface of dendritic cells, but neither on the plasma membrane of macrophages nor eosinophils was independently confirmed by flow cytometry and by incubating sorted phagocytes with highly sensitive LACK-specific hybridomas. Altogether, our results suggest that peptides derived from Leishmania proteins are loaded onto Major Histocompatibility Complex class II molecules in the phagosomes of infected phagocytes. Although these complexes are transported to the cell surface in dendritic cells, therefore allowing the stimulation of parasite-specific CD4+ T cells, this does not occur in other phagocytic cells. To our knowledge, this is the first study in which Major Histocompatibility Complex class II molecules bound to peptides derived from a parasite protein have been visualized within and at the surface of cells that were infected in vivo.


Asunto(s)
Membrana Celular/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Leishmania major/inmunología , Leishmaniasis Cutánea/inmunología , Leishmaniasis Cutánea/metabolismo , Fragmentos de Péptidos/metabolismo , Secuencia de Aminoácidos , Animales , Compartimento Celular/inmunología , Membrana Celular/inmunología , Células Cultivadas , Técnica del Anticuerpo Fluorescente/métodos , Antígenos de Histocompatibilidad Clase II/inmunología , Membranas Intracelulares/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Fragmentos de Péptidos/química , Fragmentos de Péptidos/inmunología , Fagosomas/inmunología , Fagosomas/metabolismo
12.
Front Public Health ; 10: 902724, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35875018

RESUMEN

In hindsight, the early response of liberal governments to the SARS-CoV-2 pandemic was chaotic and generally inefficient. Though one might be tempted to attribute these failures to the incompetence of certain political decision-makers, we propose another explanation. Global threats require a coordinated international response, which is only possible if the threat is perceived in the same way by all, and if government priorities are similar. The effectiveness of the response also relies on massive adhesion of citizens to the measures imposed, which in turn requires trust in government. Our hypothesis is that certain fundamental features of liberalism complicate such global and collective responses: neutrality of the state and primacy of the individual over collective society. Liberalism considers that institutions and public policy must not be designed to favor any specific conception of the common good. That which is best for all is usually determined by a "competition of opinions," which frequently leads to scientific expertise being considered as only one opinion among many. Liberalism also imposes strict respect for individual freedoms and private interests and tends to reject any form of collectivism or dictate imposed by the common good. In order to solve these structural problems and improve society's management of global threats, we make several proposals, such as the introduction of a minimal and consensual definition of the common good and the promotion of a health policy guided by One Health-like concepts. Overall, our analysis suggests that because political ideologies provide their own definitions of the common good and the place of scientific knowledge in the governance process and can thus affect the response to global threats, they should be urgently taken into consideration by public health experts.


Asunto(s)
COVID-19 , SARS-CoV-2 , COVID-19/epidemiología , Humanos , Pandemias , Salud Pública , Política Pública
13.
Eur J Immunol ; 40(7): 1995-2005, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20432233

RESUMEN

Experimental animal and human studies have demonstrated that probiotic strains have beneficial effects on allergy. Here we report that the probiotic Escherichia coli Nissle 1917 strain (EcN) is able to activate DC, as shown by important cytokine synthesis together with up-regulation of membrane expression of CD40, CD80 and CD86. This EcN-induced DC activation was strictly dependent on the TLR4 signaling pathway and was also associated with stimulation of NF-kappaB and MAPK. We next investigated the prophylactic potential of i.n. co-administration of EcN with a recombinant form of Der p 1 (ProDer p 1) in a murine model of mite allergy. I.n. vaccinations with EcN plus ProDer p 1 prevented the subsequent allergic response following Der p 1 sensitization and airway challenge with aerosolized mite extracts through the induction of an allergen-specific IgG2a response, the prevention of specific IgE production and a strong reduction of IL-5 secretion by allergen-restimulated splenocytes. EcN alone or in combination with ProDer p 1 inhibited the development of airway eosinophilia and neutrophilia. This in vivo protective effect of EcN was, in part, mediated by TLR4 signaling. Our results suggest that EcN represents an efficient adjuvant to prevent allergic responses.


Asunto(s)
Células Dendríticas/metabolismo , Desensibilización Inmunológica , Escherichia coli/inmunología , Hipersensibilidad/inmunología , Hipersensibilidad/microbiología , Probióticos/administración & dosificación , Animales , Antígenos CD/biosíntesis , Antígenos Dermatofagoides/administración & dosificación , Antígenos Dermatofagoides/inmunología , Antígenos de Diferenciación/biosíntesis , Proteínas de Artrópodos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Cisteína Endopeptidasas , Citocinas/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Hipersensibilidad/fisiopatología , Hipersensibilidad/terapia , Ratones , Ratones Endogámicos BALB C , Pyroglyphidae/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Receptor Toll-Like 4/metabolismo
14.
PLoS Pathog ; 5(6): e1000494, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19557162

RESUMEN

Leishmania major parasites reside and multiply in late endosomal compartments of host phagocytic cells. Immune control of Leishmania growth absolutely requires expression of inducible Nitric Oxide Synthase (iNOS/NOS2) and subsequent production of NO. Here, we show that CD11b+ CD11c+ Ly-6C+ MHC-II+ cells are the main iNOS-producing cells in the footpad lesion and in the draining lymph node of Leishmania major-infected C57BL/6 mice. These cells are phenotypically similar to iNOS-producing inflammatory DC (iNOS-DC) observed in the mouse models of Listeria monocytogenes and Brucella melitensis infection. The use of DsRed-expressing parasites demonstrated that these iNOS-producing cells are the major infected population in the lesions and the draining lymph nodes. Analysis of various genetically deficient mouse strains revealed the requirement of CCR2 expression for the recruitment of iNOS-DC in the draining lymph nodes, whereas their activation is strongly dependent on CD40, IL-12, IFN-gamma and MyD88 molecules with a partial contribution of TNF-alpha and TLR9. In contrast, STAT-6 deficiency enhanced iNOS-DC recruitment and activation in susceptible BALB/c mice, demonstrating a key role for IL-4 and IL-13 as negative regulators. Taken together, our results suggest that iNOS-DC represent a major class of Th1-regulated effector cell population and constitute the most frequent infected cell type during chronic Leishmania major infection phase of C57BL/6 resistant mice.


Asunto(s)
Células Dendríticas/enzimología , Leishmania major/inmunología , Leishmaniasis Cutánea/inmunología , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Animales , Enfermedad Crónica , Citocinas/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/parasitología , Citometría de Flujo , Inflamación/enzimología , Inflamación/parasitología , Leishmaniasis Cutánea/enzimología , Leishmaniasis Cutánea/parasitología , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ganglios Linfáticos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fenotipo , Receptores CCR2/metabolismo , Estadísticas no Paramétricas
15.
Front Immunol ; 12: 723516, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34566981

RESUMEN

The newborns of women infected with the parasite Trypanosoma cruzi (the agent of Chagas disease) can be infected either before birth (congenitally), or after birth (as e.g., by vector route). Congenital Chagas disease can induce high levels of neonatal morbidity and mortality. Parasite-infected pregnant women transmit antibodies to their fetus. Antibodies, by opsonizing parasites, can promote phagocytosis and killing of T. cruzi by cells expressing FcγR, on the mandatory condition that such cells are sufficiently activated in an inflammatory context. Antibody-dependent enhancement (ADE) is a mechanism well described in viral infections, by which antibodies enhance entry of infectious agents into host cells by exploiting the phagocytic FcγR pathway. Previously reported Chagas disease studies highlighted a severe reduction of the maternal-fetal/neonatal inflammatory context in parasite-transmitting pregnant women and their congenitally infected newborns. Otherwise, experimental observations brought to light ADE of T. cruzi infection (involving FcγR) in mouse pups displaying maternally transferred antibodies, out of an inflammatory context. Herein, based on such data, we discuss the previously unconsidered possibility of a role of ADE in the trans-placental parasite transmission, and/or the development of severe and mortal clinical forms of congenital/neonatal Chagas disease in newborns of T. cruzi-infected mothers.


Asunto(s)
Acrecentamiento Dependiente de Anticuerpo , Enfermedad de Chagas/inmunología , Transmisión Vertical de Enfermedad Infecciosa , Placenta/parasitología , Trypanosoma cruzi/inmunología , Animales , Enfermedad de Chagas/congénito , Enfermedad de Chagas/parasitología , Femenino , Humanos , Recién Nacido , Ratones , Placenta/inmunología , Embarazo , Complicaciones Parasitarias del Embarazo/inmunología , Mujeres Embarazadas , Trypanosoma cruzi/parasitología
17.
Front Immunol ; 11: 1085, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32655552

RESUMEN

In many infectious diseases, the immune response operates as a double-edged sword. While required for protective immunity, infection-induced inflammation can be detrimental if it is not properly controlled, causing collateral body damage and potentially leading to death. It is in this context that the potent anti-inflammatory cytokine interleukin-10 (IL-10) is required to dampen the pro-inflammatory immune response that hallmarks trypanosomosis. Effective control of this infection requires not just the action of antibodies specific for the parasite's variable surface glycoprotein (VSG) coat antigens, but also a pro-inflammatory immune response mediated mainly by IFNγ, TNF, and NO. However, strict control of inflammation is mandatory, as IL-10-deficient mice succumb from an unrestrained cytokine storm within 10 days of a Trypanosome brucei infection. The relevant cellular source of IL-10 and the associated molecular mechanisms implicated in its trypanosomosis associated production are poorly understood. Using an IL-10 reporter mouse strain (Vert-X), we demonstrate here that NK cells, CD8+ T cells and CD4+ T cells as well as B cells and plasma cells constitute potential cellular sources of IL-10 within the spleen and liver during acute infection. The IL-10 wave follows peak pro-inflammatory cytokine production, which accompanied the control of peak parasitemia. Similar results were observed following conventional experimental needle infection and physiological infections via T. brucei-infected tsetse flies. Our results show that conditional T cell-specific ablation of the IL-10 regulating Prdm1 gene (encoding for the Blimp-1 transcription factor), leads to an uncontrolled trypanosome-induced pro-inflammatory syndrome like the one observed in infected IL-10-deficient mice. This result indicates that the biological role of IL-10-derived from non-T cells, including NK cells, is of minor importance when considering host survival. The cytokine IL-27 that is also considered to be an IL-10 regulator, did not affect IL-10 production during infection. Together, these data suggest that T. brucei activates a Blimp-1-dependent IL-10 regulatory pathway in T cells that acts as a critical anti-inflammatory rheostat, mandatory for host survival during the acute phase of parasitemia.


Asunto(s)
Síndrome de Liberación de Citoquinas/prevención & control , Interleucina-10/biosíntesis , Factor 1 de Unión al Dominio 1 de Regulación Positiva/inmunología , Linfocitos T/inmunología , Trypanosoma brucei brucei , Tripanosomiasis Africana/inmunología , Animales , Síndrome de Liberación de Citoquinas/etiología , Síndrome de Liberación de Citoquinas/inmunología , Modelos Animales de Enfermedad , Femenino , Inflamación/etiología , Inflamación/inmunología , Inflamación/prevención & control , Insectos Vectores/parasitología , Interleucina-10/deficiencia , Interleucina-10/genética , Interleucinas/antagonistas & inhibidores , Interleucinas/deficiencia , Interleucinas/inmunología , Hígado/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 1 de Unión al Dominio 1 de Regulación Positiva/deficiencia , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Bazo/inmunología , Tripanosomiasis Africana/complicaciones , Tripanosomiasis Africana/parasitología , Moscas Tse-Tse/parasitología
18.
Endeavour ; 43(4): 100709, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32115245

RESUMEN

The independence of science was long seen as of prime importance. This position has become less common today. The perception of scientific research as a public service has led to the opinion that it must be accountable to citizens and produce knowledge and innovation that meet their expectations. Numerous authors have voiced the need for anticipatory ethical control of innovation focusing on the scientific research process. This control is considered as the must-have guarantee for "good science." The current article attempts to trace the ideological origins of the ethical control of innovation, examines its effectiveness against the challenge of globalization and technology-derived major threats and its compatibility with scientific methodology. It also suggests ways to both regulate the innovation process and preserve the independence of science. On the whole, we conclude that truly effective ethical regulation of innovation, i.e. one that protects the greatest number from its adverse effects, is achieved first and foremost by questioning our liberal economic model and the place given to science in our societies.

19.
Nat Commun ; 10(1): 4847, 2019 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-31649248

RESUMEN

It is assumed that intracellular pathogenic bacteria have to cope with DNA alkylating stress within host cells. Here we use single-cell reporter systems to show that the pathogen Brucella abortus does encounter alkylating stress during the first hours of macrophage infection. Genes encoding direct repair and base-excision repair pathways are required by B. abortus to face this stress in vitro and in a mouse infection model. Among these genes, ogt is found to be under the control of the conserved cell-cycle transcription factor GcrA. Our results highlight that the control of DNA repair in B. abortus displays distinct features that are not present in model organisms such as Escherichia coli.


Asunto(s)
Brucella abortus/genética , Daño del ADN/genética , Interacciones Huésped-Patógeno/genética , Macrófagos/metabolismo , Estrés Fisiológico/genética , Alquilación , Animales , Brucella abortus/metabolismo , Brucelosis , Metilación de ADN/genética , Reparación del ADN/genética , Ratones , Células RAW 264.7 , Vacuolas/metabolismo
20.
Front Immunol ; 10: 1589, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31354728

RESUMEN

Live attenuated vaccines play a key role in the control of many human and animal pathogens. Their rational development is usually helped by identification of the reservoir of infection, the lymphoid subpopulations associated with protective immunity as well as the virulence genes involved in pathogen persistence. Here, we compared the course of Brucella melitensis infection in C57BL/6 mice infected via intraperitoneal (i.p.), intranasal (i.n.) and intradermal (i.d.) route and demonstrated that the route of infection strongly impacts all of these parameters. Following i.p. and i.n. infection, most infected cells observed in the spleen or lung were F4/80+ myeloid cells. In striking contrast, infected Ly6G+ neutrophils and CD140a+ fibroblasts were also observed in the skin after i.d. infection. The virB operon encoding for the type IV secretion system is considered essential to deflecting vacuolar trafficking in phagocytic cells and allows Brucella to multiply and persist. Unexpectedly, the ΔvirB Brucella strain, which does not persist in the lung after i.n. infection, persists longer in skin tissues than the wild strain after i.d. infection. While the CD4+ T cell-mediated Th1 response is indispensable to controlling the Brucella challenge in the i.p. model, it is dispensable for the control of Brucella in the i.d. and i.n. models. Similarly, B cells are indispensable in the i.p. and i.d. models but dispensable in the i.n. model. γδ+ T cells appear able to compensate for the absence of αß+ T cells in the i.d. model but not in the other models. Taken together, our results demonstrate the crucial importance of the route of infection for the host pathogen relationship.


Asunto(s)
Brucella melitensis/inmunología , Brucelosis/inmunología , Interacciones Huésped-Patógeno/inmunología , Animales , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos Intraepiteliales/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neutrófilos/inmunología , Células TH1/inmunología , Vacunas Atenuadas/inmunología , Virulencia/inmunología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda