Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Cell ; 165(3): 679-89, 2016 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-27040495

RESUMEN

Increasing antibiotic resistance among bacterial pathogens has rendered some infections untreatable with available antibiotics. Klebsiella pneumoniae, a bacterial pathogen that has acquired high-level antibiotic resistance, is a common cause of pulmonary infections. Optimal clearance of K. pneumoniae from the host lung requires TNF and IL-17A. Herein, we demonstrate that inflammatory monocytes are rapidly recruited to the lungs of K. pneumoniae-infected mice and produce TNF, which markedly increases the frequency of IL-17-producing innate lymphoid cells. While pulmonary clearance of K. pneumoniae is preserved in neutrophil-depleted mice, monocyte depletion or TNF deficiency impairs IL-17A-dependent resolution of pneumonia. Monocyte-mediated bacterial uptake and killing is enhanced by ILC production of IL-17A, indicating that innate lymphocytes engage in a positive-feedback loop with monocytes that promotes clearance of pneumonia. Innate immune defense against a highly antibiotic-resistant bacterial pathogen depends on crosstalk between inflammatory monocytes and innate lymphocytes that is mediated by TNF and IL-17A.


Asunto(s)
Infecciones por Klebsiella/inmunología , Klebsiella pneumoniae/fisiología , Animales , Inflamación/inmunología , Interleucina-17/inmunología , Infecciones por Klebsiella/microbiología , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Linfocitos/inmunología , Ratones , Monocitos/inmunología , Factor de Necrosis Tumoral alfa/inmunología
2.
Nat Immunol ; 17(4): 379-86, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26901151

RESUMEN

The T cell antigen receptor (TCR) is unique in that its affinity for ligand is unknown before encounter and can vary by orders of magnitude. How the immune system regulates individual T cells that display very different reactivity to antigen remains unclear. Here we found that activated CD4(+) T cells, at the peak of clonal expansion, persistently downregulated their TCR expression in proportion to the strength of the initial antigen recognition. This programmed response increased the threshold for cytokine production and recall proliferation in a clone-specific manner and ultimately excluded clones with the highest antigen reactivity. Thus, programmed downregulation of TCR expression represents a negative feedback mechanism for constraining T cell effector function with a suitable time delay to thereby allow pathogen control while avoiding excess inflammatory damage.


Asunto(s)
Regulación hacia Abajo , Listeriosis/inmunología , Receptores de Antígenos de Linfocitos T/genética , Células TH1/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Linfocitos T CD4-Positivos/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Immunoblotting , Listeria monocytogenes , Activación de Linfocitos , Ratones , Ratones Transgénicos , Mycobacterium tuberculosis , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T , Transcriptoma
3.
Nature ; 572(7771): 665-669, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31435014

RESUMEN

Intestinal commensal bacteria can inhibit dense colonization of the gut by vancomycin-resistant Enterococcus faecium (VRE), a leading cause of hospital-acquired infections1,2. A four-strained consortium of commensal bacteria that contains Blautia producta BPSCSK can reverse antibiotic-induced susceptibility to VRE infection3. Here we show that BPSCSK reduces growth of VRE by secreting a lantibiotic that is similar to the nisin-A produced by Lactococcus lactis. Although the growth of VRE is inhibited by BPSCSK and L. lactis in vitro, only BPSCSK colonizes the colon and reduces VRE density in vivo. In comparison to nisin-A, the BPSCSK lantibiotic has reduced activity against intestinal commensal bacteria. In patients at high risk of VRE infection, high abundance of the lantibiotic gene is associated with reduced density of E. faecium. In germ-free mice transplanted with patient-derived faeces, resistance to VRE colonization correlates with abundance of the lantibiotic gene. Lantibiotic-producing commensal strains of the gastrointestinal tract reduce colonization by VRE and represent potential probiotic agents to re-establish resistance to VRE.


Asunto(s)
Bacteriocinas/metabolismo , Bacteriocinas/farmacología , Enterococcus faecium/efectos de los fármacos , Lactococcus lactis/metabolismo , Probióticos , Resistencia a la Vancomicina/efectos de los fármacos , Enterococos Resistentes a la Vancomicina/efectos de los fármacos , Animales , Antibacterianos/biosíntesis , Antibacterianos/aislamiento & purificación , Antibacterianos/metabolismo , Antibacterianos/farmacología , Bacteriocinas/genética , Bacteriocinas/aislamiento & purificación , Enterococcus faecium/crecimiento & desarrollo , Enterococcus faecium/aislamiento & purificación , Heces/microbiología , Femenino , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/microbiología , Vida Libre de Gérmenes , Bacterias Grampositivas/efectos de los fármacos , Bacterias Grampositivas/crecimiento & desarrollo , Humanos , Lactococcus lactis/química , Lactococcus lactis/crecimiento & desarrollo , Lactococcus lactis/fisiología , Ratones , Pruebas de Sensibilidad Microbiana , Microbiota/genética , Nisina/química , Nisina/farmacología , Simbiosis/efectos de los fármacos , Vancomicina/farmacología , Enterococos Resistentes a la Vancomicina/crecimiento & desarrollo , Enterococos Resistentes a la Vancomicina/aislamiento & purificación
4.
Immunity ; 36(2): 276-87, 2012 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-22306017

RESUMEN

Microbial penetration of the intestinal epithelial barrier triggers inflammatory responses that include induction of the bactericidal C-type lectin RegIIIγ. Systemic administration of flagellin, a bacterial protein that stimulates Toll-like receptor 5 (TLR5), induces epithelial expression of RegIIIγ and protects mice from intestinal colonization with antibiotic-resistant bacteria. Flagellin-induced RegIIIγ expression is IL-22 dependent, but how TLR signaling leads to IL-22 expression is incompletely defined. By using conditional depletion of lamina propria dendritic cell (LPDC) subsets, we demonstrated that CD103(+)CD11b(+) LPDCs, but not monocyte-derived CD103(-)CD11b(+) LPDCs, expressed high amounts of IL-23 after bacterial flagellin administration and drove IL-22-dependent RegIIIγ production. Maximal expression of IL-23 subunits IL-23p19 and IL-12p40 occurred within 60 min of exposure to flagellin. IL-23 subsequently induced a burst of IL-22 followed by sustained RegIIIγ expression. Thus, CD103(+)CD11b(+) LPDCs, in addition to promoting long-term tolerance to ingested antigens, also rapidly produce IL-23 in response to detection of flagellin in the lamina propria.


Asunto(s)
Células Dendríticas/inmunología , Flagelina/inmunología , Interleucina-23/biosíntesis , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Animales , Antígenos CD/metabolismo , Antígeno CD11b/metabolismo , Células Dendríticas/clasificación , Flagelina/administración & dosificación , Inmunidad Innata , Inmunidad Mucosa , Cadenas alfa de Integrinas/metabolismo , Interleucina-23/deficiencia , Interleucina-23/genética , Interleucinas/biosíntesis , Interleucinas/deficiencia , Interleucinas/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Pancreatitis , Proteínas/genética , Transducción de Señal/inmunología , Receptor Toll-Like 5/deficiencia , Receptor Toll-Like 5/genética , Receptor Toll-Like 5/metabolismo , Regulación hacia Arriba , Interleucina-22
5.
Infect Immun ; 88(4)2020 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-31907198

RESUMEN

Antibiotic treatment of patients undergoing complex medical treatments can deplete commensal bacterial strains from the intestinal microbiota, thereby reducing colonization resistance against a wide range of antibiotic-resistant pathogens. Loss of colonization resistance can lead to marked expansion of vancomycin-resistant Enterococcus faecium (VRE), Klebsiella pneumoniae, and Escherichia coli in the intestinal lumen, predisposing patients to bloodstream invasion and sepsis. The impact of intestinal domination by these antibiotic-resistant pathogens on mucosal immune defenses and epithelial and mucin-mediated barrier integrity is unclear. We used a mouse model to study the impact of intestinal domination by antibiotic-resistant bacterial species and strains on the colonic mucosa. Intestinal colonization with K. pneumoniae, Proteus mirabilis, or Enterobacter cloacae promoted greater recruitment of neutrophils to the colonic mucosa. To test the hypothesis that the residual microbiota influences the severity of colitis caused by infection with Clostridioides difficile, we coinfected mice that were colonized with ampicillin-resistant bacteria with a virulent strain of C. difficile and monitored colonization and pathogenesis. Despite the compositional differences in the gut microbiota, the severity of C. difficile infection (CDI) and mortality did not differ significantly between mice colonized with different ampicillin-resistant bacterial species. Our results suggest that the virulence mechanisms enabling CDI and epithelial destruction outweigh the relatively minor impact of less-virulent antibiotic-resistant intestinal bacteria on the outcome of CDI.


Asunto(s)
Antibacterianos/administración & dosificación , Infecciones por Clostridium/fisiopatología , Farmacorresistencia Bacteriana , Enterobacter cloacae/crecimiento & desarrollo , Infecciones por Enterobacteriaceae/complicaciones , Klebsiella pneumoniae/crecimiento & desarrollo , Proteus mirabilis/crecimiento & desarrollo , Animales , Infecciones por Clostridium/microbiología , Colitis/microbiología , Colitis/fisiopatología , Modelos Animales de Enfermedad , Enterobacter cloacae/efectos de los fármacos , Infecciones por Enterobacteriaceae/tratamiento farmacológico , Klebsiella pneumoniae/efectos de los fármacos , Ratones , Interacciones Microbianas , Proteus mirabilis/efectos de los fármacos , Análisis de Supervivencia
6.
Immunity ; 34(4): 590-601, 2011 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-21458307

RESUMEN

Inflammatory (Ly6C(hi) CCR2+) monocytes provide defense against infections but also contribute to autoimmune diseases and atherosclerosis. Monocytes originate from bone marrow and their entry into the bloodstream requires stimulation of CCR2 chemokine receptor by monocyte chemotactic protein-1 (MCP1). How monocyte emigration from bone marrow is triggered by remote infections remains unclear. We demonstrated that low concentrations of Toll-like receptor (TLR) ligands in the bloodstream drive CCR2-dependent emigration of monocytes from bone marrow. Bone marrow mesenchymal stem cells (MSCs) and their progeny, including CXC chemokine ligand (CXCL)12-abundant reticular (CAR) cells, rapidly expressed MCP1 in response to circulating TLR ligands or bacterial infection and induced monocyte trafficking into the bloodstream. Targeted deletion of MCP1 from MSCs impaired monocyte emigration from bone marrow. Our findings suggest that bone marrow MSCs and CAR cells respond to circulating microbial molecules and regulate bloodstream monocyte frequencies by secreting MCP1 in proximity to bone marrow vascular sinuses.


Asunto(s)
Médula Ósea/inmunología , Movimiento Celular , Células Madre Mesenquimatosas/inmunología , Monocitos/citología , Monocitos/inmunología , Receptores Toll-Like/inmunología , Animales , Ligandos , Lipopolisacáridos/inmunología , Ratones , Ratones Endogámicos C57BL , Receptores CCR2/inmunología
7.
PLoS Pathog ; 11(9): e1005132, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26334306

RESUMEN

Antibiotic resistance among enterococci and γ-proteobacteria is an increasing problem in healthcare settings. Dense colonization of the gut by antibiotic-resistant bacteria facilitates their spread between patients and also leads to bloodstream and other systemic infections. Antibiotic-mediated destruction of the intestinal microbiota and consequent loss of colonization resistance are critical factors leading to persistence and spread of antibiotic-resistant bacteria. The mechanisms underlying microbiota-mediated colonization resistance remain incompletely defined and are likely distinct for different antibiotic-resistant bacterial species. It is unclear whether enterococci or γ-proteobacteria, upon expanding to high density in the gut, confer colonization resistance against competing bacterial species. Herein, we demonstrate that dense intestinal colonization with vancomycin-resistant Enterococcus faecium (VRE) does not reduce in vivo growth of carbapenem-resistant Klebsiella pneumoniae. Reciprocally, K. pneumoniae does not impair intestinal colonization by VRE. In contrast, transplantation of a diverse fecal microbiota eliminates both VRE and K. pneumoniae from the gut. Fluorescence in situ hybridization demonstrates that VRE and K. pneumoniae localize to the same regions in the colon but differ with respect to stimulation and invasion of the colonic mucus layer. While VRE and K. pneumoniae occupy the same three-dimensional space within the gut lumen, their independent growth and persistence in the gut suggests that they reside in distinct niches that satisfy their specific in vivo metabolic needs.


Asunto(s)
Enteritis/microbiología , Enterococcus faecium/fisiología , Infecciones por Bacterias Grampositivas/microbiología , Mucosa Intestinal/microbiología , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/fisiología , Enterococos Resistentes a la Vancomicina/fisiología , Ampicilina/efectos adversos , Animales , Antibacterianos/efectos adversos , Antibacterianos/farmacología , Carbapenémicos/farmacología , Recuento de Colonia Microbiana , Farmacorresistencia Bacteriana , Enteritis/patología , Enteritis/prevención & control , Enterococcus faecium/efectos de los fármacos , Enterococcus faecium/crecimiento & desarrollo , Enterococcus faecium/aislamiento & purificación , Trasplante de Microbiota Fecal , Heces/microbiología , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Infecciones por Bacterias Grampositivas/patología , Infecciones por Bacterias Grampositivas/prevención & control , Interacciones Huésped-Patógeno , Hibridación Fluorescente in Situ , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Infecciones por Klebsiella/patología , Infecciones por Klebsiella/prevención & control , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/crecimiento & desarrollo , Klebsiella pneumoniae/aislamiento & purificación , Ratones Endogámicos C57BL , Interacciones Microbianas , Organismos Libres de Patógenos Específicos , Enterococos Resistentes a la Vancomicina/efectos de los fármacos , Enterococos Resistentes a la Vancomicina/crecimiento & desarrollo , Enterococos Resistentes a la Vancomicina/aislamiento & purificación
8.
PLoS Pathog ; 10(2): e1003940, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24586155

RESUMEN

Aspergillus fumigatus is an environmental fungus that causes invasive aspergillosis (IA) in immunocompromised patients. Although -CC-chemokine receptor-2 (CCR2) and Ly6C-expressing inflammatory monocytes (CCR2⁺Mo) and their derivatives initiate adaptive pulmonary immune responses, their role in coordinating innate immune responses in the lung remain poorly defined. Using conditional and antibody-mediated cell ablation strategies, we found that CCR2⁺Mo and monocyte-derived dendritic cells (Mo-DCs) are essential for innate defense against inhaled conidia. By harnessing fluorescent Aspergillus reporter (FLARE) conidia that report fungal cell association and viability in vivo, we identify two mechanisms by which CCR2⁺Mo and Mo-DCs exert innate antifungal activity. First, CCR2⁺Mo and Mo-DCs condition the lung inflammatory milieu to augment neutrophil conidiacidal activity. Second, conidial uptake by CCR2⁺Mo temporally coincided with their differentiation into Mo-DCs, a process that resulted in direct conidial killing. Our findings illustrate both indirect and direct functions for CCR2⁺Mo and their derivatives in innate antifungal immunity in the lung.


Asunto(s)
Células Dendríticas/inmunología , Inmunidad Innata/inmunología , Monocitos/inmunología , Aspergilosis Pulmonar/inmunología , Animales , Aspergillus fumigatus/inmunología , Diferenciación Celular/inmunología , Células Dendríticas/citología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/citología , Monocitos/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores CCR2/metabolismo , Esporas Fúngicas/inmunología
9.
J Infect Dis ; 212(10): 1656-65, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25920320

RESUMEN

Antibiotic administration disrupts the intestinal microbiota, increasing susceptibility to pathogens such as Clostridium difficile. Metronidazole or oral vancomycin can cure C. difficile infection, and administration of these agents to prevent C. difficile infection in high-risk patients, although not sanctioned by Infectious Disease Society of America guidelines, has been considered. The relative impacts of metronidazole and vancomycin on the intestinal microbiota and colonization resistance are unknown. We investigated the effect of brief treatment with metronidazole and/or oral vancomycin on susceptibility to C. difficile, vancomycin-resistant Enterococcus, carbapenem-resistant Klebsiella pneumoniae, and Escherichia coli infection in mice. Although metronidazole resulted in transient loss of colonization resistance, oral vancomycin markedly disrupted the microbiota, leading to prolonged loss of colonization resistance to C. difficile infection and dense colonization by vancomycin-resistant Enterococcus, K. pneumoniae, and E. coli. Our results demonstrate that vancomycin, and to a lesser extent metronidazole, are associated with marked intestinal microbiota destruction and greater risk of colonization by nosocomial pathogens.


Asunto(s)
Antiinfecciosos/administración & dosificación , Infecciones Bacterianas/inmunología , Resistencia a la Enfermedad/efectos de los fármacos , Metronidazol/administración & dosificación , Vancomicina/administración & dosificación , Animales , Antiinfecciosos/efectos adversos , Infecciones Bacterianas/microbiología , Clostridioides difficile/aislamiento & purificación , Modelos Animales de Enfermedad , Escherichia coli/aislamiento & purificación , Femenino , Klebsiella pneumoniae/aislamiento & purificación , Metronidazol/efectos adversos , Ratones Endogámicos C57BL , Vancomicina/efectos adversos , Enterococos Resistentes a la Vancomicina/aislamiento & purificación
10.
Infect Immun ; 83(9): 3418-27, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26056382

RESUMEN

Klebsiella pneumoniae is a common respiratory pathogen, with some strains having developed broad resistance to clinically available antibiotics. Humans can become infected with many different K. pneumoniae strains that vary in genetic background, antibiotic susceptibility, capsule composition, and mucoid phenotype. Genome comparisons have revealed differences between K. pneumoniae strains, but the impact of genomic variability on immune-mediated clearance of pneumonia remains unclear. Experimental studies of pneumonia in mice have used the rodent-adapted 43816 strain of K. pneumoniae and demonstrated that neutrophils are essential for optimal host defense. It remains unclear, however, whether CCR2(+) monocytes contribute to K. pneumoniae clearance from the lung. We selectively depleted neutrophils, CCR2(+) monocytes, or both from immunocompetent mice and determined susceptibility to infection by the 43816 strain and 4 newly isolated clinical K. pneumoniae strains. The clinical K. pneumoniae strains, including one carbapenem-resistant ST258 strain, are less virulent than 43816. Optimal clearance of each of the 5 strains required either neutrophils or CCR2(+) monocytes. Selective neutrophil depletion markedly worsened infection with K. pneumoniae strain 43816 and three clinical isolates but did not increase susceptibility of mice to infection with the carbapenem-resistant K. pneumoniae ST258 strain. Depletion of CCR2(+) monocytes delayed recovery from infection with each of the 5 K. pneumoniae strains, revealing a contribution of these cells to bacterial clearance from the lung. Our findings demonstrate strain-dependent variation in the contributions of neutrophils and CCR2(+) monocytes to clearance of K. pneumoniae pulmonary infection.


Asunto(s)
Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/inmunología , Monocitos/inmunología , Neutrófilos/inmunología , Infecciones del Sistema Respiratorio/microbiología , Animales , Modelos Animales de Enfermedad , Infecciones por Klebsiella/inmunología , Ratones , Ratones Endogámicos C57BL , Receptores CCR2/inmunología , Infecciones del Sistema Respiratorio/inmunología
11.
Proc Natl Acad Sci U S A ; 108(1): 266-71, 2011 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-21173239

RESUMEN

CTLA-4, an Ig superfamily molecule with homology to CD28, is one of the most potent negative regulators of T-cell responses. In vivo blockade of CTLA-4 exacerbates autoimmunity, enhances tumor-specific T-cell responses, and may inhibit the induction of T-cell anergy. Clinical trials of CTLA-4-blocking antibodies to augment T-cell responses to malignant melanoma are at an advanced stage; however, little is known about the effects of CTLA-4 blockade on memory CD8(+) T-cell responses and the formation and maintenance of long-term CD8(+) T-cell memory. In our studies, we show that during in vivo memory CD8(+) T-cell responses to Listeria monocytogenes infection, CTLA-4 blockade enhances bacterial clearance and increases memory CD8(+) T-cell expansion. This is followed by an accumulation of memory cells that are capable of producing the effector cytokines IFN-γ and TNF-α. We also demonstrate that in a vaccination setting, blocking CTLA-4 during CD8(+) T-cell priming leads to increased expansion and maintenance of antigen-specific memory CD8(+) T cells without adversely affecting the overall T-cell repertoire. This leads to an increase in memory cell effector function and improved protective immunity against further bacterial challenges. These results indicate that transient blockade of CTLA-4 enhances memory CD8(+) T-cell responses and support the possible use of CTLA-4-blocking antibodies during vaccination to augment memory formation and maintenance.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antígenos CD/inmunología , Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Traslado Adoptivo , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/microbiología , Antígeno CTLA-4 , Citocinas/biosíntesis , Proteínas de Unión al ADN/genética , Citometría de Flujo , Memoria Inmunológica/efectos de los fármacos , Ratones , Ratones Noqueados
12.
PLoS Pathog ; 7(3): e1001326, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21455492

RESUMEN

Host defense against the intracellular pathogen Listeria monocytogenes (Lm) requires innate and adaptive immunity. Here, we directly imaged immune cell dynamics at Lm foci established by dendritic cells in the subcapsular red pulp (scDC) using intravital microscopy. Blood borne Lm rapidly associated with scDC. Myelomonocytic cells (MMC) swarmed around non-motile scDC forming foci from which blood flow was excluded. The depletion of scDC after foci were established resulted in a 10-fold reduction in viable Lm, while graded depletion of MMC resulted in 30-1000 fold increase in viable Lm in foci with enhanced blood flow. Effector CD8+ T cells at sites of infection displayed a two-tiered reduction in motility with antigen independent and antigen dependent components, including stable interactions with infected and non-infected scDC. Thus, swarming MMC contribute to control of Lm prior to development of T cell immunity by direct killing and sequestration from blood flow, while scDC appear to promote Lm survival while preferentially interacting with CD8+ T cells in effector sites.


Asunto(s)
Inmunidad Adaptativa , Inmunidad Innata , Listeria monocytogenes/patogenicidad , Listeriosis/patología , Linfocitos T Citotóxicos/patología , Animales , Citotoxicidad Inmunológica , Células Dendríticas/inmunología , Células Dendríticas/patología , Femenino , Técnicas de Sustitución del Gen , Interacciones Huésped-Patógeno , Listeriosis/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Monocitos/inmunología , Monocitos/patología , Linfocitos T Citotóxicos/inmunología
13.
J Immunol ; 187(10): 5293-8, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21976773

RESUMEN

Listeria monocytogenes is a facultative intracellular bacterium that causes systemic infections in immunocompromised hosts. Early recruitment of myeloid cells, including inflammatory monocytes and neutrophils, to sites of L. monocytogenes infection is essential for the control of infection and host survival. Because previous experimental studies used depleting or blocking Abs that affected both inflammatory monocytes and neutrophils, the relative contributions of these cell populations to defense against L. monocytogenes infection remain incompletely defined. In this article, we used highly selective depletion strategies to either deplete inflammatory monocytes or neutrophils from L. monocytogenes-infected mice and demonstrate that neutrophils are dispensable for early and late control of infection. In contrast, inflammatory monocytes are essential for bacterial clearance during the innate and adaptive phases of the immune response to L. monocytogenes infection.


Asunto(s)
Antígenos Ly/biosíntesis , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Neutrófilos/inmunología , Animales , Anticuerpos Bloqueadores/uso terapéutico , Antígenos Ly/inmunología , Listeriosis/patología , Listeriosis/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/patología , Neutropenia/inmunología , Neutropenia/patología , Neutropenia/prevención & control , Infiltración Neutrófila/inmunología , Neutrófilos/metabolismo , Neutrófilos/patología
14.
J Immunol ; 184(11): 6266-74, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20435926

RESUMEN

Recruitment of CCR2(+)Ly6C(high) monocytes to sites of infection is essential for efficient clearance of microbial pathogens. Although CCR2-mediated signals promote monocyte emigration from bone marrow, the contribution of CCR2 to later stages of monocyte recruitment remains unresolved. In this article, we show that CCR2 deficiency markedly worsens hepatic Listeria monocytogenes infection because Ly6C(high) monocytes are retained in the bone marrow. Intravenously transferred, CCR2-deficient Ly6C(high) monocytes traffic normally to hepatic foci of infection and contribute to bacterial clearance. Pertussis toxin treatment of adoptively transferred monocytes does not impair their intrahepatic trafficking, suggesting that chemokine signaling, once CCR2(+)Ly6C(high) monocytes emigrate from the bone marrow, is not required for monocyte localization to sites of bacterial infection in the liver. Expression of ICAM-1 is induced in close proximity to foci of bacterial infection in the liver, including on CD31(+) endothelial cells, and blockade of CD11b and CD44 diminishes monocyte localization to these hepatic foci. Our studies demonstrated that Ly6C(high) monocyte recruitment from the bloodstream to the L. monocytogenes-infected liver does not require chemokine receptor-mediated signals but instead is principally dependent on integrin- and extracellular matrix-mediated monocyte adhesion.


Asunto(s)
Quimiotaxis de Leucocito/inmunología , Molécula 1 de Adhesión Intercelular/inmunología , Listeriosis/inmunología , Monocitos/inmunología , Animales , Diferenciación Celular/inmunología , Separación Celular , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Molécula 1 de Adhesión Intercelular/metabolismo , Listeria monocytogenes/inmunología , Listeriosis/metabolismo , Hígado/inmunología , Hígado/microbiología , Ratones , Ratones Transgénicos , Monocitos/citología , Monocitos/metabolismo , Receptores CCR2/inmunología , Receptores CCR2/metabolismo
15.
J Immunol ; 183(2): 1271-8, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19553532

RESUMEN

Monocytes play a central role in defense against infection, but the mechanisms promoting monocyte recruitment and activation remain incompletely defined. Defense against Listeria monocytogenes, an intracellular bacterial pathogen, requires in vivo MCP-1 induction and CCR2-dependent recruitment of Ly6C(high) monocytes from bone marrow to sites of infection. Herein, we demonstrate that infection of bone marrow-derived macrophages with virulent L. monocytogenes induces MCP-1 expression in two phases. The first phase is rapid, induces low-level production of MCP-1, and is dependent on TLR/MyD88 signaling. The second phase promotes prolonged, higher level MCP-1 secretion and is dependent on signaling via the type I IFN receptor (IFNAR). Although attenuated L. monocytogenes strains that remain confined to the phagosome trigger TLR/MyD88-mediated signals and induce low-level MCP-1 expression, only cytosol-invasive bacteria promote IFNAR-dependent MCP-1 expression. In vivo, deficiency of either MyD88 or IFNAR signaling does not impair early monocyte emigration from bone marrow and recruitment to infected spleen. Loss of both MyD88 and IFNAR-mediated MCP-1 induction, however, results in deficient Ly6C(high) monocyte recruitment and increased susceptibility to L. monocytogenes infection. Our studies demonstrate that distinct but partially overlapping signal transduction pathways provide redundancy that ensures optimal monocyte recruitment to sites of microbial infection.


Asunto(s)
Quimiocina CCL2/biosíntesis , Quimiocinas/biosíntesis , Listeriosis/inmunología , Factor 88 de Diferenciación Mieloide/fisiología , Receptor de Interferón alfa y beta/fisiología , Animales , Células de la Médula Ósea , Quimiotaxis de Leucocito , Citosol/microbiología , Listeria monocytogenes , Macrófagos/microbiología , Ratones , Ratones Noqueados , Monocitos , Factor 88 de Diferenciación Mieloide/inmunología , Receptor de Interferón alfa y beta/inmunología , Transducción de Señal , Bazo/inmunología , Bazo/microbiología
16.
J Immunol ; 183(1): 631-41, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19542472

RESUMEN

Aspergillus fumigatus, a common environmental fungus, can cause lethal invasive infections in immunocompromised hosts. In immunocompetent individuals, however, inhaled A. fumigatus spores prime CD4(+) T cells and activate immune responses that prevent invasive infection. Calibration of inflammatory responses to levels that prevent fungal invasion without inducing collateral tissue damage is essential for host survival, but the underlying regulatory mechanisms remain undefined. Although IL-10 is a validated regulatory cytokine that suppresses immune responses, and IL-10 deficiency or blockade generally enhances immune responses, we find that A. fumigatus-specific T cell frequencies are markedly reduced in airways of IL-10-deficient mice. T cell priming, proliferation, and survival were unaffected by IL-10 deficiency and did not account for decreased frequencies of A. fumigatus-specific T cells in the airways of IL-10-deficient mice. Instead, IL-10 deficiency results in redistribution of A. fumigatus-specific T cells from infected lungs to the gut, a process that is reversed by antibiotic-mediated depletion of intestinal microbes. Our studies demonstrate that disregulated immune responses in the gut can result in dramatic redistribution of pathogen-specific T cells within the host.


Asunto(s)
Aspergilosis/inmunología , Aspergilosis/patología , Aspergillus fumigatus/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/microbiología , Epítopos de Linfocito T/inmunología , Interleucina-10/deficiencia , Animales , Aspergilosis/genética , Linfocitos T CD4-Positivos/patología , Movimiento Celular/inmunología , Células Cultivadas , Interleucina-10/genética , Intestinos/inmunología , Intestinos/microbiología , Intestinos/patología , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/microbiología , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos
17.
Nat Commun ; 11(1): 4475, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32901029

RESUMEN

Tissue resident memory CD8+ T cells (Trm) are poised for immediate reactivation at sites of pathogen entry and provide optimal protection of mucosal surfaces. The intestinal tract represents a portal of entry for many infectious agents; however, to date specific strategies to enhance Trm responses at this site are lacking. Here, we present TMDI (Transient Microbiota Depletion-boosted Immunization), an approach that leverages antibiotic treatment to temporarily restrain microbiota-mediated colonization resistance, and favor intestinal expansion to high densities of an orally-delivered Listeria monocytogenes strain carrying an antigen of choice. By augmenting the local chemotactic gradient as well as the antigenic load, this procedure generates a highly expanded pool of functional, antigen-specific intestinal Trm, ultimately enhancing protection against infectious re-challenge in mice. We propose that TMDI is a useful model to dissect the requirements for optimal Trm responses in the intestine, and also a potential platform to devise novel mucosal vaccination approaches.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Inmunidad Mucosa , Administración Oral , Animales , Antígenos/administración & dosificación , Linfocitos T CD8-positivos/inmunología , Quimiotaxis/inmunología , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Interacciones Microbiota-Huesped/inmunología , Inmunidad Mucosa/efectos de los fármacos , Memoria Inmunológica , Listeria monocytogenes/crecimiento & desarrollo , Listeria monocytogenes/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ovalbúmina/administración & dosificación , Estreptomicina/administración & dosificación
18.
Comp Med ; 70(3): 277-290, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32404234

RESUMEN

Clostridioides difficile is an enteric pathogen that can cause significant clinical disease in both humans and animals. However, clinical disease arises most commonly after treatment with broad-spectrum antibiotics. The organism's ability to cause naturally occurring disease in mice is rare, and little is known about its clinical significance in highly immunocompromised mice. We report on 2 outbreaks of diarrhea associated with C. difficile in mice. In outbreak 1, 182 of approximately 2, 400 NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ (NSG) and related strains of mice became clinically ill after cessation of a 14-d course of 0.12% amoxicillin feed to control an increase in clinical signs associated with Corynebacterium bovis infection. Most mice had been engrafted with human tumors; the remainder were experimentally naïve. Affected animals exhibited 1 of 3 clinical syndromes: 1) peracute death; 2) severe diarrhea leading to euthanasia or death; or 3) mild to moderate diarrhea followed by recovery. A given cage could contain both affected and unaffected mice. Outbreak 2 involved a small breeding colony (approximately 50 mice) of NOD. CB17-Prkdcscid/NCrCrl (NOD-scid) mice that had not received antibiotics or experimental manipulations. In both outbreaks, C. difficile was isolated, and toxins A and B were detected in intestinal content or feces. Histopathologic lesions highly suggestive of C. difficile enterotoxemia included fibrinonecrotizing and neutrophilic typhlocolitis with characteristic 'volcano' erosions or pseudomembrane formation. Genomic analysis of 4 isolates (3 from outbreak 1 and 1 from outbreak 2) revealed that these isolates were closely related to a pathogenic human isolate, CD 196. To our knowledge, this report is the first to describe naturally occurring outbreaks of C. difficile-associated typhlocolitis with significant morbidity and mortality in highly immunocompromised strains of mice.


Asunto(s)
Infecciones por Clostridium/veterinaria , Diarrea/veterinaria , Amoxicilina/administración & dosificación , Amoxicilina/efectos adversos , Animales , Antibacterianos/administración & dosificación , Antibacterianos/efectos adversos , Clostridioides difficile/aislamiento & purificación , Infecciones por Clostridium/mortalidad , Diarrea/etiología , Brotes de Enfermedades/veterinaria , Huésped Inmunocomprometido , Ratones , Ratones Endogámicos NOD , Enfermedades de los Roedores
19.
Cell Host Microbe ; 28(1): 134-146.e4, 2020 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-32492369

RESUMEN

Bacteria belonging to the Lachnospiraceae family are abundant, obligate anaerobic members of the microbiota in healthy humans. Lachnospiraceae impact their hosts by producing short-chain fatty acids, converting primary to secondary bile acids, and facilitating colonization resistance against intestinal pathogens. To increase our understanding of genomic and functional diversity between members of this family, we cultured 273 Lachnospiraceae isolates representing 11 genera and 27 species from human donors and performed whole-genome sequencing assembly and annotation. This analysis revealed substantial inter- and intra-species diversity in pathways that likely influence an isolate's ability to impact host health. These differences are likely to impact colonization resistance through lantibiotic expression or intestinal acidification, influence host mucosal immune cells and enterocytes via butyrate production, or contribute to synergism within a consortium by heterogenous polysaccharide metabolism. Identification of these specific functions could facilitate development of probiotic bacterial consortia that drive and/or restore in vivo microbiome functions.


Asunto(s)
Clostridiales/clasificación , Clostridiales/genética , Microbioma Gastrointestinal/genética , Variación Genética , Redes y Vías Metabólicas/genética , Heces/microbiología , Genoma Bacteriano , Humanos , Metagenómica , Filogenia , ARN Ribosómico 16S/genética , Secuenciación Completa del Genoma
20.
mBio ; 10(2)2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30862751

RESUMEN

A diverse, antibiotic-naive microbiota prevents highly antibiotic-resistant microbes, including carbapenem-resistant Klebsiella pneumoniae (CR-Kp), from achieving dense colonization of the intestinal lumen. Antibiotic-mediated destruction of the microbiota leads to expansion of CR-Kp in the gut, markedly increasing the risk of bacteremia in vulnerable patients. While preventing dense colonization represents a rational approach to reduce intra- and interpatient dissemination of CR-Kp, little is known about pathogen-associated factors that enable dense growth and persistence in the intestinal lumen. To identify genetic factors essential for dense colonization of the gut by CR-Kp, we constructed a highly saturated transposon mutant library with >150,000 unique mutations in an ST258 strain of CR-Kp and screened for in vitro growth and in vivo intestinal colonization in antibiotic-treated mice. Stochastic and partially reversible fluctuations in the representation of different mutations during dense colonization revealed the dynamic nature of intestinal microbial populations. We identified genes that are crucial for early and late stages of dense gut colonization and confirmed their role by testing isogenic mutants in in vivo competition assays with wild-type CR-Kp Screening of the transposon library also identified mutations that enhanced in vivo CR-Kp growth. These newly identified colonization factors may provide novel therapeutic opportunities to reduce intestinal colonization by CR-KpIMPORTANCEKlebsiella pneumoniae is a common cause of bloodstream infections in immunocompromised and hospitalized patients, and over the last 2 decades, some strains have acquired resistance to nearly all available antibiotics, including broad-spectrum carbapenems. The U.S. Centers for Disease Control and Prevention has listed carbapenem-resistant K. pneumoniae (CR-Kp) as an urgent public health threat. Dense colonization of the intestine by CR-Kp and other antibiotic-resistant bacteria is associated with an increased risk of bacteremia. Reducing the density of gut colonization by CR-Kp is likely to reduce their transmission from patient to patient in health care facilities as well as systemic infections. How CR-Kp expands and persists in the gut lumen, however, is poorly understood. Herein, we generated a highly saturated mutant library in a multidrug-resistant K. pneumoniae strain and identified genetic factors that are associated with dense gut colonization by K. pneumoniae This study sheds light on host colonization by K. pneumoniae and identifies potential colonization factors that contribute to high-density persistence of K. pneumoniae in the intestine.


Asunto(s)
Antibacterianos/administración & dosificación , Carbapenémicos/administración & dosificación , Genoma Bacteriano , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/crecimiento & desarrollo , Factores de Virulencia/genética , Resistencia betalactámica , Animales , Antibacterianos/farmacología , Carbapenémicos/farmacología , Elementos Transponibles de ADN , Modelos Animales de Enfermedad , Pruebas Genéticas , Klebsiella pneumoniae/genética , Ratones , Mutagénesis Insercional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA