Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Genes Dev ; 31(8): 774-786, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28465358

RESUMEN

Gliomas harboring mutations in isocitrate dehydrogenase 1/2 (IDH1/2) have the CpG island methylator phenotype (CIMP) and significantly longer patient survival time than wild-type IDH1/2 (wtIDH1/2) tumors. Although there are many factors underlying the differences in survival between these two tumor types, immune-related differences in cell content are potentially important contributors. In order to investigate the role of IDH mutations in immune response, we created a syngeneic pair mouse model for mutant IDH1 (muIDH1) and wtIDH1 gliomas and demonstrated that muIDH1 mice showed many molecular and clinical similarities to muIDH1 human gliomas, including a 100-fold higher concentration of 2-hydroxygluratate (2-HG), longer survival time, and higher CpG methylation compared with wtIDH1. Also, we showed that IDH1 mutations caused down-regulation of leukocyte chemotaxis, resulting in repression of the tumor-associated immune system. Given that significant infiltration of immune cells such as macrophages, microglia, monocytes, and neutrophils is linked to poor prognosis in many cancer types, these reduced immune infiltrates in muIDH1 glioma tumors may contribute in part to the differences in aggressiveness of the two glioma types.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/inmunología , Glioma/genética , Glioma/inmunología , Sistema Inmunológico/fisiopatología , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo , Animales , Neoplasias Encefálicas/enzimología , Quimiotaxis/genética , Metilación de ADN , Modelos Animales de Enfermedad , Glioma/enzimología , Humanos , Antígenos Comunes de Leucocito/metabolismo , Leucocitos/patología , Ratones , Mutación , Infiltración Neutrófila/genética , Neutrófilos/patología
2.
J Immunol ; 197(11): 4493-4503, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27799309

RESUMEN

Lung cancer, the leading cause of cancer-related deaths worldwide, is a heterogeneous disease comprising multiple histologic subtypes that harbor disparate mutational profiles. Immune-based therapies have shown initial promise in the treatment of lung cancer patients but are limited by low overall response rates. We sought to determine whether the host immune response to lung cancer is dictated, at least in part, by histologic and genetic differences, because such correlations would have important clinical ramifications. Using mouse models of lung cancer, we show that small cell lung cancer (SCLC) and lung adenocarcinoma (ADCA) exhibit unique immune cell composition of the tumor microenvironment. The total leukocyte content was markedly reduced in SCLC compared with lung ADCA, which was validated in human lung cancer specimens. We further identified key differences in immune cell content using three models of lung ADCA driven by mutations in Kras, p53, and Egfr Although Egfr-mutant cancers displayed robust myeloid cell recruitment, they failed to mount a CD8+ immune response. In contrast, Kras-mutant tumors displayed significant expansion of multiple immune cell types, including CD8+ cells, regulatory T cells, IL-17A-producing lymphocytes, and myeloid cells. A human tissue microarray annotated for KRAS and EGFR mutations validated the finding of reduced CD8+ content in human lung ADCA. Taken together, these findings establish a strong foundational knowledge of the immune cell contexture of lung ADCA and SCLC and suggest that molecular and histological traits shape the host immune response to cancer.


Asunto(s)
Adenocarcinoma/inmunología , Linfocitos T CD8-positivos/inmunología , Neoplasias Pulmonares/inmunología , Proteínas de Neoplasias/inmunología , Carcinoma Pulmonar de Células Pequeñas/inmunología , Linfocitos T Reguladores/inmunología , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Linfocitos T CD8-positivos/patología , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones , Ratones Transgénicos , Mutación , Proteínas de Neoplasias/genética , Carcinoma Pulmonar de Células Pequeñas/genética , Carcinoma Pulmonar de Células Pequeñas/patología , Linfocitos T Reguladores/patología
3.
Trends Immunol ; 33(7): 333-42, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22521509

RESUMEN

During the past 10 years, much attention has been focused towards elucidating the impact of Toll-like receptors (TLRs) in central nervous system (CNS) innate immunity. TLR signaling triggers the transcriptional activation of pro-interleukin-1ß (pro-IL-1ß) and pro-IL-18 that are processed into their active forms by the inflammasome. Recent studies have demonstrated inflammasome involvement during CNS infection, autoimmune disease, and injury. This review will address inflammasome actions within the CNS and how cooperation between TLR and inflammasome signaling may influence disease outcome. In addition, the concept of alternative inflammasome functions independent of IL-1 and IL-18 processing are considered in the context of CNS disease.


Asunto(s)
Sistema Nervioso Central/inmunología , Inflamasomas/inmunología , Receptores Toll-Like/inmunología , Animales , Autoinmunidad , Sistema Nervioso Central/metabolismo , Humanos , Transducción de Señal , Receptores Toll-Like/metabolismo
4.
J Immunol ; 190(5): 2159-68, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23365077

RESUMEN

Biofilm infections often lead to significant morbidity due to their chronicity and recalcitrance to antibiotics. We have demonstrated that methicillin-resistant Staphylococcus aureus (MRSA) biofilms can evade macrophage (MΦ) antibacterial effector mechanisms by skewing MΦs toward an alternatively activated M2 phenotype. To overcome this immune evasion, we have used two complementary approaches. In the first, a proinflammatory milieu was elicited by local administration of classically activated M1 MΦs and in the second by treatment with the C5a receptor (CD88) agonist EP67, which invokes MΦ proinflammatory activity. Early administration of M1-activated MΦs or EP67 significantly attenuated biofilm formation in a mouse model of MRSA catheter-associated infection. Several proinflammatory mediators were significantly elevated in biofilm-infected tissues from MΦ- and EP67-treated animals, revealing effective reprogramming of the biofilm environment to a proinflammatory milieu. A requirement for MΦ proinflammatory activity was demonstrated by the fact that transfer of MyD88-deficient MΦs had minimal impact on biofilm growth. Likewise, neutrophil administration had no effect on biofilm formation. Treatment of established biofilm infections with M1-activated MΦs also significantly reduced catheter-associated biofilm burdens compared with antibiotic treatment. Collectively, these results demonstrate that targeting MΦ proinflammatory activity can overcome the local immune inhibitory environment created during biofilm infections and represents a novel therapeutic strategy.


Asunto(s)
Biopelículas/efectos de los fármacos , Infecciones Relacionadas con Catéteres/prevención & control , Activación de Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Oligopéptidos/farmacología , Infecciones Estafilocócicas/prevención & control , Animales , Infecciones Relacionadas con Catéteres/inmunología , Infecciones Relacionadas con Catéteres/microbiología , Células Cultivadas , Humanos , Evasión Inmune , Inflamación/inducido químicamente , Inflamación/inmunología , Inflamación/microbiología , Activación de Macrófagos/inmunología , Macrófagos/clasificación , Macrófagos/microbiología , Macrófagos/trasplante , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Neutrófilos/inmunología , Neutrófilos/trasplante , Oligopéptidos/síntesis química , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/inmunología , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología
5.
PLoS Pathog ; 8(11): e1003033, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23209408

RESUMEN

Staphylococcus aureus is a leading cause of community-associated and nosocomial infections. Imperative to the success of S. aureus is the ability to adapt and utilize nutrients that are readily available. Genomic sequencing suggests that S. aureus has the genes required for synthesis of all twenty amino acids. However, in vitro experimentation demonstrates that staphylococci have multiple amino acid auxotrophies, including arginine. Although S. aureus possesses the highly conserved anabolic pathway that synthesizes arginine via glutamate, we demonstrate here that inactivation of ccpA facilitates the synthesis of arginine via the urea cycle utilizing proline as a substrate. Mutations within putA, rocD, arcB1, argG and argH abolished the ability of S. aureus JE2 ccpA::tetL to grow in the absence of arginine, whereas an interruption in argJBCF, arcB2, or proC had no effect. Furthermore, nuclear magnetic resonance demonstrated that JE2 ccpA::ermB produced (13)C(5) labeled arginine when grown with (13)C(5) proline. Taken together, these data support the conclusion that S. aureus synthesizes arginine from proline during growth on secondary carbon sources. Furthermore, although highly conserved in all sequenced S. aureus genomes, the arginine anabolic pathway (ArgJBCDFGH) is not functional under in vitro growth conditions. Finally, a mutation in argH attenuated virulence in a mouse kidney abscess model in comparison to wild type JE2 demonstrating the importance of arginine biosynthesis in vivo via the urea cycle. However, mutations in argB, argF, and putA did not attenuate virulence suggesting both the glutamate and proline pathways are active and they, or their pathway intermediates, can complement each other in vivo.


Asunto(s)
Absceso/metabolismo , Proteínas Bacterianas/metabolismo , Enfermedades Renales/metabolismo , Prolina/metabolismo , Infecciones Estafilocócicas/metabolismo , Staphylococcus aureus/metabolismo , Factores de Transcripción/metabolismo , Absceso/genética , Absceso/microbiología , Animales , Proteínas Bacterianas/genética , Modelos Animales de Enfermedad , Genoma Bacteriano/genética , Enfermedades Renales/genética , Enfermedades Renales/microbiología , Ratones , Mutación , Prolina/genética , Infecciones Estafilocócicas/genética , Staphylococcus aureus/genética , Factores de Transcripción/genética
6.
Infect Immun ; 81(12): 4363-76, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24042108

RESUMEN

The potent phagocytic and microbicidal activities of neutrophils and macrophages are among the first lines of defense against bacterial infections. Yet Staphylococcus aureus is often resistant to innate immune defense mechanisms, especially when organized as a biofilm. To investigate how S. aureus biofilms respond to macrophages and neutrophils, gene expression patterns were profiled using Affymetrix microarrays. The addition of macrophages to S. aureus static biofilms led to a global suppression of the biofilm transcriptome with a wide variety of genes downregulated. Notably, genes involved in metabolism, cell wall synthesis/structure, and transcription/translation/replication were among the most highly downregulated, which was most dramatic at 1 h compared to 24 h following macrophage addition to biofilms. Unexpectedly, few genes were enhanced in biofilms after macrophage challenge. Unlike coculture with macrophages, coculture of S. aureus static biofilms with neutrophils did not greatly influence the biofilm transcriptome. Collectively, these experiments demonstrate that S. aureus biofilms differentially modify their gene expression patterns depending on the leukocyte subset encountered.


Asunto(s)
Macrófagos/inmunología , Neutrófilos/inmunología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Staphylococcus aureus/metabolismo , Animales , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/genética , Biopelículas , Células Cultivadas , Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica , Inmunidad Innata , Ratones , Ratones Endogámicos C57BL , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética , Transactivadores/biosíntesis , Transactivadores/genética , Transcriptoma , Factores de Virulencia/genética
7.
J Immunol ; 186(11): 6585-96, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21525381

RESUMEN

Biofilms are complex communities of bacteria encased in a matrix composed primarily of polysaccharides, extracellular DNA, and protein. Staphylococcus aureus can form biofilm infections, which are often debilitating due to their chronicity and recalcitrance to antibiotic therapy. Currently, the immune mechanisms elicited during biofilm growth and their impact on bacterial clearance remain to be defined. We used a mouse model of catheter-associated biofilm infection to assess the functional importance of TLR2 and TLR9 in the host immune response during biofilm formation, because ligands for both receptors are present within the biofilm. Interestingly, neither TLR2 nor TLR9 impacted bacterial density or inflammatory mediator secretion during biofilm growth in vivo, suggesting that S. aureus biofilms circumvent these traditional bacterial recognition pathways. Several potential mechanisms were identified to account for biofilm evasion of innate immunity, including significant reductions in IL-1ß, TNF-α, CXCL2, and CCL2 expression during biofilm infection compared with the wound healing response elicited by sterile catheters, limited macrophage invasion into biofilms in vivo, and a skewing of the immune response away from a microbicidal phenotype as evidenced by decreases in inducible NO synthase expression concomitant with robust arginase-1 induction. Coculture studies of macrophages with S. aureus biofilms in vitro revealed that macrophages successful at biofilm invasion displayed limited phagocytosis and gene expression patterns reminiscent of alternatively activated M2 macrophages. Collectively, these findings demonstrate that S. aureus biofilms are capable of attenuating traditional host proinflammatory responses, which may explain why biofilm infections persist in an immunocompetent host.


Asunto(s)
Biopelículas , Inflamación/inmunología , Macrófagos/inmunología , Fagocitosis/inmunología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Animales , Infecciones Relacionadas con Catéteres/inmunología , Infecciones Relacionadas con Catéteres/metabolismo , Infecciones Relacionadas con Catéteres/microbiología , Citocinas/inmunología , Citocinas/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Interacciones Huésped-Patógeno/inmunología , Evasión Inmune/inmunología , Inflamación/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal , Microscopía Electrónica de Rastreo , Modelos Inmunológicos , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/fisiología , Staphylococcus aureus/ultraestructura , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/inmunología
8.
J Neurosci ; 31(17): 6277-88, 2011 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-21525267

RESUMEN

Psychosocial stress is associated with altered immune function and development of psychological disorders including anxiety and depression. Here we show that repeated social defeat in mice increased c-Fos staining in brain regions associated with fear and threat appraisal and promoted anxiety-like behavior in a ß-adrenergic receptor-dependent manner. Repeated social defeat also significantly increased the number of CD11b(+)/CD45(high)/Ly6C(high) macrophages that trafficked to the brain. In addition, several inflammatory markers were increased on the surface of microglia (CD14, CD86, and TLR4) and macrophages (CD14 and CD86) after social defeat. Repeated social defeat also increased the presence of deramified microglia in the medial amygdala, prefrontal cortex, and hippocampus. Moreover, mRNA analysis of microglia indicated that repeated social defeat increased levels of interleukin (IL)-1ß and reduced levels of glucocorticoid responsive genes [glucocorticoid-induced leucine zipper (GILZ) and FK506 binding protein-51 (FKBP51)]. The stress-dependent changes in microglia and macrophages were prevented by propranolol, a ß-adrenergic receptor antagonist. Microglia isolated from socially defeated mice and cultured ex vivo produced markedly higher levels of IL-6, tumor necrosis factor-α, and monocyte chemoattractant protein-1 after stimulation with lipopolysaccharide compared with microglia from control mice. Last, repeated social defeat increased c-Fos activation in IL-1 receptor type-1-deficient mice, but did not promote anxiety-like behavior or microglia activation in the absence of functional IL-1 receptor type-1. These findings indicate that repeated social defeat-induced anxiety-like behavior and enhanced reactivity of microglia was dependent on activation of ß-adrenergic and IL-1 receptors.


Asunto(s)
Antagonistas Adrenérgicos beta/administración & dosificación , Trastornos de Ansiedad , Microglía/efectos de los fármacos , Propranolol/administración & dosificación , Conducta Social , Estrés Psicológico/complicaciones , Análisis de Varianza , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Trastornos de Ansiedad/etiología , Trastornos de Ansiedad/patología , Trastornos de Ansiedad/prevención & control , Conducta Animal/efectos de los fármacos , Células Cultivadas , Citocinas/sangre , Relación Dosis-Respuesta a Droga , Citometría de Flujo/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Corteza Prefrontal/patología , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN Mensajero/metabolismo , Receptores de Interleucina-1/deficiencia , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/metabolismo , Receptor Toll-Like 4/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
9.
Clin Sci (Lond) ; 121(9): 367-87, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21745188

RESUMEN

The discovery of mammalian TLRs (Toll-like receptors), first identified in 1997 based on their homology with Drosophila Toll, greatly altered our understanding of how the innate immune system recognizes and responds to diverse microbial pathogens. TLRs are evolutionarily conserved type I transmembrane proteins expressed in both immune and non-immune cells, and are typified by N-terminal leucine-rich repeats and a highly conserved C-terminal domain termed the TIR [Toll/interleukin (IL)-1 receptor] domain. Upon stimulation with their cognate ligands, TLR signalling elicits the production of cytokines, enzymes and other inflammatory mediators that can have an impact on several aspects of CNS (central nervous system) homoeostasis and pathology. For example, TLR signalling plays a crucial role in initiating host defence responses during CNS microbial infection. Furthermore, TLRs are targets for many adjuvants which help shape pathogen-specific adaptive immune responses in addition to triggering innate immunity. Our knowledge of TLR expression and function in the CNS has greatly expanded over the last decade, with new data revealing that TLRs also have an impact on non-infectious CNS diseases/injury. In particular, TLRs recognize a number of endogenous molecules liberated from damaged tissues and, as such, influence inflammatory responses during tissue injury and autoimmunity. In addition, recent studies have implicated TLR involvement during neurogenesis, and learning and memory in the absence of any underlying infectious aetiology. Owing to their presence and immune-regulatory role within the brain, TLRs represent an attractive therapeutic target for numerous CNS disorders and infectious diseases. However, it is clear that TLRs can exert either beneficial or detrimental effects in the CNS, which probably depend on the context of tissue homoeostasis or pathology. Therefore any potential therapeutic manipulation of TLRs will require an understanding of the signals governing specific CNS disorders to achieve tailored therapy.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encefalopatías/metabolismo , Encéfalo/fisiología , Meningitis Bacterianas/metabolismo , Neurogénesis/fisiología , Receptores Toll-Like/metabolismo , Animales , Astrocitos/metabolismo , Encéfalo/metabolismo , Sistema Nervioso Central/metabolismo , Humanos , Inmunidad Innata , Aprendizaje , Memoria , Ratones , Microglía/metabolismo , Modelos Biológicos , Neuronas/metabolismo , Oligodendroglía/metabolismo , Transducción de Señal
10.
Brain Behav Immun ; 25(1): 46-52, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20656014

RESUMEN

Dendritic cells (DCs) sample their surrounding microenvironment and consequently send immunogenic or regulatory signals to T cells during DC/T cell interactions, shaping the primary adaptive immune response to infection. The microenvironment resulting from repeated social defeat increases DC co-stimulatory molecule expression and primes DCs for enhanced cytokine responses in vitro. In this study, we show that social disruption stress (SDR) results in the generation of immunogenic DCs, capable of conferring enhanced adaptive immunity to influenza A/PR/8/34 infection. Mice infected with influenza A/PR/8/34 virus 24 h after the adoptive transfer of DCs from SDR mice had significantly increased numbers of D(b)NP(366-74)CD8(+) T cells, increased IFN-γ and IFN-α mRNA, and decreased influenza M1 mRNA expression in the lung during the peak primary response (9 days post-infection), compared to mice that received DCs from naïve mice. These data demonstrate that repeated social defeat is a significant environmental influence on immunogenic DC activation and function.


Asunto(s)
Inmunidad Adaptativa/inmunología , Conducta Competitiva/fisiología , Células Dendríticas/inmunología , Virus de la Influenza A/inmunología , Medio Social , Animales , Antígenos Virales/inmunología , Antígenos CD11/inmunología , Linfocitos T CD8-positivos/inmunología , Recuento de Células , Epítopos/inmunología , Citometría de Flujo , Memoria Inmunológica , Interferón-alfa/biosíntesis , Interferón gamma/biosíntesis , Masculino , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Predominio Social , Estrés Psicológico/inmunología
11.
Brain Behav Immun ; 24(3): 394-402, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19903521

RESUMEN

Social disruption (SDR) is a well-characterized mouse stressor that causes changes in immune cell reactivity in response to inflammatory stimuli. In this study, we found that SDR in the absence of an immune challenge induced pulmonary inflammation and increased pulmonary myeloperoxidase activity. The percentage of neutrophils within the lungs increased 2-fold after social disruption. Monocyte accumulation in the lungs was also significantly increased. In addition, SDR increased the percentage of neutrophils that expressed CD11b, indicating that more neutrophils were in an activated state. In the lungs, we observed an increased level of the inflammatory cytokine, IL-1beta, as well as higher levels of KC/CXCL1, MIP-2/CXCL2, and MCP-1/CCL2, which are chemokines responsible for neutrophil and monocyte recruitment. Furthermore, social disruption led to increased lung expression of the adhesion molecules P-selectin, E-selectin, and ICAM-1, which localize and recruit immune cells. These data support previous findings of an inflammatory environment induced by SDR. We demonstrate that this effect also occurs in the pulmonary milieu and in the absence of an inflammatory stimulus.


Asunto(s)
Neumonía/patología , Neumonía/psicología , Medio Social , Estrés Psicológico/patología , Estrés Psicológico/psicología , Animales , Permeabilidad Capilar/fisiología , Moléculas de Adhesión Celular/metabolismo , Recuento de Células , Quimiocinas/metabolismo , Citocinas/metabolismo , Azul de Evans , Citometría de Flujo , Inmunidad Celular/fisiología , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neutrófilos/fisiología , Peroxidasa/metabolismo , Neumonía/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética
12.
Open Access Rheumatol ; 12: 21-28, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32110122

RESUMEN

PURPOSE: Repository corticotropin injection (RCI) is indicated for a number of autoimmune-mediated diseases including rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), and dermatomyositis (DM)/polymyositis (PM). To better understand the practice patterns and outcomes of RCI in patients with RA, SLE, or DM/PM, we conducted a retrospective medical record analysis. PATIENTS AND METHODS: Participating providers selected deidentified medical records of patients meeting the inclusion criteria (age ≥18 years; physician-reported diagnosis of RA, SLE, or DM/PM; initiation of treatment with RCI between 1/1/2011 and 2/15/2016; ≥3 in-office visits with same site/provider). Collected data spanned 12 months before and after the first prescription date for RCI. Analyses included patient demographics and clinical history, RCI treatment patterns, and physician's impression of change. RESULTS: Data from 54 patients with RA, 30 patients with SLE, and 8 patients with DM/PM were analyzed. The most frequently reported reasons for initiating RCI were lack of efficacy with prior treatment, acute exacerbation of disease, and use as add-on to ongoing therapy. The most common initial RCI dosing, 80 U twice weekly, was used for 84% of patients with RA, 75% with SLE, and 86% with DM/PM. The mean duration of treatment was 4.8, 6.5, and 6.8 months for RA, SLE, and DM/PM, respectively. Among the 57 patients with data on physician's impression of change with RCI, 78.1% of patients with RA, 94.7% with SLE, and 66.7% with DM/PM had a rating of "improved," and the mean time to best impression of change was 3.4, 4.3, and 3.4 months for RA, SLE, and DM/PM, respectively. CONCLUSION: This study reports the real-world patient profile, use patterns, and outcomes of patients who used RCI for the treatment of RA, SLE, and DM/PM. These data can inform appropriate use and clinical expectations when using RCI.

13.
J Neuroinflammation ; 5: 15, 2008 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-18477398

RESUMEN

BACKGROUND: Activation of the peripheral innate immune system stimulates the secretion of CNS cytokines that modulate the behavioral symptoms of sickness. Excessive production of cytokines by microglia, however, may cause long-lasting behavioral and cognitive complications. The purpose of this study was to determine if minocycline, an anti-inflammatory agent and purported microglial inhibitor, attenuates lipopolysaccharide (LPS)-induced neuroinflammation, sickness behavior, and anhedonia. METHODS: In the first set of experiments the effect of minocycline pretreatment on LPS-induced microglia activation was assessed in BV-2 microglia cell cultures. In the second study, adult (3-6 m) BALB/c mice received an intraperitoneal (i.p.) injection of vehicle or minocycline (50 mg/kg) for three consecutive days. On the third day, mice were also injected (i.p.) with saline or Escherichia coli LPS (0.33 mg/kg) and behavior (i.e., sickness and anhedonia) and markers of neuroinflammation (i.e., microglia activation and inflammatory cytokines) were determined. In the final study, adult and aged BALB/c mice were treated with the same minocycline and LPS injection regimen and markers of neuroinflammation were determined. All data were analyzed using Statistical Analysis Systems General Linear Model procedures and were subjected to one-, two-, or three-way ANOVA to determine significant main effects and interactions. RESULTS: Minocycline blocked LPS-stimulated inflammatory cytokine secretion in the BV-2 microglia-derived cell line and reduced LPS-induced Toll-like-receptor-2 (TLR2) surface expression on brain microglia. Moreover, minocycline facilitated the recovery from sickness behavior (i.e., anorexia, weight loss, and social withdrawal) and prevented anhedonia in adult mice challenged with LPS. Furthermore, the minocycline associated recovery from LPS-induced sickness behavior was paralleled by reduced mRNA levels of Interleukin (IL)-1beta, IL-6, and indoleamine 2, 3 dioxygenase (IDO) in the cortex and hippocampus. Finally, in aged mice, where exaggerated neuroinflammation was elicited by LPS, minocycline pretreatment was still effective in markedly reducing mRNA levels of IL-1beta, TLR2 and IDO in the hippocampus. CONCLUSION: These data indicate that minocycline mitigates neuroinflammation in the adult and aged brain and modulates the cytokine-associated changes in motivation and behavior.


Asunto(s)
Endotoxemia/tratamiento farmacológico , Endotoxinas/toxicidad , Interleucina-1beta/sangre , Minociclina/uso terapéutico , Trastornos del Humor/tratamiento farmacológico , Rol del Enfermo , Factores de Edad , Animales , Línea Celular , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Sacarosa en la Dieta , Evaluación Preclínica de Medicamentos , Endotoxemia/complicaciones , Endotoxemia/inmunología , Endotoxemia/patología , Endotoxemia/psicología , Conducta Exploratoria/efectos de los fármacos , Preferencias Alimentarias/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Interleucina-1beta/genética , Interleucina-6/sangre , Interleucina-6/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Microglía/efectos de los fármacos , Microglía/metabolismo , Minociclina/farmacología , Trastornos del Humor/etiología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Trastorno de la Conducta Social/etiología , Trastorno de la Conducta Social/prevención & control , Organismos Libres de Patógenos Específicos , Receptor Toll-Like 2/biosíntesis
14.
Nat Commun ; 8: 14381, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28146145

RESUMEN

The response rate to immune checkpoint inhibitor therapy for non-small-cell lung cancer (NSCLC) is just 20%. To improve this figure, several early phase clinical trials combining novel immunotherapeutics with immune checkpoint blockade have been initiated. Unfortunately, these trials have been designed without a strong foundational knowledge of the immune landscape present in NSCLC. Here, we use a flow cytometry panel capable of measuring 51 immune cell populations to comprehensively identify the immune cell composition and function in NSCLC. The results show that the immune cell composition is fundamentally different in lung adenocarcinoma as compared with lung squamous cell carcinoma, and that neutrophils are the most prevalent immune cell type. Using T-cell receptor-ß sequencing and tumour reactivity assays, we predict that tumour reactive T cells are frequently present in NSCLC. These results should help to guide the design of clinical trials and the direction of future research in this area.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/inmunología , Sistema Inmunológico/inmunología , Neoplasias Pulmonares/inmunología , Neutrófilos/inmunología , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Adenocarcinoma/terapia , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/terapia , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/terapia , Recuento de Células , Citometría de Flujo , Humanos , Sistema Inmunológico/patología , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Neutrófilos/patología , Linfocitos T/inmunología , Linfocitos T/patología
15.
mBio ; 6(4)2015 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-26307164

RESUMEN

UNLABELLED: The macrophage response to planktonic Staphylococcus aureus involves the induction of proinflammatory microbicidal activity. However, S. aureus biofilms can interfere with these responses in part by polarizing macrophages toward an anti-inflammatory profibrotic phenotype. Here we demonstrate that conditioned medium from mature S. aureus biofilms inhibited macrophage phagocytosis and induced cytotoxicity, suggesting the involvement of a secreted factor(s). Iterative testing found the active factor(s) to be proteinaceous and partially agr-dependent. Quantitative mass spectrometry identified alpha-toxin (Hla) and leukocidin AB (LukAB) as critical molecules secreted by S. aureus biofilms that inhibit murine macrophage phagocytosis and promote cytotoxicity. A role for Hla and LukAB was confirmed by using hla and lukAB mutants, and synergy between the two toxins was demonstrated with a lukAB hla double mutant and verified by complementation. Independent confirmation of the effects of Hla and LukAB on macrophage dysfunction was demonstrated by using an isogenic strain in which Hla was constitutively expressed, an Hla antibody to block toxin activity, and purified LukAB peptide. The importance of Hla and LukAB during S. aureus biofilm formation in vivo was assessed by using a murine orthopedic implant biofilm infection model in which the lukAB hla double mutant displayed significantly lower bacterial burdens and more macrophage infiltrates than each single mutant. Collectively, these findings reveal a critical synergistic role for Hla and LukAB in promoting macrophage dysfunction and facilitating S. aureus biofilm development in vivo. IMPORTANCE: Staphylococcus aureus has a propensity to form multicellular communities known as biofilms. While growing in a biofilm, S. aureus displays increased tolerance to nutrient deprivation, antibiotic insult, and even host immune challenge. Previous studies have shown that S. aureus biofilms thwart host immunity in part by preventing macrophage phagocytosis. It remained unclear whether this was influenced solely by the considerable size of biofilms or whether molecules were also actively secreted to circumvent macrophage-mediated phagocytosis. This is the first report to demonstrate that S. aureus biofilms inhibit macrophage phagocytosis and induce macrophage death through the combined action of leukocidin AB and alpha-toxin. Loss of leukocidin AB and alpha-toxin expression resulted in enhanced S. aureus biofilm clearance in a mouse model of orthopedic implant infection, suggesting that these toxins could be targeted therapeutically to facilitate biofilm clearance in humans.


Asunto(s)
Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Biopelículas , Proteínas Hemolisinas/metabolismo , Leucocidinas/metabolismo , Macrófagos/fisiología , Fagocitosis , Staphylococcus aureus/fisiología , Animales , Proteínas Bacterianas/genética , Medios de Cultivo Condicionados , Modelos Animales de Enfermedad , Humanos , Leucocidinas/genética , Macrófagos/inmunología , Ratones , Mutación , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética
16.
Methods Mol Biol ; 1106: 183-91, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24222467

RESUMEN

Biofilms are adherent communities of bacteria contained within a complex matrix. Staphylococcal species are frequent etiological agents of device-associated biofilm infections in humans that are highly recalcitrant to antimicrobial therapy and alter host immune responses to facilitate bacterial persistence. Here we describe a mouse model of catheter-associated biofilm infection, which can be utilized to investigate the importance of various staphylococcal determinants on disease progression as well as the host immune response to staphylococcal biofilms.


Asunto(s)
Biopelículas , Infecciones Relacionadas con Catéteres/microbiología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/fisiología , Staphylococcus epidermidis/fisiología , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL
17.
Methods Mol Biol ; 1106: 173-81, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24222466

RESUMEN

Animal models are invaluable tools for translational research, allowing investigators to recapitulate observed clinical scenarios within the laboratory that share attributes with human disease. Here, we describe a mouse model of post-arthroplasty Staphylococcus epidermidis joint infection which mimics human disease and may be utilized to explore the complex series of events during staphylococcal implant-associated infections by identifying key immunological, bacterial, and/or therapeutic mechanisms relevant to these persistent infections.


Asunto(s)
Biopelículas , Infecciones Relacionadas con Prótesis/microbiología , Infecciones Estafilocócicas/microbiología , Staphylococcus epidermidis/fisiología , Animales , Artroplastia , Modelos Animales de Enfermedad , Humanos , Articulación de la Rodilla/microbiología , Articulación de la Rodilla/cirugía , Prótesis de la Rodilla/microbiología , Ratones , Ratones Endogámicos C57BL , Falla de Prótesis
18.
ACS Chem Biol ; 9(9): 1997-2002, 2014 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-25061850

RESUMEN

This Letter reports a family of novel antimicrobial compounds obtained by combining peptide library screening with structure-based design. Library screening led to the identification of a human LL-37 peptide resistant to chymotrypsin. This d-amino-acid-containing peptide template was active against Escherichia coli but not methicillin-resistant Staphylococcus aureus (MRSA). It possesses a unique nonclassic amphipathic structure with hydrophobic defects. By repairing the hydrophobic defects, the peptide (17BIPHE2) gained activity against the ESKAPE pathogens, including Enterococcus faecium, S. aureus, Klebsiella pneumoniae, Acinetobacter baumanii, Pseudomonas aeruginosa, and Enterobacter species. In vitro, 17BIPHE2 could disrupt bacterial membranes and bind to DNA. In vivo, the peptide prevented staphylococcal biofilm formation in a mouse model of catheter-associated infection. Meanwhile, it boosted the innate immune response to further combat the infection. Because these peptides are potent, cell-selective, and stable to several proteases, they may be utilized to combat one or more ESKAPE pathogens.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/química , Péptidos Catiónicos Antimicrobianos/farmacología , Secuencia de Aminoácidos , Animales , Infecciones Relacionadas con Catéteres/tratamiento farmacológico , Infecciones Relacionadas con Catéteres/microbiología , Modelos Animales de Enfermedad , Diseño de Fármacos , Enterococcus faecium/efectos de los fármacos , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Masculino , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana , Datos de Secuencia Molecular , Pseudomonas aeruginosa/efectos de los fármacos , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/patogenicidad , Relación Estructura-Actividad , Catelicidinas
19.
Artículo en Inglés | MEDLINE | ID: mdl-22919653

RESUMEN

Biofilms are adherent communities of bacteria contained within a complex matrix. Although host immune responses to planktonic staphylococcal species have been relatively well-characterized, less is known regarding immunity to staphylococcal biofilms and how they modulate anti-bacterial effector mechanisms when organized in this protective milieu. Previously, staphylococcal biofilms were thought to escape immune recognition on the basis of their chronic and indolent nature. Instead, we have proposed that staphylococcal biofilms skew the host immune response away from a proinflammatory bactericidal phenotype toward an anti-inflammatory, pro-fibrotic response that favors bacterial persistence. This possibility is supported by recent studies from our laboratory using a mouse model of catheter-associated biofilm infection, where S. aureus biofilms led to the accumulation of alternatively activated M2 macrophages that exhibit anti-inflammatory and pro-fibrotic properties. In addition, relatively few neutrophils were recruited into S. aureus biofilms, representing another mechanism that deviates from planktonic infections. However, it is important to recognize the diversity of biofilm infections, in that studies by others have demonstrated the induction of distinct immune responses during staphylococcal biofilm growth in other models, suggesting influences from the local tissue microenvironment. This review will discuss the immune defenses that staphylococcal biofilms evade as well as conceptual issues that remain to be resolved. An improved understanding of why the host immune response is unable to clear biofilm infections could lead to targeted therapies to reverse these defects and expedite biofilm clearance.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Evasión Inmune , Staphylococcus aureus/inmunología , Staphylococcus aureus/fisiología , Staphylococcus epidermidis/inmunología , Staphylococcus epidermidis/fisiología , Animales , Infecciones Relacionadas con Catéteres/inmunología , Infecciones Relacionadas con Catéteres/microbiología , Modelos Animales de Enfermedad , Interacciones Huésped-Patógeno , Humanos , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/patogenicidad , Staphylococcus epidermidis/patogenicidad
20.
PLoS One ; 7(8): e42476, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22879997

RESUMEN

Bacterial biofilms represent a significant therapeutic challenge based on their ability to evade host immune and antibiotic-mediated clearance. Recent studies have implicated IL-1ß in biofilm containment, whereas Toll-like receptors (TLRs) had no effect. This is intriguing, since both the IL-1 receptor (IL-1R) and most TLRs impinge on MyD88-dependent signaling pathways, yet the role of this key adaptor in modulating the host response to biofilm growth is unknown. Therefore, we examined the course of S. aureus catheter-associated biofilm infection in MyD88 knockout (KO) mice. MyD88 KO animals displayed significantly increased bacterial burdens on catheters and surrounding tissues during early infection, which coincided with enhanced dissemination to the heart and kidney compared to wild type (WT) mice. The expression of several proinflammatory mediators, including IL-6, IFN-γ, and CXCL1 was significantly reduced in MyD88 KO mice, primarily at the later stages of infection. Interestingly, immunofluorescence staining of biofilm-infected tissues revealed increased fibrosis in MyD88 KO mice concomitant with enhanced recruitment of alternatively activated M2 macrophages. Taken in the context of previous studies with IL-1ß, TLR2, and TLR9 KO mice, the current report reveals that MyD88 signaling is a major effector pathway regulating fibrosis and macrophage polarization during biofilm formation. Together these findings represent a novel example of the divergence between TLR and MyD88 action in the context of S. aureus biofilm infection.


Asunto(s)
Biopelículas , Activación de Macrófagos/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Transducción de Señal , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/fisiología , Animales , Arginasa/metabolismo , Biopelículas/crecimiento & desarrollo , Polaridad Celular , Quimiocinas/biosíntesis , Colágeno Tipo I/metabolismo , Fibrosis , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/inmunología , Ratones , Ratones Noqueados , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA