Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Immunol ; 39: 511-536, 2021 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-33577348

RESUMEN

The surfaces of all living organisms and most secreted proteins share a common feature: They are glycosylated. As the outermost-facing molecules, glycans participate in nearly all immunological processes, including driving host-pathogen interactions, immunological recognition and activation, and differentiation between self and nonself through a complex array of pathways and mechanisms. These fundamental immunologic roles are further cast into sharp relief in inflammatory, autoimmune, and cancer disease states in which immune regulation goes awry. Here, we review the broad impact of glycans on the immune system and discuss the changes and clinical opportunities associated with the onset of immunologic disease.


Asunto(s)
Interacciones Huésped-Patógeno , Polisacáridos , Animales , Diferenciación Celular , Humanos
2.
Am J Respir Cell Mol Biol ; 70(6): 457-467, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38346220

RESUMEN

Sepsis is a systemic inflammatory response that requires effective macrophage metabolic functions to resolve ongoing inflammation. Previous work showed that the mechanosensitive cation channel, transient receptor potential vanilloid 4 (TRPV4), mediates macrophage phagocytosis and cytokine production in response to lung infection. Here, we show that TRPV4 regulates glycolysis in a stiffness-dependent manner by augmenting macrophage glucose uptake by GLUT1. In addition, TRPV4 is required for LPS-induced phagolysosome maturation in a GLUT1-dependent manner. In a cecal slurry mouse model of sepsis, TRPV4 regulates sepsis-induced glycolysis as measured by BAL fluid (BALF) lactate and sepsis-induced lung injury as measured by BALF total protein and lung compliance. TRPV4 is necessary for bacterial clearance in the peritoneum to limit sepsis-induced lung injury. It is interesting that BALF lactate is increased in patients with sepsis compared with healthy control participants, supporting the relevance of lung cell glycolysis to human sepsis. These data show that macrophage TRPV4 is required for glucose uptake through GLUT1 for effective phagolysosome maturation to limit sepsis-induced lung injury. Our work presents TRPV4 as a potential target to protect the lung from injury in sepsis.


Asunto(s)
Transportador de Glucosa de Tipo 1 , Glucólisis , Lesión Pulmonar , Macrófagos , Sepsis , Canales Catiónicos TRPV , Animales , Canales Catiónicos TRPV/metabolismo , Sepsis/metabolismo , Sepsis/complicaciones , Transportador de Glucosa de Tipo 1/metabolismo , Transportador de Glucosa de Tipo 1/genética , Ratones , Lesión Pulmonar/metabolismo , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Humanos , Masculino , Glucosa/metabolismo , Fagosomas/metabolismo , Líquido del Lavado Bronquioalveolar , Lipopolisacáridos/farmacología , Fagocitosis , Modelos Animales de Enfermedad , Pulmón/metabolismo , Pulmón/patología , Pulmón/inmunología
3.
Glycobiology ; 33(11): 943-953, 2023 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-37379323

RESUMEN

The IgG antibody class forms an important basis of the humoral immune response, conferring reciprocal protection from both pathogens and autoimmunity. IgG function is determined by the IgG subclass, as defined by the heavy chain, as well as the glycan composition at N297, the conserved site of N-glycosylation within the Fc domain. For example, lack of core fucose promotes increased antibody-dependent cellular cytotoxicity, whereas α2,6-linked sialylation by the enzyme ST6Gal1 helps to drive immune quiescence. Despite the immunological significance of these carbohydrates, little is known about how IgG glycan composition is regulated. We previously reported that mice with ST6Gal1-deficient B cells have unaltered IgG sialylation. Likewise, ST6Gal1 released into the plasma by hepatocytes does not significantly impact overall IgG sialylation. Since IgG and ST6Gal1 have independently been shown to exist in platelet granules, it was possible that platelet granules could serve as a B cell-extrinsic site for IgG sialylation. To address this hypothesis, we used a platelet factor 4 (Pf4)-Cre mouse to delete ST6Gal1 in megakaryocytes and platelets alone or in combination with an albumin-Cre mouse to also remove it from hepatocytes and the plasma. The resulting mouse strains were viable and had no overt pathological phenotype. We also found that despite targeted ablation of ST6Gal1, no change in IgG sialylation was apparent. Together with our prior findings, we can conclude that in mice, neither B cells, the plasma, nor platelets have a substantial role in homeostatic IgG sialylation.


Asunto(s)
Inmunoglobulina G , Factores Inmunológicos , Animales , Ratones , Linfocitos B/metabolismo , Glicosilación , Inmunoglobulina G/metabolismo , Polisacáridos , Sialiltransferasas/genética , Sialiltransferasas/metabolismo , beta-D-Galactósido alfa 2-6-Sialiltransferasa
4.
Glycobiology ; 32(2): 136-147, 2022 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-34939104

RESUMEN

Polysaccharide A (PSA) is the immunodominant capsular carbohydrate from the gram negative commensal microbe Bacteroides fragilis that has shown remarkable potency in ameliorating many rodent models of inflammatory disease by eliciting downstream suppressive CD4+ T cells. PSA is composed of a zwitterionic repeating unit that allows it to be processed by antigen presenting cells (APCs) and presented by MHCII in a glycosylation-dependent manner. While previous work has uncovered much about the interactions between MHCII and PSA, as well as the downstream T cell response, little is known about how PSA affects the phenotype of MHCII+ APCs, including macrophages. Here, we utilized an unbiased systems approach consisting of RNAseq transcriptomics, high-throughput flow cytometry, Luminex analysis and targeted validation experiments to characterize the impact of PSA-mediated stimulation of splenic MHCII+ cells. The data revealed that PSA potently elicited the upregulation of an alternatively activated M2 macrophage transcriptomic and cell surface signature. Cell-type-specific validation experiments further demonstrated that PSA-exposed bone marrow-derived macrophages (BMDMs) induced cell surface and intracellular markers associated with M2 macrophages compared with conventional peptide ovalbumin (ova)-exposed BMDMs. In contrast to macrophages, we also found that CD11c+ dendritic cells (DCs) upregulated the pro-T cell activation costimulatory molecule CD86 following PSA stimulation. Consistent with the divergent BMDM and DC changes, PSA-exposed DCs elicited an antigen-experienced T cell phenotype in co-cultures, whereas macrophages did not. These findings collectively demonstrate that the PSA-induced immune response is characterized by both T cell stimulation via presentation by DCs, and a previously unrecognized anti-inflammatory polarization of macrophages.


Asunto(s)
Células Presentadoras de Antígenos , Antígeno Prostático Específico , Animales , Antiinflamatorios/metabolismo , Células Presentadoras de Antígenos/metabolismo , Células Dendríticas , Humanos , Macrófagos , Masculino , Ratones , Ratones Endogámicos C57BL , Polisacáridos/metabolismo , Antígeno Prostático Específico/metabolismo
5.
Glycobiology ; 32(9): 803-813, 2022 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-35746897

RESUMEN

The glycosylation of immunoglobulin G (IgG) has attracted increased attention due to the impact of N-glycan modifications at N297 on IgG function, acting primarily through modulation of Fc domain conformation and Fcγ receptor-binding affinities and signaling. However, the mechanisms regulating IgG glycosylation and especially α2,6-sialylation of its N-glycan remain poorly understood. We observed previously that IgG is normally sialylated in mice with B cells lacking the sialyltransferase ST6Gal1. This supported the hypothesis that IgG may be sialylated outside of B cells, perhaps through the action of hepatocyte-released plasma ST6Gal1. Here, we demonstrate that this model is incorrect. Animals lacking hepatocyte expressed ST6Gal1 retain normal IgG α2,6-sialylation despite the lack of detectable ST6Gal1 in plasma. Moreover, we confirmed that B cells were not a redundant source of IgG sialylation. Thus, while α2,6-sialylation is lacking in IgG from mice with germline ablation of ST6Gal1, IgG α2,6-sialylation is normal in mice lacking ST6Gal1 in either hepatocytes or B cells. These results indicate that IgG α2,6-sialylation arises after release from a B cell but is not dependent on plasma-localized ST6Gal1 activity.


Asunto(s)
Inmunoglobulina G , Sialiltransferasas , Animales , Glicosilación , Inmunoglobulina G/genética , Inmunoglobulina G/metabolismo , Ratones , Polisacáridos/química , Receptores de IgG , Sialiltransferasas/genética , Sialiltransferasas/metabolismo
6.
Trends Immunol ; 39(7): 523-535, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29759949

RESUMEN

Carbohydrates, or glycans, are as integral to biology as nucleic acids and proteins. In immunology, glycans are well known to drive diverse functions ranging from glycosaminoglycan-mediated chemokine presentation and selectin-dependent leukocyte trafficking to the discrimination of self and non-self through the recognition of sialic acids by Siglec (sialic acid-binding Ig-like lectin) receptors. In recent years, a number of key immunological discoveries are driving a renewed and burgeoning appreciation for the importance of glycans. In this review, we highlight these findings which collectively help to define and refine our knowledge of the function and impact of glycans within the immune response.


Asunto(s)
Inmunidad/inmunología , Polisacáridos/inmunología , Animales , Quimiocinas/inmunología , Humanos , Leucocitos/inmunología , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/inmunología
7.
Glycoconj J ; 37(3): 395-407, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32222873

RESUMEN

Through the catalysis of α2,6-linked sialylation, the enzyme ST6Gal1 is thought to play key roles in immune cell communication and homeostasis. Of particular importance, glycans with terminal α2,6-sialic acids are known to negatively regulate B cell receptor signaling and are associated with an immunosuppressive tumor microenvironment that promotes T cell anergy, suggesting that α2,6-sialic acids are a key immune inhibitory signal. Consistent with this model, mice harboring a hepatocyte-specific ablation of ST6Gal1 (H-cKO) develop a progressive and severe non-alcoholic fatty liver disease characterized by steatohepatitis. Using this H-cKO mouse, we have further discovered that loss of hepatocyte α2,6-sialylation not only increases the inflammatory state of the local tissue microenvironment, but also systemic T cell-dependent immune responses. H-cKO mice responded normally to innate and passively induced inflammation, but showed significantly increased morbidity in T cell-dependent house dust mite-antigen (HDM)-induced asthma and myelin oligodendrocyte glycoprotein (MOG) peptide-induced experimental autoimmune encephalomyelitis (EAE). We further discovered that H-cKO mice have a profound shift toward effector/memory T cells even among unchallenged mice, and that macrophages from both the liver and spleen expressed the inhibitory and α2,6-sialic acid-specific glycan binding molecule CD22. These findings align with previously reported pro-inflammatory changes in liver macrophages, and support a model in which the liver microenvironment sets a systemic immune tone that is regulated by tissue α2,6-sialylation and mediated by liver macrophages and systemic T cells.


Asunto(s)
Hepatocitos/metabolismo , Inmunidad Innata/fisiología , Sialiltransferasas/metabolismo , Linfocitos T/inmunología , Animales , Asma/etiología , Asma/inmunología , Colitis/inducido químicamente , Colitis/inmunología , Modelos Animales de Enfermedad , Hepatocitos/inmunología , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/toxicidad , Hígado/inmunología , Pulmón/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones Noqueados , Peritonitis/inducido químicamente , Peritonitis/inmunología , Lectina 2 Similar a Ig de Unión al Ácido Siálico/metabolismo , Sialiltransferasas/genética , Tioglicolatos/toxicidad
8.
Blood ; 128(24): 2785-2796, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27758872

RESUMEN

Alveolar macrophages are lung-resident sentinel cells that develop perinatally and protect against pulmonary infection. Molecular mechanisms controlling alveolar macrophage generation have not been fully defined. Here, we show that the actin-bundling protein L-plastin (LPL) is required for the perinatal development of alveolar macrophages. Mice expressing a conditional allele of LPL (CD11c.Crepos-LPLfl/fl) exhibited significant reductions in alveolar macrophages and failed to effectively clear pulmonary pneumococcal infection, showing that immunodeficiency results from reduced alveolar macrophage numbers. We next identified the phase of alveolar macrophage development requiring LPL. In mice, fetal monocytes arrive in the lungs during a late fetal stage, maturing to alveolar macrophages through a prealveolar macrophage intermediate. LPL was required for the transition from prealveolar macrophages to mature alveolar macrophages. The transition from prealveolar macrophage to alveolar macrophage requires the upregulation of the transcription factor peroxisome proliferator-activated receptor-γ (PPAR-γ), which is induced by exposure to granulocyte-macrophage colony-stimulating factor (GM-CSF). Despite abundant lung GM-CSF and intact GM-CSF receptor signaling, PPAR-γ was not sufficiently upregulated in developing alveolar macrophages in LPL-/- pups, suggesting that precursor cells were not correctly localized to the alveoli, where GM-CSF is produced. We found that LPL supports 2 actin-based processes essential for correct localization of alveolar macrophage precursors: (1) transmigration into the alveoli, and (2) engraftment in the alveoli. We thus identify a molecular pathway governing neonatal alveolar macrophage development and show that genetic disruption of alveolar macrophage development results in immunodeficiency.


Asunto(s)
Macrófagos Alveolares/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Alveolos Pulmonares/citología , Alveolos Pulmonares/metabolismo , Animales , Animales Recién Nacidos , Antígenos CD11/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Ratones Endogámicos C57BL , Modelos Biológicos , Monocitos/metabolismo , PPAR gamma/metabolismo , Infecciones Neumocócicas/patología , Podosomas/metabolismo , Transporte de Proteínas , Regulación hacia Arriba/efectos de los fármacos
9.
Infect Immun ; 82(5): 1982-93, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24595139

RESUMEN

We report that mice deficient for the hematopoietic-specific, actin-bundling protein L-plastin (LPL) succumb rapidly to intratracheal pneumococcal infection. The increased susceptibility of LPL(-/-) mice to pulmonary pneumococcal challenge correlated with reduced numbers of alveolar macrophages, consistent with a critical role for this cell type in the immediate response to pneumococcal infection. LPL(-/-) mice demonstrated a very early clearance defect, with an almost 10-fold-higher bacterial burden in the bronchoalveolar lavage fluid 3 h following infection. Clearance of pneumococci from the alveolar space in LPL(-/-) mice was defective compared to that in Rag1(-/-) mice, which lack all B and T lymphocytes, indicating that innate immunity is defective in LPL(-/-) mice. We did not identify defects in neutrophil or monocyte recruitment or in the production of inflammatory cytokines or chemokines that would explain the early clearance defect. However, efficient alveolar macrophage regeneration following irradiation required LPL. We thus identify LPL as being key to alveolar macrophage development and essential to an effective antipneumococcal response. Further analysis of LPL(-/-) mice will illuminate critical regulators of the generation of alveolar macrophages and, thus, effective pulmonary innate immunity.


Asunto(s)
Macrófagos Alveolares/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Neumonía Neumocócica/metabolismo , Animales , Líquido del Lavado Bronquioalveolar/citología , Proliferación Celular , Femenino , Regulación de la Expresión Génica/inmunología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
10.
Sci Immunol ; 9(95): eabq1558, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38701190

RESUMEN

Steroid resistance poses a major challenge for the management of autoimmune neuroinflammation. T helper 17 (TH17) cells are widely implicated in the pathology of steroid resistance; however, the underlying mechanisms are unknown. In this study, we identified that interleukin-1 receptor (IL-1R) blockade rendered experimental autoimmune encephalomyelitis (EAE) mice sensitive to dexamethasone (Dex) treatment. Interleukin-1ß (IL-1ß) induced a signal transducer and activator of transcription 5 (STAT5)-mediated steroid-resistant transcriptional program in TH17 cells, which promoted inflammatory cytokine production and suppressed Dex-induced anti-inflammatory genes. TH17-specific deletion of STAT5 ablated the IL-1ß-induced steroid-resistant transcriptional program and rendered EAE mice sensitive to Dex treatment. IL-1ß synergized with Dex to promote the STAT5-dependent expression of CD69 and the development of central nervous system (CNS)-resident CD69+ TH17 cells. Combined IL-1R blockade and Dex treatment ablated CNS-resident TH17 cells, reduced EAE severity, and prevented relapse. CD69+ tissue-resident TH17 cells were also detected in brain lesions of patients with multiple sclerosis. These findings (i) demonstrate that IL-1ß-STAT5 signaling in TH17 cells mediates steroid resistance and (ii) identify a therapeutic strategy for reversing steroid resistance in TH17-mediated CNS autoimmunity.


Asunto(s)
Dexametasona , Encefalomielitis Autoinmune Experimental , Interleucina-1beta , Factor de Transcripción STAT5 , Células Th17 , Animales , Células Th17/inmunología , Factor de Transcripción STAT5/metabolismo , Factor de Transcripción STAT5/inmunología , Ratones , Interleucina-1beta/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Dexametasona/farmacología , Dexametasona/uso terapéutico , Ratones Endogámicos C57BL , Resistencia a Medicamentos , Transducción de Señal/inmunología , Ratones Noqueados , Enfermedades Neuroinflamatorias/inmunología , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Femenino , Humanos
11.
Immunohorizons ; 7(6): 456-466, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37314833

RESUMEN

The intestinal mucosa is continually exposed to diverse microbial and dietary Ags, requiring coordinated efforts by specialized populations of regulatory T cells (Tregs) to maintain homeostasis. Suppressive mechanisms used by intestinal Tregs include the secretion of anti-inflammatory cytokines such as IL-10 and TGF-ß. Defects in IL-10 signaling are associated with severe infantile enterocolitis in humans, and mice deficient in IL-10 or its receptors develop spontaneous colitis. To determine the requirement of Foxp3+ Treg-specific IL-10 for protection against colitis, we generated Foxp3-specific IL-10 knockout (KO) mice (IL-10 conditional KO [cKO] mice). Colonic Foxp3+ Tregs isolated from IL-10cKO mice showed impaired ex vivo suppressive function, although IL-10cKO mice maintained normal body weights and developed only mild inflammation over 30 wk of age (in contrast to severe colitis in global IL-10KO mice). Protection from colitis in IL-10cKO mice was associated with an expanded population of IL-10-producing type 1 Tregs (Tr1, CD4+Foxp3-) in the colonic lamina propria that produced more IL-10 on a per-cell basis compared with wild-type intestinal Tr1 cells. Collectively, our findings reveal a role for Tr1 cells in the gut, as they expand to fill a tolerogenic niche in conditions of suboptimal Foxp3+ Treg-mediated suppression and provide functional protection against experimental colitis.


Asunto(s)
Colitis , Linfocitos T Reguladores , Humanos , Animales , Ratones , Interleucina-10/genética , Colitis/prevención & control , Linfocitos T CD4-Positivos , Citocinas , Ratones Noqueados , Factores de Transcripción , Factores de Transcripción Forkhead/genética
12.
Cell Host Microbe ; 31(10): 1620-1638.e7, 2023 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-37776865

RESUMEN

Immunoglobulin A (IgA) is an important factor in maintaining homeostasis at mucosal surfaces, yet luminal IgA levels vary widely. Total IgA levels are thought to be driven by individual immune responses to specific microbes. Here, we found that the prebiotic, pectin oligosaccharide (pec-oligo), induced high IgA levels in the small intestine in a T cell-dependent manner. Surprisingly, this IgA-high phenotype was retained after cessation of pec-oligo treatment, and microbiome transmission either horizontally or vertically was sufficient to retain high IgA levels in the absence of pec-oligo. Interestingly, the bacterial taxa enriched in the overall pec-oligo bacterial community differed from IgA-coated microbes in this same community. Rather, a group of ethanol-resistant microbes, highly enriched for Lachnospiraceae bacterium A2, drove the IgA-high phenotype. These findings support a model of intestinal adaptive immunity in which a limited number of microbes can promote durable changes in IgA directed to many symbionts.


Asunto(s)
Intestinos , Microbiota , Ratones , Animales , Intestinos/microbiología , Intestino Delgado , Inmunoglobulina A , Bacterias , Mucosa Intestinal/microbiología
13.
J Leukoc Biol ; 112(6): 1555-1566, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35726710

RESUMEN

The degree of α2,6-linked sialylation on IgG glycans is associated with a variety of inflammatory conditions and is thought to drive IgG anti-inflammatory activity. Previous findings revealed that ablation of ß-galactoside α2,6-sialyltransferase 1 (ST6Gal1) in B cells failed to alter IgG sialylation in vivo, yet resulted in the loss of B cell surface α2,6 sialylation, suggesting divergent pathways for IgG and cell surface glycoprotein glycosylation and trafficking. Employing both B cell hybridomas and ex vivo murine B cells, we discovered that IgG was poorly sialylated by ST6Gal1 and highly core fucosylated by α1,6-fucosyltransferase 8 (Fut8) in cell culture. In contrast, cell surface glycoproteins on IgG-producing cells showed the opposite pattern by flow cytometry, with high α2,6 sialylation and low α1,6 fucosylation. Paired studies further revealed that ex vivo B cell-produced IgG carried significantly less sialylation compared with IgG isolated from the plasma of matched animals, providing evidence that IgG sialylation increases after release in vivo. Finally, confocal analyses demonstrated that IgG poorly localized to subcellular compartments rich in sialylation and ST6Gal1, and strongly to regions rich in fucosylation and Fut8. These findings support a model in which IgG subcellular trafficking diverges from the canonical secretory pathway by promoting Fut8-mediated core fucosylation and limiting exposure to and modification by ST6Gal1, providing a mechanism for why B cell-expressed ST6Gal1 is dispensable for IgG sialylation in vivo.


Asunto(s)
Linfocitos B , Sialiltransferasas , Ratones , Animales , Sialiltransferasas/metabolismo , Glicosilación , Linfocitos B/metabolismo , Fucosiltransferasas/metabolismo , Inmunoglobulina G/metabolismo
14.
Elife ; 102021 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-33617447

RESUMEN

Cells exist within complex milieus of communicating factors, such as cytokines, that combine to generate context-specific responses, yet nearly all knowledge about the function of each cytokine and the signaling propagated downstream of their recognition is based on the response to individual cytokines. Here, we found that regulatory T cells (Tregs) integrate concurrent signaling initiated by IL-2 and IL-4 to generate a response divergent from the sum of the two pathways in isolation. IL-4 stimulation of STAT6 phosphorylation was blocked by IL-2, while IL-2 and IL-4 synergized to enhance STAT5 phosphorylation, IL-10 production, and the selective proliferation of IL-10-producing Tregs, leading to increased inhibition of conventional T cell activation and the reversal of asthma and multiple sclerosis in mice. These data define a mechanism of combinatorial cytokine signaling and lay the foundation upon which to better understand the origins of cytokine pleiotropy while informing improved the clinical use of cytokines.


The immune system is essential to defend our bodies from attacks of microbial invaders. It contains many types of cells that communicate with each other through proteins called cytokines. Cytokines act as messages that can promote or suppress the immune response. Since several types of immune cells can exist within the same environment, many different signals are sent simultaneously, from which each individual cell must tease out the correct message. So far, it has been unclear how cells decode simultaneous messages, which may be key for improving therapeutics for inflammatory diseases. To investigate this, Zhou et al. studied the effect of two cytokines (IL-2 and IL-4) on a major type of suppressive immune cell called regulatory T cell or Treg. This revealed that when Tregs grown in the laboratory were simultaneously exposed to IL-2 and IL-4, the cytokine duo boosted the production of new Tregs much more than using either cytokine alone. Together, the cytokines also increased the production of another cytokine (IL-10), and over time, Treg cells producing IL-10 divided more frequently, leading to an even more robust ability to suppress overactive immune responses. Zhou et al. then validated the experiments in cells using mouse models of asthma and multiple sclerosis. Treatment with an IL-2 and IL-4 cytokine cocktail not only prevented but reversed the symptoms of the diseases to a greater extent than either cytokine alone, suggesting that this could be a significantly improved therapy for patients affected by these conditions. This study provides important insights into how cytokines can work together to substantially improve the activity of immune cells and reduce inflammation. It also introduces a promising new treatment strategy to dampen or even eliminate a range of devastating symptoms caused by inflammatory diseases.


Asunto(s)
Interleucina-2/farmacología , Interleucina-4/farmacología , Factor de Transcripción STAT5/metabolismo , Linfocitos T Reguladores/efectos de los fármacos , Animales , Asma/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Interleucina-10/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Esclerosis Múltiple/tratamiento farmacológico , Transducción de Señal
15.
PLoS One ; 14(5): e0216893, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31120919

RESUMEN

CD4+ effector/memory T cells (Tem) represent a leading edge of the adaptive immune system responsible for protecting the body from infection, cancer, and other damaging processes. However, a subset of Tem cells with low expression of CD45Rb (RbLoTem) has been shown to suppress inflammation despite their effector surface phenotype and the lack of FoxP3 expression, the canonical transcription factor found in most regulatory T cells. In this report, we show that RbLoTem cells can suppress inflammation by influencing Treg behavior. Co-culturing activated RbLoTem and Tregs induced high expression of IL-10 in vitro, and conditioned media from RbLoTem cells induced IL-10 expression in FoxP3+ Tregs in vitro and in vivo, indicating that RbLoTem cells communicate with Tregs in a cell-contact independent fashion. Transcriptomic and multi-analyte Luminex data identified both IL-2 and IL-4 as potential mediators of RbLoTem-Treg communication, and antibody-mediated neutralization of either IL-4 or CD124 (IL-4Rα) prevented IL-10 induction in Tregs. Moreover, isolated Tregs cultured with recombinant IL-2 and IL-4 strongly induced IL-10 production. Using house dust mite (HDM)-induced airway inflammation as a model, we confirmed that the in vivo suppressive activity of RbLoTem cells was lost in IL-4-ablated RbLoTem cells. These data support a model in which RbLoTem cells communicate with Tregs using a combination of IL-2 and IL-4 to induce robust expression of IL-10 and suppression of inflammation.


Asunto(s)
Asma/inmunología , Comunicación Celular/inmunología , Interleucina-10/inmunología , Interleucina-4/inmunología , Antígenos Comunes de Leucocito/inmunología , Modelos Inmunológicos , Linfocitos T Reguladores/inmunología , Animales , Asma/genética , Asma/patología , Comunicación Celular/genética , Modelos Animales de Enfermedad , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Interleucina-10/genética , Interleucina-2/genética , Interleucina-2/inmunología , Interleucina-4/genética , Antígenos Comunes de Leucocito/genética , Ratones , Ratones Noqueados , Pyroglyphidae/inmunología , Linfocitos T Reguladores/patología
16.
Vaccine ; 35(34): 4389-4395, 2017 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-28687405

RESUMEN

BACKGROUND: The epidemiology of nasopharyngeal (NP) pneumococcal carriage varies with geography and has changed in response to pneumococcal conjugate vaccine (PCV): a low prevalence (3% or less of colonizing isolates) of colonization by vaccine-type (VT) pneumococcal serotypes after PCV introduction has been reported. The primary goal of this study was to determine the VT serotype prevalence of NP pneumococcal colonization of children residing in the St. Louis, MO, USA metropolitan area following introduction of the 13-valent PCV in 2010. The secondary goal of this study was to identify characteristics associated with NP pneumococcal carriage of any serotype. METHODS: Between July 2013 and April 2016, we enrolled 397 healthy children, aged 0-17years, who required sedation for procedures or minor surgeries at St. Louis Children's Hospital. NP swabs were collected after sedation or anesthesia and cultured for pneumococcus. Vaccine records were obtained from primary care providers or from state immunization databases. Parents/guardians completed a questionnaire to provide demographics, past medical history and household characteristics. RESULTS: Of the 88 pneumococcal isolates recovered from 84 colonized subjects (21.2% of all enrolled subjects; 95% CI 17.2-25.2%), 16 were VT. Eleven isolates were serotype 19F (12.5%), four (4.5%) were 6A and one (1.1%) was 19A. Prevalence of VT among colonizing isolates was thus 18.2% (CI 10.1-26.1%) in our cohort, despite complete PCV vaccination in 87% of colonized children. Factors associated with pneumococcal colonization by any serotype included younger age and daycare attendance. CONCLUSION: Children in St. Louis exhibit a higher prevalence of VT serotypes among pneumococcal carriage isolates than has been reported in other areas in the US, demonstrating the necessity of ongoing surveillance of local epidemiology and providing evidence that serotype 19F can remain prevalent in a pediatric population despite high vaccine uptake.


Asunto(s)
Portador Sano/epidemiología , Infecciones Neumocócicas/epidemiología , Streptococcus pneumoniae/crecimiento & desarrollo , Streptococcus pneumoniae/aislamiento & purificación , Adolescente , Portador Sano/microbiología , Niño , Preescolar , Femenino , Vacuna Neumocócica Conjugada Heptavalente/administración & dosificación , Humanos , Lactante , Recién Nacido , Masculino , Pruebas de Sensibilidad Microbiana , Missouri/epidemiología , Nasofaringe/microbiología , Infecciones Neumocócicas/microbiología , Infecciones Neumocócicas/prevención & control , Vacunas Neumococicas/administración & dosificación , Vacunas Neumococicas/clasificación , Prevalencia , Estudios Seroepidemiológicos , Serogrupo , Streptococcus pneumoniae/clasificación , Streptococcus pneumoniae/inmunología , Vacunación , Vacunas Conjugadas/administración & dosificación
17.
Mol Immunol ; 78: 79-88, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27614263

RESUMEN

Elucidating the molecular regulation of macrophage migration is essential for understanding the pathophysiology of multiple human diseases, including host responses to infection and autoimmune disorders. Macrophage migration is supported by dynamic rearrangements of the actin cytoskeleton, with formation of actin-based structures such as podosomes and lamellipodia. Here we provide novel insights into the function of the actin-bundling protein l-plastin (LPL) in primary macrophages. We found that podosome stability is disrupted in primary resident peritoneal macrophages from LPL-/- mice. Live-cell imaging of F-actin using resident peritoneal macrophages from LifeACT-RFP+ mice demonstrated that loss of LPL led to decreased longevity of podosomes, without reducing the number of podosomes initiated. Additionally, macrophages from LPL-/- mice failed to elongate in response to chemotactic stimulation. These deficiencies in podosome stabilization and in macrophage elongation correlated with impaired macrophage transmigration in culture and decreased monocyte migration into murine peritoneum. Thus, we have identified a role for LPL in stabilizing long-lived podosomes and in enabling macrophage motility.


Asunto(s)
Movimiento Celular/fisiología , Macrófagos Peritoneales/metabolismo , Fosfoproteínas/metabolismo , Podosomas/metabolismo , Animales , Proteínas del Citoesqueleto , Ratones , Ratones Noqueados , Proteínas de Microfilamentos , Microscopía Confocal
18.
Int J Infect Dis ; 39: 50-52, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26327122

RESUMEN

Nasopharyngeal (NP) pneumococcal carriage predisposes children to pneumococcal infections. Defining the proportion of pneumococcal isolates that are antibiotic-resistant enables the appropriate choice of empiric therapies. The antibiogram of NP carriage isolates derived from a pediatric population following the introduction of the 13-valent pneumococcal conjugate vaccine was defined in this study.


Asunto(s)
Farmacorresistencia Bacteriana , Nasofaringe/microbiología , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/efectos de los fármacos , Streptococcus pneumoniae/aislamiento & purificación , Adolescente , Antibacterianos/farmacología , Portador Sano/microbiología , Niño , Preescolar , Humanos , Lactante , Recién Nacido , Infecciones Neumocócicas/epidemiología , Vacunas Neumococicas , Prevalencia
19.
Rapid Commun Mass Spectrom ; 18(15): 1753-60, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15282775

RESUMEN

Dexmedetomidine (Dex) (Precedex) is a novel lipophilic imidazole derivative with a high affinity for alpha-2 adrenergic receptors, which exhibits sedative, analgesic-sparing, and sympatholytic properties. The pharmacological effects and therapeutic benefits of this drug have drawn continued interest from the medical community. Here we report a liquid chromatography/tandem mass spectrometry (LC/MS/MS) method to simultaneously measure the concentrations of dexmedetomidine and its glucoronide metabolites, G-Dex-1 and G-Dex-2, in human plasma samples. A solid-phase extraction method was developed to effectively extract Dex, G-Dex-1, and G-Dex-2 from plasma matrices. An isocratic chromatographic method was developed to achieve baseline separation of G-Dex-1 and G-Dex-2. The linear dynamic range evaluated was 19.08-1908.56 pg/mL for Dex, 65.17-6518.17 pg/mL for G-Dex-1, and 29.42-2943.28 pg/mL for G-Dex-2. The linear correlation coefficient (r) ranged from 0.9944-0.9979 for Dex, from 0.9966-0.9984 for G-Dex-1, and from 0.9939-0.9966 for G-Dex-2. The intra-assay coefficient of variation (CV) was between 2.5-12.5% for Dex, between 5.2-11.0% for G-Dex-1, and between 3.5-12.1% for G-Dex-2. The inter-assay precision of QC samples give % CV ranges from 6.5-9.3% for Dex, from 7.1-10.6% for G-Dex-1, and from 8.2-10.2% for G-Dex-2. The inter-assay accuracies ranged from 102.0-109.3% for Dex, from 95.4-105.6% for G-Dex-1, and from 98.7-115.0% for G-Dex-2.


Asunto(s)
Dexmedetomidina/sangre , Dexmedetomidina/metabolismo , Glucurónidos/sangre , Glucurónidos/metabolismo , Calibración , Cromatografía Liquida , Dexmedetomidina/química , Congelación , Glucurónidos/química , Humanos , Espectrometría de Masas , Estructura Molecular , Estándares de Referencia , Sensibilidad y Especificidad , Manejo de Especímenes , Temperatura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA