Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
J Immunol ; 208(9): 2085-2097, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35396219

RESUMEN

Asthma is a common and ubiquitous chronic respiratory disease that is associated with airway inflammation and hyperreactivity resulting in airway obstruction. It is now accepted that asthma is controlled by a combination of host genetics and environment in a rather complex fashion; however, the link between sensing of the environment and development and exacerbation of allergic lung inflammation is unclear. Human populations expressing cosegregating D299G and T399I polymorphisms in the TLR4 gene are associated with a decreased risk for asthma in adults along with hyporesponsiveness to inhaled LPS, the TLR4 ligand. However, these data do not account for other human genetic or environmental factors. Using a novel mouse strain that expresses homologous human TLR4 polymorphisms (TLR4-single nucleotide polymorphism [SNP]), we directly tested the effect of these TLR4 polymorphisms on in vivo responses to allergens using two models of induction. We report that intact TLR4 is required for allergic inflammation when using the OVA and LPS model of induction, as cellular and pathological benchmarks were diminished in both TLR4-SNP and TLR4-deficent mice. However, in the more clinically relevant model using house dust mite extract for induction, responses were enhanced in the TLR4-SNP mice, as evidenced by greater levels of eosinophilic inflammation, Th2 cytokine production, and house dust mite-specific IgG1 production compared with wild-type mice; however, mucus production and airway hyperreactivity were not affected. These results suggest that the TLR4 polymorphic variants (genes) interact differently with the allergic stimulation (environment).


Asunto(s)
Antígenos Dermatofagoides , Asma , Eosinofilia Pulmonar , Receptor Toll-Like 4 , Alérgenos , Animales , Antígenos Dermatofagoides/inmunología , Asma/genética , Asma/patología , Inflamación , Lipopolisacáridos , Ratones , Polimorfismo de Nucleótido Simple , Pyroglyphidae , Receptor Toll-Like 4/genética
2.
Cell ; 132(2): 259-72, 2008 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-18243101

RESUMEN

Interleukin-4 and Interleukin-13 are cytokines critical to the development of T cell-mediated humoral immune responses, which are associated with allergy and asthma, and exert their actions through three different combinations of shared receptors. Here we present the crystal structures of the complete set of type I (IL-4R alpha/gamma(c)/IL-4) and type II (IL-4R alpha/IL-13R alpha1/IL-4, IL-4R alpha/IL-13R alpha1/IL-13) ternary signaling complexes. The type I complex reveals a structural basis for gamma(c)'s ability to recognize six different gamma(c)-cytokines. The two type II complexes utilize an unusual top-mounted Ig-like domain on IL-13R alpha1 for a novel mode of cytokine engagement that contributes to a reversal in the IL-4 versus IL-13 ternary complex assembly sequences, which are mediated through substantially different recognition chemistries. We also show that the type II receptor heterodimer signals with different potencies in response to IL-4 versus IL-13 and suggest that the extracellular cytokine-receptor interactions are modulating intracellular membrane-proximal signaling events.


Asunto(s)
Interleucina-13/metabolismo , Interleucina-4/metabolismo , Receptores de Citocinas/metabolismo , Receptores de Interleucina-13/metabolismo , Receptores de Interleucina-4/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Línea Celular , Línea Celular Tumoral , Cristalografía por Rayos X , Dimerización , Relación Dosis-Respuesta a Droga , Histidina/metabolismo , Humanos , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Interleucina-13/genética , Interleucina-13/aislamiento & purificación , Interleucina-13/farmacología , Interleucina-4/genética , Interleucina-4/aislamiento & purificación , Interleucina-4/farmacología , Cinética , Ligandos , Modelos Moleculares , Imitación Molecular , Datos de Secuencia Molecular , Fosforilación , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptores de Citocinas/química , Receptores de Interleucina-13/química , Receptores de Interleucina-4/química , Proteínas Recombinantes/metabolismo , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT6/metabolismo , Homología de Secuencia de Aminoácido , Transducción de Señal , Termodinámica , Tirosina/metabolismo , Difracción de Rayos X
3.
Int J Mol Sci ; 23(6)2022 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-35328445

RESUMEN

Semaphorin 4A (Sema4A) exerts a stabilizing effect on human Treg cells in PBMC and CD4+ T cell cultures by engaging Plexin B1. Sema4A deficient mice display enhanced allergic airway inflammation accompanied by fewer Treg cells, while Sema4D deficient mice displayed reduced inflammation and increased Treg cell numbers even though both Sema4 subfamily members engage Plexin B1. The main objectives of this study were: 1. To compare the in vitro effects of Sema4A and Sema4D proteins on human Treg cells; and 2. To identify function-determining residues in Sema4A critical for binding to Plexin B1 based on Sema4D homology modeling. We report here that Sema4A and Sema4D display opposite effects on human Treg cells in in vitro PBMC cultures; Sema4D inhibited the CD4+CD25+Foxp3+ cell numbers and CD25/Foxp3 expression. Sema4A and Sema4D competitively bind to Plexin B1 in vitro and hence may be doing so in vivo as well. Bayesian Partitioning with Pattern Selection (BPPS) partitioned 4505 Sema domains from diverse organisms into subgroups based on distinguishing sequence patterns that are likely responsible for functional differences. BPPS groups Sema3 and Sema4 into one family and further separates Sema4A and Sema4D into distinct subfamilies. Residues distinctive of the Sema3,4 family and of Sema4A (and by homology of Sema4D) tend to cluster around the Plexin B1 binding site. This suggests that the residues both common to and distinctive of Sema4A and Sema4D may mediate binding to Plexin B1, with subfamily residues mediating functional specificity. We mutated the Sema4A-specific residues M198 and F223 to alanine; notably, F223 in Sema4A corresponds to alanine in Sema4D. Mutant proteins were assayed for Plexin B1-binding and Treg stimulation activities. The F223A mutant was unable to stimulate Treg stability in in vitro PBMC cultures despite binding Plexin B1 with an affinity similar to the WT protein. This research is a first step in generating potent mutant Sema4A molecules with stimulatory function for Treg cells with a view to designing immunotherapeutics for asthma.


Asunto(s)
Leucocitos Mononucleares , Semaforinas/metabolismo , Alanina , Animales , Teorema de Bayes , Factores de Transcripción Forkhead/genética , Humanos , Inflamación , Leucocitos Mononucleares/metabolismo , Ratones , Proteínas del Tejido Nervioso/metabolismo
4.
Am J Physiol Lung Cell Mol Physiol ; 321(2): L416-L428, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34189964

RESUMEN

Pulmonary hypertension (PH) is a devastating disease characterized by progressive elevation of pulmonary vascular resistance, right ventricular failure, and ultimately death. We have shown previously that insulin receptor substrate 2 (IRS2), a molecule highly critical to insulin resistance and metabolism, has an anti-inflammatory role in Th2-skewed lung inflammation and pulmonary vascular remodeling. Here, we investigated the hypothesis that IRS2 has an immunomodulatory role in human and experimental PH. Expression analysis showed that IRS2 was significantly decreased in the pulmonary vasculature of patients with pulmonary arterial hypertension and in rat models of PH. In mice, genetic ablation of IRS2 enhanced the hypoxia-induced signaling pathway of Akt and Forkhead box O1 (FOXO1) in the lung tissue and increased pulmonary vascular muscularization, proliferation, and perivascular macrophage recruitment. Furthermore, mice with homozygous IRS2 gene deletion showed a significant gene dosage-dependent increase in pulmonary vascular remodeling and right ventricular hypertrophy in response to hypoxia. Functional studies with bone marrow-derived macrophages isolated from homozygous IRS2 gene-deleted mice showed that hypoxia exposure led to enhancement of the Akt and ERK signaling pathway followed by increases in the pro-PH macrophage activation markers, vascular endothelial growth factor-A and arginase 1. Our data suggest that IRS2 contributes to anti-inflammatory effects by regulating macrophage activation and recruitment, which may limit the vascular inflammation, remodeling, and right ventricular hypertrophy that are seen in PH pathology. Restoring the IRS2 pathway may be an effective therapeutic approach for the treatment of PH and right heart failure.


Asunto(s)
Hipertensión Pulmonar/metabolismo , Hipoxia/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Remodelación Vascular , Animales , Modelos Animales de Enfermedad , Femenino , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Humanos , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/patología , Hipoxia/genética , Hipoxia/patología , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Proteínas Sustrato del Receptor de Insulina/genética , Masculino , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Desnudas
5.
Mol Med ; 27(1): 162, 2021 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-34961486

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a novel type b coronavirus responsible for the COVID-19 pandemic. With over 224 million confirmed infections with this virus and more than 4.6 million people dead because of it, it is critically important to define the immunological processes occurring in the human response to this virus and pathogenetic mechanisms of its deadly manifestation. This perspective focuses on the contribution of the recently discovered interaction of SARS-CoV-2 Spike protein with neuropilin 1 (NRP1) receptor, NRP1 as a virus entry receptor for SARS-CoV-2, its role in different physiologic and pathologic conditions, and the potential to target the Spike-NRP1 interaction to combat virus infectivity and severe disease manifestations.


Asunto(s)
Antivirales/farmacología , Tratamiento Farmacológico de COVID-19 , Neuropilina-1/química , Neuropilina-1/metabolismo , Enzima Convertidora de Angiotensina 2/química , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/epidemiología , COVID-19/etiología , Comorbilidad , Diabetes Mellitus/epidemiología , Diabetes Mellitus/virología , Femenino , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/inmunología , Humanos , Lactante , Terapia Molecular Dirigida/métodos , Neuropilina-1/inmunología , Embarazo , Complicaciones Infecciosas del Embarazo/tratamiento farmacológico , Complicaciones Infecciosas del Embarazo/virología , SARS-CoV-2/patogenicidad , Glicoproteína de la Espiga del Coronavirus/metabolismo
6.
Cytokine ; 113: 256-264, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30033139

RESUMEN

Psychological stress is known to have profound effects on immune function and to promote inflammatory conditions. Elevated circulating levels of cytokines associated with stress are known to increase the risk to several diseases, but little is known about this mechanism. This study assessed the role of T cells on cytokine levels after exposure to stress in the learned helplessness paradigm. Adoptive transfer of CD4+ T cells into Rag2-/- mice did not change cytokine levels to stress while CD8+ T cells resulted in an increase in TNF-α, IL-6 and IFN-γ in stressed Rag2-/- mice. Moreover, depletion of CD8+ T cells in WT mice abolished these cytokine responses to stress. Corticosterone and behavioral stress responsiveness was impaired in Rag2-/- mice reconstituted with CD8+ T cells. Notably, depletion of these cells in WT mice had no effect on behavior or corticosterone levels. Exposure to stress did not change the expression of canonical markers of T cell activation including CD62L and CD44 or modified intracellular cytokine content, suggesting that they are not the main producers of circulating cytokines in response to stress. These results show that CD8+ T cells promote TNF-α, IL-6 and IFN-γ responses to stress, possibly by stimulating non-lymphoid cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Citocinas/inmunología , Activación de Linfocitos , Estrés Psicológico/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Citocinas/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/inmunología , Receptores de Hialuranos/genética , Receptores de Hialuranos/inmunología , Selectina L/genética , Selectina L/inmunología , Ratones , Ratones Noqueados , Estrés Psicológico/genética , Estrés Psicológico/patología
7.
J Neurooncol ; 140(3): 497-507, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30132163

RESUMEN

INTRODUCTION: Emerging evidence suggests that effective treatment of glioblastoma (GBM), the most common and deadly form of adult primary brain cancer, will likely require concurrent treatment of multiple aspects of tumor pathobiology to overcome tumor heterogeneity and the complex tumor-supporting microenvironment. Recent studies in non-central nervous system (CNS) tumor cells have demonstrated that oxaliplatin (OXA) can induce multi-faceted anti-tumor effects, in particular at drug concentrations below those required to induce apoptosis. These findings motivated re-investigation of OXA for the treatment of GBM. METHODS: The effects of OXA on murine KR158 and GL261 glioma cells including cell growth, cell death, inhibition of signal transducer and activator of transcription (STAT) activity, O-6-methylguanine-DNA methyltransferase (MGMT) expression, and immunogenic cell death (ICD) initiation, were evaluated by cytotoxicity assays, Western blot analysis, STAT3-luciferase reporter assays, qRT-PCR assays, and flow cytometry. Chemical inhibitors of endoplasmic reticulum (ER) stress were used to investigate the contribution of this cell damage response to the observed OXA effects. The effect of OXA on bone marrow-derived macrophages (BMDM) exposed to glioma conditioned media (GCM) was also analyzed by Western blot analysis. RESULTS: We identified the OXA concentration threshold for induction of apoptosis and from this determined the drug dose and treatment period for sub-cytotoxic treatments of glioma cells. Under these experimental conditions, OXA reduced STAT3 activity, reduced MGMT levels and increased temozolomide sensitivity. In addition, there was evidence of immunogenic cell death (elevated EIF2α phosphorylation and calreticulin exposure) following prolonged OXA treatment. Notably, inhibition of ER stress reversed the OXA-mediated inhibition of STAT3 activity and MGMT expression in the tumor cells. In BMDMs exposed to GCM, OXA also reduced levels of phosphorylated STAT3 and decreased expression of Arginase 1, an enzyme known to contribute to pro-tumor functions in the tumor-immune environment. CONCLUSIONS: OXA can induce notable multi-faceted biological effects in glioma cells and BMDMs at relatively low drug concentrations. These findings may have significant therapeutic relevance against GBM and warrant further investigation.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Macrófagos/metabolismo , Oxaliplatino/farmacología , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Línea Celular Tumoral , Estrés del Retículo Endoplásmico , Glioma/tratamiento farmacológico , Humanos , Macrófagos/efectos de los fármacos , Ratones , Factor de Transcripción STAT3/metabolismo , Temozolomida
8.
Brain Behav Immun ; 57: 161-172, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27013354

RESUMEN

The concept of the brain as an immune privileged organ is rapidly evolving in light of new findings outlining the sophisticated relationship between the central nervous and the immune systems. The role of T cells in brain development and function, as well as modulation of behavior has been demonstrated by an increasing number of studies. Moreover, recent studies have redefined the existence of a brain lymphatic system and the presence of T cells in specific brain structures, such as the meninges and choroid plexus. Nevertheless, much information is needed to further the understanding of brain T cells and their relationship with the central nervous system under non-inflammatory conditions. In the present study we employed the Rag2(-/-) mouse model of lymphocyte deficiency and reconstitution by adoptive transfer to study the temporal and anatomical expansion of T cells in the brain under homeostatic conditions. Lymphopenic Rag2(-/-) mice were reconstituted with 10 million lymphoid cells and studied at one, two and four weeks after transfer. Moreover, lymphoid cells and purified CD4(+) and CD8(+) T cells from transgenic GFP expressing mice were used to define the neuroanatomical localization of transferred cells. T cell numbers were very low in the brain of reconstituted mice up to one week after transfer and significantly increased by 2weeks, reaching wild type values at 4weeks after transfer. CD4(+) T cells were the most abundant lymphocyte subtype found in the brain followed by CD8(+) T cells and lastly B cells. Furthermore, proliferation studies showed that CD4(+) T cells expand more rapidly than CD8(+) T cells. Lymphoid cells localize abundantly in meningeal structures, choroid plexus, and circumventricular organs. Lymphocytes were also found in vascular and perivascular spaces and in the brain parenchyma across several regions of the brain, in particular in structures rich in white matter content. These results provide proof of concept that the brain meningeal system, as well as vascular and perivascular spaces, are homing sites of lymphocytes and suggest the possibility of a brain specific T cell subtype.


Asunto(s)
Encéfalo/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Linfopenia/inmunología , Meninges/inmunología , Traslado Adoptivo , Animales , Plexo Coroideo/inmunología , Proteínas de Unión al ADN , Modelos Animales de Enfermedad , Femenino , Homeostasis/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
9.
J Immunol ; 190(9): 4763-72, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23543762

RESUMEN

Endotoxin tolerance is a complex phenomenon characterized primarily by decreased production of proinflammatory cytokines, chemokines, and other inflammatory mediators, whereas the expression of other genes are induced or unchanged. Endotoxin tolerance is induced by prior exposure of murine macrophages/human monocytes, experimental animals, or people to TLR ligands. Although recent studies reported a possible relationship between endotoxin tolerance and differentiation of alternatively activated macrophages (AA-MΦs or M2), we show in this study that LPS pretreatment of IL-4Rα(-/-) and STAT6(-/-) macrophages, which fail to develop into AA-MΦs, resulted in tolerance of proinflammatory cytokines, as well as molecules and chemokines previously associated with AA-MΦs (e.g., arginase-1, mannose receptor, CCL2, CCL17, and CCL22). In contrast to LPS, wild-type (WT) MΦs pretreated with IL-4, the prototype inducer of AA-MΦs, did not induce endotoxin tolerance with respect to proinflammatory cytokines, AA-MΦ-associated chemokines, negative regulators, NF-κB binding and subunit composition, and MAPKs; conversely, IL-13(-/-) macrophages were tolerized equivalently to WT MΦs by LPS pretreatment. Further, IL-4Rα deficiency did not affect the reversal of endotoxin tolerance exerted by the histone deacetylase inhibitor trichostatin A. Like WT mice, 100% of LPS-tolerized IL-4Rα-deficient mice survived LPS + d-galactosamine-induced lethal toxicity and exhibited decreased serum levels of proinflammatory cytokines and AA-MΦ-associated chemokines induced by LPS challenge compared with nontolerized mice. These data indicate that the signaling pathways leading to endotoxin tolerance and differentiation of AA-MΦs are dissociable.


Asunto(s)
Diferenciación Celular/inmunología , Endotoxinas/inmunología , Tolerancia Inmunológica/inmunología , Macrófagos/inmunología , Animales , Diferenciación Celular/genética , Línea Celular , Endotoxinas/metabolismo , Tolerancia Inmunológica/genética , Interleucina-13/genética , Interleucina-13/inmunología , Interleucina-13/metabolismo , Lipopolisacáridos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/inmunología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , FN-kappa B/genética , FN-kappa B/inmunología , FN-kappa B/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/inmunología , Receptores de Superficie Celular/metabolismo , Factor de Transcripción STAT6/genética , Factor de Transcripción STAT6/inmunología , Factor de Transcripción STAT6/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Transcriptoma/genética , Transcriptoma/inmunología
10.
J Immunol ; 191(4): 1517-28, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23825312

RESUMEN

STAT6 plays a central role in IL-4-mediated allergic responses. Several studies indicate that regulatory T cells (Tregs) can be modulated by IL-4 in vitro. We previously showed that STAT6(-/-) mice are highly resistant to allergic lung inflammation even when wild-type Th2 effectors were provided and that they have increased numbers of Tregs. However, the role of STAT6 in modulating Tregs in vivo during allergic lung inflammation has not been thoroughly investigated. To examine Treg and STAT6 interaction during allergic inflammation, STAT6(-/-), STAT6xRAG2(-/-), and RAG2(-/-) mice were subjected to OVA sensitization and challenge following adoptive transfer of OVA-specific, wild-type Th2 effectors with or without prior Treg depletion/inactivation, using anti-CD25 (PC61). As expected, STAT6(-/-) mice were highly resistant to airway inflammation and remodeling. In contrast, allergic lung inflammation was partially restored in STAT6(-/-) mice treated with PC61 to levels observed in STAT6xRAG2(-/-) mice. In some cases, STAT6xRAG2(-/-) mice were also given natural Tregs along with Th2 effectors. Adoptive transfer of natural Tregs caused a substantial reduction in bronchoalveolar lavage eosinophil composition and suppressed airway remodeling and T cell migration into the lung in STAT6xRAG2(-/-) mice to levels comparable to those in STAT6(-/-) mice. These results demonstrate the STAT6-dependent suppression of Tregs in vivo to promote allergic airway inflammation.


Asunto(s)
Eosinofilia Pulmonar/inmunología , Factor de Transcripción STAT6/fisiología , Linfocitos T Reguladores/inmunología , Administración Intranasal , Traslado Adoptivo , Remodelación de las Vías Aéreas (Respiratorias) , Alérgenos/administración & dosificación , Alérgenos/toxicidad , Animales , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Proteínas de Unión al ADN/deficiencia , Factores de Transcripción Forkhead/análisis , Tolerancia Inmunológica , Subunidad alfa del Receptor de Interleucina-2/análisis , Interleucina-5/análisis , Pulmón/inmunología , Pulmón/patología , Recuento de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos , Ovalbúmina/administración & dosificación , Ovalbúmina/toxicidad , Eosinofilia Pulmonar/etiología , Eosinofilia Pulmonar/patología , Factor de Transcripción STAT6/deficiencia , Factor de Transcripción STAT6/genética , Linfocitos T Reguladores/clasificación , Linfocitos T Reguladores/trasplante , Células Th2/inmunología
11.
Brain Behav Immun ; 38: 192-201, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24524915

RESUMEN

Significant evidence suggests that exposure to traumatic and/or acute stress in both mice and humans results in compromised immune function that in turn may affect associated brain processes. Additionally, recent studies in mouse models of immune deficiency have suggested that adaptive immunity may play a role during traumatic stress exposure and that impairments in lymphocyte function may contribute to increased susceptibility to various psychogenic stressors. However, rodent studies on the relationship between maladaptive stress responses and lymphocyte deficiency have been complicated by the fact that genetic manipulations in these models may also result in changes in CNS function due to the expression of targeted genes in tissues other than lymphocytes, including the brain. To address these issues we utilized mice with a deletion of recombination-activating gene 2 (Rag2), which has no confirmed expression in the CNS; thus, its loss should result in the absence of mature lymphocytes without altering CNS function directly. Stress responsiveness of immune deficient Rag2(-/-) mice on a BALB/c background was evaluated in three different paradigms: predator odor exposure (POE), fear conditioning (FC) and learned helplessness (LH). These models are often used to study different aspects of stress responsiveness after the exposure to an acute stressor. In addition, immunoblot analysis was used to assess hippocampal BDNF expression under both stressed and non-stressed conditions. Subsequent to POE, Rag2(-/-) mice exhibited a reduced acoustic startle response compared to BALB/c mice; no significant differences in behavior were observed in either FC or LH. Furthermore, analysis of hippocampal BDNF indicated that Rag2(-/-) mice have elevated levels of the mature form of BDNF compared to BALB/c mice. Results from our studies suggest that the absence of mature lymphocytes is associated with increased resilience to stress exposure in the POE and does not affect behavioral responses in the FC and LH paradigms. These findings indicate that lymphocytes play a specific role in stress responsiveness dependent upon the type, nature and intensity of the stressor.


Asunto(s)
Ansiedad/inmunología , Miedo/fisiología , Estrés Psicológico/inmunología , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Condicionamiento Psicológico , Proteínas de Unión al ADN/genética , Hipocampo/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Actividad Motora , Reflejo de Sobresalto
12.
Drug Deliv Transl Res ; 14(10): 2930-2944, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38862755

RESUMEN

Allergic disease is a major global health concern that imposes significant life-altering and economic burdens on affected individuals. However, there is still no cure. Polymer-based nanoparticles (NP) have shown the potential to induce antigen (Ag)-specific immune tolerance in various Th1/17 and Th2-mediated immune disorders including autoimmunity and allergy. Common methods by which Ags are associated with NPs are through surface conjugation or encapsulation. However, these Ag delivery strategies can be associated with several caveats that dampen their effectiveness such as uncontrolled Ag loading, a high Ag burst release, and an increased immune recognition profile. We previously developed Ag-polymer conjugate NPs (acNPs) to overcome those noted limitations, while allowing for controlled delivery of precise quantities of Ag to innate immune cells for Ag-specific CD4 T cell modulation. Here, we utilized ovalbumin (OVA) protein-poly(lactic-co-glycolic acid) (PLGA) conjugate NPs (acNP-OVA) to elucidate the impact of Ag loading on the induction of Th2 tolerance using a prophylactic and therapeutic OVA/ALUM-induced mouse model of allergic lung inflammation (ALI) in comparison to Ag-encapsulated PLGA NPs (NP(Ag)). We demonstrate that acNP-OVA formulations reduced OVA-specific IgE and inhibited Th2 cytokine secretions in an Ag loading-dependent manner when administered prophylactically. Administration of acNP-OVA to pre-sensitized mice did not affect OVA-specific IgE and Th2 cytokines tended to be reduced, however, there was no clear Ag loading dependency. acNP-OVA with medium-to-low Ag loadings were well tolerated, while formulations with high Ag loadings, including NP(Ag) resulted in anaphylaxis. Overall, our results clarify the relationship between Ag loading and Ag-specific IgE and Th2 cytokine responses in a murine model of ALI, which provides insight useful for future design of tolerogenic NP-based immunotherapies.


Asunto(s)
Antígenos , Tolerancia Inmunológica , Nanopartículas , Ovalbúmina , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Células Th2 , Animales , Células Th2/inmunología , Nanopartículas/administración & dosificación , Nanopartículas/química , Ovalbúmina/inmunología , Ovalbúmina/administración & dosificación , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Tolerancia Inmunológica/efectos de los fármacos , Antígenos/administración & dosificación , Antígenos/inmunología , Ratones , Neumonía/inmunología , Neumonía/prevención & control , Femenino , Ratones Endogámicos BALB C , Hipersensibilidad/inmunología , Citocinas/inmunología , Modelos Animales de Enfermedad
13.
J Biol Chem ; 287(38): 31948-61, 2012 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-22829596

RESUMEN

Previously, we demonstrated that the γC subunit of type I IL-4 receptor was required for robust tyrosine phosphorylation of the downstream adapter protein, IRS-2, correlating with the expression of genes (ArgI, Retnla, and Chi3l3) characteristic of alternatively activated macrophages. We located an I4R-like motif (IRS-2 docking sequence) in the γC cytoplasmic domain but not in the IL-13Rα1. Thus, we predicted that the γC tail directed enhanced IRS-2 phosphorylation. To test this, IL-4 signaling responses were examined in a mutant of the key I4R motif tyrosine residue (Y325F) and different γC truncation mutants (γ285, γ308, γ318, γ323, and γFULL LENGTH (FL)) co-expressed in L-cells or CHO cells with wild-type (WT) IL-4Rα. Surprisingly, IRS-1 phosphorylation was not diminished in Y325F L-cell mutants suggesting Tyr-325 was not required for the robust insulin receptor substrate response. IRS-2, STAT6, and JAK3 phosphorylation was observed in CHO cells expressing γ323 and γFL but not in γ318 and γ285 mutants. In addition, when CHO cells expressed γ318, γ323, or γFL with IL-2Rß, IL-2 induced phospho-STAT5 only in the γ323 and γFL clones. Our data suggest that a smaller (5 amino acid) interval than previously determined is necessary for JAK3 activation/γC-mediated signaling in response to IL-4 and IL-2. Chimeric receptor chains of the γC tail fused to the IL-13Rα1 extracellular and transmembrane domain did not elicit robust IRS-2 phosphorylation in response to IL-13 suggesting that the extracellular/transmembrane domains of the IL-4/IL-13 receptor, not the cytoplasmic domains, control signaling efficiency. Understanding this pathway fully will lead to rational drug design for allergic disease.


Asunto(s)
Subunidad gamma Común de Receptores de Interleucina/química , Subunidad alfa1 del Receptor de Interleucina-13/química , Interleucina-4/química , Animales , Células CHO , Línea Celular , Cricetinae , Cricetulus , Citocinas/metabolismo , Citoplasma/metabolismo , Citometría de Flujo , Interleucina-13/química , Ratones , Modelos Biológicos , Mutación , Fosforilación , Estructura Terciaria de Proteína , Receptores de IgE/metabolismo , Receptores de Interleucina-4/metabolismo , Factor de Transcripción STAT6/metabolismo , Transducción de Señal
14.
J Immunol ; 187(4): 1797-806, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21734075

RESUMEN

NF-κB activation is essential for receptor activator for NF-κB ligand (RANKL)-induced osteoclast formation. IL-4 is known to inhibit the RANKL-induced osteoclast differentiation while at the same time promoting macrophage fusion to form multinucleated giant cells (MNG). Several groups have proposed that IL-4 inhibition of osteoclastogenesis is mediated by suppressing the RANKL-induced activation of NF-κB. However, we found that IL-4 did not block proximal, canonical NF-κB signaling. Instead, we found that IL-4 inhibited alternative NF-κB signaling and induced p105/50 expression. Interestingly, in nfκb1(-/-) bone marrow-derived macrophages (BMM), the formation of both multinucleated osteoclast and MNG induced by RANKL or IL-4, respectively, was impaired. This suggests that NF-κB signaling also plays an important role in IL-4-induced macrophage fusion. Indeed, we found that the RANKL-induced and IL-4-induced macrophage fusion were both inhibited by the NF-κB inhibitors IκB kinase 2 inhibitor and NF-κB essential modulator inhibitory peptide. Furthermore, overexpression of p50, p65, p52, and RelB individually in nfκb1(-/-) or nfκb1(+/+) BMM enhanced both giant osteoclast and MNG formation. Interestingly, knockdown of nfκb2 in wild-type BMM dramatically enhanced both osteoclast and MNG formation. In addition, both RANKL- and IL-4-induced macrophage fusion were impaired in NF-κB-inducing kinase(-/-) BMM. These results suggest IL-4 influences NF-κB pathways by increasing p105/p50 and suppressing RANKL-induced p52 translocation and that NF-κB pathways participate in both RANKL- and IL-4-induced giant cell formation.


Asunto(s)
Células de la Médula Ósea/inmunología , Células Gigantes/inmunología , Interleucina-4/inmunología , FN-kappa B/inmunología , Osteoclastos/inmunología , Ligando RANK/inmunología , Animales , Fusión Celular , Células Cultivadas , Interleucina-4/genética , Ratones , Ratones Noqueados , FN-kappa B/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Ligando RANK/genética , Quinasa de Factor Nuclear kappa B
15.
J Immunol ; 187(1): 133-40, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21642544

RESUMEN

Memory T cells are distinguished from naive T cells by their rapid production of effector cytokines, although mechanisms for this recall response remain undefined. In this study, we investigated transcriptional mechanisms for rapid IFN-γ production by Ag-specific memory CD4 T cells. In naive CD4 T cells, IFN-γ production only occurred after sustained Ag activation and was associated with high expression of the T-bet transcription factor required for Th1 differentiation and with T-bet binding to the IFN-γ promoter as assessed by chromatin immunoprecipitation analysis. By contrast, immediate IFN-γ production by Ag-stimulated memory CD4 T cells occurred in the absence of significant nuclear T-bet expression or T-bet engagement on the IFN-γ promoter. We identified rapid induction of NF-κB transcriptional activity and increased engagement of NF-κB on the IFN-γ promoter at rapid times after TCR stimulation of memory compared with naive CD4 T cells. Moreover, pharmacologic inhibition of NF-κB activity or peptide-mediated inhibition of NF-κB p50 translocation abrogated early memory T cell signaling and TCR-mediated effector function. Our results reveal a molecular mechanism for memory T cell recall through enhanced NF-κB p50 activation and promoter engagement, with important implications for memory T cell modulation in vaccines, autoimmunity, and transplantation.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Memoria Inmunológica , Activación de Linfocitos/inmunología , Transcripción Genética/inmunología , Secuencia de Aminoácidos , Animales , Linfocitos T CD4-Positivos/citología , Células Cultivadas , Densitometría , Memoria Inmunológica/genética , Interferón gamma/biosíntesis , Interferón gamma/metabolismo , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos , Datos de Secuencia Molecular , Subunidad p50 de NF-kappa B/biosíntesis , Subunidad p50 de NF-kappa B/genética , Subunidad p50 de NF-kappa B/fisiología , Ovalbúmina/inmunología , Ovalbúmina/farmacocinética , Ovalbúmina/fisiología , Fragmentos de Péptidos/fisiología , Regiones Promotoras Genéticas/inmunología , Fase de Descanso del Ciclo Celular/genética , Fase de Descanso del Ciclo Celular/inmunología , Proteínas de Dominio T Box/biosíntesis , Proteínas de Dominio T Box/genética , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
16.
J Immunol ; 186(4): 2571-83, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21242523

RESUMEN

Th2 cells induce asthma through the secretion of cytokines. Two such cytokines, IL-4 and IL-13, are critical mediators of many features of this disease. They both share a common receptor subunit, IL-4Rα, and signal through the STAT6 pathway. STAT6(-/-) mice have impaired Th2 differentiation and reduced airway response to allergen. Transferred Th2 cells were not able to elicit eosinophilia in response to OVA in STAT6(-/-) mice. To clarify the role of STAT6 in allergic airway inflammation, we generated mouse bone marrow (BM) chimeras. We observed little to no eosinophilia in OVA-treated STAT6(-/-) mice even when STAT6(+/+) BM or Th2 cells were provided. However, when Th2 cells were transferred to STAT6×Rag2(-/-) mice, we observed an eosinophilic response to OVA. Nevertheless, the expression of STAT6 on either BM-derived cells or lung resident cells enhanced the severity of OVA-induced eosinophilia. Moreover, when both the BM donor and recipient lacked lymphocytes, transferred Th2 cells were sufficient to induce the level of eosinophilia comparable with that of wild-type (WT) mice. The expression of STAT6 in BM-derived cells was more critical for the enhanced eosinophilic response. Furthermore, we found a significantly higher number of CD4(+)CD25(+)Foxp3(+) T cells (regulatory T cells [Tregs]) in PBS- and OVA-treated STAT6(-/-) mouse lungs compared with that in WT animals suggesting that STAT6 limits both naturally occurring and Ag-induced Tregs. Tregs obtained from either WT or STAT6(-/-) mice were equally efficient in suppressing CD4(+) T cell proliferation in vitro. Taken together, our studies demonstrate multiple STAT6-dependent and -independent features of allergic inflammation, which may impact treatments targeting STAT6.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Ovalbúmina/inmunología , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/patología , Factor de Transcripción STAT6/fisiología , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/patología , Células Cultivadas , Técnicas de Cocultivo , Inmunofenotipificación , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Pulmón/inmunología , Pulmón/patología , Cooperación Linfocítica/genética , Cooperación Linfocítica/inmunología , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos , Ovalbúmina/administración & dosificación , Hipersensibilidad Respiratoria/genética , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Factor de Transcripción STAT6/deficiencia , Factor de Transcripción STAT6/genética , Bazo/inmunología , Bazo/patología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , Células Th2/inmunología , Células Th2/patología , Células Th2/trasplante
17.
J Immunol ; 187(1): 501-9, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21632710

RESUMEN

Periodontal disease (PD) is a chronic inflammation of the tooth-supporting soft tissue and alveolar bone due to infection by a select group of gram-negative microbes, which leads to tooth loss if untreated. Because mice deficient in CD4(+) cells are resistant to infection-induced alveolar bone loss, Th cells have been implicated in bone-destructive processes during PD. However, the extent to which different Th cell subtypes play roles in pathogenesis or host protection remains to be defined and is likely to vary depending on the dominant microorganism involved. By far, Porphyromonas gingivalis is the best-studied periodontal microbe in PD. Although the gram-negative anaerobe Tannerella forsythia is also a vital contributor to periodontal bone loss, almost nothing is known about immune responses to this organism. Previous studies from our laboratory revealed that T. forsythia induces periodontal bone loss in mice and that this bone loss depends on the bacterially expressed BspA protein. In this study, we showed that T. forsythia activates murine APCs primarily through TLR2-dependent signaling via BspA. Furthermore, T. forsythia infection causes a pronounced Th2 bias, evidenced by T cell expression of IL-5, but not IFN-γ or IL-17, in draining lymph nodes. Consistently, deficiencies in TLR2 or STAT6 result in resistance to T. forsythia-induced alveolar bone loss. Thus, TLR2 signaling and Th2 cells play pathogenic roles in T. forsythia-induced alveolar bone destruction.


Asunto(s)
Pérdida de Hueso Alveolar/inmunología , Infecciones por Flavobacteriaceae/inmunología , Flavobacteriaceae/inmunología , Transducción de Señal/inmunología , Células Th2/inmunología , Receptor Toll-Like 2/fisiología , Pérdida de Hueso Alveolar/genética , Pérdida de Hueso Alveolar/patología , Animales , Infecciones por Flavobacteriaceae/genética , Infecciones por Flavobacteriaceae/patología , Mediadores de Inflamación/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Distribución Aleatoria , Transducción de Señal/genética , Células Th2/metabolismo , Células Th2/patología , Receptor Toll-Like 2/deficiencia
18.
Exp Cell Res ; 318(14): 1745-58, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22652453

RESUMEN

The adapters IRS1 and IRS2 link growth factor receptors to downstream signaling pathways that regulate proliferation and survival. Both suppress factor-withdrawal-induced apoptosis and have been implicated in cancer progression. However, recent studies suggest IRS1 and IRS2 mediate differential functions in cancer pathogenesis. IRS1 promoted breast cancer proliferation, while IRS2 promoted metastasis. The role of IRS1 and IRS2 in controlling cell responses to chemotherapy is unknown. To determine the role of IRS1 and IRS2 in the sensitivity of cells to chemotherapy, we treated 32D cells lacking or expressing IRS proteins with various concentrations of chemotherapeutic agents. We found that expression of IRS1, in contrast to IRS2, enhanced the sensitivity of 32D cells to chemotherapy-induced apoptosis. When IRS2 was expressed with IRS1, the cells no longer showed enhanced sensitivity. Expression of IRS1 did not alter the expression of pro- and anti-apoptotic proteins; however, 32D-IRS1 cells expressed higher levels of Annexin A2. In 32D-IRS1 cells, IRS1 and Annexin A2 were both located in cytoplasmic and membrane fractions. We also found that IRS1 coprecipitated with Annexin A2, while IRS2 did not. Decreasing Annexin A2 levels reduced 32D-IRS1 cell sensitivity to chemotherapy. These results suggest IRS1 enhances sensitivity to chemotherapy in part through Annexin A2.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Proteínas Sustrato del Receptor de Insulina/biosíntesis , Animales , Anexina A2/metabolismo , Muerte Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Proteínas Sustrato del Receptor de Insulina/metabolismo , Ratones , Relación Estructura-Actividad
19.
Front Immunol ; 14: 1297354, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38259471

RESUMEN

We investigated the effect of global Plexin B1 deficiency on allergic airway responses to house dust mite (HDM) or ovalbumin (OVA). In the HDM model, there were higher Th2 cytokine levels in the BALF of Plexin B1 knock-out (KO) mice compared to wild type (WT), and tissue inflammation and mucus production were modestly enhanced. In the OVA model, Plexin B1 deficiency led to increases in lung inflammation, mucus production, and lung Th2 cytokines accompanied by dysregulated mucin gene expression without affecting anti-OVA IgE/IgG1 levels. Spleen cells from Plexin B1 KO mice proliferated more robustly than WT cells in vitro to a variety of stimuli. Plexin B1 KO CD4+ T cells from spleens expressed higher levels of Ki-67 and CD69 compared to WT cells. Spleen cells from naïve Plexin B1 KO mice secreted increased amounts of IL-4 and IL-6 when pulsed in vitro with OVA whereas in vivo OVA-primed spleen cells produced IL-4/IL-5 when subjected to in vitro OVA restimulation. The upregulated allergic inflammatory response in Plexin B1 KO mice was associated with a lower number of Tregs in the lung tissues. Moreover, these mice displayed lower numbers of Treg cells in the lymphoid tissues at the baseline. These results demonstrate a previously unrecognized link between Plexin B1, Treg cells, and mucus in allergic lung inflammation.


Asunto(s)
Hipersensibilidad , Proteínas del Tejido Nervioso , Receptores de Superficie Celular , Linfocitos T Reguladores , Animales , Ratones , Citocinas , Dermatophagoides pteronyssinus , Inflamación , Interleucina-4 , Mucinas , Proteínas del Tejido Nervioso/metabolismo , Ovalbúmina , Neumonía , Receptores de Superficie Celular/metabolismo
20.
mBio ; 14(5): e0120823, 2023 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-37768050

RESUMEN

IMPORTANCE: Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, impacts millions of individuals worldwide and severely impairs the quality of life for patients. Dysregulation of innate immune signaling pathways reduces barrier function and exacerbates disease progression. Macrophage (Mφ) signaling pathways are potential targets for IBD therapies. While multiple treatments are available for IBD, (i) not all patients respond, (ii) responses may diminish over time, and (iii) treatments often have undesirable side effects. Genetic studies have shown that the inheritance of two co-segregating SNPs expressed in the innate immune receptor, TLR4, is associated with human IBD. Mice expressing homologous SNPs ("TLR4-SNP" mice) exhibited more severe colitis than WT mice in a DSS-induced colonic inflammation/repair model. We identified a critical role for M2a "tissue repair" Mφ in the resolution of colitis. Our findings provide insight into potential development of novel therapies targeting Mφ signaling pathways that aim to alleviate the debilitating symptoms experienced by individuals with IBD.


Asunto(s)
Colitis , Enfermedades Inflamatorias del Intestino , Humanos , Ratones , Animales , Receptor Toll-Like 4 , Polimorfismo de Nucleótido Simple , Calidad de Vida , Colitis/inducido químicamente , Macrófagos , Enfermedades Inflamatorias del Intestino/inducido químicamente , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA