Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 380
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Stem Cells ; 37(3): 430-440, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30537419

RESUMEN

Previously, we reported that although the HSPC frequency in bone marrow cells (BMC) was comparable between ß2-/- and ß2+/+ mice, transplantation of ß2-/- BMC into lethally irradiated CD45.1 recipient resulted in more myeloid cell production than ß2+/+ BMC. The objective of this study is to address if integrin ß2 deficiency skews granulocyte/macrophage progenitor (GMP) proliferation. FACS analysis demonstrated that GMP frequency and cell number were higher and megakaryocyte/erythrocyte progenitor frequency and cell number were lower in ß2-/- mice than ß2+/+ mice. However, the common myeloid progenitors (CMP) frequency and cell number were similar between the two groups. The increased GMP number was due to GMP proliferation as evidenced by the percentage of BrdU-incorporating GMP. Whole genome transcriptome analysis identified increased FcεRIα expression in ß2-/- CMP compared to ß2+/+ CMP. FcεRIα expression on ß2-/- GMP was detected increased in ß2-/- mice by qRT-PCR and FACS. Although transplantation of FcεRIαhi GMP or FcεRIαlo GMP into lethally irradiated CD45.1 recipient resulted in comparable myeloid cell production, transplantation of ß2 deficient FcεRIαhi GMP generated more myeloid cells than ß2+/+ FcεRIαhi GMP. GATA2 expression was increased in ß2-/- GMP. Using a luciferase reporter assay, we demonstrated that mutation of the GATA2 binding site in the FcεRIα promoter region diminished FcεRIα transcription. In vitro, the addition of IgE, the ligand of FcεRIα, promoted GMP expansion, which was abrogated by inhibition of JNK phosphorylation. Integrin ß2 deficiency promoted GMP proliferation and myeloid cell production, which was mediated via FcεRIα/IgE-induced JNK phosphorylation in GMP. Stem Cells 2019;37:430-440.


Asunto(s)
Antígenos CD18/metabolismo , Proliferación Celular , Células Progenitoras de Granulocitos y Macrófagos/metabolismo , Animales , Antígenos CD18/genética , Factor de Transcripción GATA2/genética , Factor de Transcripción GATA2/metabolismo , Regulación de la Expresión Génica , MAP Quinasa Quinasa 4 , Ratones , Ratones Noqueados , Receptores de IgE/biosíntesis , Receptores de IgE/genética , Transcripción Genética
2.
J Immunol ; 198(1): 229-238, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27852746

RESUMEN

Basophils represent <1% of circulating leukocytes. They play a crucial role during allergy and helminth-induced Th2 responses. However, recent data also suggest a contribution to the pathogenesis of autoimmune diseases. Basophils from patients with systemic lupus erythematosus show an activated phenotype, correlating to disease activity. Furthermore, murine basophils or their mediators enhance memory responses and plasma cell (PC) survival, suggesting that they directly modulate the function of B cells. This is highly relevant with respect to human allergy and autoimmunity because a possible modulation of B cell differentiation by basophils could point to new therapeutic targets. Therefore, the interaction between human B cells and basophils and the mechanism underlying this interaction were investigated in detail. Using two different methods to induce PC differentiation, we found that human basophils enhance B cell proliferation, class switching, differentiation into PC, maturation of PC, and production of Igs, especially IgG. Basophil supernatants enhanced the expression of the B cell markers CD23 and CD40, which are important for B cell differentiation into IgG-producing PC. This was mainly IL-4 dependent. IL-3 amplified the number of PC in vitro, and acted synergistically with basophils in enhancing Ab production. Thus, human basophils modulate B cell differentiation into Ab-producing PC. Their contribution as modulators and effectors during allergy and autoimmunity should be considered when designing new therapeutic options.


Asunto(s)
Basófilos/inmunología , Diferenciación Celular/inmunología , Activación de Linfocitos/inmunología , Células Plasmáticas/citología , Antígenos CD40/biosíntesis , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Ensayo de Immunospot Ligado a Enzimas , Citometría de Flujo , Humanos , Receptores de IgE/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
3.
J Cell Biochem ; 119(5): 4142-4149, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29243845

RESUMEN

Despite large number of investigations, the etiology of chronic rhinosinusitis (CRS) remains unclear. Several factors are likely involved in its onset. The genetic susceptibility of IgE-responsiveness likely caused by polymorphism(s) in high affinity receptor for IgE (FcɛR1α) gene can help in understanding the pathophysiology of CRS with nasal polyposis (CRSwNP). A population-based case-control association analysis was conducted to assess the risk of CRSwNP conferred by single nucleotide polymorphisms (SNPs) in FcɛR1α gene in a North Indian cohort. Two promoter and three exonic regions of FcɛR1α gene were amplified and sequenced to investigate five SNPs: rs2427827, rs2251746, rs2298804, rs2298805, and rs2269718. BLAST analysis and subsequent multiple alignments, with known sequences available in the NCBI database, were performed. Total serum IgE and FcɛR1α antibody levels were estimated. Patient IgE level of 461.22 ± 436.43 in comparison to 83.62 ± 58.043 IU/mL in controls (P < 0.0001), and FcɛR1α antibody level of 292.38 ± 115.27 in comparison to 160.56 ± 105.9 in controls (P < 0.0001), depicts their highly significant associations with CRSwNP disease. However, no SNP showed evidence of association with CRSwNP; although relatively higher Odds ratios were observed with rs2427827, rs2251746, and rs2298804. Patient stratification revealed a significant association (P < 0.05) of rs2427827 SNP with high IgE level CRSwNP patients. Nonetheless, we found no SNP associated with low serum IgE level patients. SNP (rs2427827) in the FcɛR1α gene region and high IgE levels may confer susceptibility to CRSwNP in north Indian population. However, further studies including larger sample size, gene-gene, and gene-environment interactions are required for its elucidation.


Asunto(s)
Inmunoglobulina E/sangre , Pólipos Nasales , Polimorfismo de Nucleótido Simple , Receptores de IgE , Rinitis , Sinusitis , Adulto , Enfermedad Crónica , Femenino , Humanos , Masculino , Pólipos Nasales/sangre , Pólipos Nasales/genética , Pólipos Nasales/patología , Receptores de IgE/biosíntesis , Receptores de IgE/genética , Rinitis/sangre , Rinitis/genética , Rinitis/patología , Sinusitis/sangre , Sinusitis/genética , Sinusitis/patología
4.
J Immunol ; 194(1): 364-78, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25452561

RESUMEN

Helminth parasites provoke multicellular immune responses in their hosts that can suppress concomitant disease. The gut lumen-dwelling tapeworm Hymenolepis diminuta, unlike other parasites assessed as helminth therapy, causes no host tissue damage while potently suppressing murine colitis. With the goal of harnessing the immunomodulatory capacity of infection with H. diminuta, we assessed the putative generation of anti-colitic regulatory B cells following H. diminuta infection. Splenic CD19(+) B cells isolated from mice infected 7 [HdBc(7(d))] and 14 d (but not 3 d) previously with H. diminuta and transferred to naive mice significantly reduced the severity of dinitrobenzene sulfonic acid (DNBS)-, oxazolone-, and dextran-sodium sulfate-induced colitis. Mechanistic studies with the DNBS model, revealed the anti-colitic HdBc(7(d)) was within the follicular B cell population and its phenotype was not dependent on IL-4 or IL-10. The HdBc(7(d)) were not characterized by increased expression of CD1d, CD5, CD23, or IL-10 production, but did spontaneously, and upon LPS plus anti-CD40 stimulation, produce more TGF-ß than CD19(+) B cells from controls. DNBS-induced colitis in RAG1(-/-) mice was inhibited by administration of HdBc(7(d)), indicating a lack of a requirement for T and B cells in the recipient; however, depletion of macrophages in recipient mice abrogated the anti-colitic effect of HdBc(7(d)). Thus, in response to H. diminuta, a putatively unique splenic CD19(+) B cell with a functional immunoregulatory program is generated that promotes the suppression of colitis dominated by TH1, TH2, or TH1-plus-TH2 events, and may do so via the synthesis of TGF-ß and the generation of, or cooperation with, a regulatory macrophage.


Asunto(s)
Linfocitos B/inmunología , Colitis/inmunología , Himenolepiasis/inmunología , Hymenolepis diminuta/inmunología , Macrófagos/inmunología , Animales , Antígenos CD19/biosíntesis , Antígenos CD1d/biosíntesis , Bencenosulfonatos , Antígenos CD40/inmunología , Antígenos CD5/biosíntesis , Colitis/inducido químicamente , Colitis/terapia , Sulfato de Dextran , Proteínas de Homeodominio/genética , Himenolepiasis/parasitología , Inmunomodulación/inmunología , Inmunoterapia , Interleucina-10/biosíntesis , Interleucina-10/inmunología , Interleucina-4/inmunología , Lipopolisacáridos , Masculino , Mastocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Oxazolona , Receptores de IgE/biosíntesis , Células TH1/inmunología , Células Th2/inmunología , Factor de Crecimiento Transformador beta/biosíntesis
5.
J Immunol ; 195(7): 3427-35, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26297757

RESUMEN

FcεRI, which is composed of α, ß, and γ subunits, plays an important role in IgE-mediated allergic responses. TGF-ß1 has been reported to suppress FcεRI and stem cell factor receptor c-Kit expression on mast cell surfaces and to suppress mast cell activation induced by cross-linking of FcεRI. However, the molecular mechanism by which these expressions and activation are suppressed by TGF-ß1 remains unclear. In this study, we found that the expression of Ets homologous factor (Ehf), a member of the Ets family transcriptional factors, is upregulated by TGF-ß/Smad signaling in mouse bone marrow-derived mast cells (BMMCs). Forced expression of Ehf in BMMCs repressed the transcription of genes encoding FcεRIα, FcεRIß, and c-Kit, resulting in a reduction in cell surface FcεRI and c-Kit expression. Additionally, forced expression of Ehf suppressed FcεRI-mediated degranulation and cytokine production. Ehf inhibited the promoter activity of genes encoding FcεRIα, FcεRIß, and c-Kit by binding to these gene promoters. Furthermore, the mRNA levels of Gata1, Gata2, and Stat5b were lower in BMMCs stably expressing Ehf compared with control cells. Because GATA-1 and GATA-2 are positive regulators of FcεRI and c-Kit expression, decreased expression of GATAs may be also involved in the reduction of FcεRI and c-Kit expression. Decreased expression of Stat5 may contribute to the suppression of cytokine production by BMMCs. In part, mast cell response to TGF-ß1 was mimicked by forced expression of Ehf, suggesting that TGF-ß1 suppresses FcεRI and c-Kit expression and suppresses FcεRI-mediated activation through upregulation of Ehf.


Asunto(s)
Proteínas Proto-Oncogénicas c-kit/biosíntesis , Receptores de IgE/inmunología , Factores de Transcripción/inmunología , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Células de la Médula Ósea , Degranulación de la Célula/inmunología , Células Cultivadas , Citocinas/biosíntesis , Factor de Transcripción GATA1/biosíntesis , Factor de Transcripción GATA1/genética , Factor de Transcripción GATA2/biosíntesis , Factor de Transcripción GATA2/genética , Inmunoglobulina E/inmunología , Mastocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-kit/genética , Interferencia de ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño , Receptores de IgE/biosíntesis , Factor de Transcripción STAT5/biosíntesis , Factor de Transcripción STAT5/genética , Transducción de Señal/inmunología , Proteínas Smad/metabolismo , Factores de Transcripción/biosíntesis , Transcripción Genética/genética , Activación Transcripcional
6.
J Immunol ; 191(9): 4505-13, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24068671

RESUMEN

We previously demonstrated that TGF-ß1 suppresses IgE-mediated signaling in human and mouse mast cells in vitro, an effect that correlated with decreased expression of the high-affinity IgE receptor, FcεRI. The in vivo effects of TGF-ß1 and the means by which it suppresses mast cells have been less clear. This study shows that TGF-ß1 suppresses FcεRI and c-Kit expression in vivo. By examining changes in cytokine production concurrent with FcεRI expression, we found that TGF-ß1 suppresses TNF production independent of FcεRI levels. Rather, IgE-mediated signaling was altered. TGF-ß1 significantly reduced expression of Fyn and Stat5, proteins critical for cytokine induction. These changes may partly explain the effects of TGF-ß1, because Stat5B overexpression blocked TGF-mediated suppression of IgE-induced cytokine production. We also found that Stat5B is required for mast cell migration toward stem cell factor, and that TGF-ß1 reduced this migration. We found evidence that genetic background may alter TGF responses. TGF-ß1 greatly reduced mast cell numbers in Th1-prone C57BL/6, but not Th2-prone 129/Sv mice. Furthermore, TGF-ß1 did not suppress IgE-induced cytokine release and did increase c-Kit-mediated migration in 129/Sv mast cells. These data correlated with high basal Fyn and Stat5 expression in 129/Sv cells, which was not reduced by TGF-ß1 treatment. Finally, primary human mast cell populations also showed variable sensitivity to TGF-ß1-mediated changes in Stat5 and IgE-mediated IL-6 secretion. We propose that TGF-ß1 regulates mast cell homeostasis, and that this feedback suppression may be dependent on genetic context, predisposing some individuals to atopic disease.


Asunto(s)
Inmunoglobulina E/inmunología , Mastocitos/metabolismo , Receptores de IgE/inmunología , Factor de Transcripción STAT5/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Movimiento Celular/inmunología , Células Cultivadas , Citocinas/metabolismo , Humanos , Inmunoglobulina E/metabolismo , Mastocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Receptores de IgE/biosíntesis , Receptores de IgE/metabolismo , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/inmunología , Transducción de Señal/inmunología , Factor de Crecimiento Transformador beta1/inmunología , Factores de Necrosis Tumoral/biosíntesis
7.
Immunology ; 143(2): 230-40, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24750112

RESUMEN

The IgE Fcε3 domain is an active immunotherapeutic target for asthma and other allergic diseases. However, previous methods for preparing IgE fusion protein vaccines are complex. Antigen 43 (Ag43) is a surface protein found in Escherichia coli that contains α and ß subunits (the α subunit contains multiple T epitopes). Here we constructed a novel Ag43 surface display system (Ag43 system) to express Ag43 chimeric proteins to disrupt immune tolerance against IgE. The Ag43 system was constructed from the E. coli strain Tan109, in which the Ag43 gene was deleted and a recombinant plasmid (pETAg43) expressing a partial Ag43 gene was introduced. The Fcε3 domain of the IgE gene was then subcloned into plasmid pETAg43, resulting in a recombinant plasmid pETAg43/Fcε3, which was used to transform Tan109 for Ag43/Fcε3 surface expression. Thereafter, Ag43/Fcε3 was investigated as an asthma vaccine in a mouse model. Ag43/Fcε3 was expressed on and could be separated from the bacterial surface by heating to 60° while retaining activity. Ag43/Fcε3, as a protein vaccine, produced neutralizing autoantibodies to murine IgE, induced significant anti-asthma effects, and regulated IgE and T helper cytokines in a murine asthma model. Data show that Ag43/Fcε3 chimeric protein is a potential model vaccine for asthma treatment, and that the Ag43 system may be an effective tool for novel vaccine preparation to break immune tolerance to other self-molecules.


Asunto(s)
Adhesinas de Escherichia coli/inmunología , Asma/prevención & control , Hiperreactividad Bronquial/prevención & control , Receptores de IgE/inmunología , Vacunas Sintéticas/inmunología , Adhesinas de Escherichia coli/biosíntesis , Adhesinas de Escherichia coli/genética , Traslado Adoptivo , Animales , Anticuerpos Neutralizantes/sangre , Asma/sangre , Asma/inmunología , Asma/fisiopatología , Autoanticuerpos/sangre , Hiperreactividad Bronquial/sangre , Hiperreactividad Bronquial/inmunología , Hiperreactividad Bronquial/fisiopatología , Broncoconstricción , Células Cultivadas , Clonación Molecular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Histamina/metabolismo , Tolerancia Inmunológica , Inmunoglobulina E/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Ovalbúmina/inmunología , Receptores de IgE/biosíntesis , Receptores de IgE/genética , Proteínas Recombinantes de Fusión/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Factores de Tiempo , Vacunas Sintéticas/biosíntesis , Vacunas Sintéticas/genética
8.
Clin Exp Allergy ; 44(2): 238-49, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24118172

RESUMEN

BACKGROUND: We recently reported that the interaction between Lyn and FcεRIß is indispensable for FcεRI-mediated human mast cell (MC) activation and that FcεRIß functions as an amplifier of FcεRI-mediated activation signal. Some of FcεRIß in cytoplasm appeared not to be co-localized with FcεRIα. The function of FcεRIß in the cytoplasm remains unknown. METHODS: The localization of FcεRIß and FcεRIα in giant papillae specimens from patients with allergic keratoconjunctivitis and of FcεRIß, FcεRIα, and Lyn in cultured human MCs was examined using confocal microscopy. FcεRIß was overexpressed using an adenovirus vector system. Mediators were measured by enzyme immunoassays or enzyme-linked immunosorbent assays. RESULTS: In the subepithelial region, FcεRIß was mainly localized in the cell membrane of MCs. In the perivascular region, FcεRIß expression was scattered throughout the cytoplasm and in the cell membrane of MCs. Overexpression of FcεRIß in MCs mainly increased its cytoplasmic expression and slightly up-regulated cell surface FcεRI expression. However, overexpression of FcεRIß in MCs resulted in down-regulation of the tyrosine phosphorylation levels of FcεRIß and Syk and down-regulation of the Ca(2+) influx soon after FcεRI aggregation and then resulted in down-regulation of degranulation, PGD2 synthesis, and production of a set of cytokines. This negative regulatory effect may be due to inhibition of the redistribution of Lyn to small patches within the plasma membrane. CONCLUSION: Cytoplasmic FcεRIß, which is not co-localized with FcεRIα, may function as a negative regulator, as it can capture important signalling molecules such as Lyn.


Asunto(s)
Señalización del Calcio , Regulación hacia Abajo , Hipersensibilidad/metabolismo , Queratoconjuntivitis/metabolismo , Mastocitos/metabolismo , Receptores de IgE/biosíntesis , Adulto , Línea Celular , Citoplasma , Femenino , Humanos , Hipersensibilidad/inmunología , Hipersensibilidad/patología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Queratoconjuntivitis/inmunología , Queratoconjuntivitis/patología , Masculino , Mastocitos/inmunología , Mastocitos/patología , Proteínas Tirosina Quinasas/inmunología , Proteínas Tirosina Quinasas/metabolismo , Receptores de IgE/inmunología , Quinasa Syk , Familia-src Quinasas/inmunología , Familia-src Quinasas/metabolismo
9.
Protein Expr Purif ; 96: 1-7, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24468271

RESUMEN

Secretory human interleukin 4 (hIL4) is an N-glycosylated pleiotropic cytokine. It is unknown if these N-linked glycans are required and essential for hIL4 protein stability, expression, secretion, and activity in vivo, and hIL4 expressed from Pichia pastoris yeast has not been tested to date. In this study, we successfully expressed human hIL4 in P. pastoris, the methylotrophic yeast, with a yield of 15.0mg/L. Using the site-directed mutagenesis technique, we made two mutant hIL4 cDNA clones (N38A and N105L) and subsequently expressed them in P. pastoris to analyze the relevant function of each N-glycosylation site on hIL4. Our results demonstrate that the glycosylation only occurs at position Asn38, but not Asn105. The glycosylated form of hIL4 unexpectedly has lower biological activity and lower stability when compared to its non-glycosylated form. The implications of this are discussed.


Asunto(s)
Interleucina-4/genética , Pichia/genética , Pichia/metabolismo , Proteínas Recombinantes/genética , Clonación Molecular , Expresión Génica , Glicoproteínas/genética , Glicoproteínas/metabolismo , Glicosilación , Humanos , Interleucina-4/biosíntesis , Interleucina-4/metabolismo , Mutagénesis Sitio-Dirigida , Receptores de IgE/biosíntesis
10.
Proc Natl Acad Sci U S A ; 108(2): 710-5, 2011 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-21187409

RESUMEN

Krüppel-like factor 2 (KLF2) controls T lymphocyte egress from lymphoid organs by regulating sphingosin-1 phosphate receptor 1 (S1Pr1). Here we show that this is not the case for B cells. Instead, KLF2 controls homeostasis of B cells in peripheral lymphatic organs and homing of plasma cells to the bone marrow, presumably by controlling the expression of ß(7)-integrin. In mice with a B cell-specific deletion of KLF2, S1Pr1 expression on B cells was only slightly affected. Accordingly, all splenic B cell subsets including B1 cells were present, but their numbers were increased with a clear bias for marginal zone (MZ) B cells. In contrast, fewer peyers patches harboring fewer B cells were found, and fewer B1 cells in the peritoneal cavity as well as recirculating B cells in the bone marrow were detected. Upon thymus-dependent immunization, IgG titers were diminished, and antigen-specific plasma cells were absent in the bone marrow, although numbers of antigen-specific splenic plasmablasts were normal. KLF2 plays also a role in determining the identity of follicular B cells, as KLF2-deficient follicular B cells showed calcium responses similar to those of MZ B cells and failed to down-regulate MZ B cell signature genes, such as CD21 and CXCR7.


Asunto(s)
Linfocitos B/citología , Factores de Transcripción de Tipo Kruppel/metabolismo , Células Plasmáticas/citología , Animales , Células de la Médula Ósea/citología , Calcio/metabolismo , Clorhidrato de Fingolimod , Eliminación de Gen , Cadenas beta de Integrinas/metabolismo , Selectina L/biosíntesis , Leucosialina/biosíntesis , Ratones , Modelos Biológicos , Glicoles de Propileno/farmacología , Receptores de IgE/biosíntesis , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/farmacología
11.
J Korean Med Sci ; 29(1): 43-7, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24431904

RESUMEN

Increased FcεR1α expression with upregulated CD203c expression on peripheral basophils is seen in patients with chronic urticaria (CU). However, there has been no published report on the association between CD203c expression level and clinical disease activity in CU patients. To investigate whether the increase of basophil activation is associated with the disease activity of CU, we measured basophil CD203c expression using a tricolor flow cytometric method in 82 CU patients and 21 normal controls. The relationship between the percentage of CD203c-expressing basophils and clinical parameters was analyzed. The mean basophil CD203c expression was significantly higher in CU patients than in healthy controls (57.5% vs 11.6%, P < 0.001). The basophil CD203c expression in severe CU patients was significantly higher than in non-severe CU (66.5% ± 23.3% vs 54.0% ± 23.3%, P = 0.033). Multiple logistic regression analysis indicated that both ≥ 72% basophil CD203c expression and urticaria activity score (UAS)≥ 13 were significant predictors of severe CU (P = 0.005 and P = 0.032, respectively). These findings suggest that the quantification of basophil activation with CD203c at baseline may be used as a potential predictor of severe CU requiring another treatment option beyond antihistamines.


Asunto(s)
Basófilos/inmunología , Hidrolasas Diéster Fosfóricas/inmunología , Pirofosfatasas/inmunología , Urticaria/inmunología , Adulto , Autoanticuerpos/sangre , Femenino , Citometría de Flujo , Humanos , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Masculino , Hidrolasas Diéster Fosfóricas/biosíntesis , Pirofosfatasas/biosíntesis , Receptores de IgE/biosíntesis
13.
Cell Immunol ; 283(1-2): 38-44, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23850674

RESUMEN

There is increasing evidence that histamine regulates the immune system via histamine H4 receptors, therefore we sought to investigate the functions of the H4 receptor on mast cells. Mast cells were differentiated from murine bone marrow stem cells, and the expression of mast cell surface markers FcεRI and CD117 were measured using flow cytometry. Real-time qRT-PCR was used to determine the expression of mH4R; as a measure of antigen-dependent degranulation, ß-hexosaminidase release assay was carried out using IgE sensitized mast cells. We determined that the expression kinetics of FcεRI and mH4R can be described with a function that has one maximum value in the time range of the culture's differentiation. Antigen-dependent degranulation of murine bone marrow-derived mast cells could be inhibited by a selective H4 antagonist/inverse agonist only when it was present during the IgE sensitization phase of degranulation. In addition, flow cytometric analysis revealed that the H4 antagonist/inverse agonist also inhibited IgE induced FcεRI upregulation. The inhibition percentage of H4 antagonist on IgE induced FcεRI upregulation was determined to be dependent upon the maturity of the mast cell cultures, and this time-dependency was consistent with the expression kinetics of both mH4R and FcεRI. These results imply that H4R has regulatory roles in FcεRI expression and FcεRI mediated functions in mast cells. In conclusion the present study shows that H4 receptors potentially play a role in IgE induced FcεRI upregulation and in the sensitization phase but not the effector phase of mast cell degranulation.


Asunto(s)
Degranulación de la Célula/inmunología , Mastocitos/metabolismo , Receptores Histamínicos/metabolismo , Receptores de IgE/biosíntesis , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Citometría de Flujo , Inmunoglobulina E/inmunología , Masculino , Mastocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Histamínicos/inmunología , Receptores de IgE/inmunología , Regulación hacia Arriba
14.
J Immunol ; 186(2): 1060-7, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21160045

RESUMEN

Human peripheral blood BCRµ(+) B cells express high levels of CD23 and circulate preloaded with IgE. The Ag specificity of CD23-bound IgE presumably differs from the BCR and likely reflects the Ag-specific mix of free serum IgE. CD23-bound IgE is thought to enhance B cell Ag presentation to T cells raising the question of how a B cell might respond when presented with a broad mix of Ags and CD23-bound IgE specificities. We recently reported that an increase in CD23(+) B cells is associated with the development of resistance to schistosomiasis, highlighting the potential importance of CD23-bound IgE in mediating immunity. We sought to determine the relationship between BCR and CD23-bound IgE-mediated B cell activation in the context of schistosomiasis. We found that crude schistosome Ags downregulate basal B cell activation levels in individuals hyperexposed to infectious worms. Schistosome-specific IgE from resistant, occupationally exposed Kenyans recovered responses of B cells to schistosome Ag. Furthermore, cross-linking of CD23 overrode intracellular signals mediated via the BCR, illustrating its critical and dominating role in B cell activation. These results suggest that CD23-bound IgE augments and dominates recall responses through naive B cells.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Inmunidad Innata/inmunología , Inmunoglobulina E/metabolismo , Activación de Linfocitos/inmunología , Receptores de IgE/fisiología , Fase de Descanso del Ciclo Celular/inmunología , Esquistosomiasis mansoni/inmunología , Animales , Presentación de Antígeno/inmunología , Antígenos Helmínticos/inmunología , Antígenos Helmínticos/metabolismo , Subgrupos de Linfocitos B/citología , Subgrupos de Linfocitos B/metabolismo , Sitios de Unión de Anticuerpos , Línea Celular Tumoral , Humanos , Inmunidad Innata/genética , Inmunoglobulina E/fisiología , Memoria Inmunológica/genética , Líquido Intracelular/inmunología , Líquido Intracelular/parasitología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos NZB , Unión Proteica/inmunología , Receptores de IgE/biosíntesis , Receptores de IgE/metabolismo , Esquistosomiasis mansoni/metabolismo , Esquistosomiasis mansoni/prevención & control
15.
J Immunol ; 186(6): 3484-96, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21307287

RESUMEN

IgE-mediated allergic inflammation occurs when allergens cross-link IgE on the surface of immune cells, thereby triggering the release of inflammatory mediators as well as enhancing Ag presentations. IgE is frequently present in airway secretions, and its level can be enhanced in human patients with allergic rhinitis and bronchial asthma. However, it remains completely unknown how IgE appears in the airway secretions. In this study, we show that CD23 (FcεRII) is constitutively expressed in established or primary human airway epithelial cells, and its expression is significantly upregulated when airway epithelial cells were subjected to IL-4 stimulation. In a transcytosis assay, human IgE or IgE-derived immune complex (IC) was transported across a polarized Calu-3 monolayer. Exposure of the Calu-3 monolayer to IL-4 stimulation also enhanced the transcytosis of either human IgE or the IC. A CD23-specific Ab or soluble CD23 significantly reduced the efficiency of IgE or IC transcytosis, suggesting a specific receptor-mediated transport by CD23. Transcytosis of both IgE and the IC was further verified in primary human airway epithelial cell monolayers. Furthermore, the transcytosed Ag-IgE complexes were competent in inducing degranulation of the cultured human mast cells. Because airway epithelial cells are the first cell layer to come into contact with inhaled allergens, our study implies CD23-mediated IgE transcytosis in human airway epithelial cells may play a critical role in initiating and contributing to the perpetuation of airway allergic inflammation.


Asunto(s)
Complejo Antígeno-Anticuerpo/metabolismo , Polaridad Celular/inmunología , Inmunoglobulina E/metabolismo , Receptores de IgE/fisiología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Transcitosis/inmunología , Alérgenos/administración & dosificación , Animales , Complejo Antígeno-Anticuerpo/fisiología , Bronquios/inmunología , Bronquios/metabolismo , Bronquios/patología , Células CHO , Línea Celular , Línea Celular Tumoral , Polaridad Celular/genética , Cricetinae , Cricetulus , Células HEK293 , Células HT29 , Humanos , Inmunoglobulina E/fisiología , Mediadores de Inflamación/administración & dosificación , Mastocitos/inmunología , Mastocitos/metabolismo , Mastocitos/patología , Receptores de IgE/biosíntesis , Receptores de IgE/genética , Mucosa Respiratoria/patología , Transcitosis/genética , Células U937
16.
Clin Exp Allergy ; 42(7): 1060-70, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22702505

RESUMEN

BACKGROUND: Clinical desensitization of patients to drugs involves progressive exposure to escalating doses of drug over a period of 24 h. In prior studies, this method was re-capitulated in vitro to also demonstrate loss of mast cell or basophil responsiveness. However, most signalling studies of human basophils have identified changes in signalling by using other methods of inducing cellular desensitization. OBJECTIVE: This study examined two well-described endpoints of basophil desensitization, loss of syk or FcεRI expression, under conditions of subthreshold desensitization. METHODS: The loss of FcεRI and syk was examined in human basophils. RESULTS: It was shown that both loss of syk and FcεRI/IgE occurred during an escalating series of stimulation (anti-IgE Ab) and that expression loss occurred despite the presence of little histamine release. If basophils were first cultured for 3 days in 10 ng/mL IL-3, the concentration-dependence of histamine release shifted to 100-fold lower concentrations of stimulus. However, loss of syk did not show any change in its EC50 while loss of FcεRI also shifted 100-fold. From the perspective of early signal element activation, the marked shift in the EC50 for histamine release was not accompanied by similar shifts in the EC50s for several signalling elements. The EC50s for phospho-Src, phospho-SHIP1, phospho-Syk, or phospho-Cbl did not change while the EC50s for phospho-Erk and the cytosolic calcium response did shift 100-fold. CONCLUSIONS: These studies show that under normal conditions, subthreshold desensitization leads to loss of two critical signalling molecules (FcεRI and syk) but under at least one condition, treatment with IL-3, it is possible to markedly blunt the loss of syk, but not FcεRI, while executing a proper subthreshold titration. These data also suggest that IL-3 modifies only the sensitivity of signalling elements that are downstream of syk activation.


Asunto(s)
Basófilos/metabolismo , Desensibilización Inmunológica , Hipersensibilidad a las Drogas/metabolismo , Regulación de la Expresión Génica , Proteínas Tirosina Quinasas/biosíntesis , Receptores de IgE/biosíntesis , Anticuerpos/farmacología , Basófilos/inmunología , Células Cultivadas , Hipersensibilidad a las Drogas/inmunología , Hipersensibilidad a las Drogas/patología , Histamina/inmunología , Histamina/metabolismo , Humanos , Interleucina-3/farmacología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Proteínas Tirosina Quinasas/inmunología , Receptores de IgE/inmunología , Transducción de Señal/inmunología , Quinasa Syk
17.
Blood ; 115(23): 4734-41, 2010 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-20203264

RESUMEN

Activating transcription factor 3 (ATF3) is a basic leucine zipper transcription factor that plays a regulatory role in inflammation, cell division, and apoptosis. Mast cells (MCs) initiate many inflammatory responses and have a central role in allergy and allergic diseases. We report here that ATF3 has a central role in MC development and function. Bone marrow-derived MC populations from ATF3-deficient mice are unresponsive to interleukin-3 (IL-3)-induced maturation signals, and this correlates with increased apoptosis, diminished activation of the Akt kinase, and decreased phosphorylation of the proapoptotic protein Bad. Furthermore, ATF3-null mice lacked MCs in the peritoneum and dermis, showing that the in vitro results are recapitulated in vivo. ATF3-null MCs also showed functional defects; high-affinity immunoglobulin E receptor-mediated degranulation was significantly inhibited, whereas IL-4 and IL-6 expression was enhanced. This dual role of ATF3 provides insight into the complex interplay between MC development and its subsequent physiologic role.


Asunto(s)
Factor de Transcripción Activador 3/inmunología , Apoptosis/inmunología , Células de la Médula Ósea/inmunología , Mediadores de Inflamación/inmunología , Mastocitos/inmunología , Factor de Transcripción Activador 3/genética , Factor de Transcripción Activador 3/metabolismo , Animales , Apoptosis/genética , Células de la Médula Ósea/metabolismo , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Hipersensibilidad/genética , Hipersensibilidad/inmunología , Hipersensibilidad/metabolismo , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Mediadores de Inflamación/metabolismo , Interleucina-3/biosíntesis , Interleucina-3/genética , Interleucina-3/inmunología , Interleucina-4/genética , Interleucina-4/inmunología , Interleucina-4/metabolismo , Interleucina-6/genética , Interleucina-6/inmunología , Interleucina-6/metabolismo , Mastocitos/metabolismo , Ratones , Ratones Noqueados , Fosforilación/genética , Fosforilación/inmunología , Receptores de IgE/biosíntesis , Receptores de IgE/genética , Receptores de IgE/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología , Proteína Letal Asociada a bcl/genética , Proteína Letal Asociada a bcl/inmunología , Proteína Letal Asociada a bcl/metabolismo
18.
Ann Allergy Asthma Immunol ; 108(3): 188-94, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22374203

RESUMEN

BACKGROUND: A large body of evidence has demonstrated that treatment with omalizumab is clinically effective for the management of moderate to severe allergic asthma, emphasizing the importance of IgE in the pathogenesis of allergic asthma. We hypothesized that IgE accelerates FcεRI-mediated responsiveness of "immature" human mast cells (MCs) and that omalizumab downregulates the acceleration. OBJECTIVES: To examine when MC progenitors acquired the ability to degranulate following FcεRI aggregation, whether IgE accelerates the responsiveness of immature MCs following FcεRI aggregation, and whether omalizumab regulates such an acceleration. METHODS: Gene expression was examined using a microarray and quantitative reverse transcription polymerase chain reaction. Protein expression was investigated using FACS. Histamine release was examined using an EIA. RESULTS: The time-course analysis of the mRNA expression of MC-related genes, including FcεRI, in Kit(+) sorted cells during the differentiation and histamine experiments revealed that the expression level of FcεRI in 5 week (w)-cultured MCs was not sufficient to induce degranulation following FcεRI aggregation but that 5 w-cultured MCs were fully responsive to calcium ionophore. By addition of IgE in culture medium FcεRI expression level and FcεRI-mediated histamine release of 5 w-cultured MCs were significantly increased compared with those without addition of IgE, whereas the expression level of tryptase and number of MCs was not affected. Omalizumab significantly inhibited IgE-dependent enhancement of FcεRI expression level and FcεRI-mediated histamine release. CONCLUSIONS: High levels of IgE in the microenvironment in vivo may upregulate the responsiveness of immature MCs to allergens. Omalizumab may inhibit the IgE-mediated responsiveness of not only mature MCs, but also immature MCs.


Asunto(s)
Anticuerpos Antiidiotipos/farmacología , Anticuerpos Monoclonales Humanizados/farmacología , Inmunoglobulina E/inmunología , Mastocitos/inmunología , Receptores de IgE/inmunología , Asma/patología , Células Cultivadas , Expresión Génica , Liberación de Histamina , Humanos , Inmunoglobulina E/metabolismo , Mastocitos/metabolismo , Omalizumab , ARN Mensajero/biosíntesis , Receptores de IgE/biosíntesis
19.
J Immunol ; 184(11): 5999-6006, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20410486

RESUMEN

Plasmacytoid dendritic cells (pDCs) play essential roles in directing immune responses. These cells may be particularly important in determining the nature of immune responses to viral infections in patients with allergic asthma as well those with other atopic diseases. The purposes of this study were 1) to compare the functional capacity of pDCs in patients with one type of allergic disorder, allergic asthma, and controls; 2) to determine whether IgE cross-linking affects antiviral responses of influenza-exposed pDCs; and 3) to determine whether evidence of counterregulation of FcepsilonRIalpha and IFN-alpha pathways exists in these cells. pDC function was assessed in a subset of asthma patients and in controls by measuring IFN-alpha production after exposure of purified pDCs to influenza viruses. FcepsilonRIalpha expression on pDCs was determined by flow cytometry in blood samples from patients with allergic asthma and controls. pDCs from patients with asthma secreted significantly less IFN-alpha upon exposure to influenza A (572 versus 2815; p = 0.03), and secretion was inversely correlated with serum IgE levels. Moreover, IgE cross-linking prior to viral challenge resulted in 1) abrogation of the influenza-induced pDC IFN-alpha response; 2) diminished influenza and gardiquimod-induced TLR-7 upregulation in pDCs; and 3) interruption of influenza-induced upregulation of pDC maturation/costimulatory molecules. In addition, exposure to influenza and gardiquimod resulted in upregulation of TLR-7, with concomitant downregulation of FcepsilonRIalpha expression in pDCs. These data suggest that counterregulation of FcepsilonRI and TLR-7 pathways exists in pDCs, and that IgE cross-linking impairs pDC antiviral responses.


Asunto(s)
Células Dendríticas/inmunología , Virus de la Influenza A/inmunología , Interferón-alfa/inmunología , Receptores de IgE/inmunología , Hipersensibilidad Respiratoria/inmunología , Adolescente , Adulto , Separación Celular , Niño , Preescolar , Células Dendríticas/virología , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Humanos , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Interferón-alfa/biosíntesis , Masculino , Receptores de IgE/biosíntesis , Receptor Toll-Like 7/inmunología , Adulto Joven
20.
J Immunol ; 184(9): 4662-72, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20357250

RESUMEN

We have previously shown that TLR4 triggering promotes the generation of CD23(+)CD93(+) transitional T2-like cells in vitro from mouse B cell precursors, suggesting a possible role for this receptor in B cell maturation. In this study, we perform an extensive study of cell surface markers and functional properties of B cells matured in vitro with LPS, comparatively with the well-known B cell maturation factor B lymphocyte-activating factor (BAFF). LPS increased generation of CD23(+) transitional B cells in a TLR4-dependent way, upregulating IgD and CD21 and downregulating CD93, without inducing cell proliferation, in a manner essentially equivalent to BAFF. For both BAFF and LPS, functional maturation of the IgM(+)CD23(+)CD93(+) cells was confirmed by their higher proliferative response to anti-CD40 plus IL-4 compared with IgM(+)CD23(neg)CD93(+) cells. BAFF-R-Fc-mediated neutralization experiments showed that TLR4-induced B cell maturation was independent of BAFF. Distinct from BAFF, maturation by LPS relied on the activation of canonical NF-kappaB pathway, and the two factors together had complementary effects, leading to higher numbers of IgM(+)CD23(+)CD93(+) cells with their simultaneous addition. Importantly, BCR cross-linking abrogated the generation of CD23(+) B cells by LPS or BAFF, indicating that signals mimicking central tolerance act on both systems. Addition of cyclosporin A reverted BCR-mediated inhibition, both for BAFF and LPS, suggesting similar regulation of signaling pathways by calcineurin. Finally, LPS-injected mice showed a rapid increase of mature B cells in the bone marrow, suggesting that TLR4 signaling may effectively stimulate B cell maturation in vivo, acting as an accessory stimulus in B cell development, complementary to the BAFF physiological pathway.


Asunto(s)
Factor Activador de Células B/fisiología , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Diferenciación Celular/inmunología , Cooperación Linfocítica/inmunología , Receptor Toll-Like 4/fisiología , Animales , Subgrupos de Linfocitos B/citología , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Lipopolisacáridos/fisiología , Ratones , Ratones Endogámicos C57BL , Receptores de IgE/biosíntesis , Transducción de Señal/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA