Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Allergy Clin Immunol ; 146(3): 652-666.e11, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32445838

RESUMEN

BACKGROUND: Effector functions of IgG Abs are regulated by their Fc N-glycosylation pattern. IgG Fc glycans that lack galactose and terminal sialic acid residues correlate with the severity of inflammatory (auto)immune disorders and have also been linked to protection against viral infection and discussed in the context of vaccine-induced protection. In contrast, sialylated IgG Abs have shown immunosuppressive effects. OBJECTIVE: We sought to investigate IgG glycosylation programming during the germinal center (GC) reaction following immunization of mice with a foreign protein antigen and different adjuvants. METHODS: Mice were analyzed for GC T-cell, B-cell, and plasma cell responses, as well as for antigen-specific serum IgG subclass titers and Fc glycosylation patterns. RESULTS: Different adjuvants induce distinct IgG+ GC B-cell responses with specific transcriptomes and expression levels of the α2,6-sialyltransferase responsible for IgG sialylation that correspond to distinct serum IgG Fc glycosylation patterns. Low IgG Fc sialylation programming in GC B cells was overall highly dependent on the Foxp3- follicular helper T (TFH) cell-inducing cytokine IL-6, here in particular induced by water-in-oil adjuvants and Mycobacterium tuberculosis. Furthermore, low IgG Fc sialylation programming was dependent on adjuvants that induced IL-27 receptor-dependent IFN-γ+ TFH1 cells, IL-6/IL-23-dependent IL-17A+ TFH17 cells, and high ratios of TFH cells to Foxp3+ follicular regulatory T cells. Here, the 2 latter were dependent on M tuberculosis and its cord factor. CONCLUSION: This study's findings regarding adjuvant-dependent GC responses and IgG glycosylation programming may aid in the development of novel vaccination strategies to induce IgG Abs with both high affinity and defined Fc glycosylation patterns in the GC.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Antígenos/administración & dosificación , Centro Germinal/inmunología , Inmunoglobulina G/inmunología , Compuestos de Alumbre/administración & dosificación , Animales , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Citocinas/inmunología , Femenino , Adyuvante de Freund/administración & dosificación , Glicosilación , Lipopolisacáridos/administración & dosificación , Ratones Endogámicos C57BL , Ratones Noqueados , Aceite Mineral/administración & dosificación , Mycobacterium tuberculosis/inmunología , Ovalbúmina/administración & dosificación , Polisorbatos/administración & dosificación , Escualeno/administración & dosificación , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Vacunación
2.
Int J Mol Sci ; 22(3)2021 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-33572870

RESUMEN

As a key anti-inflammatory cytokine, IL-10 is crucial in preventing inflammatory and autoimmune diseases. However, in human and murine lupus, its role remains controversial. Our aim was to understand regulation and immunologic effects of IL-10 on different immune functions in the setting of lupus. This was explored in lupus-prone NZB/W F1 mice in vitro and vivo to understand IL-10 effects on individual immune cells as well as in the complex in vivo setting. We found pleiotropic IL-10 expression that largely increased with progressing lupus, while IL-10 receptor (IL-10R) levels remained relatively stable. In vitro experiments revealed pro- and anti-inflammatory IL-10 effects. Particularly, IL-10 decreased pro-inflammatory cytokines and slowed B cell proliferation, thereby triggering plasma cell differentiation. The frequent co-expression of ICOS, IL-21 and cMAF suggests that IL-10-producing CD4 T cells are important B cell helpers in this context. In vitro and in vivo effects of IL-10 were not fully concordant. In vivo IL-10R blockade slightly accelerated clinical lupus manifestations and immune dysregulation. Altogether, our side-by-side in vitro and in vivo comparison of the influence of IL-10 on different aspects of immunity shows that IL-10 has dual effects. Our results further reveal that the overall outcome may depend on the interplay of different factors such as target cell, inflammatory and stimulatory microenvironment, disease model and state. A comprehensive understanding of such influences is important to exploit IL-10 as a therapeutic target.


Asunto(s)
Interleucina-10/inmunología , Lupus Eritematoso Sistémico/inmunología , Inmunidad Adaptativa , Animales , Autoinmunidad , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos B/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Expresión Génica , Humanos , Inmunidad Innata , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Interleucina-10/genética , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/patología , Ratones Endogámicos NZB , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/patología
3.
J Immunol ; 197(5): 1567-76, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27439515

RESUMEN

Contact hypersensitivity (CHS) of murine skin serves as a model of allergic contact dermatitis. Hapten-specific CD8 T cells and neutrophils represent the major effector cells driving this inflammatory reaction whereas Foxp3(+) regulatory T cells (Tregs) control the severity of inflammation. However, whether in vivo expansion of endogenous Tregs can downregulate CHS-mediated inflammation remains to be elucidated. In this study, we addressed this issue by using injection of an IL-2/anti-IL-2 mAb JES6-1 complex (IL-2/JES6-1) as a means of Treg induction in 2,4,6-trinitrochlorobenzene-induced CHS. IL-2/JES6-1 injection before or after hapten sensitization led to a considerable reduction of skin inflammation, even when rechallenged up to 3 wk after the last treatment. Conversely, Treg depletion re-established the CHS response in IL-2/JES6-1-treated mice. IL-2/JES6-1 injection resulted in increased frequencies of natural and peripheral Tregs in spleen and draining lymph nodes (LNs), elevated IL-10 and TGF-ß production by CD4 T cells, reduced CD86 expression by dendritic cells, and led to lower numbers of hapten-specific IFN-γ-producing CD8 T effector cells in LNs. Neutrophil and CD8 T cell infiltration was reduced in inflamed ear tissue, whereas CTLA-4(+)Foxp3(+) Treg frequencies were augmented. Adoptive transfer of LN cells of sensitized mice into recipients treated with IL-2/JES6-1 showed impaired CHS. Our results show that in vivo Treg expansion results in a prolonged CHS suppression, a sustained reduction of hapten-specific CD8 T cells, and a decrease in effector cell influx in inflamed tissue.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Dermatitis por Contacto/inmunología , Inflamación/inmunología , Activación de Linfocitos , Piel/inmunología , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Linfocitos T CD4-Positivos/inmunología , Haptenos/inmunología , Inflamación/tratamiento farmacológico , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Interleucina-10/inmunología , Interleucina-2/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Neutrófilos/inmunología , Piel/patología
4.
Arterioscler Thromb Vasc Biol ; 36(6): 1174-85, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27102965

RESUMEN

OBJECTIVE: Explore aorta B-cell immunity in aged apolipoprotein E-deficient (ApoE(-/-)) mice. APPROACH AND RESULTS: Transcript maps, fluorescence-activated cell sorting, immunofluorescence analyses, cell transfers, and Ig-ELISPOT (enzyme-linked immunospot) assays showed multilayered atherosclerosis B-cell responses in artery tertiary lymphoid organs (ATLOs). Aging-associated aorta B-cell-related transcriptomes were identified, and transcript atlases revealed highly territorialized B-cell responses in ATLOs versus atherosclerotic lesions: ATLOs showed upregulation of bona fide B-cell genes, including Cd19, Ms4a1 (Cd20), Cd79a/b, and Ighm although intima plaques preferentially expressed molecules involved in non-B effector responses toward B-cell-derived mediators, that is, Fcgr3 (Cd16), Fcer1g (Cd23), and the C1q family. ATLOs promoted B-cell recruitment. ATLO B-2 B cells included naive, transitional, follicular, germinal center, switched IgG1(+), IgA(+), and IgE(+) memory cells, plasmablasts, and long-lived plasma cells. ATLOs recruited large numbers of B-1 cells whose subtypes were skewed toward interleukin-10(+) B-1b cells versus interleukin-10(-) B-1a cells. ATLO B-1 cells and plasma cells constitutively produced IgM and IgG and a fraction of plasma cells expressed interleukin-10. Moreover, ApoE(-/-) mice showed increased germinal center B cells in renal lymph nodes, IgM-producing plasma cells in the bone marrow, and higher IgM and anti-MDA-LDL (malondialdehyde-modified low-density lipoprotein) IgG serum titers. CONCLUSIONS: ATLOs orchestrate dichotomic, territorialized, and multilayered B-cell responses in the diseased aorta; germinal center reactions indicate generation of autoimmune B cells within the diseased arterial wall during aging.


Asunto(s)
Envejecimiento/inmunología , Aorta/inmunología , Enfermedades de la Aorta/inmunología , Apolipoproteínas E/deficiencia , Aterosclerosis/inmunología , Linfocitos B/inmunología , Estructuras Linfoides Terciarias/inmunología , Envejecimiento/genética , Envejecimiento/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Autoanticuerpos/sangre , Autoinmunidad , Linfocitos B/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Centro Germinal/inmunología , Centro Germinal/metabolismo , Inmunoglobulinas/sangre , Memoria Inmunológica , Lipoproteínas LDL/inmunología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Masculino , Malondialdehído/análogos & derivados , Malondialdehído/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Transducción de Señal , Estructuras Linfoides Terciarias/metabolismo , Estructuras Linfoides Terciarias/patología , Transcriptoma
5.
Arthritis Rheum ; 64(4): 1237-46, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22006377

RESUMEN

OBJECTIVE: Autoantibody immune complexes and cellular infiltrates drive nephritis in patients with systemic lupus erythematosus (SLE) and in murine lupus. The chemokine receptor CXCR3 is assumed to promote cellular infiltration of inflamed tissues. Moreover, CXCR3 deficiency ameliorates lupus nephritis in the MRL/MpJ-Fas(lpr) (MRL/lpr) mouse model of SLE. Hence, CXCR3 blockade has been suggested as a novel therapeutic strategy for the treatment of lupus nephritis. We undertook this study to test the effect of CXCR3 in the (NZB × NZW)F(1) (NZB/NZW) mouse model of SLE. METHODS: CXCR3(-/-) NZB/NZW mice were generated and monitored for survival, proteinuria, and kidney infiltration. Anti-double-stranded DNA (anti-dsDNA) and total IgG1, IgG2a, and IgG2b antibody levels were determined by enzyme-linked immunosorbent assay. T cell and plasma cell infiltrates in the kidneys and interferon-γ production were determined by flow cytometry. Plasma cell infiltrates were measured using enzyme-linked immunospot assay. Kidney tissue was evaluated for pathologic changes. RESULTS: CXCR3(-/-) NZB/NZW mice exhibited reduced production of total and anti-dsDNA antibodies of the IgG1 subclass, but had normal titers of IgG2a and IgG2b antibodies compared to CXCR3(+/+) NZB/NZW mice. Cellular infiltrates and glomerulonephritis were not reduced in CXCR3(-/-) mice. CONCLUSION: CXCR3 has an effect on (auto)antibody production but is not essential for lupus pathogenesis in NZB/NZW mice, indicating that the effect of CXCR3 on the development of kidney disease varies between MRL/lpr and NZB/NZW mice. These results suggest that CXCR3-dependent and -independent mechanisms can mediate lupus nephritis. Hence, therapeutic CXCR3 blockade could be beneficial for only a subgroup of patients with SLE.


Asunto(s)
Autoanticuerpos/inmunología , Inmunoglobulina G/inmunología , Nefritis Lúpica/metabolismo , Receptores CXCR3/metabolismo , Animales , Complejo Antígeno-Anticuerpo/inmunología , Nefritis Lúpica/inmunología , Ratones , Ratones Endogámicos NZB , Proteinuria/inmunología , Proteinuria/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo
6.
J Immunol ; 186(4): 2503-13, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21239719

RESUMEN

Bortezomib, an inhibitor of proteasomes, has been reported to reduce autoantibody titers and to improve clinical condition in mice suffering from lupus-like disease. Bortezomib depletes both short- and long-lived plasma cells; the latter normally survive the standard immunosuppressant treatments targeting T and B cells. These findings encouraged us to test whether bortezomib is effective for alleviating the symptoms in the experimental autoimmune myasthenia gravis (EAMG) model for myasthenia gravis, a disease that is characterized by autoantibodies against the acetylcholine receptor (AChR) of skeletal muscle. Lewis rats were immunized with saline (control, n = 36) or Torpedo AChR (EAMG, n = 54) in CFA in the first week of an experimental period of 8 wk. After immunization, rats received twice a week s.c. injections of bortezomib (0.2 mg/kg in saline) or saline injections. Bortezomib induced apoptosis in bone marrow cells and reduced the amount of plasma cells in the bone marrow by up to 81%. In the EAMG animals, bortezomib efficiently reduced the rise of anti-AChR autoantibody titers, prevented ultrastructural damage of the postsynaptic membrane, improved neuromuscular transmission, and decreased myasthenic symptoms. This study thus underscores the potential of the therapeutic use of proteasome inhibitors to target plasma cells in Ab-mediated autoimmune diseases.


Asunto(s)
Autoanticuerpos/efectos de los fármacos , Ácidos Borónicos/farmacología , Miastenia Gravis Autoinmune Experimental/tratamiento farmacológico , Miastenia Gravis Autoinmune Experimental/inmunología , Células Plasmáticas/efectos de los fármacos , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasoma , Pirazinas/farmacología , Animales , Autoanticuerpos/biosíntesis , Bortezomib , Femenino , Depleción Linfocítica/métodos , Miastenia Gravis Autoinmune Experimental/enzimología , Células Plasmáticas/enzimología , Células Plasmáticas/patología , Ratas , Ratas Endogámicas Lew
7.
Biology (Basel) ; 12(12)2023 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-38132327

RESUMEN

Food allergies are a growing public health concern worldwide, especially in children and young adults. Allergen-specific IgE plays a central role in the pathogenesis of food allergies, but their titers poorly correlate with allergy development. Host immune systems yield allergen-specific immunoglobulin (Ig)A, IgE and IgG subclasses with low or high affinities and differential Fc N-glycosylation patterns that can affect the allergic reaction to food in multiple ways. High-affinity IgE is required to induce strong mast cell activation eventually leading to allergic anaphylaxis, while low-affinity IgE can even inhibit the development of clinically relevant allergic symptoms. IgA and IgG antibodies can inhibit IgE-mediated mast cell activation through various mechanisms, thereby protecting IgE-positive individuals from allergy development. The production of IgE and IgG with differential allergenic potential seems to be affected by the signaling strength of individual B cell receptors, and by cytokines from T cells. This review provides an overview of the diversity of the B cell response and the diverse roles of antibodies in food allergy.

8.
Nat Commun ; 14(1): 7081, 2023 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-37925420

RESUMEN

B cells play a central role in humoral immunity but also have antibody-independent functions. Studies to date have focused on B cells in blood and secondary lymphoid organs but whether B cells reside in non-lymphoid organs (NLO) in homeostasis is unknown. Here we identify, using intravenous labeling and parabiosis, a bona-fide tissue-resident B cell population in lung, liver, kidney and urinary bladder, a substantial proportion of which are B-1a cells. Tissue-resident B cells are present in neonatal tissues and also in germ-free mice NLOs, albeit in lower numbers than in specific pathogen-free mice and following co-housing with 'pet-store' mice. They spatially co-localise with macrophages and regulate their polarization and function, promoting an anti-inflammatory phenotype, in-part via interleukin-10 production, with effects on bacterial clearance during urinary tract infection. Thus, our data reveal a critical role for tissue-resident B cells in determining the homeostatic 'inflammatory set-point' of myeloid cells, with important consequences for tissue immunity.


Asunto(s)
Linfocitos B , Macrófagos , Ratones , Animales , Anticuerpos , Hígado , Pulmón
9.
Autoimmun Rev ; 22(2): 103236, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36436750

RESUMEN

Approximately 5% of the world-wide population is affected by autoimmune diseases. Overall, autoimmune diseases are still difficult to treat, impose a high burden on patients, and have a significant economic impact. Like other complex diseases, e.g., cancer, autoimmune diseases develop over several years. Decisive steps in the development of autoimmune diseases are (i) the development of autoantigen-specific lymphocytes and (often) autoantibodies and (ii) potentially clinical disease manifestation at a later stage. However, not all healthy individuals with autoantibodies develop disease manifestations. Identifying autoantibody-positive healthy individuals and monitoring and inhibiting their switch to inflammatory autoimmune disease conditions are currently in their infancy. The switch from harmless to inflammatory autoantigen-specific T and B-cell and autoantibody responses seems to be the hallmark for the decisive factor in inflammatory autoimmune disease conditions. Accordingly, biomarkers allowing us to predict this progression would have a significant impact. Several factors, such as genetics and the environment, especially diet, smoking, exposure to pollutants, infections, stress, and shift work, might influence the progression from harmless to inflammatory autoimmune conditions. To inspire research directed at defining and ultimately targeting autoimmune predisease, here, we review published evidence underlying the progression from health to autoimmune predisease and ultimately to clinically manifest inflammatory autoimmune disease, addressing the following 3 questions: (i) what is the current status, (ii) what is missing, (iii) and what are the future perspectives for defining and modulating autoimmune predisease.


Asunto(s)
Enfermedades Autoinmunes , Autoinmunidad , Humanos , Enfermedades Autoinmunes/etiología , Autoanticuerpos , Autoantígenos , Linfocitos
10.
J Autoimmun ; 39(3): 180-8, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22727274

RESUMEN

Antibodies contribute to the pathogenesis of many chronic inflammatory diseases, including autoimmune disorders and allergies. They are secreted by proliferating plasmablasts, short-lived plasma cells and non-proliferating, long-lived memory plasma cells. Memory plasma cells refractory to immunosuppression are critical for the maintenance of both protective and pathogenic antibody titers. Here, we studied the response of plasma cells in spleen, bone marrow and inflamed kidneys of lupus-prone NZB/W mice to high-dose dexamethasone and/or cyclophosphamide. BrdU+, dividing plasmablasts and short-lived plasma cells in the spleen were depleted while BrdU- memory plasma cells survived. In contrast, all bone marrow plasma cells including anti-DNA secreting cells were refractory to both drugs. Unlike bone marrow and spleen, which showed a predominance of IgM-secreting plasma cells, inflamed kidneys mainly accommodated IgG-secreting plasma cells, including anti-DNA secreting cells, some of which survived the treatments. These results indicate that the bone marrow is the major site of memory plasma cells resistant to treatment with glucocorticoids and anti-proliferative drugs, and that inflamed tissues and secondary lymphoid organs can contribute to the autoreactive plasma cell memory. Therefore, new strategies targeting autoreactive plasma cell memory should be considered. This could be the key to finding a curative approach to the treatment of chronic inflammatory autoantibody-mediated diseases.


Asunto(s)
Médula Ósea/inmunología , Ciclofosfamida/uso terapéutico , Dexametasona/uso terapéutico , Inmunosupresores/uso terapéutico , Nefritis Lúpica/tratamiento farmacológico , Células Plasmáticas/inmunología , Animales , Autoanticuerpos/biosíntesis , Autoanticuerpos/inmunología , Autoinmunidad/efectos de los fármacos , Médula Ósea/efectos de los fármacos , Médula Ósea/patología , Bromodesoxiuridina/administración & dosificación , Ciclofosfamida/farmacología , ADN/inmunología , Dexametasona/farmacología , Modelos Animales de Enfermedad , Femenino , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/inmunología , Inmunoglobulina M/biosíntesis , Inmunoglobulina M/inmunología , Memoria Inmunológica/efectos de los fármacos , Inmunosupresores/farmacología , Riñón/efectos de los fármacos , Riñón/inmunología , Riñón/patología , Nefritis Lúpica/inmunología , Nefritis Lúpica/patología , Ratones , Ratones Endogámicos NZB , Especificidad de Órganos , Células Plasmáticas/efectos de los fármacos , Células Plasmáticas/patología , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/patología
11.
Blood ; 116(11): 1867-75, 2010 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-20538807

RESUMEN

Long-lived plasma cells in the bone marrow produce memory antibodies that provide immune protection persisting for decades after infection or vaccination but can also contribute to autoimmune and allergic diseases. However, the composition of the microenvironmental niches that are important for the generation and maintenance of these cells is only poorly understood. Here, we demonstrate that, within the bone marrow, plasma cells interact with the platelet precursors (megakaryocytes), which produce the prominent plasma cell survival factors APRIL (a proliferation-inducing ligand) and IL-6 (interleukin-6). Accordingly, reduced numbers of immature and mature plasma cells are found in the bone marrow of mice deficient for the thrombopoietin receptor (c-mpl) that show impaired megakaryopoiesis. After immunization, accumulation of antigen-specific plasma cells in the bone marrow is disturbed in these mice. Vice versa, injection of thrombopoietin allows the accumulation and persistence of a larger number of plasma cells generated in the course of a specific immune response in wild-type mice. These results demonstrate that megakaryocytes constitute an important component of the niche for long-lived plasma cells in the bone marrow.


Asunto(s)
Células de la Médula Ósea/metabolismo , Megacariocitos/metabolismo , Células Plasmáticas/metabolismo , Nicho de Células Madre/metabolismo , Animales , Células de la Médula Ósea/citología , Comunicación Celular/efectos de los fármacos , Recuento de Células , Células Cultivadas , Femenino , Citometría de Flujo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Masculino , Megacariocitos/citología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Ovalbúmina/inmunología , Ovalbúmina/farmacología , Células Plasmáticas/citología , Células Plasmáticas/efectos de los fármacos , Receptores de Trombopoyetina/genética , Receptores de Trombopoyetina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Nicho de Células Madre/citología , Trombopoyetina/farmacología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo
12.
Int Arch Allergy Immunol ; 158(1): 43-53, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22212634

RESUMEN

BACKGROUND: Allergen-specific immunoglobulin (Ig) E initiates the effector cascade of allergic asthma and has been identified as a valuable target for therapeutic treatment of this disease. The proteasome inhibitor bortezomib was previously shown to deplete Ig-secreting plasma cells and to efficiently suppress Ig serum titers. The present study aimed at evaluating the therapeutic potential of the proteasome inhibitor bortezomib in allergic bronchial asthma. METHODS: To address this question, a chronic experimental asthma mouse model was used in a therapeutic setting. Mice were sensitized to ovalbumin (OVA) and challenged with OVA aerosol for 12 weeks. After 6 weeks of challenge, bortezomib treatment was started and continued for 1 week (short-term) or 6 weeks (long-term) with a dosage of 0.75 mg/kg body weight twice a week. Lung function, lung histology, Ig serum titers and plasma cell numbers were assessed. RESULTS: Whereas short-term treatment lowered bronchoalveolar lavage eosinophils, long-term treatment considerably reduced serum titers of anti-OVA IgE in mice with chronic experimental asthma. However, neither short-term nor long-term treatment significantly reduced plasma cell numbers, anti-OVA IgG1 serum titers or allergic airway inflammation or ablated airway hyperresponsiveness. CONCLUSION: Our results suggest that bortezomib treatment has only limited value as plasma cell-depleting therapy against allergic bronchial asthma.


Asunto(s)
Alérgenos/inmunología , Antiasmáticos/uso terapéutico , Asma/tratamiento farmacológico , Ácidos Borónicos/uso terapéutico , Inmunoglobulina E/inmunología , Pirazinas/uso terapéutico , Animales , Asma/inmunología , Asma/fisiopatología , Bortezomib , Enfermedad Crónica , Modelos Animales de Enfermedad , Eosinófilos/efectos de los fármacos , Eosinófilos/inmunología , Femenino , Inmunoglobulina E/sangre , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Pulmón/inmunología , Pulmón/patología , Pulmón/fisiopatología , Ratones , Ratones Endogámicos BALB C , Células Plasmáticas/efectos de los fármacos , Células Plasmáticas/inmunología , Insuficiencia del Tratamiento
13.
J Am Soc Nephrol ; 22(2): 336-48, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21233415

RESUMEN

Anti-neutrophil cytoplasmic autoantibodies (ANCA) cause vasculitis and necrotizing crescentic glomerulonephritis (NCGN). Steroids and cytotoxic drugs reduce mortality but can cause significant adverse events. The proteasome inhibitor bortezomib (BTZ) prevents glomerulonephritis in mouse models of lupus but its efficacy in ANCA-associated glomerulonephritis is unknown. We induced anti-MPO IgG-mediated NCGN by transplanting wild-type bone marrow (BM) into irradiated MPO-deficient mice immunized with MPO. Four weeks after BM transplantation, we treated mice with steroid/cyclophosphamide (S/CYC) or BTZ. Compared with untreated control mice, both S/CYC and BTZ significantly reduced urine abnormalities, NCGN, and infiltration of neutrophils and macrophages. Response to BTZ depended on timing of administration: BTZ abrogated NCGN if begun 3 weeks, but not 5 weeks, after BM transplantation. BTZ treatment significantly reduced total and MPO-specific plasma cells in both the spleen and bone marrow, resulting in significantly reduced anti-MPO titers. Furthermore, BTZ affected neither B cells nor total CD4 and CD8 T cells, including their naive and effector subsets. In contrast, S/CYC reduced the total number of cells in the spleen, including total and MPO-specific plasma cells and B cells. In contrast to BTZ, S/CYC did not affect total and MPO-specific plasma cells in the bone marrow. Three of 23 BTZ-treated mice died within 36 hours after BTZ administration. In summary, BTZ depletes MPO-specific plasma cells, reduces anti-MPO titers, and prevents NCGN in mice.


Asunto(s)
Anticuerpos Anticitoplasma de Neutrófilos/inmunología , Ácidos Borónicos/farmacología , Glomerulonefritis/prevención & control , Peroxidasa/inmunología , Células Plasmáticas/fisiología , Inhibidores de Proteasas/farmacología , Pirazinas/farmacología , Animales , Autoanticuerpos/sangre , Células de la Médula Ósea/fisiología , Ácidos Borónicos/toxicidad , Bortezomib , Ensayo de Immunospot Ligado a Enzimas , Glomerulonefritis/etiología , Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila , Pirazinas/toxicidad , Bazo/inmunología
14.
Mucosal Immunol ; 15(6): 1375-1388, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-36114245

RESUMEN

Mutated and unmutated IgE and IgG play different and partly opposing roles in allergy development, but the mechanisms controlling their relative production are incompletely understood. Here, we analyzed the IgE-response in murine food allergy. Deep sequencing of the complementary-determining region (CDR) repertoires indicated that an ongoing unmutated extrafollicular IgE response coexists with a germinal center response, even after long-lasting allergen challenges. Despite overall IgG1-dominance, a significant proportion of clonotypes contained several-fold more IgE than IgG1. Clonotypes with differential bias to either IgE or IgG1 showed distinct hypermutation and clonal expansion. Hypermutation rates were associated with different physiochemical binding properties of individual B-cell receptors (BCR). Increasing BCR signaling strength inhibited class switching from IgG1 to IgE in vitro, preferentially constraining IgE formation. These data indicate that antigen-binding properties of individual BCRs determine differential IgE hypermutation and IgE versus IgG1 production on the level of single B-cell clones.


Asunto(s)
Hipersensibilidad al Huevo , Ratones , Animales , Hipersensibilidad al Huevo/metabolismo , Inmunoglobulina G/metabolismo , Inmunoglobulina E/metabolismo , Receptores de Antígenos de Linfocitos B/genética , Linfocitos B , Alérgenos
15.
Blood ; 113(11): 2461-9, 2009 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-18987362

RESUMEN

Providing humoral immunity, antibody-secreting plasma cells and their immediate precursors, the plasmablasts, are generated in systemic and mucosal immune reactions. Despite their key role in maintaining immunity and immunopathology, little is known about their homeostasis. Here we show that plasmablasts and plasma cells are always detectable in human blood at low frequency in any unimmunized donor. In this steady state, 80% of plasmablasts and plasma cells express immunoglobulin A (IgA). Expression of a functional mucosal chemokine receptor, C-C motif receptor 10 (CCR10) and the adhesion molecule beta(7) integrin suggests that these cells come from mucosal immune reactions and can return to mucosal tissue. These blood-borne, CCR10(+) plasmablasts also are attracted by CXCL12. Approximately 40% of plasma cells in human bone marrow are IgA(+), nonmigratory, and express beta(7) integrin and CCR10, suggesting a substantial contribution of mucosal plasma cells to bone marrow resident, long-lived plasma cells. Six to 8 days after parenteral tetanus/diphtheria vaccination, intracellular IgG(+) cells appear in blood, both CD62L(+), beta(7) integrin(-), dividing, vaccine-specific, migratory plasmablasts and nondividing, nonmigratory, CD62L(-) plasma cells of different specificities. Systemic vaccination does not impact on peripheral IgA(+) plasmablast numbers, indicating that mucosal and systemic humoral immune responses are regulated independent of each other.


Asunto(s)
Diferenciación Celular/inmunología , Inmunidad Mucosa/fisiología , Células Plasmáticas/fisiología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Vacuna contra Difteria y Tétanos/farmacología , Humanos , Inmunidad Mucosa/efectos de los fármacos , Inmunoglobulina A/metabolismo , Cadenas beta de Integrinas/metabolismo , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/fisiología , Persona de Mediana Edad , Células Plasmáticas/citología , Células Plasmáticas/efectos de los fármacos , Células Plasmáticas/metabolismo , Receptores CCR10/metabolismo , Vacunación , Adulto Joven
16.
J Immunol ; 182(4): 2113-23, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19201864

RESUMEN

IL-6 and APRIL influence the growth, differentiation, and survival of normal and neoplastic Ab-forming cells (AFC). In this study, we identify two subsets of myeloid cells that associate with the AFC and are the main producers of these factors during a T-dependent Ab response to alum-precipitated protein in mouse lymph nodes. First CD11c(+)CD8alpha(-) dendritic cells located in the perivascular area of the T zone provide about half of the IL-6 mRNA produced in the node together with significant amounts of APRIL mRNA. The number of these cells increases during the response, at least in part due to local proliferation. The second subset comprises Gr1(+)CD11b(+)F4/80(+) monocyte/macrophages. These colonize the medullary cords during the response and are the other main IL-6 mRNA producers and the greatest source of APRIL mRNA. This medullary cord monocyte/macrophage subset results in local increase of APRIL mRNA that mirrors the polarity of CXCL12 expression in the node. The distribution of these myeloid cell subsets correlates with a gradient of AFC maturation assessed by progressive loss of Ki67 as AFC pass from the B cell follicle along the perivascular areas to the medullary cords.


Asunto(s)
Células Dendríticas/citología , Interleucina-6/inmunología , Leucocitos Mononucleares/citología , Ganglios Linfáticos/citología , Células Plasmáticas/citología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Traslado Adoptivo , Animales , Diferenciación Celular/inmunología , Proliferación Celular , Células Dendríticas/inmunología , Citometría de Flujo , Leucocitos Mononucleares/inmunología , Ganglios Linfáticos/inmunología , Subgrupos Linfocitarios/citología , Subgrupos Linfocitarios/inmunología , Macrófagos/citología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microdisección , Microscopía Confocal , Células Mieloides/citología , Células Mieloides/inmunología , Células Plasmáticas/inmunología , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
J Exp Med ; 199(11): 1577-84, 2004 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-15173206

RESUMEN

The current view holds that chronic autoimmune diseases are driven by the continuous activation of autoreactive B and T lymphocytes. However, despite the use of potent immunosuppressive drugs designed to interfere with this activation the production of autoantibodies often persists and contributes to progression of the immunopathology. In the present study, we analyzed the life span of (auto)antibody-secreting cells in the spleens of NZB x NZW F1 (NZB/W) mice, a murine model of systemic lupus erythematosus. The number of splenic ASCs increased in mice aged 1-5 mo and became stable thereafter. Less than 60% of the splenic (auto)antibody-secreting cells were short-lived plasmablasts, whereas 40% were nondividing, long-lived plasma cells with a half-life of >6 mo. In NZB/W mice and D42 Ig heavy chain knock-in mice, a fraction of DNA-specific plasma cells were also long-lived. Although antiproliferative immunosuppressive therapy depleted short-lived plasmablasts, long-lived plasma cells survived and continued to produce (auto)antibodies. Thus, long-lived, autoreactive plasma cells are a relevant target for researchers aiming to develop curative therapies for autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes/etiología , Células Plasmáticas/fisiología , Animales , Enfermedades Autoinmunes/inmunología , Bromodesoxiuridina/metabolismo , Enfermedad Crónica , ADN/inmunología , Femenino , Glicoproteínas de Membrana/análisis , Ratones , Ratones Endogámicos NZB , Ovalbúmina/inmunología , Proteoglicanos/análisis , Sindecanos
18.
J Allergy Clin Immunol ; 124(4): 819-26.e4, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19815119

RESUMEN

BACKGROUND: Allergen-specific IgE antibodies are responsible for the pathogenesis of type I hypersensitivity. In patients with allergy, IgE titers can persist in the apparent absence of allergen for years. Seasonal allergen exposure triggers clinical symptoms and enhances allergen-specific IgE. Whether allergen-specific plasma cells originating from seasonal allergen exposures can survive and become long-lived is so far unclear. OBJECTIVE: We analyzed the localization and lifetimes of allergen-specific IgE-secreting, IgA-secreting, and IgG(1)-secreting plasma cells after allergen inhalation in an ovalbumin-induced murine model of allergic asthma. METHODS: Ovalbumin-specific IgG(1)-secreting, IgA-secreting, and IgE-secreting cells in lungs, spleen, and bone marrow were isolated and tested for antibody secretion by the ELISpot technique. Longevity of ovalbumin-specific plasma cells was determined by cyclophosphamide treatment, which depletes proliferating plasmablasts but leaves plasma cells untouched. Ovalbumin aerosol-induced infiltrates in lungs were localized by confocal microscopy. RESULTS: Long-lived ovalbumin-specific plasma cells were generated by systemic sensitization and survived in bone marrow and spleen, maintaining systemic ovalbumin-specific titers of IgG, IgA, and IgE. On inhalation of ovalbumin-containing aerosol, sensitized mice developed airway inflammation and more ovalbumin-specific IgG(1)-secreting, IgA-secreting, and IgE-secreting cells in the lungs and in secondary lymphoid organs. These plasma cells joined the pool of ovalbumin-specific plasma cells in the bone marrow and became long-lived-that is, they are resistant to cyclophosphamide. Termination of ovalbumin inhalation depleted ovalbumin-specific plasma cells from the lungs, but they persisted in spleen and bone marrow. CONCLUSION: Our results show that inhalation of aerosolized allergen generates long-lived, allergen-specific IgG(1)-secreting, IgA-secreting, and IgE-secreting plasma cells that survive cytostatic treatment.


Asunto(s)
Alérgenos/inmunología , Asma/inmunología , Inmunoglobulina E/sangre , Células Plasmáticas/inmunología , Animales , Asma/metabolismo , Médula Ósea/inmunología , Médula Ósea/metabolismo , Ciclofosfamida/farmacología , Modelos Animales de Enfermedad , Femenino , Inmunidad Mucosa , Inmunoglobulina A/sangre , Inmunoglobulina G/sangre , Inmunosupresores/farmacología , Pulmón/inmunología , Pulmón/metabolismo , Ratones , Ratones Endogámicos BALB C , Ovalbúmina/inmunología , Células Plasmáticas/efectos de los fármacos , Bazo/inmunología , Bazo/metabolismo
19.
Front Immunol ; 11: 596772, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33362780

RESUMEN

Food allergies are common, costly and potentially life-threatening disorders. They are driven by Th2, but inhibited by Th1 reactions. There is also evidence indicating that IL-2 agonist treatment inhibits allergic sensitization through expansion of regulatory T cells. Here, we tested the impact of an IL-2 agonist in a novel model for food allergy to hen´s egg in mice sensitized without artificial adjuvants. Prophylactic IL-2 agonist treatment expanded Treg populations and inhibited allergen-specific sensitization. However, IL-2 agonist treatment of already sensitized mice increased mast cell responses and allergic anaphylaxis upon allergen re-challenge. These effects depended on allergen-specific IgE and were mediated through IFN-γ, as shown by IgE transfer and blockade of IFN-γ with monoclonal antibodies. These results suggest that although shifting the allergic reaction toward a Treg/Th1 response inhibits allergic sensitization, the prototypic Th1 cytokine IFN-γ promotes mast cell activation and allergen-induced anaphylaxis in individuals that are already IgE-sensitized. Hence, while a Th1 response can prevent the development of food allergy, IFN-γ has the ability to exacerbate already established food allergy.


Asunto(s)
Alérgenos/inmunología , Anafilaxia/etiología , Anafilaxia/metabolismo , Alimentos/efectos adversos , Interferón gamma/metabolismo , Interleucina-2/agonistas , Animales , Pollos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Clara de Huevo/efectos adversos , Femenino , Hipersensibilidad a los Alimentos/inmunología , Inmunización , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Ratones
20.
Curr Opin Immunol ; 18(3): 265-70, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16616478

RESUMEN

Contacts made with other cells and stroma have a major impact on proliferation, differentiation, survival, migration and immunoglobulin class switching of plasma cell precursors as well as on the lifespan of the antibody-secreting cells. Induction of tissue-specific chemokine receptors and adhesion molecules directs migratory plasma cell precursors to tissues close to those in which the original immune stimulation occurred. This mechanism focuses the production of specific antibodies within a particular type of tissue, thus providing a means for the most efficient protection against tissue-specific pathogens. Relocation does not apply to long-lived plasma cells responsible for sustained titers of high-affinity systemic antibody. These are formed in germinal centers and migrate to specific niches in the bone marrow that support their further differentiation and long-term survival.


Asunto(s)
Diferenciación Celular/inmunología , Células Plasmáticas/inmunología , Células del Estroma/inmunología , Animales , Supervivencia Celular/inmunología , Humanos , Modelos Inmunológicos , Células Plasmáticas/citología , Transducción de Señal/inmunología , Células del Estroma/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA