Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Cell ; 168(6): 1041-1052.e18, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28283060

RESUMEN

Most secreted growth factors and cytokines are functionally pleiotropic because their receptors are expressed on diverse cell types. While important for normal mammalian physiology, pleiotropy limits the efficacy of cytokines and growth factors as therapeutics. Stem cell factor (SCF) is a growth factor that acts through the c-Kit receptor tyrosine kinase to elicit hematopoietic progenitor expansion but can be toxic when administered in vivo because it concurrently activates mast cells. We engineered a mechanism-based SCF partial agonist that impaired c-Kit dimerization, truncating downstream signaling amplitude. This SCF variant elicited biased activation of hematopoietic progenitors over mast cells in vitro and in vivo. Mouse models of SCF-mediated anaphylaxis, radioprotection, and hematopoietic expansion revealed that this SCF partial agonist retained therapeutic efficacy while exhibiting virtually no anaphylactic off-target effects. The approach of biasing cell activation by tuning signaling thresholds and outputs has applications to many dimeric receptor-ligand systems.


Asunto(s)
Anafilaxia/metabolismo , Células Madre Hematopoyéticas/inmunología , Mastocitos/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Transducción de Señal , Factor de Células Madre/metabolismo , Anafilaxia/inmunología , Animales , Dimerización , Humanos , Mastocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Ingeniería de Proteínas , Proteínas Proto-Oncogénicas c-kit/agonistas , Proteínas Proto-Oncogénicas c-kit/química , Factor de Células Madre/química , Factor de Células Madre/genética
2.
Nat Immunol ; 20(11): 1435-1443, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31591569

RESUMEN

Allergic skin diseases, such as atopic dermatitis, are clinically characterized by severe itching and type 2 immunity-associated hypersensitivity to widely distributed allergens, including those derived from house dust mites (HDMs). Here we found that HDMs with cysteine protease activity directly activated peptidergic nociceptors, which are neuropeptide-producing nociceptive sensory neurons that express the ion channel TRPV1 and Tac1, the gene encoding the precursor for the neuropeptide substance P. Intravital imaging and genetic approaches indicated that HDM-activated nociceptors drive the development of allergic skin inflammation by inducing the degranulation of mast cells contiguous to such nociceptors, through the release of substance P and the activation of the cationic molecule receptor MRGPRB2 on mast cells. These data indicate that, after exposure to HDM allergens, activation of TRPV1+Tac1+ nociceptor-MRGPRB2+ mast cell sensory clusters represents a key early event in the development of allergic skin reactions.


Asunto(s)
Alérgenos/inmunología , Dermatitis Atópica/inmunología , Mastocitos/inmunología , Nociceptores/inmunología , Pyroglyphidae/inmunología , Animales , Comunicación Celular/inmunología , Dermatitis Atópica/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Mastocitos/metabolismo , Ratones Noqueados , Nociceptores/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Piel/citología , Piel/inmunología , Canales Catiónicos TRPV/metabolismo , Taquicininas/genética , Taquicininas/metabolismo
3.
Immunity ; 53(4): 793-804.e9, 2020 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-32910906

RESUMEN

Allergies are considered to represent mal-directed type 2 immune responses against mostly innocuous exogenous compounds. Immunoglobulin E (IgE) antibodies are a characteristic feature of allergies and mediate hypersensitivity against allergens through activation of effector cells, particularly mast cells (MCs). Although the physiological functions of this dangerous branch of immunity have remained enigmatic, recent evidence shows that allergic immune reactions can help to protect against the toxicity of venoms. Because bacteria are a potent alternative source of toxins, we assessed the possible role of allergy-like type 2 immunity in antibacterial host defense. We discovered that the adaptive immune response against Staphylococcus aureus (SA) skin infection substantially improved systemic host defense against secondary SA infections in mice. Moreover, this acquired protection depended on IgE effector mechanisms and MCs. Importantly, our results reveal a previously unknown physiological function of allergic immune responses, IgE antibodies, and MCs in host defense against a pathogenic bacterium.


Asunto(s)
Inmunidad Adaptativa/inmunología , Inmunoglobulina E/inmunología , Mastocitos/inmunología , Infecciones Estafilocócicas/inmunología , Infecciones Cutáneas Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Alérgenos/inmunología , Animales , Femenino , Hipersensibilidad/inmunología , Hipersensibilidad/microbiología , Mastocitos/microbiología , Ratones , Ratones Endogámicos C57BL , Piel/inmunología , Piel/microbiología , Infecciones Estafilocócicas/microbiología , Infecciones Cutáneas Estafilocócicas/microbiología
5.
Allergy ; 77(2): 499-512, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-33840121

RESUMEN

BACKGROUND: In contrast to their clearly defined roles in allergic diseases, the physiologic functions of Immunoglobulin E antibodies (IgEs) and mast cells (MCs) remain enigmatic. Recent research supports the toxin hypothesis, showing that MCs and IgE-related type 2 immune responses can enhance host defense against certain noxious substances, including honeybee venom (BV). However, the mechanisms by which MCs can interfere with BV toxicity are unknown. In this study, we assessed the role of IgE and certain MC products in MC-mediated BV detoxification. METHODS: We applied in vitro and in vivo fluorescence microscopyimaging, and flow cytometry, fibroblast-based toxicity assays and mass spectrometry to investigate IgE-mediated detoxification of BV cytotoxicity by mouse and human MCs in vitro. Pharmacologic strategies to interfere with MC-derived heparin and proteases helped to define the importance of specific detoxification mechanisms. RESULTS: Venom-specific IgE increased the degranulation and cytokine responses of MCs to BV in vitro. Passive serum sensitization enhanced MC degranulation in vivo. IgE-activated mouse or human MCs exhibited enhanced potential for detoxifying BV by both proteolytic degradation and heparin-related interference with toxicity. Mediators released by IgE-activated human MCs efficiently degraded multiple BV toxins. CONCLUSIONS: Our results both reveal that IgE sensitization enhances the MC's ability to detoxify BV and also assign efficient toxin-neutralizing activity to MC-derived heparin and proteases. Our study thus highlights the potential importance of IgE, MCs, and particular MC products in defense against BV.


Asunto(s)
Venenos de Abeja , Mastocitos , Alérgenos/metabolismo , Animales , Degranulación de la Célula , Heparina/metabolismo , Humanos , Inmunoglobulina E , Ratones , Péptido Hidrolasas/metabolismo
6.
Immunol Rev ; 282(1): 35-46, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29431204

RESUMEN

Mast cells (MCs) are derived from committed precursors that leave the hematopoietic tissue, migrate in the blood, and colonize peripheral tissues where they terminally differentiate under microenvironment stimuli. They are distributed in almost all vascularized tissues where they act both as immune effectors and housekeeping cells, contributing to tissue homeostasis. Historically, MCs were classified into 2 subtypes, according to tryptic enzymes expression. However, MCs display a striking heterogeneity that reflects a complex interplay between different microenvironmental signals delivered by various tissues, and a differentiation program that decides their identity. Moreover, tissue-specific MCs show a trained memory, which contributes to shape their function in a specific microenvironment. In this review, we summarize the current state of our understanding of MC heterogeneity that reflects their different tissue experiences. We describe the discovery of unique cell molecules that can be used to distinguish specific MC subsets in vivo, and discuss how the improved ability to recognize these subsets provided new insights into the biology of MCs. These recent advances will be helpful for the understanding of the specific role of individual MC subsets in the control of tissue homeostasis, and in the regulation of pathological conditions such as infection, autoimmunity, and cancer.


Asunto(s)
Mastocitos/fisiología , Triptasas/metabolismo , Animales , Diferenciación Celular , Microambiente Celular , Homeostasis , Humanos , Inmunomodulación , Fenotipo
7.
Lab Invest ; 100(4): 516-526, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31857699

RESUMEN

Mast cell-deficient mice are widely used to identify and quantify contributions of mast cells to diverse biological responses in vivo, including allergic inflammation. However, despite the fact that scores of genes have been identified as modifiers of allergic inflammation, most mast cell-deficient models have been available only on a single genetic background. We transferred the KitW-sh allele onto the BALB/c background to generate BALB/c mast cell-deficient mice (BALB/c-KitW-sh/W-sh). BALB/c-KitW-sh/W-sh mice have dramatically reduced mast cell numbers (0-2% of wild type) in all tissues examined, as well as subtle hematologic differences from the corresponding wild type mice, including splenomegaly with evidence of increased splenic hematopoiesis. We examined in BALB/c-KitW-sh/W-sh mice models of allergic inflammation that are substantially diminished in C57BL/6-KitW-sh/W-sh mast cell-deficient mice. In a model of acute allergic inflammation, i.e., IgE-dependent passive cutaneous anaphylaxis, both ear swelling and leukocyte infiltration were largely or entirely absent in BALB/c-KitW-sh/W-sh mice. In contrast, in two different models of allergic airway inflammation, airway hyperresponsiveness, lung inflammation, and airway remodeling developed robustly in mast cell-deficient BALB/c-KitW-sh/W-sh mice. These results support the conclusion that the importance of mast cell contributions in various models of allergic inflammation may be at least partially determined by genetic background.


Asunto(s)
Asma , Modelos Animales de Enfermedad , Animales , Asma/inducido químicamente , Asma/patología , Asma/fisiopatología , Mastocitos/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-kit/genética
8.
J Allergy Clin Immunol ; 139(2): 584-596.e10, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27555460

RESUMEN

BACKGROUND: Conflicting results have been obtained regarding the roles of Fc receptors and effector cells in models of active systemic anaphylaxis (ASA). In part, this might reflect the choice of adjuvant used during sensitization because various adjuvants might differentially influence the production of particular antibody isotypes. OBJECTIVE: We developed an "adjuvant-free" mouse model of ASA and assessed the contributions of components of the "classical" and "alternative" pathways in this model. METHODS: Mice were sensitized intraperitoneally with ovalbumin at weekly intervals for 6 weeks and challenged intraperitoneally with ovalbumin 2 weeks later. RESULTS: Wild-type animals had immediate hypothermia and late-phase intraperitoneal inflammation in this model. These features were reduced in mice lacking the IgE receptor FcεRI, the IgG receptor FcγRIII or the common γ-chain FcRγ. FcγRIV blockade resulted in a partial reduction of inflammation without any effect on hypothermia. Depletion of monocytes/macrophages with clodronate liposomes significantly reduced the hypothermia response. By contrast, depletion of neutrophils or basophils had no significant effects in this ASA model. Both the hypothermia and inflammation were dependent on platelet-activating factor and histamine and were reduced in 2 types of mast cell (MC)-deficient mice. Finally, engraftment of MC-deficient mice with bone marrow-derived cultured MCs significantly exacerbated the hypothermia response and restored inflammation to levels similar to those observed in wild-type mice. CONCLUSION: Components of the classical and alternative pathways contribute to anaphylaxis in this adjuvant-free model, with key roles for MCs and monocytes/macrophages.


Asunto(s)
Anafilaxia/inmunología , Movimiento Celular , Hipotermia/inmunología , Leucocitos/inmunología , Macrófagos/inmunología , Mastocitos/inmunología , Adyuvantes Inmunológicos , Animales , Células Cultivadas , Vía Alternativa del Complemento , Vía Clásica del Complemento , Modelos Animales de Enfermedad , Humanos , Inmunización , Mastocitos/trasplante , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de IgE/genética , Receptores de IgE/metabolismo , Receptores de IgG/genética , Receptores de IgG/metabolismo
9.
J Allergy Clin Immunol ; 139(1): 269-280.e7, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27246523

RESUMEN

BACKGROUND: Animal models have demonstrated that allergen-specific IgG confers sensitivity to systemic anaphylaxis that relies on IgG Fc receptors (FcγRs). Mouse IgG2a and IgG2b bind activating FcγRI, FcγRIII, and FcγRIV and inhibitory FcγRIIB; mouse IgG1 binds only FcγRIII and FcγRIIB. Although these interactions are of strikingly different affinities, these 3 IgG subclasses have been shown to enable induction of systemic anaphylaxis. OBJECTIVE: We sought to determine which pathways control the induction of IgG1-, IgG2a-, and IgG2b-dependent passive systemic anaphylaxis. METHODS: Mice were sensitized with IgG1, IgG2a, or IgG2b anti-trinitrophenyl mAbs and challenged with trinitrophenyl-BSA intravenously to induce systemic anaphylaxis that was monitored by using rectal temperature. Anaphylaxis was evaluated in mice deficient for FcγRs injected with mediator antagonists or in which basophils, monocytes/macrophages, or neutrophils had been depleted. FcγR expression was evaluated on these cells before and after anaphylaxis. RESULTS: Activating FcγRIII is the receptor primarily responsible for all 3 models of anaphylaxis, and subsequent downregulation of this receptor was observed. These models differentially relied on histamine release and the contribution of mast cells, basophils, macrophages, and neutrophils. Strikingly, basophil contribution and histamine predominance in mice with IgG1- and IgG2b-induced anaphylaxis correlated with the ability of inhibitory FcγRIIB to negatively regulate these models of anaphylaxis. CONCLUSION: We propose that the differential expression of inhibitory FcγRIIB on myeloid cells and its differential binding of IgG subclasses controls the contributions of mast cells, basophils, neutrophils, and macrophages to IgG subclass-dependent anaphylaxis. Collectively, our results unravel novel complexities in the involvement and regulation of cell populations in IgG-dependent reactions in vivo.


Asunto(s)
Anafilaxia/inmunología , Inmunoglobulina G/inmunología , Subunidades de Proteína/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Femenino , Haptenos/inmunología , Histamina/inmunología , Inmunoglobulina E/inmunología , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Mieloides/inmunología , Receptores de IgG/genética , Receptores de IgG/inmunología , Albúmina Sérica Bovina/inmunología
10.
J Allergy Clin Immunol ; 137(1): 246-257.e11, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26410782

RESUMEN

BACKGROUND: Type 2 cytokine-related immune responses associated with development of antigen-specific IgE antibodies can contribute to pathology in patients with allergic diseases and to fatal anaphylaxis. However, recent findings in mice indicate that IgE also can enhance defense against honeybee venom. OBJECTIVE: We tested whether IgE antibodies, IgE-dependent effector mechanisms, and a local anaphylactic reaction to an unrelated antigen can enhance defense against Russell viper venom (RVV) and determined whether such responses can be influenced by immunization protocol or mouse strain. METHODS: We compared the resistance of RVV-immunized wild-type, IgE-deficient, and Fcer1a-deficient mice after injection of a potentially lethal dose of RVV. RESULTS: A single prior exposure to RVV enhanced the ability of wild-type mice, but not mice lacking IgE or functional FcεRI, to survive challenge with a potentially lethal amount of RVV. Moreover, IgE-dependent local passive cutaneous anaphylaxis in response to challenge with an antigen not naturally present in RVV significantly enhanced resistance to the venom. Finally, we observed different effects on resistance to RVV or honeybee venom in BALB/c versus C57BL/6 mice that had received a second exposure to that venom before challenge with a high dose of that venom. CONCLUSION: These observations illustrate the potential benefit of IgE-dependent effector mechanisms in acquired host defense against venoms. The extent to which type 2 immune responses against venoms can decrease pathology associated with envenomation seems to be influenced by the type of venom, the frequency of venom exposure, and the genetic background of the host.


Asunto(s)
Anafilaxia/inmunología , Inmunoglobulina E/inmunología , Receptores de IgE/inmunología , Venenos de Víboras/inmunología , Animales , Venenos de Abeja/inmunología , Degranulación de la Célula , Femenino , Inmunización , Mastocitos/inmunología , Mastocitos/fisiología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de IgE/genética
11.
Eur J Immunol ; 44(9): 2558-66, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25066089

RESUMEN

Mast cells regulate the immunological responses causing allergy and autoimmunity, and contribute to the tumor microenvironment through generation and secretion of a broad array of preformed, granule-stored and de novo synthesized bioactive compounds. The release and production of mast cell mediators is the result of a coordinated signaling machinery, followed by the FcεRI and FcγR antigen ligation. In this review, we present the latest understanding of FcεRI and FcγR signaling, required for the canonical mast cell activation during allergic responses and anaphylaxis. We then describe the cooperation between the signaling of FcR and other recently characterized membrane-bound receptors (i.e., IL-33R and thymic stromal lymphopoietin receptor) and their role in the chronic settings, where mast cell activation is crucial for the development and the sustainment of chronic diseases, such as asthma or airway inflammation. Finally, we report how the FcR activation could be used as a therapeutic approach to treat allergic and atopic diseases by mast cell inactivation. Understanding the magnitude and the complexity of mast cell signaling is necessary to identify the mechanisms underlying the potential effector and regulatory roles of mast cells in the biology and pathology of those disease settings in which mast cells are activated.


Asunto(s)
Anafilaxia/inmunología , Mastocitos/inmunología , Receptores de IgE/inmunología , Receptores de IgG/inmunología , Receptores de Interleucina/inmunología , Transducción de Señal/inmunología , Anafilaxia/patología , Animales , Humanos , Mastocitos/patología
12.
J Immunol ; 189(1): 120-7, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22649193

RESUMEN

The aryl hydrocarbon receptor (AhR) is a ligand-dependent transcription factor whose activity is modulated by xenobiotics as well as physiological ligands. These compounds may modulate inflammatory responses and contribute to the rising prevalence of allergic diseases observed in industrialized countries. Mast cells (MCs), located within tissues at the boundary of the external environment, represent a potential target of AhR ligands. In this study, we report that murine and human MCs constitutively express AhR, and its activation by the high-affinity ligand 6-formylindolo[3,2-b]carbazole (FICZ) determines a boost in degranulation. On the contrary, repeated exposure to FICZ inhibits MC degranulation. Accordingly, histamine release, in an in vivo passive systemic anaphylactic model, is exacerbated by a single dose and is attenuated by repetitive stimulation of AhR. FICZ-exposed MCs produce reactive oxygen species and IL-6 in response to cAMP-dependent signals. Moreover, AhR-activated MCs produce IL-17, a critical player in chronic inflammation and autoimmunity, suggesting a novel pathway for MC activation in the pathogenesis of these diseases. Indeed, histological analysis of patients with chronic obstructive pulmonary disease revealed an enrichment in AhR/IL-6 and AhR/IL-17 double-positive MCs within bronchial lamina propria. Thus, tissue-resident MCs could translate external chemical challenges through AhR by modulating allergic responses and contributing to the generation of inflammation-related diseases.


Asunto(s)
Degranulación de la Célula/inmunología , Mastocitos/inmunología , Mastocitos/metabolismo , Receptores de Hidrocarburo de Aril/fisiología , Anafilaxia/inmunología , Anafilaxia/metabolismo , Anafilaxia/patología , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Degranulación de la Célula/genética , Línea Celular , Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Humanos , Interleucina-17/biosíntesis , Interleucina-6/biosíntesis , Ligandos , Mastocitos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Hidrocarburo de Aril/deficiencia , Receptores de Hidrocarburo de Aril/metabolismo , Receptores de IgE/fisiología , Factores de Tiempo , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
13.
J Allergy Clin Immunol ; 130(3): 751-760.e2, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22564682

RESUMEN

BACKGROUND: The interaction of mast cells (MCs) with regulatory T cells through the OX40 ligand (OX40L):OX40 axis downregulates FcεRI-dependent immediate hypersensitivity responses both in vitro and in vivo. Little is known on OX40L-mediated intracellular signaling or on the mechanism by which OX40L engagement suppresses MC degranulation. OBJECTIVE: We explored the role of OX40L engagement on IgE/antigen-triggered MCs both in vitro and in vivo. METHODS: The soluble form of OX40 molecule was used to selectively trigger OX40L on MCs in vitro and was used to dissect OX40L contribution in an in vivo model of systemic anaphylaxis. RESULTS: OX40L:OX40 interaction led to the recruitment of C-terminal src kinase into lipid rafts, causing a preferential suppression of Fyn kinase activity and subsequent reduction in the phosphorylation of Gab2, the phosphatidylinositol 3-OH kinase regulatory subunit p85, and Akt, without affecting the Lyn pathway. Dampening of Fyn kinase activity also inhibited RhoA activation and microtubule nucleation, key regulators of MC degranulation. The in vivo administration of a blocking antibody to OX40L in wild-type mice caused enhanced immediate hypersensitivity, whereas the administration of soluble OX40 to regulatory T-cell-depleted or OX40-deficient mice reduced MC degranulation. CONCLUSIONS: The engagement of OX40L selectively suppresses Fyn-initiated signals required for MC degranulation and serves to limit immediate hypersensitivity. Our data suggest that soluble OX40 can restore the aberrant or absent regulatory T-cell activity, revealing a previously unappreciated homeostatic role for OX40L in setting the basal threshold of MC response.


Asunto(s)
Mastocitos/fisiología , Glicoproteínas de Membrana/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas c-fyn/fisiología , Receptores de IgE/fisiología , Factores de Necrosis Tumoral/fisiología , Proteínas de Unión al GTP rho/fisiología , Proteínas Adaptadoras Transductoras de Señales , Anafilaxia/etiología , Animales , Microdominios de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Microtúbulos/fisiología , Ligando OX40 , Fosfoproteínas/fisiología , Fosforilación , Proteína de Unión al GTP rhoA
14.
Eur J Immunol ; 41(7): 1872-82, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21509780

RESUMEN

The biological behavior of immune cells is determined by their intrinsic properties and interactions with other cell populations within their microenvironment. Several studies have confirmed the existence of tight spatial interactions between mast cells (MCs) and Tregs in different settings. For instance, we have recently identified the functional cross-talk between MCs and Tregs, through the OX40L-OX40 axis, as a new mechanism of reciprocal influence. However, there is scant information regarding the single-cell dynamics of this process. In this study, time-lapse video microscopy revealed direct interactions between Tregs and MCs in both murine and human cell co-cultures, resulting in the inhibition of the MC degranulation response. MCs incubated with WT, but not OX40-deficient, Tregs mediated numerous and long-lasting interactions and displayed different morphological features lacking the classical signs of exocytosis. MC degranulation and Ca2+ mobilization upon activation were inhibited by Tregs on a single-cell basis, without affecting overall cytokine secretion. Transmission electron microscopy showed ultrastructural evidence of vesicle-mediated secretion reconcilable with the morphological pattern of piecemeal degranulation. Our results suggest that MC morphological and functional changes following MC-Treg interactions can be ascribed to cell-cell contact and represent a transversal, non-species-specific mechanism of immune response regulation. Further research, looking at the molecular composition of this interaction will broaden our understanding of its contribution to immunity.


Asunto(s)
Comunicación Celular , Mastocitos/inmunología , Linfocitos T Reguladores/inmunología , Animales , Antígenos CD4/análisis , Calcio/metabolismo , Degranulación de la Célula , Línea Celular Tumoral , Técnicas de Cocultivo , Citocinas/metabolismo , Humanos , Subunidad alfa del Receptor de Interleucina-2/análisis , Mastocitos/fisiología , Mastocitos/ultraestructura , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión , Microscopía por Video , Ligando OX40/metabolismo , Receptores OX40/metabolismo , Análisis de la Célula Individual , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/ultraestructura
15.
JCI Insight ; 2(9)2017 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-28469089

RESUMEN

Contact hypersensitivity (CHS) is a common skin disease induced by epicutaneous sensitization to haptens. Conflicting results have been obtained regarding pathogenic versus protective roles of mast cells (MCs) in CHS, and this has been attributed in part to the limitations of certain models for studying MC functions in vivo. Here we describe a fluorescent imaging approach that enables in vivo selective labeling and tracking of MC secretory granules by real-time intravital 2-photon microscopy in living mice, and permits the identification of such MCs as a potential source of cytokines in different disease models. We show using this method that dermal MCs release their granules progressively into the surrounding microenvironment, but also represent an initial source of the antiinflammatory cytokine IL-10, during the early phase of severe CHS reactions. Finally, using 3 different types of MC-deficient mice, as well as mice in which IL-10 is ablated specifically in MCs, we show that IL-10 production by MCs can significantly limit the inflammation and tissue pathology observed in severe CHS reactions.

16.
J Exp Med ; 214(5): 1249-1258, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28385925

RESUMEN

Neutrophils have crucial antimicrobial functions but are also thought to contribute to tissue injury upon exposure to bacterial products, such as lipopolysaccharide (LPS). To study the role of neutrophils in LPS-induced endotoxemia, we developed a new mouse model, PMNDTR mice, in which injection of diphtheria toxin induces selective neutrophil ablation. Using this model, we found, surprisingly, that neutrophils serve to protect the host from LPS-induced lethal inflammation. This protective role was observed in conventional and germ-free animal facilities, indicating that it does not depend on a particular microbiological environment. Blockade or genetic deletion of myeloperoxidase (MPO), a key neutrophil enzyme, significantly increased mortality after LPS challenge, and adoptive transfer experiments confirmed that neutrophil-derived MPO contributes importantly to protection from endotoxemia. Our findings imply that, in addition to their well-established antimicrobial properties, neutrophils can contribute to optimal host protection by limiting the extent of endotoxin-induced inflammation in an MPO-dependent manner.


Asunto(s)
Endotoxemia/inmunología , Lipopolisacáridos/toxicidad , Neutrófilos/fisiología , Peroxidasa/fisiología , Animales , Anticuerpos/inmunología , Modelos Animales de Enfermedad , Endotoxemia/mortalidad , Ratones , Neutrófilos/enzimología , Sepsis/inmunología
17.
PLoS One ; 12(10): e0185704, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28982129

RESUMEN

Gouty arthritis is caused by the deposition of monosodium urate (MSU) crystals in joints. Despite many treatment options for gout, there is a substantial need for alternative treatments for patients unresponsive to current therapies. Tyrosine kinase inhibitors have demonstrated therapeutic benefit in experimental models of antibody-dependent arthritis and in rheumatoid arthritis in humans, but to date, the potential effects of such inhibitors on gouty arthritis has not been evaluated. Here we demonstrate that treatment with the tyrosine kinase inhibitor imatinib mesylate (imatinib) can suppress inflammation induced by injection of MSU crystals into subcutaneous air pouches or into the ankle joint of wild type mice. Moreover, imatinib treatment also largely abolished the lower levels of inflammation which developed in IL-1R1-/- or KitW-sh/W-sh mice, indicating that this drug can inhibit IL-1-independent pathways, as well as mast cell-independent pathways, contributing to pathology in this model. Imatinib treatment not only prevented ankle swelling and synovial inflammation when administered before MSU crystals but also diminished these features when administrated after the injection of MSU crystals, a therapeutic protocol more closely mimicking the clinical situation in which treatment occurs after the development of an acute gout flare. Finally, we also assessed the efficiency of local intra-articular injections of imatinib-loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles in this model of acute gout. Treatment with low doses of this long-acting imatinib:PLGA formulation was able to reduce ankle swelling in a therapeutic protocol. Altogether, these results raise the possibility that tyrosine kinase inhibitors might have utility in the treatment of acute gout in humans.


Asunto(s)
Artritis Gotosa/prevención & control , Mesilato de Imatinib/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Ácido Úrico/efectos adversos , Animales , Cristalización , Mesilato de Imatinib/administración & dosificación , Inyecciones Intraperitoneales , Ratones , Ratones Endogámicos C57BL , Inhibidores de Proteínas Quinasas/administración & dosificación , Ácido Úrico/química
18.
Nat Commun ; 7: 13696, 2016 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-27982078

RESUMEN

Asthma has multiple features, including airway hyperreactivity, inflammation and remodelling. The TNF superfamily member TNFSF14 (LIGHT), via interactions with the receptor TNFRSF14 (HVEM), can support TH2 cell generation and longevity and promote airway remodelling in mouse models of asthma, but the mechanisms by which TNFSF14 functions in this setting are incompletely understood. Here we find that mouse and human mast cells (MCs) express TNFRSF14 and that TNFSF14:TNFRSF14 interactions can enhance IgE-mediated MC signalling and mediator production. In mouse models of asthma, TNFRSF14 blockade with a neutralizing antibody administered after antigen sensitization, or genetic deletion of Tnfrsf14, diminishes plasma levels of antigen-specific IgG1 and IgE antibodies, airway hyperreactivity, airway inflammation and airway remodelling. Finally, by analysing two types of genetically MC-deficient mice after engrafting MCs that either do or do not express TNFRSF14, we show that TNFRSF14 expression on MCs significantly contributes to the development of multiple features of asthma pathology.


Asunto(s)
Asma/inducido químicamente , Asma/metabolismo , Mastocitos/fisiología , Receptores de IgE/metabolismo , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Remodelación de las Vías Aéreas (Respiratorias) , Animales , Anticuerpos , Antígenos Dermatofagoides/inmunología , Antígenos Dermatofagoides/toxicidad , Asma/patología , Líquido del Lavado Bronquioalveolar/citología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Genotipo , Inmunoglobulina E , Inmunoglobulina G , Ratones , Ratones Noqueados , Ovalbúmina/inmunología , Ovalbúmina/toxicidad , Receptores de IgE/genética , Miembro 14 de Receptores del Factor de Necrosis Tumoral/genética
19.
J Clin Invest ; 126(10): 3981-3998, 2016 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-27643442

RESUMEN

Mast cells (MCs) influence intercellular communication during inflammation by secreting cytoplasmic granules that contain diverse mediators. Here, we have demonstrated that MCs decode different activation stimuli into spatially and temporally distinct patterns of granule secretion. Certain signals, including substance P, the complement anaphylatoxins C3a and C5a, and endothelin 1, induced human MCs rapidly to secrete small and relatively spherical granule structures, a pattern consistent with the secretion of individual granules. Conversely, activating MCs with anti-IgE increased the time partition between signaling and secretion, which was associated with a period of sustained elevation of intracellular calcium and formation of larger and more heterogeneously shaped granule structures that underwent prolonged exteriorization. Pharmacological inhibition of IKK-ß during IgE-dependent stimulation strongly reduced the time partition between signaling and secretion, inhibited SNAP23/STX4 complex formation, and switched the degranulation pattern into one that resembled degranulation induced by substance P. IgE-dependent and substance P-dependent activation in vivo also induced different patterns of mouse MC degranulation that were associated with distinct local and systemic pathophysiological responses. These findings show that cytoplasmic granule secretion from MCs that occurs in response to different activating stimuli can exhibit distinct dynamics and features that are associated with distinct patterns of MC-dependent inflammation.


Asunto(s)
Degranulación de la Célula , Mastocitos/fisiología , Animales , Señalización del Calcio , Células Cultivadas , Quimiocinas/metabolismo , Gránulos Citoplasmáticos/metabolismo , Humanos , Inmunoglobulina E/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Sustancia P/fisiología
20.
J Clin Invest ; 126(12): 4497-4515, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27820702

RESUMEN

Epidermal keratinocytes form a structural and immune barrier that is essential for skin homeostasis. However, the mechanisms that regulate epidermal barrier function are incompletely understood. Here we have found that keratinocyte-specific deletion of the gene encoding RAB guanine nucleotide exchange factor 1 (RABGEF1, also known as RABEX-5) severely impairs epidermal barrier function in mice and induces an allergic cutaneous and systemic phenotype. RABGEF1-deficient keratinocytes exhibited aberrant activation of the intrinsic IL-1R/MYD88/NF-κB signaling pathway and MYD88-dependent abnormalities in expression of structural proteins that contribute to skin barrier function. Moreover, ablation of MYD88 signaling in RABGEF1-deficient keratinocytes or deletion of Il1r1 restored skin homeostasis and prevented development of skin inflammation. We further demonstrated that epidermal RABGEF1 expression is reduced in skin lesions of humans diagnosed with either atopic dermatitis or allergic contact dermatitis as well as in an inducible mouse model of allergic dermatitis. Our findings reveal a key role for RABGEF1 in dampening keratinocyte-intrinsic MYD88 signaling and sustaining epidermal barrier function in mice, and suggest that dysregulation of RABGEF1 expression may contribute to epidermal barrier dysfunction in allergic skin disorders in mice and humans. Thus, RABGEF1-mediated regulation of IL-1R/MYD88 signaling might represent a potential therapeutic target.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/inmunología , Queratinocitos/inmunología , Transducción de Señal/inmunología , Animales , Dermatitis por Contacto/genética , Dermatitis por Contacto/inmunología , Dermatitis por Contacto/patología , Epidermis/patología , Femenino , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Queratinocitos/patología , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , FN-kappa B/genética , FN-kappa B/inmunología , Receptores Tipo I de Interleucina-1/genética , Receptores Tipo I de Interleucina-1/inmunología , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA