Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
J Control Release ; 359: 287-301, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37301267

RESUMEN

Although nebulizers have been developed for delivery of small molecules in human patients, no tunable device has been purpose-built for targeted delivery of modern large molecule and temperature-sensitive therapeutics to mice. Mice are used most of all species in biomedical research and have the highest number of induced models for human-relevant diseases and transgene models. Regulatory approval of large molecule therapeutics, including antibody therapies and modified RNA highlight the need for quantifiable dose delivery in mice to model human delivery, proof-of-concept studies, efficacy, and dose-response. To this end, we developed and characterized a tunable nebulization system composed of an ultrasonic transducer equipped with a mesh nebulizer fitted with a silicone restrictor plate modification to control the nebulization rate. We have identified the elements of design that influence the most critical factors to targeted delivery to the deep lungs of BALB/c mice. By comparing an in silico model of the mouse lung with experimental data, we were able to optimize and confirm the targeted delivery of over 99% of the initial volume to the deep portions of the mouse lung. The resulting nebulizer system provides targeted lung delivery efficiency far exceeding conventional nebulizers preventing waste of expensive biologics and large molecules during proof-of-concept and pre-clinical experiments involving mice. (Word Count =207).


Asunto(s)
Pulmón , Nebulizadores y Vaporizadores , Humanos , Animales , Ratones , Aerosoles , Administración por Inhalación , Sistemas de Liberación de Medicamentos/métodos , Diseño de Equipo
2.
Front Immunol ; 14: 1006741, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36798116

RESUMEN

Introduction: Anaphylaxis represents the most extreme and life-threatening form of allergic disease and is considered a medical emergency requiring immediate intervention. Additionally, some people with mastocytosis experience recurrent episodes of anaphylaxis during normal daily activities without exposure to known triggers. While acute therapy consists primarily of epinephrine and supportive care, chronic therapy relies mostly on desensitization and immunotherapy against the offending allergen, which is a time-consuming and sometimes unsuccessful process. These treatments also necessitate identification of the triggering allergen which is not always possible, and thus highlighting a need for alternative treatments for mast cell-mediated diseases. Methods: The exon-skipping oligonucleotide KitStop was administered to mice intradermally, intraperitoneally, or systemically at a dose of 12.5 mg/kg. Local mast cell numbers were enumerated via peritoneal lavage or skin histology, and passive systemic anaphylaxis was induced to evaluate KitStop's global systemic effect. A complete blood count and biochemistry panel were performed to assess the risk of acute toxicity following KitStop administration. Results: Here, we report the use of an exon-skipping oligonucleotide, which we have previously termed KitStop, to safely reduce the severity and duration of the anaphylactic response via mast cell depopulation in tissues. KitStop administration results in the integration of a premature stop codon within the mRNA transcript of the KIT receptor-a receptor tyrosine kinase found primarily on mast cells and whose gain-of-function mutation can lead to systemic mastocytosis. Following either local or systemic KitStop treatment, mice had significantly reduced mast cell numbers in the skin and peritoneum. In addition, KitStop-treated mice experienced a significantly diminished anaphylactic response using a model of passive systemic anaphylaxis when compared with control mice. Discussion: KitStop treatment results in a significant reduction in systemic mast cell responses, thus offering the potential to serve as a powerful additional treatment modality for patients that suffer from anaphylaxis.


Asunto(s)
Anafilaxia , Mastocitosis , Ratones , Animales , Mastocitos , Anafilaxia/genética , Anafilaxia/terapia , Alérgenos
3.
Allergy ; 78(5): 1204-1217, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36424895

RESUMEN

BACKGROUND: Allergic diseases are triggered by signaling through the high-affinity IgE receptor, FcεRI. In both mast cells (MCs) and basophils, FcεRI is a tetrameric receptor complex comprising a ligand-binding α subunit (FcεRIα), a tetraspan ß subunit (FcεRIß, MS4A2) responsible for trafficking and signal amplification, and a signal transducing dimer of single transmembrane γ subunits (FcεRIγ). However, FcεRI also exists as presumed trimeric complexes that lack FcεRIß and are expressed on several cell types outside the MC and basophil lineages. Despite known differences between humans and mice in the presence of the trimeric FcεRI complex, questions remain as to how it traffics and whether it signals in the absence of FcεRIß. We have previously reported that targeting FcεRIß with exon-skipping oligonucleotides eliminates IgE-mediated degranulation in mouse MCs, but equivalent targeting in human MCs was not effective at reducing degranulation. RESULTS: Here, we report that the FcεRIß-like protein MS4A6A exists in human MCs and compensates for FcεRIß in FcεRI trafficking and signaling. Human MS4A6A promotes surface expression of FcεRI complexes and facilitates degranulation. MS4A6A and FcεRIß are encoded by highly related genes within the MS4A gene family that cluster within the human gene loci 11q12-q13, a region linked to allergy and asthma susceptibility. CONCLUSIONS: Our data suggest the presence of either FcεRIß or MS4A6A is sufficient for degranulation, indicating that MS4A6A could be an elusive FcεRIß-like protein in human MCs that performs compensatory functions in allergic disease.


Asunto(s)
Hipersensibilidad , Receptores de IgE , Animales , Humanos , Ratones , Basófilos/metabolismo , Degranulación de la Célula , Exones , Hipersensibilidad/metabolismo , Mastocitos/metabolismo , Receptores de IgE/genética , Receptores de IgE/metabolismo , Transducción de Señal
4.
Front Allergy ; 3: 1029184, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36452260

RESUMEN

Eosinophilic esophagitis (EoE) is a chronic allergy-mediated condition with an increasing incidence in both children and adults. Despite EoE's strong impact on human health and welfare, there is a large unmet need for treatments with only one recently FDA-approved medication for EoE. The goal of this study was to establish swine as a relevant large animal model for translational biomedical research in EoE with the potential to facilitate development of therapeutics. We recently showed that after intraperitoneal sensitization and oral challenge with the food allergen hen egg white protein (HEWP), swine develop esophageal eosinophilia-a hallmark of human EoE. Herein, we used a similar sensitization and challenge treatment and evaluated immunological and pathological markers associated with human EoE. Our data demonstrate that the incorporated sensitization and challenge treatment induces (i) a systemic T-helper 2 and IgE response, (ii) a local expression of eotaxin-1 and other allergy-related immune markers, (iii) esophageal eosinophilia (>15 eosinophils/0.24 mm2), and (iv) esophageal endoscopic findings including linear furrows and white exudates. Thereby, we demonstrate that our sensitization and oral challenge protocol not only induces the underlying immune markers but also the micro- and macro-pathological hallmarks of human EoE. This swine model for EoE represents a novel relevant large animal model that can drive translational biomedical research to develop urgently needed treatment strategies for EoE.

5.
Int J Mol Sci ; 23(2)2022 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-35054974

RESUMEN

Mast cells are tissue-resident immune cells that function in both innate and adaptive immunity through the release of both preformed granule-stored mediators, and newly generated proinflammatory mediators that contribute to the generation of both the early and late phases of the allergic inflammatory response. Although mast cells can be activated by a vast array of mediators to contribute to homeostasis and pathophysiology in diverse settings and contexts, in this review, we will focus on the canonical setting of IgE-mediated activation and allergic inflammation. IgE-dependent activation of mast cells occurs through the high affinity IgE receptor, FcεRI, which is a multimeric receptor complex that, once crosslinked by antigen, triggers a cascade of signaling to generate a robust response in mast cells. Here, we discuss FcεRI structure and function, and describe established and emerging roles of the ß subunit of FcεRI (FcεRIß) in regulating mast cell function and FcεRI trafficking and signaling. We discuss current approaches to target IgE and FcεRI signaling and emerging approaches that could target FcεRIß specifically. We examine how alternative splicing of FcεRIß alters protein function and how manipulation of splicing could be employed as a therapeutic approach. Targeting FcεRI directly and/or IgE binding to FcεRI are promising approaches to therapeutics for allergic inflammation. The characteristic role of FcεRIß in both trafficking and signaling of the FcεRI receptor complex, the specificity to IgE-mediated activation pathways, and the preferential expression in mast cells and basophils, makes FcεRIß an excellent, but challenging, candidate for therapeutic strategies in allergy and asthma, if targeting can be realized.


Asunto(s)
Regulación de la Expresión Génica , Hipersensibilidad/etiología , Hipersensibilidad/metabolismo , Empalme del ARN , Receptores de IgE/genética , Receptores de IgE/metabolismo , Transducción de Señal , Empalme Alternativo , Animales , Biomarcadores , Degranulación de la Célula/genética , Degranulación de la Célula/inmunología , Susceptibilidad a Enfermedades , Humanos , Hipersensibilidad/diagnóstico , Hipersensibilidad/terapia , Mastocitos/inmunología , Mastocitos/metabolismo , Receptores de IgE/química , Relación Estructura-Actividad
6.
Mol Ther ; 30(1): 295-310, 2022 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-34371183

RESUMEN

Activating mutations in c-KIT are associated with the mast cell (MC) clonal disorders cutaneous mastocytosis and systemic mastocytosis and its variants, including aggressive systemic mastocytosis, MC leukemia, and MC sarcoma. Currently, therapies inhibiting KIT signaling are a leading strategy to treat MC proliferative disorders. However, these approaches may have off-target effects, and in some patients, complete remission or improved survival time cannot be achieved. These limitations led us to develop an approach using chemically stable exon skipping oligonucleotides (ESOs) that induce exon skipping of precursor (pre-)mRNA to alter gene splicing and introduce a frameshift into mature KIT mRNA transcripts. The result of this alternate approach results in marked downregulation of KIT expression, diminished KIT signaling, inhibition of MC proliferation, and rapid induction of apoptosis in neoplastic HMC-1.2 MCs. We demonstrate that in vivo administration of KIT targeting ESOs significantly inhibits tumor growth and systemic organ infiltration using both an allograft mastocytosis model and a humanized xenograft MC tumor model. We propose that our innovative approach, which employs well-tolerated, chemically stable oligonucleotides to target KIT expression through unconventional pathways, has potential as a KIT-targeted therapeutic alone, or in combination with agents that target KIT signaling, in the treatment of KIT-associated malignancies.


Asunto(s)
Mastocitos , Mastocitosis , Humanos , Mastocitos/metabolismo , Mastocitos/patología , Mastocitosis/genética , Mastocitosis/patología , Mastocitosis/terapia , Proteínas Proto-Oncogénicas c-kit/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo
7.
JCI Insight ; 6(21)2021 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-34546976

RESUMEN

Ozone is a highly reactive environmental pollutant with well-recognized adverse effects on lung health. Bronchial hyperresponsiveness (BHR) is one consequence of ozone exposure, particularly for individuals with underlying lung disease. Our data demonstrated that ozone induced substantial ATP release from human airway epithelia in vitro and into the airways of mice in vivo and that ATP served as a potent inducer of mast cell degranulation and BHR, acting through P2X7 receptors on mast cells. Both mast cell-deficient and P2X7 receptor-deficient (P2X7-/-) mice demonstrated markedly attenuated BHR to ozone. Reconstitution of mast cell-deficient mice with WT mast cells and P2X7-/- mast cells restored ozone-induced BHR. Despite equal numbers of mast cells in reconstituted mouse lungs, mice reconstituted with P2X7-/- mast cells demonstrated significantly less robust BHR than mice reconstituted with WT mast cells. These results support a model where P2X7 on mast cells and other cell types contribute to ozone-induced BHR.


Asunto(s)
Adenosina Trifosfato/metabolismo , Hiperreactividad Bronquial/metabolismo , Mastocitos/metabolismo , Ozono/efectos adversos , Animales , Femenino , Humanos , Ratones
8.
Tissue Eng Part A ; 27(15-16): 1008-1022, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33003982

RESUMEN

Mast cells (MCs) are pro-inflammatory tissue-resident immune cells that play a key role in inflammation. MCs circulate in peripheral blood as progenitors and undergo terminal differentiation in the tissue microenvironment where they can remain for many years. This in situ maturation results in tissue- and species-specific MC phenotypes, culminating in significant variability in response to environmental stimuli. There are many challenges associated with studying mature tissue-derived MCs, particularly in humans. In cases where cultured MCs are able to differentiate in two-dimensional in vitro cultures, there remains an inability for full maturation. Extracellular matrix (ECM) scaffolds provide for a more physiologically relevant environment for cells in vitro and have been shown to modulate the response of other immune cells such as T cells, monocytes, and macrophages. To improve current in vitro testing platforms of MCs and to assess future use of ECM scaffolds for MC regulation, we studied the in vitro response of human MCs cultured on decellularized porcine dermis hydrogels (dermis extracellular matrix hydrogel [dECM-H]). This study investigated the effect of dECM-H on cellular metabolic activity, cell viability, and receptor expression compared to collagen type I hydrogel (Collagen-H). Human MCs showed different metabolic activity when cultured in the dECM-H and also upregulated immunoglobulin E (IgE) receptors associated with MC maturation/activation compared to collagen type I. These results suggest an overall benefit in the long-term culture of human MCs in the dECM-H compared to Collagen-H providing important steps toward a model that is more representative of in vivo conditions. Graphical abstract [Formula: see text] Impact statement Mast cells (MCs) are difficult to culture in vitro as current culture conditions and substrates fail to promote similar phenotypic features observed in vivo. Extracellular matrix (ECM)-based biomaterials offer three-dimensional, tissue-specific environments that more closely resemble in vivo conditions. Our study explores the use of dermal ECM hydrogels for MC culture and shows significant upregulation of metabolic activity, cell viability, and gene expression of markers associated with MC maturation or activation compared to collagen type I-hydrogel and tissue culture plastic controls at 7 days. These results are among the first to describe MC behavior in response to ECM hydrogels.


Asunto(s)
Matriz Extracelular , Mastocitos , Animales , Diferenciación Celular , Colágeno , Humanos , Hidrogeles , Porcinos
9.
Tissue Eng Part B Rev ; 27(6): 590-603, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33164714

RESUMEN

Tissue engineers often use biomaterials to provide structural support along with mechanical and chemical signals to modulate the wound healing process. Biomaterials that are implanted into the body interact with a heterogeneous and dynamic inflammatory environment that is present at the site of injury. Whether synthetically derived, naturally derived, or a combination of both, it is important to assess biomaterials for their ability to modulate inflammation to understand their potential clinical use. One important, but underexplored cell in the context of biomaterials is the mast cell (MC). MCs are granulocytic leukocytes that engage in a variety of events in both the innate and adaptive immune systems. Although highly recognized for their roles in allergic reactions, MCs play an important role in wound healing by recognizing antigens through pattern recognition receptors and the high-affinity immunoglobulin E receptor (FceRI) and releasing granules that affect cell recruitment, fibrosis, extracellular matrix deposition, angiogenesis, and vasculogenesis. MCs also mediate the foreign body response, contributing to the incorporation or rejection of implants. Studies of MC-biomaterial interactions can aid in the elucidation of MC roles during the host tissue response and tissue repair. This review is designed for those in the tissue engineering and biomaterial fields who are interested in exploring the role MCs may play in wound-biomaterial interactions and wound healing. With this review, we hope to inspire more research in the MC-biomaterial space to accelerate the design and construction of optimized implants. Impact statement Mast cells (MCs) are highly specialized inflammatory cells that have crucial, but not fully understood, roles in wound healing and tissue repair. Upon stimulation, they recognize foreign antigens and release granules that help orchestrate the inflammatory response after tissue damage or biomaterial implantation. This review summarizes the current use of MCs in biomaterial research along with literature from the past decade focusing on MC interactions with materials used for tissue repair and regeneration. Studying MC-biomaterial interactions will help (i) further understand the process of inflammation and (ii) design biomaterials and tissue-engineered constructs for optimal repair and regeneration.


Asunto(s)
Materiales Biocompatibles , Mastocitos , Comunicación Celular , Fibrosis , Humanos , Mastocitos/patología , Ingeniería de Tejidos
10.
Cell Rep ; 31(1): 107472, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32268102

RESUMEN

Chronic allergic itch is a common symptom affecting millions of people and animals, but its pathogenesis is not fully explained. Herein, we show that periostin, abundantly expressed in the skin of patients with atopic dermatitis (AD), induces itch in mice, dogs, and monkeys. We identify the integrin αVß3 expressed on a subset of sensory neurons as the periostin receptor. Using pharmacological and genetic approaches, we inhibited the function of neuronal integrin αVß3, which significantly reduces periostin-induced itch in mice. Furthermore, we show that the cytokine TSLP, the application of AD-causing MC903 (calcipotriol), and house dust mites all induce periostin secretion. Finally, we establish that the JAK/STAT pathway is a key regulator of periostin secretion in keratinocytes. Altogether, our results identify a TSLP-periostin reciprocal activation loop that links the skin to the spinal cord via peripheral sensory neurons, and we characterize the non-canonical functional role of an integrin in itch.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Integrinas/metabolismo , Prurito/metabolismo , Animales , Moléculas de Adhesión Celular/fisiología , Dermatitis Atópica/etiología , Dermatitis Atópica/metabolismo , Dermatitis Atópica/patología , Perros , Femenino , Hipersensibilidad/fisiopatología , Integrina alfa5/metabolismo , Integrina beta3/metabolismo , Queratinocitos/metabolismo , Masculino , Ratones , Primates , Prurito/patología , Células Receptoras Sensoriales/metabolismo , Piel/metabolismo
11.
Cell Signal ; 71: 109617, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32240745

RESUMEN

Members of the membrane spanning 4A (MS4A) gene family are clustered around 11q12-13, a region linked to allergy and asthma susceptibility. Other than the known functions of FcεRIß (MS4A2) and CD20 (MS4A1) in mast cell and B cell signaling, respectively, functional studies for the remaining MS4A proteins are lacking. We thus explored whether MS4A4A, a mast cell expressed homologue of FcεRIß, has related functions to FcεRIß in FcεRI signaling. We establish in this study that MS4A4A promotes phosphorylation of PLCγ1, calcium flux and degranulation in response to IgE-mediated crosslinking of FcεRI. We previously demonstrated that MS4A4A promotes recruitment of KIT into caveolin-1-enriched microdomains and signaling through PLCγ1. Caveolin-1 itself is an important regulator of IgE-dependent store-operated Ca2+ entry (SOCE) and promotes expression of the store-operated Ca2+ channel pore-forming unit, Orai1. We thus further report that MS4A4A functions through interaction with caveolin-1 and recruitment of FcεRI and KIT into lipid rafts. In addition to proximal FcεRI signaling, we similarly show that MS4A4A regulates Orai1-mediated calcium entry downstream of calcium release from stores. Both MS4A4A and Orai1 had limited effects with compound 48/80 stimulation, demonstrating some degree of selectivity of both proteins to FcεRI receptor signaling over Mas-related G Protein coupled receptor X2 signaling. Overall, our data are consistent with the conclusion that MS4A4A performs a related function to the homologous FcεRIß to promote PLCγ1 signaling, SOCE, and degranulation through FcεRI in human mast cells and thus represents a new target in the regulation of IgE-mediated mast cell activation.


Asunto(s)
Calcio/metabolismo , Mastocitos/metabolismo , Proteínas de la Membrana/metabolismo , Receptores de IgE/metabolismo , Homología de Secuencia de Aminoácido , Transducción de Señal , Señalización del Calcio , Degranulación de la Célula , Línea Celular , Colesterol/metabolismo , Sangre Fetal/metabolismo , Humanos , Mastocitos/fisiología , Microdominios de Membrana/metabolismo , Proteína ORAI1/metabolismo , Fosfolipasa C gamma/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo
12.
J Biomed Mater Res B Appl Biomater ; 108(6): 2599-2609, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32100966

RESUMEN

Platelets crucially facilitate wound healing but can become depleted in traumatic injury or chronic wounds. Previously, our group developed injectable platelet-like particles (PLPs) comprised of highly deformable, ultralow crosslinked pNIPAm microgels (ULCs) coupled to fibrin binding antibodies to treat post-trauma bleeding. PLP fibrin-binding facilitates homing to sites of injury, promotes clot formation, and, due to high particle deformability, induces clot retraction. Clot retraction augments healing by increasing clot stability, enhancing clot stiffness, and promoting cell migration into the wound bed. Because post-traumatic healing is often complicated by infection, the objective of these studies was to develop antimicrobial nanosilver microgel composite PLPs to augment hemostasis, fight infection, and promote healing post-trauma. A key goal was to maintain particle deformability following silver incorporation to preserve PLP-mediated clot retraction. Clot retraction, antimicrobial activity, hemostasis after trauma, and healing after injury were evaluated via confocal microscopy, colony-forming unit assays, a murine liver trauma model, and a murine full-thickness injury model in the absence or presence of infection, respectively. We found that nanosilver incorporation does not affect base PLP performance while bestowing significant antimicrobial activity and enhancing infected wound healing outcomes. Therefore, Ag-PLPs have great promise for treating hemorrhage and improving healing following trauma.


Asunto(s)
Resinas Acrílicas/química , Antiinfecciosos/farmacología , Plaquetas , Nanopartículas del Metal , Plata/administración & dosificación , Animales , Antiinfecciosos/química , Infecciones Bacterianas/tratamiento farmacológico , Infecciones Bacterianas/microbiología , Retracción del Coagulo , Ensayo de Unidades Formadoras de Colonias , Fibrina/química , Fibrina/inmunología , Geles , Hemorragia/tratamiento farmacológico , Hemostasis/efectos de los fármacos , Hígado/lesiones , Masculino , Ratones , Ratones Endogámicos C57BL , Microgeles , Plata/química , Cicatrización de Heridas
13.
Methods Mol Biol ; 1828: 503-518, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30171564

RESUMEN

Mast cells are key effector cells in allergic inflammation and consequently are ideal targets for new therapeutics. The high-affinity IgE receptor complex, FcεRI, plays a critical role in mast cell and basophil activation by allergens to drive the immediate allergic inflammatory response. The ß subunit of FcεRI is critical for trafficking the FcεRI complex to the cell membrane and amplifies the FcεRI signaling cascade. We have utilized splice switching antisense oligonucleotides to force expression of a truncated isoform of FcεRIß, which we have shown does not associate with the FcεRI complex. This approach eliminates surface FcεRI expression in mast cells by targeting protein-protein interactions. Exon skipping has several therapeutic applications, and our findings demonstrate a novel application to alter receptor trafficking and dampen allergic inflammation. Here, we describe the methods of exon skipping in mast cells and the assays used to examine the responses of mast cells in vitro and in vivo.


Asunto(s)
Exones , Regulación de la Expresión Génica , Hipersensibilidad/genética , Empalme del ARN , Receptores de IgE/genética , Anafilaxia , Animales , Calcio/metabolismo , Señalización del Calcio , Degranulación de la Célula/inmunología , Predisposición Genética a la Enfermedad , Humanos , Hipersensibilidad/inmunología , Mastocitos/inmunología , Mastocitos/metabolismo , Ratones , Oligonucleótidos Antisentido/administración & dosificación , Oligonucleótidos Antisentido/genética , Transfección
14.
Proc Natl Acad Sci U S A ; 113(49): 14115-14120, 2016 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-27872312

RESUMEN

Allergic diseases are driven by activation of mast cells and release of mediators in response to IgE-directed antigens. However, there are no drugs currently available that can specifically down-regulate mast cell function in vivo when chronically administered. Here, we describe an innovative approach for targeting mast cells in vitro and in vivo using antisense oligonucleotide-mediated exon skipping of the ß-subunit of the high-affinity IgE receptor (FcεRIß) to eliminate surface high-affinity IgE receptor (FcεRI) expression and function, rendering mast cells unresponsive to IgE-mediated activation. As FcεRIß expression is restricted to mast cells and basophils, this approach would selectively target these cell types. Given the success of exon skipping in clinical trials to treat genetic diseases such as Duchenne muscular dystrophy, we propose that exon skipping of FcεRIß is a potential approach for mast cell-specific treatment of allergic diseases.


Asunto(s)
Degranulación de la Célula/efectos de los fármacos , Dermatitis Alérgica por Contacto/terapia , Mastocitos/efectos de los fármacos , Oligonucleótidos Antisentido/uso terapéutico , Empalme del ARN/efectos de los fármacos , Receptores de IgE/metabolismo , Animales , Calcio/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Femenino , Humanos , Mastocitos/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Oligonucleótidos Antisentido/farmacología , Anafilaxis Cutánea Pasiva/genética , Receptores de IgE/genética
15.
PLoS One ; 11(7): e0159177, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27459687

RESUMEN

Hermansky-Pudlak Syndrome type-1 (HPS-1) is an autosomal recessive disorder caused by mutations in HPS1 which result in reduced expression of the HPS-1 protein, defective lysosome-related organelle (LRO) transport and absence of platelet delta granules. Patients with HPS-1 exhibit oculocutaneous albinism, colitis, bleeding and pulmonary fibrosis postulated to result from a dysregulated immune response. The effect of the HPS1 mutation on human mast cells (HuMCs) is unknown. Since HuMC granules classify as LROs along with platelet granules and melanosomes, we set out to determine if HPS-1 cutaneous and CD34+ culture-derived HuMCs have distinct granular and cellular characteristics. Cutaneous and cultured CD34+-derived HuMCs from HPS-1 patients were compared with normal cutaneous and control HuMCs, respectively, for any morphological and functional differences. One cytokine-independent HPS-1 culture was expanded, cloned, designated the HP proMastocyte (HPM) cell line and characterized. HPS-1 and idiopathic pulmonary fibrosis (IPF) alveolar interstitium showed numerous HuMCs; HPS-1 dermal mast cells exhibited abnormal granules when compared to healthy controls. HPS-1 HuMCs showed increased CD63, CD203c and reduced mediator release following FcɛRI aggregation when compared with normal HuMCs. HPM cells also had the duplication defect, expressed FcɛRI and intracytoplasmic proteases and exhibited less mediator release following FcɛRI aggregation. HPM cells constitutively released IL-6, which was elevated in patients' serum, in addition to IL-8, fibronectin-1 (FN-1) and galectin-3 (LGALS3). Transduction with HPS1 rescued the abnormal HPM morphology, cytokine and matrix secretion. Microarray analysis of HPS-1 HuMCs and non-transduced HPM cells confirmed upregulation of differentially expressed genes involved in fibrogenesis and degranulation. Cultured HPS-1 HuMCs appear activated as evidenced by surface activation marker expression, a decrease in mediator content and impaired releasibility. The near-normalization of constitutive cytokine and matrix release following rescue by HPS1 transduction of HPM cells suggests that HPS-1 HuMCs may contribute to pulmonary fibrosis and constitute a target for therapeutic intervention.


Asunto(s)
Síndrome de Hermanski-Pudlak/diagnóstico , Síndrome de Hermanski-Pudlak/metabolismo , Mastocitos/metabolismo , Mastocitos/ultraestructura , Adulto , Biomarcadores , Estudios de Casos y Controles , Línea Celular , Células Cultivadas , Quimiotaxis , Análisis por Conglomerados , Matriz Extracelular/metabolismo , Femenino , Perfilación de la Expresión Génica , Síndrome de Hermanski-Pudlak/genética , Humanos , Mesilato de Imatinib/farmacología , Inmunofenotipificación , Pulmón/metabolismo , Pulmón/patología , Masculino , Mastocitos/efectos de los fármacos , Proteínas de la Membrana/genética , Persona de Mediana Edad , Mutación , Fenotipo , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología , Fibrosis Pulmonar/etiología , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Adulto Joven
16.
N Engl J Med ; 374(7): 656-63, 2016 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-26841242

RESUMEN

Patients with autosomal dominant vibratory urticaria have localized hives and systemic manifestations in response to dermal vibration, with coincident degranulation of mast cells and increased histamine levels in serum. We identified a previously unknown missense substitution in ADGRE2 (also known as EMR2), which was predicted to result in the replacement of cysteine with tyrosine at amino acid position 492 (p.C492Y), as the only nonsynonymous variant cosegregating with vibratory urticaria in two large kindreds. The ADGRE2 receptor undergoes autocatalytic cleavage, producing an extracellular subunit that noncovalently binds a transmembrane subunit. We showed that the variant probably destabilizes an autoinhibitory subunit interaction, sensitizing mast cells to IgE-independent vibration-induced degranulation. (Funded by the National Institutes of Health.).


Asunto(s)
Mutación Missense , Receptores Acoplados a Proteínas G/genética , Urticaria/genética , Vibración/efectos adversos , Biopsia , Degranulación de la Célula/genética , Femenino , Histamina/sangre , Humanos , Líbano , Masculino , Mastocitos/fisiología , Persona de Mediana Edad , Linaje , Receptores Acoplados a Proteínas G/metabolismo , Piel/patología , Urticaria/sangre , Urticaria/etiología
17.
Eur J Pharmacol ; 778: 125-38, 2016 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-25959386

RESUMEN

Mast cells are major effector cells of inflammation and there is strong evidence that mast cells play a significant role in asthma pathophysiology. There is also a growing body of evidence that mast cells contribute to other inflammatory and fibrotic lung diseases such as chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis. This review discusses the role that mast cells play in airway diseases and highlights how mast cell microlocalisation within specific lung compartments and their cellular interactions are likely to be critical for their effector function in disease.


Asunto(s)
Enfermedades Pulmonares Intersticiales/inmunología , Mastocitos/patología , Animales , Proliferación Celular , Humanos , Enfermedades Pulmonares Intersticiales/microbiología , Mastocitos/microbiología
18.
Mol Biol Cell ; 26(9): 1711-27, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25717186

RESUMEN

MS4A family members differentially regulate the cell cycle, and aberrant, or loss of, expression of MS4A family proteins has been observed in colon and lung cancer. However, the precise functions of MS4A family proteins and their mechanistic interactions remain unsolved. Here we report that MS4A4 facilitates trafficking of the receptor tyrosine kinase KIT through endocytic recycling rather than degradation pathways by a mechanism that involves recruitment of KIT to caveolin-1-enriched microdomains. Silencing of MS4A4 in human mast cells altered ligand-induced KIT endocytosis pathways and reduced receptor recycling to the cell surface, thus promoting KIT signaling in the endosomes while reducing that in the plasma membrane, as exemplified by Akt and PLCγ1 phosphorylation, respectively. The altered endocytic trafficking of KIT also resulted in an increase in SCF-induced mast cell proliferation and migration, which may reflect altered signaling in these cells. Our data reveal a novel function for MS4A family proteins in regulating trafficking and signaling, which could have implications in both proliferative and immunological diseases.


Asunto(s)
Clatrina/metabolismo , Mastocitos/metabolismo , Proteínas de la Membrana/fisiología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Antígenos CD20/fisiología , Línea Celular , Membrana Celular/metabolismo , Endocitosis , Endosomas/metabolismo , Expresión Génica , Humanos , Fosfolipasa C gamma/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Transducción de Señal , Proteínas de Unión al GTP rab5/metabolismo
19.
Methods Mol Biol ; 1220: 365-79, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25388263

RESUMEN

Mast cell activation is a central process in the initiation of allergic disorders. As described elsewhere in this volume, this process can be readily monitored by biochemical, antibody-based, and enzyme-based formats when the cell population examined is homogenous. When dealing with mixed and transfected cell populations however, such approaches may not be appropriate. Hence alternative methods are required. Here we describe flow-cytometry-based assays that can be utilized to examine signaling processes and degranulation in both pure mast cell populations and, following appropriate selection, in populations where the mast cells of interest may only represent a fraction of the total cell population.


Asunto(s)
Citometría de Flujo/métodos , Mastocitos/citología , Actinas/química , Calcio/metabolismo , Degranulación de la Célula , Proteínas Fluorescentes Verdes/genética , Humanos , Cinética , Mastocitos/metabolismo , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Transducción de Señal , Transfección
20.
J Immunol ; 193(12): 5924-32, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25378594

RESUMEN

Rictor is a regulatory component of the mammalian target of rapamycin (mTOR) complex 2 (mTORC2). We have previously demonstrated that rictor expression is substantially downregulated in terminally differentiated mast cells as compared with their immature or transformed counterparts. However, it is not known whether rictor and mTORC2 regulate mast cell activation. In this article, we show that mast cell degranulation induced by aggregation of high-affinity receptors for IgE (FcεRI) is negatively regulated by rictor independently of mTOR. We found that inhibition of mTORC2 by the dual mTORC1/mTORC2 inhibitor Torin1 or by downregulation of mTOR by short hairpin RNA had no impact on FcεRI-induced degranulation, whereas downregulation of rictor itself resulted in an increased sensitivity (∼50-fold) of cells to FcεRI aggregation with enhancement of degranulation. This was linked to a similar enhancement in calcium mobilization and cytoskeletal rearrangement attributable to increased phosphorylation of LAT and PLCγ1. In contrast, degranulation and calcium responses elicited by the G protein-coupled receptor ligand, C3a, or by thapsigargin, which induces a receptor-independent calcium signal, was unaffected by rictor knockdown. Overexpression of rictor, in contrast with knockdown, suppressed FcεRI-mediated degranulation. Taken together, these data provide evidence that rictor is a multifunctional signaling regulator that can regulate FcεRI-mediated degranulation independently of mTORC2.


Asunto(s)
Proteínas Portadoras/metabolismo , Degranulación de la Célula/inmunología , Mastocitos/inmunología , Mastocitos/metabolismo , Receptores de IgE/metabolismo , Actinas/metabolismo , Señalización del Calcio , Proteínas Portadoras/genética , Degranulación de la Célula/genética , Línea Celular , Activación Enzimática , Técnicas de Silenciamiento del Gen , Humanos , Agregado de Proteínas , Transporte de Proteínas , Interferencia de ARN , Proteína Asociada al mTOR Insensible a la Rapamicina , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA