Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
2.
J Allergy Clin Immunol ; 148(3): 889-894, 2021 09.
Article in English | MEDLINE | ID: mdl-33667475

ABSTRACT

BACKGROUND: Mastocytosis encompasses a heterogeneous group of disorders characterized by accumulation of clonal mast cells (MCs) in the skin and/or internal organs. Patients typically present with a broad variety of recurrent mediator-related clinical symptoms, including severe anaphylaxis. However, not all patients with mastocytosis experience anaphylactic reactions. OBJECTIVE: We sought to identify disease-specific biomarkers in plasma that could be used to predict patients with mastocytosis with increased risk of anaphylaxis. METHODS: Nineteen patients (≥18 years) and 2 control groups (11 subjects with allergic asthma and 13 healthy volunteers without history of atopy) were recruited. In total, 248 plasma proteins were analyzed by Proximity Extension Assay using Olink Proseek Multiplex panels. RESULTS: We identified 4 novel proteins, in addition to tryptase, E-selectin, adrenomedullin, T-cell immunoglobulin, and mucin domain 1, and CUB domain-containing protein 1/CD138 to be significantly increased in patients with mastocytosis compared with both patients with asthma and healthy controls. Furthermore, we investigated whether we could discriminate between patients with mastocytosis with or without anaphylaxis. In addition to tryptase, we identified 3 novel proteins, that is, allergin-1, pregnancy-associated plasma protein-A, and galectin-3, with significantly different levels in patients with mastocytosis with anaphylaxis compared with those without anaphylaxis. CONCLUSIONS: Newly identified proteomic biomarkers may be used to predict patients with mastocytosis with increased risk of anaphylaxis.


Subject(s)
Anaphylaxis/etiology , Blood Proteins/analysis , Mastocytosis/blood , Adult , Aged , Biomarkers/blood , Female , Humans , Male , Mastocytosis/complications , Mastocytosis/diagnosis , Middle Aged , Proteomics , Risk , Young Adult
4.
Oncotarget ; 8(6): 9647-9659, 2017 Feb 07.
Article in English | MEDLINE | ID: mdl-28038453

ABSTRACT

Systemic mastocytosis (SM) is a clonal bone marrow disorder, where therapeutical options are limited. Over 90% of the patients carry the D816V point mutation in the KIT receptor that renders this receptor constitutively active. We assessed the sensitivity of primary mast cells (MC) and mast cell lines HMC1.2 (D816V mutated), ROSA (KIT WT) and ROSA (KIT D816V) cells to histone deacetylase inhibitor (HDACi) treatment. We found that of four HDACi, suberoyl anilide hydroxamic acid (SAHA) was the most effective in killing mutated MC. SAHA downregulated KIT, followed by major MC apoptosis. Primary SM patient MC cultured ex vivo were even more sensitive to SAHA than HMC1.2 cells, whereas primary MC from healthy subjects were less affected. There was a correlation between cell death and SM disease severity, where cell death was more pronounced in the case of aggressive SM, with almost 100% cell death among MC from the mast cell leukemia patient. Additionally, ROSA (KIT D816V) was more affected by HDACi than ROSA (KIT WT) cells. Using ChIP qPCR, we found that the level of active chromatin mark H3K18ac/H3 decreased significantly in the KIT region. This epigenetic silencing was seen only in the KIT region and not in control genes upstream and downstream of KIT, indicating that the downregulation of KIT is exerted by specific epigenetic silencing. In conclusion, KIT D816V mutation sensitized MC to HDACi mediated killing, and SAHA may be of value as specific treatment for SM, although the specific mechanism of action requires further investigation.


Subject(s)
DNA Methylation/drug effects , Gene Silencing/drug effects , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Mast Cells/drug effects , Mastocytosis, Systemic/drug therapy , Mutation , Proto-Oncogene Proteins c-kit/genetics , Acetylation , Case-Control Studies , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Female , Histones/metabolism , Humans , Male , Mast Cells/enzymology , Mast Cells/pathology , Mastocytosis, Systemic/enzymology , Mastocytosis, Systemic/genetics , Mastocytosis, Systemic/pathology , Phosphorylation , Proto-Oncogene Proteins c-kit/metabolism , Time Factors , Vorinostat
5.
J Immunol ; 194(3): 1316-22, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25548219

ABSTRACT

Many forms of hypersensitivity reactions and allergic responses depend on deregulated mast cell activity. Several reports suggested that the antiapoptotic Bcl-2 family protein Bcl2a1/Bfl-1/A1 plays a critical role in mast cell survival upon activation. However, its in vivo relevance is poorly understood because of quadruplication of the Bcl2a1 gene locus in mice, hindering conventional knockout studies. In this study, we used a mouse model allowing traceable constitutive knockdown of all A1 isoforms expressed in the hematopoietic system by RNA interference. Knockdown of A1 reduced mast cell numbers in the skin and impaired connective tissue-like mast cell survival upon FcεRI-mediated activation in vitro. In contrast, A1 was dispensable for mucosa-like mast cell differentiation and survival. Moreover, knockdown of A1 prevented IgE-mediated passive systemic and cutaneous anaphylaxis in vivo. Our findings demonstrate that A1 is essential for the homeostasis of connective tissue mast cells, identifying A1 as a possible therapeutic target for therapy of certain types of mast cell-driven allergy symptoms.


Subject(s)
Anaphylaxis/genetics , Anaphylaxis/immunology , Proto-Oncogene Proteins c-bcl-2/genetics , Animals , Apoptosis/genetics , Apoptosis/immunology , Gene Expression , Gene Knockdown Techniques , Hematopoiesis/genetics , Immunoglobulin E/immunology , Mast Cells/immunology , Mast Cells/metabolism , Mice , Mice, Transgenic , Minor Histocompatibility Antigens , Promoter Regions, Genetic , Proto-Oncogene Proteins c-vav/genetics , RNA, Small Interfering/genetics , Receptors, IgE/metabolism
6.
J Immunol ; 192(12): 5852-62, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24829410

ABSTRACT

Exosomes, nano-sized membrane vesicles, are released by various cells and are found in many human body fluids. They are active players in intercellular communication and have immune-suppressive, immune-regulatory, and immune-stimulatory functions. EBV is a ubiquitous human herpesvirus that is associated with various lymphoid and epithelial malignancies. EBV infection of B cells in vitro induces the release of exosomes that harbor the viral latent membrane protein 1 (LMP1). LMP1 per se mimics CD40 signaling and induces proliferation of B lymphocytes and T cell-independent class-switch recombination. Constitutive LMP1 signaling within B cells is blunted through the shedding of LMP1 via exosomes. In this study, we investigated the functional effect of exosomes derived from the DG75 Burkitt's lymphoma cell line and its sublines (LMP1 transfected and EBV infected), with the hypothesis that they might mimic exosomes released during EBV-associated diseases. We show that exosomes released during primary EBV infection of B cells harbored LMP1, and similar levels were detected in exosomes from LMP1-transfected DG75 cells. DG75 exosomes efficiently bound to human B cells within PBMCs and were internalized by isolated B cells. In turn, this led to proliferation, induction of activation-induced cytidine deaminase, and the production of circle and germline transcripts for IgG1 in B cells. Finally, exosomes harboring LMP1 enhanced proliferation and drove B cell differentiation toward a plasmablast-like phenotype. In conclusion, our results suggest that exosomes released from EBV-infected B cells have a stimulatory capacity and interfere with the fate of human B cells.


Subject(s)
B-Lymphocytes/immunology , Burkitt Lymphoma/immunology , Cell Differentiation/immunology , Cell Proliferation , Exosomes/immunology , Immunoglobulin Class Switching/immunology , B-Lymphocytes/pathology , Burkitt Lymphoma/pathology , Cell Line, Tumor , Exosomes/pathology , Female , Humans , Male , Viral Matrix Proteins/immunology
7.
PLoS One ; 7(6): e39117, 2012.
Article in English | MEDLINE | ID: mdl-22720045

ABSTRACT

Mast cells are best known for their role in allergic reactions, where aggregation of FcεRI leads to the release of mast cell mediators causing allergic symptoms. The activation also induces a survival program in the cells, i.e., activation-induced mast cell survival. The aim of the present study was to investigate how the activation-induced survival is mediated. Cord blood-derived mast cells and the mast cell line LAD-2 were activated through FcεRI crosslinking, with or without addition of chemicals that inhibit the activity or expression of selected Bcl-2 family members (ABT-737; roscovitine). Cell viability was assessed using staining and flow cytometry. The expression and function of Bcl-2 family members BFL-1 and MCL-1 were investigated using real-time quantitative PCR and siRNA treatment. The mast cell expression of Bfl-1 was investigated in skin biopsies. FcεRI crosslinking promotes activation-induced survival of human mast cells and this is associated with an upregulation of the anti-apoptotic Bcl-2 family member Bfl-1. ABT-737 alone or in combination with roscovitine decreases viability of human mast cells although activation-induced survival is sustained, indicating a minor role for Bcl-X(L), Bcl-2, Bcl-w and Mcl-1. Reducing BFL-1 but not MCL-1 levels by siRNA inhibited activation-induced mast cell survival. We also demonstrate that mast cell expression of Bfl-1 is elevated in birch-pollen-provocated skin and in lesions of atopic dermatitis and psoriasis patients. Taken together, our results highlight Bfl-1 as a major effector in activation-induced human mast cell survival.


Subject(s)
Cell Survival/physiology , Mast Cells/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , Flow Cytometry , Humans , Minor Histocompatibility Antigens
8.
Mamm Genome ; 22(5-6): 353-60, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21465164

ABSTRACT

The syndrome Multiple Congenital Ocular Anomalies (MCOA) is the collective name ascribed to heritable congenital eye defects in horses. Individuals homozygous for the disease allele (MCOA phenotype) have a wide range of eye anomalies, while heterozygous horses (Cyst phenotype) predominantly have cysts that originate from the temporal ciliary body, iris, and/or peripheral retina. MCOA syndrome is highly prevalent in the Rocky Mountain Horse but the disease is not limited to this breed. Affected horses most often have a Silver coat color; however, a pleiotropic link between these phenotypes is yet to be proven. Locating and possibly isolating these traits would provide invaluable knowledge to scientists and breeders. This would favor maintenance of a desirable coat color while addressing the health concerns of the affected breeds, and would also provide insight into the genetic basis of the disease. Identical-by-descent mapping was used to narrow the previous 4.6-Mb region to a 264-kb interval for the MCOA locus. One haplotype common to four breeds showed complete association to the disease (Cyst phenotype, n = 246; MCOA phenotype, n = 83). Candidate genes from the interval, SMARCC2 and IKZF4, were screened for polymorphisms and genotyped, and segregation analysis allowed the MCOA syndrome region to be shortened to 208 kb. This interval also harbors PMEL17, the gene causative for Silver coat color. However, by shortening the MCOA locus by a factor of 20, 176 other genes have been unlinked from the disease and only 15 genes remain.


Subject(s)
Chromosomes, Mammalian/genetics , Eye Abnormalities/veterinary , Genetic Loci/genetics , Horse Diseases/genetics , Animals , Base Sequence , Chromosome Mapping , Eye Abnormalities/genetics , Genotype , Haplotypes/genetics , Horses , Microsatellite Repeats/genetics , Molecular Sequence Data , Polymorphism, Single Nucleotide/genetics , Sequence Analysis, DNA , Species Specificity , Syndrome , Transcription Factors/genetics
9.
J Immunol ; 186(4): 2523-8, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21239713

ABSTRACT

In response to cell injury, caused, for example, by trauma, several processes must be initiated simultaneously to achieve an acute inflammatory response designed to prevent sustained tissue damage and infection and to restore and maintain tissue homeostasis. Detecting cell injury is facilitated by the fact that damaged cells release intracellular molecules not normally present in the extracellular space. However, potential underlying mechanisms for the recognition of endogenous danger signals released upon cell injury have yet to be elucidated. In this study, we demonstrate that mast cells, potent promoters of acute inflammation, play a key role in responding to cell injury by recognizing IL-33 released from necrotic structural cells. In an in vitro model of cell injury, this recognition was shown to involve the T1/ST2 receptor and result in the secretion of proinflammatory leukotrienes and cytokines by mouse mast cells. Remarkably, of all of the components released upon necrosis, our results show that IL-33 alone is a key component responsible for initiating proinflammatory responses in mast cells reacting to cell injury. Our findings identify IL-33 as a key danger signal released by necrotic structural cells capable of activating mast cells, thus providing novel insights concerning the role of mast cells as sensors of cell injury.


Subject(s)
Interleukins/metabolism , Mast Cells/immunology , Mast Cells/pathology , Animals , Cell-Free System/immunology , Cell-Free System/metabolism , Cell-Free System/pathology , Cells, Cultured , Cytokines/metabolism , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Interleukin-33 , Interleukins/deficiency , Interleukins/genetics , Leukotrienes/metabolism , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Necrosis
SELECTION OF CITATIONS
SEARCH DETAIL
...