Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
1.
Exp Mol Med ; 56(6): 1331-1339, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38825637

ABSTRACT

Interleukin-9 (IL-9) is a multifunctional cytokine with roles in a broad cross-section of human diseases. Like many cytokines, IL-9 is transcriptionally regulated by a group of noncoding regulatory elements (REs) surrounding the IL9 gene. These REs modulate IL-9 transcription by forming 3D loops that recruit transcriptional machinery. IL-9-promoting transcription factors (TFs) can bind REs to increase locus accessibility and permit chromatin looping, or they can be recruited to already accessible chromatin to promote transcription. Ample mechanistic and genome-wide association studies implicate this interplay between IL-9-modulating TFs and IL9 cis-REs in human physiology, homeostasis, and disease.


Subject(s)
Epigenesis, Genetic , Interleukin-9 , Humans , Interleukin-9/genetics , Interleukin-9/metabolism , Animals , Gene Expression Regulation , Chromatin/metabolism , Chromatin/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Regulatory Sequences, Nucleic Acid , Disease Susceptibility
2.
J Extracell Biol ; 3(1): e139, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38938682

ABSTRACT

The receptor tyrosine kinase (RTK) KIT and its ligand stem cell factor (SCF) are essential for human mast cell (huMC) survival and proliferation. HuMCs expressing oncogenic KIT variants secrete large numbers of extracellular vesicles (EVs). The role KIT plays in regulating EV secretion has not been examined. Here, we investigated the effects of stimulation or inhibition of KIT activity on the secretion of small EVs (sEVs). In huMCs expressing constitutively active KIT, the quantity and quality of secreted sEVs positively correlated with the activity status of KIT. SCF-mediated stimulation of KIT in huMCs or murine MCs, or of transiently expressed KIT in HeLa cells, enhanced the release of sEVs expressing exosome markers. In contrast, ligand-mediated stimulation of the RTK EGFR in HeLa cells did not affect sEV secretion. The release of sEVs induced by either constitutively active or ligand-activated KIT was remarkably decreased when cells were treated with KIT inhibitors, concomitant with reduced exosome markers in sEVs. Similarly, inhibition of oncogenic KIT signalling kinases like PI3K, and MAPK significantly reduced the secretion of sEVs. Thus, activation of KIT and its early signalling cascades stimulate the secretion of exosome-like sEVs in a regulated fashion, which may have implications for KIT-driven functions.

6.
Nat Immunol ; 24(6): 1036-1048, 2023 06.
Article in English | MEDLINE | ID: mdl-37106040

ABSTRACT

Allergic diseases are a major global health issue. Interleukin (IL)-9-producing helper T (TH9) cells promote allergic inflammation, yet TH9 cell effector functions are incompletely understood because their lineage instability makes them challenging to study. Here we found that resting TH9 cells produced IL-9 independently of T cell receptor (TCR) restimulation, due to STAT5- and STAT6-dependent bystander activation. This mechanism was seen in circulating cells from allergic patients and was restricted to recently activated cells. STAT5-dependent Il9/IL9 regulatory elements underwent remodeling over time, inactivating the locus. A broader 'allergic TH9' transcriptomic and epigenomic program was also unstable. In vivo, TH9 cells induced airway inflammation via TCR-independent, STAT-dependent mechanisms. In allergic patients, TH9 cell expansion was associated with responsiveness to JAK inhibitors. These findings suggest that TH9 cell instability is a negative checkpoint on bystander activation that breaks down in allergy and that JAK inhibitors should be considered for allergic patients with TH9 cell expansion.


Subject(s)
Hypersensitivity , Janus Kinase Inhibitors , Humans , Interleukin-9/genetics , T-Lymphocytes, Helper-Inducer , STAT5 Transcription Factor/genetics , Chromatin/genetics , Inflammation , Hypersensitivity/genetics , Cell Differentiation , STAT6 Transcription Factor
7.
Front Immunol ; 14: 1078958, 2023.
Article in English | MEDLINE | ID: mdl-37025992

ABSTRACT

The HMC-1.2 human mast cell (huMC) line is often employed in the study of attributes of neoplastic huMCs as found in patients with mastocytosis and their sensitivity to interventional drugs in vitro and in vivo. HMC-1.2 cells express constitutively active KIT, an essential growth factor receptor for huMC survival and function, due to the presence of two oncogenic mutations (D816V and V560G). However, systemic mastocytosis is commonly associated with a single D816V-KIT mutation. The functional consequences of the coexisting KIT mutations in HMC-1.2 cells are unknown. We used CRISPR/Cas9-engineering to reverse the V560G mutation in HMC-1.2 cells, resulting in a subline (HMC-1.3) with a single mono-allelic D816V-KIT variant. Transcriptome analyses predicted reduced activity in pathways involved in survival, cell-to-cell adhesion, and neoplasia in HMC-1.3 compared to HMC-1.2 cells, with differences in expression of molecular components and cell surface markers. Consistently, subcutaneous inoculation of HMC-1.3 into mice produced significantly smaller tumors than HMC-1.2 cells, and in colony assays, HMC-1.3 formed less numerous and smaller colonies than HMC-1.2 cells. However, in liquid culture conditions, the growth of HMC-1.2 and HMC-1.3 cells was comparable. Phosphorylation levels of ERK1/2, AKT and STAT5, representing pathways associated with constitutive oncogenic KIT signaling, were also similar between HMC-1.2 and HMC-1.3 cells. Despite these similarities in liquid culture, survival of HMC-1.3 cells was diminished in response to various pharmacological inhibitors, including tyrosine kinase inhibitors used clinically for treatment of advanced systemic mastocytosis, and JAK2 and BCL2 inhibitors, making HMC-1.3 more susceptible to these drugs than HMC-1.2 cells. Our study thus reveals that the additional V560G-KIT oncogenic variant in HMC-1.2 cells modifies transcriptional programs induced by D816V-KIT, confers a survival advantage, alters sensitivity to interventional drugs, and increases the tumorigenicity, suggesting that engineered huMCs with a single D816V-KIT variant may represent an improved preclinical model for mastocytosis.


Subject(s)
Mastocytosis, Systemic , Mastocytosis , Humans , Animals , Mice , Mastocytosis, Systemic/drug therapy , Mastocytosis, Systemic/genetics , Mastocytosis, Systemic/pathology , CRISPR-Cas Systems , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Mastocytosis/genetics , Mutation , Cell Line
8.
J Extracell Vesicles ; 11(10): e12272, 2022 09.
Article in English | MEDLINE | ID: mdl-36239715

ABSTRACT

Activating mutations in the receptor KIT promote the dysregulated proliferation of human mast cells (huMCs). The resulting neoplastic huMCs secrete extracellular vesicles (EVs) that can transfer oncogenic KIT among other cargo into recipient cells. Despite potential contributions to diseases, KIT-containing EVs have not been thoroughly investigated. Here, we isolated and characterized KIT-EV subpopulations released by neoplastic huMCs using an immunocapture approach that selectively isolates EVs containing KIT in its proper topology. Immunocapture of EVs on KIT antibody-coated electron microscopy (EM) affinity grids allowed to assess the morphology and size of KIT-EVs. Immunoblot analysis demonstrated KIT-EVs have a distinct protein profile from KIT-depleted EVs, contain exosome and microvesicle markers, and are separated into these subtypes by ultracentrifugation. Cell treatment with sphingomyelinase inhibitors shifted the protein content among KIT-EV subtypes, suggesting different biogenesis routes. Proteomic analysis revealed huMC KIT-EVs are enriched in proteins involved in signalling, immune responses, and cell migration, suggesting diverse biological functions, and indicated neoplastic huMCs disseminate KIT via shuttling in heterogeneous microvesicle- and exosome-like EVs. Further, selective KIT-immunocapture will enable the enrichment of specific huMC-derived EVs from complex human biosamples and facilitate an understanding of their in vivo functions and potential to serve as biomarkers of specific biological pathologies.


Subject(s)
Exosomes , Extracellular Vesicles , Biomarkers/metabolism , Exosomes/metabolism , Extracellular Vesicles/metabolism , Humans , Mast Cells/metabolism , Proteomics , Sphingomyelin Phosphodiesterase/metabolism
9.
Mol Ther ; 30(1): 295-310, 2022 01 05.
Article in English | MEDLINE | ID: mdl-34371183

ABSTRACT

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.


Subject(s)
Mast Cells , Mastocytosis , Humans , Mast Cells/metabolism , Mast Cells/pathology , Mastocytosis/genetics , Mastocytosis/pathology , Mastocytosis/therapy , Proto-Oncogene Proteins c-kit/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/metabolism
10.
Parasite Immunol ; 43(4): e12820, 2021 04.
Article in English | MEDLINE | ID: mdl-33434287

ABSTRACT

The goal of this study was to analyse the effects of a protein-deficient (PD) diet on antibody-dependent cell-mediated cytotoxicity (ADCC) in vitro against newborn larvae (NBL) of Trichinella spiralis in the lungs of infected rats. Two groups of weaning Wistar rats received a PD diet (6.5% casein) and other two received a control diet (C, 20% casein). After ten days, one group of each diet was infected (PDI and CI ) with muscle larvae. Lung tissue extracts (LTE) and lung cell suspension (LCS) were obtained. PDI had lower titres of anti-NBL antibodies in LTE than CI . In ADCC assays using control cells, NBL mortality percentage was lower with LTE from PDI than LTE from CI (P < .01). In assays using control cytotoxic sera, ADCC was exerted by LCS from CI at all days post-infection (p.i.), but only by LCS from 13 days p.i. from PDI . ADCC assays combining LTE and LCS from the same group showed a lower response for PDI than for CI (P < .0001). LCS from PDI contained lower numbers of neutrophils, eosinophils and FcεRI+ cells than CI . PD may diminish ADCC activity against T spiralis NBL in lungs through alterations in specific antibodies and effector cells.


Subject(s)
Lung/immunology , Protein Deficiency/complications , Trichinella spiralis , Trichinellosis/complications , Animals , Antibodies, Helminth/blood , Antigens, Helminth/immunology , Female , Larva , Lung/parasitology , Rats , Rats, Wistar , Trichinella spiralis/immunology , Weaning
11.
Pharmacol Ther ; 220: 107718, 2021 04.
Article in English | MEDLINE | ID: mdl-33130192

ABSTRACT

Mast cells are tissue-resident immune cells that play key roles in the initiation and perpetuation of allergic inflammation, usually through IgE-mediated mechanisms. Mast cells are, however, evolutionary ancient immune cells that can be traced back to urochordates and before the emergence of IgE antibodies, suggesting their involvement in antibody-independent biological functions, many of which are still being characterized. Herein, we summarize recent advances in understanding the roles of mast cells in health and disease, partly through the study of emerging non-IgE receptors such as the Mas-related G protein-coupled receptor X2, implicated in pseudo-allergic reactions as well as in innate defense and neuronal sensing; the mechano-sensing adhesion G protein-coupled receptor E2, variants of which are associated with familial vibratory urticaria; and purinergic receptors, which orchestrate tissue damage responses similarly to the IL-33 receptor. Recent evidence also points toward novel mechanisms that contribute to mast cell-mediated pathophysiology. Thus, in addition to releasing preformed mediators contained in granules and synthesizing mediators de novo, mast cells also secrete extracellular vesicles, which convey biological functions. Understanding their release, composition and uptake within a variety of clinical conditions will contribute to the understanding of disease specific pathology and likely lead the way to novel therapeutic approaches. We also discuss recent advances in the development of therapies targeting mast cell activity, including the ligation of inhibitory ITIM-containing receptors, and other strategies that suppress mast cells or responses to mediators for the management of mast cell-related diseases.


Subject(s)
Inflammation , Mast Cells , Humans , Immunoglobulin E , Inflammation/drug therapy , Mast Cells/pathology , Receptors, G-Protein-Coupled
12.
Nutrition ; 60: 161-169, 2019 04.
Article in English | MEDLINE | ID: mdl-30599460

ABSTRACT

OBJECTIVES: We aimed to analyze the effect of a protein-deficient diet on mucosal and systemic immunity during a Trichinella spiralis infection. METHODS: Two groups of weaning Wistar rats received a protein-deficient diet (6.5% casein) and the other two groups received a control diet (20% casein). After 10 d, one group of each diet was infected (PDI and CI) with muscle larvae (infecting stage). Food intake and body weight were assessed over time. Blood eosinophils counts, antibodies in serum, and tissue extracts were assessed at different days postinfection. Histologic studies were done in the lungs and intestines, and adult worm (AW) fecundity index score and muscle parasite burden were determined. RESULTS: Food and protein intake were lower in PDI than in CI. Body weight was lower in PDI than in a non-infected protein-deficient diet. Eosinophils counts were lower in PDI than in CI. Total and specific antibodies were lower in PDI than CI. PDI had a reduced number of mast and goblet cells in the lungs and intestines compared with CI. The persistence of AW in the intestines and migrant larvae at the lungs was longer in PDI than in CI.. The AW fecundity index score was higher in PDI than in CI. Finally, PDI evidenced a higher muscular parasite burden than CI. CONCLUSIONS: Protein deficiency affects the mucosal and systemic immune response to Trichinella spiralis and delays the expulsion and increases the fecundity index score of AW, which leads to a higher parasite burden in the muscles.


Subject(s)
Antibodies, Helminth/immunology , Diet, Protein-Restricted/adverse effects , Dietary Proteins/immunology , Trichinella spiralis/immunology , Trichinellosis/immunology , Animals , Intestinal Mucosa/immunology , Intestinal Mucosa/parasitology , Rats , Rats, Wistar , Trichinellosis/parasitology
13.
Vet Parasitol ; 231: 47-52, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27211081

ABSTRACT

Parasitic infection caused by Trichinella spiralis provokes an early stimulation of the mucosal immune system which causes an allergic inflammatory response in the lungs. The present work was intended to characterize the kinetics of emergence of regulatory parameters in Wistar rat lungs during this early inflammatory response, between days 0 and 13p.i. The presence of regulatory cells such as regulatory T cells (Tregs) and alternatively activated macrophages (AAM) was analyzed in lung cell suspensions. Moreover, a regulatory cytokine (TGF-ß) was studied in lung tissue extracts. Considering that newborn larvae (NBL) travel along the pulmonary microvasculature, the ability of this parasite stage to modulate the activation of lung macrophages was evaluated. For this purpose, lung macrophages from non-infected or infected rats (day 6p.i.) were cultured with live or dead NBL. Arginase activity (characteristic of AAM) and nitric oxide (NO produced by iNOS, characteristic of classical activated macrophages) were measured after 48h. Our results revealed a significant increase in the percentage of Tregs on days 6 and 13p.i., arginase activity on day 13p.i. and TGF-ß levels on days 6 and 13p.i. Lung macrophages from non-infected rats cultured with live NBL showed a significant increase in arginase activity and NO levels. Live and dead NBL induced a significant increase in arginase activity in lung macrophages from infected rats. Only live NBL significantly increased NO levels in these macrophages. The present work demonstrates for the first time, the emergence of regulatory parameters in the early lung immune response during T. spiralis infection. The immumodulatory properties exerted by NBL during its passage through this organ could be the cause of such regulation. Moreover, we have shown the ability of NBL to activate macrophages from the lung parenchyma by the classical and alternative pathways.


Subject(s)
Lung Diseases, Parasitic/immunology , Lung/immunology , Trichinellosis/immunology , Animals , Female , Inflammation/immunology , Inflammation/parasitology , Larva , Lung Diseases, Parasitic/parasitology , Lung Diseases, Parasitic/pathology , Macrophage Activation , Macrophages , Rats , Rats, Wistar , Trichinellosis/pathology
14.
Parasitol Res ; 114(2): 679-85, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25416332

ABSTRACT

The migratory stage of Trichinella spiralis, the newborn larva (NBL), travels along the pulmonary microvascular system on its way to the skeletal muscle cells. The present work studies the capability of lung cells to kill NBL. For this purpose, in vitro cytotoxicity assays were performed using NBL, lung cell suspensions from Wistar rats, rat anti-NBL surface sera, and fresh serum as complement source. The cytotoxic activity of lung cells from rats infected on day 6 p.i. was compared with that from noninfected rats. Two and 20 h-old NBL (NBL2 and NBL20) were used as they had shown to exhibit different surface antigens altering their biological activity. Sera antibodies were analyzed by indirect immunofluorescence assay, and cell populations used in each assay were characterized by histological staining. The role of IgE in the cytotoxic attack against NBL was analyzed using heated serum. The FcεRI expression on cell suspensions was examined by flow cytometry. Results showed that lung cells were capable of killing NBL by antibody-dependent cell-mediated cytotoxicity (ADCC). Lung cells from infected animals yielded the highest mortality percentages of NBL, with NBL20 being the most susceptible to such attack. IgE yielded a critical role in the cytotoxic attack. Regarding the analysis of cell suspensions, cells from infected rats showed an increase in the percentage of eosinophils, neutrophils, and the number of cells expressing the FcεRI receptor. We conclude that lung cells are capable of killing NBL in the presence of specific antibodies, supporting the idea that the lung is one of the sites where the NBL death occurs due to ADCC.


Subject(s)
Antigens, Helminth/immunology , Trichinella spiralis/immunology , Trichinellosis/immunology , Animals , Antigens, Surface/immunology , Biological Assay , Cell Survival , Eosinophils/immunology , Female , Larva , Lung/immunology , Neutrophils/immunology , Rats , Rats, Wistar , Trichinella spiralis/physiology , Trichinellosis/parasitology
SELECTION OF CITATIONS
SEARCH DETAIL