Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters








Publication year range
1.
Life Sci Alliance ; 7(8)2024 Aug.
Article in English | MEDLINE | ID: mdl-38830770

ABSTRACT

Post-transcriptional regulation of immune-related transcripts by RNA-binding proteins (RBPs) impacts immune cell responses, including mast cell functionality. Despite their importance in immune regulation, the functional role of most RBPs remains to be understood. By manipulating the expression of specific RBPs in murine mast cells, coupled with mass spectrometry and transcriptomic analyses, we found that the Regnase family of proteins acts as a potent regulator of mast cell physiology. Specifically, Regnase-1 is required to maintain basic cell proliferation and survival, whereas both Regnase-1 and -3 cooperatively regulate the expression of inflammatory transcripts upon activation, with Tnf being a primary target in both human and mouse cells. Furthermore, Regnase-3 directly interacts with Regnase-1 in mast cells and is necessary to restrain Regnase-1 expression through the destabilization of its transcript. Overall, our study identifies protein interactors of endogenously expressed Regnase factors, characterizes the regulatory interplay between Regnase family members in mast cells, and establishes their role in the control of mast cell homeostasis and inflammatory responses.


Subject(s)
Cell Survival , Cytokines , Mast Cells , Mast Cells/metabolism , Animals , Mice , Humans , Cytokines/metabolism , Cell Survival/genetics , Ribonuclease, Pancreatic/metabolism , Ribonuclease, Pancreatic/genetics , Ribonucleases/metabolism , Ribonucleases/genetics , Gene Expression Regulation , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Mice, Inbred C57BL , Cell Proliferation , Inflammation/metabolism , Transcription Factors
2.
Nat Commun ; 14(1): 3862, 2023 06 29.
Article in English | MEDLINE | ID: mdl-37386028

ABSTRACT

Mast cells are central players in allergy and asthma, and their dysregulated responses lead to reduced quality of life and life-threatening conditions such as anaphylaxis. The RNA modification N6-methyladenosine (m6A) has a prominent impact on immune cell functions, but its role in mast cells remains unexplored. Here, by optimizing tools to genetically manipulate primary mast cells, we reveal that the m6A mRNA methyltransferase complex modulates mast cell proliferation and survival. Depletion of the catalytic component Mettl3 exacerbates effector functions in response to IgE and antigen complexes, both in vitro and in vivo. Mechanistically, deletion of Mettl3 or Mettl14, another component of the methyltransferase complex, lead to the enhanced expression of inflammatory cytokines. By focusing on one of the most affected mRNAs, namely the one encoding the cytokine IL-13, we find that it is methylated in activated mast cells, and that Mettl3 affects its transcript stability in an enzymatic activity-dependent manner, requiring consensus m6A sites in the Il13 3'-untranslated region. Overall, we reveal that the m6A machinery is essential in mast cells to sustain growth and to restrain inflammatory responses.


Subject(s)
Cytokines , Mast Cells , Cytokines/genetics , RNA, Messenger/genetics , Quality of Life , Interleukin-13/genetics , RNA Stability/genetics , Methyltransferases/genetics
3.
Cancer Immunol Res ; 10(12): 1525-1541, 2022 12 02.
Article in English | MEDLINE | ID: mdl-36206577

ABSTRACT

During melanoma metastasis, tumor cells originating in the skin migrate via lymphatic vessels to the sentinel lymph node (sLN). This process facilitates tumor cell spread across the body. Here, we characterized the innate inflammatory response to melanoma in the metastatic microenvironment of the sLN. We found that macrophages located in the subcapsular sinus (SS) produced protumoral IL1α after recognition of tumoral antigens. Moreover, we confirmed that the elimination of LN macrophages or the administration of an IL1α-specific blocking antibody reduced metastatic spread. To understand the mechanism of action of IL1α in the context of the sLN microenvironment, we applied single-cell RNA sequencing to microdissected metastases obtained from animals treated with the IL1α-specific blocking antibody. Among the different pathways affected, we identified STAT3 as one of the main targets of IL1α signaling in metastatic tumor cells. Moreover, we found that the antitumoral effect of the anti-IL1α was not mediated by lymphocytes because Il1r1 knockout mice did not show significant differences in metastasis growth. Finally, we found a synergistic antimetastatic effect of the combination of IL1α blockade and STAT3 inhibition with stattic, highlighting a new immunotherapy approach to preventing melanoma metastasis.


Subject(s)
Lymphatic Vessels , Melanoma , Sentinel Lymph Node , Skin Neoplasms , Animals , Mice , Sentinel Lymph Node Biopsy , Sentinel Lymph Node/pathology , Lymphatic Metastasis/pathology , Melanoma/pathology , Macrophages/metabolism , Lymphatic Vessels/metabolism , Lymphatic Vessels/pathology , Lymph Nodes/pathology , Skin Neoplasms/pathology , Tumor Microenvironment
4.
Nat Immunol ; 23(8): 1208-1221, 2022 08.
Article in English | MEDLINE | ID: mdl-35879451

ABSTRACT

T cell antigen-receptor (TCR) signaling controls the development, activation and survival of T cells by involving several layers and numerous mechanisms of gene regulation. N6-methyladenosine (m6A) is the most prevalent messenger RNA modification affecting splicing, translation and stability of transcripts. In the present study, we describe the Wtap protein as essential for m6A methyltransferase complex function and reveal its crucial role in TCR signaling in mouse T cells. Wtap and m6A methyltransferase functions were required for the differentiation of thymocytes, control of activation-induced death of peripheral T cells and prevention of colitis by enabling gut RORγt+ regulatory T cell function. Transcriptome and epitranscriptomic analyses reveal that m6A modification destabilizes Orai1 and Ripk1 mRNAs. Lack of post-transcriptional repression of the encoded proteins correlated with increased store-operated calcium entry activity and diminished survival of T cells with conditional genetic inactivation of Wtap. These findings uncover how m6A modification impacts on TCR signal transduction and determines activation and survival of T cells.


Subject(s)
Cell Cycle Proteins , Methyltransferases , Adenosine/analogs & derivatives , Animals , Cell Cycle Proteins/metabolism , Methylation , Methyltransferases/genetics , Mice , RNA Splicing Factors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
5.
Immunol Rev ; 304(1): 51-61, 2021 11.
Article in English | MEDLINE | ID: mdl-34523134

ABSTRACT

RNA-binding proteins (RBPs) regulate all aspects of the life of mRNA transcripts. They are critically important in regulating immune responses, most notably by restraining excessive inflammation that can potentially lead to tissue damage. RBPs are also crucial for pathogen sensing, for instance for the recognition of viral nucleic acids. Concordant with these central regulatory roles, the dysregulated activity of many RBPs can give rise to disease. The expression and function of RBPs are therefore highly controlled by an elaborate network of transcriptional, post-transcriptional and post-translational mechanisms, including the ability of different RBPs to cross-regulate each other's expression. With an emphasis on macrophages and mast cells, we review current knowledge on the role of selected RBPs that have been shown to directly impact the expression of inflammatory transcripts. By focusing specifically on proteins of the Regnase and ZFP36 family, as well as on factors involved in N6 -methyladenosine (m6 A) deposition and recognition, we discuss mechanism of action, regulatory feedback, and impact of these selected proteins on immune responses. Finally, we include examples of the role of m6 A and RBPs in the recognition of viral RNAs. Overall, we provide a general overview of the impact of selected RBPs on the myeloid compartment, followed by a discussion of outstanding questions and challenges for the future.


Subject(s)
Immunity , RNA-Binding Proteins , Methylation , Myeloid Cells/metabolism , RNA , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
6.
PLoS One ; 16(2): e0247232, 2021.
Article in English | MEDLINE | ID: mdl-33600503

ABSTRACT

The appropriate regulation of T lymphocyte functions is key to achieve protective immune responses, while at the same time limiting the risks of tissue damage and chronic inflammation. Deciphering the mechanisms underpinning T cell responses in humans may therefore be beneficial for a range of infectious and chronic diseases. Recently, the development of methods based on CRISPR-Cas9 gene-editing has greatly expanded the available tool-box for the mechanistic studies of primary human T cell responses. While the deletion of a surface protein has become a relatively straightforward task, as long as an antibody for detection is available, the identification and selection of cells lacking an intracellular protein, a non-coding RNA or a protein for which no antibody is available, remain more problematic. Here, we discuss the options currently available to scientists interested in performing gene-editing in primary human T lymphocytes and we describe the optimization of a workflow for the screening and analysis of lymphocytes following gene-editing with CRISPR-Cas9 based on T cell cloning and T7 endonuclease I cleavage assay.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Editing/methods , Membrane Proteins/genetics , Transcription Factors/genetics , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/genetics , Cells, Cultured , Endoribonucleases/deficiency , Endoribonucleases/genetics , Humans , Membrane Proteins/deficiency , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism , Receptors, Antigen, T-Cell, alpha-beta/deficiency , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Box Domain Proteins/deficiency , T-Box Domain Proteins/genetics , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Transcription Factors/deficiency
7.
Nat Immunol ; 21(4): 388-399, 2020 04.
Article in English | MEDLINE | ID: mdl-32205878

ABSTRACT

Understanding the mechanisms that modulate helper T lymphocyte functions is crucial to decipher normal and pathogenic immune responses in humans. To identify molecular determinants influencing the pathogenicity of T cells, we separated ex vivo-isolated primary human memory T lymphocytes on the basis of their ability to produce high levels of inflammatory cytokines. We found that the inflammatory, cytokine-producing phenotype of memory T lymphocytes was defined by a specific core gene signature and was mechanistically regulated by the constitutive activation of the NF-κB pathway and by the expression of the transcriptional repressor BHLHE40. BHLHE40 attenuated the expression of anti-inflammatory factors, including miR-146a, a negative regulator of NF-κB activation and ZC3H12D, an RNase of the Regnase-1 family able to degrade inflammatory transcripts. Our data reveal a molecular network regulating the proinflammatory phenotype of human memory T lymphocytes, with the potential to contribute to disease.


Subject(s)
Gene Expression Regulation/immunology , Immunologic Memory/immunology , Inflammation/immunology , Cell Line , Cell Line, Tumor , Cytokines/immunology , HEK293 Cells , Humans , Jurkat Cells , Lymphocyte Activation/immunology , NF-kappa B/immunology , Phenotype , T-Lymphocytes/immunology
8.
Eur J Immunol ; 49(4): 611-625, 2019 04.
Article in English | MEDLINE | ID: mdl-30698829

ABSTRACT

In mammals, the 5'-methylcytosine (5mC) modification in the genomic DNA contributes to the dynamic control of gene expression. 5mC erasure is required for the activation of developmental programs and occurs either by passive dilution through DNA replication, or by enzymatic oxidation of the methyl mark to 5-hydroxymethylcytosine (5hmC), which can persist as such or undergo further oxidation and enzymatic removal. The relative contribution of each mechanism to epigenetic control in dynamic biological systems still remains a compelling question. To explore this critical issue, we used primary human T lymphocytes, in which two cellular states can be clearly identified, namely quiescent naïve T cells, which are slowly or rarely proliferating, and rapidly proliferating activated T cells. We found that active mechanisms of methylation removal were selectively at work in naïve T cells, while memory T lymphocytes entirely relied on passive, replication-dependent dilution, suggesting that proliferative capacity influences the choice of the preferential demethylation mechanism. Active processes of demethylation appear to be critical in quiescent naïve T lymphocytes for the maintenance of regulatory regions poised for rapid responses to physiological stimuli.


Subject(s)
5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Cell Differentiation/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Biomarkers , Cell Differentiation/genetics , DNA Methylation , Humans , Immunophenotyping , Lymphocyte Activation/genetics , Receptors, Antigen, T-Cell/metabolism
9.
F1000Res ; 6: 2064, 2017.
Article in English | MEDLINE | ID: mdl-29225792

ABSTRACT

Mast cells are tissue-resident, innate immune cells present in most tissues of the body and are important effector and immunomodulatory cells. Differentiated mast cells typically are characterized by the surface expression of the receptors KIT and FcεRI, the latter especially being important for stimulation through IgE antibodies, although these cells have the ability to respond to a wide variety of environmental signals, to which they can variably react by releasing pre-stored or de novo-synthesized mediators or both. Since mast cells terminate their differentiation in their tissue of residence in response to specific microenvironmental cues, each tissue may comprise unique mast cell subtypes, and responses are tailored to the danger signals that are likely to be encountered in each anatomical location. From a transcriptional point of view, these cells therefore must be endowed with epigenetic and transcriptional programs that allow them to maintain a stable identity and at the same time allow sufficient plasticity to adapt to different environmental challenges. In this commentary, we highlight some of the recent findings that advanced our understanding of the transcriptional and epigenetic programs regulating mast cell functions.

11.
Proc Natl Acad Sci U S A ; 114(8): E1490-E1499, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28167789

ABSTRACT

DNA methylation and specifically the DNA methyltransferase enzyme DNMT3A are involved in the pathogenesis of a variety of hematological diseases and in regulating the function of immune cells. Although altered DNA methylation patterns and mutations in DNMT3A correlate with mast cell proliferative disorders in humans, the role of DNA methylation in mast cell biology is not understood. By using mast cells lacking Dnmt3a, we found that this enzyme is involved in restraining mast cell responses to acute and chronic stimuli, both in vitro and in vivo. The exacerbated mast cell responses observed in the absence of Dnmt3a were recapitulated or enhanced by treatment with the demethylating agent 5-aza-2'-deoxycytidine as well as by down-modulation of Dnmt1 expression, further supporting the role of DNA methylation in regulating mast cell activation. Mechanistically, these effects were in part mediated by the dysregulated expression of the scaffold protein IQGAP2, which is characterized by the ability to regulate a wide variety of biological processes. Altogether, our data demonstrate that DNMT3A and DNA methylation are key modulators of mast cell responsiveness to acute and chronic stimulation.


Subject(s)
Cell Proliferation/physiology , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/physiology , Dermatitis, Contact/immunology , Epigenesis, Genetic/physiology , Mast Cells/physiology , Passive Cutaneous Anaphylaxis/immunology , ras GTPase-Activating Proteins/metabolism , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Degranulation/genetics , Cell Proliferation/drug effects , Cells, Cultured , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation/drug effects , DNA Methyltransferase 3A , Decitabine , Dermatitis, Contact/etiology , Disease Models, Animal , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Immunoglobulin E/immunology , Interleukin-3/metabolism , Mast Cells/drug effects , Mastocytosis, Systemic/immunology , Mice , Mice, Knockout , Mutation , Oxazolone/toxicity , RNA Interference , RNA, Small Interfering/metabolism , ras GTPase-Activating Proteins/genetics
12.
Cell Rep ; 15(7): 1566-1579, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27160912

ABSTRACT

Dioxygenases of the TET family impact genome functions by converting 5-methylcytosine (5mC) in DNA to 5-hydroxymethylcytosine (5hmC). Here, we identified TET2 as a crucial regulator of mast cell differentiation and proliferation. In the absence of TET2, mast cells showed disrupted gene expression and altered genome-wide 5hmC deposition, especially at enhancers and in the proximity of downregulated genes. Impaired differentiation of Tet2-ablated cells could be relieved or further exacerbated by modulating the activity of other TET family members, and mechanistically it could be linked to the dysregulated expression of C/EBP family transcription factors. Conversely, the marked increase in proliferation induced by the loss of TET2 could be rescued exclusively by re-expression of wild-type or catalytically inactive TET2. Our data indicate that, in the absence of TET2, mast cell differentiation is under the control of compensatory mechanisms mediated by other TET family members, while proliferation is strictly dependent on TET2 expression.


Subject(s)
Biocatalysis , Cell Differentiation , DNA-Binding Proteins/metabolism , Mast Cells/cytology , Mast Cells/metabolism , Proto-Oncogene Proteins/metabolism , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Ascorbic Acid/pharmacology , Biocatalysis/drug effects , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cytokines/metabolism , DNA-Binding Proteins/deficiency , Dioxygenases , Gene Deletion , Gene Expression Regulation , Gene Knockdown Techniques , Genome , HEK293 Cells , Humans , Male , Mast Cells/drug effects , Proto-Oncogene Proteins/deficiency , Sequence Analysis, RNA , Transcription, Genetic
13.
Swiss Med Wkly ; 145: w14191, 2015.
Article in English | MEDLINE | ID: mdl-26496689

ABSTRACT

The risk of developing autoimmune diseases depends on both genetic and environmental factors, with epigenetic mechanisms of regulation potentially translating environmental cues into stable modifications in gene expression. Such stable memory of a functional state has been deciphered into a number of molecular mechanisms that collectively define the epigenetic status of a cell. In recent years, it has become increasingly clear that epigenetic modifications are highly dynamic and are able to adapt to the changing environment, with important impact on the onset and development of a number of diseases. Here, we describe some of the epigenetic mechanisms of regulation of cellular functional states in T lymphocytes, with a particular focus on DNA methylation. We will also discuss current knowledge on the role of epigenetics in autoimmunity and consider open questions in the field.


Subject(s)
Epigenesis, Genetic , T-Lymphocytes/physiology , Adaptation, Physiological/genetics , Autoimmunity/genetics , DNA Methylation , Gene-Environment Interaction , Humans
14.
Nat Commun ; 6: 6431, 2015 Mar 16.
Article in English | MEDLINE | ID: mdl-25775432

ABSTRACT

T helper (TH) cell polarization during priming is modulated by a number of signals, but whether polarization to a given phenotype also influences recall responses of memory TH cells is relatively unknown. Here we show that miR-181a is selectively induced in both human and mouse naive T cells differentiating into the TH17, but not TH1 or TH2 subset. In human memory TH17 cells, miR-181a regulates responses to cognate antigens through modulation of ERK phosphorylation. By enhancing the signalling cascade from the T-cell receptor, such molecular network reduces the threshold of TH17 cell activation. Moreover, at a late time point, the same network induces a self-regulatory mechanism dependent on ID3, a negative regulator of transcription factors that control RORC expression, thus modulating TH17 activity. Our results demonstrate that the phenotype acquired by TH cells during priming contributes to their threshold of activation to secondary antigenic stimulations, thus influencing memory responses.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Immunologic Memory , MicroRNAs/metabolism , Th17 Cells/cytology , Animals , Antigens/chemistry , Candida albicans/metabolism , Cell Differentiation , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , Phenotype , Phosphorylation , RNA Interference , Signal Transduction
15.
Eur J Haematol ; 95(6): 566-75, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25688802

ABSTRACT

OBJECTIVE: As disruption of epigenetic control is a frequent event in solid tumors and leukemia, we investigated changes in DNA methylation (5mC) and hydroxymethylation (5hmC) in patients with systemic mastocytosis (SM), a rare myeloproliferative disease with a wide spectrum of severity, characterized by the accumulation of mast cells in various organs. METHODS: We measured overall genomic levels of 5hmC and 5mC in patients with SM by dot blot, as well as by quantitative immunofluorescence in samples of cutaneous mastocytosis. RESULTS: Overall 5hmC levels were reduced in all patients with SM, but to a greater extent in the presence of higher D816V mutational load in the KIT oncogene, which affects prognosis and therapeutic options in these patients. Loss of 5hmC was likely due to systemic effects of SM as it did not correlate with overall mast cell burden in these patients, nor it was due to inactivating mutations of TET2 or reduced TET2 expression. CONCLUSIONS: The correlation between SM diagnosis and significantly low 5hmC levels suggests that reduction of 5hmC represents a systemic effect of SM that may be useful for patient stratification and that measurements of 5hmC levels may serve as a better prognostic marker than TET2 mutations.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Mastocytosis, Systemic/genetics , Biopsy , Bone Marrow/pathology , Cell Line , DNA-Binding Proteins/genetics , Dioxygenases , Female , Humans , Immunophenotyping , Male , Mast Cells/metabolism , Mast Cells/pathology , Mastocytosis, Systemic/diagnosis , Mutation , Proto-Oncogene Proteins/genetics
16.
J Immunol ; 193(5): 2196-206, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25063866

ABSTRACT

We identified two mast cell subsets characterized by the differential expression of surface CD25 (IL-2Rα) and by different abilities to produce cytokines and to proliferate, both in vitro and in vivo. CD25 can be expressed on the surface of immune cells in the absence of the other chains of the IL-2R, which are indispensable for IL-2 signaling. We show that functional differences between the two mast cell populations were dependent on CD25 itself, which directly modulated proliferation and cytokine responses. These effects were completely independent from IL-2 or the expression of the other chains of the high-affinity IL-2R, indicating an autonomous and previously unappreciated role for CD25 in regulating cell functions. Cells genetically ablated for CD25 completely recapitulated the CD25-negative phenotype and never acquired the properties characteristic of CD25-positive mast cells. Finally, adoptive transfer experiments in the mouse demonstrated a different impact of these populations in models of anaphylaxis and contact sensitivity. Our findings indicate a general role for CD25 in contexts where IL-2 signaling is not involved, and may have important implications for all mast cell-related diseases, as well as in all cell types expressing CD25 independently of its IL-2-related functions.


Subject(s)
Gene Expression Regulation/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-2/immunology , Mast Cells/immunology , Signal Transduction/immunology , 3T3 Cells , Animals , Gene Expression Regulation/genetics , Interleukin-2/genetics , Interleukin-2 Receptor alpha Subunit/genetics , Mast Cells/cytology , Mice , Mice, Knockout , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL