Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Annu Rev Immunol ; 35: 53-84, 2017 04 26.
Article in English | MEDLINE | ID: mdl-27912316

ABSTRACT

Helper T (Th) cell subsets direct immune responses by producing signature cytokines. Th2 cells produce IL-4, IL-5, and IL-13, which are important in humoral immunity and protection from helminth infection and are central to the pathogenesis of many allergic inflammatory diseases. Molecular analysis of Th2 cell differentiation and maintenance of function has led to recent discoveries that have refined our understanding of Th2 cell biology. Epigenetic regulation of Gata3 expression by chromatin remodeling complexes such as Polycomb and Trithorax is crucial for maintaining Th2 cell identity. In the context of allergic diseases, memory-type pathogenic Th2 cells have been identified in both mice and humans. To better understand these disease-driving cell populations, we have developed a model called the pathogenic Th population disease induction model. The concept of defined subsets of pathogenic Th cells may spur new, effective strategies for treating intractable chronic inflammatory disorders.


Subject(s)
Helminthiasis/immunology , Hypersensitivity/immunology , Th2 Cells/immunology , Animals , Cell Differentiation , Disease Models, Animal , Epigenesis, Genetic , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Immunity, Humoral , Immunologic Memory , Interleukin-13/metabolism , Interleukin-4/metabolism , Interleukin-5/metabolism , Mice , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/metabolism
2.
Nat Immunol ; 20(11): 1469-1480, 2019 11.
Article in English | MEDLINE | ID: mdl-31591568

ABSTRACT

Tissue-resident memory T cells (TRM cells) are crucial mediators of adaptive immunity in nonlymphoid tissues. However, the functional heterogeneity and pathogenic roles of CD4+ TRM cells that reside within chronic inflammatory lesions remain unknown. We found that CD69hiCD103lo CD4+ TRM cells produced effector cytokines and promoted the inflammation and fibrotic responses induced by chronic exposure to Aspergillus fumigatus. Simultaneously, immunosuppressive CD69hiCD103hiFoxp3+ CD4+ regulatory T cells were induced and constrained the ability of pathogenic CD103lo TRM cells to cause fibrosis. Thus, lung tissue-resident CD4+ T cells play crucial roles in the pathology of chronic lung inflammation, and CD103 expression defines pathogenic effector and immunosuppressive tissue-resident cell subpopulations in the inflamed lung.


Subject(s)
Cell Communication/immunology , Immune Tolerance , Immunologic Memory , Pulmonary Fibrosis/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, CD/metabolism , Antigens, Fungal/immunology , Aspergillus fumigatus/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Integrin alpha Chains/metabolism , Lung/cytology , Lung/immunology , Lung/pathology , Male , Mice, Transgenic , Pulmonary Fibrosis/pathology , T-Lymphocytes, Regulatory/metabolism
3.
J Allergy Clin Immunol ; 153(3): 672-683.e6, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37931708

ABSTRACT

BACKGROUND: Patients with severe asthma can present with eosinophilic type 2 (T2), neutrophilic, or mixed inflammation that drives airway remodeling and exacerbations and represents a major treatment challenge. The common ß (ßc) receptor signals for 3 cytokines, GM-CSF, IL-5, and IL-3, which collectively mediate T2 and neutrophilic inflammation. OBJECTIVE: To determine the pathogenesis of ßc receptor-mediated inflammation and remodeling in severe asthma and to investigate ßc antagonism as a therapeutic strategy for mixed granulocytic airway disease. METHODS: ßc gene expression was analyzed in bronchial biopsy specimens from patients with mild-to-moderate and severe asthma. House dust mite extract and Aspergillus fumigatus extract (ASP) models were used to establish asthma-like pathology and airway remodeling in human ßc transgenic mice. Lung tissue gene expression was analyzed by RNA sequencing. The mAb CSL311 targeting the shared cytokine binding site of ßc was used to block ßc signaling. RESULTS: ßc gene expression was increased in patients with severe asthma. CSL311 potently reduced lung neutrophils, eosinophils, and interstitial macrophages and improved airway pathology and lung function in the acute steroid-resistant house dust mite extract model. Chronic intranasal ASP exposure induced airway inflammation and fibrosis and impaired lung function that was inhibited by CSL311. CSL311 normalized the ASP-induced fibrosis-associated extracellular matrix gene expression network and strongly reduced signatures of cellular inflammation in the lung. CONCLUSIONS: ßc cytokines drive steroid-resistant mixed myeloid cell airway inflammation and fibrosis. The anti-ßc antibody CSL311 effectively inhibits mixed T2/neutrophilic inflammation and severe asthma-like pathology and reverses fibrosis gene signatures induced by exposure to commonly encountered environmental allergens.


Subject(s)
Asthma , Receptors, Cytokine , Mice , Animals , Humans , Receptors, Cytokine/metabolism , Airway Remodeling , Lung , Cytokines/metabolism , Mice, Transgenic , Inflammation , Allergens , Steroids/therapeutic use , Fibrosis , Pyroglyphidae
4.
J Clin Immunol ; 44(2): 55, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38270687

ABSTRACT

A homozygous missense mutation in the transferrin receptor 1 (TfR1), also known as CD71, leads to a rare inborn error of immunity (IEI) characterized by the impaired lymphocyte activation and proliferation due to defective iron uptake of cells. However, only one causative mutation (c.58T > C, p.Y20H) in the TFRC gene coding for TfR1 has been reported so far. We herein identified a new disease-causing homozygous germline mutation in the TFRC gene (c.64C > T, p.R22W) (referred to as TfR1R22W from now on) in a Turkish patient with combined immunodeficiency (CID). TfR1R22W results in impaired TfR1 internalization similar to previously defined TfR1Y20H mutation. We found that TfR1R22W is associated with severely restricted B and T lymphocyte clonal diversity and impaired T cell activation and cytokine production as well as defective mitochondrial oxidative phosphorylation in helper T cells. In addition, circulating NK, Treg, and MAIT cell populations were significantly decreased in the patient. Using whole transcriptome analysis, we found dysregulated immune homeostasis and novel biological processes associated with TfR1R22W. We also identified a considerable expansion of circulating low-density neutrophils (LDNs) in patient's PBMCs. Overall, TfR1R22W mutation expands the current understanding of the IEI associated with TfR1 dysfunction and provides new insights underlying impaired immune function, lymphocyte diversity, and granulocyte homeostasis.


Subject(s)
Germ-Line Mutation , Primary Immunodeficiency Diseases , Humans , Gene Expression Profiling , Iron
5.
Trends Immunol ; 42(6): 523-535, 2021 06.
Article in English | MEDLINE | ID: mdl-33962887

ABSTRACT

Mast cells (MC)s are evolutionarily conserved, tissue-resident immune cells with diverse roles in allergy, cancer, and protection from infection by helminths and microorganisms. The significant diversity in MC development and tissue-specific functional characteristics has recently begun to be understood. Exciting developments in single-cell-based RNA, protein, and chromatin profiling technologies offer new opportunities to characterize MC heterogeneity and to uncover novel MC functions and subtypes; these developments might lead to new and clinically effective therapies for certain pathologies. In this review, we provide an overview of the current understanding of MC development and heterogeneity and discuss new insights gained from single-cell-based studies that may lead to future research directions and therapeutic opportunities.


Subject(s)
Mast Cells , RNA , Cell Differentiation
6.
Immunity ; 42(2): 294-308, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25692703

ABSTRACT

Memory CD4(+) T helper (Th) cells provide long-term protection against pathogens and are essential for the development of vaccines; however, some antigen-specific memory Th cells also drive immune-related pathology, including asthma. The mechanisms regulating the pathogenicity of memory Th cells remain poorly understood. We found that interleukin-33 (IL-33)-ST2 signals selectively licensed memory Th2 cells to induce allergic airway inflammation via production of IL-5 and that the p38 MAP kinase pathway was a central downstream target of IL-33-ST2 in memory Th2 cells. In addition, we found that IL-33 induced upregulation of IL-5 by memory CD4(+) T cells isolated from nasal polyps of patients with eosinophilic chronic rhinosinusitis. Thus, IL-33-ST2-p38 signaling appears to directly instruct pathogenic memory Th2 cells to produce IL-5 and induce eosinophilic inflammation.


Subject(s)
Asthma/immunology , Interleukin-5/immunology , Interleukins/immunology , Receptors, Interleukin/immunology , Th2 Cells/immunology , p38 Mitogen-Activated Protein Kinases/immunology , Animals , Asthma/pathology , Cells, Cultured , Humans , Immunologic Memory/immunology , Inflammation/immunology , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Interleukin-5/biosynthesis , Interleukins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nasal Polyps/immunology , Pulmonary Eosinophilia/immunology , RNA Interference , RNA, Small Interfering , Receptors, Antigen, T-Cell/immunology , Receptors, Interleukin/genetics , Sinusitis/immunology , p38 Mitogen-Activated Protein Kinases/genetics
7.
J Allergy Clin Immunol ; 151(2): 324-344, 2023 02.
Article in English | MEDLINE | ID: mdl-36424209

ABSTRACT

The family of cytokines that comprises IL-3, IL-5, and GM-CSF was discovered over 30 years ago, and their biological activities and resulting impact in clinical medicine has continued to expand ever since. Originally identified as bone marrow growth factors capable of acting on hemopoietic progenitor cells to induce their proliferation and differentiation into mature blood cells, these cytokines are also recognized as key mediators of inflammation and the pathobiology of diverse immunologic diseases. This increased understanding of the functional repertoire of IL-3, IL-5, and GM-CSF has led to an explosion of interest in modulating their functions for clinical management. Key to the successful clinical translation of this knowledge is the recognition that these cytokines act by engaging distinct dimeric receptors and that they share a common signaling subunit called ß-common or ßc. The structural determination of how IL-3, IL-5, and GM-CSF interact with their receptors and linking this to their differential biological functions on effector cells has unveiled new paradigms of cell signaling. This knowledge has paved the way for novel mAbs and other molecules as selective or pan inhibitors for use in different clinical settings.


Subject(s)
Clinical Medicine , Granulocyte-Macrophage Colony-Stimulating Factor , Humans , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Cytokines/metabolism , Interleukin-3/metabolism , Interleukin-5/metabolism , Eosinophils , Biology
8.
J Allergy Clin Immunol ; 152(3): 725-735.e10, 2023 09.
Article in English | MEDLINE | ID: mdl-37127225

ABSTRACT

BACKGROUND: Mast cells (MCs) are tissue-resident immune cells that mediate IgE-dependent allergic responses. Downstream of FcεRI, an intricate network of receptor-specific signaling pathways and adaptor proteins govern MC function. The 14-3-3 family of serine-threonine phosphorylation-dependent adapter proteins are known to organize intracellular signaling. However, the role of 14-3-3 in IgE-dependent activation remains poorly defined. OBJECTIVE: We sought to determine whether 14-3-3 proteins are required for IgE-dependent MC activation and whether 14-3-3 is a viable target for the treatment of MC-mediated inflammatory diseases. METHODS: Genetic manipulation of 14-3-3ζ expression in human and mouse MCs was performed and IgE-dependent mediator release assessed. Pharmacologic inhibitors of 14-3-3 and 14-3-3ζ knockout mice were used to assess 14-3-3ζ function in a MC-dependent in vivo passive cutaneous anaphylaxis (PCA) model of allergic inflammation. Expression and function of 14-3-3ζ were assessed in human nasal polyp tissue MCs. RESULTS: IgE-dependent mediator release from human MCs was decreased by 14-3-3ζ knockdown and increased by 14-3-3ζ overexpression. Deletion of the 14-3-3ζ gene decreased IgE-dependent activation of mouse MCs in vitro and PCA responses in vivo. Furthermore, the 14-3-3 inhibitor, RB-11, which impairs dimerization of 14-3-3, inhibited cultured MC and polyp tissue MC activation and signaling downstream of the FcεRI receptor and dose-dependently attenuated PCA responses. CONCLUSION: IgE/FcεRI-mediated MC activation is positively regulated by 14-3-3ζ. We identify a critical role for this p-Ser/Thr-binding protein in the regulation of MC FcεRI signaling and IgE-dependent immune responses and show that this pathway may be amenable to pharmacologic targeting.


Subject(s)
Anaphylaxis , Receptors, IgE , Humans , Mice , Animals , 14-3-3 Proteins/genetics , 14-3-3 Proteins/metabolism , Mast Cells , Adaptor Proteins, Signal Transducing/metabolism , Immunoglobulin E , Inflammation/metabolism , Cell Degranulation
9.
Immunity ; 39(5): 819-32, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24238339

ABSTRACT

After antigen encounter by CD4(+) T cells, polarizing cytokines induce the expression of master regulators that control differentiation. Inactivation of the histone methyltransferase Ezh2 was found to specifically enhance T helper 1 (Th1) and Th2 cell differentiation and plasticity. Ezh2 directly bound and facilitated correct expression of Tbx21 and Gata3 in differentiating Th1 and Th2 cells, accompanied by substantial trimethylation at lysine 27 of histone 3 (H3K27me3). In addition, Ezh2 deficiency resulted in spontaneous generation of discrete IFN-γ and Th2 cytokine-producing populations in nonpolarizing cultures, and under these conditions IFN-γ expression was largely dependent on enhanced expression of the transcription factor Eomesodermin. In vivo, loss of Ezh2 caused increased pathology in a model of allergic asthma and resulted in progressive accumulation of memory phenotype Th2 cells. This study establishes a functional link between Ezh2 and transcriptional regulation of lineage-specifying genes in terminally differentiated CD4(+) T cells.


Subject(s)
Gene Expression Regulation , Histone-Lysine N-Methyltransferase/physiology , Polycomb Repressive Complex 2/physiology , T-Lymphocyte Subsets/cytology , Th1 Cells/cytology , Th2 Cells/cytology , Animals , Asthma/genetics , Asthma/immunology , Asthma/pathology , Cell Differentiation , Cells, Cultured/cytology , Cells, Cultured/immunology , Cells, Cultured/metabolism , Enhancer of Zeste Homolog 2 Protein , Female , GATA3 Transcription Factor/metabolism , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/deficiency , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Immunologic Memory , Interferon-gamma Release Tests , Lymphokines/biosynthesis , Lymphokines/genetics , Male , Methylation , Mice , Mice, Inbred C57BL , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/deficiency , Polycomb Repressive Complex 2/genetics , Protein Processing, Post-Translational , Sequence Deletion , T-Box Domain Proteins/biosynthesis , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , T-Lymphocyte Subsets/immunology , Th1 Cells/immunology , Th2 Cells/immunology
10.
Br J Cancer ; 125(3): 337-350, 2021 08.
Article in English | MEDLINE | ID: mdl-33927352

ABSTRACT

BACKGROUND: Glioblastoma is the most aggressive type of brain cancer with high-levels of intra- and inter-tumour heterogeneity that contribute to its rapid growth and invasion within the brain. However, a spatial characterisation of gene signatures and the cell types expressing these in different tumour locations is still lacking. METHODS: We have used a deep convolutional neural network (DCNN) as a semantic segmentation model to segment seven different tumour regions including leading edge (LE), infiltrating tumour (IT), cellular tumour (CT), cellular tumour microvascular proliferation (CTmvp), cellular tumour pseudopalisading region around necrosis (CTpan), cellular tumour perinecrotic zones (CTpnz) and cellular tumour necrosis (CTne) in digitised glioblastoma histopathological slides from The Cancer Genome Atlas (TCGA). Correlation analysis between segmentation results from tumour images together with matched RNA expression data was performed to identify genetic signatures that are specific to different tumour regions. RESULTS: We found that spatially resolved gene signatures were strongly correlated with survival in patients with defined genetic mutations. Further in silico cell ontology analysis along with single-cell RNA sequencing data from resected glioblastoma tissue samples showed that these tumour regions had different gene signatures, whose expression was driven by different cell types in the regional tumour microenvironment. Our results further pointed to a key role for interactions between microglia/pericytes/monocytes and tumour cells that occur in the IT and CTmvp regions, which may contribute to poor patient survival. CONCLUSIONS: This work identified key histopathological features that correlate with patient survival and detected spatially associated genetic signatures that contribute to tumour-stroma interactions and which should be investigated as new targets in glioblastoma. The source codes and datasets used are available in GitHub: https://github.com/amin20/GBM_WSSM .


Subject(s)
Brain Neoplasms/diagnostic imaging , Gene Expression Profiling/methods , Gene Regulatory Networks , Glioblastoma/diagnostic imaging , Radiographic Image Interpretation, Computer-Assisted/methods , Brain Neoplasms/genetics , Deep Learning , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Humans , Neural Networks, Computer , Single-Cell Analysis , Stem Cell Niche , Survival Analysis , Tumor Microenvironment
11.
Immunol Rev ; 278(1): 8-19, 2017 07.
Article in English | MEDLINE | ID: mdl-28658556

ABSTRACT

An estimated 300 million people currently suffer from asthma, which causes approximately 250 000 deaths a year. Allergen-specific T-helper (Th) cells produce cytokines that induce many of the hallmark features of asthma including airways hyperreactivity, eosinophilic and neutrophilic inflammation, mucus hypersecretion, and airway remodeling. Cytokine-producing Th subsets including Th1 (IFN-γ), Th2 (IL-4, IL-5, IL-13), Th9 (IL-9), Th17 (IL-17), Th22 (IL-22), and T regulatory (IL-10) cells have all been suggested to play a role in the development of asthma. Th differentiation involves genetic regulation of gene expression through the concerted action of cytokines, transcription factors, and epigenetic regulators. We describe how Th differentiation and plasticity is regulated by epigenetic histone and DNA modifications, with a focus on the regulation of histone methylation by members of the polycomb and trithorax complexes. In addition, we outline environmental influences that could influence epigenetic regulation of Th cells and discuss the potential to regulate Th plasticity and function through drugs targeting the epigenetic machinery. It is also becoming apparent that epigenetic regulation of allergen-specific memory Th cells may be important in the development and persistence of chronic allergies. Finally, we describe how epigenetic modifiers regulate cytokine memory in Th cells and describe recently identified hybrid, plastic, and pathogenic memory Th subsets the context of allergic asthma.


Subject(s)
Asthma/genetics , Asthma/immunology , Cell Differentiation , Cell Plasticity , Epigenesis, Genetic , Immunologic Memory , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Animals , Asthma/pathology , Cell Communication , Cell Differentiation/drug effects , Cytokines/biosynthesis , Disease Susceptibility , Environment , Epigenesis, Genetic/drug effects , Gene Expression Regulation/drug effects , Humans , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
12.
Immunity ; 35(5): 733-45, 2011 Nov 23.
Article in English | MEDLINE | ID: mdl-22118525

ABSTRACT

The regulation of memory CD4(+) helper T (Th) cell function, such as polarized cytokine production, remains unclear. Here we show that memory T helper 2 (Th2) cells are divided into four subpopulations by CD62L and CXCR3 expression. All four subpopulations produced interleukin-4 (IL-4) and IL-13, whereas only the CD62L(lo)CXCR3(lo) population produced IL-5 accompanied by increased H3-K4 methylation at the Il5 gene locus. The transcription factor Eomesodermin (encoded by Eomes) was highly expressed in memory Th2 cells, whereas its expression was selectively downregulated in the IL-5-producing cells. Il5 expression was enhanced in Eomes-deficient cells, and Eomesodermin was shown to interact with the transcription factor GATA3, preventing GATA3 binding to the Il5 promoter. Memory Th2 cell-dependent airway inflammation was attenuated in the absence of the CD62L(lo)CXCR3(lo) population but was enhanced by Eomes-deficient memory Th2 cells. Thus, IL-5 production in memory Th2 cells is regulated by Eomesodermin via the inhibition of GATA3 activity.


Subject(s)
GATA3 Transcription Factor/metabolism , Immunologic Memory/immunology , Interleukin-5/biosynthesis , T-Box Domain Proteins/metabolism , Th2 Cells/immunology , Animals , Cells, Cultured , GATA3 Transcription Factor/antagonists & inhibitors , Gene Expression , Inflammation/immunology , L-Selectin/metabolism , Lymphocyte Depletion , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Promoter Regions, Genetic , Receptors, CXCR3/metabolism , Respiratory System/immunology , T-Box Domain Proteins/genetics , Th2 Cells/metabolism , Transcription, Genetic
13.
Proc Natl Acad Sci U S A ; 113(20): E2842-51, 2016 May 17.
Article in English | MEDLINE | ID: mdl-27140620

ABSTRACT

Memory CD4(+) T helper (Th) cells are central to long-term protection against pathogens, but they can also be pathogenic and drive chronic inflammatory disorders. How these pathogenic memory Th cells are maintained, particularly at sites of local inflammation, remains unclear. We found that ectopic lymphoid-like structures called inducible bronchus-associated lymphoid tissue (iBALT) are formed during chronic allergic inflammation in the lung, and that memory-type pathogenic Th2 (Tpath2) cells capable of driving allergic inflammation are maintained within the iBALT structures. The maintenance of memory Th2 cells within iBALT is supported by Thy1(+)IL-7-producing lymphatic endothelial cells (LECs). The Thy1(+)IL-7-producing LECs express IL-33 and T-cell-attracting chemokines CCL21 and CCL19. Moreover, ectopic lymphoid structures consisting of memory CD4(+) T cells and IL-7(+)IL-33(+) LECs were found in nasal polyps of patients with eosinophilic chronic rhinosinusitis. Thus, Thy1(+)IL-7-producing LECs control chronic allergic airway inflammation by providing a survival niche for memory-type Tpath2 cells.


Subject(s)
Endothelial Cells/physiology , Rhinitis, Allergic/immunology , Sinusitis/immunology , Tertiary Lymphoid Structures/immunology , Animals , Cell Survival , Interleukin-7/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Tertiary Lymphoid Structures/pathology , Th2 Cells/immunology , Thy-1 Antigens/metabolism
14.
Trends Immunol ; 35(2): 69-78, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24332592

ABSTRACT

Immunological memory is a hallmark of adaptive immunity. Memory CD4 T helper (Th) cells are central to acquired immunity, and vaccines for infectious diseases are developed based on this concept. However, memory Th cells also play a critical role in the pathogenesis of various chronic inflammatory diseases, including asthma. We refer to these populations as 'pathogenic memory Th cells.' Here, we review recent developments highlighting the functions and characteristics of several pathogenic memory type Th2 cell subsets in allergic inflammation. Also discussed are the similarities and differences between pathogenic memory Th2 cells and recently identified type 2 innate lymphoid cells (ILC2), focusing on cytokine production and phenotypic profiles.


Subject(s)
Asthma/immunology , Th2 Cells/immunology , Animals , Dermatitis/immunology , Humans , Immunity, Innate , Immunologic Memory , Interleukin-17/biosynthesis , Interleukin-5/biosynthesis , Mice , Models, Immunological
15.
Proc Natl Acad Sci U S A ; 111(35): 12829-34, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25136117

ABSTRACT

Epigenetic modifications, such as posttranslational modifications of histones, play an important role in gene expression and regulation. These modifications are in part mediated by the Trithorax group (TrxG) complex and the Polycomb group (PcG) complex, which activate and repress transcription, respectively. We herein investigate the role of Menin, a component of the TrxG complex in T helper (Th) cell differentiation and show a critical role for Menin in differentiation and maintenance of Th17 cells. Menin(-/-) T cells do not efficiently differentiate into Th17 cells, leaving Th1 and Th2 cell differentiation intact in in vitro cultures. Menin deficiency resulted in the attenuation of Th17-induced airway inflammation. In differentiating Th17 cells, Menin directly bound to the Il17a gene locus and was required for the deposition of permissive histone modifications and recruitment of the RNA polymerase II transcriptional complex. Interestingly, although Menin bound to the Rorc locus, Menin was dispensable for the induction of Rorc expression and permissive histone modifications in differentiating Th17 cells. In contrast, Menin was required to maintain expression of Rorc in differentiated Th17 cells, indicating that Menin is essential to stabilize expression of the Rorc gene. Thus, Menin orchestrates Th17 cell differentiation and function by regulating both the induction and maintenance of target gene expression.


Subject(s)
Asthma/immunology , Epigenesis, Genetic/immunology , Interleukin-17/immunology , Proto-Oncogene Proteins/immunology , Th17 Cells/immunology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Chromatin/immunology , Chromatin/metabolism , Epigenesis, Genetic/genetics , Gene Expression Regulation/immunology , Histone-Lysine N-Methyltransferase/immunology , Histone-Lysine N-Methyltransferase/metabolism , Interleukin-17/genetics , Interleukin-17/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myeloid-Lymphoid Leukemia Protein/immunology , Myeloid-Lymphoid Leukemia Protein/metabolism , Neutrophils/drug effects , Neutrophils/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Ovalbumin/immunology , Ovalbumin/pharmacology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Polymerase II/immunology , RNA Polymerase II/metabolism , Th17 Cells/metabolism
18.
Proc Natl Acad Sci U S A ; 109(19): 7409-14, 2012 May 08.
Article in English | MEDLINE | ID: mdl-22474373

ABSTRACT

Memory T-helper (Th) lymphocytes are crucial for the maintenance of acquired immunity to eliminate infectious pathogens. We have previously demonstrated that most memory Th lymphocytes reside and rest on stromal niches of the bone marrow (BM). Little is known, however, regarding the molecular basis for the generation and maintenance of BM memory Th lymphocytes. Here we show that CD69-deficient effector CD4 T lymphocytes fail to relocate into and persist in the BM and therefore to differentiate into memory cells. Consequently, CD69-deficient CD4 T cells fail to facilitate the production of high-affinity antibodies and the generation of BM long-lived plasma cells in the late phase of immune responses. Thus, CD69 is critical for the generation and maintenance of professional memory Th lymphocytes, which can efficiently help humoral immunity in the late phase. The deficit of immunological memory in CD69-deficient mice also highlights the essential role of BM for the establishment of Th memory.


Subject(s)
Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Immunologic Memory/immunology , Lectins, C-Type/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adoptive Transfer , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/genetics , Antigens, Differentiation, T-Lymphocyte/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Bone Marrow/immunology , Bone Marrow/metabolism , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/transplantation , Female , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Mice, Transgenic , Microscopy, Confocal , Stromal Cells/immunology , Stromal Cells/metabolism , T-Lymphocytes, Helper-Inducer/metabolism
19.
J Immunol ; 184(8): 4510-20, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20237291

ABSTRACT

Polycomb group (PcG) gene products regulate the maintenance of homeobox gene expression in Drosophila and vertebrates. In the immune system, PcG molecules control cell cycle progression of thymocytes, Th2 cell differentiation, and the generation of memory CD4 T cells. In this paper, we extended the study of PcG molecules to the regulation of in vivo Th2 responses, especially allergic airway inflammation, by using conditional Ring1B-deficient mice with a CD4 T cell-specific deletion of the Ring1B gene (Ring1B(-/-) mice). In Ring1B(-/-) mice, CD4 T cell development appeared to be normal, whereas the differentiation of Th2 cells but not Th1 cells was moderately impaired. In an Ag-induced Th2-driven allergic airway inflammation model, eosinophilic inflammation was attenuated in Ring1B(-/-) mice. Interestingly, Ring1B(-/-) effector Th2 cells were highly susceptible to apoptosis in comparison with wild-type effector Th2 cells in vivo and in vitro. The in vitro experiments revealed that the expression of Bim was increased at both the transcriptional and protein levels in Ring1B(-/-) effector Th2 cells, and the enhanced apoptosis in Ring1B(-/-) Th2 cells was rescued by the knockdown of Bim but not the other proapoptotic genes, such as Perp, Noxa, or Bax. The enhanced apoptosis detected in the transferred Ring1B(-/-) Th2 cells in the lung of the recipient mice was also rescued by knockdown of Bim. Therefore, these results indicate that Ring1B plays an important role in Th2-driven allergic airway inflammation through the control of Bim-dependent apoptosis of effector Th2 cells in vivo.


Subject(s)
Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis/immunology , Inflammation Mediators/physiology , Lung/immunology , Lung/pathology , Membrane Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Repressor Proteins/physiology , Th2 Cells/immunology , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/physiology , Bcl-2-Like Protein 11 , Cells, Cultured , Down-Regulation/genetics , Down-Regulation/immunology , Immunophenotyping , Lung/metabolism , Membrane Proteins/deficiency , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Polycomb Repressive Complex 1 , Polycomb-Group Proteins , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/physiology , Th2 Cells/metabolism , Th2 Cells/pathology
20.
Curr Protoc ; 2(11): e589, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36373983

ABSTRACT

Recent advances in flow cytometry have allowed high-dimensional characterization of biological phenomena, enabling breakthroughs in a multitude of fields. Despite the appreciation of the unique properties of antigens and fluorophores in high-parameter panel design, staining conditions are often standardized for short surface stains, regardless of antibody affinity or antigen accessibility. Here, we demonstrate how increasing antibody incubation times can lead to substantial improvements in sensitivity, maintaining specificity, and reducing background, while also significantly reducing the costs of high-parameter cytometry panels. Furthermore, overnight staining reduces the influence of interexperimental variability, assisting accurate pooling over experiments over extended time courses. We provide guidance on how to optimize staining conditions for diverse antigens, including how different fixation strategies can affect epitope accessibility. Overnight staining can thus substantially improve the resolution, repeatability, and cost-effectiveness of high-parameter cytometry. © 2022 The Authors. Current Protocols published by Wiley Periodicals LLC.


Subject(s)
Antibodies , Antigens , Flow Cytometry/methods , Staining and Labeling , Fluorescent Dyes
SELECTION OF CITATIONS
SEARCH DETAIL