Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Adv Sci (Weinh) ; 11(10): e2304702, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38145969

ABSTRACT

The DNA damage response is essential for preserving genome integrity and eliminating damaged cells. Although cellular metabolism plays a central role in cell fate decision between proliferation, survival, or death, the metabolic response to DNA damage remains largely obscure. Here, this work shows that DNA damage induces fatty acid oxidation (FAO), which is required for DNA damage-induced cell death. Mechanistically, FAO induction increases cellular acetyl-CoA levels and promotes N-alpha-acetylation of caspase-2, leading to cell death. Whereas chemotherapy increases FAO related genes through peroxisome proliferator-activated receptor α (PPARα), accelerated hypoxia-inducible factor-1α stabilization by tumor cells in obese mice impedes the upregulation of FAO, which contributes to its chemoresistance. Finally, this work finds that improving FAO by PPARα activation ameliorates obesity-driven chemoresistance and enhances the outcomes of chemotherapy in obese mice. These findings reveal the shift toward FAO induction is an important metabolic response to DNA damage and may provide effective therapeutic strategies for cancer patients with obesity.


Subject(s)
Fatty Acids , PPAR alpha , Mice , Animals , Humans , Oxidation-Reduction , Fatty Acids/metabolism , PPAR alpha/metabolism , Mice, Obese , Drug Resistance, Neoplasm , Obesity/metabolism , Cell Death
2.
Front Immunol ; 14: 1332386, 2023.
Article in English | MEDLINE | ID: mdl-38239368

ABSTRACT

γδT17 cells are a subset of γδT cells producing IL-17, which is crucial for protection against bacterial and fungal infections. It has recently been shown that γδT17 cells have enriched lipid storage and lipid metabolism. However, the regulation of γδT17 cell function and differentiation with respect to lipids remains unknown. Here, we report that PRDM16 is a critical regulator of γδT17 cell differentiation, controlling type 17 immunity gene expression program and lipid-dependent cell fitness. We demonstrated that γδT17 cells have higher lipid-dependent cell fitness, which is negatively correlated with the expression of Prdm16. Loss of Prdm16 enhances the function and differentiation of γδT17 cells, and increases their fitness in lipid-rich environments. Specifically, loss of Prdm16 exacerbates development of psoriasis in the skin, a lipid-rich organ, and Prdm16 controls lipid-mediated differentiation of Vγ4+ γδT17 cells, which are the major source of IL-17 during the onset of psoriasis. Our study highlights the potential impact of PRDM16 on lipid-dependent fitness and protective immune function of γδT cells and also on the immunotherapy of psoriasis and inflammatory diseases.


Subject(s)
Interleukin-17 , Psoriasis , Humans , Interleukin-17/metabolism , Skin , Cell Differentiation , Lipids , DNA-Binding Proteins/genetics , Transcription Factors
3.
Sci Adv ; 8(47): eadc9346, 2022 11 25.
Article in English | MEDLINE | ID: mdl-36427304

ABSTRACT

Exhausted CD8 T cells during chronic inflammatory responses against viral infections and cancer are phenotypically and functionally heterogeneous. In particular, CD8 T cells with cytolytic effector function have been recently identified among the exhausted CD8 T cell subsets. However, the regulation of their differentiation and function remains largely unknown. Here, we report that Krüppel-like factor 4 (KLF4) is a critical regulator of the exhaustion process, promoting the cytolytic effector function of exhausted CD8 T cells. KLF4-expressing CD8 T cells in exhaustion contexts showed the features of transitory effector CD8 T cells. Enforced KLF4 expression increased CD8 T cell differentiation into transitory effector subsets and enhanced their antitumor immunity. We further demonstrated that KLF4 also showed a capacity of reinvigorating exhausted CD8 T cells. Last, high KLF4 expression was positively correlated with a favorable prognosis in human patients with cancer. Our study highlights the potential impacts of KLF4 on CD8 T cell exhaustion and antitumor immune therapy.


Subject(s)
CD8-Positive T-Lymphocytes , Kruppel-Like Factor 4 , Humans , Cell Death , Cell Differentiation , Lymphocyte Activation
4.
Proc Natl Acad Sci U S A ; 119(27): e2121520119, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35776543

ABSTRACT

Activated Foxp3+ regulatory T (Treg) cells differentiate into effector Treg (eTreg) cells to maintain peripheral immune homeostasis and tolerance. T cell receptor (TCR)-mediated induction and regulation of store-operated Ca2+ entry (SOCE) is essential for eTreg cell differentiation and function. However, SOCE regulation in Treg cells remains unclear. Here, we show that inositol polyphosphate multikinase (IPMK), which generates inositol tetrakisphosphate and inositol pentakisphosphate, is a pivotal regulator of Treg cell differentiation downstream of TCR signaling. IPMK is highly expressed in TCR-stimulated Treg cells and promotes a TCR-induced Treg cell program. IPMK-deficient Treg cells display aberrant T cell activation and impaired differentiation into RORγt+ Treg cells and tissue-resident Treg cells. Mechanistically, IPMK controls the generation of higher-order inositol phosphates, thereby promoting Ca2+ mobilization and Treg cell effector functions. Our findings identify IPMK as a critical regulator of TCR-mediated Ca2+ influx and highlight the importance of IPMK in Treg cell-mediated immune homeostasis.


Subject(s)
Calcium , Homeostasis , Phosphotransferases (Alcohol Group Acceptor) , Polyphosphates , T-Lymphocytes, Regulatory , Animals , Calcium/metabolism , Cell Differentiation , Homeostasis/immunology , Mice , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Polyphosphates/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology
5.
Sci Adv ; 7(38): eabc6792, 2021 Sep 17.
Article in English | MEDLINE | ID: mdl-34524839

ABSTRACT

Increase in the size of human neocortex­acquired in evolution­accounts for the unique cognitive capacity of humans. This expansion reflects the evolutionarily enhanced proliferative ability of basal progenitors (BPs), including the basal radial glia and basal intermediate progenitors (bIPs) in mammalian cortex, which may have been acquired through epigenetic alterations in BPs. However, how the epigenome in BPs differs across species is not known. Here, we report that histone H3 acetylation is a key epigenetic regulation in bIP amplification and cortical expansion. Through epigenetic profiling of sorted bIPs, we show that histone H3 lysine 9 acetylation (H3K9ac) is low in murine bIPs and high in human bIPs. Elevated H3K9ac preferentially increases bIP proliferation, increasing the size and folding of the normally smooth mouse neocortex. H3K9ac drives bIP amplification by increasing expression of the evolutionarily regulated gene, Trnp1, in developing cortex. Our findings demonstrate a previously unknown mechanism that controls cortical architecture.

6.
Cell Rep ; 33(7): 108395, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33207205

ABSTRACT

The mammalian SWitch/Sucrose Non-Fermentable (SWI/SNF) chromatin-remodeling BAF (BRG1/BRM-associated factor) complex plays an essential role in developmental and pathological processes. We show that the deletion of Baf155, which encodes a subunit of the BAF complex, in the Tie2(+) lineage (Baf155 (CKO) leads to defects in yolk sac myeloid and definitive erythroid (EryD) lineage differentiation from erythromyeloid progenitors (EMPs). The chromatin of myeloid gene loci in Baf155 CKO EMPs is mostly inaccessible and enriched mainly by the ETS binding motif. BAF155 interacts with PU.1 and is recruited to PU.1 target gene loci together with p300 and KDM6a. Treatment of Baf155 CKO embryos with GSK126, an H3K27me2/3 methyltransferase EZH2 inhibitor, rescues myeloid lineage gene expression. This study uncovers indispensable BAF-mediated chromatin remodeling of myeloid gene loci at the EMP stage. Future studies exploiting epigenetics in the generation and application of EMP derivatives for tissue repair, regeneration, and disease are warranted.


Subject(s)
Cell Lineage/physiology , Chromatin Assembly and Disassembly/physiology , Transcription Factors/metabolism , Animals , Cell Differentiation/physiology , Chromatin/metabolism , Chromatin Assembly and Disassembly/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Epigenesis, Genetic/genetics , Erythroid Cells/metabolism , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mouse Embryonic Stem Cells/metabolism , Myeloid Cells/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism
7.
iScience ; 23(5): 101106, 2020 May 22.
Article in English | MEDLINE | ID: mdl-32434140

ABSTRACT

Epigenetic regulation, including chromatin accessibility and posttranslational modifications of histones, is of importance for T cell lineage decision. TH17 cells play a critical role in protective mucosal immunity and pathogenic multiple autoimmune diseases. The differentiation of TH17 cells is dictated by a master transcription factor, RORγt. However, the epigenetic mechanism that controls TH17 cell differentiation remains poorly understood. Here we show that the Swi/Snf complex is required for TH17-mediated cytokine production both in vitro and in vivo. We demonstrate that RORγt recruits and forms a complex with the Swi/Snf complex to cooperate for the RORγt-mediated epigenetic modifications of target genes, including both permissive and repressive ones for TH17 cell differentiation. Our findings thus highlight the Swi/Snf complex as an essential epigenetic regulator of TH17 cell differentiation and provide a basis for the understanding of how a master transcription factor RORγt collaborates with the Swi/Snf complex to govern epigenetic regulation.

8.
EMBO Rep ; 19(12)2018 12.
Article in English | MEDLINE | ID: mdl-30348891

ABSTRACT

Proper control of immune responses by Foxp3+ regulatory T cells at inflamed sites is crucial for the prevention of immunopathology. TGF-ß-induced Foxp3+ regulatory T (Treg) cells are generated in inflammatory environments as well as in steady-state conditions. Inflammatory cytokines such as IFN-γ and IL-4 have an antagonistic effect on Treg cell conversion. However, it is not known how naive CD4+ T cells overcome the inhibitory environment in inflamed sites to differentiate into Treg cells. Here, we show that CCAAT/enhancer-binding protein (C/EBP) functions as a safeguard that enhances Treg cell generation by dampening the inhibitory effect of IFN-γ and IL-4 on Foxp3 expression. We find that C/EBPß is induced by retinoic acid and binds to the methyl-CRE sequence in the Foxp3 TSDR to sustain its expression. C/EBPß-transduced iTreg cells show more potent suppressive activity in mouse disease models. We also reveal that C/EBPß-transduced human iTreg cells exhibit more enhanced suppressor function. These results establish C/EBP as a new molecular target for enhancing the formation and stability of Treg cells in inflammatory environments.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Forkhead Transcription Factors/metabolism , Inflammation/metabolism , Inflammation/pathology , T-Lymphocytes, Regulatory/metabolism , Adult , Animals , Base Sequence , Colitis/immunology , Colitis/pathology , Cytokines/pharmacology , DNA Methylation/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Mice, Transgenic , Protein Binding , Response Elements/genetics , T-Lymphocytes, Regulatory/drug effects , Tretinoin/pharmacology , Up-Regulation/drug effects
9.
iScience ; 4: 109-126, 2018 Jun 29.
Article in English | MEDLINE | ID: mdl-30240734

ABSTRACT

The abundance of basal progenitors (BPs), basal radial glia progenitors (bRGs) and basal intermediate progenitors (bIPs), in primate brain has been correlated to the high degree of cortical folding. Here we examined the role of BAF155, a subunit of the chromatin remodeling BAF complex, in generation of cortical progenitor heterogeneity. The conditional deletion of BAF155 led to diminished bIP pool and increased number of bRGs, due to delamination of apical RGs. We found that BAF155 is required for normal activity of neurogenic transcription factor PAX6, thus controlling the expression of genes that are involved in bIP specification, cell-cell interaction, and establishment of adherens junction. In a PAX6-dependent manner, BAF155 regulates the expression of the CDC42 effector protein CEP4, thereby controlling progenitor delamination. Furthermore, BAF155-dependent chromatin remodeling seems to exert a specific role in the genesis of BPs through the regulation of human RG-specific genes (such as Foxn4) that possibly acquired evolutionary significance.

10.
Stem Cell Reports ; 10(6): 1734-1750, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29779894

ABSTRACT

During early cortical development, neural stem cells (NSCs) divide symmetrically to expand the progenitor pool, whereas, in later stages, NSCs divide asymmetrically to self-renew and produce other cell types. The timely switch from such proliferative to differentiative division critically determines progenitor and neuron numbers. However, the mechanisms that limit proliferative division in late cortical development are not fully understood. Here, we show that the BAF (mSWI/SNF) complexes restrict proliferative competence and promote neuronal differentiation in late corticogenesis. Inactivation of BAF complexes leads to H3K27me3-linked silencing of neuronal differentiation-related genes, with concurrent H3K4me2-mediated activation of proliferation-associated genes via de-repression of Wnt signaling. Notably, the deletion of BAF complexes increased proliferation of neuroepithelial cell-like NSCs, impaired neuronal differentiation, and exerted a Wnt-dependent effect on neocortical and hippocampal development. Thus, these results demonstrate that BAF complexes act as both activators and repressors to control global epigenetic and gene expression programs in late corticogenesis.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Embryonic Development/genetics , Epigenesis, Genetic , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Ribonucleoproteins/metabolism , Wnt Signaling Pathway , Animals , Cell Differentiation , Cell Proliferation , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/genetics , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Hippocampus/embryology , Hippocampus/metabolism , Mice , Neurogenesis , Neurons/cytology , Neurons/metabolism , Protein Binding , Ribonucleoproteins/genetics
11.
Front Immunol ; 8: 1793, 2017.
Article in English | MEDLINE | ID: mdl-29326694

ABSTRACT

Dysfunction of T helper 17 (Th17) cells leads to chronic inflammatory disorders. Signal transducer and activator of transcription 3 (STAT3) orchestrates the expression of proinflammatory cytokines and pathogenic cell differentiation from interleukin (IL)-17-producing Th17 cells. However, the pathways mediated by STAT3 signaling are not fully understood. Here, we observed that Fos-related antigen 1 (FRA1) and JUNB are directly involved in STAT3 binding to sites in the promoters of Fosl1 and Junb. Promoter binding increased expression of IL-17 and the development of Th17 cells. Overexpression of Fra1 and Junb in mice resulted in susceptibility to collagen-induced arthritis and an increase in Th17 cell numbers and inflammatory cytokine production. In patients with rheumatoid arthritis, FRA1 and JUNB were colocalized with STAT3 in the inflamed synovium. These observations suggest that FRA1 and JUNB are associated closely with STAT3 activation, and that this activation leads to Th17 cell differentiation in autoimmune diseases and inflammation.

12.
PLoS Genet ; 12(9): e1006274, 2016 09.
Article in English | MEDLINE | ID: mdl-27611684

ABSTRACT

Neurogenesis is a key developmental event through which neurons are generated from neural stem/progenitor cells. Chromatin remodeling BAF (mSWI/SNF) complexes have been reported to play essential roles in the neurogenesis of the central nervous system. However, whether BAF complexes are required for neuron generation in the olfactory system is unknown. Here, we identified onscBAF and ornBAF complexes, which are specifically present in olfactory neural stem cells (oNSCs) and olfactory receptor neurons (ORNs), respectively. We demonstrated that BAF155 subunit is highly expressed in both oNSCs and ORNs, whereas high expression of BAF170 subunit is observed only in ORNs. We report that conditional deletion of BAF155, a core subunit in both onscBAF and ornBAF complexes, causes impaired proliferation of oNSCs as well as defective maturation and axonogenesis of ORNs in the developing olfactory epithelium (OE), while the high expression of BAF170 is important for maturation of ORNs. Interestingly, in the absence of BAF complexes in BAF155/BAF170 double-conditional knockout mice (dcKO), OE is not specified. Mechanistically, BAF complex is required for normal activation of Pax6-dependent transcriptional activity in stem cells/progenitors of the OE. Our findings unveil a novel mechanism mediated by the mSWI/SNF complex in OE neurogenesis and development.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , Neurogenesis , Olfactory Mucosa/metabolism , Transcription Factors/genetics , Animals , Cells, Cultured , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins , Gene Deletion , Mice , Mice, Inbred C57BL , Olfactory Mucosa/cytology , Olfactory Mucosa/embryology , Olfactory Receptor Neurons/cytology , Olfactory Receptor Neurons/metabolism , Transcription Factors/metabolism
13.
Development ; 143(8): 1271-83, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26952987

ABSTRACT

Dynamic control of gene expression is essential for the development of a totipotent zygote into an embryo with defined cell lineages. The accessibility of genes responsible for cell specification to transcriptional machinery is dependent on chromatin remodelling complexes such as the SWI\SNF (BAF) complex. However, the role of the BAF complex in early mouse development has remained unclear. Here, we demonstrate that BAF155, a major BAF complex subunit, regulates the assembly of the BAF complex in vivo and regulates lineage specification of the mouse blastocyst. We find that associations of BAF155 with other BAF complex subunits become enriched in extra-embryonic lineages just prior to implantation. This enrichment is attributed to decreased mobility of BAF155 in extra-embryonic compared with embryonic lineages. Downregulation of BAF155 leads to increased expression of the pluripotency marker Nanog and its ectopic expression in extra-embryonic lineages, whereas upregulation of BAF155 leads to the upregulation of differentiation markers. Finally, we show that the arginine methyltransferase CARM1 methylates BAF155, which differentially influences assembly of the BAF complex between the lineages and the expression of pluripotency markers. Together, our results indicate a novel role of BAF-dependent chromatin remodelling in mouse development via regulation of lineage specification.


Subject(s)
Cell Lineage/genetics , Embryonic Development/genetics , Epigenesis, Genetic , Transcription Factors/physiology , Animals , Blastocyst/cytology , Chromatin Assembly and Disassembly , Female , Gene Expression , Gene Expression Regulation, Developmental , Humans , Male , Methylation , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Protein-Arginine N-Methyltransferases/metabolism , Transcription Factors/genetics
14.
Cell Rep ; 13(9): 1842-54, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26655900

ABSTRACT

BAF (Brg/Brm-associated factors) complexes play important roles in development and are linked to chromatin plasticity at selected genomic loci. Nevertheless, a full understanding of their role in development and chromatin remodeling has been hindered by the absence of mutants completely lacking BAF complexes. Here, we report that the loss of BAF155/BAF170 in double-conditional knockout (dcKO) mice eliminates all known BAF subunits, resulting in an overall reduction in active chromatin marks (H3K9Ac), a global increase in repressive marks (H3K27me2/3), and downregulation of gene expression. We demonstrate that BAF complexes interact with H3K27 demethylases (JMJD3 and UTX) and potentiate their activity. Importantly, BAF complexes are indispensable for forebrain development, including proliferation, differentiation, and cell survival of neural progenitor cells. Our findings reveal a molecular mechanism mediated by BAF complexes that controls the global transcriptional program and chromatin state in development.


Subject(s)
Chromatin/metabolism , Chromosomal Proteins, Non-Histone/genetics , Transcription Factors/genetics , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cerebellar Cortex/metabolism , Chromosomal Proteins, Non-Histone/deficiency , Chromosomal Proteins, Non-Histone/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins , Down-Regulation , Embryo, Mammalian/metabolism , Histone Demethylases/genetics , Histone Demethylases/metabolism , Histones/genetics , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Nuclear Proteins/metabolism , Transcription Factors/deficiency , Transcription Factors/metabolism
15.
Eur J Immunol ; 45(1): 167-79, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25348202

ABSTRACT

Foxp3(+) Treg cells are crucial for maintaining T-cell homeostasis, but their role in B-cell homeostasis remains unclear. Here, we found that Foxp3 mutant scurfy mice had fewer B-lineage cells and progenitors, including common lymphoid progenitors and lymphoid-primed multipotent progenitors, but higher myeloid-lineage cell numbers in BM compared with WT littermates. Homeostasis within the HSC compartment was also compromised with apparent expansion of long- and short-term HSCs. This abnormality was due to the lack of Treg cells, but not to the Treg-cell extrinsic functions of Foxp3 or cell-autonomous defects. Among cytokines enriched in the BM of scurfy mice, IFN-γ affected only B lymphopoiesis, but GM-CSF, TNF, and IL-6 collectively promoted granulopoiesis at the expense of B lymphopoiesis. Neutralization of these three cytokines reversed the hematopoietic defects on early B-cell progenitors in scurfy mice. Treg cells ensured B lymphopoiesis by reducing the production of these cytokines by effector T cells, but not by directly affecting B lymphopoiesis. These results suggest that Treg cells occupy an important niche in the BM to protect B-lineage progenitor cells from excessive exposure to a lymphopoiesis-regulating milieu.


Subject(s)
B-Lymphocytes/cytology , Bone Marrow Cells/cytology , Forkhead Transcription Factors/immunology , Granulocytes/cytology , Stem Cells/cytology , T-Lymphocytes, Regulatory/cytology , Animals , Antibodies, Neutralizing/pharmacology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Cell Lineage/immunology , Cell Proliferation , Forkhead Transcription Factors/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocytes/drug effects , Granulocytes/immunology , Homeostasis/immunology , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/immunology , Interleukin-6/antagonists & inhibitors , Interleukin-6/immunology , Male , Mice , Mice, Transgenic , Primary Cell Culture , Stem Cells/drug effects , Stem Cells/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/immunology
16.
Mol Cells ; 36(4): 333-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23996527

ABSTRACT

Mammalian SWI/SNF complexes are evolutionary conserved, ATP-dependent chromatin remodeling units. BAF155 in the SWI/SNF complex contains several highly conserved domains, including SANT, SWIRM, and leucine zipper domains. The biological roles of the SWIRM domain remain unclear; however, both structural and biochemical analyses of this domain have suggested that it could mediate protein-protein or protein-DNA interactions during the chromatin remodeling process. The human BAF155 SWIRM domain was cloned into the Escherichia coli expression vector pMAL-c2X and purified using affinity chromatography for structural analysis. We report the backbone (1)H, (15)N, and (13)C resonance assignments and secondary structure of this domain using nuclear magnetic resonance (NMR) spectroscopy and the TALOS+ program. The secondary structure consists of five α-helices that form a typical histone fold for DNA interactions. Our data suggest that the BAF155 SWIRM domain interacts with nucleosome DNA (Kd = 0.47 µM).


Subject(s)
Chromatin/chemistry , Transcription Factors/chemistry , Amino Acid Sequence , Chromatin/metabolism , Chromatin Assembly and Disassembly , DNA-Binding Proteins/metabolism , Escherichia coli/genetics , Genetic Vectors , Humans , Magnetic Resonance Imaging , Molecular Sequence Data , Nucleosomes/metabolism , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Transcription Factors/metabolism
17.
J Immunol ; 188(8): 3791-803, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22427636

ABSTRACT

During the process of B cell development, transcription factors, such as E2A and Ebf1, have been known to play key roles. Although transcription factors and chromatin regulators work in concert to direct the expression of B lineage-specific genes, little is known about the involvement of regulators for chromatin structure during B lymphopoiesis. In this article, we show that deletion of Srg3/mBaf155, a scaffold subunit of the SWI/SNF-like BAF complex, in the hematopoietic lineage caused defects at both the common lymphoid progenitor stage and the transition from pre-pro-B to early pro-B cells due to failures in the expression of B lineage-specific genes, such as Ebf1 and Il7ra, and their downstream target genes. Moreover, mice that were deficient in the expression of Brg1, a subunit of the complex with ATPase activity, also showed defects in early B cell development. We also found that the expression of Ebf1 and Il7ra is directly regulated by the SWI/SNF-like BAF complex. Thus, our results suggest that the SWI/SNF-like BAF complex facilitates early B cell development by regulating the expression of B lineage-specific genes.


Subject(s)
Cell Differentiation/immunology , Chromatin/immunology , Chromosomal Proteins, Non-Histone/immunology , Precursor Cells, B-Lymphoid/immunology , Transcription Factors/immunology , Adenovirus E2 Proteins/genetics , Adenovirus E2 Proteins/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/immunology , Cell Differentiation/genetics , Cell Lineage/immunology , Chromatin/genetics , Chromosomal Proteins, Non-Histone/deficiency , Chromosomal Proteins, Non-Histone/genetics , DNA Helicases/deficiency , DNA Helicases/genetics , DNA Helicases/immunology , Gene Expression Regulation , Mice , Mice, Knockout , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Poly I-C/pharmacology , Precursor Cells, B-Lymphoid/cytology , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/immunology , Signal Transduction , Trans-Activators/genetics , Trans-Activators/immunology , Transcription Factors/deficiency , Transcription Factors/genetics
18.
Biochem Biophys Res Commun ; 411(3): 599-606, 2011 Aug 05.
Article in English | MEDLINE | ID: mdl-21763281

ABSTRACT

Recent reports have provided evidence for cross-talk between regulatory T (Treg) cells and natural killer T (NKT) cells. However, it is unclear whether NKT cells play a role in the differentiation of Treg cells. By employing NKT cell-abundant Vα14 TCR transgenic (Tg) and NKT cell-deficient CD1d knock-out (KO) mice, we examined the effects of NKT cells on the in vitro differentiation of induced Treg (iTreg) cells with IL2 and TGFß. We found that iTreg induction from CD1d KO mice was significantly increased compared to the control. Also, the addition of isolated NKT cells from Vα14 TCR Tg mice to naïve CD4(+) T cells from CD1d KO mice during iTreg differentiation caused a remarkable reduction of iTreg cells. Through IFNγ neutralization, we showed that this reduction was mediated by IFNγ. Furthermore, the main source of IFNγ during iTreg differentiation was NK1.1(-)CD4(+)Foxp3(-) T cells. This finding implied that early-activated NKT cells induced Th1-type cells and subsequently underwent apoptosis. Taken together, our results suggest that NKT cells inhibit the in vitro development of iTreg cells by increasing IFNγ.


Subject(s)
Interferon-gamma/immunology , Lymphocyte Activation , Natural Killer T-Cells/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, CD1d/genetics , CD4 Antigens/immunology , Cells, Cultured , Forkhead Transcription Factors/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Mice , Mice, Knockout , Receptors, Antigen, T-Cell, alpha-beta/genetics
19.
Exp Neurobiol ; 19(1): 39-48, 2010 Jun.
Article in English | MEDLINE | ID: mdl-22110340

ABSTRACT

SRG3 (SWI3-related gene) is a core subunit of mouse SWI/SNF complex and is known to play a critical role in stabilizing the SWI/SNF complex by attenuating its proteasomal degradation. SWI/SNF chromatin remodeling complex is reported to act as an important endogenous regulator in the proliferation and differentiation of mammalian neural stem cells. Because limited expression of SRG3 occurs in the brain and thymus during mouse embryogenesis, it was hypothesized that the altered SRG3 expression level might affect the process of adult hippocampal neurogenesis. Due to the embryonic lethality of homozygous knockout mice, this study focuses on dissecting the effect of overexpressed SRG3 on adult hippocampal neurogenesis. The BrdU incorporation assay, immunostaing with neuronal markers for each differentiation stage, and imunoblotting analysis with intracellular molecules involved in survival in adult hippocampal neurogenesis found no alteration, suggesting that the overexpression of SRG3 protein in mature neurons had no effect on the entire process of adult hippocampal neurogenesis including proliferation, differentiation, and survival.

20.
Mol Cells ; 28(6): 553-8, 2009 Dec 31.
Article in English | MEDLINE | ID: mdl-19937140

ABSTRACT

In the periphery, a galectin-1 receptor, CD7, plays crucial roles in galectin-1-mediated apoptosis of activated T-cells as well as progression of T-lymphoma. Previously, we demonstrated that NF-kappaB downregulated CD7 gene expression through the p38 MAPK pathway in developing immature thymocytes. However, its regulatory pathway is not well understood in functional mature T-cells. Here, we show that CD7 expression was downregulated by Twist2 in Jurkat cells, a human acute T-cell lymphoma cell line, and in EL4 cells, a mature murine T-cell lymphoma cell line. Furthermore, ectopic expression of Twist2 in Jurkat cells reduced galectin-1-induced apoptosis. While full-length Twist2 decreased CD7 promoter activity, a C-terminal deletion form of Twist2 reversed its inhibition, suggesting an important role of the C-terminus in CD7 regulation. In addition, CD7 expression was enhanced by histone deacetylase inhibitors such as trichostatin A and sodium butyrate, which indicates that Twist2 might be one of candidate factors involved in histone deacetylation. Based on these results, we conclude that upregulation of Twist2 increases the resistance to galectin-1-mediated-apoptosis, which may have significant implications for the progression of some T-cells into tumors such as Sezary cells.


Subject(s)
Antigens, CD7/metabolism , Repressor Proteins/metabolism , Sezary Syndrome/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/metabolism , Twist-Related Protein 1/metabolism , Animals , Antigens, CD7/genetics , Antigens, CD7/immunology , Apoptosis/drug effects , Apoptosis/genetics , Chromatin Assembly and Disassembly/drug effects , Chromatin Assembly and Disassembly/genetics , Disease Progression , Galectin 1/immunology , Galectin 1/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/immunology , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Jurkat Cells , Mice , Protein Engineering , Protein Structure, Tertiary/genetics , Repressor Proteins/genetics , Repressor Proteins/immunology , Sequence Deletion , Sezary Syndrome/genetics , Sezary Syndrome/pathology , Sezary Syndrome/physiopathology , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Transfection , Twist-Related Protein 1/genetics , Twist-Related Protein 1/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...