Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Nat Immunol ; 15(8): 767-76, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24973820

ABSTRACT

Regulatory T (Treg) cells suppress the development of inflammatory disease, but our knowledge of transcriptional regulators that control this function remains incomplete. Here we show that expression of Id2 and Id3 in Treg cells was required to suppress development of fatal inflammatory disease. We found that T cell antigen receptor (TCR)-driven signaling initially decreased the abundance of Id3, which led to the activation of a follicular regulatory T (TFR) cell-specific transcription signature. However, sustained lower abundance of Id2 and Id3 interfered with proper development of TFR cells. Depletion of Id2 and Id3 expression in Treg cells resulted in compromised maintenance and localization of the Treg cell population. Thus, Id2 and Id3 enforce TFR cell checkpoints and control the maintenance and homing of Treg cells.


Subject(s)
Inflammation/immunology , Inhibitor of Differentiation Protein 2/immunology , Inhibitor of Differentiation Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Base Sequence , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Movement/immunology , Cell Proliferation , Female , Forkhead Transcription Factors/biosynthesis , Gene Expression Regulation/immunology , Green Fluorescent Proteins/genetics , Inflammation/genetics , Inhibitor of Differentiation Protein 2/biosynthesis , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Proteins/biosynthesis , Inhibitor of Differentiation Proteins/genetics , Interleukin-10/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell/immunology , Receptors, CXCR5/biosynthesis , Sequence Analysis, RNA
2.
Immunity ; 46(5): 818-834.e4, 2017 05 16.
Article in English | MEDLINE | ID: mdl-28514688

ABSTRACT

Innate and adaptive lymphoid development is orchestrated by the activities of E proteins and their antagonist Id proteins, but how these factors regulate early T cell progenitor (ETP) and innate lymphoid cell (ILC) development remains unclear. Using multiple genetic strategies, we demonstrated that E proteins E2A and HEB acted in synergy in the thymus to establish T cell identity and to suppress the aberrant development of ILCs, including ILC2s and lymphoid-tissue-inducer-like cells. E2A and HEB orchestrated T cell fate and suppressed the ILC transcription signature by activating the expression of genes associated with Notch receptors, T cell receptor (TCR) assembly, and TCR-mediated signaling. E2A and HEB acted in ETPs to establish and maintain a T-cell-lineage-specific enhancer repertoire, including regulatory elements associated with the Notch1, Rag1, and Rag2 loci. On the basis of these and previous observations, we propose that the E-Id protein axis specifies innate and adaptive lymphoid cell fate.


Subject(s)
Adaptive Immunity , Immunity, Innate , Immunomodulation , Lymphocyte Subsets/immunology , Thymocytes/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/immunology , Cluster Analysis , Gene Expression , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Immunophenotyping , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/metabolism , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocyte Subsets/cytology , Lymphocyte Subsets/metabolism , Lymphoid Progenitor Cells/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Phenotype , T-Cell Antigen Receptor Specificity/immunology , Thymocytes/cytology , Thymocytes/metabolism , Transcriptome
3.
Nat Immunol ; 13(12): 1196-204, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23064439

ABSTRACT

The genome is folded into domains located in compartments that are either transcriptionally inert or transcriptionally permissive. Here we used genome-wide strategies to characterize domains during B cell development. Structured interaction matrix analysis showed that occupancy by the architectural protein CTCF was associated mainly with intradomain interactions, whereas sites bound by the histone acetyltransferase p300 or the transcription factors E2A or PU.1 were associated with intra- and interdomain interactions that are developmentally regulated. We identified a spectrum of genes that switched nuclear location during early B cell development. In progenitor cells, the transcriptionally inactive locus encoding early B cell factor (Ebf1) was sequestered at the nuclear lamina, which thereby preserved their multipotency. After development into the pro-B cell stage, Ebf1 and other genes switched compartments to establish new intra- and interdomain interactions associated with a B lineage-specific transcription signature.


Subject(s)
B-Lymphocytes/physiology , Cell Lineage , Cell Nucleus/genetics , Lymphopoiesis , Precursor Cells, B-Lymphoid/physiology , Animals , B-Lymphocytes/cytology , Basic Helix-Loop-Helix Transcription Factors/genetics , CCCTC-Binding Factor , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Mice , Mice, Inbred C57BL , Nuclear Lamina/metabolism , Precursor Cells, B-Lymphoid/cytology , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription, Genetic , p300-CBP Transcription Factors/genetics
4.
Adv Exp Med Biol ; 1459: 97-113, 2024.
Article in English | MEDLINE | ID: mdl-39017841

ABSTRACT

Helix-loop-helix (HLH) transcription factors (TFs) play a key role in various cellular differentiation and function through the regulation of enhancer activity. E2A, a member of the mammalian E-protein family (class I HLH protein), is well known to play an important role in hematopoiesis, especially in adaptive lymphocyte development. E2A instructs B- and T-cell lineage development through the regulation of enhancer activity for B- or T-cell signature gene expression, including Rag1 and Rag2 (Rag1/2) genes. In this chapter, we mainly focus on the function of E2A in B-cell development and on the roles of E2A in establishing the enhancer landscape through the recruitment of EP300/KAT3B, chromatin remodeling complex, mediator, cohesion, and TET proteins. Finally, we demonstrate how E2A orchestrates the assembly of the Rag1/2 gene super-enhancer (SE) formation by changing the chromatin conformation across the Rag gene locus.


Subject(s)
B-Lymphocytes , Homeodomain Proteins , Humans , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Enhancer Elements, Genetic/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Chromatin Assembly and Disassembly , Cell Differentiation/genetics , Chromatin/metabolism , Chromatin/genetics , E1A-Associated p300 Protein/metabolism , E1A-Associated p300 Protein/genetics , DNA-Binding Proteins , Nuclear Proteins
5.
Nat Immunol ; 12(10): 992-1001, 2011 Aug 21.
Article in English | MEDLINE | ID: mdl-21857655

ABSTRACT

It is established that the transcription factor E2A and its antagonist Id3 modulate the checkpoints consisting of the precursor to the T cell antigen receptor (pre-TCR) and the TCR. Here we demonstrate that Id3 expression was higher beyond the pre-TCR checkpoint, remained high in naive T cells and showed a bimodal pattern in the effector-memory population. We show how E2A promoted T lineage specification and how pre-TCR-mediated signaling affected E2A genome-wide occupancy. Thymi in Id3-deficient mice had aberrant development of effector-memory cells, higher expression of the chemokine receptor CXCR5 and the transcriptional repressor Bcl-6 and, unexpectedly, T cell-B cell conjugates and B cell follicles. Collectively, our data show how E2A acted globally to orchestrate development into the T lineage and that Id3 antagonized E2A activity beyond the pre-TCR checkpoint to enforce the naive fate of T cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Inhibitor of Differentiation Proteins/physiology , T-Lymphocytes/immunology , Animals , Immunologic Memory , Immunophenotyping , Leukocyte Common Antigens/analysis , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/physiology , Receptors, CXCR5/analysis , Spleen/immunology , Thymus Gland/immunology
6.
Genes Dev ; 29(4): 409-25, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25691468

ABSTRACT

It is now well established that the E and Id protein axis regulates multiple steps in lymphocyte development. However, it remains unknown how E and Id proteins mechanistically enforce and maintain the naïve T-cell fate. Here we show that Id2 and Id3 suppressed the development and expansion of innate variant follicular helper T (TFH) cells. Innate variant TFH cells required major histocompatibility complex (MHC) class I-like signaling and were associated with germinal center B cells. We found that Id2 and Id3 induced Foxo1 and Foxp1 expression to antagonize the activation of a TFH transcription signature. We show that Id2 and Id3 acted upstream of the Hif1a/Foxo/AKT/mTORC1 pathway as well as the c-myc/p19Arf module to control cellular expansion. We found that mice depleted for Id2 and Id3 expression developed colitis and αß T-cell lymphomas. Lymphomas depleted for Id2 and Id3 expression displayed elevated levels of c-myc, whereas p19Arf abundance declined. Transcription signatures of Id2- and Id3-depleted lymphomas revealed similarities to genetic deficiencies associated with Burkitt lymphoma. We propose that, in response to antigen receptor and/or cytokine signaling, the E-Id protein axis modulates the activities of the PI3K-AKT-mTORC1-Hif1a and c-myc/p19Arf pathways to control cellular expansion and homeostatic proliferation.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Differentiation , Inhibitor of Differentiation Proteins/metabolism , Lymphoma/physiopathology , T-Lymphocytes, Helper-Inducer/cytology , Thymocytes/cytology , Animals , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inhibitor of Differentiation Proteins/genetics , Lymphoid Tissue/cytology , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes/metabolism , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-myc/metabolism , STAT1 Transcription Factor , TOR Serine-Threonine Kinases/metabolism
7.
Int J Mol Sci ; 22(11)2021 May 31.
Article in English | MEDLINE | ID: mdl-34072618

ABSTRACT

Adaptive immunity relies on the V(D)J DNA recombination of immunoglobulin (Ig) and T cell receptor (TCR) genes, which enables the recognition of highly diverse antigens and the elicitation of antigen-specific immune responses. This process is mediated by recombination-activating gene (Rag) 1 and Rag2 (Rag1/2), whose expression is strictly controlled in a cell type-specific manner; the expression of Rag1/2 genes represents a hallmark of lymphoid lineage commitment. Although Rag genes are known to be evolutionally conserved among jawed vertebrates, how Rag genes are regulated by lineage-specific transcription factors (TFs) and how their regulatory system evolved among vertebrates have not been fully elucidated. Here, we reviewed the current body of knowledge concerning the cis-regulatory elements (CREs) of Rag genes and the evolution of the basic helix-loop-helix TF E protein regulating Rag gene CREs, as well as the evolution of the antagonist of this protein, the Id protein. This may help to understand how the adaptive immune system develops along with the evolution of responsible TFs and enhancers.


Subject(s)
Adaptive Immunity/genetics , Enhancer Elements, Genetic , Evolution, Molecular , Homeodomain Proteins/genetics , Transcription Factors/genetics , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Gene Expression Regulation , Humans , Regulatory Sequences, Nucleic Acid , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transcription Factors/metabolism , V(D)J Recombination
8.
Biochem Biophys Res Commun ; 526(1): 128-134, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32199616

ABSTRACT

Androgen receptor (AR)-negative castration-resistant prostate cancer (CRPC) is highly aggressive and is resistant to most of the current therapies. Bromodomain and extra terminal domain (BET) protein BRD4 binds to super-enhancers (SEs) that drive high expression of oncogenes in many cancers. A BET inhibitor, JQ1, has been found to suppress the malignant phenotypes of prostate cancer cells, however, the target genes of JQ1 remain largely unknown. Here we show that SE-associated genes specific for AR-negative CRPC PC3 cells include genes involved in migration and invasion, and that JQ1 impairs migration and invasion of PC3 cells. We identified a long non-coding RNA, MANCR, which was markedly down-regulated by JQ1, and found that BRD4 binds to the MANCR locus. MANCR knockdown led to a significant decrease in migration and invasion of PC3 cells. Furthermore, RNA sequencing analysis revealed that expression of the genes involved in migration and invasion was altered by MANCR knockdown. In summary, our data demonstrate that MANCR plays a critical role in migration and invasion of PC3 cells.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Movement , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Untranslated/metabolism , Transcription Factors/metabolism , Azepines/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Down-Regulation/drug effects , Down-Regulation/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Neoplasm Invasiveness , Prostatic Neoplasms/genetics , RNA, Untranslated/genetics , Triazoles/pharmacology
9.
Trends Immunol ; 35(5): 205-10, 2014 May.
Article in English | MEDLINE | ID: mdl-24679436

ABSTRACT

In B cell progenitors, E-proteins E2A and HEB (HeLa E-box binding protein) are crucial for the induction of a B lineage-specific program of gene expression and for orchestrating the assembly of the immunoglobulin loci. In the thymus E2A and HEB act differently, activating the expression of genes closely associated with the establishment of T cell identity and promoting the rearrangement of T cell receptor (TCR) loci. These findings have raised the question as to how E-proteins exert these different activities. We review here the distinct regulatory networks that establish B versus T cell identity, and how genomic architecture and location of genes is modulated in these lineage decisions. We conclude by proposing a model wherein stochasticity in the nuclear location of the early B cell factor 1 (Ebf1) locus in multipotent progenitors determines this lineage choice.


Subject(s)
B-Lymphocytes/metabolism , Cell Lineage , T-Lymphocytes/metabolism , Alleles , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Lineage/genetics , Cell Lineage/immunology , Gene Expression Regulation , Gene Regulatory Networks , Genome , Humans , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Transcription, Genetic
10.
J Immunol ; 188(11): 5547-60, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22544934

ABSTRACT

V(D)J recombination of Ig and TCR genes is strictly regulated in a lineage- and stage-specific manner by the accessibility of target gene chromatin to the recombinases RAG1 and RAG2. It has been shown that enforced expression of the basic helix-loop-helix protein, E2A, together with RAG1/2 in a nonlymphoid cell line BOSC23 can induce V(D)J recombination in endogenous Igκ and TCR loci by increasing chromatin accessibility of target gene segments. In this study, we demonstrate that ectopically expressed E2A proteins in BOSC23 cells have the ability to bind directly to the promoter and recombination signal sequence of Vκ genes and to recruit histone acetyltransferase CBP/p300. Overexpression of CBP/p300 in conjunction with E2A results in enhancement of E2A-induced histone acetylation, germline transcription, and Igκ rearrangement. Conversely, knockdown of endogenous CBP/p300 expression by small interfering RNA leads to a decrease in histone acetylation, germline transcription and Igκ rearrangement. Furthermore, analyses using a mouse pre-B cell line revealed that endogenous E2A proteins also bind to a distinct set of Vκ genes and regulatory regions in the mouse Igκ locus and act to increase histone acetylation by recruiting p300, confirming the similar findings observed with BOSC23 cells. These observations indicate that E2A plays critical roles in inducing Igκ rearrangement by directly binding to and increasing chromatin accessibility at target gene segments.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Chromatin/genetics , Immunoglobulin kappa-Chains/genetics , p300-CBP Transcription Factors/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line , Chromatin/metabolism , Drug Synergism , Enhancer Elements, Genetic/genetics , Enhancer Elements, Genetic/immunology , Germ Cells/enzymology , Germ Cells/immunology , Germ Cells/metabolism , Histone Deacetylases/metabolism , Humans , Immunoglobulin kappa-Chains/metabolism , Mice , V(D)J Recombination/genetics , p300-CBP Transcription Factors/biosynthesis , p300-CBP Transcription Factors/genetics
11.
Proc Natl Acad Sci U S A ; 108(6): 2468-73, 2011 Feb 08.
Article in English | MEDLINE | ID: mdl-21252303

ABSTRACT

To clarify the molecular pathways governing hematopoietic stem cell (HSC) development, we screened a fetal liver (FL) HSC cDNA library and identified a unique gene, hematopoietic expressed mammalian polycomb (hemp), encoding a protein with a zinc-finger domain and four malignant brain tumor (mbt) repeats. To investigate its biological role, we generated mice lacking Hemp (hemp(-/-)). Hemp(-/-) mice exhibited a variety of skeletal malformations and died soon after birth. In the FL, hemp was preferentially expressed in the HSC and early progenitor cell fractions, and analyses of fetal hematopoiesis revealed that the number of FL mononuclear cells, including HSCs, was reduced markedly in hemp(-/-) embryos, especially during early development. In addition, colony-forming and competitive repopulation assays demonstrated that the proliferative and reconstitution abilities of hemp(-/-) FL HSCs were significantly impaired. Microarray analysis revealed alterations in the expression levels of several genes implicated in hematopoietic development and differentiation in hemp(-/-) FL HSCs. These results demonstrate that Hemp, an mbt-containing protein, plays essential roles in HSC function and skeletal formation. It is also hypothesized that Hemp might be involved in certain congenital diseases, such as Klippel-Feil anomaly.


Subject(s)
Bone Development/physiology , Bone and Bones/embryology , Chromosomal Proteins, Non-Histone/metabolism , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental/physiology , Hematopoietic Stem Cells/metabolism , Repressor Proteins/metabolism , Animals , Cell Differentiation/physiology , Chromosomal Proteins, Non-Histone/genetics , Embryo, Mammalian/cytology , Gene Expression Profiling , Hematopoiesis/physiology , Klippel-Feil Syndrome/genetics , Klippel-Feil Syndrome/metabolism , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Repressor Proteins/genetics
12.
Cell Rep ; 42(9): 113127, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37729919

ABSTRACT

Natural killer (NK) cells are innate immune cells critical for protective immune responses against infection and cancer. Although NK cells differentiate in the bone marrow (BM) in an interleukin-15 (IL-15)-dependent manner, the cellular source of IL-15 remains elusive. Using NK cell reporter mice, we show that NK cells are localized in the BM in scattered and clustered manners. NK cell clusters overlap with monocyte and dendritic cell accumulations, whereas scattered NK cells require CXCR4 signaling. Using cell-specific IL-15-deficient mice, we show that hematopoietic cells, but not stromal cells, support NK cell development in the BM through IL-15. In particular, IL-15 produced by monocytes and dendritic cells appears to contribute to NK cell development. These results demonstrate that hematopoietic cells are the IL-15 niche for NK cell development in the BM and that BM NK cells are present in scattered and clustered compartments by different mechanisms, suggesting their distinct functions in the immune response.


Subject(s)
Bone Marrow , Interleukin-15 , Mice , Animals , Bone Marrow Cells , Cell Differentiation , Killer Cells, Natural
13.
J Immunol ; 185(12): 7330-9, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21057086

ABSTRACT

Naturally occurring regulatory T (Treg) cells play a central role in the maintenance of immune homeostasis and in restraining the development of spontaneous inflammatory responses. However, the underlying mechanisms of Treg homeostasis remain incompletely understood. Of particular note, the IL-2Rα (CD25) is crucial for the homeostasis of Treg cells and the prevention of lymphoproliferative autoimmune disease. In this paper, we report that the basic helix-loop-helix transcription factor Dec1 is involved in the homeostasis of Treg cells and plays a role in their survival or expansion after adoptive transfer to lymphopenic recipients. Hence, it is crucial for the suppression of effector T cell-mediated inflammatory responses. Enforced expression of Dec1 upregulates CD25 expression during thymocyte development and increases the number of Treg cells in the periphery. Dec1 binds the transcription factor Runx1 and colocalizes with Runx1 in Treg cells. Specifically, we demonstrate that in Treg cells the Dec1/Runx1 complex binds to regulatory elements present in the Il-2rα locus. Collectively, these data show how Dec1 mechanistically acts in Treg cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/immunology , Homeodomain Proteins/immunology , Homeostasis/physiology , Interleukin-2 Receptor alpha Subunit/immunology , Up-Regulation/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Survival/physiology , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/immunology , Core Binding Factor Alpha 2 Subunit/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , Mice , Mice, Transgenic , Protein Binding/genetics , Protein Binding/immunology , T-Lymphocytes, Regulatory
14.
J Biochem ; 172(5): 259-264, 2022 Oct 19.
Article in English | MEDLINE | ID: mdl-36000775

ABSTRACT

Our bodies are constantly threatened with the invasion of pathogens, such as bacteria and virus. Immune responses against pathogens are evoked in collaboration with adaptive and innate immune systems. Adaptive immune cells including T and B cells recognize various antigens from pathogens through the antigen recognition receptors such as immunoglobulin (Ig) and T-cell receptor (TCR), and they evoke antigen-specific immune responses to eliminate the pathogens. This specific recognition of a variety of antigens relies on the V(D)J DNA recombination of Ig and TCR genes, which is generated by the Rag (recombination activation gene) 1/Rag2 protein complex. The expression of Rag1/2 genes is stringently controlled during the T and B cell development; Rag1/2 gene expression indicates the commitment towards adaptive lymphocyte lineages. In this review article, we will discuss the developmental bifurcation between adaptive and innate lymphoid cells, and the role of transcription factors, especially the E and Id proteins, upon the lineage commitment, and the regulation of Rag gene locus.


Subject(s)
Immunity, Innate , Lymphocytes , Lymphocytes/metabolism , Transcription Factors , Receptors, Antigen, T-Cell , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism
15.
Front Immunol ; 13: 890056, 2022.
Article in English | MEDLINE | ID: mdl-35603170

ABSTRACT

Immune responses are primarily mediated by adaptive and innate immune cells. Adaptive immune cells, such as T and B cells, evoke antigen-specific responses through the recognition of specific antigens. This antigen-specific recognition relies on the V(D)J recombination of immunoglobulin (Ig) and T cell receptor (TCR) genes mediated by recombination-activating gene (Rag)1 and Rag2 (Rag1/2). In addition, T and B cells employ cell type-specific developmental pathways during their activation processes, and the regulation of these processes is strictly regulated by the transcription factor network. Among these factors, members of the basic helix-loop-helix (bHLH) transcription factor mammalian E protein family, including E12, E47, E2-2, and HEB, orchestrate multiple adaptive immune cell development, while their antagonists, Id proteins (Id1-4), function as negative regulators. It is well established that a majority of T and B cell developmental trajectories are regulated by the transcriptional balance between E and Id proteins (the E-Id axis). E2A is critically required not only for B cell but also for T cell lineage commitment, whereas Id2 and Id3 enforce the maintenance of naïve T cells and naïve regulatory T (Treg) cells. Here, we review the current knowledge of E- and Id-protein function in T cell lineage commitment and Treg cell differentiation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Transcription Factors , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Hematopoiesis , Lymphocyte Activation , Mammals/metabolism , Transcription Factors/metabolism
16.
PLoS One ; 17(9): e0273683, 2022.
Article in English | MEDLINE | ID: mdl-36083881

ABSTRACT

Surgical site infection (SSI) is a common but potentially serious maternal complication of cesarean section (CS). C-reactive protein (CRP) can be used in early detection of SSI. However, its predictive value for post-cesarean SSI has never been investigated. This study aims to evaluate the predictive value of CRP for the development of SSI. This was a hospital-based retrospective cohort study of 748 pregnant women who underwent CS at our university hospital between January 2017 and December 2019. CRP was measured on postoperative days 1, 3, and 6. The predictive values of CRP for SSI were evaluated using receiver operating characteristics analysis. Forty-seven (6.3%) patients developed SSI, of whom 38 (80.9%) underwent emergency CS. Serum CRP levels were significantly higher in the SSI group than in the non-SSI group from postoperative day 1 (64 vs. 81 mg/L, p = 0.001); the difference became more evident on postoperative days 3 and 6. The area under the receiver operating characteristic curve (AUC) for CRP on days 1, 3, and 6 was 0.58 (95% confidence interval [CI], 0.49 to 0.68), 0.70 (0.62 to 0.78) and 0.73 (0.65 to 0.81), respectively. The optimal cutoff value for day 3 and 6 CRP was 66.4 mg/L (sensitivity = 76.1% and specificity = 54.4%) and 22.2 mg/L (sensitivity = 76.5% and specificity = 63.2%), respectively. CRP on postoperative days 3 and 6 can be used as a predictive marker for the development of SSI after CS. Further studies to validate the predictive value in different populations is essential.


Subject(s)
C-Reactive Protein , Surgical Wound Infection , Biomarkers , C-Reactive Protein/metabolism , Cesarean Section/adverse effects , Female , Humans , Japan , Pregnancy , ROC Curve , Retrospective Studies , Surgical Wound Infection/diagnosis , Surgical Wound Infection/etiology
17.
Dev Biol ; 345(2): 226-36, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20647008

ABSTRACT

Although internal ribosome entry site (IRES)-mediated translation is considered important for proper cellular function, its precise biological role is not fully understood. Runx1 gene, which encodes a transcription factor implicated in hematopoiesis, angiogenesis, and leukemogenesis, contains IRES sequences in the 5' untranslated region. To clarify the roles of the IRES element in Runx1 function, we generated knock-in mice for either wild-type Runx1 or Runx1/Evi1, a Runx1 fusion protein identified in human leukemia. In both cases, native promoter-dependent transcription was retained, whereas IRES-mediated translation was eliminated. Interestingly, homozygotes expressing wild-type Runx1 deleted for the IRES element (Runx1(Delta IRES/Delta IRES)) died in utero with prominent dilatation of peripheral blood vessels due to impaired pericyte development. In addition, hematopoietic cells in the Runx1(Delta IRES/Delta IRES) fetal liver were significantly decreased, and exhibited an altered differentiation pattern, a reduced proliferative activity, and an impaired reconstitution ability. On the other hand, heterozygotes expressing Runx1/Evi1 deleted for the IRES element (Runx1(+/RE Delta IRES)) were born normally and did not show any hematological abnormalities, in contrast that conventional Runx1/Evi1 heterozygotes die in utero with central nervous system hemorrhage and Runx1/Evi1 chimeric mice develop acute leukemia. The findings reported here demonstrate the essential roles of the IRES element in Runx1 function under physiological and pathological conditions.


Subject(s)
5' Untranslated Regions , Core Binding Factor Alpha 2 Subunit/genetics , Hematopoiesis/genetics , Leukemia/genetics , Neovascularization, Physiologic/genetics , Animals , Cell Differentiation , Gene Expression Regulation, Leukemic , Gene Knock-In Techniques , Heterozygote , Liver/pathology , Mice , Models, Animal , Ribosomes/metabolism
18.
Blood ; 113(19): 4702-10, 2009 May 07.
Article in English | MEDLINE | ID: mdl-19234145

ABSTRACT

Chronic myelogenous leukemia (CML) is a hematopoietic disorder originating from p210BCR/ABL-transformed stem cells, which begins as indolent chronic phase (CP) but progresses into fatal blast crisis (BC). To investigate molecular mechanism(s) underlying disease evolution, CML-exhibiting p210BCR/ABL transgenic mice were crossed with BXH2 mice that transmit a replication-competent retrovirus. Whereas nontransgenic mice in the BXH2 background exclusively developed acute myeloid leukemia, p210BCR/ABL transgenic littermates developed nonmyeloid leukemias, in which inverse polymerase chain reaction detected 2 common viral integration sites (CISs). Interestingly, one CIS was transgene's own promoter, which up-regulated p210BCR/ABL expression. The other was the 5' noncoding region of a transcription factor, Zfp423, which induced aberrant Zfp423 expression. The cooperative activities of Zfp423 and p210BCR/ABL were demonstrated as follows: (1) introduction of Zfp423 in p210BCR/ABL transgenic bone marrow (BM) cells increased colony-forming ability, (2) suppression of ZNF423 (human homologue of Zfp423) in ZNF423-expressing, p210BCR/ABL-positive hematopoietic cells retarded cell growth, (3) mice that received a transplant of BM cells transduced with Zfp423 and p210BCR/ABL developed acute leukemia, and (4) expression of ZNF423 was found in human BCR/ABL-positive cell lines and CML BC samples. These results demonstrate that enhanced expression of p210BCR/ABL and deregulated expression of Zfp423/ZNF423 contribute to CML BC.


Subject(s)
B-Lymphocytes/pathology , Blast Crisis/genetics , DNA-Binding Proteins/physiology , Fusion Proteins, bcr-abl/physiology , Gene Expression Regulation, Leukemic , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Transcription Factors/physiology , Animals , Blotting, Northern , Blotting, Southern , Blotting, Western , Bone Marrow Transplantation , Cell Proliferation , Colony-Forming Units Assay , DNA-Binding Proteins/antagonists & inhibitors , Female , Flow Cytometry , Gene Rearrangement , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Male , Mice , Mice, Knockout , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Retroviridae , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Transcription Factors/antagonists & inhibitors , Zinc Fingers
19.
Front Immunol ; 12: 659761, 2021.
Article in English | MEDLINE | ID: mdl-33796120

ABSTRACT

Cell type-specific gene expression is driven through the interplay between lineage-specific transcription factors (TFs) and the chromatin architecture, such as topologically associating domains (TADs), and enhancer-promoter interactions. To elucidate the molecular mechanisms of the cell fate decisions and cell type-specific functions, it is important to understand the interplay between chromatin architectures and TFs. Among enhancers, super-enhancers (SEs) play key roles in establishing cell identity. Adaptive immunity depends on the RAG-mediated assembly of antigen recognition receptors. Hence, regulation of the Rag1 and Rag2 (Rag1/2) genes is a hallmark of adaptive lymphoid lineage commitment. Here, we review the current knowledge of 3D genome organization, SE formation, and Rag1/2 gene regulation during B cell and T cell differentiation.


Subject(s)
Cell Lineage/immunology , Chromatin Assembly and Disassembly/immunology , Chromatin/immunology , DNA-Binding Proteins/immunology , Homeodomain Proteins/immunology , Nuclear Proteins/immunology , Transcription Factors/immunology , Adaptive Immunity/genetics , Adaptive Immunity/immunology , Cell Lineage/genetics , Chromatin/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation/immunology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
20.
Sci Immunol ; 5(51)2020 09 04.
Article in English | MEDLINE | ID: mdl-32887843

ABSTRACT

Cell type-specific gene expression is driven by the interplay between lineage-specific transcription factors and cis-regulatory elements to which they bind. Adaptive immunity relies on RAG-mediated assembly of T cell receptor (TCR) and immunoglobulin (Ig) genes. Although Rag1 and Rag2 expression is largely restricted to adaptive lymphoid lineage cells, it remains unclear how Rag gene expression is regulated in a cell lineage-specific manner. Here, we identified three distinct cis-regulatory elements, a T cell lineage-specific enhancer (R-TEn) and the two B cell-specific elements, R1B and R2B By generating mice lacking either R-TEn or R1B and R2B, we demonstrate that these distinct sets of regulatory elements drive the expression of Rag genes in developing T and B cells. What these elements have in common is their ability to bind the transcription factor E2A. By generating a mouse strain that carries a mutation within the E2A binding site of R-TEn, we demonstrate that recruitment of E2A to this site is essential for orchestrating changes in chromatin conformation that drive expression of Rag genes in T cells. By mapping cis-regulatory elements and generating multiple mouse strains lacking distinct enhancer elements, we demonstrate expression of Rag genes in developing T and B cells to be driven by distinct sets of E2A-dependent cis-regulatory modules.


Subject(s)
B-Lymphocytes/immunology , Basic Helix-Loop-Helix Transcription Factors/immunology , DNA-Binding Proteins/immunology , Homeodomain Proteins/immunology , T-Lymphocytes/immunology , Animals , Animals, Genetically Modified , Basic Helix-Loop-Helix Transcription Factors/genetics , DNA-Binding Proteins/genetics , Female , Homeodomain Proteins/genetics , Male , Mice
SELECTION OF CITATIONS
SEARCH DETAIL