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










Publication year range
1.
Mol Ther Oncolytics ; 24: 77-86, 2022 Mar 17.
Article in English | MEDLINE | ID: mdl-35024435

ABSTRACT

To develop effective adoptive cell transfer therapy using T cell receptor (TCR)-engineered T cells, it is critical to isolate tumor-reactive TCRs that have potent anti-tumor activity. In humans, tumor-infiltrating lymphocytes (TILs) have been reported to contain CD8+PD-1+ T cells that express tumor-reactive TCRs. Characterization of tumor reactivity of TILs from non-human primate tumors could improve anti-tumor activity of TCR-engineered T cells in preclinical research. In this study, we sought to isolate TCR genes from CD8+PD-1+ T cells among TILs in a cynomolgus macaque model of tumor transplantation in which the tumors were infiltrated with CD8+ T cells and were eventually rejected. We analyzed the repertoire of TCRα and ß pairs obtained from single CD8+PD-1+ T cells in TILs and circulating lymphocytes and identified multiple TCR pairs with high frequency, suggesting that T cells expressing these recurrent TCRs were clonally expanded in response to tumor cells. We further showed that the recurrent TCRs exhibited cytotoxic activity to tumor cells in vitro and potent anti-tumor activity in mice transplanted with tumor cells. These results imply that this tumor transplantation macaque model recapitulates key features of human TILs and can serve as a platform toward preclinical studies of non-human primate tumor models.

2.
Mol Ther Methods Clin Dev ; 19: 250-260, 2020 Dec 11.
Article in English | MEDLINE | ID: mdl-33102617

ABSTRACT

In the current adoptive T cell therapy, T cells from a patient are given back to that patient after ex vivo activation, expansion, or genetic manipulation. However, such strategy depends on the quality of the patient's T cells, sometimes leading to treatment failure. It would therefore be ideal to use allogeneic T cells as "off-the-shelf" T cells. To this aim, we have been developing a strategy where potent tumor-antigen-specific cytotoxic T lymphocytes (CTLs) are regenerated from T-cell-derived induced pluripotent stem cells (T-iPSCs). However, certain issues still remain that make it difficult to establish highly potent T-iPSCs: poor reprogramming efficiency of T cells into iPSCs and high variability in the differentiation capability of each T-iPSC clone. To expand the versatility of this approach, we thought of a method to produce iPSCs equivalent to T-iPSCs, namely, iPSCs transduced with exogenous T cell receptor (TCR) genes (TCR-iPSCs). To test this idea, we first cloned TCR genes from WT1-specific CTLs regenerated from T-iPSCs and then established WT1-TCR-iPSCs. We show that the regenerated CTLs from TCR-iPSCs exerted cytotoxic activity comparable to those from T-iPSCs against WT1 peptide-loaded cell line in in vitro model. These results collectively demonstrate the feasibility of the TCR-iPSC strategy.

3.
Sci Rep ; 10(1): 8414, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32439888

ABSTRACT

Immunotherapy has emerged as a promising and effective treatment for cancer, yet the clinical benefit is still variable, in part due to insufficient accumulation of immune effector cells in the tumour microenvironment. Better understanding of tumour-infiltrating lymphocytes (TILs) from nonhuman primate tumours could provide insights into improving effector cell accumulation in tumour tissues during immunotherapy. Here, we characterize TILs in a cynomolgus macaque tumour model in which the tumours were infiltrated with CD4+ and CD8+ T cells and were eventually rejected. The majority of CD4+ and CD8+ TILs exhibited a CD45RA-CCR7- effector memory phenotype, but unlike circulating T cells, they expressed CD69, a marker for tissue-resident memory T (TRM) cells. CD69-expressing CD8+ TILs expressed high levels of the cytotoxic molecule granzyme B and the co-inhibitory receptor PD-1. Consistent with the TRM cell phenotype, CD8+ TILs minimally expressed CX3CR1 but expressed CXCR3 at higher levels than circulating CD8+ T cells. Meanwhile, CXCL9, CXCL10 and CXCL11, chemokine ligands for CXCR3, were expressed at high levels in the tumours, thus attracting CXCR3+CD8+ T cells. These results indicate that tumour-transplanted macaques can be a useful preclinical model for studying and optimizing T cell accumulation in tumours for the development of new immunotherapies.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive/methods , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms/immunology , Animals , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , CD4-Positive T-Lymphocytes/transplantation , CD8-Positive T-Lymphocytes/transplantation , CX3C Chemokine Receptor 1/metabolism , Cell Line, Tumor , Chemokine CXCL10/metabolism , Chemokine CXCL11/metabolism , Chemokine CXCL9/metabolism , Lectins, C-Type/metabolism , Lymphocytes, Tumor-Infiltrating/transplantation , Macaca fascicularis , Models, Animal , Neoplasms/therapy , Receptors, CXCR3/metabolism
4.
iScience ; 23(4): 100998, 2020 Apr 24.
Article in English | MEDLINE | ID: mdl-32259478

ABSTRACT

Current adoptive T cell therapies conducted in an autologous setting are costly, time consuming, and depend on the quality of the patient's T cells. To address these issues, we developed a strategy in which cytotoxic T lymphocytes (CTLs) are regenerated from iPSCs that were originally derived from T cells and succeeded in regenerating CTLs specific for the WT1 antigen, which exhibited therapeutic efficacy in a xenograft model of leukemia. In this study, we extended our strategy to solid tumors. The regenerated WT1-specific CTLs had a strong therapeutic effect in orthotopic xenograft model using a renal cell carcinoma (RCC) cell line. To make our method more generally applicable, we developed an allogeneic approach by transducing HLA-haplotype homozygous iPSCs with WT1-specific TCR α/ß genes that had been tested clinically. The regenerated CTLs antigen-specifically suppressed tumor growth in a patient-derived xenograft model of RCC, demonstrating the feasibility of our strategy against solid tumors.

5.
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
6.
Genes Dev ; 32(2): 112-126, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29440259

ABSTRACT

Stem cell fate is orchestrated by core transcription factors (TFs) and epigenetic modifications. Although regulatory genes that control cell type specification are identified, the transcriptional circuit and the cross-talk among regulatory factors during cell fate decisions remain poorly understood. To identify the "time-lapse" TF networks during B-lineage commitment, we used multipotent progenitors harboring a tamoxifen-inducible form of Id3, an in vitro system in which virtually all cells became B cells within 6 d by simply withdrawing 4-hydroxytamoxifen (4-OHT). Transcriptome and epigenome analysis at multiple time points revealed that ∼10%-30% of differentially expressed genes were virtually controlled by the core TFs, including E2A, EBF1, and PAX5. Strikingly, we found unexpected transcriptional priming before the onset of the key TF program. Inhibition of the immediate early genes such as Nr4a2, Klf4, and Egr1 severely impaired the generation of B cells. Integration of multiple data sets, including transcriptome, protein interactome, and epigenome profiles, identified three representative transcriptional circuits. Single-cell RNA sequencing (RNA-seq) analysis of lymphoid progenitors in bone marrow strongly supported the three-step TF network model during specification of multipotent progenitors toward B-cell lineage in vivo. Thus, our findings will provide a blueprint for studying the normal and neoplastic development of B lymphocytes.


Subject(s)
B-Lymphocytes/metabolism , Multipotent Stem Cells/metabolism , Transcription, Genetic , Animals , Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Lineage/genetics , Cells, Cultured , Epigenesis, Genetic , Gene Expression Profiling , Gene Regulatory Networks , Histone Code , Kruppel-Like Factor 4 , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , PAX5 Transcription Factor/physiology , Single-Cell Analysis , Trans-Activators/physiology , Transcriptome
7.
Sci Rep ; 7: 45175, 2017 03 24.
Article in English | MEDLINE | ID: mdl-28338057

ABSTRACT

Many metazoans start germ cell development during embryogenesis, while some metazoans possessing pluripotent stem cells undergo postembryonic germ cell development. The latter reproduce asexually but develop germ cells from pluripotent stem cells or dormant primordial germ cells when they reproduce sexually. Sexual induction of the planarian Dugesia ryukyuensis is an important model for postembryonic germ cell development. In this experimental system, hermaphroditic reproductive organs are differentiated in presumptive gonadal regions by the administration of a crude extract from sexual planarians to asexual ones. However, the substances involved in the first event during postembryonic germ cell development, i.e., ovarian development, remain unknown. Here, we aimed to identify a bioactive compound associated with postembryonic ovarian development. Bioassay-guided fractionation identified ʟ-tryptophan (Trp) on the basis of electrospray ionization-mass spectrometry, circular dichroism, and nuclear magnetic resonance spectroscopy. Originally masked by a large amount of ʟ-Trp, ᴅ-Trp was detected by reverse-phase high-performance liquid chromatography. The ovary-inducing activity of ᴅ-Trp was 500 times more potent than that of ʟ-Trp. This is the first report describing a role for an intrinsic ᴅ-amino acid in postembryonic germ cell development. Our findings provide a novel insight into the mechanisms of germ cell development regulated by low-molecular weight bioactive compounds.


Subject(s)
Oogenesis , Ovary/metabolism , Planarians/metabolism , Tryptophan/metabolism , Animals , Female , Hermaphroditic Organisms/growth & development , Hermaphroditic Organisms/metabolism , Male , Ovary/cytology , Ovary/diagnostic imaging , Planarians/growth & development
8.
Genes Dev ; 30(22): 2475-2485, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27913604

ABSTRACT

In general, cell fate is determined primarily by transcription factors, followed by epigenetic mechanisms fixing the status. While the importance of transcription factors controlling cell fate has been well characterized, epigenetic regulation of cell fate maintenance remains to be elucidated. Here we provide an obvious fate conversion case, in which the inactivation of polycomb-medicated epigenetic regulation results in conversion of T-lineage progenitors to the B-cell fate. In T-cell-specific Ring1A/B-deficient mice, T-cell development was severely blocked at an immature stage. We found that these developmentally arrested T-cell precursors gave rise to functional B cells upon transfer to immunodeficient mice. We further demonstrated that the arrest was almost completely canceled by additional deletion of Pax5 These results indicate that the maintenance of T-cell fate critically requires epigenetic suppression of the B-lineage gene program.


Subject(s)
B-Lymphocytes/cytology , Cell Transformation, Neoplastic/genetics , Epigenesis, Genetic/genetics , Gene Silencing , Polycomb-Group Proteins/metabolism , T-Lymphocytes/cytology , Animals , Cell Lineage , Gene Deletion , Gene Expression Regulation, Developmental , Immunoglobulin Heavy Chains/genetics , Mice, Inbred C57BL , PAX5 Transcription Factor/genetics , PAX5 Transcription Factor/metabolism , Polycomb Repressive Complex 1/genetics , Promoter Regions, Genetic/genetics , Ubiquitin-Protein Ligases/genetics
9.
Biol Reprod ; 94(2): 30, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26658710

ABSTRACT

Spermatogenesis is controlled by hormonal secretions from the hypothalamus and pituitary gland, by factors produced locally in the testis, and by direct interaction between germ cells and Sertoli cells in seminiferous tubules. Although the mammalian testis contains high levels of D-aspartate (D-Asp), and D-Asp is known to stimulate the secretion of testosterone in cultured Leydig cells, its role in testis is unclear. We describe here biochemical, immunohistochemical, and flow cytometric studies designed to elucidate developmental changes in testicular D-Asp levels and the direct effect of D-Asp on germ cells. We found that the concentration of D-Asp in mouse testis increased with growth and that fluctuations in D-Asp levels were controlled in part by its degradative enzyme, D-aspartate oxidase expressed in Sertoli cells. In vitro sperm production studies showed that mitosis in premeiotic germ cells was strongly inhibited by the addition of D-Asp to the culture medium. Moreover, immunohistochemical analysis demonstrated that d-Asp accumulated in the differentiated spermatids, indicating either transport of D-Asp to spermatids or its de novo synthesis in these cells. Such compartmentation seems to prevent premeiotic germ cells in mouse testis from being exposed to the excess amount of D-Asp. In concert, our results indicate that in mouse testis, levels of D-Asp are regulated in a spatiotemporal manner and that D-Asp functions as a modulator of spermatogenesis.


Subject(s)
D-Aspartic Acid/pharmacology , Spermatogenesis/drug effects , Spermatozoa/drug effects , Testis/drug effects , Animals , D-Aspartic Acid/metabolism , Male , Mice , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Spermatogenesis/physiology , Spermatozoa/cytology , Spermatozoa/metabolism , Testis/cytology , Testis/metabolism
10.
Stem Cell Reports ; 5(5): 716-727, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26607950

ABSTRACT

Self-renewal potential and multipotency are hallmarks of a stem cell. It is generally accepted that acquisition of such stemness requires rejuvenation of somatic cells through reprogramming of their genetic and epigenetic status.We show here that a simple block of cell differentiation is sufficient to induce and maintain stem cells. By overexpression of the transcriptional inhibitor ID3 in murine hematopoietic progenitor cells and cultivation under B cell induction conditions, the cells undergo developmental arrest and enter a self-renewal cycle. These cells can be maintained in vitro almost indefinitely, and the long-term cultured cells exhibit robust multi-lineage reconstitution when transferred into irradiated mice. These cells can be cloned and re-expanded with 50% plating efficiency, indicating that virtually all cells are self-renewing. Equivalent progenitors were produced from human cord blood stem cells, and these will ultimately be useful as a source of cells for immune cell therapy.


Subject(s)
Cell Cycle Checkpoints , Hematopoietic Stem Cells/cytology , Leukocytes/cytology , Animals , Cell Lineage , Cells, Cultured , Fetal Blood/cytology , Hematopoiesis , Hematopoietic Stem Cell Transplantation , Induced Pluripotent Stem Cells/cytology , Mice , Mice, Inbred C57BL
11.
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
12.
Sci Rep ; 5: 7978, 2015 Jan 23.
Article in English | MEDLINE | ID: mdl-25613394

ABSTRACT

The Rap G protein signal regulates Notch activation in early thymic progenitor cells, and deregulated Rap activation (Rap(high)) results in the development of Notch-dependent T-cell acute lymphoblastic leukemia (T-ALL). We demonstrate that the Rap signal is required for the proliferation and leukemogenesis of established Notch-dependent T-ALL cell lines. Attenuation of the Rap signal by the expression of a dominant-negative Rap1A17 or Rap1GAP, Sipa1, in a T-ALL cell line resulted in the reduced Notch processing at site 2 due to impaired maturation of Adam10. Inhibition of the Rap1 prenylation with a geranylgeranyl transferase inhibitor abrogated its membrane-anchoring to Golgi-network and caused reduced proprotein convertase activity required for Adam10 maturation. Exogenous expression of a mature form of Adam10 overcame the Sipa1-induced inhibition of T-ALL cell proliferation. T-ALL cell lines expressed Notch ligands in a Notch-signal dependent manner, which contributed to the cell-autonomous Notch activation. Although the initial thymic blast cells barely expressed Notch ligands during the T-ALL development from Rap(high) hematopoietic progenitors in vivo, the ligands were clearly expressed in the T-ALL cells invading extrathymic vital organs. These results reveal a crucial role of the Rap signal in the Notch-dependent T-ALL development and the progression.


Subject(s)
Cell Proliferation , GTPase-Activating Proteins/metabolism , Nuclear Proteins/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Receptors, Notch/metabolism , Signal Transduction , rap1 GTP-Binding Proteins/metabolism , Animals , Cell Line, Tumor , GTPase-Activating Proteins/genetics , Mice , Mice, Knockout , Nuclear Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptors, Notch/genetics , rap1 GTP-Binding Proteins/genetics
13.
Acta Histochem ; 116(5): 702-7, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24529545

ABSTRACT

Chicken D-serine dehydratase (DSD) degrades d-serine to pyruvate and ammonia. The enzyme requires both pyridoxal 5'-phosphate and Zn(2+) for its activity. d-Serine is a physiological coagonist that regulates the activity of the N-methyl-d-aspartate receptor (NMDAR) for l-glutamate. We have recently found in chickens that d-serine is degraded only by DSD in the brain, whereas it is also degraded to 3-hydroxypyruvate by d-amino acid oxidase (DAO) in the kidney and liver. In mammalian brains, d-serine is degraded only by DAO. It has not been clarified why chickens selectively use DSD for the control of d-serine concentrations in the brain. In the present study, we measured DSD activity in chicken tissues, and examined the cellular localization of DSD using a specific anti-chicken DSD antibody. The highest activity was found in kidney. Skeletal muscles and heart showed no activity. In chicken brain, cerebellum showed about 6-fold-higher activity (1.1 ± 0.3 U/g protein) than cerebrum (0.19 ± 0.03 U/g protein). At the cellular level DSD was demonstrated in proximal tubule cells of the kidney, in hepatocytes, in Bergmann-glia cells of the cerebellum and in astrocytes. The finding of DSD in glial cells seems to be important because d-serine is involved in NMDAR-dependent brain functions.


Subject(s)
Brain/enzymology , Immunohistochemistry , Kidney/enzymology , L-Serine Dehydratase/metabolism , Liver/enzymology , Animals , Blotting, Western , Chickens , Male
14.
J Immunol ; 189(9): 4426-36, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23018457

ABSTRACT

After receiving a TCR-mediated differentiation signal, CD4 and CD8 double-positive thymocytes diverge into CD4 or CD8 single-positive T cells, for which Th-POK and Runx3 have been identified as pivotal transcription factors, respectively. The cross-antagonistic regulation of Th-POK and Runx3 seems to be essential for CD4/8 thymocyte lineage commitment. However, the process for determining which pivotal factor acts dominantly has not been established. To explore the determining process, we used an in vitro culture system in which CD4 or CD8 single-positive cells are selectively induced from CD4/8 double-positive cells. Surprisingly, we found that control of G(1) cell cycle phase progression is critical for the determination. In the CD4 pathway, sustained TCR signal, as well as Th-POK, induces G(1)-phase extension and represses CD8 expression in a G(1) extension-dependent manner. In the CD8 pathway, after receiving a transient TCR signal, the IL-7R signal, as well as Runx3, antagonizes TCR signal-mediated G(1) extension and CD8 repression. Importantly, forced G(1) extension cancels the functions of Runx3 to repress Th-POK and CD4 and to reactivate CD8. In contrast, it is suggested that forced G(1) progression inhibits Th-POK function to repress CD8. Collectively, Th-POK and Runx3 are reciprocally involved in the control of G(1)-phase progression, on which they exert their functions dependently. These findings may provide novel insight into how CD4/CD8 cell lineages are determined by Th-POK and Runx3.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Lineage/immunology , Core Binding Factor Alpha 3 Subunit/physiology , G1 Phase/immunology , Transcription Factors/physiology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Organ Culture Techniques , Tumor Cells, Cultured
15.
PLoS One ; 7(8): e43340, 2012.
Article in English | MEDLINE | ID: mdl-22912857

ABSTRACT

While NF90 has been known to participate in transcription, translation and microRNA biogenesis, physiological functions of this protein still remain unclear. To uncover this, we generated transgenic (Tg) mice using NF90 cDNA under the control of ß-actin promoter. The NF90 Tg mice exhibited a reduction in body weight compared with wild-type mice, and a robust expression of NF90 was detected in skeletal muscle, heart and eye of the Tg mice. To evaluate the NF90 overexpression-induced physiological changes in the tissues, we performed a number of analyses including CT-analysis and hemodynamic test, revealing that the NF90 Tg mice developed skeletal muscular atrophy and heart failure. To explore causes of the abnormalities in the NF90 Tg mice, we performed histological and biochemical analyses for the skeletal and cardiac muscles of the Tg mice. Surprisingly, these analyses demonstrated that mitochondria in those muscular tissues of the Tg mice were degenerated by autophagy. To gain further insight into the cause for the mitochondrial degeneration, we identified NF90-associated factors by peptide mass fingerprinting. Of note, approximately half of the NF90-associated complexes were ribosome-related proteins. Interestingly, protein synthesis rate was significantly suppressed by high-expression of NF90. These observations suggest that NF90 would negatively regulate the function of ribosome via its interaction with the factors involved in the ribosome function. Furthermore, we found that the translations or protein stabilities of PGC-1 and NRF-1, which are critical transcription factors for expression of mitochondrial genes, were significantly depressed in the skeletal muscles of the NF90 Tg mice. Taken together, these findings suggest that the mitochondrial degeneration engaged in the skeletal muscle atrophy and the heart failure in the NF90 Tg mice may be caused by NF90-induced posttranscriptional repression of transcription factors such as PGC-1 and NRF-1 for regulating nuclear-encoded genes relevant to mitochondrial function.


Subject(s)
Autophagy/physiology , Mitophagy/physiology , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Myocardium/metabolism , Nuclear Factor 90 Proteins/metabolism , Actins/genetics , Animals , Blood Pressure , Blotting, Western , Body Weight/genetics , DNA, Complementary/genetics , Genotype , HEK293 Cells , Humans , Mice , Mice, Transgenic , Nuclear Factor 90 Proteins/genetics , Nuclear Respiratory Factor 1/metabolism , Promoter Regions, Genetic/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Ribosomal Proteins/metabolism , Tomography, X-Ray Computed , Transcription Factors/metabolism
16.
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
17.
Blood ; 119(13): 3123-7, 2012 Mar 29.
Article in English | MEDLINE | ID: mdl-22337716

ABSTRACT

Activation-induced cytidine deaminase (AID) is essential for class switch recombination and somatic hypermutation. Its deregulated expression acts as a genomic mutator that can contribute to the development of various malignancies. During treatment with imatinib mesylate (IM), patients with chronic myeloid leukemia often develop hypogammaglobulinemia, the mechanism of which has not yet been clarified. Here, we provide evidence that class switch recombination on B-cell activation is apparently inhibited by IM through down-regulation of AID. Furthermore, expression of E2A, a key transcription factor for AID induction, was markedly suppressed by IM. These results elucidate not only the underlying mechanism of IM-induced hypogammaglobulinemia but also its potential efficacy as an AID suppressor.


Subject(s)
Cytidine Deaminase/antagonists & inhibitors , Immunoglobulin Class Switching/drug effects , Piperazines/pharmacology , Pyrimidines/pharmacology , Animals , Benzamides , Cytidine Deaminase/metabolism , Down-Regulation/drug effects , Down-Regulation/immunology , Drug Evaluation, Preclinical , Imatinib Mesylate , Immunosuppressive Agents/pharmacology , Mice , Mice, Inbred C57BL , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Recombination, Genetic/drug effects , Recombination, Genetic/immunology , Sheep , Somatic Hypermutation, Immunoglobulin/drug effects , Treatment Outcome
18.
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
19.
Int Immunol ; 23(5): 297-305, 2011 May.
Article in English | MEDLINE | ID: mdl-21421735

ABSTRACT

V(D)J recombination of Ig and TCR genes is strictly regulated by the accessibility of target gene chromatin in a lineage- and stage-specific manner. In the mouse TCRγ locus, rearrangement of the Vγ2 gene predominates over Vγ3 rearrangement in the adult thymus. This preferential rearrangement is likely due to the differential accessibility of the individual Vγ genes, because the levels of germ line transcription and histone acetylation of the Vγ genes are well correlated with the rearrangement frequency in adult thymocytes. However, factors responsible for the differential regulation of the Vγ gene rearrangement have been largely unknown. In this study, we demonstrated that Vγ2 rearrangement in the adult thymus was substantially reduced in mice deficient for the basic helix-loop-helix protein, E2A. The decreased rearrangement is likely caused by the reduced accessibility of Vγ2 chromatin, since germ line transcription and histone acetylation of the Vγ2 gene were reduced in an E2A dosage-dependent manner. We further showed that E2A bound around the Vγ2 gene in vivo and we identified two canonical E-box sites downstream of Vγ2, to which E2A can bind in vitro. Furthermore, these two E-box sites had the ability to activate transcription upon E2A over-expression. These data suggest that E2A directly binds to and increases accessibility of Vγ2 chromatin, thereby facilitating Vγ2 rearrangement in the adult thymus.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Rearrangement, gamma-Chain T-Cell Antigen Receptor/genetics , Receptors, Antigen, T-Cell, gamma-delta/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Mice , Mice, Knockout
20.
Proc Natl Acad Sci U S A ; 108(10): 4075-80, 2011 Mar 08.
Article in English | MEDLINE | ID: mdl-21325057

ABSTRACT

Claudins (Clds) are crucial constituents of tight-junction strands in epithelial cells and have a central role in barrier functions. We show that Cld4 is unexpectedly expressed in normal thymic lymphocytes independently of tight junctions. The Cld4 expression was mostly confined to a portion of the CD4/CD8 double-positive (DP) cells. The proportion of Cld4(+) DP cells was markedly increased in MHC-I(-/-) II(-/-) mice but decreased in Rorγ(-/-) mice, and Cld4(+) DP cells contained higher levels of the rearranged Tcra transcripts involving the most distal Va and Ja segments than Cld4(-) DP cells. The Cld4 expression levels were reduced in E47-deficient mice in a gene dose-dependent manner, and ChIP analysis indicated that E2A and HEB were bound to the E-box sites of the putative Cldn4 promoter region. Functionally, Cld4 showed a potent T-cell receptor costimulatory activity by coligation with CD3. The Cld4 was distributed diffusely on the cell surface and associated with CD4/lck independently of CD3 in the resting thymocytes. However, Cld4 was strongly recruited to the immunological synapse on specific T-cell receptor engagement through antigen-presenting cells. In the fetal thymic organ culture, knockdown of Cldn4 resulted in the reduced generation of CD4/CD8 single-positive cells from the DP cells. These results suggest that Cld4 is induced by E-protein activity in the later stages of DP cells to increase the efficiency of positive selection, uncovering a hitherto unrecognized function of a Cld family protein.


Subject(s)
CD4 Antigens/immunology , CD8 Antigens/immunology , Membrane Proteins/biosynthesis , Thymus Gland/immunology , Animals , Cell Adhesion , Claudin-4 , Gene Knockdown Techniques , Immunological Synapses , Membrane Proteins/genetics , Mice , Thymus Gland/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...