Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
J Immunol ; 208(2): 303-320, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34930780

ABSTRACT

The deficiency of Aire, a transcriptional regulator whose defect results in the development of autoimmunity, is associated with reduced expression of tissue-restricted self-Ags (TRAs) in medullary thymic epithelial cells (mTECs). Although the mechanisms underlying Aire-dependent expression of TRAs need to be explored, the physical identification of the target(s) of Aire has been hampered by the low and promiscuous expression of TRAs. We have tackled this issue by engineering mice with augmented Aire expression. Integration of the transcriptomic data from Aire-augmented and Aire-deficient mTECs revealed that a large proportion of so-called Aire-dependent genes, including those of TRAs, may not be direct transcriptional targets downstream of Aire. Rather, Aire induces TRA expression indirectly through controlling the heterogeneity of mTECs, as revealed by single-cell analyses. In contrast, Ccl25 emerged as a canonical target of Aire, and we verified this both in vitro and in vivo. Our approach has illuminated the Aire's primary targets while distinguishing them from the secondary targets.


Subject(s)
Autoantigens/immunology , Autoimmunity/immunology , Chemokines, CC/metabolism , Thymus Gland/immunology , Transcription Factors/metabolism , Animals , Autoimmunity/genetics , Chemokines, CC/genetics , Epithelial Cells/immunology , Gene Expression Regulation , Gene Knock-In Techniques , Gene Knockout Techniques , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Thymus Gland/cytology , Transcription Factors/genetics , Transcription, Genetic/genetics , AIRE Protein
2.
Adv Exp Med Biol ; 1444: 197-205, 2024.
Article in English | MEDLINE | ID: mdl-38467981

ABSTRACT

Programmed cell death-1 (PD-1) is one of the most famous coinhibitory receptors that are expressed on effector T cells to regulate their function. The PD-1 ligands, PD-L1 and PD-L2, are expressed by various cells throughout the body at steady state and their expression was further regulated within different pathological conditions such as tumor-bearing and chronic inflammatory diseases. In recent years, immune checkpoint inhibitor (ICI) therapies with anti-PD-1 or anti-PD-L1 has become a standard treatment for various malignancies and has shown remarkable antitumor effects. Since the discovery of PD-1 in 1992, a huge number of studies have been conducted to elucidate the function of PD-1. Herein, this paper provides an overview of PD-1 biological findings and sheds some light on the current technology for molecular imaging of PD-1.


Subject(s)
Neoplasms , Programmed Cell Death 1 Receptor , Humans , Programmed Cell Death 1 Receptor/metabolism , Neoplasms/metabolism , T-Lymphocytes/metabolism , B7-H1 Antigen/metabolism , Immunotherapy/methods , Molecular Imaging
3.
Immunol Cell Biol ; 100(5): 371-377, 2022 05.
Article in English | MEDLINE | ID: mdl-35313042

ABSTRACT

Deficiency for AIRE/Aire in both humans and mice results in the development of organ-specific autoimmune disease. We tested whether augmented and/or dysregulated AIRE/Aire expression might be also prone to the breakdown of self-tolerance. To define the effect of augmented Aire expression on the development of autoimmunity, antigen-specific clonal deletion and production of clonotypic regulatory T cells (Tregs) in the thymus were examined using mice expressing two additional copies of Aire in a heterozygous state (3xAire-knockin mice: 3xAire-KI). We found that both clonal deletion of autoreactive T cells and production of clonotypic Tregs in the thymus from 3xAire-KI were impaired in a T-cell receptor-transgenic system. Furthermore, 3xAire-KI females showed higher scores of experimental autoimmune encephalomyelitis induced by myelin oligodendrocyte glycoprotein than wild-type littermates, suggesting that augmented Aire expression exacerbates organ-specific autoimmunity under disease-prone conditions. In humans, we found that one patient with amyopathic dermatomyositis showed CD3- CD19- cells expressing AIRE in the peripheral blood before the treatment but not during the remission phase treated with immunosuppressive drugs. Thus, not only loss of function of AIRE/Aire but also augmented and/or dysregulated expression of AIRE/Aire should be considered for the pathogenesis of organ-specific autoimmunity. We suggest that further analyses should be pursued to establish a novel link between organ-specific autoimmune disease and dysregulated AIRE expression in clinical settings.


Subject(s)
Autoimmunity , Encephalomyelitis, Autoimmune, Experimental , Animals , Clonal Deletion , Female , Humans , Immune Tolerance , Mice , Myelin-Oligodendrocyte Glycoprotein , Thymus Gland
4.
Int Immunol ; 32(2): 117-131, 2020 02 07.
Article in English | MEDLINE | ID: mdl-31586207

ABSTRACT

Tissue-specific autoimmune diseases are assumed to arise through malfunction of two checkpoints for immune tolerance: defective elimination of autoreactive T cells in the thymus and activation of these T cells by corresponding autoantigens in the periphery. However, evidence for this model and the outcome of such alterations in each or both of the tolerance mechanisms have not been sufficiently investigated. We studied these issues by expressing human AIRE (huAIRE) as a modifier of tolerance function in NOD mice wherein the defects of thymic and peripheral tolerance together cause type I diabetes (T1D). Additive huAIRE expression in the thymic stroma had no major impact on the production of diabetogenic T cells in the thymus. In contrast, huAIRE expression in peripheral antigen-presenting cells (APCs) rendered the mice resistant to T1D, while maintaining other tissue-specific autoimmune responses and antibody production against an exogenous protein antigen, because of the loss of Xcr1+ dendritic cells, an essential component for activating diabetogenic T cells in the periphery. These results contrast with our recent demonstration that huAIRE expression in both the thymic stroma and peripheral APCs resulted in the paradoxical development of muscle-specific autoimmunity. Our results reveal that tissue-specific autoimmunity is differentially controlled by a combination of thymic function and peripheral tolerance, which can be manipulated by expression of huAIRE/Aire in each or both of the tolerance mechanisms.


Subject(s)
Autoimmunity/immunology , Peripheral Tolerance/immunology , Thymus Gland/immunology , Transcription Factors/immunology , Animals , Autoantigens/immunology , Diabetes Mellitus, Type 1/immunology , Disease Models, Animal , Humans , Islets of Langerhans/immunology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , T-Lymphocytes/immunology , Transcription Factors/genetics , AIRE Protein
5.
J Immunol ; 201(11): 3244-3257, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30389776

ABSTRACT

Medullary thymic epithelial cells (mTECs), which express a wide range of tissue-restricted Ags (TRAs), contribute to the establishment of self-tolerance by eliminating autoreactive T cells and/or inducing regulatory T cells. Aire controls a diverse set of TRAs within Aire-expressing cells by employing various transcriptional pathways. As Aire has a profound effect on transcriptomes of mTECs, including TRAs not only at the single-cell but also the population level, we suspected that Aire (Aire+ mTECs) might control the cellular composition of the thymic microenvironment. In this study, we confirmed that this is indeed the case by identifying a novel mTEC subset expressing Ly-6 family protein whose production was defective in Aire-deficient thymi. Reaggregated thymic organ culture experiments demonstrated that Aire did not induce the expression of Ly-6C/Ly-6G molecules from mTECs as Aire-dependent TRAs in a cell-intrinsic manner. Instead, Aire+ mTECs functioned in trans to maintain Ly-6C/Ly-6G+ mTECs. Thus, Aire not only controls TRA expression transcriptionally within the cell but also controls the overall composition of mTECs in a cell-extrinsic manner, thereby regulating the transcriptome from mTECs on a global scale.


Subject(s)
Epithelial Cells/pathology , Thymus Gland/physiology , Transcription Factors/metabolism , Animals , Antigens, Ly/metabolism , Cells, Cultured , Cellular Microenvironment , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Organ Culture Techniques , Transcription Factors/genetics , Transcriptional Activation , AIRE Protein
6.
J Autoimmun ; 86: 75-92, 2018 01.
Article in English | MEDLINE | ID: mdl-28931462

ABSTRACT

Autoimmunity is prevented by the function of the autoimmune regulator [AIRE (Aire in mice)], which promotes the expression of a wide variety of tissue-restricted antigens (TRAs) from medullary thymic epithelial cells (mTECs) and from a subset of peripheral antigen-presenting cells (APCs). We examined the effect of additive expression of human AIRE (huAIRE) in a model of autoimmune diabetes in NOD mice. Unexpectedly, we observed that mice expressing augmented AIRE/Aire developed muscle-specific autoimmunity associated with incomplete maturation of mTECs together with impaired expression of Aire-dependent TRAs. This led to failure of deletion of autoreactive T cells together with dramatically reduced production of regulatory T cells in the thymus. In peripheral APCs, expression of costimulatory molecules was augmented. We suggest that levels of Aire expression need to be tightly controlled for maintenance of immunological tolerance. Our results also highlight the importance of coordinated action between central tolerance and peripheral tolerance under the common control of Aire.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Muscles/immunology , Polymyositis/immunology , Thymus Gland/immunology , Transcription Factors/metabolism , Animals , Autoantigens/metabolism , Autoimmunity , Disease Models, Animal , Humans , Immune Tolerance , Mice , Mice, Inbred NOD , Mice, Transgenic , Organ Specificity , Transcription Factors/genetics , AIRE Protein
7.
J Immunol ; 195(11): 5149-58, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26503950

ABSTRACT

Aire in medullary thymic epithelial cells (mTECs) plays an important role in the establishment of self-tolerance. Because Aire(+) mTECs appear to be a limited subset, they may constitute a unique lineage(s) among mTECs. An alternative possibility is that all mTECs are committed to express Aire in principle, but Aire expression by individual mTECs is conditional. To investigate this issue, we established a novel Aire reporter strain in which endogenous Aire is replaced by the human AIRE-GFP-Flag tag (Aire/hAGF-knockin) fusion gene. The hAGF reporter protein was produced and retained very efficiently within mTECs as authentic Aire nuclear dot protein. Remarkably, snapshot analysis revealed that mTECs expressing hAGF accounted for >95% of mature mTECs, suggesting that Aire expression does not represent a particular mTEC lineage(s). We confirmed this by generating Aire/diphtheria toxin receptor-knockin mice in which long-term ablation of Aire(+) mTECs by diphtheria toxin treatment resulted in the loss of most mature mTECs beyond the proportion of those apparently expressing Aire. These results suggest that Aire expression is inherent to all mTECs but may occur at particular stage(s) and/or cellular states during their differentiation, thus accounting for the broad impact of Aire on the promiscuous gene expression of mTECs.


Subject(s)
Epithelial Cells/metabolism , Thymus Gland/metabolism , Transcription Factors/biosynthesis , Animals , Cell Differentiation , Diphtheria Toxin/pharmacology , Epithelial Cells/cytology , Gene Expression , Gene Expression Regulation , Gene Knock-In Techniques , Green Fluorescent Proteins/genetics , Heparin-binding EGF-like Growth Factor/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Thymus Gland/cytology , Transcription Factors/genetics , AIRE Protein
8.
J Immunol ; 195(10): 4641-9, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26453754

ABSTRACT

Cortical thymic epithelial cells (cTECs) and medullary thymic epithelial cells (mTECs) play essential roles in the positive and negative selection of developing thymocytes, respectively. Aire in mTECs plays an essential role in the latter process through expression of broad arrays of tissue-restricted Ags. To determine whether the location of Aire within the medulla is absolutely essential or whether Aire could also function within the cortex for establishment of self-tolerance, we used bacterial artificial chromosome technology to establish a semiknockin strain of NOD-background (ß5t/Aire-transgenic) mice expressing Aire under control of the promoter of ß5t, a thymoproteasome expressed exclusively in the cortex. Although Aire was expressed in cTECs as typical nuclear dot protein in ß5t/Aire-Tg mice, cTECs expressing Aire ectopically did not confer transcriptional expression of either Aire-dependent or Aire-independent tissue-restricted Ag genes. We then crossed ß5t/Aire-Tg mice with Aire-deficient NOD mice, generating a strain in which Aire expression was confined to cTECs. Despite the presence of Aire(+) cTECs, these mice succumbed to autoimmunity, as did Aire-deficient NOD mice. The thymic microenvironment harboring Aire(+) cTECs, within which many Aire-activated genes were present, also showed no obvious alteration of positive selection, suggesting that Aire's unique property of generating a self-tolerant T cell repertoire is functional only in mTECs.


Subject(s)
Autoimmunity/genetics , Self Tolerance/genetics , Thymocytes/immunology , Thymus Gland/immunology , Transcription Factors/genetics , Animals , Autoimmunity/immunology , Cell Differentiation/immunology , Chromosomes, Artificial, Bacterial/genetics , Epithelial Cells/cytology , Epithelial Cells/immunology , Gene Knock-In Techniques , Mice , Mice, Inbred NOD , Mice, Knockout , Self Tolerance/immunology , T-Lymphocytes/immunology , Thymocytes/cytology , Thymus Gland/cytology , Transcription Factors/metabolism , AIRE Protein
9.
J Immunol ; 193(9): 4356-67, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25261487

ABSTRACT

Essential roles of NF-κB-inducing kinase (NIK) for the development of medullary thymic epithelial cells (mTECs) and regulatory T cells have been highlighted by studies using a strain of mouse bearing a natural mutation of the NIK gene (aly mice). However, the exact mechanisms underlying the defect in thymic cross-talk leading to the breakdown of self-tolerance in aly mice remain elusive. In this study, we demonstrated that production of regulatory T cells and the final maturation process of positively selected conventional αß T cells are impaired in aly mice, partly because of a lack of mature mTECs. Of note, numbers of thymic dendritic cells and their expression of costimulatory molecules were also affected in aly mice in a thymic stroma-dependent manner. The results suggest a pivotal role of NIK in the thymic stroma in establishing self-tolerance by orchestrating cross-talk between mTECs and dendritic cells as well as thymocytes. In addition, we showed that negative selection was impaired in aly mice as a result of the stromal defect, which accounts for the development of organ-specific autoimmunity through a lack of normal NIK.


Subject(s)
Cell Communication/immunology , Dendritic Cells/immunology , Protein Serine-Threonine Kinases/metabolism , Self Tolerance/immunology , Thymocytes/immunology , Animals , B7-1 Antigen/metabolism , Cell Differentiation , Dendritic Cells/metabolism , Epithelial Cells/metabolism , Gene Expression , Immunophenotyping , Male , Mice , Mice, Transgenic , Models, Immunological , Mutation , Phenotype , Protein Serine-Threonine Kinases/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Stromal Cells/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Thymocytes/metabolism , Thymus Gland/immunology , Thymus Gland/metabolism , NF-kappaB-Inducing Kinase
10.
J Immunol ; 192(6): 2585-92, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24516201

ABSTRACT

Understanding the cellular dynamics of Aire-expressing lineage(s) among medullary thymic epithelial cells (AEL-mTECs) is essential for gaining insight into the roles of Aire in establishment of self-tolerance. In this study, we monitored the maturation program of AEL-mTECs by temporal lineage tracing, in which bacterial artificial chromosome transgenic mice expressing tamoxifen-inducible Cre recombinase under control of the Aire regulatory element were crossed with reporter strains. We estimated that the half-life of AEL-mTECs subsequent to Aire expression was ∼7-8 d, which was much longer than that reported previously, owing to the existence of a post-Aire stage. We found that loss of Aire did not alter the overall lifespan of AEL-mTECs, inconsistent with the previous notion that Aire expression in medullary thymic epithelial cells (mTECs) might result in their apoptosis for efficient cross-presentation of self-antigens expressed by AEL-mTECs. In contrast, Aire was required for the full maturation program of AEL-mTECs, as exemplified by the lack of physiological downregulation of CD80 during the post-Aire stage in Aire-deficient mice, thus accounting for the abnormally increased CD80(high) mTECs seen in such mice. Of interest, increased CD80(high) mTECs in Aire-deficient mice were not mTEC autonomous and were dependent on cross-talk with thymocytes. These results further support the roles of Aire in the differentiation program of AEL-mTECs.


Subject(s)
Cell Differentiation/immunology , Cell Lineage/immunology , Epithelial Cells/immunology , Transcription Factors/immunology , Animals , Apoptosis/genetics , Apoptosis/immunology , Autoantigens/immunology , Autoantigens/metabolism , B7-1 Antigen/immunology , B7-1 Antigen/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Cells, Cultured , Cross-Priming/genetics , Cross-Priming/immunology , Epithelial Cells/metabolism , Flow Cytometry , Immunohistochemistry , Kinetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Thymocytes/cytology , Thymocytes/immunology , Thymocytes/metabolism , Thymus Gland/cytology , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism , AIRE Protein
11.
J Cell Sci ; 125(Pt 1): 166-75, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22250197

ABSTRACT

The nucleus in eukaryotic cells is a highly organized and dynamic structure containing numerous subnuclear bodies. The morphological appearance of nuclear bodies seems to be a reflection of ongoing functions, such as DNA replication, transcription, repair, RNA processing and RNA transport. The integrator complex mediates processing of small nuclear RNA (snRNA), so it might play a role in nuclear body formation. Here, we show that the integrator complex is essential for integrity of the Cajal body. Depletion of INTS4, an integrator complex subunit, abrogated 3'-end processing of snRNA. A defect in this activity caused a significant accumulation of the Cajal body marker protein coilin in nucleoli. Some fractions of coilin still formed nucleoplasmic foci; however, they were free of other Cajal body components, such as survival of motor neuron protein (SMN), Sm proteins and snRNAs. SMN and Sm proteins formed striking cytoplasmic granules. These findings demonstrate that the integrator complex is essential for snRNA maturation and Cajal body homeostasis.


Subject(s)
Coiled Bodies/metabolism , Nuclear Proteins/metabolism , Active Transport, Cell Nucleus , Cell Nucleolus/metabolism , Cytoplasmic Granules/metabolism , Gene Knockdown Techniques , HeLa Cells , Humans , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Phenotype , Protein Transport , RNA, Small Nuclear/metabolism , Ribonucleases/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , SMN Complex Proteins/metabolism
12.
Nat Commun ; 14(1): 3157, 2023 06 06.
Article in English | MEDLINE | ID: mdl-37280233

ABSTRACT

With recent advances in immune checkpoint inhibitors (ICIs), immunotherapy has become the standard treatment for various malignant tumors. Their indications and dosages have been determined empirically, taking individually conducted clinical trials into consideration, but without a standard method to evaluate them. Here we establish an advanced imaging system to visualize human PD-1 microclusters, in which a minimal T cell receptor (TCR) signaling unit co-localizes with the inhibitory co-receptor PD-1 in vitro. In these microclusters PD-1 dephosphorylates both the TCR/CD3 complex and its downstream signaling molecules via the recruitment of a phosphatase, SHP2, upon stimulation with the ligand hPD-L1. In this system, blocking antibodies for hPD-1-hPD-L1 binding inhibits hPD-1 microcluster formation, and each therapeutic antibody (pembrolizumab, nivolumab, durvalumab and atezolizumab) is characterized by a proprietary optimal concentration and combinatorial efficiency enhancement. We propose that our imaging system could digitally evaluate PD-1-mediated T cell suppression to evaluate their clinical usefulness and to develop the most suitable combinations among ICIs or between ICIs and conventional cancer treatments.


Subject(s)
Neoplasms , Programmed Cell Death 1 Receptor , Humans , Single Molecule Imaging , Nivolumab/pharmacology , Nivolumab/therapeutic use , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Receptors, Antigen, T-Cell , B7-H1 Antigen/metabolism , Immunotherapy/methods
13.
Genes Cells ; 14(8): 975-90, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19695025

ABSTRACT

The interphase nucleus is a highly ordered and compartmentalized organelle. Little is known regarding what elaborate mechanisms might exist to explain these properties of the nucleus. Also unresolved is whether some architectural components might facilitate the formation of functional intranuclear compartments or higher order chromatin structure. As the first step to address these questions, we performed an in-depth proteome analysis of nuclear insoluble fractions of human HeLa-S3 cells prepared by two different approaches: a high-salt/detergent/nuclease-resistant fraction and a lithium 3,5-diiodosalicylate/nuclease-resistant fraction. Proteins of the fractions were analyzed by liquid chromatography electrospray ionization tandem mass spectrometry, identifying 333 and 330 proteins from each fraction respectively. Among the insoluble nuclear proteins, we identified 37 hitherto unknown or functionally uncharacterized proteins. The RNA recognition motif, WD40 repeats, HEAT repeats and the SAP domain were often found in these identified proteins. The subcellular distribution of selected proteins, including DEK protein and SON protein, demonstrated their novel associations with nuclear insoluble materials, corroborating our MS-based analysis. This study establishes a comprehensive catalog of the nuclear insoluble proteins in human cells. Further functional analysis of the proteins identified in our study will significantly improve our understanding of the dynamic organization of the interphase nucleus.


Subject(s)
Cell Nucleus/metabolism , Interphase , Nuclear Proteins , Proteome , Cell Nucleus/chemistry , Chromatography, Liquid , Fluorescent Antibody Technique , HeLa Cells , Humans , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Solubility , Spectrometry, Mass, Electrospray Ionization , Subcellular Fractions/chemistry , Subcellular Fractions/metabolism , Tandem Mass Spectrometry
14.
Genes Cells ; 13(6): 571-82, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18422602

ABSTRACT

Snf2SR, a suppressor of rna1(ts), which is a temperature-sensitive mutation in Schizosaccharomyces pombe RanGAP (GTPase activating protein), possesses both the SNF2 and the helicase domains conserved in the chromatin remodeling SNF2 ATPase/helicase protein family. We have now clarified a function of Snf2SR. Snf2SR indeed showed DNA-stimulated ATPase activity, proving that it is a member of the SNF2 ATPase/helicase family. Consistent with this role, Snf2SR was localized in the nucleus and cell fractionation analysis revealed that Snf2SR was tightly associated with the nuclear matrix. The disruption of snf2SR(+) was detrimental for a cell proliferation of S. pombe. Snf2SR that did not enhance RanGAP activity by itself, but abolished histone-H3-mediated RanGAP inhibition, as previously reported for the histone H3 methyltransferase, Clr4, another rna1(ts) suppressor. In contrast to Clr4, Snf2SR directly bound to the GDP-bound form of the S. pombe Ran homologue Spi1 and enhanced the nucleotide exchange activity of Pim1, the S. pombe RanGEF (guanine nucleotide exchange factor). Over-expression of Spi1-G18V, a Ran GTPase mutant fixed in the GTP-bound form, was lethal to S. pombe Deltasnf2SR. Together, our results indicate that Snf2SR is involved in the Ran GTPase cycle in vivo.


Subject(s)
Adenosine Triphosphatases/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/metabolism , ran GTP-Binding Protein/metabolism , Adenosine Triphosphatases/analysis , Adenosine Triphosphatases/genetics , Cell Nucleus/chemistry , Chromosomal Proteins, Non-Histone/metabolism , Gene Deletion , Histones/metabolism , Schizosaccharomyces/cytology , Schizosaccharomyces pombe Proteins/analysis , Schizosaccharomyces pombe Proteins/genetics
15.
Mol Biol Cell ; 17(6): 2524-36, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16540522

ABSTRACT

Although the Ran GTPase-activating protein RanGAP mainly functions in the cytoplasm, several lines of evidence indicate a nuclear function of RanGAP. We found that Schizosaccharomyces pombe RanGAP, SpRna1, bound the core of histone H3 (H3) and enhanced Clr4-mediated H3-lysine 9 (K9) methylation. This enhancement was not observed for methylation of the H3-tail containing K9 and was independent of SpRna1-RanGAP activity, suggesting that SpRna1 itself enhances Clr4-mediated H3-K9 methylation via H3. Although most SpRna1 is in the cytoplasm, some cofractionated with H3. Sprna1(ts) mutations caused decreases in Swi6 localization and H3-K9 methylation at all three heterochromatic regions of S. pombe. Thus, nuclear SpRna1 seems to be involved in heterochromatin assembly. All core histones bound SpRna1 and inhibited SpRna1-RanGAP activity. In contrast, Clr4 abolished the inhibitory effect of H3 on the RanGAP activity of SpRna1 but partially affected the other histones. SpRna1 formed a trimeric complex with H3 and Clr4, suggesting that nuclear SpRna1 is reciprocally regulated by histones, especially H3, and Clr4 on the chromatin to function for higher order chromatin assembly. We also found that SpRna1 formed a stable complex with Xpo1/Crm1 plus Ran-GTP, in the presence of H3.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , GTPase-Activating Proteins/metabolism , Heterochromatin/metabolism , Heterochromatin/physiology , Histones/metabolism , Methyltransferases/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/metabolism , Base Sequence , Cell Cycle Proteins/genetics , DNA Primers , Genetic Markers , Histone-Lysine N-Methyltransferase , Kinetics , Methyltransferases/genetics , Plasmids , Polymerase Chain Reaction , Recombinant Fusion Proteins/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/genetics
16.
Biochem Biophys Res Commun ; 373(4): 624-30, 2008 Sep 05.
Article in English | MEDLINE | ID: mdl-18601901

ABSTRACT

Amounts of soluble histones in cells are tightly regulated to ensure supplying them for the newly synthesized DNA and preventing the toxic effect of excess histones. Prior to incorporation into chromatin, newly synthesized histones H3 and H4 are highly acetylated in pre-deposition complex, wherein H4 is di-acetylated at Lys-5 and Lys-12 residues by histone acetyltransferase-1 (Hat1), but their role in histone metabolism is still unclear. Here, using chicken DT 40 cytosolic extracts, we found that histones H3/H4 and their chaperone Asf1, including RbAp48, a regulatory subunit of Hat1 enzyme, were associated with Hat1. Interestingly, in HAT1-deficient cells, cytosolic histones H3/H4 fractions on sucrose gradient centrifugation, having a sedimentation coefficient of 5-6S in DT40 cells, were shifted to lower molecular mass fractions, with Asf1. Further, sucrose gradient fractionation of semi-purified tagged Asf1-complexes showed the presence of Hat1, RbAp48 and histones H3/H4 at 5-6S fractions in the complexes. These findings suggest the possible involvement of Hat1 in regulating cytosolic H3/H4 pool mediated by Asf1-containing cytosolic H3/H4 pre-deposition complex.


Subject(s)
Acetyltransferases/metabolism , Cytosol/metabolism , Histones/metabolism , Acetylation , Acetyltransferases/genetics , Animals , Catalysis , Chickens/metabolism , Histone Acetyltransferases , Molecular Chaperones
17.
Mol Biol Cell ; 15(11): 4960-70, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15317843

ABSTRACT

We isolated 11 independent temperature-sensitive (ts) mutants of Schizosaccharomyces pombe RanGAP, SpRna1 that have several amino acid changes in the conserved domains of RanGAP. Resulting Sprna1ts showed a strong defect in mitotic chromosome segregation, but did not in nucleocytoplasmic transport and microtubule formation. In addition to Sprna1+ and Spksp1+, the clr4+ (histone H3-K9 methyltransferase), the S. pombe gene, SPAC25A8.01c, designated snf2SR+ (a member of the chromatin remodeling factors, Snf2 family with DNA-dependent ATPase activity), but not the spi1+ (S. pombe Ran homolog), rescued a lethality of Sprna1ts. Both Clr4 and Snf2 were reported to be involved in heterochromatin formation essential for building the centromeres. Consistently, Sprna1ts was defective in gene-silencing at the centromeres. But a silencing at the telomere, another heterochromatic region, was normal in all of Sprna1ts strains, indicating SpRna1 in general did not function for a heterochromatin formation. snf2SR+ rescued a centromeric silencing defect and Deltaclr4+ was synthetic lethal with Sprna1ts. Taken together, SpRna1 was suggested to function for constructing the centromeres, by cooperating with Clr4 and Snf2SR. Loss of SpRna1 activity, therefore, caused chromosome missegregation.


Subject(s)
Centromere/ultrastructure , GTPase-Activating Proteins/biosynthesis , GTPase-Activating Proteins/physiology , Gene Silencing , Schizosaccharomyces/metabolism , Active Transport, Cell Nucleus , Cell Nucleus/metabolism , Chromosome Segregation , Cytoplasm/metabolism , DNA Mutational Analysis , Green Fluorescent Proteins/metabolism , Heterochromatin/metabolism , Histones , Microscopy, Fluorescence , Mitosis , Models, Biological , Models, Molecular , Mutation , Protein Structure, Tertiary , Schizosaccharomyces pombe Proteins , Temperature
18.
Front Genet ; 8: 17, 2017.
Article in English | MEDLINE | ID: mdl-28270832

ABSTRACT

Nuclear actin family proteins, comprising of actin and actin-related proteins (Arps), are essential functional components of the multiple chromatin remodeling complexes. The INO80 chromatin remodeling complex, which is evolutionarily conserved and has roles in transcription, DNA replication and repair, consists of actin and actin-related proteins Arp4, Arp5, and Arp8. We generated Arp5 knockout (KO) and Arp8 KO cells from the human Nalm-6 pre-B cell line and used these KO cells to examine the roles of Arp5 and Arp8 in the transcriptional regulation mediated by the INO80 complex. In both of Arp5 KO and Arp8 KO cells, the oxidative stress-induced expression of HMOX1 gene, encoding for heme oxygenase-1 (HO-1), was significantly impaired. Consistent with these observations, chromatin immunoprecipitation (ChIP) assay revealed that oxidative stress caused an increase in the binding of the INO80 complex to the regulatory sites of HMOX1 in wild-type cells. The binding of INO80 complex to chromatin was reduced in Arp8 KO cells compared to that in the wild-type cells. On the other hand, the binding of INO80 complex to chromatin in Arp5 KO cells was similar to that in the wild-type cells even under the oxidative stress condition. However, both remodeling of chromatin at the HMOX1 regulatory sites and binding of a transcriptional activator to these sites were impaired in Arp5 KO cells, indicating that Arp5 is required for the activation of the INO80 complex. Collectively, these results suggested that these nuclear Arps play indispensable roles in the function of the INO80 chromatin remodeling complex.

19.
PLoS One ; 9(10): e108354, 2014.
Article in English | MEDLINE | ID: mdl-25299602

ABSTRACT

Actin and actin-related proteins (Arps), which are members of the actin family, are essential components of many of these remodeling complexes. Actin, Arp4, Arp5, and Arp8 are found to be evolutionarily conserved components of the INO80 chromatin remodeling complex, which is involved in transcriptional regulation, DNA replication, and DNA repair. A recent report showed that Arp8 forms a module in the INO80 complex and this module can directly capture a nucleosome. In the present study, we showed that recombinant human Arp8 binds to DNAs, and preferentially binds to single-stranded DNA. Analysis of the binding of adenine nucleotides to Arp8 mutants suggested that the ATP-binding pocket, located in the evolutionarily conserved actin fold, plays a regulatory role in the binding of Arp8 to DNA. To determine the cellular function of Arp8, we derived tetracycline-inducible Arp8 knockout cells from a cultured human cell line. Analysis of results obtained after treating these cells with aphidicolin and camptothecin revealed that Arp8 is involved in DNA repair. Together with the previous observation that Arp8, but not γ-H2AX, is indispensable for recruiting INO80 complex to DSB in human, results of our study suggest an individual role for Arp8 in DNA repair.


Subject(s)
DNA Repair/physiology , DNA-Binding Proteins/metabolism , Microfilament Proteins/metabolism , Actins/metabolism , Cell Line, Tumor , Chromatin Assembly and Disassembly/physiology , DNA Replication/physiology , DNA, Single-Stranded/genetics , Humans
20.
Front Immunol ; 4: 210, 2013.
Article in English | MEDLINE | ID: mdl-23885257

ABSTRACT

The discovery of Aire-dependent transcriptional control of many tissue-restricted self-antigen (TRA) genes in thymic epithelial cells in the medulla (medullary thymic epithelial cells, mTECs) has raised the intriguing question of how the single Aire gene can influence the transcription of such a large number of TRA genes within mTECs. From a mechanistic viewpoint, there are two possible models to explain the function of Aire in this action. In the first model, TRAs are considered to be the direct target genes of Aire's transcriptional activity. In this scenario, the lack of Aire protein within cells would result in the defective TRA gene expression, while the maturation program of mTECs would be unaffected in principle. The second model hypothesizes that Aire is necessary for the maturation program of mTECs. In this case, we assume that the mTEC compartment does not mature normally in the absence of Aire. If acquisition of the properties of TRA gene expression depends on the maturation status of mTECs, a defect of such an Aire-dependent maturation program in Aire-deficient mTECs can also result in impaired TRA gene expression. In this brief review, we will focus on these two contrasting models for the roles of Aire in controlling the expression of TRAs within mTECs.

SELECTION OF CITATIONS
SEARCH DETAIL