Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Genes Cells ; 28(12): 929-941, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37909727

ABSTRACT

One hallmark of some autoimmune diseases is the variability of symptoms among individuals. Organs affected by the disease differ between patients, posing a challenge in diagnosing the affected organs. Although numerous studies have investigated the correlation between T cell antigen receptor (TCR) repertoires and the development of infectious and immune diseases, the correlation between TCR repertoires and variations in disease symptoms among individuals remains unclear. This study aimed to investigate the correlation of TCRα and ß repertoires in blood T cells with the extent of autoimmune signs that varies among individuals. We sequenced TCRα and ß of CD4+ CD44high CD62Llow T cells in the blood and stomachs of mice deficient in autoimmune regulator (Aire) (AIRE KO), a mouse model of human autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy. Data analysis revealed that the degree of similarity in TCR sequences between the blood and stomach varied among individual AIRE KO mice and reflected the extent of T cell infiltration in the stomach. We identified a set of TCR sequences whose frequencies in blood might correlate with extent of the stomach manifestations. Our results propose a potential of using TCR repertoires not only for diagnosing disease development but also for diagnosing affected organs in autoimmune diseases.


Subject(s)
Autoimmune Diseases , Polyendocrinopathies, Autoimmune , Humans , Mice , Animals , CD4-Positive T-Lymphocytes , Receptors, Antigen, T-Cell/genetics
2.
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
3.
Biochem Biophys Res Commun ; 501(3): 745-750, 2018 06 27.
Article in English | MEDLINE | ID: mdl-29753741

ABSTRACT

Hindlimb unloading (HU) of rodents has been used as a ground-based model of spaceflight. In this study, we investigated the detailed impact of 14-day HU on the murine thymus. Thymic mass and cell number were significantly reduced after 14 days of hindlimb unloading, which was accompanied by an increment of plasma corticosterone. Although corticosterone reportedly causes selective apoptosis of CD4+CD8+ thymocytes (CD4+CD8+DPs) in mice treated with short-term HU, the reduction of thymocyte cellularity after the 14-day HU was not selective for CD4+CD8+DPs. In addition to the thymocyte reduction, the cellularity of thymic epithelial cells (TECs) was also reduced by the 14-day HU. Flow cytometric and RNA-sequencing analysis suggested that medullary TECs (mTECs) were preferentially reduced after HU. Moreover, immunohistochemical staining suggested that the 14-day HU caused a reduction of the mTECs expressing autoimmune regulator (Aire). Our data suggested that HU impacts both thymocytes and TECs. Consequently, these data imply that thymic T cell repertoire formation could be disturbed during spaceflight-like stress.


Subject(s)
Epithelial Cells/cytology , Hindlimb Suspension/methods , Thymocytes/cytology , Thymus Gland/physiology , Transcription Factors/analysis , Animals , CD4 Antigens/analysis , CD8 Antigens/analysis , Cell Count , Male , Mice, Inbred C57BL , Organ Size , Thymus Gland/cytology , Time Factors , AIRE Protein
4.
Nat Commun ; 15(1): 953, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38296961

ABSTRACT

Autophagy is primarily activated by cellular stress, such as starvation or mitochondrial damage. However, stress-independent autophagy is activated by unclear mechanisms in several cell types, such as thymic epithelial cells (TECs). Here we report that the mitochondrial protein, C15ORF48, is a critical inducer of stress-independent autophagy. Mechanistically, C15ORF48 reduces the mitochondrial membrane potential and lowers intracellular ATP levels, thereby activating AMP-activated protein kinase and its downstream Unc-51-like kinase 1. Interestingly, C15ORF48-dependent induction of autophagy upregulates intracellular glutathione levels, promoting cell survival by reducing oxidative stress. Mice deficient in C15orf48 show a reduction in stress-independent autophagy in TECs, but not in typical starvation-induced autophagy in skeletal muscles. Moreover, C15orf48-/- mice develop autoimmunity, which is consistent with the fact that the stress-independent autophagy in TECs is crucial for the thymic self-tolerance. These results suggest that C15ORF48 induces stress-independent autophagy, thereby regulating oxidative stress and self-tolerance.


Subject(s)
Autoimmunity , Mitochondrial Proteins , Mice , Animals , Mitochondrial Proteins/metabolism , Oxidative Stress , Autophagy , Epithelial Cells/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism
5.
Bio Protoc ; 13(1): e4588, 2023 Jan 05.
Article in English | MEDLINE | ID: mdl-36789086

ABSTRACT

Accessible chromatin regions modulate gene expression by acting as cis-regulatory elements. Understanding the epigenetic landscape by mapping accessible regions of DNA is therefore imperative to decipher mechanisms of gene regulation under specific biological contexts of interest. The assay for transposase-accessible chromatin sequencing (ATAC-seq) has been widely used to detect accessible chromatin and the recent introduction of single-cell technology has increased resolution to the single-cell level. In a recent study, we used droplet-based, single-cell ATAC-seq technology (scATAC-seq) to reveal the epigenetic profile of the transit-amplifying subset of thymic epithelial cells (TECs), which was identified previously using single-cell RNA-sequencing technology (scRNA-seq). This protocol allows the preparation of nuclei from TECs in order to perform droplet-based scATAC-seq and its integrative analysis with scRNA-seq data obtained from the same cell population. Integrative analysis has the advantage of identifying cell types in scATAC-seq data based on cell cluster annotations in scRNA-seq analysis.

6.
Front Immunol ; 14: 1186154, 2023.
Article in English | MEDLINE | ID: mdl-38022666

ABSTRACT

The thymus has the ability to regenerate from acute injury caused by radiation, infection, and stressors. In addition to thymocytes, thymic epithelial cells in the medulla (mTECs), which are crucial for T cell self-tolerance by ectopically expressing and presenting thousands of tissue-specific antigens (TSAs), are damaged by these insults and recover thereafter. However, given recent discoveries on the high heterogeneity of mTECs, it remains to be determined whether the frequency and properties of mTEC subsets are restored during thymic recovery from radiation damage. Here we demonstrate that acute total body irradiation with a sublethal dose induces aftereffects on heterogeneity and gene expression of mTECs. Single-cell RNA-sequencing (scRNA-seq) analysis showed that irradiation reduces the frequency of mTECs expressing AIRE, which is a critical regulator of TSA expression, 15 days after irradiation. In contrast, transit-amplifying mTECs (TA-mTECs), which are progenitors of AIRE-expressing mTECs, and Ccl21a-expressing mTECs, were less affected. Interestingly, a detailed analysis of scRNA-seq data suggested that the proportion of a unique mTEC cluster expressing Ccl25 and a high level of TSAs was severely decreased by irradiation. In sum, we propose that the effects of acute irradiation disrupt the heterogeneity and properties of mTECs over an extended period, which potentially leads to an impairment of thymic T cell selection.


Subject(s)
Transcription Factors , Transcriptome , Mice , Animals , Transcription Factors/metabolism , Cell Differentiation , Mice, Inbred C57BL , Epithelial Cells/metabolism
7.
BMC Immunol ; 13: 58, 2012 Nov 02.
Article in English | MEDLINE | ID: mdl-23116172

ABSTRACT

BACKGROUND: Medullary thymic epithelial cells (mTECs) are characterized by ectopic expression of self-antigens during the establishment of central tolerance. The autoimmune regulator (Aire), which is specifically expressed in mTECs, is responsible for the expression of a large repertoire of tissue-restricted antigens (TRAs) and plays a role in the development of mTECs. However, Aire-deficient mTECs still express TRAs. Moreover, a subset of mTECs, which are considered to be at a stage of terminal differentiation, exists in the Aire-deficient thymus. The phenotype of a specific cell type in a multicellular organism is governed by the epigenetic regulation system. DNA methylation modification is an important component of this system. Every cell or tissue type displays a DNA methylation profile, consisting of tissue-dependent and differentially methylated regions (T-DMRs), and this profile is involved in cell-type-specific genome usage. The aim of this study was to examine the DNA methylation profile of mTECs by using Aire-deficient mTECs as a model. RESULTS: We identified the T-DMRs of mTECs (mTEC-T-DMRs) via genome-wide DNA methylation analysis of Aire(-/-) mTECs by comparison with the liver, brain, thymus, and embryonic stem cells. The hypomethylated mTEC-T-DMRs in Aire(-/-) mTECs were associated with mTEC-specific genes, including Aire, CD80, and Trp63, as well as other genes involved in the RANK signaling pathway. While these mTEC-T-DMRs were also hypomethylated in Aire(+/+) mTECs, they were hypermethylated in control thymic stromal cells. We compared the pattern of DNA methylation levels at a total of 55 mTEC-T-DMRs and adjacent regions and found that the DNA methylation status was similar for Aire(+/+) and Aire(-/-) mTECs but distinct from that of athymic cells and tissues. CONCLUSIONS: These results indicate a unique DNA methylation profile that is independent of Aire in mTECs. This profile is distinct from other cell types in the thymic microenvironment and is indicated to be involved in the differentiation of the mTEC lineage.


Subject(s)
DNA Methylation/genetics , Epithelial Cells/metabolism , Thymus Gland/cytology , Transcription Factors/deficiency , Animals , Biomarkers/metabolism , Cell Separation , Gene Expression Profiling , Gene Expression Regulation , Leukocyte Common Antigens/metabolism , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Organ Specificity/genetics , Stromal Cells/metabolism , Transcription Factors/metabolism , Transcription Initiation Site , AIRE Protein
8.
Elife ; 112022 05 17.
Article in English | MEDLINE | ID: mdl-35578835

ABSTRACT

Medullary thymic epithelial cells (mTECs) are critical for self-tolerance induction in T cells via promiscuous expression of tissue-specific antigens (TSAs), which are controlled by the transcriptional regulator, AIRE. Whereas AIRE-expressing (Aire+) mTECs undergo constant turnover in the adult thymus, mechanisms underlying differentiation of postnatal mTECs remain to be discovered. Integrative analysis of single-cell assays for transposase-accessible chromatin (scATAC-seq) and single-cell RNA sequencing (scRNA-seq) suggested the presence of proliferating mTECs with a specific chromatin structure, which express high levels of Aire and co-stimulatory molecules, CD80 (Aire+CD80hi). Proliferating Aire+CD80hi mTECs detected using Fucci technology express a minimal number of Aire-dependent TSAs and are converted into quiescent Aire+CD80hi mTECs expressing high levels of TSAs after a transit amplification. These data provide evidence for the existence of transit-amplifying Aire+mTEC precursors during the Aire+mTEC differentiation process of the postnatal thymus.


Subject(s)
Chromatin , Single-Cell Analysis , Animals , Cell Differentiation/genetics , Chromatin/metabolism , Epithelial Cells/metabolism , Mice , Mice, Inbred C57BL , Thymus Gland , Transposases/metabolism
9.
Biochem Biophys Res Commun ; 414(4): 773-8, 2011 Nov 04.
Article in English | MEDLINE | ID: mdl-22005462

ABSTRACT

NF-κB-inducing kinase (NIK) plays critical roles in the development of lymph nodes and Peyer's patches, and microarchitecture of the thymus and spleen via NF-κB activation. Alymphoplasia (aly/aly) mice have a point mutation in the NIK gene that causes a defect in the activation of an NF-κB member RelB. Here, we developed a novel method to determine the aly mutation by genetic typing using PCR. This method facilitated the easy establishment of a congeneic aly/aly mouse line. Indeed, we generated a mouse line with aly mutation on a BALB/cA background (BALB/cA-aly/aly). BALB/cA-aly/aly mice showed significant splenomegaly with extramedullary hemopoiesis, which was not significant in aly/aly mice on a C57BL/6 background. Interestingly, the splenomegaly and extramedullary hemopoiesis caused by the aly mutation was gender-dependent. These data together with previous reports on extramedullary hemopoiesis in RelB-deficient mice suggest that NIK-RelB signaling may be involved in the suppression of extramedullary hemopoiesis in adult mice.


Subject(s)
Gene Expression Regulation, Enzymologic , Hematopoiesis, Extramedullary/genetics , Protein Serine-Threonine Kinases/genetics , Transcription Factor RelB/metabolism , Animals , Base Sequence , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mutation , Spleen/physiology , Splenomegaly/genetics , Transcription Factor RelB/genetics , NF-kappaB-Inducing Kinase
10.
Biochem Biophys Res Commun ; 408(4): 530-6, 2011 May 20.
Article in English | MEDLINE | ID: mdl-21527253

ABSTRACT

Medullary thymic epithelial cells (mTECs) are essential for thymic negative selection to prevent autoimmunity. Previous studies show that mTEC development is dependent on the signal transducers TRAF6 and NIK. However, the downstream target genes of signals controlled by these molecules remain unknown. We performed a microarray analysis on mRNAs down-regulated by deficiencies in TRAF6 or functional NIK in an in vitro organ culture of fetal thymic stromata (2DG-FTOC). An in silico analysis of transcription factor binding sites in plausible promoter regions of differentially expressed genes suggests that STAT1 is involved in TRAF6- and NIK-dependent gene expression. Indeed, the signal of RANK, a TNF receptor family member that activates TRAF6 and NIK, induces the activation of STAT1 in 2DG-FTOC. Moreover, RANK signaling induces the up-regulation of interferon (IFN)-stimulated gene (ISG) expression, suggesting that the RANKL-dependent activation of STAT1 up-regulates ISG expression. The RANKL-dependent expression levels of ISGs were reduced but not completely abolished in interferon α receptor 1-deficient (Ifnar1(-/-)) 2DG-FTOC. Our data suggest that RANK signaling induces ISG expression in both type I interferon-independent and interferon-dependent mechanisms.


Subject(s)
Epithelial Cells/immunology , Gene Expression Regulation , Interferon Type I/immunology , Receptor Activator of Nuclear Factor-kappa B/metabolism , Self Tolerance/genetics , Thymus Gland/immunology , Animals , Fetus , Mice , Oligonucleotide Array Sequence Analysis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor, Interferon alpha-beta/genetics , Signal Transduction , Stromal Cells/immunology , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism , NF-kappaB-Inducing Kinase
11.
Front Immunol ; 12: 621824, 2021.
Article in English | MEDLINE | ID: mdl-33717123

ABSTRACT

Peripheral T cells capable of discriminating between self and non-self antigens are major components of a robust adaptive immune system. The development of self-tolerant T cells is orchestrated by thymic epithelial cells (TECs), which are localized in the thymic cortex (cortical TECs, cTECs) and medulla (medullary TECs, mTECs). cTECs and mTECs are essential for differentiation, proliferation, and positive and negative selection of thymocytes. Recent advances in single-cell RNA-sequencing technology have revealed a previously unknown degree of TEC heterogeneity, but we still lack a clear picture of the identity of TEC progenitors in the adult thymus. In this review, we describe both earlier and recent findings that shed light on features of these elusive adult progenitors in the context of tissue homeostasis, as well as recovery from stress-induced thymic atrophy.


Subject(s)
Adult Stem Cells/physiology , Autoantigens/immunology , Epithelial Cells/physiology , Immunologic Deficiency Syndromes/immunology , T-Lymphocytes/immunology , Thymus Gland/cytology , Animals , Cell Differentiation , Clonal Selection, Antigen-Mediated , Humans , Immune Tolerance , Immunity, Cellular
12.
Nat Commun ; 11(1): 6169, 2020 12 02.
Article in English | MEDLINE | ID: mdl-33268794

ABSTRACT

A repertoire of T cells with diverse antigen receptors is selected in the thymus. However, detailed mechanisms underlying this thymic positive selection are not clear. Here we show that the CCR4-NOT complex limits expression of specific genes through deadenylation of mRNA poly(A) tails, enabling positive selection. Specifically, the CCR4-NOT complex is up-regulated in thymocytes before initiation of positive selection, where in turn, it inhibits up-regulation of pro-apoptotic Bbc3 and Dab2ip. Elimination of the CCR4-NOT complex permits up-regulation of Bbc3 during a later stage of positive selection, inducing thymocyte apoptosis. In addition, CCR4-NOT elimination up-regulates Dab2ip at an early stage of positive selection. Thus, CCR4-NOT might control thymocyte survival during two-distinct stages of positive selection by suppressing expression levels of pro-apoptotic molecules. Taken together, we propose a link between CCR4-NOT-mediated mRNA decay and T cell selection in the thymus.


Subject(s)
Apoptosis/genetics , Exoribonucleases/genetics , Repressor Proteins/genetics , Thymocytes/immunology , Thymus Gland/immunology , Animals , Apoptosis/immunology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/immunology , Cell Differentiation , Cell Lineage/genetics , Cell Lineage/immunology , Exoribonucleases/immunology , Gene Expression Regulation, Developmental , Mice , Poly A/genetics , Poly A/immunology , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/immunology , Repressor Proteins/immunology , Signal Transduction , Thymocytes/cytology , Thymus Gland/cytology , Thymus Gland/growth & development , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/immunology , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/immunology
13.
Antimicrob Agents Chemother ; 53(6): 2510-6, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19289519

ABSTRACT

In our previous study, we found that the antibacterial peptide KLKLLLLLKLK-NH(2) (L5) and its d-enantiomer (DL5) activate neutrophils to produce superoxide anions (O(2)(-)) and prevent death due to infection by methicillin-resistant Staphylococcus aureus, suggesting that these peptides may elicit in vivo antimicrobial activities through host inflammatory responses mediated by neutrophils. In this study, we investigated the mechanisms behind in vivo antimicrobial prophylaxis by the use of L5 for the treatment of bacterial infection introduced via intra-abdominal implantation. We found that the intraperitoneal treatment with L5 before bacterial infection markedly reduced rates of death due to infection. Treatments with L5 were highly effective in preventing death due to intraperitoneal inoculation of not only S. aureus Smith but also Enterococcus faecalis SR1004 and Escherichia coli EC14. The intra-abdominal administration of L5 induced accumulation of neutrophils, increased levels of reactive oxygen species, and augmented antibacterial activity in the abdominal cavity. In addition, administration of L5 upregulated the expression of the Mig/CXCL9 chemokine gene in thioglycolate-elicited peritoneal macrophages. Our results suggested that the prevention of death by treatment of infected mice with L5 might occur primarily through the activation of a host immune response.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Bacterial Infections/immunology , Macrophage Activation/drug effects , Neutrophil Activation/drug effects , Oligopeptides/therapeutic use , Animals , Bacterial Infections/mortality , Chemokine CXCL9/genetics , Dose-Response Relationship, Drug , Female , Injections, Intraperitoneal , Mice , Mice, Inbred ICR , Reactive Oxygen Species/metabolism , Stereoisomerism
14.
Commun Biol ; 2: 444, 2019.
Article in English | MEDLINE | ID: mdl-31815199

ABSTRACT

Thymic crosstalk, a set of reciprocal regulations between thymocytes and the thymic environment, is relevant for orchestrating appropriate thymocyte development as well as thymic recovery from various exogenous insults. In this work, interactions shaping thymic crosstalk and the resultant dynamics of thymocytes and thymic epithelial cells are inferred based on quantitative analysis and modeling of the recovery dynamics induced by irradiation. The analysis identifies regulatory interactions consistent with known molecular evidence and reveals their dynamic roles in the recovery process. Moreover, the analysis also predicts, and a subsequent experiment verifies, a previously unrecognized regulation of CD4+CD8+ double positive thymocytes which temporarily increases their proliferation rate upon the decrease in their population size. Our model establishes a pivotal step towards the dynamic understanding of thymic crosstalk as a regulatory network system.


Subject(s)
Cell Communication , Cellular Microenvironment , Models, Biological , Thymocytes/metabolism , Thymus Gland/physiology , Algorithms , Animals , Cell Proliferation , Epithelial Cells/metabolism , Mice , Radiation, Ionizing , Recovery of Function , Thymocytes/radiation effects , Thymus Gland/radiation effects
15.
Sci Rep ; 9(1): 19866, 2019 12 27.
Article in English | MEDLINE | ID: mdl-31882694

ABSTRACT

The environment experienced during spaceflight may impact the immune system and the thymus appears to undergo atrophy during spaceflight. However, molecular aspects of this thymic atrophy remain to be elucidated. In this study, we analysed the thymi of mice on board the international space station (ISS) for approximately 1 month. Thymic size was significantly reduced after spaceflight. Notably, exposure of mice to 1 × g using centrifugation cages in the ISS significantly mitigated the reduction in thymic size. Although spaceflight caused thymic atrophy, the global thymic structure was not largely changed. However, RNA sequencing analysis of the thymus showed significantly reduced expression of cell cycle-regulating genes in two independent spaceflight samples. These reductions were partially countered by 1 × g exposure during the space flights. Thus, our data suggest that spaceflight leads to reduced proliferation of thymic cells, thereby reducing the size of the thymus, and exposure to 1 × g might alleviate the impairment of thymus homeostasis induced by spaceflight.


Subject(s)
Gravity, Altered , Space Flight , Thymus Gland/metabolism , Animals , Base Sequence , Enzyme-Linked Immunosorbent Assay , Male , Mice , Mice, Inbred C57BL , Principal Component Analysis , RNA-Seq
16.
Sci Rep ; 9(1): 7654, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31114014

ABSTRACT

Secondary lymphoid organs are critical for regulating acquired immune responses. The aim of this study was to characterize the impact of spaceflight on secondary lymphoid organs at the molecular level. We analysed the spleens and lymph nodes from mice flown aboard the International Space Station (ISS) in orbit for 35 days, as part of a Japan Aerospace Exploration Agency mission. During flight, half of the mice were exposed to 1 g by centrifuging in the ISS, to provide information regarding the effect of microgravity and 1 g exposure during spaceflight. Whole-transcript cDNA sequencing (RNA-Seq) analysis of the spleen suggested that erythrocyte-related genes regulated by the transcription factor GATA1 were significantly down-regulated in ISS-flown vs. ground control mice. GATA1 and Tal1 (regulators of erythropoiesis) mRNA expression was consistently reduced by approximately half. These reductions were not completely alleviated by 1 g exposure in the ISS, suggesting that the combined effect of space environments aside from microgravity could down-regulate gene expression in the spleen. Additionally, plasma immunoglobulin concentrations were slightly altered in ISS-flown mice. Overall, our data suggest that spaceflight might disturb the homeostatic gene expression of the spleen through a combination of microgravity and other environmental changes.


Subject(s)
GATA1 Transcription Factor/metabolism , Space Flight , Spleen/metabolism , Transcriptome , Animals , Down-Regulation , Erythropoiesis , GATA1 Transcription Factor/genetics , Mice , T-Cell Acute Lymphocytic Leukemia Protein 1/genetics , T-Cell Acute Lymphocytic Leukemia Protein 1/metabolism , Weightlessness/adverse effects
17.
J Biochem ; 142(1): 41-8, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17684029

ABSTRACT

N-beta-Alanyl-5-S-glutathionyl-3,4-dihydroxyphenylalanine (5-S-GAD), an insect-derived antibacterial peptide, generates hydrogen peroxide (H(2)O(2)) that exerts antitumour activity. We have investigated the precise mechanism of H(2)O(2) production from 5-S-GAD by autoxidation aiming to understand its action toward tumour cells. Using the electron spin resonance (ESR) technique, we detected a strong signal due to radical formation from 5-S-GAD. Surprisingly, the ESR signal of the radical derived from 5-S-GAD appeared after incubation for 30 min at 37 degrees C in the buffer at pH 7.4; the signal was persistently detected for 10 h in the absence of catalytic metal ions. The computer simulation of the observed ESR spectrum together with the theoretical calculation of the spin density of the radical species indicates that an o-semiquinone radical anion was formed from 5-S-GAD. We demonstrated that H(2)O(2) is produced via the formation of superoxide anion O2(.-) by the electron-transfer reduction of molecular oxygen by the 5-S-GAD anion, which is in equilibrium with 5-S-GAD in the aqueous solution. The radical formation and the subsequent H(2)O(2) production were inhibited by superoxide dismutase (SOD), when the antitumour activity of 5-S-GAD was inhibited by SOD. Thus, the formation of the o-semiquinone radical anion would be necessary for the antitumour activity of 5-S-GAD as an intermediate in the production of cytotoxic H(2)O(2).


Subject(s)
Antineoplastic Agents/chemistry , Benzoquinones/chemistry , Dihydroxyphenylalanine/analogs & derivatives , Glutathione/analogs & derivatives , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Antineoplastic Agents/metabolism , Antioxidants/chemistry , Antioxidants/metabolism , Benzoquinones/metabolism , Dihydroxyphenylalanine/chemistry , Dihydroxyphenylalanine/metabolism , Electron Spin Resonance Spectroscopy , Free Radicals/chemistry , Free Radicals/metabolism , Glutathione/chemistry , Glutathione/metabolism , Hydrogen Peroxide/metabolism , Hydrogen-Ion Concentration , Models, Molecular , Oxidation-Reduction , Superoxides/chemistry , Superoxides/metabolism , Time Factors
18.
Eur J Pharmacol ; 539(3): 151-7, 2006 Jun 13.
Article in English | MEDLINE | ID: mdl-16725138

ABSTRACT

N-beta-alanyl-5-S-glutathionyl-3,4-dihydroxyphenylalanine (5-S-GAD), an antibacterial substance isolated from the flesh fly, inhibits human tumor growth in the nude mice model; however, the mechanism of its action is unclear. The in vivo antitumor effect includes the inhibition of tumor cell proliferation and suppression of angiogenesis. Angiogenesis is essential for tumor growth in vivo. In this study, we examined whether 5-S-GAD inhibits tumor cell-induced angiogenesis by performing the mouse dorsal air sac assay. We found that intraperitoneal administration of 5-S-GAD inhibited the angiogenesis induced by S180 mouse sarcoma cells. Furthermore, 5-S-GAD also inhibited vascular endothelial growth factor-induced angiogenesis in the Matrigel plug assay and embryonic angiogenesis in the chick embryo chorioallantoic membrane assay. However, 5-S-GAD did not show any effect on the proliferation, migration, and tube formation of vascular endothelial cells. These results provide the first evidence that a bioactive substance derived from the flesh fly has antiangiogenic activity in vivo, although the mechanisms involved could not be explained.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Dihydroxyphenylalanine/analogs & derivatives , Glutathione/analogs & derivatives , Neovascularization, Pathologic/drug therapy , Angiogenesis Inhibitors/pharmacology , Animals , Cell Line, Tumor , Chick Embryo , Dihydroxyphenylalanine/pharmacology , Dihydroxyphenylalanine/therapeutic use , Dose-Response Relationship, Drug , Female , Glutathione/pharmacology , Glutathione/therapeutic use , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Xenograft Model Antitumor Assays/methods
19.
J Exp Med ; 213(8): 1441-58, 2016 07 25.
Article in English | MEDLINE | ID: mdl-27401343

ABSTRACT

Medullary thymic epithelial cells (mTECs) expressing autoimmune regulator (Aire) are critical for preventing the onset of autoimmunity. However, the differentiation program of Aire-expressing mTECs (Aire(+) mTECs) is unclear. Here, we describe novel embryonic precursors of Aire(+) mTECs. We found the candidate precursors of Aire(+) mTECs (pMECs) by monitoring the expression of receptor activator of nuclear factor-κB (RANK), which is required for Aire(+) mTEC differentiation. pMECs unexpectedly expressed cortical TEC molecules in addition to the mTEC markers UEA-1 ligand and RANK and differentiated into mTECs in reaggregation thymic organ culture. Introduction of pMECs in the embryonic thymus permitted long-term maintenance of Aire(+) mTECs and efficiently suppressed the onset of autoimmunity induced by Aire(+) mTEC deficiency. Mechanistically, pMECs differentiated into Aire(+) mTECs by tumor necrosis factor receptor-associated factor 6-dependent RANK signaling. Moreover, nonclassical nuclear factor-κB activation triggered by RANK and lymphotoxin-ß receptor signaling promoted pMEC induction from progenitors exhibiting lower RANK expression and higher CD24 expression. Thus, our findings identified two novel stages in the differentiation program of Aire(+) mTECs.


Subject(s)
Cell Differentiation/immunology , Epithelial Cells/immunology , Gene Expression Regulation/immunology , Mouse Embryonic Stem Cells/immunology , Thymus Gland/immunology , Transcription Factors/immunology , Animals , Cell Differentiation/genetics , Epithelial Cells/cytology , Gene Expression Regulation/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Mouse Embryonic Stem Cells/cytology , Plant Lectins/genetics , Plant Lectins/immunology , Thymus Gland/cytology , Transcription Factors/genetics , AIRE Protein
20.
Exp Anim ; 65(2): 175-87, 2016 May 20.
Article in English | MEDLINE | ID: mdl-26822934

ABSTRACT

The Japan Aerospace Exploration Agency developed the mouse Habitat Cage Unit (HCU) for installation in the Cell Biology Experiment Facility (CBEF) onboard the Japanese Experimental Module ("Kibo") on the International Space Station. The CBEF provides "space-based controls" by generating artificial gravity in the HCU through a centrifuge, enabling a comparison of the biological consequences of microgravity and artificial gravity of 1 g on mice housed in space. Therefore, prior to the space experiment, a ground-based study to validate the habitability of the HCU is necessary to conduct space experiments using the HCU in the CBEF. Here, we investigated the ground-based effect of a 32-day housing period in the HCU breadboard model on male mice in comparison with the control cage mice. Morphology of skeletal muscle, the thymus, heart, and kidney, and the sperm function showed no critical abnormalities between the control mice and HCU mice. Slight but significant changes caused by the HCU itself were observed, including decreased body weight, increased weights of the thymus and gastrocnemius, reduced thickness of cortical bone of the femur, and several gene expressions from 11 tissues. Results suggest that the HCU provides acceptable conditions for mouse phenotypic analysis using CBEF in space, as long as its characteristic features are considered. Thus, the HCU is a feasible device for future space experiments.


Subject(s)
Gravitation , Housing, Animal , Phenotype , Space Flight , Weightlessness , Animals , Femur/anatomy & histology , Heart/anatomy & histology , Kidney/anatomy & histology , Male , Mice, Inbred C57BL , Muscle, Skeletal/anatomy & histology , Spermatozoa/physiology , Thymus Gland/anatomy & histology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL