Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 4418, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38806459

ABSTRACT

The mechanisms by which the number of memory CD8 T cells is stably maintained remains incompletely understood. It has been postulated that maintaining them requires help from CD4 T cells, because adoptively transferred memory CD8 T cells persist poorly in MHC class II (MHCII)-deficient mice. Here we show that chronic interferon-γ signals, not CD4 T cell-deficiency, are responsible for their attrition in MHCII-deficient environments. Excess IFN-γ is produced primarily by endogenous colonic CD8 T cells in MHCII-deficient mice. IFN-γ neutralization restores the number of memory CD8 T cells in MHCII-deficient mice, whereas repeated IFN-γ administration or transduction of a gain-of-function STAT1 mutant reduces their number in wild-type mice. CD127high memory cells proliferate actively in response to IFN-γ signals, but are more susceptible to attrition than CD127low terminally differentiated effector memory cells. Furthermore, single-cell RNA-sequencing of memory CD8 T cells reveals proliferating cells that resemble short-lived, terminal effector cells and documents global downregulation of gene signatures of long-lived memory cells in MHCII-deficient environments. We propose that chronic IFN-γ signals deplete memory CD8 T cells by compromising their long-term survival and by diverting self-renewing CD127high cells toward terminal differentiation.


Subject(s)
CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Immunologic Memory , Interferon-gamma , STAT1 Transcription Factor , Animals , CD8-Positive T-Lymphocytes/immunology , Interferon-gamma/metabolism , Interferon-gamma/immunology , CD4-Positive T-Lymphocytes/immunology , Mice , STAT1 Transcription Factor/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/deficiency , Mice, Inbred C57BL , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Signal Transduction , Mice, Knockout , Memory T Cells/immunology , Memory T Cells/metabolism , Interleukin-7 Receptor alpha Subunit/metabolism , Cell Proliferation , Adoptive Transfer
2.
Immunity ; 47(2): 268-283.e9, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28778586

ABSTRACT

Foxp3 controls the development and function of regulatory T (Treg) cells, but it remains elusive how Foxp3 functions in vivo. Here, we established mouse models harboring three unique missense Foxp3 mutations that were identified in patients with the autoimmune disease IPEX. The I363V and R397W mutations were loss-of-function mutations, causing multi-organ inflammation by globally compromising Treg cell physiology. By contrast, the A384T mutation induced a distinctive tissue-restricted inflammation by specifically impairing the ability of Treg cells to compete with pathogenic T cells in certain non-lymphoid tissues. Mechanistically, repressed BATF expression contributed to these A384T effects. At the molecular level, the A384T mutation altered Foxp3 interactions with its specific target genes including Batf by broadening its DNA-binding specificity. Our findings identify BATF as a critical regulator of tissue Treg cells and suggest that sequence-specific perturbations of Foxp3-DNA interactions can influence specific facets of Treg cell physiology and the immunopathologies they regulate.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Diabetes Mellitus, Type 1/congenital , Diarrhea/genetics , Forkhead Transcription Factors/metabolism , Genetic Diseases, X-Linked/genetics , Immune System Diseases/congenital , Inflammation/genetics , T-Lymphocytes, Regulatory/physiology , Alleles , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Cell Differentiation , Cell Movement , Cells, Cultured , DNA Mutational Analysis , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Diarrhea/immunology , Forkhead Transcription Factors/genetics , Genetic Diseases, X-Linked/immunology , Humans , Immune System Diseases/genetics , Immune System Diseases/immunology , Inflammation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mutation, Missense/genetics , Organ Specificity/genetics
3.
Int Immunol ; 28(6): 293-305, 2016 06.
Article in English | MEDLINE | ID: mdl-26857736

ABSTRACT

Human CCR7(low)CD45RA(high) effector memory CD8(+) T cells (terminally differentiated TEMRA) are reportedly a functionally compromised population with characteristics of cellular senescence when examined ex vivo Although their frequencies are increased in elderly subjects in association with declined immune competence, however, it remains unclear whether their impaired functions can be reversed so that they contribute to immune responses in vivo Here, I show that, in contrast to TCR stimulation, stimulation of TEMRA with IL-15 induced a unique transcriptional signature, promoted IFN-γ production and cell cycle entry, and reduced chemotaxis toward sphingosine-1-phosphate (S1P). TEMRA preferentially accumulated in non-lymphoid tissues when transferred into IL-15-treated NOD.SCID.γc-deficient mice compared with non-treated mice. This accumulation was impaired by S1P receptor 1 over-expression. These results suggest that TEMRA act as functional effector T cells in non-lymphoid tissues when IL-15 is abundant and that IL-15 treatment may be beneficial in enhancing vaccine efficacy in elderly people.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Interleukin-15/immunology , T-Lymphocyte Subsets/immunology , Adjuvants, Immunologic , Adult , Aged , Animals , Cell Differentiation , Cells, Cultured , Cellular Senescence , Chemotaxis , Humans , Immunologic Memory , Interferon-gamma/metabolism , Lysophospholipids/metabolism , Mice, SCID , Middle Aged , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Transcriptome
4.
Eur J Immunol ; 45(3): 893-902, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25476730

ABSTRACT

Human CD8(+) T cells are functionally heterogeneous and can be divided into phenotypically and functionally distinct subsets according to CCR7 and CD45RA expression levels. Among these, CCR7(low) CD45RA(low) effector memory CD8(+) T cells (Tem) and CCR7(low) CD45RA(high) CD8(+) T cells, which are designated as Temra and considered to be terminally differentiated cells, are Ag-experienced T cells but show different functionalities. Here, we show that, while Tem proliferate vigorously and produce IFN-γ persistently and robustly, Temra proliferate poorly and lose the ability to produce IFN-γ over time after TCR stimulation. Temra showed impaired cell growth upon TCR stimulation, which was associated with defective activation of the mammalian target of rapamycin (mTOR) signaling. Furthermore, rapamycin, an inhibitor of mTOR signaling, interfered with the robust and continuous proliferation of and IFN-γ production by Tem at later time points after TCR stimulation. Thus, these data collectively indicate that activation of mTOR signaling is required for the robust functions of Tem cells in humans and suggest that defective mTOR signaling in Temra contributes to their functional impairment.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Immunologic Memory , Interferon-gamma/immunology , Signal Transduction/immunology , TOR Serine-Threonine Kinases/immunology , Female , Humans , Immunosuppressive Agents/pharmacology , Leukocyte Common Antigens/immunology , Male , Receptors, Antigen, T-Cell/immunology , Receptors, CCR7/immunology , Signal Transduction/drug effects , Sirolimus/pharmacology
5.
Immunity ; 36(2): 262-75, 2012 Feb 24.
Article in English | MEDLINE | ID: mdl-22326580

ABSTRACT

The emerging notion of environment-induced reprogramming of Foxp3(+) regulatory T (Treg) cells into helper T (Th) cells remains controversial. By genetic fate mapping or adoptive transfers, we have identified a minor population of nonregulatory Foxp3(+) T cells exhibiting promiscuous and transient Foxp3 expression, which gave rise to Foxp3(-) ("exFoxp3") Th cells and selectively accumulated in inflammatory cytokine milieus or in lymphopenic environments including those in early ontogeny. In contrast, Treg cells did not undergo reprogramming under those conditions irrespective of their thymic or peripheral origins. Moreover, although a few Treg cells transiently lose Foxp3 expression, such "latent" Treg cells retained their memory and robustly re-expressed Foxp3 and suppressive function upon activation. This study establishes that Treg cells constitute a stable cell lineage, whose committed state in a changing environment is ensured by DNA demethylation of the Foxp3 locus irrespectively of ongoing Foxp3 expression.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Forkhead Transcription Factors/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , CD2 Antigens/genetics , CD2 Antigens/metabolism , CD4-Positive T-Lymphocytes/cytology , Cell Differentiation , Cell Lineage/genetics , Cell Lineage/immunology , DNA Methylation , Epigenesis, Genetic , Forkhead Transcription Factors/genetics , Gene Expression , Humans , Immunologic Memory , In Vitro Techniques , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocyte Activation , Lymphopenia/immunology , Lymphopenia/metabolism , Lymphopenia/pathology , Mice , Mice, Knockout , Mice, Transgenic , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/cytology
6.
J Immunol ; 183(7): 4467-74, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19734230

ABSTRACT

In the thymus, the transcription factor ThPOK is essential for the development of the CD4 helper T cell lineage, whereas active repression of ThPOK is critical for the development of the CD8 cytotoxic T cell lineage. ThPOK gene silencing is thought to be irreversible in peripheral CD8 T cells. We noticed that ThPOK repression is readily abrogated upon in vitro TCR stimulation of peripheral CD8 T cells. This observation prompted us to investigate a role for ThPOK in the CD8 T cell response to an acute viral infection. We observed that a functional deficiency of ThPOK does not affect CD8 T cell differentiation into effector T cells and the long-term persistence of Ag-specific memory T cells. However, in the absence of functional ThPOK, clonal expansion is significantly less in both primary and secondary CD8 T cell responses. Long-lived, Ag-specific CD8 T cells with a functional deficiency in ThPOK fail to produce high amounts of IL-2 and also fail to express high levels of granzyme B upon rechallenge. Our data reveal an unexpected role for ThPOK in CD8 T cells in promoting expansion and boosting the response to antigenic challenge.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Transcription Factors/biosynthesis , Transcription Factors/deficiency , Acute Disease , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Gene Knock-In Techniques , Immunologic Memory/genetics , Lymphocytic Choriomeningitis/genetics , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Transcription Factors/genetics , Transcription Factors/physiology
7.
Science ; 319(5864): 822-5, 2008 Feb 08.
Article in English | MEDLINE | ID: mdl-18258917

ABSTRACT

Mouse CD4+CD8+ double-positive (DP) thymocytes differentiate into CD4+ helper-lineage cells upon expression of the transcription factor Th-POK but commit to the CD8+ cytotoxic lineage in its absence. We report the redirected differentiation of class I-restricted thymocytes into CD4+CD8- helper-like T cells upon loss of Runx transcription factor complexes. A Runx-binding sequence within the Th-POK locus acts as a transcriptional silencer that is essential for Th-POK repression and for development of CD8+ T cells. Thus, Th-POK expression and genetic programming for T helper cell development are actively inhibited by Runx-dependent silencer activity, allowing for cytotoxic T cell differentiation. Identification of the transcription factors network in CD4 and CD8 lineage choice provides insight into how distinct T cell subsets are developed for regulating the adaptive immune system.


Subject(s)
Core Binding Factor Alpha 2 Subunit/physiology , Core Binding Factor Alpha 3 Subunit/physiology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Cytotoxic/immunology , Transcription Factors/physiology , Animals , Cell Differentiation , Cell Lineage , Chromatin Immunoprecipitation , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/genetics , Core Binding Factor beta Subunit/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/immunology , Mice , Mice, Transgenic , Molecular Sequence Data , Silencer Elements, Transcriptional , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Transcription Factors/genetics
8.
Bioinformatics ; 23(21): 2934-41, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17893089

ABSTRACT

MOTIVATION: Although a huge amount of mammalian genomic data does become publicly available, there are still hurdles for biologists to overcome before such data can be fully exploited. One of the challenges for gaining biological insight from genomic data has been the inability to cross-reference transcriptomic and proteomic data using a single informational platform. To address this, we constructed an open-access database that enabled us to cross-reference transcriptomic and proteomic data obtained from immune cells. RESULTS: The database, named RefDIC (Reference genomics Database of Immune Cells), currently contains: (i) quantitative mRNA profiles for human and mouse immune cells/tissues obtained using Affymetrix GeneChip technology; (ii) quantitative protein profiles for mouse immune cells obtained using two-dimensional gel electrophoresis (2-DE) followed by image analysis and mass spectrometry and (iii) various visualization tools to cross-reference the mRNA and protein profiles of immune cells. RefDIC is the first open-access database for immunogenomics and serves as an important information-sharing platform, enabling a focused genomic approach in immunology. AVAILABILITY: All raw data and information can be accessed from http://refdic.rcai.riken.jp/. The microarray data is also available at http://cibex.nig.ac.jp/ under CIBEX accession no. CBX19, and http://www.ebi.ac.uk/pride/ under PRIDE accession numbers 2354-2378 and 2414.


Subject(s)
Database Management Systems , Databases, Factual , Information Storage and Retrieval/methods , Internet , Lymphocytes/immunology , Proteome/immunology , Transcription Factors/immunology , Animals , Humans , Systems Integration
9.
J Exp Med ; 204(8): 1749-55, 2007 Aug 06.
Article in English | MEDLINE | ID: mdl-17646405

ABSTRACT

Interferon gamma (IFN gamma) is the hallmark cytokine produced by T helper type 1 (Th1) cells, whereas interleukin (IL)-4 is the hallmark cytokine produced by Th2 cells. Although previous studies have revealed the roles of cytokine signaling and of transcription factors during differentiation of Th1 or Th2 cells, it is unclear how the exclusive expression pattern of each hallmark cytokine is established. The DNaseI hypersensitivity site IV within the mouse Il4 locus plays an important role in the repression of Il4 expression in Th1 cells, and it has been named the Il4 silencer. Using Cbf beta- or Runx3-deficient T cells, we show that loss of Runx complex function results in derepression of IL-4 in Th1 cells. Binding of Runx complexes to the Il4 silencer was detected in naive CD4(+) T cells and Th1 cells, but not in Th2 cells. Furthermore, enforced expression of GATA-3 in Th1 cells inhibited binding of Runx complexes to the Il4 silencer. Interestingly, T cell-specific inactivation of the Cbf beta gene in mice led to elevated serum immunoglobulin E and airway infiltration. These results demonstrate critical roles of Runx complexes in regulating immune responses, at least in part, through the repression of the Il4 gene.


Subject(s)
CCAAT-Binding Factor/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Enhancer Elements, Genetic , Interleukin-4/genetics , Interleukin-4/metabolism , Th1 Cells/metabolism , Animals , Asthma/metabolism , CD28 Antigens/biosynthesis , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation , Gene Silencing , Immune System , Mice , Protein Binding , T-Lymphocytes/metabolism
10.
Immunol Rev ; 212: 8-27, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16903903

ABSTRACT

Naturally arising CD25+ CD4+ regulatory T (Treg) cells, most of which are produced by the normal thymus as a functionally mature T-cell subpopulation, play key roles in the maintenance of immunologic self-tolerance and negative control of a variety of physiological and pathological immune responses. Natural Tregs specifically express Foxp3, a transcription factor that plays a critical role in their development and function. Complete depletion of Foxp3-expressing natural Tregs, whether they are CD25+ or CD25-, activates even weak or rare self-reactive T-cell clones, inducing severe and widespread autoimmune/inflammatory diseases. Natural Tregs are highly dependent on exogenously provided interleukin (IL)-2 for their survival in the periphery. In addition to Foxp3 and IL-2/IL-2 receptor, deficiency or functional alteration of other molecules, expressed by T cells or non-T cells, may affect the development/function of Tregs or self-reactive T cells, or both, and consequently tip the peripheral balance between the two populations toward autoimmunity. Elucidation of the molecular and cellular basis of this Treg-mediated active maintenance of self-tolerance will facilitate both our understanding of the pathogenetic mechanism of autoimmune disease and the development of novel methods of autoimmune disease prevention and treatment via enhancing and re-establishing Treg-mediated dominant control over self-reactive T cells.


Subject(s)
Autoimmune Diseases/immunology , Forkhead Transcription Factors/physiology , Receptors, Interleukin-2/analysis , Self Tolerance , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmune Diseases/therapy , CD4 Antigens/analysis , Forkhead Transcription Factors/metabolism , Humans , Lymphocyte Activation , Mice , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/drug effects
11.
J Exp Med ; 201(5): 723-35, 2005 Mar 07.
Article in English | MEDLINE | ID: mdl-15753206

ABSTRACT

Interleukin (IL)-2 plays a crucial role in the maintenance of natural immunologic self-tolerance. Neutralization of circulating IL-2 by anti-IL-2 monoclonal antibody for a limited period elicits autoimmune gastritis in BALB/c mice. Similar treatment of diabetes-prone nonobese diabetic mice triggers early onset of diabetes and produces a wide spectrum of T cell-mediated autoimmune diseases, including gastritis, thyroiditis, sialadenitis, and notably, severe neuropathy. Such treatment selectively reduces the number of Foxp3-expressing CD25(+) CD4(+) T cells, but not CD25(-) CD4(+) T cells, in the thymus and periphery of normal and thymectomized mice. IL-2 neutralization inhibits physiological proliferation of peripheral CD25(+) CD4(+) T cells that are presumably responding to normal self-antigens, whereas it is unable to inhibit their lymphopenia-induced homeostatic expansion in a T cell-deficient environment. In normal naive mice, CD25(low) CD4(+) nonregulatory T cells actively transcribe the IL-2 gene and secrete IL-2 protein in the physiological state. IL-2 is thus indispensable for the peripheral maintenance of natural CD25(+) CD4(+) regulatory T cells (T reg cells). The principal physiological source of IL-2 for the maintenance of T reg cells appears to be other T cells, especially CD25(low) CD4(+) activated T cells, which include self-reactive T cells. Furthermore, impairment of this negative feedback loop via IL-2 can be a cause and a predisposing factor for autoimmune disease.


Subject(s)
Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , DNA-Binding Proteins/metabolism , Homeostasis , Interleukin-2/antagonists & inhibitors , Interleukin-2/immunology , Receptors, Interleukin-2/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Autoimmune Diseases/pathology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Cell Proliferation , Diabetes Mellitus/immunology , Diabetes Mellitus/pathology , Forkhead Transcription Factors , Gastritis/immunology , Gastritis/pathology , Interleukin-2/genetics , Lymphopenia/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Neuritis, Autoimmune, Experimental/immunology , Neuritis, Autoimmune, Experimental/pathology , Receptors, Interleukin-2/immunology , Transcription, Genetic/genetics
12.
Int Immunol ; 16(8): 1189-201, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15237110

ABSTRACT

Naturally arising CD25(+)CD4(+) regulatory T (T(R)) cells can be exploited to establish immunologic tolerance to non-self antigens. In vivo exposure of CD25(+)CD4(+) T cells from normal naive mice to alloantigen in a T cell-deficient environment elicited spontaneous expansion of alloantigen-specific CD25(+)CD4(+) T(R) cells, which suppressed allograft rejection mediated by subsequently transferred naive T cells, leading to long-term graft tolerance. The expanded T(R) cells, which became CD25(low) in the absence of other T cells, stably sustained suppressive activity, maintained expression levels of other T(R) cell-associated molecules, including Foxp3, CTLA-4 and GITR, and could adoptively transfer tolerance to normal mice. Furthermore, specific removal of the T(R) cells derived from originally transferred CD25(+)CD4(+) T(R) cells evoked graft rejection in the long-term tolerant mice, indicating that any T(R) cells deriving from CD25(-)CD4(+) naive T cells minimally contribute to graft tolerance and that natural T(R) cells are unable to infectiously confer significant suppressive activity to other T cells. Similar antigen-specific expansion of T(R) cells can also be achieved in vitro by stimulating naturally present CD25(+)CD4(+) T cells with alloantigen in the presence of IL-2. The expanded CD25(+)CD4(+) T cells potently suppressed even secondary MLR in vitro and, by in vivo transfer, established antigen-specific long-term graft tolerance. Thus, in vivo or in vitro, direct or indirect ways of antigen-specific expansion of naturally arising Foxp3(+)CD25(+)CD4(+) T(R) cells can establish antigen-specific dominant tolerance to non-self antigens, and would also be instrumental in re-establishing self-tolerance in autoimmune disease and antigen-specific negative control of pathological immune responses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , DNA-Binding Proteins/immunology , Receptors, Interleukin-2/immunology , Skin Transplantation/immunology , Transplantation Tolerance/immunology , Transplantation, Homologous/immunology , Adoptive Transfer , Animals , Antigens/immunology , Antigens, CD , Antigens, Differentiation/immunology , CD4-Positive T-Lymphocytes/transplantation , CTLA-4 Antigen , Cell Count , Cells, Cultured , Forkhead Transcription Factors , Glucocorticoid-Induced TNFR-Related Protein , Mice , Mice, Inbred BALB C , Receptors, Nerve Growth Factor/immunology , Receptors, Tumor Necrosis Factor/immunology
13.
Microbiol Immunol ; 48(1): 7-13, 2004.
Article in English | MEDLINE | ID: mdl-14734853

ABSTRACT

The effects of tacrolimus on insulin-dependent diabetes mellitus (IDDM) induced by the D-variant of encephalomyocarditis virus (D-EMCV) have been investigated. Male BALB/c mice were treated with tacrolimus before viral inoculation, and then were inoculated with 10 plaque forming units (PFU) of DEMCV. The mice continued to be treated with tacrolimus until the animals were sacrificed. D-EMCV-infected mice, which were treated with saline as controls, showed abnormal glucose tolerance test (GTT) values, whereas all infected mice with tacrolimus pretreatment were normal on 7 days-post inoculation (DPI). Histological observations revealed that non-treated tacrolimus D-EMCV-infected mice and which developed diabetes showed severe insulitis in their islets of Langerhans. On the other hand, D-EMCV-infected mice treated with tacrolimus were normal. In D-EMCV-infected mice, viruses in the pancreata were detected at the same level regardless of treatment with tacrolimus or saline. Expressions of TNF-alpha and IFN-gamma mRNA in spleens of tacrolimus-treated D-EMCV-infected mice were lower than that of non-treated tacrolimus DEMCV-infected mice on 7 DPI. The results suggest that tacrolimus suppresses expressions of TNF-alpha and IFN-gamma mRNAs to prevent the onset of D-EMCV-induced IDDM.


Subject(s)
Cardiovirus Infections/complications , Diabetes Mellitus, Type 1/prevention & control , Diabetes Mellitus, Type 1/virology , Encephalomyocarditis virus , Tacrolimus/pharmacology , Animals , Blood Glucose/analysis , Cardiovirus Infections/pathology , Diabetes Mellitus, Type 1/pathology , Encephalomyocarditis virus/growth & development , Encephalomyocarditis virus/isolation & purification , Gene Expression , Glucose Tolerance Test , Immunosuppressive Agents/pharmacology , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred BALB C , Pancreas/pathology , Pancreas/virology , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Spleen/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Viral Load
14.
Biochem Biophys Res Commun ; 311(2): 501-5, 2003 Nov 14.
Article in English | MEDLINE | ID: mdl-14592444

ABSTRACT

Tumor necrosis factor-alpha (TNF-alpha) is a cytokine considered to play a key role in beta-cell destruction in insulin-dependent diabetes mellitus (IDDM). Serum thymic factor (Facteur thymique serique; FTS) is a nonapeptide thymus hormone known to inhibit IDDM in a mouse model. In this study, the effect of TNF-alpha on the murine pancreatic beta-cell line MIN6 was examined. Cell shrinkage and detachment were seen in cells treated with 0-50 ng/ml TNF-alpha for 12h. Oligonucleosomal DNA fragmentation was determined from non-adherent cells, indicating that the TNF-alpha-induced cell destruction was attributed to apoptosis. Fragmented DNA was quantified by enzyme-linked immunosorbent assay to measure the amount of histone-bound oligonucleosomes. FTS was treated with TNF-alpha and the percentage of fragmented DNA was analyzed. The data indicate a distinct reduction of fragmented DNA at a concentration of 1 ng/ml FTS. Expression of TNF receptor I, inducible form of nitric oxide synthase (iNOS), interleukin-1 beta-converting enzyme (ICE), Bcl-2, and nuclear factor kappa B (NF-kappa B) was analyzed by reverse transcriptase-polymerase chain reaction to investigate the suppressor mechanism of FTS on TNF-alpha-induced apoptosis. FTS treatment suppressed the expression of iNOS and Bcl-2 mRNA in TNF-alpha-treated cells. The expression of NF-kappa B mRNA in TNF-alpha-treated cells was enhanced after FTS treatment, while that of ICE mRNA did not change in TNF-alpha-treated cells with or without FTS treatment. These results suggest that the inhibition of MIN6 cell death by FTS on TNF-alpha-induced apoptosis is caused by a negative feedback mechanism involving the inhibition of iNOS induction.


Subject(s)
Apoptosis/drug effects , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Thymic Factor, Circulating/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cell Line , DNA Damage , Dose-Response Relationship, Drug , Drug Interactions , Islets of Langerhans/cytology , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...