Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Nat Cell Biol ; 23(12): 1255-1270, 2021 12.
Article in English | MEDLINE | ID: mdl-34876687

ABSTRACT

Circadian rhythms align physiological functions with the light-dark cycle through oscillatory changes in the abundance of proteins in the clock transcriptional programme. Timely removal of these proteins by different proteolytic systems is essential to circadian strength and adaptability. Here we show a functional interplay between the circadian clock and chaperone-mediated autophagy (CMA), whereby CMA contributes to the rhythmic removal of clock machinery proteins (selective chronophagy) and to the circadian remodelling of a subset of the cellular proteome. Disruption of this autophagic pathway in vivo leads to temporal shifts and amplitude changes of the clock-dependent transcriptional waves and fragmented circadian patterns, resembling those in sleep disorders and ageing. Conversely, loss of the circadian clock abolishes the rhythmicity of CMA, leading to pronounced changes in the CMA-dependent cellular proteome. Disruption of this circadian clock/CMA axis may be responsible for both pathways malfunctioning in ageing and for the subsequently pronounced proteostasis defect.


Subject(s)
ARNTL Transcription Factors/genetics , CLOCK Proteins/metabolism , Chaperone-Mediated Autophagy/physiology , Circadian Clocks/physiology , Circadian Rhythm/physiology , Lysosomal-Associated Membrane Protein 2/genetics , Aging/physiology , Animals , Lysosomes/chemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Photoperiod , Proteome/genetics , Proteostasis/physiology , Sleep Deprivation/physiopathology , Transcription, Genetic/genetics
2.
J Immunol ; 196(4): 1964-76, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26755821

ABSTRACT

Immune responses against cancer cells are often hindered by immunosuppressive mechanisms that are developed in the tumor microenvironment. Induction of a hyporesponsive state in tumor Ag-specific T cells is one of the major events responsible for the inability of the adaptive immune system to mount an efficient antitumor response and frequently contributes to lessen the efficacy of immunotherapeutic approaches. Treatment of localized tumors by focused ultrasound (FUS) is a minimally invasive therapy that uses a range of input energy for in situ tumor ablation through the generation of thermal and cavitation effect. Using a murine B16 melanoma tumor model, we show that a variant of FUS that delivers a reduced level of energy at the focal point and generates mild mechanical and thermal stress in target cells has the ability to increase immunogenic presentation of tumor Ags, which results in reversal of tumor-induced T cell tolerance. Furthermore, we show that the combination of nonablative low-energy FUS with an ablative hypofractionated radiation therapy results in synergistic control of primary tumors and leads to a dramatic reduction in spontaneous pulmonary metastases while prolonging recurrence-free survival only in immunocompetent mice.


Subject(s)
Melanoma, Experimental/immunology , Skin Neoplasms/immunology , T-Lymphocytes/immunology , Tumor Escape/immunology , Ultrasonic Therapy/methods , Animals , Combined Modality Therapy , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Metastasis/pathology , Neoplasm Metastasis/therapy , Radiotherapy , Real-Time Polymerase Chain Reaction , Skin Neoplasms/pathology , Skin Neoplasms/therapy
3.
EMBO Rep ; 15(9): 991-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25074018

ABSTRACT

Regulatory T cells (Tregs) control autoreactive T cells by inhibiting activation-induced proliferation and cytokine expression. The molecular mechanisms responsible for the inactivation of effector T cells by Tregs remain yet to be fully characterized. We report that T-helper cells stimulated in the presence of Tregs quickly activate NFAT1 and have increased NFAT1-dependent expression of the transcription repressor Ikaros. NFAT1 deficiency or dominant-negative Ikaros compromises Treg-mediated inhibition of T-helper cells in vitro and in vivo. Thus, our results place NFAT-dependent mechanisms as general regulators of T-cell tolerance and show that Treg-mediated suppression of T-helper cells results from the activation of NFAT-regulated gene expression.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Ikaros Transcription Factor/biosynthesis , NFATC Transcription Factors/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation , Cytokines/biosynthesis , Gene Expression Regulation , Ikaros Transcription Factor/genetics , Lymphocyte Activation/immunology , Mice , NFATC Transcription Factors/biosynthesis , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/metabolism
4.
Cancer Res ; 74(14): 3695-706, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24830722

ABSTRACT

Tumor cells must overcome apoptosis to survive throughout metastatic dissemination and distal organ colonization. Here, we show in the Polyoma Middle T mammary tumor model that N-cadherin (Cdh2) expression causes Slug (Snai2) upregulation, which in turn promotes carcinoma cell survival. Slug was dramatically upregulated in metastases relative to primary tumors. Consistent with a role in metastasis, Slug knockdown in carcinoma cells suppressed lung colonization by decreasing cell survival at metastatic sites, but had no effect on tumor cell invasion or extravasation. In support of this idea, Slug inhibition by shRNA sensitized tumor cells to apoptosis by DNA damage, resulting in caspase-3 and PARP cleavage. The prosurvival effect of Slug was found to be caused by direct repression of the proapoptotic gene, Puma (Bbc3), by Slug. Consistent with a pivotal role for a Slug-Puma axis in metastasis, inhibition of Puma by RNA interference in Slug-knockdown cells rescued lung colonization, whereas Puma overexpression in control tumor cells suppressed lung metastasis. The survival function of the Slug-Puma axis was confirmed in human breast cancer cells, where Slug knockdown increased Puma expression and inhibited lung colonization. This study demonstrates a pivotal role for Slug in carcinoma cell survival, implying that disruption of the Slug-Puma axis may impinge on the survival of metastatic cells.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Apoptosis/genetics , Neoplasms/genetics , Neoplasms/pathology , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Animals , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Survival/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Lung Neoplasms/secondary , Neoplasm Metastasis , RNA Interference , Receptors, Fibroblast Growth Factor/metabolism , Snail Family Transcription Factors , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism
5.
Mol Cell Biol ; 34(2): 233-45, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24190972

ABSTRACT

In response to suboptimal activation, T cells become hyporesponsive, with a severely reduced capacity to proliferate and produce cytokines upon reencounter with antigen. Chromatin analysis of T cells made tolerant by use of different in vitro and in vivo approaches reveals that the expression of gamma interferon (IFN-γ) is epigenetically silenced in anergic effector TH1 cells. In those T cells, calcium signaling triggers the expression of Tle4, a member of the Groucho family of corepressors, which is then recruited to a distal regulatory element in the Ifng locus and causes the establishment of repressive epigenetic marks at the Ifng gene regulatory elements. Consequently, impaired Tle4 activity results in a markedly reduced capacity to inhibit IFN-γ production in tolerized T cells. We propose that Blimp1-dependent recruitment of Tle4 to the Ifng locus causes epigenetic silencing of the expression of the Ifng gene in anergic TH1 cells. These results define a novel function of Groucho family corepressors in peripheral T cells and demonstrate that specific mechanisms are activated in tolerant T helper cells to directly repress expression of effector cytokines, supporting the hypothesis that stable epigenetic imprinting contributes to the maintenance of the tolerance-associated hyporesponsive phenotype in T cells.


Subject(s)
Epigenetic Repression , Interferon-gamma/genetics , Repressor Proteins/physiology , Th1 Cells/immunology , Th2 Cells/immunology , Acetylation , Animals , Base Sequence , Cell Proliferation , Cells, Cultured , Clonal Anergy , Conserved Sequence , Down-Regulation , Female , Histones/metabolism , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Positive Regulatory Domain I-Binding Factor 1 , Promoter Regions, Genetic , Protein Processing, Post-Translational , Transcription Factors/metabolism
6.
Eur J Immunol ; 42(9): 2471-83, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22684523

ABSTRACT

Anergy is induced in T cells as a consequence of a partial or suboptimal stimulation. Anergic T cells become unresponsive and fail to proliferate and produce cytokines. We had previously shown that in anergic CD4(+) T cells, Ikaros participates in the transcriptional repression of the Il2 gene by recruiting histone deacetylases that cause core histone deacetylation at the Il2 promoter. Here we show that deacetylation at the Il2 promoter is the initial step in a process that leads to the stable silencing of the Il2 gene transcription in anergic T cells. We have found that anergy-induced deacetylation of the Il2 promoter permits binding of the histone methyl-transferase Suv39H1, which trimethylates lysine-9 of histone H3 (Me3H3-K9). Furthermore, the establishment of the Me3H3-K9 mark allows the recruitment of the heterochromatin protein HP1, allowing the silenced Il2 loci to reposition close to heterochromatin-rich regions. Our results indicate that silencing of Il2 transcription in anergic T cells is attained through a series of epigenetic changes that involve the establishment of repressive marks and the subsequent nuclear repositioning of the Il2 loci, which become juxtaposed to transcriptionally silent regions. This mechanism may account for the stable nature of the inhibition of IL-2 production in anergic cells.


Subject(s)
Interleukin-2/genetics , T-Lymphocytes/physiology , Animals , Cells, Cultured , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA Methylation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Gene Expression Regulation , Gene Silencing , Heterochromatin/genetics , Heterochromatin/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Histones/metabolism , Interleukin-2/biosynthesis , Interleukin-2/metabolism , Methyltransferases/genetics , Methyltransferases/metabolism , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Repressor Proteins/genetics , Repressor Proteins/metabolism , T-Lymphocytes/metabolism , Transcription, Genetic
7.
J Exp Med ; 206(4): 867-76, 2009 Apr 13.
Article in English | MEDLINE | ID: mdl-19307325

ABSTRACT

In T cells, anergy can be induced after T cell receptor engagement in the absence of costimulation. Under these conditions, the expression of a specific set of anergy-associated genes is activated. Several lines of evidence suggest that nuclear factor of activated T cells (NFAT) proteins may regulate the expression of many of those genes; however, the nature of the complexes responsible for the induction of this new program of gene expression is unknown. Here, we show that transcriptional complexes formed by NFAT homodimers are directly responsible for the activation of at least two anergy-inducing genes, Grail and Caspase3. Our data shows that Grail expression is activated by direct binding of NFAT dimers to the Grail promoter at two different sites. Consequently, a mutant NFAT protein with impaired ability to dimerize is not able to induce an unresponsive state in T cells. Our results not only identify a new biological function for NFAT dimers but also reveal the different nature of NFAT-containing complexes that induce anergy versus those that are activated during a productive immune response. These data also establish a basis for the design of immunomodulatory strategies that specifically target each type of complex.


Subject(s)
Immune Tolerance , NFATC Transcription Factors/genetics , T-Lymphocytes/immunology , Transcription, Genetic , Caspase 3/genetics , Dimerization , Gene Expression Regulation , Humans , Jurkat Cells/immunology , Mutation , Promoter Regions, Genetic , Receptors, Antigen, T-Cell/immunology , Ubiquitin-Protein Ligases/genetics
8.
Semin Immunol ; 19(3): 180-7, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17387022

ABSTRACT

Self-reactive T cells that escape negative selection in the thymus must be kept under control in the periphery. Mechanisms of peripheral tolerance include deletion or functional inactivation of self-reactive T cells and mechanisms of dominant tolerance mediated by regulatory T cells. In the absence of costimulation, T cell receptor (TCR) engagement results in unopposed calcium signaling that leads to the activation of a cell-intrinsic program of inactivation, which makes T cells hyporesponsive to subsequent stimulations. The activation of this program in anergic T cells is a consequence of the induction of a nuclear factor of activated T cells (NFAT)-dependent program of gene expression. Recent studies have offered new insights into the mechanisms responsible for the implementation and maintenance of T cell anergy and have provided evidence that the proteins encoded by the genes upregulated in anergic T cells are responsible for the implementation of anergy by interfering with TCR signaling or directly inhibiting cytokine gene transcription.


Subject(s)
Immune Tolerance/physiology , T-Lymphocytes/immunology , Transcription, Genetic , Animals , Clonal Anergy , Gene Expression Regulation/immunology , Humans
9.
Blood ; 109(7): 2878-86, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17148585

ABSTRACT

In T cells anergy may be evoked by an unbalanced stimulation of the T-cell receptor in the absence of costimulation. Anergic T cells are unresponsive to new antigen receptor engagement and do not produce interleukin 2. We present evidence that anergizing stimuli induce changes in histone acetylation, which mediates transcriptional repression of interleukin 2 expression. In response to calcium signaling, anergic T cells up-regulate the expression of Ikaros, a zinc finger transcription factor essential for lymphoid lineage determination. Ikaros binds to the interleukin 2 promoter where it induces histone deacetylation. Confirming the role of Ikaros in the induction of T-cell anergy, cells with reduced Ikaros activity show defective inactivation in response to an anergizing stimulus. We propose a model in which tolerizing stimuli induce epigenetic changes on the interleukin 2 locus that are responsible for the stable inhibition of the expression of this cytokine in anergic T cells.


Subject(s)
Clonal Anergy/genetics , Clonal Anergy/immunology , Histones/metabolism , Ikaros Transcription Factor/metabolism , Interleukin-2/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Acetylation , Animals , Epigenesis, Genetic , Humans , Ikaros Transcription Factor/genetics , In Vitro Techniques , Jurkat Cells , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Models, Immunological , Promoter Regions, Genetic , Th1 Cells/immunology , Th1 Cells/metabolism , Transcription, Genetic
10.
Eur J Haematol ; 75(1): 47-53, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15946310

ABSTRACT

OBJECTIVE: To correlate different polymorphisms of the beta-globin cluster with fetal hemoglobin (HbF) level in beta-thalassemia and E-beta thalassemia patients. METHODS: Fifteen thalassemia patients, seven with high HbF and not requiring transfusion, eight with lower HbF and requiring transfusion were studied for beta-globin mutation, concurrent inheritance of alpha-thalassemia, RFLP haplotype, a C-->T polymorphism at -158 of Ggamma and configuration of an (AT)(x)T(y) motif at -540 of beta-globin gene. RESULTS: Senegal 5'beta-haplotype and the polymorphism at -158 of G(gamma) was (P = 0.063) was linked to the high-HbF phenotype but the (AT)(9)T(5) configuration of the (AT)(x)T(y) motif was not (P = 0.6). Study of 30 chromosomes revealed 7 different configurations of the (AT)(x)T(y) motif. Association of these motifs with specific beta-globin mutations of this region has also been determined. CONCLUSION: The senegal haplotype and the polymorphism at -158 of G(gamma) was linked to the high-HbF phenotype.


Subject(s)
Fetal Hemoglobin , Hemoglobins, Abnormal/genetics , Phenotype , Point Mutation , Polymorphism, Restriction Fragment Length , alpha-Thalassemia/genetics , Female , Fetal Hemoglobin/analysis , Genetic Linkage , Humans , Male , Multigene Family/genetics , Quantitative Trait Loci/genetics , alpha-Thalassemia/blood
11.
Prenat Diagn ; 24(12): 992-6, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15614841

ABSTRACT

OBJECTIVE: To control the birth of thalassemic children in India. METHODS: Mutations present in the population of eastern India and in carrier parents seeking prenatal diagnosis were detected by the PCR-based technique of ARMS (amplification refractory mutation system) or gap-PCR. To screen for maternal tissue contamination in CVS, haplotypes associated with the beta-globin gene clusters were constructed using six polymorphic restriction sites. Prenatal diagnosis was accomplished by checking presence of parental mutation in the DNA from chorionic villus sampling (CVS) collected at 8 to 10 weeks' gestation by appropriate technique. RESULTS: Six hundred and fifty (650) unrelated beta-thalassemia chromosomes were screened for 11 common mutations to characterize the mutation distribution in this population. Starting from early 2000, 63 families from different parts of West Bengal and from surrounding areas have been offered prenatal counseling for beta-thalassemia. CONCLUSION: The population of this region is conscious and willing to accept prenatal diagnosis as a means of control of thalassemia.


Subject(s)
Prenatal Diagnosis , beta-Thalassemia/diagnosis , beta-Thalassemia/genetics , Chorionic Villi Sampling , DNA/analysis , False Negative Reactions , False Positive Reactions , Female , Genetic Counseling , Gestational Age , Globins/genetics , Haplotypes , Humans , India , Male , Mutation , Polymerase Chain Reaction , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...