Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Front Immunol ; 12: 703632, 2021.
Article in English | MEDLINE | ID: mdl-34290714

ABSTRACT

The Mads/Mef2 (Mef2a/b/c/d) family of transcription factors (TFs) regulates differentiation of muscle cells, neurons and hematopoietic cells. By functioning in physiological feedback loops, Mef2 TFs promote the transcription of their repressor, Hdac9, thereby providing temporal control of Mef2-driven differentiation. Disruption of this feedback is associated with the development of various pathologic states, including cancer. Beside their direct involvement in oncogenesis, Mef2 TFs indirectly control tumor progression by regulating antitumor immunity. We recently reported that in CD4+CD25+Foxp3+ T-regulatory (Treg) cells, Mef2d is required for the acquisition of an effector Treg (eTreg) phenotype and for the activation of an epigenetic program that suppresses the anti-tumor immune responses of conventional T and B cells. We now report that as with Mef2d, the deletion of Mef2c in Tregs switches off the expression of Il10 and Icos and leads to enhanced antitumor immunity in syngeneic models of lung cancer. Mechanistically, Mef2c does not directly bind the regulatory elements of Icos and Il10, but its loss-of-function in Tregs induces the expression of the transcriptional repressor, Hdac9. As a consequence, Mef2d, the more abundant member of the Mef2 family, is converted by Hdac9 into a transcriptional repressor on these loci. This leads to the impairment of Treg suppressive properties in vivo and to enhanced anti-cancer immunity. These data further highlight the central role played by the Mef2/Hdac9 axis in the regulation of CD4+Foxp3+ Treg function and adds a new level of complexity to the analysis and study of Treg biology.


Subject(s)
Histone Deacetylases/immunology , Immune Tolerance , Lung Neoplasms/immunology , Neoplasms, Experimental/immunology , Repressor Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Histone Deacetylases/genetics , Immunity, Cellular , Lung Neoplasms/genetics , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Neoplasms, Experimental/genetics , Repressor Proteins/genetics
2.
J Clin Invest ; 130(12): 6242-6260, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32790649

ABSTRACT

The transcription factor MEF2D is important in the regulation of differentiation and adaptive responses in many cell types. We found that among T cells, MEF2D gained new functions in Foxp3+ T regulatory (Treg) cells due to its interactions with the transcription factor Foxp3 and its release from canonical partners, like histone/protein deacetylases. Though not necessary for the generation and maintenance of Tregs, MEF2D was required for the expression of IL-10, CTLA4, and Icos, and for the acquisition of an effector Treg phenotype. At these loci, MEF2D acted both synergistically and additively to Foxp3, and downstream of Blimp1. Mice with the conditional deletion in Tregs of the gene encoding MEF2D were unable to maintain long-term allograft survival despite costimulation blockade, had enhanced antitumor immunity in syngeneic models, but displayed only minor evidence of autoimmunity when maintained under normal conditions. The role played by MEF2D in sustaining effector Foxp3+ Treg functions without abrogating their basal actions suggests its suitability for drug discovery efforts in cancer therapy.


Subject(s)
Graft Survival/immunology , Heart Transplantation , Lymphocyte Activation , Neoplasms, Experimental/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Graft Survival/genetics , HEK293 Cells , Humans , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Neoplasms, Experimental/genetics , T-Lymphocytes, Regulatory/pathology , Transplantation, Isogeneic
3.
J Clin Invest ; 130(4): 1830-1842, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31917688

ABSTRACT

Foxp3+ Tregs are key to immune homeostasis, but the contributions of various large, multiprotein complexes that regulate gene expression remain unexplored. We analyzed the role in Tregs of the evolutionarily conserved CoREST complex, consisting of a scaffolding protein, Rcor1 or Rcor2, plus Hdac1 or Hdac2 and Lsd1 enzymes. Rcor1, Rcor2, and Lsd1 were physically associated with Foxp3, and mice with conditional deletion of Rcor1 in Foxp3+ Tregs had decreased proportions of Tregs in peripheral lymphoid tissues and increased Treg expression of IL-2 and IFN-γ compared with what was found in WT cells. Mice with conditional deletion of the gene encoding Rcor1 in their Tregs had reduced suppression of homeostatic proliferation, inability to maintain long-term allograft survival despite costimulation blockade, and enhanced antitumor immunity in syngeneic models. Comparable findings were seen in WT mice treated with CoREST complex bivalent inhibitors, which also altered the phenotype of human Tregs and impaired their suppressive function. Our data point to the potential for therapeutic modulation of Treg functions by pharmacologic targeting of enzymatic components of the CoREST complex and contribute to an understanding of the biochemical and molecular mechanisms by which Foxp3 represses large gene sets and maintains the unique properties of this key immune cell.


Subject(s)
Co-Repressor Proteins/immunology , Immunity, Cellular , Multiprotein Complexes/immunology , Neoplasms, Experimental/immunology , Nerve Tissue Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Line, Tumor , Co-Repressor Proteins/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Multiprotein Complexes/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Nerve Tissue Proteins/genetics , T-Lymphocytes, Regulatory/pathology
4.
Nucleic Acids Res ; 48(2): 646-664, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31754707

ABSTRACT

Transcriptional networks supervising class IIa HDAC expression are poorly defined. Here we demonstrate that MEF2D is the key factor controlling HDAC9 transcription. This control, which is part of a negative feed-back loop during muscle differentiation, is hijacked in cancer. In leiomyosarcomas the MEF2D/HDAC9 vicious circuit sustains proliferation and cell survival, through the repression of the death receptor FAS. Comprehensive genome-wide studies demonstrate that HDAC4 and HDAC9 control different genetic programs and show both specific and common genomic binding sites. Although the number of MEF2-target genes commonly regulated is similar, only HDAC4 represses many additional genes that are not MEF2D targets. As expected, HDAC4-/- and HDAC9-/- cells increase H3K27ac levels around the TSS of the respective repressed genes. However, these genes rarely show binding of the HDACs at their promoters. Frequently HDAC4 and HDAC9 bind intergenic regions. We demonstrate that these regions, recognized by MEF2D/HDAC4/HDAC9 repressive complexes, show the features of active enhancers. In these regions HDAC4 and HDAC9 can differentially influence H3K27 acetylation. Our studies describe new layers of class IIa HDACs regulation, including a dominant positional effect, and can contribute to explain the pleiotropic actions of MEF2 TFs.


Subject(s)
Histone Deacetylases/genetics , Leiomyosarcoma/genetics , Repressor Proteins/genetics , Acetylation , Cell Differentiation/genetics , Cell Line, Tumor , Cell Survival/genetics , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Leiomyosarcoma/pathology , MEF2 Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...