Thymic selection involves conversion of a continuous antigen receptor signal to a discrete set of developmental outcomes: death, positive selection or negative selection. This highly regulated developmental process plays a critical role in the selection of a population of T cells carrying a variety of functional T cell receptors (positive selection), while eliminating autoreactive T cells (negative selection). The class II histone deacetylase HDAC7 is expressed at high levels in the nucleus of double positive thymocytes. HDAC7 is exported from the nucleus to the cytoplasm in response to T cell receptor activation and allows changes in gene expression associated with positive selection. Loss of stageappropriate gene expression in HDAC7-deficient DP thymocytes shortens their survival in vivo and impairs maturation to the single positive stage. Transgenic expression of an HDAC7 mutant that cannot be exported from the nucleus increases single positive thymocyte numbers and renders thymocytes resistant to negative selection. Mice expressing this HDAC7 mutant protein in the thymus develop a striking autoimmune exocrine pancreatitis. Finally, both knockout and transgenic mice show distinct and abnormal distributions of CD4 effector T cells. These observations support the model that HDAC7 participates in the transcriptional state changes associated with both positive and negative selection and the subsequent differentiation of CD4 na?ve T cells in effector T cells. We propose: 1. To map the transcriptional cascade downstream of HDAC7 in thymocytes. We will use chIP-seq to identify the direct primary targets of binding of HDAC7 in the genome. This gene list will be compared with the genes that are transcriptionally activated when HDAC7 is knocked out in vivo. This analysis will be followed by pathway analysis both in silico and in the laboratory to establish the connectivity of the primary and secondary targets of HDAC7. 2. To characterize the autoimmune phenotype of transgenic mice expressing a dominant positive HDAC7 mutant protein (HDAC7-��P). We have observed a highly penetrant autoimmune exocrine pancreatitis syndrome in transgenic mice expressing a mutant HDAC7 protein (HADC7-��P) that is not exported from the nucleus in response to TCR signaling. HADC7-��P may allow autoreactive T cells to persist by suppressing the death of cells that normally undergo negative selection. However, a disturbance in the balance of effector and regulatory T cell types in the periphery is another possible mechanism. We propose to further characterize this phenotype and in particular to define the contribution of HDAC7 to the induction of central vs. peripheral tolerance. 3. To examine the physiological function of HDAC7 in murine models of T cell differentiation. After exiting the thymus, Na?ve CD4+ T cells can undergo further differentiation in the periphery into distinct effector T cell subsets, depending on TCR activation as well as signaling through specific cytokine receptors. Perturbing the balance among these effector CD4+ T cells can lead to inflammation and/or autoimmunity, by inappropriate expansion of pro-inflammatory T cells and by insufficient production and expansion of regulatory T cells. We have obtained several lines of evidence supporting the hypothesis that HDAC7 also plays an important role in the regulation of effector T cell differentiation in the periphery in addition to its specific effects in the thymus. We propose to functionally characterize the role of HDAC7 in effector T cell differentiation in mice, to determine the effect of perturbation of HDAC7 function on changes in gene expression during effector T cell differentiation and to examine the role of HDAC7- mediated effector differentiation in murine disease models.

Public Health Relevance

The vertebrate adaptive immune system must strike a delicate balance between generating lymphocytes that can mount vigorous responses against foreign pathogens and insuring that the same lymphocytes do not attack the self (self-tolerance). Breakdown of these mechanisms can manifest in autoimmune diseases. T cell self-tolerance is established in part during T cell development in the thymus (central tolerance) by the elimination of self-reactive T cells. We have identified HDAC7, a histone deacetylase that represses gene expression, as a critical molecular link that regulates survival of thymocytes during their development. We have also generated a new mouse expressing a mutated HDAC7 protein. This mouse presents with symptoms of autoimmunity. Here, we propose to further test our hypothesis that HDAC7 is a critical regulator of central T cell tolerance. We will identify the signals that regulate the activity of HDAC7 and its association with target genes. We will map the transcriptional cascade downstream of HDAC7 in thymocytes. We will characterize the physiologic function of HDAC7 in murine models of T cell development and in immune function using mice that lack HDAC7 in the thymus and using transgenic mice in which various mutant versions of HDAC7 are expressed in the thymus. We anticipate that these experiments will further our understanding of the mechanism of immune tolerance and the development of autoimmunity.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
High Priority, Short Term Project Award (R56)
Project #
1R56AI076664-01A2
Application #
8023197
Study Section
Cellular and Molecular Immunology - B Study Section (CMIB)
Program Officer
Prabhudas, Mercy R
Project Start
2010-03-01
Project End
2012-02-28
Budget Start
2010-03-01
Budget End
2012-02-28
Support Year
1
Fiscal Year
2010
Total Cost
$482,500
Indirect Cost
Name
J. David Gladstone Institutes
Department
Type
DUNS #
099992430
City
San Francisco
State
CA
Country
United States
Zip Code
94158
Calvanese, Vincenzo; Chavez, Leonard; Laurent, Timothy et al. (2013) Dual-color HIV reporters trace a population of latently infected cells and enable their purification. Virology 446:283-92
Kasler, Herbert G; Young, Bryan D; Mottet, Denis et al. (2011) Histone deacetylase 7 regulates cell survival and TCR signaling in CD4/CD8 double-positive thymocytes. J Immunol 186:4782-93