Store-operated Ca2+ entry (SOCE) is the process whereby loss of Ca2+ in the endoplasmic reticulum (ER) leads to Ca2+ entry into cells across the plasma membrane (PM). STIM1 and STIM2 are critical ER Ca2+ sensors that control the activation of Orai1, the store-operated Ca2+ channel. STIM1/2 are critical for T cell receptor (TCR)- mediated signaling and their deficiency in humans results in lymphoproliferative disorder and autoimmunity sug- gesting defects in T cell development and activation. In addition to ER Ca2+ release and SOCE, Ca2+ signals involve Ca2+ extrusion mechanisms. In lymphocytes, Plasma Membrane Ca2+/ATPase 4 (PMCA4) is the primary mediator of Ca2+ extrusion and is essential for normal function. Recently, we identified PMCA4 as a further target of STIM1 (but not STIM2) during lymphocyte activation. Unlike SOCE, STIM1-induced PMCA inhibition is initi- ated by Early Growth Response 1 (EGR1) and EGR4-mediated STIM1 upregulation. EGRs are ubiquitous and rapidly upregulated zinc finger transcription factors. The current proposal is based on the following hypothesis: TCR engagement stimulates EGR-mediated STIM1 upregulation in nave T cells that initiates Ca2+ clearance inhibition, critical for the maintenance of NFAT/NF-?B activation and subsequent T cell activation. This work is organized into 3 aims.
Aim 1 is to characterize EGR1/4-mediated control of T cell diversity and function. While EGR1KO mice exhibit previously described defects in early T cell development, preliminary findings re- ported here are the first to reveal a T cell phenotype in EGR4KO mice. We have observed altered progression through the double-negative stages, dysregulation of Ca2+ clearance during T cell activation and defective T cell expansion. These investigations will be expanded, both to include EGR1/4dKO mice and to include analysis of developmental, conventional and non-conventional subtypes.
Aim 2 is to assess how STIM1?597 knockin af- fects T cell activation. As established in our recent study, STIM1 truncation at amino acid 597 (STIM1?597) eliminates PMCA association without affecting Orai1 activation13. Preliminary analysis of T cells obtained from a recently generated STIM1?597 knock-in mouse reveals intriguing T cell subset-specific changes in Ca re- 2+ sponses. Implications to T cell activation will be determined by measuring transcription factor activation, cytokine expression patterns and T cell diversity.
Aim 3 is to define the mechanisms whereby POST coordinates functional STIM1-PMCA4 interactions. We revealed that EGR-induced STIM1 upregulation causes decreased PMCA function within the immunological synapse during T cell activation. However, a third protein termed POST interacts with both proteins and regulates Ca2+ clearance during store depletion. Our preliminary findings provide new insight into how these interactions are coordinated during T cell activation and define their functional impli- cations. The dynamics of these interactions will be determined using mutational analysis, FRET and Ca2+ en- try/clearance assays. Collectively, these studies reveal a novel new concept for how Ca2+ signals contribute to events that occur beyond the seconds to minutes time periods encompassing a single Ca2+ transient.

Public Health Relevance

EGR-MEDIATED STIM1-PMCA EXPRESSION AND FUNCTION IN T CELL SUBSETS ? NARRATIVE STIM1-dependent Ca2+ signals represent the primary means of Ca2+ entry in T cells, with critical contributions to their activation. We have recently shown that, in addition to activating Ca2+ channels, STIM1 also inhibits the plasma membrane Ca2+ pump during T cell activation. The current proposal focuses on defining the molecular events that mediate this process and determining the impact of this new STIM1-mediated function on T cell- mediated immunity.

Agency
National Institute of Health (NIH)
Institute
National Institute of General Medical Sciences (NIGMS)
Type
Research Project (R01)
Project #
5R01GM117907-02
Application #
9229047
Study Section
Cellular and Molecular Immunology - B Study Section (CMIB)
Program Officer
Marino, Pamela
Project Start
2016-03-01
Project End
2019-12-31
Budget Start
2017-01-01
Budget End
2017-12-31
Support Year
2
Fiscal Year
2017
Total Cost
Indirect Cost
Name
Temple University
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
057123192
City
Philadelphia
State
PA
Country
United States
Zip Code
19122
Mo, Xuan; Zhang, Hanghang; Preston, Sarah et al. (2018) Interferon-? Signaling in Melanocytes and Melanoma Cells Regulates Expression of CTLA-4. Cancer Res 78:436-450
Duong-Ly, Krisna C; Kuo, Yin-Ming; Johnson, Matthew C et al. (2018) T cell activation triggers reversible inosine-5'-monophosphate dehydrogenase assembly. J Cell Sci 131:
Soboloff, Jonathan; Gligorijevic, Bojana; Zaidi, M Raza (2018) STIM1 (c)AMPs up melanogenesis. EMBO J 37:
Go, Christina K; Soboloff, Jonathan (2018) Hold the door: hPMCA1/neuroplastin interactions regulate Ca2+-binding site accessibility. Cell Calcium 76:135-136
He, Shan; Liu, Yongnian; Meng, Lijun et al. (2017) Ezh2 phosphorylation state determines its capacity to maintain CD8+ T memory precursors for antitumor immunity. Nat Commun 8:2125
Troupes, Constantine D; Wallner, Markus; Borghetti, Giulia et al. (2017) Role of STIM1 (Stromal Interaction Molecule 1) in Hypertrophy-Related Contractile Dysfunction. Circ Res 121:125-136
Dong, Zhiwei; Shanmughapriya, Santhanam; Tomar, Dhanendra et al. (2017) Mitochondrial Ca2+Uniporter Is a Mitochondrial Luminal Redox Sensor that Augments MCU Channel Activity. Mol Cell 65:1014-1028.e7
Hooper, Robert; Zaidi, M Raza; Soboloff, Jonathan (2016) The heterogeneity of store-operated calcium entry in melanoma. Sci China Life Sci 59:764-9
Samakai, Elsie; Hooper, Robert; Martin, Kayla A et al. (2016) Novel STIM1-dependent control of Ca2+ clearance regulates NFAT activity during T-cell activation. FASEB J 30:3878-3886
Hooper, Robert; Rothberg, Brad S; Soboloff, Jonathan (2016) Sterol hindrance of Orai activation. Sci Signal 9:fs4