PMCA2 is a calcium pump expressed in the apical membrane of mammary epithelial cells (MECs) during lactation. It is responsible for transporting 60-70% of milk calcium. After weaning, milk stasis rapidly decreases PMCA2 expression, causing a sustained increase in intracellular calcium. Preliminary data suggest that this rise in intracellular calcium may serve as the biochemical signal that couples milk stasis to activation of lysosome biogenesis, STAT3 phosphorylation and lysosome-dependent cell death (LDCD). In the last cycle of this grant, we documented that PMCA2 is also re-expressed in breast cancers and correlates with ErbB2/HER2 levels and increased mortality. We have now defined interactions between PMCA2, NHERF1, Ezrin, HSP90 and ErbB2 that are required for membrane retention of ErbB2 and its downstream biochemical signaling. Inhibition of PMCA2 increases intracellular calcium and causes a PKCa-dependent disassembly of this multiprotein complex as well as the ubiquitinylation, internalization and degradation of ErbB2. In normal MECs, NHERF1, ezrin and PMCA2 interact within the apical plasma membrane while ErbB2 interacts with a different scaffolding protein called Erbin, at the basolateral membrane. Preliminary data show that, in DCIS lesions and in invasive breast cancer cells, PMCA2, Ezrin and NHERF1 intermix with Erbin and ErbB2 throughout the plasma membrane and that this complex is greatly upregulated in trastuzumab-resistant breast cancer cells. The premise/hypothesis of this grant application is three-fold: 1) that milk stasis rapidly decreases PMCA2 expression causing sustained increases in intracellular calcium, which, in turn, trigger LDCD; 2) that loss of cell polarity enables novel interactions between the apical, PMCA2-NHERF1-Ezrin complex and the basolateral, Erbin-ErbB2 complex that accelerate ErbB2-mediated transformation; and 3) that upregulation of interactions between PMCA2 and ErbB2 contribute to the development of trastuzumab resistance. We propose 3 specific aims.
Aim 1 will examine the mechanisms whereby increased intracellular calcium concentrations activate lysosome biogenesis, STAT3 and LDCD.
Aim 2 will test whether alterations in PMCA2 localization accelerate malignant transformation by ErbB2/HER2.
Aim 3 will test whether further upregulation of interactions between PMCA2, NHERF1, Ezrin, Erbin, HSP90 and ErbB2 contributes to trastuzumab resistance. These experiments will provide important new knowledge of the mechanisms by which milk stasis triggers involution. They will contribute important insight into the early steps of transformation of MECs by ErbB2. They will validate novel cancer cell-specific drug targets for ErbB2/HER2-positive tumors and will test the potential therapeutic value of disrupting these interactions in trastuzumab-resistant cancers.

Public Health Relevance

The plasma membrane calcium ATPase 2 (PMCA2) is a calcium pump found on the apical membrane of mammary epithelial cells that transports 60%-70% of milk calcium from within these cells into the alveolar lumen. We have found that PMCA2 expression is rapidly downregulated by milk stasis after weaning and that it is re-expressed in ErbB2/HER2-positive breast cancers. This renewal application seeks to continue our studies of PMCA2 during mammary gland involution and in breast cancer by: 1) examining how the decline of PMCA2 and resulting increase of intracellular calcium triggers new lysosome formation, STAT3 activation and cell death during early involution; and 2) examining how interactions between PMCA2, Erbin and NHERF1 form as a result of disturbed cell polarity to promote ErbB2 signaling, malignant transformation and resistance to trastuzumab.

Agency
National Institute of Health (NIH)
Institute
Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD)
Type
Research Project (R01)
Project #
2R01HD076248-06A1
Application #
10120306
Study Section
Integrative and Clinical Endocrinology and Reproduction Study Section (ICER)
Program Officer
Bremer, Andrew
Project Start
2014-04-10
Project End
2025-06-30
Budget Start
2020-09-18
Budget End
2021-06-30
Support Year
6
Fiscal Year
2020
Total Cost
Indirect Cost
Name
Yale University
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
043207562
City
New Haven
State
CT
Country
United States
Zip Code
06520
Zwick, Rachel K; Rudolph, Michael C; Shook, Brett A et al. (2018) Adipocyte hypertrophy and lipid dynamics underlie mammary gland remodeling after lactation. Nat Commun 9:3592
Jeong, Jaekwang; VanHouten, Joshua N; Kim, Wonnam et al. (2017) The scaffolding protein NHERF1 regulates the stability and activity of the tyrosine kinase HER2. J Biol Chem 292:6555-6568
Kim, Wonnam; Wysolmerski, John J (2016) Calcium-Sensing Receptor in Breast Physiology and Cancer. Front Physiol 7:440
Jeong, Jaekwang; VanHouten, Joshua N; Dann, Pamela et al. (2016) PMCA2 regulates HER2 protein kinase localization and signaling and promotes HER2-mediated breast cancer. Proc Natl Acad Sci U S A 113:E282-90
Kim, Wonnam; Takyar, Farzin M; Swan, Karena et al. (2016) Calcium-Sensing Receptor Promotes Breast Cancer by Stimulating Intracrine Actions of Parathyroid Hormone-Related Protein. Cancer Res 76:5348-60
Ardeshirpour, Laleh; Dumitru, Cristina; Dann, Pamela et al. (2015) OPG Treatment Prevents Bone Loss During Lactation But Does Not Affect Milk Production or Maternal Calcium Metabolism. Endocrinology 156:2762-73
Boras-Granic, Kata; Dann, Pamela; Wysolmerski, John J (2014) Embryonic cells contribute directly to the quiescent stem cell population in the adult mouse mammary gland. Breast Cancer Res 16:487