Abstract

A majority of breast cancers (BC) are age-related and we seek to determine what cellular and molecular changes occur in breast tissue with age that make women more susceptible to cancer initiation. Immune-epithelial cell interactions are important during mammary gland development and the immune system plays an important role in BC progression. The composition of human immune cell populations is known to change in peripheral blood with age and in breast tissue during BC progression. Less is known about changes in immune populations in normal breast tissue and how their interactions with mammary epithelia change with age. We quantified densities of T cells, B cells, and macrophage subsets in pathologically normal breast tissue from 122 different women who ranged in age from 24 to 74 years old. Donor-matched peripheral blood from a subset of 20 donors was analyzed by flow cytometry. Tissue immune cell densities and localizations relative to the epithelium were quantified in situ with machine learning-based image analyses of multiplex immunohistochemistry-stained tissue sections. In situ results were corroborated with flow cytometry analyses of peri-epithelial immune cells from primary breast tissue preparations and transcriptome analyses of public data from bulk tissue reduction mammoplasties. Proportions of immune cell subsets in breast tissue and donor-matched peripheral blood were not correlated. Density (cells/mm2) of T and B lymphocytes in situ decreased with age. T cells and macrophages preferentially localized near or within epithelial bilayers, rather than the intralobular stroma. M2 macrophage density was higher than M1 macrophage density and this difference was due to higher density of M2 in the intralobular stroma. Transcriptional signature analyses suggested age-dependent decline in adaptive immune cell populations and functions and increased innate immune cell activity. T cells and macrophages are so intimately associated with the epithelia that they are embedded within the bilayer, suggesting an important role for immune-epithelial cell interactions. Age-associated decreased T cell density in peri-epithelial regions, and increased M2 macrophage density in intralobular stroma suggests the emergence of a tissue microenvironment that is simultaneously immune-senescent and immunosuppressive with age.

Highlights

  • More than 75% of breast cancers (BC) are diagnosed in women after age 50y [1], the mechanisms underlying age-related BC susceptibility are not well understood

  • As described in the original article (NCBI GEO GSE102088 [24]), frozen breast tissue obtained from reduction mammoplasty (RM) of 121 healthy women underwent RNA extraction and was profiled via the GeneChip® Human Transcriptome Array 2.0 (Affymetrix Inc, Santa Clara, CA)

  • Aging is the greatest risk factor for BC, and we performed a survey of immune cell subsets in normal breast tissue as a function of age to better understand whether there were changes that could be related to breast cancer susceptibility

Read more

Summary

Introduction

More than 75% of breast cancers (BC) are diagnosed in women after age 50y [1], the mechanisms underlying age-related BC susceptibility are not well understood. The increased incidence is thought to be driven by a combination of accumulated somatic mutations, and changes in the breast tissue microenvironment that unleash malignant cells and increase susceptibility of the tissue to cancer initiation [2]. Age-dependent tissue composition changes in breast include decreased connective tissue, increased adipose cell proportions [3, 4], reduced proportions of myoepithelial cells, accumulation of luminal epithelial cells and dysfunctional progenitor cells with a basal differentiation bias [5,6,7]. Immune cells can impose potent phenotypic and compositional changes during mammary gland development [10,11,12,13]. Mouse models suggest that the innate and adaptive immune cell types exert opposing effects during mammary gland development. In addition to sculpting developmental processes, the immune system is infamous for its role in tumor progression [14], for example tumor-associated macrophages (TAMs) are polarized to an M2-like phenotype, giving them immunosuppressive functions and further facilitating tumor progression and cancer cell proliferation [15]

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call