Skip to main content

Advertisement

Log in

Parity reduces mammary repopulating activity but does not affect mammary stem cells defined as CD24 + CD29/CD49fhi in mice

  • Preclinical study
  • Published:
Breast Cancer Research and Treatment Aims and scope Submit manuscript

Abstract

Background

Breast cancer (BCa) mortality is decreasing with early detection and improvement in therapies. The incidence of BCa, however, continues to increase, particularly estrogen-receptor-positive (ER +) subtypes. One of the greatest modifiers of ER + BCa risk is childbearing (parity), with BCa risk halved in young multiparous mothers. Despite convincing epidemiological data, the biology that underpins this protection remains unclear. Parity-induced protection has been postulated to be due to a decrease in mammary stem cells (MaSCs); however, reports to date have provided conflicting data.

Methods

We have completed rigorous functional testing of repopulating activity in parous mice using unfractionated and MaSC (CD24midCD49fhi)-enriched populations. We also developed a novel serial transplant method to enable us to assess self-renewal of MaSC following pregnancy. Lastly, as each pregnancy confers additional BCa protection, we subjected mice to multiple rounds of pregnancy to assess whether additional pregnancies impact MaSC activity.

Results

Here, we report that while repopulating activity in the mammary gland is reduced by parity in the unfractionated gland, it is not due to a loss in the classically defined MaSC (CD24+CD49fhi) numbers or function. Self-renewal was unaffected by parity and additional rounds of pregnancy also did not lead to a decrease in MaSC activity.

Conclusions

Our data show instead that parity impacts on the stem-like activity of cells outside the MaSC population.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

Data availability

All data generated or analysed during this study are included in this published article and its supplementary information files.

References

  1. Bigaard J et al (2012) Breast cancer incidence by estrogen receptor status in Denmark from 1996 to 2007. Breast Cancer Res Treat 136(2):559–564

    CAS  PubMed  Google Scholar 

  2. Glass AG, Hoover RN (1990) Rising incidence of breast cancer: relationship to stage and receptor status. J Natl Cancer Inst 82(8):693–696

    CAS  PubMed  Google Scholar 

  3. Li CI, Daling JR, Malone KE (2003) Incidence of invasive breast cancer by hormone receptor status from 1992 to 1998. J Clin Oncol 21(1):28–34

    CAS  PubMed  Google Scholar 

  4. Rosenberg PS, Barker KA, Anderson WF (2015) Estrogen Receptor status and the future burden of invasive and in situ breast cancers in the United States. J Natl Cancer Inst 107(9):djv159

    PubMed  PubMed Central  Google Scholar 

  5. Howalder N et al (2016) SEER cancer statistics review, 1975–2013. National Cancer Institute, Bethesda, MD

    Google Scholar 

  6. Youlden DR et al (2014) Incidence and mortality of female breast cancer in the Asia-Pacific region. Cancer Biol Med 11(2):101–115

    PubMed  PubMed Central  Google Scholar 

  7. AIHW (2012) Breast Cancer in Australia: an overview in Cancer Series no 71 Cat no CAN67. Australian Institute of Health and Welfare and National Breast and Ovarian Cancer Centre, Canberra

    Google Scholar 

  8. Dall GV, Britt KL (2017) Estrogen effects on the mammary gland in early and late life and breast cancer risk. Front Oncol 7:110

    PubMed  PubMed Central  Google Scholar 

  9. Collaborative Group on Hormonal Factors in Breast, C (2012) Menarche, menopause, and breast cancer risk: individual participant meta-analysis, including 118 964 women with breast cancer from 117 epidemiological studies. Lancet Oncol 13(11):1141–1151

    Google Scholar 

  10. Collaborative Group on Hormonal Factors in Breast, C (2002) Breast cancer and breastfeeding: collaborative reanalysis of individual data from 47 epidemiological studies in 30 countries. Lancet 360:187–195

    Google Scholar 

  11. MacMahon B et al (1970) Age at first birth and breast cancer risk. Bull World Health Organ 43(2):209–221

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Ewertz M et al (1990) Age at first birth, parity and risk of breast cancer: a meta-analysis of 8 studies from the Nordic countries. Int J Cancer 46(4):597–603

    CAS  PubMed  Google Scholar 

  13. Britt K, Ashworth A, Smalley M (2007) Pregnancy and the risk of breast cancer. Endocr Relat Cancer 14(4):907–933

    CAS  PubMed  Google Scholar 

  14. Chang CC et al (2001) A human breast epithelial cell type with stem cell characteristics as target cells for carcinogenesis. Radiat Res 155(1 Pt 2):201–207

    CAS  PubMed  Google Scholar 

  15. Russo J, Tay LK, Russo IH (1982) Differentiation of the mammary gland and susceptibility to carcinogenesis. Breast Cancer Res Treat 2(1):5–73

    CAS  PubMed  Google Scholar 

  16. Russo J, Russo IH (1978) DNA labeling index and structure of the rat mammary gland as determinants of its susceptibility to carcinogenesis. J Natl Cancer Inst 61(6):1451–1459

    CAS  PubMed  Google Scholar 

  17. Tokunaga M et al (1994) Incidence of female breast cancer among atomic bomb survivors, 1950–1985. Radiat Res 138(2):209–223

    CAS  PubMed  Google Scholar 

  18. Land CE et al (2003) Incidence of female breast cancer among atomic bomb survivors, Hiroshima and Nagasaki, 1950–1990. Radiat Res 160(6):707–717

    CAS  PubMed  Google Scholar 

  19. Siwko SK et al (2008) Evidence that an early pregnancy causes a persistent decrease in the number of functional mammary epithelial stem cells–implications for pregnancy-induced protection against breast cancer. Stem Cells 26(12):3205–3209

    PubMed  PubMed Central  Google Scholar 

  20. Meier-Abt F et al (2013) Parity induces differentiation and reduces Wnt/Notch signaling ratio and proliferation potential of basal stem/progenitor cells isolated from mouse mammary epithelium. Breast Cancer Res 15(2):R36

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Britt KL et al (2009) Pregnancy in the mature adult mouse does not alter the proportion of mammary epithelial stem/progenitor cells. Breast Cancer Res 11(2):R20

    PubMed  PubMed Central  Google Scholar 

  22. Vaillant F, Lindeman GJ, Visvader JE (2011) Jekyll or Hyde: does Matrigel provide a more or less physiological environment in mammary repopulating assays? Breast Cancer Res 13(3):108

    PubMed  PubMed Central  Google Scholar 

  23. Gupta PB et al (2007) Systemic stromal effects of estrogen promote the growth of estrogen receptor-negative cancers. Cancer Res 67(5):2062–2071

    CAS  PubMed  Google Scholar 

  24. Schedin P (2006) Pregnancy-associated breast cancer and metastasis. Nat Rev Cancer 6(4):281–291

    CAS  PubMed  Google Scholar 

  25. Tikoo A et al (2012) Physiological levels of Pik3ca(H1047R) mutation in the mouse mammary gland results in ductal hyperplasia and formation of ERalpha-positive tumors. PLoS ONE 7(5):e36924

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Flurkey, C.a.H., The mouse in Biomedical Research, ed. J. Fox. 2007: American College of Laboratory Animal Medicine (Elsevier).

  27. Lilla JN et al (2009) Active plasma kallikrein localizes to mast cells and regulates epithelial cell apoptosis, adipocyte differentiation, and stromal remodeling during mammary gland involution. J Biol Chem 284(20):13792–13803

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Sleeman KE et al (2006) CD24 staining of mouse mammary gland cells defines luminal epithelial, myoepithelial/basal and non-epithelial cells. Breast Cancer Res 8(1):R7

    PubMed  Google Scholar 

  29. Sleeman KE et al (2007) Dissociation of estrogen receptor expression and in vivo stem cell activity in the mammary gland. J Cell Biol 176(1):19–26

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Dall GV et al (2017) SCA-1 Labels a subset of estrogen-responsive bipotential repopulating cells within the CD24+ CD49fhi mammary stem cell-enriched compartment. Stem Cell Rep 8(2):417–431

    CAS  Google Scholar 

  31. Hu Y, Smyth GK (2009) ELDA: extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays. J Immunol Methods 347(1–2):70–78

    CAS  PubMed  Google Scholar 

  32. Dall G, Risbridger G, Britt K (2016) Mammary stem cells and parity-induced breast cancer protection- new insights. J Steroid Biochem Mol Biol 170:54–60

    PubMed  Google Scholar 

  33. Shackleton M et al (2006) Generation of a functional mammary gland from a single stem cell. Nature 439(7072):84–88

    CAS  PubMed  Google Scholar 

  34. Stingl J et al (2006) Purification and unique properties of mammary epithelial stem cells. Nature 439(7079):993–997

    CAS  PubMed  Google Scholar 

  35. Prater MD et al (2014) Mammary stem cells have myoepithelial cell properties. Nat Cell Biol 16(10):942–950

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Tamakoshi K et al (2005) Impact of menstrual and reproductive factors on breast cancer risk in Japan: results of the JACC study. Cancer Sci 96(1):57–62

    CAS  PubMed  Google Scholar 

  37. Suh JS et al (1996) Menstrual and reproductive factors related to the risk of breast cancer in Korea. Ovarian hormone effect on breast cancer. J Korean Med Sci 11(6):501–508

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Kato I et al (1992) A case-control study of breast cancer among Japanese women: with special reference to family history and reproductive and dietary factors. Breast Cancer Res Treat 24(1):51–59

    CAS  PubMed  Google Scholar 

  39. Hirose K et al (1995) A large-scale, hospital-based case-control study of risk factors of breast cancer according to menopausal status. Jpn J Cancer Res 86(2):146–154

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Russo J, Wilgus G, Russo IH (1979) Susceptibility of the mammary gland to carcinogenesis: I Differentiation of the mammary gland as determinant of tumor incidence and type of lesion. Am J Pathol 96(3):721–736

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Russo IH, Russo J (1978) Developmental stage of the rat mammary gland as determinant of its susceptibility to 7,12-dimethylbenz[a]anthracene. J Natl Cancer Inst 61(6):1439–1449

    CAS  PubMed  Google Scholar 

  42. Boice JD Jr et al (1991) Frequent chest X-ray fluoroscopy and breast cancer incidence among tuberculosis patients in Massachusetts. Radiat Res 125(2):214–222

    PubMed  Google Scholar 

  43. Cohn BA et al (2007) DDT and breast cancer in young women: new data on the significance of age at exposure. Environ Health Perspect 115(10):1406–1414

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Hoffman DA et al (1989) Breast cancer in women with scoliosis exposed to multiple diagnostic x rays. J Natl Cancer Inst 81(17):1307–1312

    CAS  PubMed  Google Scholar 

  45. McGregor H et al (1977) Breast cancer incidence among atomic bomb survivors, Hiroshima and Nagasaki, 1950–69. J Natl Cancer Inst 59(3):799–811

    CAS  PubMed  Google Scholar 

  46. Albrektsen G et al (2005) Breast cancer risk by age at birth, time since birth and time intervals between births: exploring interaction effects. Br J Cancer 92(1):167–175

    CAS  PubMed  Google Scholar 

  47. Press DJ, Pharoah P (2010) Risk factors for breast cancer: a reanalysis of two case-control studies from 1926 and 1931. Epidemiology 21(4):566–572

    PubMed  Google Scholar 

  48. Ursin G et al (2005) Reproductive factors and subtypes of breast cancer defined by hormone receptor and histology. Br J Cancer 93(3):364–371

    CAS  PubMed  PubMed Central  Google Scholar 

  49. van Amerongen R, Bowman AN, Nusse R (2012) Developmental stage and time dictate the fate of Wnt/beta-catenin-responsive stem cells in the mammary gland. Cell Stem Cell 11(3):387–400

    PubMed  Google Scholar 

  50. Rios AC et al (2014) In situ identification of bipotent stem cells in the mammary gland. Nature 506(7488):322–327

    CAS  PubMed  Google Scholar 

  51. Woodward WA et al (2007) WNT/beta-catenin mediates radiation resistance of mouse mammary progenitor cells. Proc Natl Acad Sci U S A 104(2):618–623

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Insinga A et al (2013) DNA damage in stem cells activates p21, inhibits p53, and induces symmetric self-renewing divisions. Proc Natl Acad Sci U S A 110(10):3931–3936

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Huh SJ et al (2015) Age- and pregnancy-associated DNA methylation changes in mammary epithelial cells. Stem Cell Rep 4(2):297–311

    CAS  Google Scholar 

  54. Plaks V et al (2013) Lgr5-expressing cells are sufficient and necessary for postnatal mammary gland organogenesis. Cell Rep 3(1):70–78

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Lim E et al (2010) Transcriptome analyses of mouse and human mammary cell subpopulations reveal multiple conserved genes and pathways. Breast Cancer Res 12(2):R21

    PubMed  PubMed Central  Google Scholar 

  56. Shehata M et al (2012) Phenotypic and functional characterisation of the luminal cell hierarchy of the mammary gland. Breast Cancer Res 14(5):R134

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This research was supported by an Australian Postgraduate Award scholarship for GD, Pfizer Australia and Victorian Cancer Agency (VCA) Clinical Research Fellowships for MS, an NBCF Early Career fellowship, NHMRC New Investigator grant and VCA Early Career fellowship for KB, and the Australian Cancer Research Foundation (for the Peter Mac Flow Cytometry and Centre for Advanced Histology and Microscopy core facilities). We thank the following Peter MacCallum Cancer Centre core facilities for provision of instrumentation, training and general support: Flow Cytometry, Centre for Advanced Histology and Microscopy and Animal House core facilities.

Author information

Authors and Affiliations

Authors

Contributions

GD: Concept and design, collection and/or assembly of data, data analysis and interpretation, manuscript writing, final approval of manuscript. JV: Collection and/or assembly of data, Data analysis and interpretation, final approval of manuscript. YS-R: Collection and/or assembly of data, Data analysis and interpretation, final approval of manuscript. NG: Collection and/or assembly of data, final approval of manuscript. ML-M: Collection and/or assembly of data, Data analysis and interpretation, final approval of manuscript. SR: Financial support, data analysis and interpretation, manuscript writing, final approval of manuscript. AA: Data analysis and interpretation, final approval of manuscript. RA: Financial support, data analysis and interpretation, manuscript writing, final approval of manuscript. GR: Financial support, data analysis and interpretation, manuscript writing, final approval of manuscript. MS: Concept and design, data analysis and interpretation, manuscript writing, final approval of manuscript. KB: Concept and design, financial support, collection and/or assembly of data, data analysis and interpretation, manuscript writing, final approval of manuscript.

Corresponding author

Correspondence to Kara L. Britt.

Ethics declarations

Conflicts of interest

Alan Ashworth has the following disclosures: A.A. is a shareholder in Tango Therapeutics and consultant for AtlasMDX, Third Rock Ventures, Pfizer, ProLynx and Bluestar, a SAB Member of Genentech and Gladiator, and receives grant support from Sun Pharma and AstraZeneca. A.A. holds patents on the use of PARP inhibitors held jointly with AstraZeneca from which he has benefitted financially (and may do so in the future) through the ICR Rewards to Inventors Scheme.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

10549_2020_5804_MOESM1_ESM.tif

Supplementary file1 Figure S1. Mouse model of early parity. Related to Figure 1. (A) Schematic of the mouse model used to mimic early parity in women, which is associated with decreased breast cancer risk. Mice were mated when young (6 weeks old) and the mammary epithelial cells analysed and/or isolated 10 weeks after involution. (B) Mammary glands from age-matched nulliparous control mice (left) and from mice at day 4 involution (involution), day 21 involution (repair) and 10 weeks after involution (10wks post Inv) were stained for morphology evaluation (H&E), mast cell infiltration using toluidine blue, and Ki67 to assess proliferation. (TIF 13302 kb)

10549_2020_5804_MOESM2_ESM.tif

Supplementary file2 Figure S2. Gating strategy used to assess mammary cell populations via flow cytometry. Related to Figure 1 and 3 (A) Gating strategy used to identify morphologically normal (i), single (ii), viable (iii) lineage negative (iv) mammary cells. CD24 and CD49f are then used together to distinguish luminal, basal and CD49fhi MaSC-enriched cells as well as the stroma (v). B We stained cells with both CD29 (i) and CD49f (ii) (as well as the other lineage and epithelial markers) and used flowlogic to analyse the populations. When coloured as populations in the analysis using CD29 and CD24, we could backgate and assess where the CD24 and CD29 populations sit on the CD24 and CD49f plot. The luminal and basal populations isolated with CD29 and CD24 were similarly isolated with CD49f and CD24. Moreover the CD29hi population labels the same cells as the CD49f hi population. (TIF 25521 kb)

10549_2020_5804_MOESM3_ESM.tif

Supplementary file3 Figure S3 Outgrowth size does not affect results of transplant assay from uniparous mice. Related to Table 1. (A) Results of mammary fat pad transplant assay with unfractionated cells isolated from uniparous and nulliparous mice. (B) Results of mammary fat pad transplant assay with MaSCs isolated from uniparous and nulliparous mice. For both tables circles show proportions of filling of each engrafted fat pad with repopulated epithelium. ELDA (Hu and Smyth, 2009) was used to determine frequencies of repopulating cells (MRCs) in cell suspensions from which aliquots were drawn for injection. Results show stem cell enrichment when all outgrowths are considered, or when only extensive outgrowths filling more than 25% of the fat pad are considered. (TIF 26856 kb)

10549_2020_5804_MOESM4_ESM.tif

Supplementary file4 Figure S4 Outgrowth size does not affect results of transplant assay from multiparous mice. Related to Table 3. Results of mammary fat pad transplant assay with MaSCs isolated from multiparous and nulliparous mice. Circles show proportions of filling of each engrafted fat pad with repopulated epithelium. ELDA (Hu and Smyth, 2009) was used to determine frequencies of repopulating cells (MRCs) in cell suspensions from which aliquots were drawn for injection. Results show stem cell enrichment when all outgrowths are considered, or when only extensive outgrowths filling more than 25% of the fat pad are considered. (TIF 25523 kb)

10549_2020_5804_MOESM5_ESM.docx

Supplementary file5 Supplementary Table 1. Results from primary transplantation using CD49fhi vs CD49flo cells (DOCX 13 kb)

10549_2020_5804_MOESM6_ESM.docx

Supplementary file6 Supplementary Table 2. Results from primary transplantation of CD49flo cells of parous and nulliparous mice (DOCX 13 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dall, G.V., Vieusseux, J., Seyed-Razavi, Y. et al. Parity reduces mammary repopulating activity but does not affect mammary stem cells defined as CD24 + CD29/CD49fhi in mice. Breast Cancer Res Treat 183, 565–575 (2020). https://doi.org/10.1007/s10549-020-05804-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10549-020-05804-1

Keywords

Navigation