Skip to main content
Top
Published in: Breast Cancer Research 1/2021

Open Access 01-12-2021 | Metastasis | Research article

Transcriptome analysis of heterogeneity in mouse model of metastatic breast cancer

Authors: Anastasia A. Ionkina, Gabriela Balderrama-Gutierrez, Krystian J. Ibanez, Steve Huy D. Phan, Angelique N. Cortez, Ali Mortazavi, Jennifer A. Prescher

Published in: Breast Cancer Research | Issue 1/2021

Login to get access

Abstract

Background

Cancer metastasis is a complex process involving the spread of malignant cells from a primary tumor to distal organs. Understanding this cascade at a mechanistic level could provide critical new insights into the disease and potentially reveal new avenues for treatment. Transcriptome profiling of spontaneous cancer models is an attractive method to examine the dynamic changes accompanying tumor cell spread. However, such studies are complicated by the underlying heterogeneity of the cell types involved. The purpose of this study was to examine the transcriptomes of metastatic breast cancer cells using the well-established MMTV-PyMT mouse model.

Methods

Organ-derived metastatic cell lines were harvested from 10 female MMTV-PyMT mice. Cancer cells were isolated and sorted based on the expression of CD44low/EpCAMhigh or CD44high/EpCAMhigh surface markers. RNA from each cell line was extracted and sequenced using the NextSeq 500 Illumina platform. Tissue-specific genes were compared across the different metastatic and primary tumor samples. Reads were mapped to the mouse genome using STAR, and gene expression was quantified using RSEM. Single-cell RNA-seq (scRNA-seq) was performed on select samples using the ddSeq platform by BioRad and analyzed using Seurat v3.2.3. Monocle2 was used to infer pseudo-time progression.

Results

Comparison of RNA sequencing data across all cell populations produced distinct gene clusters. Differential gene expression patterns related to CD44 expression, organ tropism, and immunomodulatory signatures were observed. scRNA-seq identified expression profiles based on tissue-dependent niches and clonal heterogeneity. These cohorts of data were narrowed down to identify subsets of genes with high expression and known metastatic propensity. Dot plot analyses further revealed clusters expressing cancer stem cell and cancer dormancy markers. Changes in relevant genes were investigated across pseudo-time and tissue origin using Monocle2. These data revealed transcriptomes that may contribute to sub-clonal evolution and treatment evasion during cancer progression.

Conclusions

We performed a comprehensive transcriptome analysis of tumor heterogeneity and organ tropism during breast cancer metastasis. These data add to our understanding of metastatic progression and highlight targets for breast cancer treatment. These markers could also be used to image the impact of tumor heterogeneity on metastases.
Appendix
Available only for authorised users
Literature
4.
7.
go back to reference Cai Y, et al. Transcriptomic dynamics of breast cancer progression in the MMTV-PyMT mouse model. BMC Genom. 2017;18(1):185.CrossRef Cai Y, et al. Transcriptomic dynamics of breast cancer progression in the MMTV-PyMT mouse model. BMC Genom. 2017;18(1):185.CrossRef
8.
go back to reference Zheng W, et al. Lung mammary metastases but not primary tumors induce accumulation of atypical large platelets and their chemokine expression. Cell Rep. 2019;29(7):1747-1755e4.PubMedPubMedCentralCrossRef Zheng W, et al. Lung mammary metastases but not primary tumors induce accumulation of atypical large platelets and their chemokine expression. Cell Rep. 2019;29(7):1747-1755e4.PubMedPubMedCentralCrossRef
9.
go back to reference Olsen TK, Baryawno N. Introduction to single-cell RNA sequencing. Curr Protoc Mol Biol. 2018;122(1):57.CrossRef Olsen TK, Baryawno N. Introduction to single-cell RNA sequencing. Curr Protoc Mol Biol. 2018;122(1):57.CrossRef
10.
go back to reference Yeo S.K. et al. Single-cell RNA-sequencing reveals distinct patterns of cell state heterogeneity in mouse models of breast cancer. Elife 2020;9. Yeo S.K. et al. Single-cell RNA-sequencing reveals distinct patterns of cell state heterogeneity in mouse models of breast cancer. Elife 2020;9.
11.
go back to reference Li H, et al. Reference component analysis of single-cell transcriptomes elucidates cellular heterogeneity in human colorectal tumors. Nat Genet. 2017;49(5):708–18.PubMedCrossRef Li H, et al. Reference component analysis of single-cell transcriptomes elucidates cellular heterogeneity in human colorectal tumors. Nat Genet. 2017;49(5):708–18.PubMedCrossRef
12.
17.
go back to reference Ma J, et al. Characterization of mammary cancer stem cells in the MMTV-PyMT mouse model. Tumour Biol. 2012;33(6):1983–96.PubMedCrossRef Ma J, et al. Characterization of mammary cancer stem cells in the MMTV-PyMT mouse model. Tumour Biol. 2012;33(6):1983–96.PubMedCrossRef
18.
go back to reference Kersten K, Salvagno C, de Visser KE. Exploiting the immunomodulatory properties of chemotherapeutic drugs to improve the success of cancer immunotherapy. Front Immunol. 2015;6:516.PubMedPubMedCentralCrossRef Kersten K, Salvagno C, de Visser KE. Exploiting the immunomodulatory properties of chemotherapeutic drugs to improve the success of cancer immunotherapy. Front Immunol. 2015;6:516.PubMedPubMedCentralCrossRef
19.
go back to reference Crabtree JS, Miele L. Breast cancer stem cells. Biomedicines. 2018;6(3):94. Crabtree JS, Miele L. Breast cancer stem cells. Biomedicines. 2018;6(3):94.
20.
go back to reference Fico F, et al. Breast cancer stem cells with tumor- versus metastasis-initiating capacities are modulated by TGFBR1 inhibition. Stem Cell Rep. 2019;13(1):1–9.CrossRef Fico F, et al. Breast cancer stem cells with tumor- versus metastasis-initiating capacities are modulated by TGFBR1 inhibition. Stem Cell Rep. 2019;13(1):1–9.CrossRef
21.
go back to reference Lobba AR, et al. Differential expression of CD90 and CD14 stem cell markers in malignant breast cancer cell lines. Cytometry A. 2012;81(12):1084–91.PubMedCrossRef Lobba AR, et al. Differential expression of CD90 and CD14 stem cell markers in malignant breast cancer cell lines. Cytometry A. 2012;81(12):1084–91.PubMedCrossRef
22.
go back to reference Haynes B, et al. Breast cancer complexity: implications of intratumoral heterogeneity in clinical management. Cancer Metastasis Rev. 2017;36(3):547–55.PubMedPubMedCentralCrossRef Haynes B, et al. Breast cancer complexity: implications of intratumoral heterogeneity in clinical management. Cancer Metastasis Rev. 2017;36(3):547–55.PubMedPubMedCentralCrossRef
25.
26.
go back to reference Liao X, Makris M, Luo XM. Fluorescence-activated cell sorting for purification of plasmacytoid dendritic cells from the mouse bone marrow. J Vis Exp. 2016;117:54641. Liao X, Makris M, Luo XM. Fluorescence-activated cell sorting for purification of plasmacytoid dendritic cells from the mouse bone marrow. J Vis Exp. 2016;117:54641.
27.
go back to reference Marhaba R, et al. CD44 and EpCAM: cancer-initiating cell markers. Curr Mol Med. 2008;8(8):784–804.PubMedCrossRef Marhaba R, et al. CD44 and EpCAM: cancer-initiating cell markers. Curr Mol Med. 2008;8(8):784–804.PubMedCrossRef
28.
go back to reference Guy CT, Cardiff RD, Muller WJ. Induction of mammary tumors by expression of polyomavirus middle T oncogene: a transgenic mouse model for metastatic disease. Mol Cell Biol. 1992;12(3):954–61.PubMedPubMedCentral Guy CT, Cardiff RD, Muller WJ. Induction of mammary tumors by expression of polyomavirus middle T oncogene: a transgenic mouse model for metastatic disease. Mol Cell Biol. 1992;12(3):954–61.PubMedPubMedCentral
29.
30.
go back to reference Li B, Dewey CN. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinf. 2011;12:323.CrossRef Li B, Dewey CN. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinf. 2011;12:323.CrossRef
31.
go back to reference Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26(1):139–40.PubMedCrossRef Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26(1):139–40.PubMedCrossRef
32.
go back to reference Bray NL, et al. Near-optimal probabilistic RNA-seq quantification. Nat Biotechnol. 2016;34(5):525–7.CrossRefPubMed Bray NL, et al. Near-optimal probabilistic RNA-seq quantification. Nat Biotechnol. 2016;34(5):525–7.CrossRefPubMed
33.
go back to reference Hen O, Barkan D. Dormant disseminated tumor cells and cancer stem/progenitor-like cells: Similarities and opportunities. Semin Cancer Biol. 2020;60:157–65.PubMedCrossRef Hen O, Barkan D. Dormant disseminated tumor cells and cancer stem/progenitor-like cells: Similarities and opportunities. Semin Cancer Biol. 2020;60:157–65.PubMedCrossRef
34.
go back to reference Cardoso F, et al. 70-gene signature as an aid to treatment decisions in early-stage breast cancer. N Engl J Med. 2016;375(8):717–29.PubMedCrossRef Cardoso F, et al. 70-gene signature as an aid to treatment decisions in early-stage breast cancer. N Engl J Med. 2016;375(8):717–29.PubMedCrossRef
35.
go back to reference van 't Veer LJ, et al. Gene expression profiling predicts clinical outcome of breast cancer. Nature. 2002;415(6871):530–6. van 't Veer LJ, et al. Gene expression profiling predicts clinical outcome of breast cancer. Nature. 2002;415(6871):530–6.
37.
go back to reference Takebe N, Warren RQ, Ivy SP. Breast cancer growth and metastasis: interplay between cancer stem cells, embryonic signaling pathways and epithelial-to-mesenchymal transition. Breast Cancer Res. 2011;13(3):211.PubMedPubMedCentralCrossRef Takebe N, Warren RQ, Ivy SP. Breast cancer growth and metastasis: interplay between cancer stem cells, embryonic signaling pathways and epithelial-to-mesenchymal transition. Breast Cancer Res. 2011;13(3):211.PubMedPubMedCentralCrossRef
38.
go back to reference Feng Y, et al. Breast cancer development and progression: risk factors, cancer stem cells, signaling pathways, genomics, and molecular pathogenesis. Genes Dis. 2018;5(2):77–106.PubMedPubMedCentralCrossRef Feng Y, et al. Breast cancer development and progression: risk factors, cancer stem cells, signaling pathways, genomics, and molecular pathogenesis. Genes Dis. 2018;5(2):77–106.PubMedPubMedCentralCrossRef
39.
go back to reference Evans MK, et al. X-linked inhibitor of apoptosis protein mediates tumor cell resistance to antibody-dependent cellular cytotoxicity. Cell Death Dis. 2016;7:2073.CrossRef Evans MK, et al. X-linked inhibitor of apoptosis protein mediates tumor cell resistance to antibody-dependent cellular cytotoxicity. Cell Death Dis. 2016;7:2073.CrossRef
40.
go back to reference Altieri DC. Survivin, cancer networks and pathway-directed drug discovery. Nat Rev Cancer. 2008;8(1):61–70.PubMedCrossRef Altieri DC. Survivin, cancer networks and pathway-directed drug discovery. Nat Rev Cancer. 2008;8(1):61–70.PubMedCrossRef
41.
go back to reference Wasinski B, et al. Discoidin domain receptors, DDR1b and DDR2, promote tumour growth within collagen but DDR1b suppresses experimental lung metastasis in HT1080 xenografts. Sci Rep. 2020;10(1):2309.PubMedPubMedCentralCrossRef Wasinski B, et al. Discoidin domain receptors, DDR1b and DDR2, promote tumour growth within collagen but DDR1b suppresses experimental lung metastasis in HT1080 xenografts. Sci Rep. 2020;10(1):2309.PubMedPubMedCentralCrossRef
42.
go back to reference Xu X, et al. Matrix metalloproteinase-2 contributes to cancer cell migration on collagen. Cancer Res. 2005;65(1):130–6.PubMedCrossRef Xu X, et al. Matrix metalloproteinase-2 contributes to cancer cell migration on collagen. Cancer Res. 2005;65(1):130–6.PubMedCrossRef
43.
go back to reference Morandi EM, et al. ITGAV and ITGA5 diversely regulate proliferation and adipogenic differentiation of human adipose derived stem cells. Sci Rep. 2016;6:28889.PubMedPubMedCentralCrossRef Morandi EM, et al. ITGAV and ITGA5 diversely regulate proliferation and adipogenic differentiation of human adipose derived stem cells. Sci Rep. 2016;6:28889.PubMedPubMedCentralCrossRef
45.
go back to reference Atsumi T, et al. High expression of inducible 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (iPFK-2; PFKFB3) in human cancers. Cancer Res. 2002;62(20):5881–7.PubMed Atsumi T, et al. High expression of inducible 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (iPFK-2; PFKFB3) in human cancers. Cancer Res. 2002;62(20):5881–7.PubMed
47.
go back to reference Nguyen DX, Bos PD, Massague J. Metastasis: from dissemination to organ-specific colonization. Nat Rev Cancer. 2009;9(4):274–84.PubMedCrossRef Nguyen DX, Bos PD, Massague J. Metastasis: from dissemination to organ-specific colonization. Nat Rev Cancer. 2009;9(4):274–84.PubMedCrossRef
50.
go back to reference Westbrook JA, et al. CAPG and GIPC1: breast cancer biomarkers for bone metastasis development and Treatment. J Natl Cancer Inst. 2016;108(4):djv360.PubMedCentralCrossRef Westbrook JA, et al. CAPG and GIPC1: breast cancer biomarkers for bone metastasis development and Treatment. J Natl Cancer Inst. 2016;108(4):djv360.PubMedCentralCrossRef
51.
53.
go back to reference Ross C, et al. Metastasis-specific gene expression in autochthonous and allograft mouse mammary tumor models: stratification and identification of targetable signatures. Mol Cancer Res. 2020;18(9):1278–89.PubMedPubMedCentralCrossRef Ross C, et al. Metastasis-specific gene expression in autochthonous and allograft mouse mammary tumor models: stratification and identification of targetable signatures. Mol Cancer Res. 2020;18(9):1278–89.PubMedPubMedCentralCrossRef
54.
go back to reference Wood SL, Brown JE. Personal medicine and bone metastases: biomarkers, micro-RNAs and bone metastases. Cancers (Basel). 2020;12(8):2109.PubMedCentralCrossRef Wood SL, Brown JE. Personal medicine and bone metastases: biomarkers, micro-RNAs and bone metastases. Cancers (Basel). 2020;12(8):2109.PubMedCentralCrossRef
55.
go back to reference Yang C, et al. Inducible formation of leader cells driven by CD44 switching gives rise to collective invasion and metastases in luminal breast carcinomas. Oncogene. 2019;38(46):7113–32.PubMedCrossRef Yang C, et al. Inducible formation of leader cells driven by CD44 switching gives rise to collective invasion and metastases in luminal breast carcinomas. Oncogene. 2019;38(46):7113–32.PubMedCrossRef
57.
58.
go back to reference Sachidanandam R, et al. A map of human genome sequence variation containing 1.42 million single nucleotide polymorphisms. Nature 2001;409(6822):928–33. Sachidanandam R, et al. A map of human genome sequence variation containing 1.42 million single nucleotide polymorphisms. Nature 2001;409(6822):928–33.
59.
go back to reference Chen K, Fraley SI. Abstract 5706: Identifying regulators of cancer heterogeneity with phenotypic sorting and single cell sequencing. Can Res. 2020;80(16 Supplement):5706.CrossRef Chen K, Fraley SI. Abstract 5706: Identifying regulators of cancer heterogeneity with phenotypic sorting and single cell sequencing. Can Res. 2020;80(16 Supplement):5706.CrossRef
60.
go back to reference Weigelt B, Peterse JL, van 't Veer LJ. Breast cancer metastasis: markers and models. Nat Rev Cancer. 2005;5(8):591–602. Weigelt B, Peterse JL, van 't Veer LJ. Breast cancer metastasis: markers and models. Nat Rev Cancer. 2005;5(8):591–602.
62.
go back to reference Nagai T, et al. Tactics of cancer invasion: solitary and collective invasion. J Biochem. 2020;167(4):347–55.PubMedCrossRef Nagai T, et al. Tactics of cancer invasion: solitary and collective invasion. J Biochem. 2020;167(4):347–55.PubMedCrossRef
64.
go back to reference Iliopoulos D, et al. Inducible formation of breast cancer stem cells and their dynamic equilibrium with non-stem cancer cells via IL6 secretion. Proc Natl Acad Sci USA. 2011;108(4):1397–402.PubMedPubMedCentralCrossRef Iliopoulos D, et al. Inducible formation of breast cancer stem cells and their dynamic equilibrium with non-stem cancer cells via IL6 secretion. Proc Natl Acad Sci USA. 2011;108(4):1397–402.PubMedPubMedCentralCrossRef
65.
go back to reference Plaks V, Kong N, Werb Z. The cancer stem cell niche: HOW essential is the niche in regulating stemness of tumor cells? Cell Stem Cell. 2015;16(3):225–38.PubMedPubMedCentralCrossRef Plaks V, Kong N, Werb Z. The cancer stem cell niche: HOW essential is the niche in regulating stemness of tumor cells? Cell Stem Cell. 2015;16(3):225–38.PubMedPubMedCentralCrossRef
66.
go back to reference Yi Y, et al. Transcriptional suppression of AMPKalpha1 promotes breast cancer metastasis upon oncogene activation. Proc Natl Acad Sci USA. 2020;117(14):8013–21.PubMedPubMedCentralCrossRef Yi Y, et al. Transcriptional suppression of AMPKalpha1 promotes breast cancer metastasis upon oncogene activation. Proc Natl Acad Sci USA. 2020;117(14):8013–21.PubMedPubMedCentralCrossRef
71.
go back to reference Kim RS, et al. Dormancy signatures and metastasis in estrogen receptor positive and negative breast cancer. PLoS ONE 2012;7(4):e35569. Kim RS, et al. Dormancy signatures and metastasis in estrogen receptor positive and negative breast cancer. PLoS ONE 2012;7(4):e35569.
72.
73.
go back to reference Ibrahim SA, et al. Syndecan-1 is a novel molecular marker for triple negative inflammatory breast cancer and modulates the cancer stem cell phenotype via the IL-6/STAT3, Notch and EGFR signaling pathways. Mol Cancer. 2017;16(1):57.PubMedPubMedCentralCrossRef Ibrahim SA, et al. Syndecan-1 is a novel molecular marker for triple negative inflammatory breast cancer and modulates the cancer stem cell phenotype via the IL-6/STAT3, Notch and EGFR signaling pathways. Mol Cancer. 2017;16(1):57.PubMedPubMedCentralCrossRef
74.
go back to reference Gotte M, et al. Predictive value of syndecan-1 expression for the response to neoadjuvant chemotherapy of primary breast cancer. Anticancer Res. 2006;26(1B):621–7.PubMed Gotte M, et al. Predictive value of syndecan-1 expression for the response to neoadjuvant chemotherapy of primary breast cancer. Anticancer Res. 2006;26(1B):621–7.PubMed
75.
go back to reference Grimm D, et al. The role of SOX family members in solid tumours and metastasis. Semin Cancer Biol. 2020;67(Pt 1):122–53.PubMedCrossRef Grimm D, et al. The role of SOX family members in solid tumours and metastasis. Semin Cancer Biol. 2020;67(Pt 1):122–53.PubMedCrossRef
77.
go back to reference Aslakson CJ, Miller FR. Selective events in the metastatic process defined by analysis of the sequential dissemination of subpopulations of a mouse mammary tumor. Cancer Res. 1992;52(6):1399–405.PubMed Aslakson CJ, Miller FR. Selective events in the metastatic process defined by analysis of the sequential dissemination of subpopulations of a mouse mammary tumor. Cancer Res. 1992;52(6):1399–405.PubMed
78.
go back to reference Lifsted T, et al. Identification of inbred mouse strains harboring genetic modifiers of mammary tumor age of onset and metastatic progression. Int J Cancer. 1998;77(4):640–4.PubMedCrossRef Lifsted T, et al. Identification of inbred mouse strains harboring genetic modifiers of mammary tumor age of onset and metastatic progression. Int J Cancer. 1998;77(4):640–4.PubMedCrossRef
79.
go back to reference Le Voyer T, et al. An epistatic interaction controls the latency of a transgene-induced mammary tumor. Mamm Genome. 2000;11(10):883–9.PubMedCrossRef Le Voyer T, et al. An epistatic interaction controls the latency of a transgene-induced mammary tumor. Mamm Genome. 2000;11(10):883–9.PubMedCrossRef
Metadata
Title
Transcriptome analysis of heterogeneity in mouse model of metastatic breast cancer
Authors
Anastasia A. Ionkina
Gabriela Balderrama-Gutierrez
Krystian J. Ibanez
Steve Huy D. Phan
Angelique N. Cortez
Ali Mortazavi
Jennifer A. Prescher
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Breast Cancer Research / Issue 1/2021
Electronic ISSN: 1465-542X
DOI
https://doi.org/10.1186/s13058-021-01468-x

Other articles of this Issue 1/2021

Breast Cancer Research 1/2021 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine