1
|
Ran F, Chen ST, Li MY, Jin DD, Yang ZM. Effect of Diethylstilbestrol on Implantation and Decidualization in Mice. Int J Mol Sci 2025; 26:4122. [PMID: 40362361 PMCID: PMC12071748 DOI: 10.3390/ijms26094122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/23/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Diethylstilbestrol (DES) is a synthetic non-steroidal estrogen, which was widely used to prevent preterm birth and abortion from the 1940s to the 1970s. DES can increase the incidence of infertility, the abnormal reproductive tract, and autoimmune diseases. However, the mechanism underlying DES on early pregnancy in mice is unclear. This study evaluated the effects of DES on early pregnancy in mice, especially on uterine receptivity and decidualization. Newborn female mice were subcutaneously injected with 0.1 mg/kg DES, 1 mg/kg DES, or sesame oil as controls for 5 consecutive days. At 6 weeks old, these female mice were mated with 8-12-week-old fertile males to obtain pregnancy. The uteri of these mice were collected on days 4, 5, and 8 of pregnancy for further analysis. On days 5 and 8 of pregnancy, the number of implantation sites in 0.1 mg/kg DES group is similar to the control group, while almost no implantation sites are detected in the 1 mg/kg DES group. On day 4 of pregnancy, there was no significant difference in uterine receptive molecules between the control group and the 0.1 mg/kg DES group. However, the levels of uterine receptive molecules in the 1 mg/kg DES group are abnormal. In addition, 6 μM DES significantly inhibits mouse in vitro decidualization. The excessive activation of pyroptosis may lead to pregnancy failure. The pyroptosis-related molecules in the 1 mg/kg DES group were significantly up-regulated, suggesting that DES may contribute to pregnancy failure by over-activating pyroptosis.
Collapse
Affiliation(s)
- Feng Ran
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Si-Ting Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Meng-Yuan Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Dan-Dan Jin
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Zeng-Ming Yang
- College of Animal Science, Guizhou University, Guiyang 550025, China
| |
Collapse
|
2
|
Liao S, Zhang X, Chen L, Zhang J, Lu W, Rao M, Zhang Y, Ye Z, Ivanova D, Li F, Chen X, Wang Y, Song A, Xie B, Wang M. KRT14 is a promising prognostic biomarker of breast cancer related to immune infiltration. Mol Immunol 2025; 180:55-73. [PMID: 40014952 DOI: 10.1016/j.molimm.2025.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 01/12/2025] [Accepted: 02/19/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Breast cancer (BC) is the leading cancer among women globally, which has the highest incidence and mortality rate in over a hundred countries. This study was intended to discover a new prognostic biomarker, facilitating personalized treatment approaches. METHODS RNA sequencing data from The Cancer Genome Atlas database and Gene Expression Omnibus database were utilized to download to evaluate expression levels and prognostic significance of Keratin 14 (KRT14). Methylation of KRT14 was also assessed. The CIBERSORT and single-sample gene set enrichment analysis algorithms were applied to explore the connection between KRT14 and the tumor microenvironment. Primary drugs' sensitivity and potential small molecule therapeutic compounds were analyzed through the "pRRophetic" R package and the Connectivity Map. The prognostic value of KRT14 was additionally corroborated through a comparison of protein levels in peritumoral and cancerous tissues via immunohistochemistry. Moreover, an immune-related prognostic model based on KRT14 was designed to enhance the prediction accuracy for the prognosis of BC patients. RESULTS The study found that KRT14 expression was generally downregulated in BC, correlating strongly with poor prognosis. Compared to normal tissues, the methylation level of KRT14 was higher in BC tissues. Lower expression of KRT14 was linked to decreased anti-tumoral immune cells infiltration and increased immunosuppressive cells infiltration. Sensitivity to various key therapeutic drugs was lower in groups with diminished KRT14 expression. In addition, several potential anti-BC small molecule compounds were identified. The model designed in this study significantly enhanced the predictive capability for BC patients compared to predictions based solely on KRT14 expression levels. CONCLUSION Overall, KRT14 was closely correlated with the prognosis in BC, making it a reliable biomarker.
Collapse
Affiliation(s)
- Siqi Liao
- Department of Physiology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xin Zhang
- Department of Physiology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Lanhui Chen
- Department of Physiology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Jianning Zhang
- Department of Physiology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Weiyu Lu
- Department of Physiology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Mengou Rao
- Department of Physiology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yifan Zhang
- Department of Physiology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Zijian Ye
- Department of Physiology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Deyana Ivanova
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston MA02115, USA
| | - Fangfang Li
- Joint International Research Laboratory of Reproduction, Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Xuemei Chen
- Joint International Research Laboratory of Reproduction, Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Yingxiong Wang
- Joint International Research Laboratory of Reproduction, Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Anchao Song
- Department of Biostatistics, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Biao Xie
- Department of Biostatistics, School of Public Health, Chongqing Medical University, Chongqing 400016, China.
| | - Meijiao Wang
- Department of Physiology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China; Joint International Research Laboratory of Reproduction, Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
3
|
Cyr DG, Gregory M, Hermo L, Dufresne J. Molecular Pathways Implicated in the Differentiation and Function of Epididymal Basal Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1469:89-113. [PMID: 40301254 DOI: 10.1007/978-3-031-82990-1_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2025]
Abstract
The postnatal development of the epididymis is a complex and poorly understood process. Our recent studies have shown that undifferentiated primitive small columnar cells are stem cells and can differentiate in vitro into basal and principal cells. This process represents a key aspect of early epididymal development. As such, the genes and signaling pathways implicated in the differentiation of stem cells are critical. In the rat, epididymal development has been subdivided into three phases consisting of an undifferentiated epithelium (birth to day 14), differentiation (days 14 to 44), and expansion (day 45 to adult). During this period, changes in gene expression in the epididymis are the most significant, as almost 1500 genes are differentially expressed between epididymides of 7 and 18 days of age. In the adult rat, basal cells appear to represent a quiescent adult stem cell population that can be cultured under 3D conditions and can differentiate into principal cells. Gene expression in basal cells of adults compared with epididymides from day 7 rats reveals approximately 400 genes that are common to both. Analyses of these genes predict multiple signaling pathways and master regulator genes. Their roles in early epididymal development suggest that the process is complex and involves multiple regulators, cell surface factors, signaling pathways, and hormones that are interconnected and which promote the differentiation of epididymal basal cells into other epididymal cell types.
Collapse
Affiliation(s)
- Daniel G Cyr
- Laboratory for Reproductive Toxicology, INRS-Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, QC, Canada.
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada.
- Department of Obstetrics, Gynecology and Reproduction, Université Laval, Québec, QC, Canada.
| | - Mary Gregory
- Laboratory for Reproductive Toxicology, INRS-Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, QC, Canada
| | - Louis Hermo
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | - Julie Dufresne
- Laboratory for Reproductive Toxicology, INRS-Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, QC, Canada
| |
Collapse
|
4
|
Yun Y, Kim S, Lee SN, Cho HY, Choi JW. Nanomaterial-based detection of circulating tumor cells and circulating cancer stem cells for cancer immunotherapy. NANO CONVERGENCE 2024; 11:56. [PMID: 39671082 PMCID: PMC11645384 DOI: 10.1186/s40580-024-00466-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 12/04/2024] [Indexed: 12/14/2024]
Abstract
Nanomaterials have emerged as transformative tools for detecting circulating tumor cells (CTCs) and circulating cancer stem cells (CCSCs), significantly enhancing cancer diagnostics and immunotherapy. Nanomaterials, including those composed of gold, magnetic materials, and silica, have enhanced the sensitivity, specificity, and efficiency of isolating these rare cells from blood. These developments are of paramount importance for the early detection of cancer and for providing real-time insights into metastasis and treatment resistance, which are essential for the development of personalized immunotherapies. The combination of nanomaterial-based platforms with phenotyping techniques, such as Raman spectroscopy and microfluidics, enables researchers to enhance immunotherapy protocols targeting specific CTC and CCSC markers. Nanomaterials also facilitate the targeted delivery of immunotherapeutic agents, including immune checkpoint inhibitors and therapeutic antibodies, directly to tumor cells. This synergistic approach has the potential to enhance therapeutic efficacy and mitigate the risk of metastasis and relapse. In conclusion, this review critically examines the use of nanomaterial-driven detection systems for detecting CTCs and CCSCs, their application in immunotherapy, and suggests future directions, highlighting their potential to transform the integration of diagnostics and treatment, thereby paving the way for more precise and personalized cancer therapies.
Collapse
Affiliation(s)
- Yeochan Yun
- Department of Bio and Fermentation Convergence Technology, Kookmin University, 77 Jeongneung-ro, Seongbuk-gu, Seoul, 02707, Republic of Korea
| | - Seewoo Kim
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107, Republic of Korea
| | - Sang-Nam Lee
- Uniance Gene Inc., 273, Digital-ro, Guro-gu, Seoul, 08381, Republic of Korea.
| | - Hyeon-Yeol Cho
- Department of Bio and Fermentation Convergence Technology, Kookmin University, 77 Jeongneung-ro, Seongbuk-gu, Seoul, 02707, Republic of Korea.
| | - Jeong-Woo Choi
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107, Republic of Korea.
| |
Collapse
|
5
|
Thi K, Del Toro K, Licon-Munoz Y, Sayaman RW, Hines WC. Comprehensive identification, isolation, and culture of human breast cell types. J Biol Chem 2024; 300:107637. [PMID: 39122004 PMCID: PMC11459906 DOI: 10.1016/j.jbc.2024.107637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 07/03/2024] [Accepted: 07/16/2024] [Indexed: 08/12/2024] Open
Abstract
Tissues are formed and shaped by cells of many different types and are orchestrated through countless interactions. Deciphering a tissue's biological complexity thus requires studying it at cell-level resolution, where molecular and biochemical features of different cell types can be explored and thoroughly dissected. Unfortunately, the lack of comprehensive methods to identify, isolate, and culture each cell type from many tissues has impeded progress. Here, we present a method for the breadth of cell types composing the human breast. Our goal has long been to understand the essence of each of these different breast cell types, to reveal the underlying biology explaining their intrinsic features, the consequences of interactions, and their contributions to the tissue. This biological exploration has required cell purification, deep-RNA sequencing, and a thorough dissection of the genes and pathways defining each cell type. While the molecular analysis is presented in an adjoining article, we present here an exhaustive cellular dissection of the human breast and explore its cellular composition and histological organization. Moreover, we introduce a novel FACS antibody panel and rigorous gating strategy capable of isolating each of the 12 major breast cell types to purity. Finally, we describe the creation of primary cell models from nearly every breast cell type-some the first of their kind-and submit these as critical tools for studying the dynamic cellular interactions within breast tissues and tumors. Together, this body of work delivers a unique perspective of the breast, revealing insights into its cellular, molecular, and biochemical composition.
Collapse
Affiliation(s)
- Kate Thi
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - Katelyn Del Toro
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Yamhilette Licon-Munoz
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Rosalyn W Sayaman
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - William C Hines
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA.
| |
Collapse
|
6
|
Grasset EM, Deshpande A, Lee JW, Cho Y, Shin SM, Coyne EM, Hernandez A, Yuan X, Zhang Z, Cimino-Mathews A, Ewald AJ, Ho WJ. Mapping the breast tumor microenvironment: proximity analysis reveals spatial relationships between macrophage subtypes and metastasis-initiating cancer cells. Oncogene 2024; 43:2927-2937. [PMID: 39164522 DOI: 10.1038/s41388-024-03127-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/30/2024] [Accepted: 08/06/2024] [Indexed: 08/22/2024]
Abstract
Metastasis is responsible for the majority of cancer-related fatalities. We previously identified specific cancer cell populations responsible for metastatic events which are cytokeratin-14 (CK14) and E-cadherin positive in luminal tumors, and E-cadherin and vimentin positive in triple-negative tumors. Since cancer cells evolve within a complex ecosystem comprised of immune cells and stromal cells, we sought to decipher the spatial interactions of these aggressive cancer cell populations within the tumor microenvironment (TME). We used imaging mass cytometry to detect 36 proteins in tumor microarrays containing paired primary and metastatic lesions from luminal or triple-negative breast cancers (TNBC), resulting in a dataset of 1,477,337 annotated cells. Focusing on metastasis-initiating cell populations, we observed close proximity to specific fibroblast and macrophage subtypes, a relationship maintained between primary and metastatic tumors. Notably, high CK14 in luminal cancer cells and high vimentin in TNBC cells correlated with close proximity to specific macrophage subtypes (CD163intCD206intPDL1intHLA-DR+ or PDL1highARG1high). Our in-depth spatial analysis demonstrates that metastasis-initiating cancer cells consistently colocalizes with distinct cell populations within the TME, suggesting a role for these cell-cell interactions in promoting metastasis.
Collapse
Affiliation(s)
- Eloïse M Grasset
- Université de Nantes, INSERM, CNRS, CRCI2NA, Nantes, France.
- Équipe Labellisée LIGUE Contre le Cancer CRCI2NA, Nantes, France.
- Department of Cell Biology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
| | - Atul Deshpande
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University, Baltimore, MD, USA
- Department of Electrical and Computer Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jae W Lee
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Yeonju Cho
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Sarah M Shin
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Erin M Coyne
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Alexei Hernandez
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Xuan Yuan
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Zhehao Zhang
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Ashley Cimino-Mathews
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Andrew J Ewald
- Department of Cell Biology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University, Baltimore, MD, USA
- Giovanis Institute for Translational Cell Biology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Won Jin Ho
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.
- Convergence Institute, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
7
|
Sato N, Tsujimoto M, Nakatsuji M, Tsuji H, Sugama Y, Shimazu K, Shimoda M, Ishihara H. Flow cytometric analysis for Ki67 assessment in formalin-fixed paraffin-embedded breast cancer tissue. BMC Biol 2024; 22:181. [PMID: 39183273 PMCID: PMC11346000 DOI: 10.1186/s12915-024-01980-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 08/13/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Pathologists commonly employ the Ki67 immunohistochemistry labelling index (LI) when deciding appropriate therapeutic strategies for patients with breast cancer. However, despite several attempts at standardizing the Ki67 LI, inter-observer and inter-laboratory bias remain problematic. We developed a flow cytometric assay that employed tissue dissociation, enzymatic treatment and a gating process to analyse Ki67 in formalin-fixed paraffin-embedded (FFPE) breast cancer tissue. RESULTS We demonstrated that mechanical homogenizations combined with thrombin treatment can be used to recover efficiently intact single-cell nuclei from FFPE breast cancer tissue. Ki67 in the recovered cell nuclei retained reactivity against the MIB-1 antibody, which has been widely used in clinical settings. Additionally, since the method did not alter the nucleoskeletal structure of tissues, the nuclei of cancer cells can be enriched in data analysis based on differences in size and complexity of nuclei of lymphocytes and normal mammary cells. In a clinical study using the developed protocol, Ki67 positivity was correlated with the Ki67 LI obtained by hot spot analysis by a pathologist in Japan (rho = 0.756, P < 0.0001). The number of cancer cell nuclei subjected to the analysis in our assay was more than twice the number routinely checked by pathologists in clinical settings. CONCLUSIONS The findings of this study showed the application of this new flow cytometry method could potentially be used to standardize Ki67 assessments in breast cancer.
Collapse
Affiliation(s)
- Natsuki Sato
- Nitto Boseki Co., Ltd, 2-4-1, Kojimachi, Chiyoda-ku, Tokyo, 102-8489, Japan
| | - Masahiko Tsujimoto
- Department of Diagnostic Pathology, Daini Osaka Police Hospital, 2-6-40 Karasugatsuji, Tennoji-Ku, Osaka, 543-8922, Japan
- Present Address: Osaka Pathology and Cytology Laboratory, 2-2-26 Kunijima, Higashiyodogawa-Ku, Osaka, 533-0024, Japan
| | - Masatoshi Nakatsuji
- Nitto Boseki Co., Ltd, 2-4-1, Kojimachi, Chiyoda-ku, Tokyo, 102-8489, Japan
- Department of Pathobiochemistry, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Hiromi Tsuji
- Department of Diagnostic Pathology, Osaka Police Hospital, 10-31 Kitayamacho, Tennoji-Ku, Osaka, Japan
| | - Yuji Sugama
- Nitto Boseki Co., Ltd, 2-4-1, Kojimachi, Chiyoda-ku, Tokyo, 102-8489, Japan
| | - Kenzo Shimazu
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masafumi Shimoda
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Hideki Ishihara
- Nitto Boseki Co., Ltd, 2-4-1, Kojimachi, Chiyoda-ku, Tokyo, 102-8489, Japan.
- Department of Research Support, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8, Saito-Asagi, Ibaraki City, Osaka, 567-0085, Japan.
| |
Collapse
|
8
|
Whitham D, Bruno P, Haaker N, Arcaro KF, Pentecost BT, Darie CC. Deciphering a proteomic signature for the early detection of breast cancer from breast milk: the role of quantitative proteomics. Expert Rev Proteomics 2024; 21:81-98. [PMID: 38376826 PMCID: PMC11694492 DOI: 10.1080/14789450.2024.2320158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 12/26/2023] [Indexed: 02/21/2024]
Abstract
INTRODUCTION Breast cancer is one of the most prevalent cancers among women in the United States. Current research regarding breast milk has been focused on the composition and its role in infant growth and development. There is little information about the proteins, immune cells, and epithelial cells present in breast milk which can be indicative of the emergence of BC cells and tumors. AREAS COVERED We summarize all breast milk studies previously done in our group using proteomics. These studies include 1D-PAGE and 2D-PAGE analysis of breast milk samples, which include within woman and across woman comparisons to identify dysregulated proteins in breast milk and the roles of these proteins in both the development of BC and its diagnosis. Our projected outlook for the use of milk for cancer detection is also discussed. EXPERT OPINION Analyzing the samples by multiple methods allows one to interrogate a set of samples with various biochemical methods that complement each other, thus providing a more comprehensive proteome. Complementing methods like 1D-PAGE, 2D-PAGE, in-solution digestion and proteomics analysis with PTM-omics, peptidomics, degradomics, or interactomics will provide a better understanding of the dysregulated proteins, but also the modifications or interactions between these proteins.
Collapse
Affiliation(s)
- Danielle Whitham
- Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY, USA
| | - Pathea Bruno
- Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY, USA
| | - Norman Haaker
- Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY, USA
| | - Kathleen F. Arcaro
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Brian T. Pentecost
- Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY, USA
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Costel C. Darie
- Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY, USA
| |
Collapse
|
9
|
Sahoo S, Ramu S, Nair MG, Pillai M, San Juan BP, Milioli HZ, Mandal S, Naidu CM, Mavatkar AD, Subramaniam H, Neogi AG, Chaffer CL, Prabhu JS, Somarelli JA, Jolly MK. Multi-modal transcriptomic analysis unravels enrichment of hybrid epithelial/mesenchymal state and enhanced phenotypic heterogeneity in basal breast cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.30.558960. [PMID: 37873432 PMCID: PMC10592858 DOI: 10.1101/2023.09.30.558960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Intra-tumoral phenotypic heterogeneity promotes tumor relapse and therapeutic resistance and remains an unsolved clinical challenge. It manifests along multiple phenotypic axes and decoding the interconnections among these different axes is crucial to understand its molecular origins and to develop novel therapeutic strategies to control it. Here, we use multi-modal transcriptomic data analysis - bulk, single-cell and spatial transcriptomics - from breast cancer cell lines and primary tumor samples, to identify associations between epithelial-mesenchymal transition (EMT) and luminal-basal plasticity - two key processes that enable heterogeneity. We show that luminal breast cancer strongly associates with an epithelial cell state, but basal breast cancer is associated with hybrid epithelial/mesenchymal phenotype(s) and higher phenotypic heterogeneity. These patterns were inherent in methylation profiles, suggesting an epigenetic crosstalk between EMT and lineage plasticity in breast cancer. Mathematical modelling of core underlying gene regulatory networks representative of the crosstalk between the luminal-basal and epithelial-mesenchymal axes recapitulate and thus elucidate mechanistic underpinnings of the observed associations from transcriptomic data. Our systems-based approach integrating multi-modal data analysis with mechanism-based modeling offers a predictive framework to characterize intra-tumor heterogeneity and to identify possible interventions to restrict it.
Collapse
Affiliation(s)
- Sarthak Sahoo
- Department of Bioengineering, Indian Institute of Science, Bangalore, 560012, India
| | - Soundharya Ramu
- Department of Bioengineering, Indian Institute of Science, Bangalore, 560012, India
| | - Madhumathy G Nair
- Division of Molecular Medicine, St. John’s Research Institute, St. John’s Medical College, Bangalore, 560012, India
| | - Maalavika Pillai
- Department of Bioengineering, Indian Institute of Science, Bangalore, 560012, India
- Current affiliation: Feinberg School of Medicine, Northwestern University, Chicago, 60611, USA
| | - Beatriz P San Juan
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
| | | | - Susmita Mandal
- Department of Bioengineering, Indian Institute of Science, Bangalore, 560012, India
| | - Chandrakala M Naidu
- Division of Molecular Medicine, St. John’s Research Institute, St. John’s Medical College, Bangalore, 560012, India
| | - Apoorva D Mavatkar
- Division of Molecular Medicine, St. John’s Research Institute, St. John’s Medical College, Bangalore, 560012, India
| | - Harini Subramaniam
- Department of Bioengineering, Indian Institute of Science, Bangalore, 560012, India
| | - Arpita G Neogi
- Department of Bioengineering, Indian Institute of Science, Bangalore, 560012, India
| | - Christine L Chaffer
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
- University of New South Wales, UNSW Medicine, UNSW Sydney, NSW, 2052, Australia
| | - Jyothi S Prabhu
- Division of Molecular Medicine, St. John’s Research Institute, St. John’s Medical College, Bangalore, 560012, India
| | | | - Mohit Kumar Jolly
- Department of Bioengineering, Indian Institute of Science, Bangalore, 560012, India
| |
Collapse
|
10
|
Najafabadi MG, Gray GK, Kong LR, Gupta K, Perera D, Naylor H, Brugge JS, Venkitaraman AR, Shehata M. A transcriptional response to replication stress selectively expands a subset of Brca2-mutant mammary epithelial cells. Nat Commun 2023; 14:5206. [PMID: 37626143 PMCID: PMC10457340 DOI: 10.1038/s41467-023-40956-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Germline BRCA2 mutation carriers frequently develop luminal-like breast cancers, but it remains unclear how BRCA2 mutations affect mammary epithelial subpopulations. Here, we report that monoallelic Brca2mut/WT mammary organoids subjected to replication stress activate a transcriptional response that selectively expands Brca2mut/WT luminal cells lacking hormone receptor expression (HR-). While CyTOF analyses reveal comparable epithelial compositions among wildtype and Brca2mut/WT mammary glands, Brca2mut/WT HR- luminal cells exhibit greater organoid formation and preferentially survive and expand under replication stress. ScRNA-seq analysis corroborates the expansion of HR- luminal cells which express elevated transcript levels of Tetraspanin-8 (Tspan8) and Thrsp, plus pathways implicated in replication stress survival including Type I interferon responses. Notably, CRISPR/Cas9-mediated deletion of Tspan8 or Thrsp prevents Brca2mut/WT HR- luminal cell expansion. Our findings indicate that Brca2mut/WT cells activate a transcriptional response after replication stress that preferentially favours outgrowth of HR- luminal cells through the expression of interferon-responsive and mammary alveolar genes.
Collapse
Affiliation(s)
| | - G Kenneth Gray
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA, USA
| | - Li Ren Kong
- MRC Cancer Unit, University of Cambridge, Cambridge, UK
- The Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Pharmacology, NUS School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research, National University of Singapore, Singapore, Singapore
| | - Komal Gupta
- MRC Cancer Unit, University of Cambridge, Cambridge, UK
- The Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research, National University of Singapore, Singapore, Singapore
| | - David Perera
- MRC Cancer Unit, University of Cambridge, Cambridge, UK
| | - Huw Naylor
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Joan S Brugge
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA, USA
| | - Ashok R Venkitaraman
- MRC Cancer Unit, University of Cambridge, Cambridge, UK.
- The Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.
- Institute of Molecular & Cellular Biology Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.
| | - Mona Shehata
- Department of Oncology, University of Cambridge, Cambridge, UK.
- MRC Cancer Unit, University of Cambridge, Cambridge, UK.
| |
Collapse
|
11
|
Martínez-Illescas NG, Leal S, González P, Graña-Castro O, Muñoz-Oliveira JJ, Cortés-Peña A, Gómez-Gil M, Vega Z, Neva V, Romero A, Quintela-Fandino M, Ciruelos E, Sanz C, Aragón S, Sotolongo L, Jiménez S, Caleiras E, Mulero F, Sánchez C, Malumbres M, Salazar-Roa M. miR-203 drives breast cancer cell differentiation. Breast Cancer Res 2023; 25:91. [PMID: 37542268 PMCID: PMC10401798 DOI: 10.1186/s13058-023-01690-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 07/25/2023] [Indexed: 08/06/2023] Open
Abstract
A hallmark of many malignant tumors is dedifferentiated (immature) cells bearing slight or no resemblance to the normal cells from which the cancer originated. Tumor dedifferentiated cells exhibit a higher capacity to survive to chemo and radiotherapies and have the ability to incite tumor relapse. Inducing cancer cell differentiation would abolish their self-renewal and invasive capacity and could be combined with the current standard of care, especially in poorly differentiated and aggressive tumors (with worst prognosis). However, differentiation therapy is still in its early stages and the intrinsic complexity of solid tumor heterogeneity demands innovative approaches in order to be efficiently translated into the clinic. We demonstrate here that microRNA 203, a potent driver of differentiation in pluripotent stem cells (ESCs and iPSCs), promotes the differentiation of mammary gland tumor cells. Combining mouse in vivo approaches and both mouse and human-derived tridimensional organoid cultures, we report that miR-203 influences the self-renewal capacity, plasticity and differentiation potential of breast cancer cells and prevents tumor cell growth in vivo. Our work sheds light on differentiation-based antitumor therapies and offers miR-203 as a promising tool for directly confronting the tumor-maintaining and regeneration capability of cancer cells.
Collapse
Affiliation(s)
- Nuria G Martínez-Illescas
- Department of Biochemistry and Molecular Biology, School of Biology, Complutense University, Madrid, Spain
- Breast and Gynecologic Cancer Group, Research Institute i+12, Madrid, Spain
- Cell Division and Cancer Group, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | | | - Osvaldo Graña-Castro
- Bioinformatics Unit, CNIO, Madrid, Spain
- Department of Basic Medical Sciences, Institute of Applied Molecular Medicine (IMMA-Nemesio Díez), San Pablo-CEU University, Madrid, Spain
| | | | - Alfonso Cortés-Peña
- Flow Cytometry and Fluorescence Microscopy Unit (CAI), Complutense University, Madrid, Spain
| | | | - Zaira Vega
- Histopathology Unit, CNIO, Madrid, Spain
| | | | | | | | - Eva Ciruelos
- Breast and Gynecologic Cancer Group, Research Institute i+12, Madrid, Spain
- Hospital 12 de Octubre, Madrid, Spain
| | - Consuelo Sanz
- Breast and Gynecologic Cancer Group, Research Institute i+12, Madrid, Spain
- Hospital 12 de Octubre, Madrid, Spain
| | - Sofía Aragón
- Breast and Gynecologic Cancer Group, Research Institute i+12, Madrid, Spain
- Hospital 12 de Octubre, Madrid, Spain
| | - Leisy Sotolongo
- Breast and Gynecologic Cancer Group, Research Institute i+12, Madrid, Spain
- Hospital 12 de Octubre, Madrid, Spain
| | - Sara Jiménez
- Breast and Gynecologic Cancer Group, Research Institute i+12, Madrid, Spain
- Hospital 12 de Octubre, Madrid, Spain
| | | | | | - Cristina Sánchez
- Department of Biochemistry and Molecular Biology, School of Biology, Complutense University, Madrid, Spain.
- Breast and Gynecologic Cancer Group, Research Institute i+12, Madrid, Spain.
| | - Marcos Malumbres
- Cell Division and Cancer Group, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.
- Cancer Cell Cycle Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
- ICREA, Passeig Lluís Companys 23, Barcelona, Spain.
| | - María Salazar-Roa
- Department of Biochemistry and Molecular Biology, School of Biology, Complutense University, Madrid, Spain.
- Breast and Gynecologic Cancer Group, Research Institute i+12, Madrid, Spain.
- Cell Division and Cancer Group, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.
| |
Collapse
|
12
|
Mohamed GA, Mahmood S, Ognjenovic NB, Lee MK, Wilkins OM, Christensen BC, Muller KE, Pattabiraman DR. Lineage plasticity enables low-ER luminal tumors to evolve and gain basal-like traits. Breast Cancer Res 2023; 25:23. [PMID: 36859337 PMCID: PMC9979432 DOI: 10.1186/s13058-023-01621-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 02/15/2023] [Indexed: 03/03/2023] Open
Abstract
Stratifying breast cancer into specific molecular or histologic subtypes aids in therapeutic decision-making and predicting outcomes; however, these subtypes may not be as distinct as previously thought. Patients with luminal-like, estrogen receptor (ER)-expressing tumors have better prognosis than patients with more aggressive, triple-negative or basal-like tumors. There is, however, a subset of luminal-like tumors that express lower levels of ER, which exhibit more basal-like features. We have found that breast tumors expressing lower levels of ER, traditionally considered to be luminal-like, represent a distinct subset of breast cancer characterized by the emergence of basal-like features. Lineage tracing of low-ER tumors in the MMTV-PyMT mouse mammary tumor model revealed that basal marker-expressing cells arose from normal luminal epithelial cells, suggesting that luminal-to-basal plasticity is responsible for the evolution and emergence of basal-like characteristics. This plasticity allows tumor cells to gain a new lumino-basal phenotype, thus leading to intratumoral lumino-basal heterogeneity. Single-cell RNA sequencing revealed SOX10 as a potential driver for this plasticity, which is known among breast tumors to be almost exclusively expressed in triple-negative breast cancer (TNBC) and was also found to be highly expressed in low-ER tumors. These findings suggest that basal-like tumors may result from the evolutionary progression of luminal tumors with low ER expression.
Collapse
Affiliation(s)
- Gadisti Aisha Mohamed
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
| | - Sundis Mahmood
- Department of Pathology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, 03756, USA
| | - Nevena B Ognjenovic
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
| | - Min Kyung Lee
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
| | - Owen M Wilkins
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
- Dartmouth Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH, 03756, USA
| | - Brock C Christensen
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
- Dartmouth Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH, 03756, USA
| | - Kristen E Muller
- Department of Pathology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, 03756, USA.
| | - Diwakar R Pattabiraman
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA.
- Dartmouth Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH, 03756, USA.
| |
Collapse
|
13
|
Aylon Y, Furth N, Mallel G, Friedlander G, Nataraj NB, Dong M, Hassin O, Zoabi R, Cohen B, Drendel V, Salame TM, Mukherjee S, Harpaz N, Johnson R, Aulitzky WE, Yarden Y, Shema E, Oren M. Breast cancer plasticity is restricted by a LATS1-NCOR1 repressive axis. Nat Commun 2022; 13:7199. [PMID: 36443319 PMCID: PMC9705295 DOI: 10.1038/s41467-022-34863-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 11/10/2022] [Indexed: 11/29/2022] Open
Abstract
Breast cancer, the most frequent cancer in women, is generally classified into several distinct histological and molecular subtypes. However, single-cell technologies have revealed remarkable cellular and functional heterogeneity across subtypes and even within individual breast tumors. Much of this heterogeneity is attributable to dynamic alterations in the epigenetic landscape of the cancer cells, which promote phenotypic plasticity. Such plasticity, including transition from luminal to basal-like cell identity, can promote disease aggressiveness. We now report that the tumor suppressor LATS1, whose expression is often downregulated in human breast cancer, helps maintain luminal breast cancer cell identity by reducing the chromatin accessibility of genes that are characteristic of a "basal-like" state, preventing their spurious activation. This is achieved via interaction of LATS1 with the NCOR1 nuclear corepressor and recruitment of HDAC1, driving histone H3K27 deacetylation near NCOR1-repressed "basal-like" genes. Consequently, decreased expression of LATS1 elevates the expression of such genes and facilitates slippage towards a more basal-like phenotypic identity. We propose that by enforcing rigorous silencing of repressed genes, the LATS1-NCOR1 axis maintains luminal cell identity and restricts breast cancer progression.
Collapse
Affiliation(s)
- Yael Aylon
- grid.13992.300000 0004 0604 7563Department of Molecular Cell Biology, The Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Noa Furth
- grid.13992.300000 0004 0604 7563Department of Immunology and Regenerative Biology, The Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Giuseppe Mallel
- grid.13992.300000 0004 0604 7563Department of Molecular Cell Biology, The Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Gilgi Friedlander
- grid.13992.300000 0004 0604 7563Department of Life Sciences Core Facilities, The Nancy & Stephen Grand Israel National Center for Personalized Medicine (G-INCPM), The Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Nishanth Belugali Nataraj
- grid.13992.300000 0004 0604 7563Department of Immunology and Regenerative Biology, The Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Meng Dong
- grid.502798.10000 0004 0561 903XDr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology and University of Tuebingen, Stuttgart, Germany
| | - Ori Hassin
- grid.13992.300000 0004 0604 7563Department of Molecular Cell Biology, The Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Rawan Zoabi
- grid.13992.300000 0004 0604 7563Department of Molecular Cell Biology, The Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Benjamin Cohen
- grid.13992.300000 0004 0604 7563Department of Immunology, The Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Vanessa Drendel
- grid.416008.b0000 0004 0603 4965Department of Pathology, Robert Bosch Hospital, Stuttgart, Germany
| | - Tomer Meir Salame
- grid.13992.300000 0004 0604 7563Flow Cytometry Unit, Department of Life Sciences Core Facilities, The Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Saptaparna Mukherjee
- grid.13992.300000 0004 0604 7563Department of Molecular Cell Biology, The Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Nofar Harpaz
- grid.13992.300000 0004 0604 7563Department of Immunology and Regenerative Biology, The Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Randy Johnson
- grid.240145.60000 0001 2291 4776Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Walter E. Aulitzky
- grid.416008.b0000 0004 0603 4965Department of Hematology, Oncology and Palliative Medicine, Robert Bosch Hospital, Stuttgart, Germany
| | - Yosef Yarden
- grid.13992.300000 0004 0604 7563Department of Immunology and Regenerative Biology, The Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Efrat Shema
- grid.13992.300000 0004 0604 7563Department of Immunology and Regenerative Biology, The Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Moshe Oren
- grid.13992.300000 0004 0604 7563Department of Molecular Cell Biology, The Weizmann Institute of Science, 76100 Rehovot, Israel
| |
Collapse
|
14
|
Firouzi J, Sotoodehnejadnematalahi F, Shokouhifar A, Rahimi M, Sodeifi N, Sahranavardfar P, Azimi M, Janzamin E, Safa M, Ebrahimi M. Silibinin exhibits anti-tumor effects in a breast cancer stem cell model by targeting stemness and induction of differentiation and apoptosis. BIOIMPACTS : BI 2022; 12:415-429. [PMID: 36381630 PMCID: PMC9596878 DOI: 10.34172/bi.2022.23336] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 08/27/2021] [Accepted: 09/18/2021] [Indexed: 06/16/2023]
Abstract
Introduction: Malignant breast cancer (BC) frequently contains a rare population of cells called cancer stem cells which underlie tumor relapse and metastasis, and targeting these cells may improve treatment options and outcomes for patients with BC. The aim of the present study was to determine the effect of silibinin on the self-renewal capacity, tumorgenicity, and metastatic potential of mammospheres. Methods: The effect of silibinin on viability and proliferation of MCF-7, MDA-MB-231 mammospheres, and MDA-MB-468 cell aggregation was determined after 72-120 hours of treatment. Colony and sphere formation ability, and the expression of stemness, differentiation, and epithelial-mesenchymal-transition (EMT)-associated genes were assessed by reverse transcription-quantitative polymerase chain reaction (qRT-PCR) in mammospheres treated with an IC50 dose of silibinin. Additionally, the antitumor capacity of silibinin was assessed in vivo, in mice. Results: The results of the present study showed that silibinin decreased the viability of all mammospheres derived from MCF-7, MDA-MB-231, and MDA-MB-468 cell aggregation in a dose-dependent manner. Colony and sphere-forming ability, as well as the expression of genes associated with EMT were reduced in mammospheres treated with silibinin. Additionally, the expression of genes associated with stemness and metastasis was also decreased and the expression of genes associated with differentiation were increased. Intra-tumoral injection of 2 mg/kg silibinin decreased tumor volumes in mice by 2.8 fold. Conclusion: The present study demonstrated that silibinin may have exerted its anti-tumor effects in BC by targeting the BC stem cells, reducing the tumorgenicity and metastasis. Therefore, silibinin may be a potential adjuvant for treatment of BC.
Collapse
Affiliation(s)
- Javad Firouzi
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148
| | | | - Alireza Shokouhifar
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148
| | - Mahsa Rahimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148
| | - Niloufar Sodeifi
- Department of Pathology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran 16635-148, Iran
| | - Parisa Sahranavardfar
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148
| | - Masoumeh Azimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148
| | - Ehsan Janzamin
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148
| | - Majid Safa
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran
- Department of Hematology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148
| |
Collapse
|
15
|
Morrissey RL, Thompson AM, Lozano G. Is loss of p53 a driver of ductal carcinoma in situ progression? Br J Cancer 2022; 127:1744-1754. [PMID: 35764786 DOI: 10.1038/s41416-022-01885-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 05/17/2022] [Accepted: 06/01/2022] [Indexed: 11/09/2022] Open
Abstract
Ductal carcinoma in situ (DCIS) is a non-obligate precursor of invasive carcinoma. Multiple studies have shown that DCIS lesions typically possess a driver mutation associated with cancer development. Mutation in the TP53 tumour suppressor gene is present in 15-30% of pure DCIS lesions and in ~30% of invasive breast cancers. Mutations in TP53 are significantly associated with high-grade DCIS, the most likely form of DCIS to progress to invasive carcinoma. In this review, we summarise published evidence on the prevalence of mutant TP53 in DCIS (including all DCIS subtypes), discuss the availability of mouse models for the study of DCIS and highlight the need for functional studies of the role of TP53 in the development of DCIS and progression from DCIS to invasive disease.
Collapse
Affiliation(s)
- Rhiannon L Morrissey
- Genetics and Epigenetics Program at The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.,Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alastair M Thompson
- Division of Surgical Oncology, Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Guillermina Lozano
- Genetics and Epigenetics Program at The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA. .,Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
16
|
Gray GK, Li CMC, Rosenbluth JM, Selfors LM, Girnius N, Lin JR, Schackmann RCJ, Goh WL, Moore K, Shapiro HK, Mei S, D'Andrea K, Nathanson KL, Sorger PK, Santagata S, Regev A, Garber JE, Dillon DA, Brugge JS. A human breast atlas integrating single-cell proteomics and transcriptomics. Dev Cell 2022; 57:1400-1420.e7. [PMID: 35617956 DOI: 10.1016/j.devcel.2022.05.003] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/23/2022] [Accepted: 05/02/2022] [Indexed: 12/12/2022]
Abstract
The breast is a dynamic organ whose response to physiological and pathophysiological conditions alters its disease susceptibility, yet the specific effects of these clinical variables on cell state remain poorly annotated. We present a unified, high-resolution breast atlas by integrating single-cell RNA-seq, mass cytometry, and cyclic immunofluorescence, encompassing a myriad of states. We define cell subtypes within the alveolar, hormone-sensing, and basal epithelial lineages, delineating associations of several subtypes with cancer risk factors, including age, parity, and BRCA2 germline mutation. Of particular interest is a subset of alveolar cells termed basal-luminal (BL) cells, which exhibit poor transcriptional lineage fidelity, accumulate with age, and carry a gene signature associated with basal-like breast cancer. We further utilize a medium-depletion approach to identify molecular factors regulating cell-subtype proportion in organoids. Together, these data are a rich resource to elucidate diverse mammary cell states.
Collapse
Affiliation(s)
- G Kenneth Gray
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Carman Man-Chung Li
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Jennifer M Rosenbluth
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA; Department of Medical Oncology, Dana-Farber Cancer Institute (DFCI), Boston, MA 02115, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Laura M Selfors
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Nomeda Girnius
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA; The Laboratory of Systems Pharmacology (LSP), HMS, Boston, MA 02115, USA
| | - Jia-Ren Lin
- The Laboratory of Systems Pharmacology (LSP), HMS, Boston, MA 02115, USA
| | - Ron C J Schackmann
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Walter L Goh
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Kaitlin Moore
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Hana K Shapiro
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Shaolin Mei
- The Laboratory of Systems Pharmacology (LSP), HMS, Boston, MA 02115, USA
| | - Kurt D'Andrea
- Department of Medicine, Division of Translation Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Katherine L Nathanson
- Department of Medicine, Division of Translation Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Peter K Sorger
- The Laboratory of Systems Pharmacology (LSP), HMS, Boston, MA 02115, USA
| | - Sandro Santagata
- The Laboratory of Systems Pharmacology (LSP), HMS, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital (BWH), Boston, MA 02115, USA
| | - Aviv Regev
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Judy E Garber
- Department of Medical Oncology, Dana-Farber Cancer Institute (DFCI), Boston, MA 02115, USA
| | - Deborah A Dillon
- Department of Pathology, Brigham and Women's Hospital (BWH), Boston, MA 02115, USA
| | - Joan S Brugge
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA.
| |
Collapse
|
17
|
Ali S, Hamam D, Liu X, Lebrun JJ. Terminal differentiation and anti-tumorigenic effects of prolactin in breast cancer. Front Endocrinol (Lausanne) 2022; 13:993570. [PMID: 36157462 PMCID: PMC9499354 DOI: 10.3389/fendo.2022.993570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/19/2022] [Indexed: 11/13/2022] Open
Abstract
Breast cancer is a major disease affecting women worldwide. A woman has 1 in 8 lifetime risk of developing breast cancer, and morbidity and mortality due to this disease are expected to continue to rise globally. Breast cancer remains a challenging disease due to its heterogeneity, propensity for recurrence and metastasis to distant vital organs including bones, lungs, liver and brain ultimately leading to patient death. Despite the development of various therapeutic strategies to treat breast cancer, still there are no effective treatments once metastasis has occurred. Loss of differentiation and increased cellular plasticity and stemness are being recognized molecularly and clinically as major derivers of heterogeneity, tumor evolution, relapse, metastasis, and therapeutic failure. In solid tumors, breast cancer is one of the leading cancer types in which tumor differentiation state has long been known to influence cancer behavior. Reprograming and/or restoring differentiation of cancer cells has been proposed to provide a viable approach to reverse the cancer through differentiation and terminal maturation. The hormone prolactin (PRL) is known to play a critical role in mammary gland lobuloalveolar development/remodeling and the terminal differentiation of the mammary epithelial cells promoting milk proteins gene expression and lactation. Here, we will highlight recent discoveries supporting an anti-tumorigenic role for PRL in breast cancer as a "pro/forward-differentiation" pathway restricting plasticity, stemness and tumorigenesis.
Collapse
|
18
|
Yue D, Liu W, Gao L, Zhang L, Wang T, Xiao S, Fu Y, Li N, Lin R, Hu Y, Ding L, Zhang Z, Zhang B, Wang C. Integrated Multiomics Analyses Revealing Different Molecular Profiles Between Early- and Late-Stage Lung Adenocarcinoma. Front Oncol 2021; 11:746943. [PMID: 34745971 PMCID: PMC8567144 DOI: 10.3389/fonc.2021.746943] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 10/04/2021] [Indexed: 12/24/2022] Open
Abstract
The molecular differences in genetic and epigenetic profiling between early-stage (ES) and late-stage (LS) lung adenocarcinoma (LUAD), which might help to understand cancer progression and biomarker guided precision treatment, need further be investigated. In this study, we performed comprehensive analysis using multi-omics next-generation sequencing (NGS) on tissue samples from 7 ES (stage I) and 10 LS (stage III/IV) LUAD patients to study molecular characteristics between the two groups. Characterization of the genomic and transcriptomic profiles showed stage-specific somatic mutations, copy number variations (CNVs) and differentially expressed genes (DEGs). LS samples tend to have more TP53, ERBB2 and CHD4 mutations. Gene copy number loss occurs in immune-related gene pathways in the late stage of LUAD. ATAC-seq analysis showed that LS samples harbored more open chromatin peaks around promoter regions and transcription start sites (TSS) than ES samples. We then identified the known transcription factor (TF) binding motifs for the differentially abundant ATAC-seq peaks between the ES and LS samples and found distinct regulatory mechanisms related to each stage. Furthermore, integrative analysis of ATAC-seq with WGS and RNA-seq data showed that the degree of chromatin accessibility is related to copy number changes, and the open chromatin regions could directly regulate the expression of some DEGs. In conclusion, we performed a comprehensive multi-omics analysis of the early and late stages of LUAD and highlighted some important molecular differences in regulatory mechanisms during cancer progression. Those findings help to further understand mechanism and biomarker related targeted therapy.
Collapse
Affiliation(s)
- Dongsheng Yue
- Department of Lung Cancer, Lung Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Weiran Liu
- Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Liuwei Gao
- Department of Enhanced Recovery After Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Lianmin Zhang
- Department of Lung Cancer, Lung Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Tao Wang
- Department of R&D, Hangzhou Repugene Technology Co., Ltd., Hangzhou, China
| | - Shanshan Xiao
- Department of R&D, Hangzhou Repugene Technology Co., Ltd., Hangzhou, China
| | - Yingxue Fu
- Department of R&D, Hangzhou Repugene Technology Co., Ltd., Hangzhou, China
| | - Nan Li
- Department of R&D, Hangzhou Repugene Technology Co., Ltd., Hangzhou, China
| | - Rui Lin
- Department of R&D, Hangzhou Repugene Technology Co., Ltd., Hangzhou, China
| | - Yao Hu
- Department of R&D, Hangzhou Repugene Technology Co., Ltd., Hangzhou, China
| | - Lieming Ding
- Department of Medical, Betta Pharmaceutical Co., Ltd, Hangzhou, China
| | - Zhenfa Zhang
- Department of Lung Cancer, Lung Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Bin Zhang
- Department of Lung Cancer, Lung Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Changli Wang
- Department of Lung Cancer, Lung Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
19
|
Affiliation(s)
- K. Hughes
- Department of Veterinary Medicine University of Cambridge Cambridge UK
| |
Collapse
|
20
|
Virtanen S, Schulte R, Stingl J, Caldas C, Shehata M. High-throughput surface marker screen on primary human breast tissues reveals further cellular heterogeneity. Breast Cancer Res 2021; 23:66. [PMID: 34120626 PMCID: PMC8201685 DOI: 10.1186/s13058-021-01444-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 05/31/2021] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Normal human breast tissues are a heterogeneous mix of epithelial and stromal subtypes in different cell states. Delineating the spectrum of cellular heterogeneity will provide new insights into normal cellular properties within the breast tissue that might become dysregulated in the initial stages of cancer. Investigation of surface marker expression provides a valuable approach to resolve complex cell populations. However, the majority of cell surface maker expression of primary breast cells have not been investigated. METHODS To determine the differences in expression of a range of uninvestigated cell surface markers between the normal breast cell subpopulations, primary human breast cells were analysed using high-throughput flow cytometry for the expression of 242 cell surface proteins in conjunction with EpCAM/CD49f staining. RESULTS We identified 35 surface marker proteins expressed on normal breast epithelial and/or stromal subpopulations that were previously unreported. We also show multiple markers were equally expressed in all cell populations (e.g. CD9, CD59, CD164) while other surface markers were confirmed to be enriched in different cell lineages: CD24, CD227 and CD340 in the luminal compartment, CD10 and CD90 in the basal population, and CD34 and CD140b on stromal cells. CONCLUSIONS Our dataset of CD marker expression in the normal breast provides better definition for breast cellular heterogeneity.
Collapse
Affiliation(s)
- Siru Virtanen
- CRUK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK
| | - Reiner Schulte
- Cambridge Institute for Medical Research, Cambridge University, Cambridge, CB2 0XY, UK
| | - John Stingl
- CRUK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK
| | - Carlos Caldas
- CRUK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK
- Cambridge Breast Unit, Addenbrookes Hospital, Cambridge University Hospital NHS Foundation Trust and NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Mona Shehata
- CRUK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK.
- Medical Research Council Cancer Unit, University of Cambridge, Hills Road, Cambridge, CB2 0XZ, UK.
| |
Collapse
|
21
|
Abstract
An understanding of the anatomy, histology, and development of the equine mammary gland underpins study of the pathology of diseases including galactorrhoea, agalactia, mastitis, and mammary tumour development. This review examines the prenatal development of the equine mammary gland and the striking degree to which the tissue undergoes postnatal development associated with the reproductive cycle. The gland is characterised by epithelial structures arranged in terminal duct lobular units, similar to those of the human breast, supported by distinct zones of intra- and interlobular collagenous stroma. Mastitis and mammary carcinomas are two of the most frequently described equine mammary pathologies and have an overlap in associated clinical signs. Mastitis is most frequently associated with bacterial aetiologies, particularly Streptococcus spp., and knowledge of the process of post-lactational regression can be applied to preventative husbandry strategies. Equine mammary tumours are rare and carry a poor prognosis in many cases. Recent studies have used mammosphere assays to reveal novel insights into the identification and potential behaviour of mammary stem/progenitor cell populations. These suggest that mammospheres derived from equine cells have different growth dynamics compared to those from other species. In parallel with studying the equine mammary gland in order to advance knowledge of equine mammary disease at the interface of basic and clinical science, there is a need to better understand equine lactational biology. This is driven in part by the recognition of the potential value of horse and donkey milk for human consumption, particularly donkey milk in children with 'Cow Milk Protein Allergy'.
Collapse
Affiliation(s)
- Katherine Hughes
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
22
|
Yoshimura H, Moriya M, Yoshida A, Yamamoto M, Machida Y, Ochiai K, Michishita M, Nakagawa T, Matsuda Y, Takahashi K, Kamiya S, Ishiwata T. Involvement of Nestin in the Progression of Canine Mammary Carcinoma. Vet Pathol 2021; 58:994-1003. [PMID: 34056976 DOI: 10.1177/03009858211018656] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Nestin, a class VI intermediate filament protein, is known to be expressed in various types of human neoplasms, including breast cancer, and is associated with their progression. However, its expression and role in canine mammary tumors remain unknown. We analyzed nestin expression in canine mammary tumors using in situ hybridization and immunohistochemistry. We also investigated its role in a canine mammary carcinoma cell line using RNA interference. Nestin expression was not observed in luminal epithelial cells of any of the 62 cases of benign mammary lesions examined, although myoepithelial cells showed its expression in most cases. In 16/50 (32%) primary mammary carcinomas and 6/15 (40%) metastases of mammary carcinomas, cytoplasmic nestin expression was detected in luminal epithelial cells. In luminal cells of primary mammary carcinomas, its expression was positively related to several pathological parameters that indicate high-grade malignancy, including histological grading (P < .01), vascular/lymphatic invasion (P < .01), Ki-67 index (P < .01), and metastasis (P < .05). Immunohistochemistry revealed that nestin expression was related to vimentin expression in mammary carcinomas (P < .01). This relationship was confirmed using reverse transcription-quantitative polymerase chain reaction using 9 cell lines derived from canine mammary carcinoma (P < .01). Finally, nestin knockdown in canine mammary carcinoma cells using small interfering RNA inhibited cell proliferation and migration based on WST-8, Boyden chamber, and cell-tracking assays. These findings suggest that nestin may at least partially mediate these behaviors of canine mammary carcinoma cells.
Collapse
Affiliation(s)
| | - Maiko Moriya
- 12989Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Ayaka Yoshida
- 12989Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Masami Yamamoto
- 12989Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Yukino Machida
- 12989Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Kazuhiko Ochiai
- 12989Nippon Veterinary and Life Science University, Tokyo, Japan
| | | | | | | | | | - Shinji Kamiya
- 12989Nippon Veterinary and Life Science University, Tokyo, Japan
| | | |
Collapse
|
23
|
Genetic Variation and Immunohistochemical Localization of the Glucocorticoid Receptor in Breast Cancer Cases from the Breast Cancer Care in Chicago Cohort. Cancers (Basel) 2021; 13:cancers13102261. [PMID: 34068181 PMCID: PMC8152982 DOI: 10.3390/cancers13102261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 04/27/2021] [Accepted: 05/10/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Glucocorticoid, one of the primary mediators of stress, acts via its receptor, the glucocorticoid receptor (GCR/NR3C1), to regulate a myriad of physiological processes. We measured the genetic variation and protein expression of GCR, and the genes that regulate GCR function or response and examined whether these alterations were associated with breast cancer clinicopathological characteristics. METHOD We used samples from a multiracial cohort of breast cancer patients to assess the association between breast cancer characteristics and the genetic variants of single nucleotide polymorphisms (SNPs) in GCR/NR3C1, FKBP5, Sgk1, IL-6, ADIPOQ, LEPR, SOD2, CAT, and BCL2. RESULTS Several SNPs were associated with breast cancer characteristics, but statistical significance was lost after adjustment for multiple comparisons. GCR was detected in all normal breast tissues and was predominantly located in the nuclei of the myoepithelial cell layer, whereas the luminal layer was negative for GCR. GCR expression was significantly decreased in all breast cancer tissue types, compared to nontumor tissue, but was not associated with breast cancer characteristics. We found that high nuclear GCR expression was associated with basal cell marker cytokeratin 5/6 positivity. CONCLUSION GCR expression is reduced in breast cancer tissue and correlates with the basal cell marker CK5/6.
Collapse
|
24
|
Abe-Fukasawa N, Watanabe R, Gen Y, Nishino T, Itasaki N. A liquid culture cancer spheroid model reveals low PI3K/Akt pathway activity and low adhesiveness to the extracellular matrix. FEBS J 2021; 288:5650-5667. [PMID: 33837641 DOI: 10.1111/febs.15867] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 02/16/2021] [Accepted: 03/29/2021] [Indexed: 01/13/2023]
Abstract
Three-dimensional (3D) cultures of cancer cells in liquid without extracellular matrix (ECM) offer in vitro models for metastasising conditions such as those in vessels and effusion. However, liquid culturing is often hindered by cell adhesiveness, which causes large cell clumps. We previously described a liquid culture material, LA717, which prevents nonclonal cell adhesion and subsequent clumping, thus allowing clonal growth of spheroids in an anchorage-independent manner. Here, we examined such liquid culture cancer spheroids for the acquisition of apical-basal polarity, sensitivity to an Akt inhibitor (anticancer drug MK-2206) and interaction with ECM. The spheroids present apical plasma membrane on the surface, which originated from the failure of polarisation at the single-cell stage and subsequent defects in phosphorylated ezrin accumulation at the cell boundary during the first cleavage, failing internal lumen formation. At the multicellular stage, liquid culture spheroids presented bleb-like protrusion on the surface, which was enhanced by the activation of the PI3K/Akt pathway and reduced by PI3K/Akt inhibitors. Liquid culture spheroids exhibited slow proliferation speed and low endogenous pAkt levels compared with gel-cultured spheroids and 2D-cultured cells, explaining the susceptibility to the Akt-inhibiting anticancer drug. Subcutaneous xenografting and in vitro analysis demonstrated low viability and adhesive property of liquid culture spheroids to ECM, while migratory and invasive capacities were comparable with gel-cultured spheroids. These features agree with the low efficacy of circulating tumour spheroids in the settling step of metastasis. This study demonstrates the feature of anchorage-independent spheroids and validates liquid cultures as a useful method in cancer spheroid research.
Collapse
Affiliation(s)
| | - Rina Watanabe
- Biological Research Laboratories, Nissan Chemical Corporation, Saitama, Japan
| | - Yuki Gen
- Faculty of Health Sciences, University of Bristol, UK
| | - Taito Nishino
- Biological Research Laboratories, Nissan Chemical Corporation, Saitama, Japan
| | - Nobue Itasaki
- Faculty of Health Sciences, University of Bristol, UK
| |
Collapse
|
25
|
Hu L, Su L, Cheng H, Mo C, Ouyang T, Li J, Wang T, Fan Z, Fan T, Lin B, Zhang J, Xie Y. Single-Cell RNA Sequencing Reveals the Cellular Origin and Evolution of Breast Cancer in BRCA1 Mutation Carriers. Cancer Res 2021; 81:2600-2611. [PMID: 33727227 DOI: 10.1158/0008-5472.can-20-2123] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 10/29/2020] [Accepted: 03/11/2021] [Indexed: 11/16/2022]
Abstract
The cell of origin and the development of breast cancer are not fully elucidated in BRCA1 mutation carriers, especially for estrogen receptor (ER)-positive breast cancers. Here, we performed single-cell RNA sequencing (RNA-seq) on 82,122 cells isolated from the breast cancer tissues and adjacent or prophylactic normal breast tissues from four BRCA1 mutation carriers and three noncarriers. Whole-exome sequencing was performed on breast tumors from the four BRCA1 mutation carriers; for validation, bulk RNA-seq was performed on adjacent normal breast tissues from eight additional BRCA1 mutation carriers and 14 noncarriers. Correlation analyses suggested that breast cancers in BRCA1 mutation carriers might originate from luminal cells. The aberrant luminal progenitor cells with impaired differentiation were significantly increased in normal breast tissues in BRCA1 mutation carriers compared with noncarriers. These observations were further validated by the bulk RNA-seq data from additional BRCA1 mutation carriers. These data suggest that the cell of origin of basal-like breast tumors (ERneg) in BRCA1 mutation carriers might be luminal progenitor cells. The expression of TP53 and BRCA1 was decreased in luminal progenitor cells from normal breast tissue in BRCA1 mutation carriers, which might trigger the basal/mesenchymal transition of luminal progenitors and might result in basal-like tumor development. Furthermore, ERhigh luminal tumors might originate from mature luminal cells. Our study provides in-depth evidence regarding the cells of origin of different breast cancer subtypes in BRCA1 mutation carriers. SIGNIFICANCE: Single-cell RNA-seq data indicate that basal-like breast cancer (ERneg) might originate from luminal progenitors, and ERhigh luminal breast cancer might originate from mature luminal cells in BRCA1 mutation carriers.
Collapse
Affiliation(s)
- Li Hu
- Breast Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Liming Su
- Breast Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Hainan Cheng
- Berry Oncology Co., Ltd. (Berry Genomics Group), Beijing, P.R. China
| | - Chunling Mo
- Berry Oncology Co., Ltd. (Berry Genomics Group), Beijing, P.R. China
| | - Tao Ouyang
- Breast Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Jinfeng Li
- Breast Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Tianfeng Wang
- Breast Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Zhaoqing Fan
- Breast Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Tie Fan
- Breast Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Benyao Lin
- Breast Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Jianguang Zhang
- Berry Oncology Co., Ltd. (Berry Genomics Group), Beijing, P.R. China.
| | - Yuntao Xie
- Breast Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, P.R. China.
| |
Collapse
|
26
|
García Solá ME, Stedile M, Beckerman I, Kordon EC. An Integrative Single-cell Transcriptomic Atlas of the Post-natal Mouse Mammary Gland Allows Discovery of New Developmental Trajectories in the Luminal Compartment. J Mammary Gland Biol Neoplasia 2021; 26:29-42. [PMID: 33913090 DOI: 10.1007/s10911-021-09488-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 04/22/2021] [Indexed: 12/13/2022] Open
Abstract
The mammary gland is a highly dynamic organ which undergoes periods of expansion, differentiation and cell death in each reproductive cycle. Partly because of the dynamic nature of the gland, mammary epithelial cells (MECs) are extraordinarily heterogeneous. Single cell RNA-seq (scRNA-seq) analyses have contributed to understand the cellular and transcriptional heterogeneity of this complex tissue. Here, we integrate scRNA-seq data from three foundational reports that have explored the mammary gland cell populations throughout development at single-cell level using 10× Chromium Drop-Seq. We center our analysis on post-natal development of the mammary gland, from puberty to post-involution. The new integrated study corresponds to RNA sequences from 53,686 individual cells, which greatly outnumbers the three initial data sets. The large volume of information provides new insights, as a better resolution of the previously detected Procr+ stem-like cell subpopulation or the identification of a novel group of MECs expressing immune-like markers. Moreover, here we present new pseudo-temporal trajectories of MEC populations at two resolution levels, that is either considering all mammary cell subtypes or focusing specifically on the luminal lineages. Interestingly, the luminal-restricted analysis reveals distinct expression patterns of various genes that encode milk proteins, suggesting specific and non-redundant roles for each of them. In summary, our data show that the application of bioinformatic tools to integrate multiple scRNA-seq data-sets helps to describe and interpret the high level of plasticity involved in gene expression regulation throughout mammary gland post-natal development.
Collapse
Affiliation(s)
- Martín E García Solá
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIByNE), CONICET, Departamento de Fisiología y Biología Molecular y Celular. Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.
| | - Micaela Stedile
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIByNE), CONICET, Departamento de Fisiología y Biología Molecular y Celular. Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Inés Beckerman
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIByNE), CONICET, Departamento de Fisiología y Biología Molecular y Celular. Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Edith C Kordon
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIByNE), CONICET, Departamento de Fisiología y Biología Molecular y Celular. Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
27
|
Peng S, Hebert LL, Eschbacher JM, Kim S. Single-Cell RNA Sequencing of a Postmenopausal Normal Breast Tissue Identifies Multiple Cell Types That Contribute to Breast Cancer. Cancers (Basel) 2020; 12:cancers12123639. [PMID: 33291647 PMCID: PMC7761899 DOI: 10.3390/cancers12123639] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/27/2020] [Accepted: 12/02/2020] [Indexed: 12/26/2022] Open
Abstract
Simple Summary The human body is composed of multiple cell types that form structures and carry out the functions of specific tissues. The human breast is mainly known for the milk ducts organized by epithelial cells, but also contains many other cell types of little-known identity. In this study, we employed the single-cell sequencing technology to ascertain the various cell types present in the normal breast. The results showed 10 distinct cell types that included three epithelial and other novel cell types. The gene signatures of five cell types (three epithelial, one fibroblast subset, and immune cells) matched to the gene expression profiles of >85% breast tumors cataloged in The Cancer Gene Atlas dataset, suggesting their significant contribution to breast cancer. These findings provide a framework for the better mapping of the cellular composition in the breast and its relationship to breast disease. Abstract The human breast is composed of diverse cell types. Studies have delineated mammary epithelial cells, but the other cell types in the breast have scarcely been characterized. In order to gain insight into the cellular composition of the tissue, we performed droplet-mediated RNA sequencing of 3193 single cells isolated from a postmenopausal breast tissue without enriching for epithelial cells. Unbiased clustering analysis identified 10 distinct cell clusters, seven of which were nonepithelial devoid of cytokeratin expression. The remaining three cell clusters expressed cytokeratins (CKs), representing breast epithelial cells; Cluster 2 and Cluster 7 cells expressed luminal and basal CKs, respectively, whereas Cluster 9 cells expressed both luminal and basal CKs, as well as other CKs of unknown specificity. To assess which cell type(s) potentially contributes to breast cancer, we used the differential gene expression signature of each cell cluster to derive gene set variation analysis (GSVA) scores and classified breast tumors in The Cancer Gene Atlas (TGGA) dataset (n = 1100) by assigning the highest GSVA scoring cell cluster number for each tumor. The results showed that five clusters (Clusters 2, 3, 7, 8, and 9) could categorize >85% of breast tumors collectively. Notably, Cluster 2 (luminal epithelial) and Cluster 3 (fibroblast) tumors were equally prevalent in the luminal breast cancer subtypes, whereas Cluster 7 (basal epithelial) and Cluster 9 (other epithelial) tumors were present primarily in the triple-negative breast cancer (TNBC) subtype. Cluster 8 (immune) tumors were present in all subtypes, indicating that immune cells may contribute to breast cancer regardless of the subtypes. Cluster 9 tumors were significantly associated with poor patient survival in TNBC, suggesting that this epithelial cell type may give rise to an aggressive TNBC subset.
Collapse
Affiliation(s)
- Sen Peng
- Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ 85004, USA;
| | - Lora L. Hebert
- Department of Surgery, St. Joseph’s Hospital, Dignity Health, Phoenix, AZ 85013, USA; (L.L.H.); (J.M.E.)
- Surgical Breast Oncology Division, University of Arizona Cancer Center-Phoenix, Phoenix, AZ 85004, USA
| | - Jennifer M. Eschbacher
- Department of Surgery, St. Joseph’s Hospital, Dignity Health, Phoenix, AZ 85013, USA; (L.L.H.); (J.M.E.)
- Department of Neuropathology, Barrow Neurological Institute, Dignity Health, Phoenix, AZ 85013, USA
| | - Suwon Kim
- Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ 85004, USA;
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA
- Correspondence: ; Tel.: +1-602-343-8762
| |
Collapse
|
28
|
Regan JL, Smalley MJ. Integrating single-cell RNA-sequencing and functional assays to decipher mammary cell states and lineage hierarchies. NPJ Breast Cancer 2020; 6:32. [PMID: 32793804 PMCID: PMC7391676 DOI: 10.1038/s41523-020-00175-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 07/02/2020] [Indexed: 12/13/2022] Open
Abstract
The identification and molecular characterization of cellular hierarchies in complex tissues is key to understanding both normal cellular homeostasis and tumorigenesis. The mammary epithelium is a heterogeneous tissue consisting of two main cellular compartments, an outer basal layer containing myoepithelial cells and an inner luminal layer consisting of estrogen receptor-negative (ER−) ductal cells and secretory alveolar cells (in the fully functional differentiated tissue) and hormone-responsive estrogen receptor-positive (ER+) cells. Recent publications have used single-cell RNA-sequencing (scRNA-seq) analysis to decipher epithelial cell differentiation hierarchies in human and murine mammary glands, and reported the identification of new cell types and states based on the expression of the luminal progenitor cell marker KIT (c-Kit). These studies allow for comprehensive and unbiased analysis of the different cell types that constitute a heterogeneous tissue. Here we discuss scRNA-seq studies in the context of previous research in which mammary epithelial cell populations were molecularly and functionally characterized, and identified c-Kit+ progenitors and cell states analogous to those reported in the recent scRNA-seq studies.
Collapse
Affiliation(s)
- Joseph L Regan
- Charité Comprehensive Cancer Centre, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Matthew J Smalley
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Wales, CF24 4HQ UK
| |
Collapse
|
29
|
Biology of Oestrogen-Receptor Positive Primary Breast Cancer in Older Women with Utilisation of Core Needle Biopsy Samples and Correlation with Clinical Outcome. Cancers (Basel) 2020; 12:cancers12082067. [PMID: 32726924 PMCID: PMC7465346 DOI: 10.3390/cancers12082067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 07/23/2020] [Indexed: 01/16/2023] Open
Abstract
The majority of biological profiling studies use surgical excision (SE) samples, excluding patients receiving nonsurgical and neoadjuvant therapy. We propose using core needle biopsy (CNB) for biological profiling in older women. Over 37 years (1973–2010), 1 758 older (≥70 years) women with operable primary breast cancer attended a dedicated clinic. Of these, 693 had sufficient quality CNB to construct tissue microarray (TMA). The pattern of biomarkers was analysed in oestrogen receptor (ER)-positive cases, using immunohistochemistry and partitional clustering analysis. The biomarkers measured were: progesterone receptor (PgR), Ki67, Epidermal Growth Factor Receptor (EGFR), Human Epidermal Growth Factor Receptor (HER)-2, HER3, HER4, p53, cytokeratins CK5/6 and CK7/8, Mucin (MUC)1, liver kinase B1 (LKB1), Breast Cancer Associated gene (BRCA) 1, B-Cell Lymphoma (BCL)-2, phosphate and tensin homolog (PTEN), vascular endothelial growth factor (VEGF), and Amplified in breast cancer 1 (AIB1). CNB TMA construction was possible in 536 ER-positive cases. Multivariate analysis showed progesterone receptor (PgR) (p = 0.015), Ki67 (p = 0.001), and mucin (MUC)1 (p = 0.033) as independent predictors for breast-cancer-specific survival (BCSS). Cluster analysis revealed three biological clusters, which were consistent with luminal A, luminal B, and low-ER luminal. The low-ER luminal cluster had lower BCSS compared to luminal A and B. The presence of the low-ER luminal cluster unique to older women, identified in a previous study in SE TMAs in the same cohort, is confirmed. This present study is novel in its use of core needle biopsy tissue microarrays to profile the biology of breast cancer in older women.
Collapse
|
30
|
Vala H, Carvalho T, Pinto C, Pereira MA, Mesquita JR, Peleteiro MC, Ferrer L, Fondevila D. Immunohistochemical Studies of Cytokeratins and Differentiation Markers in Bovine Ocular Squamous Cell Carcinoma. Vet Sci 2020; 7:70. [PMID: 32485997 PMCID: PMC7355659 DOI: 10.3390/vetsci7020070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/25/2020] [Accepted: 05/27/2020] [Indexed: 11/16/2022] Open
Abstract
Bovine Ocular Squamous Cell Carcinoma is considered the most common bovine tumour, causing significant economic losses, mainly by abattoir condemnations. To obtain a better insight into the genesis and neoplastic transformation, 19 samples collected at slaughter from Holstein Friesian cattle and diagnosed as Ocular Squamous Cell Carcinoma were studied. Tumours were histologically classified into three categories: poorly (26.3%), moderately (26.3%), and well differentiated (47.4%). Expression of keratins (MNF116 and LP34) and of cornified envelope precursors (involucrin and profilaggrin) was studied. Expression of MNF116 was observed in all carcinomas. LP34 immunostaining was seen in all but three carcinomas, one from each degree. Involucrin immunoreaction was observed in all but one poorly differentiated carcinoma. Profilaggrin was present in only two moderately differentiated carcinomas, in all but one well differentiated, and in all but one poorly differentiated. MNF116 is a useful marker to confirm the epithelial origin of the tumour and stain most neoplastic cells in these tumours. The expression of involucrin and LP34 demonstrates that, in all tumours, cells have reached the final program of differentiation, regardless of the grade. The expression of profilaggrin could indicate molecular changes during malignant transformation but their expression does not seem to be of diagnostic value.
Collapse
Affiliation(s)
- Helena Vala
- Agrarian School of the Polytechnic Institute of Viseu, Quinta da Alagoa-Estrada de Nelas Ranhados, 3500-606 Viseu, Portugal;
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro, 5001-801 Vila Real, Portugal
| | - Tânia Carvalho
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal;
| | - Carlos Pinto
- Faculdade de Ciências Agrárias e do Ambiente—Universidade dos Açores, Rua Capitão João d’Ávila—Pico da Urze, 9700-042 Angra do Heroísmo, Portugal;
| | - Maria A. Pereira
- Agrarian School of the Polytechnic Institute of Viseu, Quinta da Alagoa-Estrada de Nelas Ranhados, 3500-606 Viseu, Portugal;
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa (UNL), R. da Junqueira 100, 1349-008 Lisboa, Portugal
| | - João R. Mesquita
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal;
| | - Maria C. Peleteiro
- Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal;
| | - Lluís Ferrer
- Departament de Medicina i Cirurgia Animals, Facultat de Veterinària, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (L.F.); (D.F.)
| | - Dolores Fondevila
- Departament de Medicina i Cirurgia Animals, Facultat de Veterinària, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (L.F.); (D.F.)
| |
Collapse
|
31
|
Cytokeratin 5 alters β-catenin dynamics in breast cancer cells. Oncogene 2020; 39:2478-2492. [PMID: 31988452 PMCID: PMC7085458 DOI: 10.1038/s41388-020-1164-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 12/26/2019] [Accepted: 01/15/2020] [Indexed: 12/13/2022]
Abstract
Estrogen receptor (ER) positive breast cancers often contain subpopulations of cells that express the intermediate filament protein cytokeratin 5 (CK5). CK5+ cells are enriched in cancer stem cell (CSC) properties, can be induced by progestins, and predict poor prognosis in ER+ breast cancer. We established through CK5 knockout and overexpression in ER+ breast cancer cell lines that CK5 is important for tumorsphere formation, prompting us to speculate that CK5 has regulatory activity in CSCs. To interrogate CK5 interacting proteins that may be functionally cooperative, we performed immunoprecipitation-mass spectrometry for CK5 in ER+ breast cancer cells. Focusing on proteins with signaling activity, we identified β-catenin, a key transcription factor of the Wnt signaling pathway and cell adhesion molecule, as a CK5 interactor, which we confirmed by co-immunoprecipitation in several breast cancer models. We interrogated the dual functions of β-catenin in relation to CK5. Knockout or knockdown of CK5 ablated β-catenin transcriptional activity in response to progestins and Wnt stimuli. Conversely, CK5 induced by progestins or overexpression was sufficient to promote loss of β-catenin at the cell membrane and total E-cadherin loss. A breast cancer patient-derived xenograft showed similar loss of membrane β-catenin and E-cadherin in CK5+ but not intratumoral CK5− cells and single cell RNA sequencing found the top enriched pathways in the CK5+ cell cluster were cell junction remodeling and signaling. This report highlights that CK5 actively remodels cell morphology and that blockade of CK5-β-catenin interaction may reverse the detrimental properties of CK5+ breast cancer cells.
Collapse
|
32
|
Anillin regulates breast cancer cell migration, growth, and metastasis by non-canonical mechanisms involving control of cell stemness and differentiation. Breast Cancer Res 2020; 22:3. [PMID: 31910867 PMCID: PMC6947866 DOI: 10.1186/s13058-019-1241-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 12/17/2019] [Indexed: 12/21/2022] Open
Abstract
Background Breast cancer metastasis is driven by a profound remodeling of the cytoskeleton that enables efficient cell migration and invasion. Anillin is a unique scaffolding protein regulating major cytoskeletal structures, such as actin filaments, microtubules, and septin polymers. It is markedly overexpressed in breast cancer, and high anillin expression is associated with poor prognosis. The aim of this study was to investigate the role of anillin in breast cancer cell migration, growth, and metastasis. Methods CRISPR/Cas9 technology was used to deplete anillin in highly metastatic MDA-MB-231 and BT549 cells and to overexpress it in poorly invasive MCF10AneoT cells. The effects of anillin depletion and overexpression on breast cancer cell motility in vitro were examined by wound healing and Matrigel invasion assays. Assembly of the actin cytoskeleton and matrix adhesion were evaluated by immunofluorescence labeling and confocal microscopy. In vitro tumor development was monitored by soft agar growth assays, whereas cancer stem cells were examined using a mammosphere formation assay and flow cytometry. The effects of anillin knockout on tumor growth and metastasis in vivo were determined by injecting control and anillin-depleted breast cancer cells into NSG mice. Results Loss-of-function and gain-of-function studies demonstrated that anillin is necessary and sufficient to accelerate migration, invasion, and anchorage-independent growth of breast cancer cells in vitro. Furthermore, loss of anillin markedly attenuated primary tumor growth and metastasis of breast cancer in vivo. In breast cancer cells, anillin was localized in the nucleus; however, knockout of this protein affected the cytoplasmic/cortical events, e.g., the organization of actin cytoskeleton and cell-matrix adhesions. Furthermore, we observed a global transcriptional reprogramming of anillin-depleted breast cancer cells that resulted in suppression of their stemness and induction of the mesenchymal to epithelial trans-differentiation. Such trans-differentiation was manifested by the upregulation of basal keratins along with the increased expression of E-cadherin and P-cadherin. Knockdown of E-cadherin restored the impaired migration and invasion of anillin-deficient breast cancer cells. Conclusion Our study demonstrates that anillin plays essential roles in promoting breast cancer growth and metastatic dissemination in vitro and in vivo and unravels novel functions of anillin in regulating breast cancer stemness and differentiation.
Collapse
|
33
|
Jimenez-Rojo L, Pagella P, Harada H, Mitsiadis TA. Dental Epithelial Stem Cells as a Source for Mammary Gland Regeneration and Milk Producing Cells In Vivo. Cells 2019; 8:cells8101302. [PMID: 31652655 PMCID: PMC6830078 DOI: 10.3390/cells8101302] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/17/2019] [Accepted: 10/18/2019] [Indexed: 12/17/2022] Open
Abstract
The continuous growth of rodent incisors is ensured by clusters of mesenchymal and epithelial stem cells that are located at the posterior part of these teeth. Genetic lineage tracing studies have shown that dental epithelial stem cells (DESCs) are able to generate all epithelial cell populations within incisors during homeostasis. However, it remains unclear whether these cells have the ability to adopt alternative fates in response to extrinsic factors. Here, we have studied the plasticity of DESCs in the context of mammary gland regeneration. Transplantation of DESCs together with mammary epithelial cells into the mammary stroma resulted in the formation of chimeric ductal epithelial structures in which DESCs adopted all the possible mammary fates including milk-producing alveolar cells. In addition, when transplanted without mammary epithelial cells, DESCs developed branching rudiments and cysts. These in vivo findings demonstrate that when outside their niche, DESCs redirect their fates according to their new microenvironment and thus can contribute to the regeneration of non-dental tissues.
Collapse
Affiliation(s)
- Lucia Jimenez-Rojo
- Department of Orofacial Development and Regeneration, Institute of Oral Biology, Centre for Dental Medicine, University of Zurich, 8032 Zurich, Switzerland.
| | - Pierfrancesco Pagella
- Department of Orofacial Development and Regeneration, Institute of Oral Biology, Centre for Dental Medicine, University of Zurich, 8032 Zurich, Switzerland.
| | - Hidemitsu Harada
- Division of Developmental Biology and Regenerative Medicine, Department of Anatomy, Iwate Medical University Yahaba, Morioka 020-0023, Japan.
| | - Thimios A Mitsiadis
- Department of Orofacial Development and Regeneration, Institute of Oral Biology, Centre for Dental Medicine, University of Zurich, 8032 Zurich, Switzerland.
| |
Collapse
|
34
|
Basal-like Breast Cancers: From Pathology to Biology and Back Again. Stem Cell Reports 2019; 10:1676-1686. [PMID: 29874626 PMCID: PMC6117459 DOI: 10.1016/j.stemcr.2018.04.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 04/23/2018] [Accepted: 04/25/2018] [Indexed: 12/17/2022] Open
Abstract
Human breast cancers referred to as "basal-like" are of interest because they lack effective therapies and their biology is poorly understood. The term basal-like derives from studies demonstrating tumor gene expression profiles that include some transcripts characteristic of the basal cells of the normal adult human mammary gland and others associated with a subset of normal luminal cells. Elucidating the mechanisms responsible for the profiles of basal-like tumors is an active area of investigation. More refined molecular analysis of patients' samples and genetic strategies to produce breast cancers de novo from defined populations of normal mouse mammary cells have served as complementary approaches to identify relevant pathway alterations. However, both also have limitations. Here, we review some of the underlying reasons, including the unifying concept that some normal luminal cells have both luminal and basal features, as well as some emerging new avenues of investigation.
Collapse
|
35
|
Perone Y, Farrugia AJ, Rodríguez-Meira A, Győrffy B, Ion C, Uggetti A, Chronopoulos A, Marrazzo P, Faronato M, Shousha S, Davies C, Steel JH, Patel N, Del Rio Hernandez A, Coombes C, Pruneri G, Lim A, Calvo F, Magnani L. SREBP1 drives Keratin-80-dependent cytoskeletal changes and invasive behavior in endocrine-resistant ERα breast cancer. Nat Commun 2019; 10:2115. [PMID: 31073170 PMCID: PMC6509342 DOI: 10.1038/s41467-019-09676-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 03/22/2019] [Indexed: 01/03/2023] Open
Abstract
Approximately 30% of ERα breast cancer patients relapse with metastatic disease following adjuvant endocrine therapies. The connection between acquisition of drug resistance and invasive potential is poorly understood. In this study, we demonstrate that the type II keratin topological associating domain undergoes epigenetic reprogramming in aromatase inhibitors (AI)-resistant cells, leading to Keratin-80 (KRT80) upregulation. KRT80 expression is driven by de novo enhancer activation by sterol regulatory element-binding protein 1 (SREBP1). KRT80 upregulation directly promotes cytoskeletal rearrangements at the leading edge, increased focal adhesion and cellular stiffening, collectively promoting cancer cell invasion. Shearwave elasticity imaging performed on prospectively recruited patients confirms KRT80 levels correlate with stiffer tumors. Immunohistochemistry showed increased KRT80-positive cells at relapse and, using several clinical endpoints, KRT80 expression associates with poor survival. Collectively, our data uncover an unpredicted and potentially targetable direct link between epigenetic and cytoskeletal reprogramming promoting cell invasion in response to chronic AI treatment.
Collapse
MESH Headings
- Antineoplastic Agents, Hormonal/pharmacology
- Antineoplastic Agents, Hormonal/therapeutic use
- Aromatase Inhibitors/pharmacology
- Aromatase Inhibitors/therapeutic use
- Breast/pathology
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Breast Neoplasms/mortality
- Breast Neoplasms/pathology
- Cell Movement/drug effects
- Cell Movement/genetics
- Cytoskeleton/genetics
- Cytoskeleton/pathology
- Drug Resistance, Neoplasm/genetics
- Enhancer Elements, Genetic/genetics
- Epigenesis, Genetic
- Estrogen Receptor alpha/metabolism
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Keratins, Type II/genetics
- Keratins, Type II/metabolism
- MCF-7 Cells
- Neoplasm Invasiveness/genetics
- Neoplasm Invasiveness/pathology
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/mortality
- Neoplasm Recurrence, Local/pathology
- Prognosis
- Protein Domains/genetics
- Sterol Regulatory Element Binding Protein 1/metabolism
- Up-Regulation
Collapse
Affiliation(s)
- Ylenia Perone
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Aaron J Farrugia
- Division of Cancer Biology, Tumour Microenvironment Team, Institute of Cancer Research, London, UK
| | - Alba Rodríguez-Meira
- Department of Surgery and Cancer, Imperial College London, London, UK
- MRC Molecular Haematology Unit, Haematopoietic Stem Cell Biology Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Balázs Győrffy
- MTA TTK Lendület Cancer Biomarker Research Group, Institute of Enzymology, Hungarian Academy of Sciences, Budapest, Hungary
- 2nd Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Charlotte Ion
- Department of Surgery and Cancer, Imperial College London, London, UK
| | | | - Antonios Chronopoulos
- Faculty of Engineering, Department of Bioengineering, Imperial College London, London, UK
| | - Pasquale Marrazzo
- Department for Life Quality Studies, Alma Mater Studiorum, University of Bologna, Rimini, Italy
| | - Monica Faronato
- Department of Chemistry, Imperial College London, London, UK
| | - Sami Shousha
- Histopathology Department, Imperial College London, Charing Cross Hospital NHS Trust, London, UK
| | - Claire Davies
- ECMC Imperial College. Department of Surgery and Cancer, Imperial College London, London, UK
| | - Jennifer H Steel
- ECMC Imperial College. Department of Surgery and Cancer, Imperial College London, London, UK
| | - Naina Patel
- ECMC Imperial College. Department of Surgery and Cancer, Imperial College London, London, UK
| | | | - Charles Coombes
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Giancarlo Pruneri
- Pathology Department, Fondazione IRCCS Istituto Nazionale Tumori and University of Milan, School of Medicine, Milan, Italy
| | - Adrian Lim
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Fernando Calvo
- Division of Cancer Biology, Tumour Microenvironment Team, Institute of Cancer Research, London, UK.
- Instituto de Biomedicina y Biotecnologia de Cantabria, Santander, Spain.
| | - Luca Magnani
- Department of Surgery and Cancer, Imperial College London, London, UK.
| |
Collapse
|
36
|
Djomehri SI, Burman B, Gonzalez ME, Takayama S, Kleer CG. A reproducible scaffold-free 3D organoid model to study neoplastic progression in breast cancer. J Cell Commun Signal 2019; 13:129-143. [PMID: 30515709 PMCID: PMC6381373 DOI: 10.1007/s12079-018-0498-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 11/21/2018] [Indexed: 12/21/2022] Open
Abstract
While 3D cellular models are useful to study biological processes, gel-embedded organoids have large variability. This paper describes high-yield production of large (~1 mm diameter), scaffold-free, highly-spherical organoids in a one drop-one organoid format using MCF10A cells, a non-tumorigenic breast cell line. These organoids display a hollow lumen and secondary acini, and express mammary gland-specific and progenitor markers, resembling normal human breast acini. When subjected to treatment with TGF-β, the hypoxia-mimetic reagent CoCl2, or co-culture with mesenchymal stem/stromal cells (MSC), the organoids increase collagen I production and undergo large phenotypic and morphological changes of neoplastic progression, which were reproducible and quantifiable. Advantages of this scaffold-free, 3D breast organoid model include high consistency and reproducibility, ability to measure cellular collagen I production without noise from exogenous collagen, and capacity to subject the organoid to various stimuli from the microenvironment and exogenous treatments with precise timing without concern of matrix binding. Using this system, we generated organoids from primary metaplastic mammary carcinomas of MMTV-Cre;Ccn6fl/fl mice, which retained the high grade spindle cell morphology of the primary tumors. The platform is envisioned to be useful as a standardized 3D cellular model to study how microenvironmental factors influence breast tumorigenesis, and to potential therapeutics.
Collapse
Affiliation(s)
- Sabra I Djomehri
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Molecular and Cellular Pathology Training Program, University of Michigan, Ann Arbor, MI, 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Boris Burman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Maria E Gonzalez
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Shuichi Takayama
- Department of Biomedical Engineering, Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
| | - Celina G Kleer
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
37
|
Spatially correlated phenotyping reveals K5-positive luminal progenitor cells and p63-K5/14-positive stem cell-like cells in human breast epithelium. J Transl Med 2018; 98:1065-1075. [PMID: 29743728 DOI: 10.1038/s41374-018-0054-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 01/19/2018] [Accepted: 02/18/2018] [Indexed: 01/15/2023] Open
Abstract
Understanding the mechanisms regulating human mammary epithelium requires knowledge of the cellular constituents of this tissue. Different and partially contradictory definitions and concepts describing the cellular hierarchy of mammary epithelium have been proposed, including our studies of keratins K5 and/or K14 as markers of progenitor cells. Furthermore, we and others have suggested that the p53 homolog p63 is a marker of human breast epithelial stem cells. In this investigation, we expand our previous studies by testing whether immunohistochemical staining with monospecific anti-keratin antibodies in combination with an antibody against the stem cell marker p63 might help refine the different morphologic phenotypes in normal breast epithelium. We used in situ multilabel staining for p63, different keratins, the myoepithelial marker smooth muscle actin (SMA), the estrogen receptor (ER), and Ki67 to dissect and quantify the cellular components of 16 normal pre- and postmenopausal human breast epithelial tissue samples at the single-cell level. Importantly, we confirm the existence of K5+ only cells and suggest that they, in contrast to the current view, are key luminal precursor cells from which K8/18+ progeny cells evolve. These cells are further modified by the expression of ER and Ki67. We have also identified a population of p63+K5+ cells that are only found in nipple ducts. Based on our findings, we propose a new concept of the cellular hierarchy of human breast epithelium, including K5 luminal lineage progenitors throughout the ductal-lobular axis and p63+K5+ progenitors confined to the nipple ducts.
Collapse
|
38
|
Bonde GV, Yadav SK, Chauhan S, Mittal P, Ajmal G, Thokala S, Mishra B. Lapatinib nano-delivery systems: a promising future for breast cancer treatment. Expert Opin Drug Deliv 2018. [DOI: 10.1080/17425247.2018.1449832] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Gunjan Vasant Bonde
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Sarita Kumari Yadav
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
- Department of Pharmacy, Moti Lal Nehru Medical College, Allahabad, India
| | - Sheetal Chauhan
- Department of Pharmacology, Melaka Manipal Medical College, Manipal University, Manipal, India
| | - Pooja Mittal
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Gufran Ajmal
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Sathish Thokala
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Brahmeshwar Mishra
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| |
Collapse
|
39
|
Liu ZB, Wu J, Ping B, Feng LQ, Di GH, Lu JS, Shen KW, Shen ZZ, Shao ZM. Basal Cytokeratin Expression in Relation to Immunohistochemical and Clinical Characterization in Breast Cancer Patients with Triple Negative Phenotype. TUMORI JOURNAL 2018; 95:53-62. [DOI: 10.1177/030089160909500110] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Aims and background To evaluate the immunohistochemical characterization of CK5/6 and CK17 and whether the expression level of the two markers was correlated with clinical outcome or pathological feature in triple negative (ER-, PR-, HER-2-) patients with breast cancer. Methods and study design We carried out an immunohistochemical assay for CK5/6 and CK17 markers on formalin-fixed invasive carcinoma samples from 112 patients who were diagnosed between 2000 and 2002. All of them had an immunohistochemical triple negative status and follow-up information available. Results Of the 112 patients characterized by triple negative immunohistochemical status, 82 (73.2%) were disease free with no relapse or metastasis. In total, CK5/6 and CK17 were both determined positive in 33.9% (38/112) of the 112 tumor samples, and 46.4% (52/112) were regarded as positive for CK5/6 or CK17. The Kaplan-Meier curve showed that positive staining for CK5/6, CK17, or CK which means CK5/6 positive or CK17 positive, was associated with worse disease-free survival (P = 0.020, P = 0.032, P = 0.003), and positive staining for CK5/6 or CK was associated with worse overall survival (P = 0.027, P = 0.015). When we considered 91 patients whose pathological type was invasive ductal carcinoma, we found that there was also an association between CK5/6 or CK17 immunostaining and high grade (P = 0.030). In addition, these two markers were also associated with axillary lymph node status (P = 0.044). The Cox regression multiple-factor analysis showed that pathological stage, grade and expression of CK were the factors affecting both disease-free and overall survival, whereas age and menopausal status were independent factors affecting disease-free and overall survival, respectively. Conclusions Positive staining for CK5/6 or CK17 was associated with a worse prognosis, high tumor grade and positive axillary lymph nodes.
Collapse
Affiliation(s)
- Zhe-Bin Liu
- Department of Breast Surgery, Breast Cancer Institute, Fudan University, Shanghai
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, Republic of China
| | - Jiong Wu
- Department of Breast Surgery, Breast Cancer Institute, Fudan University, Shanghai
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, Republic of China
| | - Bo Ping
- Department of Pathology, Cancer Hospital/Cancer Institute, Fudan University, Shanghai
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, Republic of China
| | - Li-Qing Feng
- Department of Pathology, Cancer Hospital/Cancer Institute, Fudan University, Shanghai
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, Republic of China
| | - Gen-Hong Di
- Department of Breast Surgery, Breast Cancer Institute, Fudan University, Shanghai
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, Republic of China
| | - Jin-Song Lu
- Department of Breast Surgery, Breast Cancer Institute, Fudan University, Shanghai
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, Republic of China
| | - Kun-Wei Shen
- Department of Breast Surgery, Breast Cancer Institute, Fudan University, Shanghai
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, Republic of China
| | - Zhen-Zhou Shen
- Department of Breast Surgery, Breast Cancer Institute, Fudan University, Shanghai
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, Republic of China
| | - Zhi-Min Shao
- Department of Breast Surgery, Breast Cancer Institute, Fudan University, Shanghai
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, Republic of China
| |
Collapse
|
40
|
Expression of GHRH-R, a Potentially Targetable Biomarker, in Triple-negative Breast Cancer. Appl Immunohistochem Mol Morphol 2017; 26:1-5. [PMID: 29206714 DOI: 10.1097/pai.0000000000000622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
PURPOSE Growth hormone-releasing hormone (GHRH) has been shown to modify the growth behavior of many cancers, including breast. GHRH is produced by tumor cells, acts in an autocrine/paracrine manner, and requires the presence of GHRH receptor (GHRH-R) on the tumor cells to exert its effects. GHRH activity can be effectively blocked by synthetic antagonists of its receptor and hence, the expression of GHRH-R by tumor cells could serve as a predictor of response to GHRH-R antagonist therapy. In this study, we investigated the expression of GHRH-R in triple-negative breast cancers (TNBC). As TNBCs are morphologically and immunophenotypically heterogenous, the staining results were also correlated with the histologic subtypes of these tumors. MATERIALS AND METHODS On the basis of histomorphology and immunophenotype, 134 cases of primary TNBCs were further subdivided into medullary, metaplastic, apocrine, and invasive ductal carcinomas of no special type (IDC-NST). Immunohistochemistry for GHRH-R was performed on paraffin sections and the staining results were assessed semiquantitatively as negative, low expression, moderate, and high expression. RESULTS Of the 134 TNBCs, 85 were classified as IDC-NST, 25 as metaplastic, 16 as medullary, and 8 as apocrine carcinoma. Overall, positive reaction for GHRH-R was seen in 77 (57%) of tumors including 66 (77.6%) of IDC-NST. All medullary carcinomas were negative for GHRH-R and, with the exception of 1 case with low expression, none of the metaplastic carcinomas expressed GHRH-R (P<0.005). CONCLUSIONS A considerable number of TNBCs are positive for GHRH-R as a predictor of potential response to anti-GHRH-R treatment. This expression however, varies considerably between histologic subtypes of triple-negative breast cancers. Although most medullary and metaplastic carcinomas do not express GHRH-R, three fourths of the IDC-NST show a positive reaction. Testing for GHRH-R expression is therefore advisable if anti-GHRH-R therapy is being considered.
Collapse
|
41
|
Obesity reversibly depletes the basal cell population and enhances mammary epithelial cell estrogen receptor alpha expression and progenitor activity. Breast Cancer Res 2017; 19:128. [PMID: 29187227 PMCID: PMC5707907 DOI: 10.1186/s13058-017-0921-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 11/15/2017] [Indexed: 12/15/2022] Open
Abstract
Background Obesity is correlated with an increased risk for developing postmenopausal breast cancer. Since obesity rates continue to rise worldwide, it is important to understand how the obese microenvironment influences normal mammary tissue to increase breast cancer risk. We hypothesized that obesity increases the proportion of luminal progenitor cells, which are thought to be the cells of origin for the most common types of breast cancer, potentially leading to an increased risk for breast cancer. Methods To study the obese microenvironment within the mammary gland, we used a high-fat diet mouse model of obesity and human breast tissue from reduction mammoplasty surgery. We identified changes in breast epithelial cell populations using flow cytometry for cell surface markers, in vitro functional assays and expression of markers on breast tissue sections. Results In both obese female mice and women, mammary epithelial cell populations demonstrated significant decreases in basal/myoepithelial cells, using either flow cytometry or cell-type-specific markers (SMA and p63). Estrogen receptor alpha (ERα) expression was significantly increased in luminal cells in obese mammary tissue, compared with control mice or breast tissue from lean women. Functional assays demonstrated significantly enhanced mammary epithelial progenitor activity in obese mammary epithelial cells and elevated numbers of ERα-positive epithelial cells that were co-labeled with markers of proliferation. Weight loss in a group of obese mice reversed increases in progenitor activity and ERα expression observed in obese mammary tissue. Conclusions Obesity enhances ERα-positive epithelial cells, reduces the number of basal/myoepithelial cells, and increases stem/progenitor activity within normal mammary tissue in both women and female mice. These changes in epithelial cell populations induced by obesity are reversible with weight loss. Our findings support further studies to examine how obesity-induced changes in stem/progenitor cells impact breast tumor incidence and histologic tumor types. Electronic supplementary material The online version of this article (doi:10.1186/s13058-017-0921-7) contains supplementary material, which is available to authorized users.
Collapse
|
42
|
Ricciardelli C, Lokman NA, Pyragius CE, Ween MP, Macpherson AM, Ruszkiewicz A, Hoffmann P, Oehler MK. Keratin 5 overexpression is associated with serous ovarian cancer recurrence and chemotherapy resistance. Oncotarget 2017; 8:17819-17832. [PMID: 28147318 PMCID: PMC5392289 DOI: 10.18632/oncotarget.14867] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 01/16/2017] [Indexed: 02/06/2023] Open
Abstract
This study investigated the clinical significance of keratin 5 and 6 expression in serous ovarian cancer progression and chemotherapy resistance. KRT5 and KRT6 (KRT6A, KRT6B & KRT6C) gene expression was assessed in publically available serous ovarian cancer data sets, ovarian cancer cell lines and primary serous ovarian cancer cells. Monoclonal antibodies which detect both K5/6 or only K5 were used to assess protein expression in ovarian cancer cell lines and a cohort of high grade serous ovarian carcinomas at surgery (n = 117) and after neoadjuvant chemotherapy (n = 21). Survival analyses showed that high KRT5 mRNA in stage III/IV serous ovarian cancers was significantly associated with reduced progression-free (HR 1.38, P < 0.0001) and overall survival (HR 1.28, P = 0.013) whilst high KRT6 mRNA was only associated with reduced progression-free survival (HR 1.2, P = 0.031). Both high K5/6 (≥ 10%, HR 1.78 95% CI; 1.03−2.65, P = 0.017) and high K5 (≥ 10%, HR 1.90, 95% CI; 1.12−3.19, P = 0.017) were associated with an increased risk of disease recurrence. KRT5 but not KRT6C mRNA expression was increased in chemotherapy resistant primary serous ovarian cancer cells compared to chemotherapy sensitive cells. The proportion of serous ovarian carcinomas with high K5/6 or high K5 immunostaining was significantly increased following neoadjuvant chemotherapy. K5 can be used to predict serous ovarian cancer prognosis and identify cancer cells that are resistant to chemotherapy. Developing strategies to target K5 may therefore improve serous ovarian cancer survival.
Collapse
Affiliation(s)
- Carmela Ricciardelli
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, University of Adelaide, Adelaide, 5000, South Australia, Australia
| | - Noor A Lokman
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, University of Adelaide, Adelaide, 5000, South Australia, Australia
| | - Carmen E Pyragius
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, University of Adelaide, Adelaide, 5000, South Australia, Australia
| | - Miranda P Ween
- Lung Research Laboratory, Hanson Institute, Adelaide, South Australia, Australia.,Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, 5000, South Australia, Australia
| | - Anne M Macpherson
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, University of Adelaide, Adelaide, 5000, South Australia, Australia
| | - Andrew Ruszkiewicz
- Centre of Cancer Biology, University of South Australia and Department of Anatomical Pathology, SA Pathology, Adelaide, 5000, South Australia, Australia
| | - Peter Hoffmann
- Adelaide Proteomics Centre, School of Biological Sciences, University of Adelaide, Adelaide, 5005, South Australia, Australia
| | - Martin K Oehler
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, University of Adelaide, Adelaide, 5000, South Australia, Australia.,Department of Gynaecological Oncology, Royal Adelaide Hospital, Adelaide, 5000, South Australia, Australia
| |
Collapse
|
43
|
Zeng Z, Hou CJ, Hu QH, Liu Y, Wang C, Wei R, Fan XM. Mammography and ultrasound effective features in differentiating basal-like and normal-like subtypes of triple negative breast cancer. Oncotarget 2017; 8:79670-79679. [PMID: 29108347 PMCID: PMC5668080 DOI: 10.18632/oncotarget.19053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 06/26/2017] [Indexed: 12/31/2022] Open
Abstract
The aim of our study was to find effective features of mammography and ultrasound in differentiating Basal-like breast cancer (BBC) and Normal-like breast cancer (NBC), two subtypes of triple negative breast cancer (TNBC). From January 2014 to March 2017, we retrospectively reviewed 91 patients who were pathologically confirmed as TNBC. According to immunohistochemical cytokeratin 5/6 (CK5/6) and Epidermal Growth Factor Receptor (EGFR), TNBCs were classified into BBCs group and NBCs group. Both CK5/6 and EGFR were negative defined to be NBC, whereas if any of CK5/6 or EGFR was positive then defined as BBC. BBCs group concluded 65 (71.4%) cases and NBCs group concluded26 (28.6%) cases. Ultrasound images and mammograms were reevaluated by breast imaging experts according to the breast imaging reporting and data system (BI-RADS) 4th edition. On mammography, masses margins had significant differences between BBCs group and NBCs group (P = 0.024). Most BBCs margins exhibited microlobulated (30/64, 46.9%) or spiculated (25/64, 39.0%), whereas most NBCs margins exhibited microlobulated (17/23, 73.9%). On ultrasound, BBCs were more frequently to present as larger than 20mm lesions (52/65, 80.0%) and more likely to have angular or spiculated margins (35/65, 53.8%), additionally, compared with NBCs, BBCs were less likely to have calcification (1/65, 1.5%). Other mammography and ultrasound features showed no significant differences between the two groups. In conclusion, we have found some effective features of mammography and ultrasound that could be helpful in differentiating BBC and NBC, which will provide some useful references for clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Zeng Zeng
- Department of Ultrasound, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - Chun Jie Hou
- Department of Ultrasound, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - Qiao Hong Hu
- Department of Ultrasound, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - Ying Liu
- Department of Ultrasound, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - Ceng Wang
- Department of Ultrasound, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - Ran Wei
- Department of Ultrasound, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - Xiao Ming Fan
- Department of Ultrasound, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
44
|
Saad E, Milley K, Al-Khan A, Nimmo J, Bacci B, Tayebi M, Day M, Richardson S, Danks J. Canine Mixed Mammary Tumour as a Model for Human Breast Cancer with Osseous Metaplasia. J Comp Pathol 2017; 156:352-365. [DOI: 10.1016/j.jcpa.2017.03.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 02/17/2017] [Accepted: 03/27/2017] [Indexed: 01/04/2023]
|
45
|
Carter EP, Gopsill JA, Gomm JJ, Jones JL, Grose RP. A 3D in vitro model of the human breast duct: a method to unravel myoepithelial-luminal interactions in the progression of breast cancer. Breast Cancer Res 2017; 19:50. [PMID: 28427436 PMCID: PMC5399380 DOI: 10.1186/s13058-017-0843-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 04/11/2017] [Indexed: 11/10/2022] Open
Abstract
Background 3D modelling fulfils a critical role in research, allowing for complex cell behaviour and interactions to be studied in physiomimetic conditions. With tissue banks becoming established for a number of cancers, researchers now have access to primary patient cells, providing the perfect building blocks to recreate and interrogate intricate cellular systems in the laboratory. The ducts of the human breast are composed of an inner layer of luminal cells supported by an outer layer of myoepithelial cells. In early-stage ductal carcinoma in situ, cancerous luminal cells are confined to the ductal space by an intact myoepithelial layer. Understanding the relationship between myoepithelial and luminal cells in the development of cancer is critical for the development of new therapies and prognostic markers. This requires the generation of new models that allows for the manipulation of these two cell types in a physiological setting. Methods Using access to the Breast Cancer Now Tissue Bank, we isolated pure populations of myoepithelial and luminal cells from human reduction mammoplasty specimens and placed them into 2D culture. These cells were infected with lentiviral particles encoding either fluorescent proteins, to facilitate cell tracking, or an inducible human epidermal growth factor receptor 2 (HER2) expression construct. Myoepithelial and luminal cells were then recombined in collagen gels, and the resulting cellular structures were analysed by confocal microscopy. Results Myoepithelial and luminal cells isolated from reduction mammoplasty specimens can be grown separately in 2D culture and retain their differentiated state. When recombined in collagen gels, these cells reform into physiologically reflective bilayer structures. Inducible expression of HER2 in the luminal compartment, once the bilayer has formed, leads to robust luminal filling, recapitulating ductal carcinoma in situ, and can be blocked with anti-HER2 therapies. Conclusions This model allows for the interaction between myoepithelial and luminal cells to be investigated in an in-vitro environment and paves the way to study early events in breast cancer development with the potential to act as a powerful drug discovery platform. Electronic supplementary material The online version of this article (doi:10.1186/s13058-017-0843-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Edward P Carter
- Centre for Tumour Biology, Barts Cancer Institute - a Cancer Research UK Centre of Excellence, Queen Mary University of London, London, EC1M 6BQ, UK.
| | - James A Gopsill
- Department of Mechanical Engineering, University of Bristol, Bristol, BS8 1TR, UK
| | - Jennifer J Gomm
- Centre for Tumour Biology, Barts Cancer Institute - a Cancer Research UK Centre of Excellence, Queen Mary University of London, London, EC1M 6BQ, UK
| | - J Louise Jones
- Centre for Tumour Biology, Barts Cancer Institute - a Cancer Research UK Centre of Excellence, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Richard P Grose
- Centre for Tumour Biology, Barts Cancer Institute - a Cancer Research UK Centre of Excellence, Queen Mary University of London, London, EC1M 6BQ, UK.
| |
Collapse
|
46
|
Keating P, Cambrosio A, Nelson NC. "Triple negative breast cancer": Translational research and the (re)assembling of diseases in post-genomic medicine. STUDIES IN HISTORY AND PHILOSOPHY OF BIOLOGICAL AND BIOMEDICAL SCIENCES 2016; 59:20-34. [PMID: 27235853 DOI: 10.1016/j.shpsc.2016.05.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 05/09/2016] [Accepted: 05/12/2016] [Indexed: 06/05/2023]
Abstract
The paper examines the debate about the nature and status of "Triple-negative breast cancer", a controversial biomedical entity whose existence illustrates a number of features of post-genomic translational research. The emergence of TNBC is intimately linked to the rise of molecular oncology, and, more generally, to the changing configuration of the life sciences at the turn of the new century. An unprecedented degree of integration of biological and clinical practices has led to the proliferation of bio-clinical entities emerging from translational research. These translations take place between platforms rather than between clinical and laboratory settings. The complexity and heterogeneity of TNBC, its epistemic and technical, biological and clinical dualities, result from its multiple instantiations via different platforms, and from the uneven distribution of biological materials, techniques, and objects across clinical research settings. The fact that TNBC comes in multiple forms, some of which seem to be incompatible or, at least, only partially overlapping, appears to be less a threat to the whole endeavor, than an aspect of an ongoing translational research project. Discussions of translational research that rest on a distinction between basic research and its applications fail to capture the dynamics of this new domain of activity, insofar as application is built-in from the very beginning in the bio-clinical entities that emerge from the translational research domain.
Collapse
Affiliation(s)
- Peter Keating
- Department of History, University of Quebec at Montreal, Canada
| | - Alberto Cambrosio
- Department of Social Studies of Medicine, McGill University, Canada.
| | - Nicole C Nelson
- Department of the History of Science, University of Wisconsin-Madison, United States
| |
Collapse
|
47
|
Pourteimoor V, Mohammadi-Yeganeh S, Paryan M. Breast cancer classification and prognostication through diverse systems along with recent emerging findings in this respect; the dawn of new perspectives in the clinical applications. Tumour Biol 2016; 37:14479-14499. [DOI: 10.1007/s13277-016-5349-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 09/06/2016] [Indexed: 01/10/2023] Open
|
48
|
Baker LA, Holliday H, Swarbrick A. ID4 controls luminal lineage commitment in normal mammary epithelium and inhibits BRCA1 function in basal-like breast cancer. Endocr Relat Cancer 2016; 23:R381-92. [PMID: 27412917 DOI: 10.1530/erc-16-0196] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 07/13/2016] [Indexed: 12/21/2022]
Abstract
Inhibitor of differentiation (ID) proteins are key regulators of development and tumorigenesis. One member of this family, ID4, controls lineage commitment during mammary gland development by acting upstream of key developmental pathways. Recent evidence suggests an emerging role for ID4 as a lineage-dependent proto-oncogene that is overexpressed and amplified in a subset of basal-like breast cancers (BLBCs), conferring poor prognosis. Several lines of evidence suggest ID4 may suppress BRCA1 function in BLBC and in doing so, define a subset of BLBC patients who may respond to therapies traditionally used in BRCA1-mutant cancers. This review highlights recent advances in our understanding of the requirement for ID4 in mammary lineage commitment and the role for ID4 in BLBC. We address current shortfalls in this field and identify important areas of future research.
Collapse
Affiliation(s)
- Laura A Baker
- The Kinghorn Cancer Centre and Cancer Research DivisionGarvan Institute of Medical Research, Darlinghurst, New South Wales, Australia St Vincent's Clinical SchoolFaculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Holly Holliday
- The Kinghorn Cancer Centre and Cancer Research DivisionGarvan Institute of Medical Research, Darlinghurst, New South Wales, Australia St Vincent's Clinical SchoolFaculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Alexander Swarbrick
- The Kinghorn Cancer Centre and Cancer Research DivisionGarvan Institute of Medical Research, Darlinghurst, New South Wales, Australia St Vincent's Clinical SchoolFaculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
49
|
Eivani D, Mortazavi P. The relationship between basal and luminal cytokeratins with histopathologic characteristics of canine mammary gland cancer. Pol J Vet Sci 2016; 19:261-9. [PMID: 27487499 DOI: 10.1515/pjvs-2016-0033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Neoplasia occurs mostly in mammary glands in female dogs and mammary gland cancer is one of the causes of death in these animals cytokeratins are one of the most important of tumor markers for identification of tumor prognosis. In this study, 120 canine malignant tumor samples of mammary glands were studied. From each sample, a section was taken for hematoxylin-eosin staining and two sections for immunohistochemical staining of markers CK5/6 and CK7. Histopathology slides was evaluated by light microscope. The results show that the presence of markers CK7 and CK5/6 had no significant relationship with tumor grade and type (p<0.05). However, it seems that unlike humans, CK5/6 and CK7 is not an independent prognostic factor in canine mammary gland tumors.
Collapse
|
50
|
Streckfus CF, Bigler L. A Catalogue of Altered Salivary Proteins Secondary to Invasive Ductal Carcinoma: A Novel In Vivo Paradigm to Assess Breast Cancer Progression. Sci Rep 2016; 6:30800. [PMID: 27477923 PMCID: PMC4967869 DOI: 10.1038/srep30800] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 07/11/2016] [Indexed: 12/13/2022] Open
Abstract
The objective of this manuscript is to introduce a catalogue of salivary proteins that are altered secondary to carcinoma of the breast. The catalogue of salivary proteins is a compilation of twenty years of research by the authors and consists of 233 high and low abundant proteins which have been identified by LC-MS/MS mass spectrometry, 2D-gel analysis and by enzyme-linked immunosorbent assay. The body of research suggests that saliva is a fluid suffused with solubilized by-products of oncogenic expression and that these proteins may be useful in the study of breast cancer progress, treatment efficacy and the tailoring of individualized patient care.
Collapse
Affiliation(s)
- Charles F. Streckfus
- University of Texas School of Dentistry at Houston Department of Diagnostic and Behavioral Sciences Behavioral & Biomedical Sciences Building, Rm. 5322 Houston, Texas 77054, USA
| | - Lenora Bigler
- University of Texas School of Dentistry at Houston Department of Diagnostic and Behavioral Sciences Behavioral & Biomedical Sciences Building, Rm. 5322 Houston, Texas 77054, USA
| |
Collapse
|