1
|
Yu X, Tian Y, Zhang R, Yang Y. Decoding the Tumor Microenvironment of Myoepithelial Cells in Triple-Negative Breast Cancer Through Single-Cell and Transcriptomic Sequencing and Establishing a Prognostic Model Based on Key Myoepithelial Cell Genes. Int J Genomics 2025; 2025:6454413. [PMID: 40365116 PMCID: PMC12074843 DOI: 10.1155/ijog/6454413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Accepted: 04/09/2025] [Indexed: 05/15/2025] Open
Abstract
Background: Triple-negative breast cancer (TNBC) is an aggressive subtype with high malignancy, rapid progression, and a poor 5-year survival rate of ~77%. Due to the lack of targeted therapies, treatment options are limited, highlighting the urgent need for novel therapeutic strategies. Myoepithelial cells (MECs) in the tumor microenvironment may significantly influence TNBC development and progression. Methods: This study used single-cell RNA sequencing to compare the MEC gene expression in the normal versus TNBC tissues. TNBC-associated MECs showed increased expression of proliferation- and immune-related genes (e.g., FDCSP, KRT14, and KRT17) and decreased expression of inflammatory and extracellular matrix-related genes (e.g., CXCL8, SRGN, and DCN). Copy number variation and pseudotime analyses revealed genomic alterations and phenotypic dynamics in MECs. A CoxBoost-based prognostic model was developed and validated across 20 survival cohorts, integrating immune profiling, pathway enrichment, and drug sensitivity analyses. Mendelian randomization identified TPD52 as a TNBC risk-associated gene. siRNA knockdown of TPD52 was performed in TNBC cell lines to evaluate its effects on proliferation and migration. Results: TNBC MECs displayed significant changes in the gene expression and genomic integrity, impacting immune responses and tumor invasion. The prognostic model effectively predicted 1-, 3-, and 5-year survival outcomes, stratifying high-risk patients with enriched cell cycle and DNA replication pathways, reduced immune checkpoint expression, and chemotherapy resistance. TPD52 was identified as a tumor-promoting gene, and its knockdown suppressed TNBC cell proliferation and migration. Conclusion: This study highlights MECs' role in TNBC progression, provides a CoxBoost prognostic model for personalized treatment, and identifies TPD52 as a potential therapeutic target for TNBC intervention.
Collapse
Affiliation(s)
- Xiaocheng Yu
- Department of Thyroid and Breast Surgery, Wuhan No.1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ye Tian
- Department of Thyroid and Breast Surgery, Wuhan No.1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Zhang
- Department of Thyroid and Breast Surgery, Wuhan No.1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Yang
- Department of Thyroid and Breast Surgery, Wuhan No.1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 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
|
Watson KL, Moorehead RA. Transgenic overexpression of the miR-200b/200a/429 cluster prevents mammary tumor initiation in Neu/Erbb2 transgenic mice. Int J Cancer 2025; 156:993-1004. [PMID: 39369448 DOI: 10.1002/ijc.35211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/26/2024] [Accepted: 09/19/2024] [Indexed: 10/08/2024]
Abstract
Although significant progress in the treatment of breast cancer has been achieved, toxic therapies would not be required if breast cancer could be prevented from developing in the first place. While breast cancer prevention is difficult to study in humans due to long disease latency and stochastic cancer development, transgenic mouse models with 100% incidence and defined mammary tumor onset, provide excellent models for tumor prevention studies. In this study, we used Neu/Erbb2 transgenic mice (MTB-TAN) as a model of human HER2+ breast cancer to investigate whether a family of microRNAs, known as the miR-200 family, can prevent mammary tumor development. Overexpression of Neu induced palpable mammary tumors in 100% of the mice within 38 days of Neu overexpression. When the miR-200b/200a/429 cluster was co-overexpressed with Neu in the same mammary epithelial cells (MTB-TANba429 mice), the miR-200b/200a/429 cluster prevented Neu from inducing mammary epithelial hyperplasia and mammary tumor development. RNA sequencing revealed alterations in the extracellular matrix of the mammary gland and a decrease in stromal cells including myoepithelial cells in Neu transgenic mice. Immunohistochemistry for smooth muscle actin confirmed that mammary epithelial cells in control and MTB-TANba429 mice were surrounded by a layer of myoepithelial cells and these myoepithelial cells were lost in MTB-TAN mice with hyperplasia. Thus, we have shown for the first time that elevated expression of miR-200 family members in mammary epithelial cells can completely prevent mammary tumor development in Neu transgenic mice possibly through regulating myoepithelial cells.
Collapse
Affiliation(s)
- Katrina L Watson
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, USA
| | - Roger A Moorehead
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, USA
| |
Collapse
|
4
|
Roozitalab MR, Prekete N, Allen M, Grose RP, Louise Jones J. The Microenvironment in DCIS and Its Role in Disease Progression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:211-235. [PMID: 39821028 DOI: 10.1007/978-3-031-70875-6_12] [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: 01/19/2025]
Abstract
Ductal carcinoma in situ (DCIS) accounts for ~20% of all breast cancer diagnoses but whilst known to be a precursor of invasive breast cancer (IBC), evidence suggests only one in six patients will ever progress. A key challenge is to distinguish between those lesions that will progress and those that will remain indolent. Molecular analyses of neoplastic epithelial cells have not identified consistent differences between lesions that progressed and those that did not, and this has focused attention on the tumour microenvironment (ME).The DCIS ME is unique, complex and dynamic. Myoepithelial cells form the wall of the ductal-lobular tree and exhibit broad tumour suppressor functions. However, in DCIS they acquire phenotypic changes that bestow them with tumour promoter properties, an important evolution since they act as the primary barrier for invasion. Changes in the peri-ductal stromal environment also arise in DCIS, including transformation of fibroblasts into cancer-associated fibroblasts (CAFs). CAFs orchestrate other changes in the stroma, including the physical structure of the extracellular matrix (ECM) through altered protein synthesis, as well as release of a plethora of factors including proteases, cytokines and chemokines that remodel the ECM. CAFs can also modulate the immune ME as well as impact on tumour cell signalling pathways. The heterogeneity of CAFs, including recognition of anti-tumourigenic populations, is becoming evident, as well as heterogeneity of immune cells and the interplay between these and the adipocyte and vascular compartments. Knowledge of the impact of these changes is more advanced in IBC but evidence is starting to accumulate for a role in DCIS. Detailed in vitro, in vivo and tissue studies focusing on the interplay between DCIS epithelial cells and the ME should help to define features that can better predict DCIS behaviour.
Collapse
Affiliation(s)
- Mohammad Reza Roozitalab
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK
| | - Niki Prekete
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK
| | - Michael Allen
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK
| | - Richard P Grose
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK
| | - J Louise Jones
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK.
| |
Collapse
|
5
|
Li A, Ruan M, Fei X, Xu H, Deng S, Bi R, Yang W, Dong L. Altered cytokeratin 5 expression in breast lobular myoepithelial cells. J Clin Pathol 2024; 77:536-543. [PMID: 37116947 DOI: 10.1136/jcp-2023-208835] [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: 02/11/2023] [Accepted: 04/11/2023] [Indexed: 04/30/2023]
Abstract
AIMS Cytokeratin 5 (CK5) is a surrogate maker of progenitor cells and early glandular and myoepithelial cells (MECs) in the breast, and CK5 expression in breast MECs varies from ducts to lobules, and from normal to diseased tissue. However, the mechanisms underlying immunophenotypic alterations of CK5 expression in MECs remain unclear. METHODS CK5 expression in MECs of 20 normal breast samples, 58 ductal carcinoma in situ (DCIS; including 21 DCIS with extensive lobular involvement), 11 atypical ductal hyperplasia (ADH), 18 non-invasive lobular neoplasia consisting of 11 atypical lobular hyperplasia (ALH) and 7 lobular carcinoma in situ (LCIS), 20 cystic lobules and 10 usual ductal hyperplasia (UDH) involving lobules were observed to evaluate the effects of contact with benign hyperplastic or cancerous luminal cells and pressure of dilated glands on CK5 expression. RESULTS CK5 expression in normal ductal MECs was exclusively positive, whereas most normal lobular MECs were negative. In DCIS, cancerous ducts were primarily surrounded by CK5-positive MECs (91.0%), as were lobular acini involved by DCIS (89.2%), while the remaining normal acini maintained CK5-negative. CK5-positive MECs were found in 57.5% of acini in ALH and were more prevalent in LCIS (70.7%). CK5 expression was occasionally positive in both cystic lobules (16.7%) and lobules involved by UDH (14.3%), while an increase of CK5-positive MECs was found in ADH (38.2%). CONCLUSIONS These results suggest that CK5 expression in lobular MECs may be altered by contact with cancerous luminal cells rather than benign hyperplastic luminal cells or pressure from dilated glands.
Collapse
Affiliation(s)
- Anqi Li
- Department of Pathology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Miao Ruan
- Department of Pathology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Xiaochun Fei
- Department of Pathology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Haimin Xu
- Department of Pathology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Shijie Deng
- Department of Pathology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Rui Bi
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Wentao Yang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Lei Dong
- Department of Pathology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| |
Collapse
|
6
|
Jariani A, Kakroodi ST, Arabfard M, Jamialahmadi T, Rahimi M, Sahebkar A. Identification of Key Genes in Angiogenesis of Breast and Prostate Cancers in the Context of Different Cell Types. Curr Med Chem 2024; 31:1595-1605. [PMID: 36999716 DOI: 10.2174/0929867330666230331101458] [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: 10/13/2022] [Revised: 12/27/2022] [Accepted: 02/03/2023] [Indexed: 04/01/2023]
Abstract
INTRODUCTION Angiogenesis involves the development of new blood vessels. Biochemical signals start this process in the body, which is followed by migration, growth, and differentiation of endothelial cells that line the inside wall of blood vessels. This process is vital for the growth of cancer cells and tumors. MATERIALS AND METHODS We started our analysis by composing a list of genes that have a validated impact in humans with respect to angiogenesis-related phenotypes. Here, we have investigated the expression patterns of angiogenesis-related genes in the context of previously published single-cell RNA-Seq data from prostate and breast cancer samples. RESULTS Using a protein-protein interaction network, we showed how different modules of angiogenesis-related genes are overexpressed in different cell types. In our results, genes, such as ACKR1, AQP1, and EGR1, showed a strong cell type-dependent overexpression pattern in the two investigated cancer types, which can potentially be helpful in the diagnosis and follow-up of patients with prostate and breast cancer. CONCLUSION Our work demonstrates how different biological processes in distinct cell types contribute to the angiogenesis process, which can provide clues regarding the potential application of targeted inhibition of the angiogenesis process.
Collapse
Affiliation(s)
- Abbas Jariani
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | - Masoud Arabfard
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Tannaz Jamialahmadi
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Rahimi
- Clinical care and Health Promotion Research Center, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Medicine, The University of Western Australia, Perth, Australia
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
7
|
Gibson SV, Roozitalab RM, Allen MD, Jones JL, Carter EP, Grose RP. Everybody needs good neighbours: the progressive DCIS microenvironment. Trends Cancer 2023; 9:326-338. [PMID: 36739265 DOI: 10.1016/j.trecan.2023.01.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 02/05/2023]
Abstract
Ductal carcinoma in situ (DCIS) is a pre-invasive form of breast cancer where neoplastic luminal cells are confined to the ductal tree. While as many as 70% of DCIS cases will remain indolent, most women are treated with surgery, often combined with endocrine and radiotherapies. Overtreatment is therefore a major issue, demanding new methods to stratify patients. Somewhat paradoxically, the neoplastic cells in DCIS are genetically comparable to those in invasive disease, suggesting the tumour microenvironment is the driving force for progression. Clinical and mechanistic studies highlight the complex DCIS microenvironment, with multiple cell types competing to regulate progression. Here, we examine recent studies detailing distinct aspects of the DCIS microenvironment and discuss how these may inform more effective care.
Collapse
Affiliation(s)
- Shayin V Gibson
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Reza M Roozitalab
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Michael D Allen
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - J Louise Jones
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Edward P Carter
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK.
| | - Richard P Grose
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK.
| |
Collapse
|
8
|
Sourouni M, Opitz C, Radke I, Kiesel L, Tio J, Götte M, von Wahlde M. Establishment of a
3D
co‐culture model to investigate the role of primary fibroblasts in ductal carcinoma in situ of the breast. Cancer Rep (Hoboken) 2022; 6:e1771. [PMID: 36534078 PMCID: PMC10075300 DOI: 10.1002/cnr2.1771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 12/01/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Ductal carcinoma in situ (DCIS) is a precursor form of breast cancer. 13%-50% of these lesions will progress to invasive breast cancer, but the individual progression risk cannot be estimated. Therefore, all patients receive the same therapy, resulting in potential overtreatment of a large proportion of patients. AIMS The role of the tumor microenvironment (TME) and especially of fibroblasts appears to be critical in DCIS development and a better understanding of their role may aid individualized treatment. METHODS AND RESULTS Primary fibroblasts isolated from benign or malignant punch biopsies of the breast and MCF10DCIS.com cells were seeded in a 3D cell culture system. The fibroblasts were cultured in a type I collagen layer beneath a Matrigel layer with MCF10DCIS.com cells. Dye-quenched (DQ) fluorescent collagen I and IV were used in collagen and Matrigel layer respectively to demonstrate proteolysis. Confocal microscopy was performed on day 2, 7, and 14 to reveal morphological changes, which could indicate the transition to an invasive phenotype. MCF10DCIS.com cells form smooth, round spheroids in co-culture with non-cancer associated fibroblasts (NAFs). Spheroids in co-culture with tumor-associated fibroblasts (TAFs) appear irregularly shaped and with an uneven surface; similar to spheroids formed from invasive cells. Therefore, these morphological changes represent the progression of an in situ to an invasive phenotype. In addition, TAFs show a higher proteolytic activity compared to NAFs. The distance between DCIS cells and fibroblasts decreases over time. CONCLUSION The TAFs seem to play an important role in the progression of DCIS to invasive breast cancer. The better characterization of the TME could lead to the identification of DCIS lesions with high or low risk of progression. This could enable personalized oncological therapy, prevention of overtreatment and individualized hormone replacement therapy after DCIS.
Collapse
Affiliation(s)
- Marina Sourouni
- Department of Obstetrics and Gynecology, Breast Center University Hospital Münster Münster Germany
| | - Carl Opitz
- Department of Obstetrics and Gynecology, Breast Center University Hospital Münster Münster Germany
| | - Isabel Radke
- Department of Obstetrics and Gynecology, Breast Center University Hospital Münster Münster Germany
| | - Ludwig Kiesel
- Department of Obstetrics and Gynecology, Breast Center University Hospital Münster Münster Germany
| | - Joke Tio
- Department of Obstetrics and Gynecology, Breast Center University Hospital Münster Münster Germany
| | - Martin Götte
- Department of Obstetrics and Gynecology, Breast Center University Hospital Münster Münster Germany
| | - Marie‐Kristin von Wahlde
- Department of Obstetrics and Gynecology, Breast Center University Hospital Münster Münster Germany
| |
Collapse
|
9
|
Shams A. Re-evaluation of the myoepithelial cells roles in the breast cancer progression. Cancer Cell Int 2022; 22:403. [PMID: 36510219 PMCID: PMC9746125 DOI: 10.1186/s12935-022-02829-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Over the past decades, luminal epithelial cell lineage has gained considerable attraction as the functionally milk-secreting units and as the most fruitful acreage for breast cancer launching. Recognition of the effective involvement of the myoepithelial cells in mammary gland development and in hampering tumorigenesis has renewed the interest in investigating the biological roles of this second main mammary lineage. The human breast is made up of an extensively branching ductal system intervening by copious lobular units. The ductal system is coated by a chain of luminal epithelial cells (LECs) situated on a layer of myoepithelial cells (MECs) and encompassed by a distinguished basement membrane. Ductal contractility during lactation is a well-known function delivered by the MECs however this is not the only assignment mediated by these cellular populations. It has been well appreciated that the MECs exhibit a natural paracrine power in defeating cancer development and advancement. MECs were found to express numerous proteinase inhibitors, anti-angiogenic factors, and tumour suppressors proteins. Additionally, MECs contributed effectively to maintaining the right luminal cells' polarization and further separating them from the adjacent stroma by making an integrated fence. Indeed, disruption of the MECs layer was reported to facilitate the invasion of the cancer cells to the surrounding stroma. Nonetheless, MECs were also found to exhibit cancer-promoting effects and provoke tumour invasion and dissemination by displaying distinct cancer chemokines. Herein in this review, we aimed to address the roles delivered by MECs in breast cancer progression and decipher the molecular mechanisms regulating proper MECs' physiology, integrity, and terminal differentiation.
Collapse
Affiliation(s)
- Anwar Shams
- grid.412895.30000 0004 0419 5255Department of Pharmacology, College of Medicine, Taif University, P.O. BOX 11099, Taif, 21944 Saudi Arabia
| |
Collapse
|
10
|
Mechanostimulation of breast myoepithelial cells induces functional changes associated with DCIS progression to invasion. NPJ Breast Cancer 2022; 8:109. [PMID: 36127361 PMCID: PMC9489768 DOI: 10.1038/s41523-022-00464-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 07/13/2022] [Indexed: 11/16/2022] Open
Abstract
Women with ductal carcinoma in situ (DCIS) have an increased risk of progression to invasive breast cancer. Although not all women with DCIS will progress to invasion, all are treated as such, emphasising the need to identify prognostic biomarkers. We have previously shown that altered myoepithelial cells in DCIS predict disease progression and recurrence. By analysing DCIS duct size in sections of human breast tumour samples, we identified an associated upregulation of integrin β6 and an increase in periductal fibronectin deposition with increased DCIS duct size that associated with the progression of DCIS to invasion. Our modelling of the mechanical stretching myoepithelial cells undergo during DCIS progression confirmed the upregulation of integrin β6 and fibronectin expression in isolated primary and cell line models of normal myoepithelial cells. Our studies reveal that this mechanostimulated DCIS myoepithelial cell phenotype enhances invasion in a TGFβ-mediated upregulation of MMP13. Immunohistochemical analysis identified that MMP13 was specifically upregulated in DCIS, and it was associated with progression to invasion. These findings implicate tissue mechanics in altering the myoepithelial cell phenotype in DCIS, and that these alterations may be used to stratify DCIS patients into low and high risk for invasive progression.
Collapse
|
11
|
Expression of Proteinase-activated Receptor 2 (PAR2) as a Correlate of Concern in Triple-negative Breast Cancer (TNBC). Appl Immunohistochem Mol Morphol 2022; 30:446-452. [DOI: 10.1097/pai.0000000000001025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 02/18/2022] [Indexed: 01/18/2023]
|
12
|
Risom T, Glass DR, Averbukh I, Liu CC, Baranski A, Kagel A, McCaffrey EF, Greenwald NF, Rivero-Gutiérrez B, Strand SH, Varma S, Kong A, Keren L, Srivastava S, Zhu C, Khair Z, Veis DJ, Deschryver K, Vennam S, Maley C, Hwang ES, Marks JR, Bendall SC, Colditz GA, West RB, Angelo M. Transition to invasive breast cancer is associated with progressive changes in the structure and composition of tumor stroma. Cell 2022; 185:299-310.e18. [PMID: 35063072 PMCID: PMC8792442 DOI: 10.1016/j.cell.2021.12.023] [Citation(s) in RCA: 198] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 08/05/2021] [Accepted: 12/16/2021] [Indexed: 01/16/2023]
Abstract
Ductal carcinoma in situ (DCIS) is a pre-invasive lesion that is thought to be a precursor to invasive breast cancer (IBC). To understand the changes in the tumor microenvironment (TME) accompanying transition to IBC, we used multiplexed ion beam imaging by time of flight (MIBI-TOF) and a 37-plex antibody staining panel to interrogate 79 clinically annotated surgical resections using machine learning tools for cell segmentation, pixel-based clustering, and object morphometrics. Comparison of normal breast with patient-matched DCIS and IBC revealed coordinated transitions between four TME states that were delineated based on the location and function of myoepithelium, fibroblasts, and immune cells. Surprisingly, myoepithelial disruption was more advanced in DCIS patients that did not develop IBC, suggesting this process could be protective against recurrence. Taken together, this HTAN Breast PreCancer Atlas study offers insight into drivers of IBC relapse and emphasizes the importance of the TME in regulating these processes.
Collapse
Affiliation(s)
- Tyler Risom
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Department of Research Pathology, Genentech, South San Francisco, CA, USA
| | - David R Glass
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Inna Averbukh
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Candace C Liu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Alex Baranski
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Adam Kagel
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Erin F McCaffrey
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Noah F Greenwald
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Siri H Strand
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sushama Varma
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Alex Kong
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Leeat Keren
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sucheta Srivastava
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Chunfang Zhu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Zumana Khair
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Deborah J Veis
- Departments of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Katherine Deschryver
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Sujay Vennam
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Carlo Maley
- Biodesign institute, Arizona State University, Tempe, AZ, USA
| | | | | | - Sean C Bendall
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Graham A Colditz
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Robert B West
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Michael Angelo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Departments of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
13
|
Habanjar O, Diab-Assaf M, Caldefie-Chezet F, Delort L. 3D Cell Culture Systems: Tumor Application, Advantages, and Disadvantages. Int J Mol Sci 2021; 22:12200. [PMID: 34830082 PMCID: PMC8618305 DOI: 10.3390/ijms222212200] [Citation(s) in RCA: 251] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/05/2021] [Accepted: 11/07/2021] [Indexed: 01/09/2023] Open
Abstract
The traditional two-dimensional (2D) in vitro cell culture system (on a flat support) has long been used in cancer research. However, this system cannot be fully translated into clinical trials to ideally represent physiological conditions. This culture cannot mimic the natural tumor microenvironment due to the lack of cellular communication (cell-cell) and interaction (cell-cell and cell-matrix). To overcome these limitations, three-dimensional (3D) culture systems are increasingly developed in research and have become essential for tumor research, tissue engineering, and basic biology research. 3D culture has received much attention in the field of biomedicine due to its ability to mimic tissue structure and function. The 3D matrix presents a highly dynamic framework where its components are deposited, degraded, or modified to delineate functions and provide a platform where cells attach to perform their specific functions, including adhesion, proliferation, communication, and apoptosis. So far, various types of models belong to this culture: either the culture based on natural or synthetic adherent matrices used to design 3D scaffolds as biomaterials to form a 3D matrix or based on non-adherent and/or matrix-free matrices to form the spheroids. In this review, we first summarize a comparison between 2D and 3D cultures. Then, we focus on the different components of the natural extracellular matrix that can be used as supports in 3D culture. Then we detail different types of natural supports such as matrigel, hydrogels, hard supports, and different synthetic strategies of 3D matrices such as lyophilization, electrospiding, stereolithography, microfluid by citing the advantages and disadvantages of each of them. Finally, we summarize the different methods of generating normal and tumor spheroids, citing their respective advantages and disadvantages in order to obtain an ideal 3D model (matrix) that retains the following characteristics: better biocompatibility, good mechanical properties corresponding to the tumor tissue, degradability, controllable microstructure and chemical components like the tumor tissue, favorable nutrient exchange and easy separation of the cells from the matrix.
Collapse
Affiliation(s)
- Ola Habanjar
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France; (O.H.); (F.C.-C.)
| | - Mona Diab-Assaf
- Equipe Tumorigénèse Pharmacologie Moléculaire et Anticancéreuse, Faculté des Sciences II, Université Libanaise Fanar, Beyrouth 1500, Liban;
| | - Florence Caldefie-Chezet
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France; (O.H.); (F.C.-C.)
| | - Laetitia Delort
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France; (O.H.); (F.C.-C.)
| |
Collapse
|
14
|
Liu CC, McCaffrey EF, Greenwald NF, Soon E, Risom T, Vijayaragavan K, Oliveria JP, Mrdjen D, Bosse M, Tebaykin D, Bendall SC, Angelo M. Multiplexed Ion Beam Imaging: Insights into Pathobiology. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2021; 17:403-423. [PMID: 34752710 DOI: 10.1146/annurev-pathmechdis-030321-091459] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Next-generation tools for multiplexed imaging have driven a new wave of innovation in understanding how single-cell function and tissue structure are interrelated. In previous work, we developed multiplexed ion beam imaging by time of flight, a highly multiplexed platform that uses secondary ion mass spectrometry to image dozens of antibodies tagged with metal reporters. As instrument throughput has increased, the breadth and depth of imaging data have increased as well. To extract meaningful information from these data, we have developed tools for cell identification, cell classification, and spatial analysis. In this review, we discuss these tools and provide examples of their application in various contexts, including ductal carcinoma in situ, tuberculosis, and Alzheimer's disease. We hope the synergy between multiplexed imaging and automated image analysis will drive a new era in anatomic pathology and personalized medicine wherein quantitative spatial signatures are used routinely for more accurate diagnosis, prognosis, and therapeutic selection. Expected final online publication date for the Annual Review of Pathology: Mechanisms of Disease, Volume 17 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Candace C Liu
- Department of Pathology, Stanford University, Stanford, California 94304 USA; , , , , , , , , , , ,
| | - Erin F McCaffrey
- Department of Pathology, Stanford University, Stanford, California 94304 USA; , , , , , , , , , , ,
| | - Noah F Greenwald
- Department of Pathology, Stanford University, Stanford, California 94304 USA; , , , , , , , , , , ,
| | - Erin Soon
- Department of Pathology, Stanford University, Stanford, California 94304 USA; , , , , , , , , , , ,
| | - Tyler Risom
- Department of Pathology, Stanford University, Stanford, California 94304 USA; , , , , , , , , , , , .,Current affiliation: Genentech, South San Francisco, California 94080; USA
| | - Kausalia Vijayaragavan
- Department of Pathology, Stanford University, Stanford, California 94304 USA; , , , , , , , , , , ,
| | - John-Paul Oliveria
- Department of Pathology, Stanford University, Stanford, California 94304 USA; , , , , , , , , , , , .,Current affiliation: Genentech, South San Francisco, California 94080; USA
| | - Dunja Mrdjen
- Department of Pathology, Stanford University, Stanford, California 94304 USA; , , , , , , , , , , ,
| | - Marc Bosse
- Department of Pathology, Stanford University, Stanford, California 94304 USA; , , , , , , , , , , ,
| | - Dmitry Tebaykin
- Department of Pathology, Stanford University, Stanford, California 94304 USA; , , , , , , , , , , ,
| | - Sean C Bendall
- Department of Pathology, Stanford University, Stanford, California 94304 USA; , , , , , , , , , , ,
| | - Michael Angelo
- Department of Pathology, Stanford University, Stanford, California 94304 USA; , , , , , , , , , , ,
| |
Collapse
|
15
|
Ganz HM, Buchmann B, Engelbrecht LK, Jesinghaus M, Eichelberger L, Gabka CJ, Schmidt GP, Muckenhuber A, Weichert W, Bausch AR, Scheel CH. Generation of ductal organoids from normal mammary luminal cells reveals invasive potential. J Pathol 2021; 255:451-463. [PMID: 34467523 DOI: 10.1002/path.5790] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 08/12/2021] [Accepted: 08/28/2021] [Indexed: 11/09/2022]
Abstract
Here we present an experimental model for human luminal progenitor cells that enables single, primary cells isolated from normal tissue to generate complex branched structures resembling the ductal morphology of low-grade carcinoma of no special type. Thereby, we find that ductal structures are generated through invasive branching morphogenesis via matrix remodeling and identify reduced actomyosin contractility as a prerequisite for invasion. In addition, we show that knockout of E-cadherin causes a dissolution of duct formation as observed in invasive lobular carcinoma, a subtype of invasive carcinomas where E-cadherin function is frequently lost. Thus, our model shows that invasive capacity can be elicited from normal luminal cells in specific environments, which results in low-grade no special type morphology. This assay offers a platform to investigate the dynamics of luminal cell invasion and unravel the impact of genetic and non-genetic aberrations on invasive morphology. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Hilary M Ganz
- Institute of Stem Cell Research, Helmholtz Center for Health and Environmental Research Munich, Neuherberg, Germany
| | - Benedikt Buchmann
- Chair of Cellular Biophysics E27, Technical University Munich, Garching, Germany
| | - Lisa K Engelbrecht
- Chair of Cellular Biophysics E27, Technical University Munich, Garching, Germany
| | - Moritz Jesinghaus
- Institute of Pathology, Technical University of Munich, Munich, Germany.,Institute of Pathology, University Hospital Marburg, Marburg, Germany
| | - Laura Eichelberger
- Center for Functional Protein Assemblies, Technical University of Munich, Munich, Germany.,Clinic and Polyclinic for Internal Medicine II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Christian J Gabka
- Nymphenburg Clinic for Plastic and Aesthetic Surgery, Munich, Germany
| | - Georg P Schmidt
- Department of Obstetrics and Gynecology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | | | - Wilko Weichert
- Institute of Pathology, Technical University of Munich, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Andreas R Bausch
- Chair of Cellular Biophysics E27, Technical University Munich, Garching, Germany
| | - Christina H Scheel
- Institute of Stem Cell Research, Helmholtz Center for Health and Environmental Research Munich, Neuherberg, Germany.,Department of Dermatology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
16
|
Ismerim AB, de Oliveira Araújo IB, de Aquino Xavier FC, Rocha CAG, Macedo CL, Cangussu MCT, Freitas VS, Della Coletta R, Cury PR, Santos JN. Mast Cells and Proteins Related to Myofibroblast Differentiation (PAR-2, IL-6, and TGFβ1) in Salivary Cancers: A Preliminary Study. Appl Immunohistochem Mol Morphol 2021; 29:e57-e67. [PMID: 33797435 DOI: 10.1097/pai.0000000000000924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 01/27/2021] [Indexed: 11/26/2022]
Abstract
Salivary gland neoplasms represent an important group of cancers in the head and neck and myoepithelial cells play a key role on the development these tumors. This study evaluated the distribution of mast cells and related proteins (PAR-2, TGFβ1, IL-6) to the myofibroblastic differentiation in malignant tumors of salivary glands with and without myoepithelial differentiation. Immunohistochemical assessement for tryptase mast cells, SMA, PAR-2, TGFβ1, IL-6 was performed in 10 cases of polymorphous low-grade adenocarcinoma, 14 cases of mucoepidermoid carcinoma (MEC) and 10 cases of adenoid cystic carcinoma. When the density of mast cells were compared between tumors, their density was significantly higher in MEC (P=0.08). Tumors with high expression of PAR-2 (79.4%) exhibited a high density of mast cells. Myofibroblasts were more frequent in malignant tumors with low expression (<50%) of cell masts. Individual analysis of the tumors showed no significant difference between the expression of PAR-2, IL-6, TGFβ1, and myofibroblasts. When the density of mast cells, myofibroblasts and the expression of PAR-2 protein, IL-6, and TGFβ1 were compared, it was no statistically significant difference between tumors with and without myoepithelial differentiation. The results of present study suggest a possible participation of mast cells and especially of PAR-2 in the development and progression of malignant salivary cancers, regardless of myoepithelial content.
Collapse
Affiliation(s)
- Adna B Ismerim
- Department of Health, School of Dentistry, Southwestern State University of Bahia, Jequié
| | | | | | | | | | - Maria C T Cangussu
- Department of Dental Public Health, School of Dentistry, Federal University of Bahia
| | - Valéria S Freitas
- Department of Health, School of Dentistry, State University of Feira de Santana, Feira de Santana
| | - Ricardo Della Coletta
- Department of Oral Diagnosis, School of Dentistry, University of Campinas, Piracicaba, São Paulo, Brazil
| | - Patrícia R Cury
- Department of Periodontic, School of Dentistry, State University of Feira de Santana, Feira de Santana, Bahia
| | | |
Collapse
|
17
|
Karpinets TV, Mitani Y, Liu B, Zhang J, Pytynia KB, Sellen LD, Karagiannis DT, Ferrarotto R, Futreal AP, El-Naggar AK. Whole-Genome Sequencing of Common Salivary Gland Carcinomas: Subtype-Restricted and Shared Genetic Alterations. Clin Cancer Res 2021; 27:3960-3969. [PMID: 34011559 DOI: 10.1158/1078-0432.ccr-20-4071] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/27/2021] [Accepted: 05/14/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Salivary gland carcinomas (SGCs) are pathologically classified into several widely diverse subtypes, of which adenoid cystic carcinoma (ACC), mucoepidermoid carcinoma (MEC), and salivary duct carcinoma (SDC) are the most commonly encountered. A comparative genetic analysis of these subtypes provides detailed information on the genetic alterations that are associated with their tumorigenesis and may lead to the identification of biomarkers to guide tumor-specific clinical trials. EXPERIMENTAL DESIGN Whole-genome sequencing of 58 common SGCs (20 ACCs, 20 SDCs, and 18 MECs) was performed to catalog structural variations, copy number, rearrangements, and driver mutations. Data were bioinformatically analyzed and correlated with clinicopathologic parameters, and selected targets were validated. RESULTS Novel and recurrent type-specific and shared genetic alterations were identified within and among 3 subtypes. Mutually exclusive canonical fusion and nonfusion genomic alterations were identified in both ACC and MEC. In ACCs, loss of chromosome 12q was dominant in MYB or MYBL1 fusion-positive tumors and mutations of NOTCH pathway were more common in these fusion negatives. In MECs, CRTC1-MAML2 fusion-positive tumors showed frequent BAP1 mutation, and tumors lacking this fusion were enriched with LRFN1 mutation. SDCs displayed considerable genetic instability, lacked recurrent chromosomal rearrangements, and demonstrated nonoverlapping TP53 mutation and ERBB2 amplification in a subset of tumors. Limited genetic alterations, including focal amplifications of 8q21-q23, were shared by all subtypes and were associated with poor survival. CONCLUSIONS This study delineates type-specific and shared genetic alterations that are associated with early phenotypic commitment and the biologic progression of common SGCs. These alterations, upon validation, could serve as biomarkers in tumor-specific clinical trials.
Collapse
Affiliation(s)
- Tatiana V Karpinets
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yoshitsugu Mitani
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bin Liu
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jianhua Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kristen B Pytynia
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Linton D Sellen
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Danice T Karagiannis
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Renata Ferrarotto
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Andrew P Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Adel K El-Naggar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
18
|
Marousez L, Hanssens S, Butruille L, Petit C, Pourpe C, Besengez C, Rakza T, Storme L, Deruelle P, Lesage J, Eberlé D. Breast milk apelin level increases with maternal obesity and high-fat feeding during lactation. Int J Obes (Lond) 2021; 45:1052-1060. [PMID: 33594258 DOI: 10.1038/s41366-021-00772-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 12/14/2020] [Accepted: 01/21/2021] [Indexed: 01/31/2023]
Abstract
OBJECTIVE Recent evidence indicates that levels of breast milk (BM) hormones such as leptin can fluctuate with maternal adiposity, suggesting that BM hormones may signal maternal metabolic and nutritional environments to offspring during postnatal development. The hormone apelin is highly abundant in BM but its regulation during lactation is completely unknown. Here, we evaluated whether maternal obesity and overnutrition impacted BM apelin and leptin levels in clinical cohorts and lactating rats. METHODS BM and plasma samples were collected from normal-weight and obese breastfeeding women, and from lactating rats fed a control or a high fat (HF) diet during lactation. Apelin and leptin levels were assayed by ELISA. Mammary gland (MG) apelin expression and its cellular localization in lactating rats was measured by quantitative RT-PCR and immunofluorescence, respectively. RESULTS BM apelin levels increased with maternal BMI, whereas plasma apelin levels decreased. BM apelin was also positively correlated with maternal insulin and C-peptide levels. In rats, maternal HF feeding exclusively during lactation was sufficient to increase BM apelin levels and decrease its plasma concentration without changing body weight. In contrast, BM leptin levels increased with maternal BMI in humans, but did not change with maternal HF feeding during lactation in rats. Apelin is highly expressed in the rat MG during lactation and was mainly localized to mammary myoepithelial cells. We found that MG apelin gene expression was up-regulated by maternal HF diet and positively correlated with BM apelin content and maternal insulinemia. CONCLUSIONS Our study indicates that BM apelin levels increase with long- and short-term overnutrition, possibly via maternal hyperinsulinemia and transcriptional upregulation of MG apelin expression in myoepithelial cells. Apelin regulates many physiological processes, including energy metabolism, digestive function, and development. Further studies are needed to unravel the consequences of such changes in offspring development.
Collapse
Affiliation(s)
- Lucie Marousez
- Univ. Lille, EA4489 Environnement Périnatal et Santé, Lille, France.,Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France
| | - Sandy Hanssens
- Univ. Lille, EA4489 Environnement Périnatal et Santé, Lille, France.,CHU Lille, Jeanne de Flandre Hospital, Gynecology-Obstetrics, Lille, France
| | - Laura Butruille
- Univ. Lille, EA4489 Environnement Périnatal et Santé, Lille, France.,Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Céline Petit
- CHU Lille, Jeanne de Flandre Hospital, Gynecology-Obstetrics, Lille, France
| | - Charlène Pourpe
- Univ. Lille, EA4489 Environnement Périnatal et Santé, Lille, France.,Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | | | - Thameur Rakza
- Univ. Lille, EA4489 Environnement Périnatal et Santé, Lille, France.,CHU Lille, Jeanne de Flandre Hospital, Neonatology and Pediatrics, Lille, France
| | - Laurent Storme
- Univ. Lille, EA4489 Environnement Périnatal et Santé, Lille, France.,CHU Lille, Jeanne de Flandre Hospital, Neonatology and Pediatrics, Lille, France
| | - Philippe Deruelle
- Univ. Lille, EA4489 Environnement Périnatal et Santé, Lille, France.,CHU Lille, Jeanne de Flandre Hospital, Gynecology-Obstetrics, Lille, France
| | - Jean Lesage
- Univ. Lille, EA4489 Environnement Périnatal et Santé, Lille, France.,Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France
| | - Delphine Eberlé
- Univ. Lille, EA4489 Environnement Périnatal et Santé, Lille, France. .,Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France.
| |
Collapse
|
19
|
Holliday H, Roden D, Junankar S, Wu SZ, Baker LA, Krisp C, Chan CL, McFarland A, Skhinas JN, Cox TR, Pal B, Huntington ND, Ormandy CJ, Carroll JS, Visvader J, Molloy MP, Swarbrick A. Inhibitor of Differentiation 4 (ID4) represses mammary myoepithelial differentiation via inhibition of HEB. iScience 2021; 24:102072. [PMID: 33554073 PMCID: PMC7851187 DOI: 10.1016/j.isci.2021.102072] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/24/2020] [Accepted: 01/12/2021] [Indexed: 12/17/2022] Open
Abstract
Inhibitor of differentiation (ID) proteins dimerize with basic HLH (bHLH) transcription factors, repressing transcription of lineage-specification genes across diverse cellular lineages. ID4 is a key regulator of mammary stem cells; however, the mechanism by which it achieves this is unclear. Here, we show that ID4 has a cell autonomous role in preventing myoepithelial differentiation of basal cells in mammary organoids and in vivo. ID4 positively regulates proliferative genes and negatively regulates genes involved in myoepithelial function. Mass spectrometry reveals that ID4 interacts with the bHLH protein HEB, which binds to E-box motifs in regulatory elements of basal developmental genes involved in extracellular matrix and the contractile cytoskeleton. We conclude that high ID4 expression in mammary basal stem cells antagonizes HEB transcriptional activity, preventing myoepithelial differentiation and allowing for appropriate tissue morphogenesis. Downregulation of ID4 during pregnancy modulates gene regulated by HEB, promoting specialization of basal cells into myoepithelial cells.
Collapse
Affiliation(s)
- Holly Holliday
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - Daniel Roden
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - Simon Junankar
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - Sunny Z. Wu
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - Laura A. Baker
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - Christoph Krisp
- Australian Proteome Analysis Facility, Macquarie University, Sydney, NSW 2109, Australia
- Institute of Clinical Chemistry and Laboratory Medicine, Mass Spectrometric Proteomics, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
| | - Chia-Ling Chan
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Andrea McFarland
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Joanna N. Skhinas
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Thomas R. Cox
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - Bhupinder Pal
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3084, Australia
| | - Nicholas D. Huntington
- Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3168, Australia
| | - Christopher J. Ormandy
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - Jason S. Carroll
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UK
| | - Jane Visvader
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Mark P. Molloy
- Australian Proteome Analysis Facility, Macquarie University, Sydney, NSW 2109, Australia
| | - Alexander Swarbrick
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| |
Collapse
|
20
|
Hillers-Ziemer LE, Arendt LM. Weighing the Risk: effects of Obesity on the Mammary Gland and Breast Cancer Risk. J Mammary Gland Biol Neoplasia 2020; 25:115-131. [PMID: 32519090 PMCID: PMC7933979 DOI: 10.1007/s10911-020-09452-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/02/2020] [Accepted: 06/03/2020] [Indexed: 12/17/2022] Open
Abstract
Obesity is a preventable risk factor for breast cancer following menopause. Regardless of menopausal status, obese women who develop breast cancer have a worsened prognosis. Breast tissue is comprised of mammary epithelial cells organized into ducts and lobules and surrounded by adipose-rich connective tissue. Studies utilizing multiple in vivo models of obesity as well as human breast tissue have contributed to our understanding of how obesity alters mammary tissue. Localized changes in mammary epithelial cell populations, elevated secretion of adipokines and angiogenic mediators, inflammation within mammary adipose tissue, and remodeling of the extracellular matrix may result in an environment conducive to breast cancer growth. Despite these significant alterations caused by obesity within breast tissue, studies have suggested that some, but not all, obesity-induced changes may be mitigated with weight loss. Here, we review our current understanding regarding the impact of obesity on the breast microenvironment, how obesity-induced changes may contribute to breast tumor progression, and the impact of weight loss on the breast microenvironment.
Collapse
Affiliation(s)
- Lauren E Hillers-Ziemer
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, 1525 Linden Drive, Madison, WI, 53706, USA
| | - Lisa M Arendt
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, 1525 Linden Drive, Madison, WI, 53706, USA.
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI, 53706, USA.
| |
Collapse
|
21
|
Masood S. Is it ductal carcinoma in situ with microinvasion or "Ductogenesis"? The role of myoepithelial cell markers. Breast J 2020; 26:1138-1147. [PMID: 32447817 DOI: 10.1111/tbj.13897] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 05/01/2020] [Indexed: 11/29/2022]
Abstract
Mammary myoepithelial cells have been under-recognized for many years since they were considered less important in breast cancer tumorigenesis compared to luminal epithelial cells. However, in recent years with advances in genomics, cell biology, and research in breast cancer microenvironment, more emphasis has been placed on better understanding of the role that myoepithelial cells play in breast cancer progression. As the result, it has been recognized that the presence or absence of myoepithelial cells play a critical role in the assessment of tumor invasion in diagnostic breast pathology. In addition, advances in screening mammography and breast imaging has resulted in increased detection of ductal carcinoma in situ and consequently more diagnosis of ductal carcinoma in situ with microinvasion. In the present review, we discuss the characteristics of myoepithelial cells, their genomic markers and their role in the accurate diagnosis of ductal carcinoma in situ with microinvasion. We also share our experience with reporting of various morphologic features of ductal carcinoma in situ that may mimic microinvasion and introduce the term of ductogenesis.
Collapse
Affiliation(s)
- Shahla Masood
- Department of Pathology, University of Florida College of Medicine - Jax, Jacksonville, FL, USA
| |
Collapse
|
22
|
Dessources K, Sebastiao APM, Pareja F, Weigelt B, Reis-Filho JS. How Did We Get There? The Progression from Ductal Carcinoma In Situ to Invasive Ductal Carcinoma. CURRENT BREAST CANCER REPORTS 2019. [DOI: 10.1007/s12609-019-00318-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
23
|
Brock EJ, Ji K, Shah S, Mattingly RR, Sloane BF. In Vitro Models for Studying Invasive Transitions of Ductal Carcinoma In Situ. J Mammary Gland Biol Neoplasia 2019; 24:1-15. [PMID: 30056557 PMCID: PMC6641861 DOI: 10.1007/s10911-018-9405-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 07/13/2018] [Indexed: 12/11/2022] Open
Abstract
About one fourth of all newly identified cases of breast carcinoma are diagnoses of breast ductal carcinoma in situ (DCIS). Since we cannot yet distinguish DCIS cases that would remain indolent from those that may progress to life-threatening invasive ductal carcinoma (IDC), almost all women undergo aggressive treatment. In order to allow for more rational individualized treatment, we and others are developing in vitro models to identify and validate druggable pathways that mediate the transition of DCIS to IDC. These models range from conventional two-dimensional (2D) monolayer cultures on plastic to 3D cultures in natural or synthetic matrices. Some models consist solely of DCIS cells, either cell lines or primary cells. Others are co-cultures that include additional cell types present in the normal or cancerous human breast. The 3D co-culture models more accurately mimic structural and functional changes in breast architecture that accompany the transition of DCIS to IDC. Mechanistic studies of the dynamic and temporal changes associated with this transition are facilitated by adapting the in vitro models to engineered microfluidic platforms. Ultimately, the goal is to create in vitro models that can serve as a reproducible preclinical screen for testing therapeutic strategies that will reduce progression of DCIS to IDC. This review will discuss the in vitro models that are currently available, as well as the progress that has been made using them to understand DCIS pathobiology.
Collapse
MESH Headings
- Breast/pathology
- Breast Neoplasms/drug therapy
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/drug therapy
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Intraductal, Noninfiltrating/drug therapy
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Cell Line, Tumor
- Coculture Techniques/methods
- Drug Screening Assays, Antitumor/methods
- Female
- Humans
- Neoplasm Invasiveness/pathology
- Neoplasm Invasiveness/prevention & control
- Primary Cell Culture/methods
Collapse
Affiliation(s)
- Ethan J Brock
- Program in Cancer Biology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Kyungmin Ji
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Seema Shah
- Program in Cancer Biology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Raymond R Mattingly
- Program in Cancer Biology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Bonnie F Sloane
- Program in Cancer Biology, Wayne State University School of Medicine, Detroit, MI, 48201, USA.
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, 48201, USA.
- Department of Pharmacology, Wayne State University, 540 E. Canfield, Detroit, MI, 48201, USA.
| |
Collapse
|
24
|
Nelson AC, Machado HL, Schwertfeger KL. Breaking through to the Other Side: Microenvironment Contributions to DCIS Initiation and Progression. J Mammary Gland Biol Neoplasia 2018; 23:207-221. [PMID: 30168075 PMCID: PMC6237657 DOI: 10.1007/s10911-018-9409-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/22/2018] [Indexed: 01/08/2023] Open
Abstract
Refinements in early detection, surgical and radiation therapy, and hormone receptor-targeted treatments have improved the survival rates for breast cancer patients. However, the ability to reliably identify which non-invasive lesions and localized tumors have the ability to progress and/or metastasize remains a major unmet need in the field. The current diagnostic and therapeutic strategies focus on intrinsic alterations within carcinoma cells that are closely associated with proliferation. However, substantial accumulating evidence has indicated that permissive changes in the stromal tissues surrounding the carcinoma play an integral role in breast cancer tumor initiation and progression. Numerous studies have suggested that the stromal environment surrounding ductal carcinoma in situ (DCIS) lesions actively contributes to enhancing tumor cell invasion and immune escape. This review will describe the current state of knowledge regarding the mechanisms through which the microenvironment interacts with DCIS lesions focusing on recent studies that describe the contributions of myoepithelial cells, fibroblasts and immune cells to invasion and subsequent progression. These mechanisms will be considered in the context of developing biomarkers for identifying lesions that will progress to invasive carcinoma and/or developing approaches for therapeutic intervention.
Collapse
Affiliation(s)
- Andrew C Nelson
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, 2231 6th St SE, Minneapolis, MN, 55455, USA
| | - Heather L Machado
- Department of Biochemistry and Molecular Biology, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA, USA
| | - Kathryn L Schwertfeger
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA.
- Masonic Cancer Center, University of Minnesota, 2231 6th St SE, Minneapolis, MN, 55455, USA.
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
25
|
Fleming JM, Yeyeodu ST, McLaughlin A, Schuman D, Taylor DK. In Situ Drug Delivery to Breast Cancer-Associated Extracellular Matrix. ACS Chem Biol 2018; 13:2825-2840. [PMID: 30183254 DOI: 10.1021/acschembio.8b00396] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The extracellular matrix (ECM) contributes to tumor progression through changes induced by tumor and stromal cell signals that promote increased ECM density and stiffness. The increase in ECM stiffness is known to promote tumor cell invasion into surrounding tissues and metastasis. In addition, this scar-like ECM creates a protective barrier around the tumor that reduces the effectiveness of innate and synthetic antitumor agents. Herein, clinically approved breast cancer therapies as well as novel experimental approaches that target the ECM are discussed, including in situ hydrogel drug delivery systems, an emerging technology the delivers toxic chemotherapeutics, gene-silencing microRNAs, and tumor suppressing immune cells directly inside the tumor. Intratumor delivery of therapeutic agents has the potential to drastically reduce systemic side effects experienced by the patient and increase the efficacy of these agents. This review also describes the opposing effects of ECM degradation on tumor progression, where some studies report improved drug delivery and delayed cancer progression and others report enhanced metastasis and decreased patient survival. Given the recent increase in ECM-targeting drugs entering preclinical and clinical trials, understanding and addressing the factors that impact the effect of the ECM on tumor progression is imperative for the sake of patient safety and survival outcome.
Collapse
Affiliation(s)
- Jodie M. Fleming
- Department of Biological and Biomedical Sciences, North Carolina Central University, Durham, North Carolina, United States
| | - Susan T. Yeyeodu
- Charles River Discovery Services, Morrisville, North Carolina, United States
| | - Ashley McLaughlin
- Department of Biological and Biomedical Sciences, North Carolina Central University, Durham, North Carolina, United States
| | - Darren Schuman
- Department of Chemistry and Biochemistry, North Carolina Central University, Durham, North Carolina, United States
| | - Darlene K. Taylor
- Department of Chemistry and Biochemistry, North Carolina Central University, Durham, North Carolina, United States
| |
Collapse
|
26
|
Epigenetic Silencing of THY1 Tracks the Acquisition of the Notch1–EGFR Signaling in a Xenograft Model of CD44+/CD24low/CD90+ Myoepithelial Cells. Mol Cancer Res 2018; 17:628-641. [DOI: 10.1158/1541-7786.mcr-17-0324] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/13/2017] [Accepted: 09/13/2018] [Indexed: 11/16/2022]
|
27
|
Abstract
Goodwin and Nelson discuss the recent discovery by Sirka et al. that remodeling activity of myoepithelial cells can control breast cancer cell invasion. Smooth muscle–like cells can actively remodel epithelia, a mechanism common across developing tissues. In this issue, new work from Sirka et al. (2018. J. Cell Biol.https://doi.org/10.1083/jcb.201802144) demonstrates a novel mechanism for tumor suppression by smooth muscle–like myoepithelial cells of the mammary gland.
Collapse
Affiliation(s)
- Katharine Goodwin
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ
| | - Celeste M Nelson
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ .,Department of Molecular Biology, Princeton University, Princeton, NJ
| |
Collapse
|
28
|
Luminal MCF-12A & myoepithelial-like Hs 578Bst cells form bilayered acini similar to human breast. Future Sci OA 2018; 4:FSO315. [PMID: 30112185 PMCID: PMC6088263 DOI: 10.4155/fsoa-2018-0010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 05/08/2018] [Indexed: 12/21/2022] Open
Abstract
The epithelium's functional unit is the bilayered acinus, made of a layer of luminal cells, surrounded by a layer of basal cells mainly composed of myoepithelial cells. Aim The aim of this study was to develop a reproducible and manipulable 3D co-culture model of the bilayered acinus in vitro to study the interactions between the two layers. Materials & methods Two different combinations of cell lines were co-cultured in Matrigel: SCp2 and SCg6 mice cells, or MCF-12A and Hs 578Bst human cell lines. Results Confocal microscopy analysis showed that only MCF-12A and Hs 578Bst cells could form some bilayered acini. This in vitro bilayered acini model will allow us to understand the role of interactions between luminal and myoepithelial cells in the normal breast development.
Collapse
|
29
|
Sirka OK, Shamir ER, Ewald AJ. Myoepithelial cells are a dynamic barrier to epithelial dissemination. J Cell Biol 2018; 217:3368-3381. [PMID: 30061105 PMCID: PMC6168248 DOI: 10.1083/jcb.201802144] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/12/2018] [Accepted: 06/28/2018] [Indexed: 01/10/2023] Open
Abstract
Myoepithelial cells function collectively as a dynamic barrier to the invasion and dissemination of Twist1+ luminal epithelial cells and both luminal and basal phenotype breast cancer cells. Barrier function depends on myoepithelial abundance and both smooth muscle contractility and intercellular adhesion within the myoepithelium. The mammary epithelium is composed of an inner luminal and surrounding myoepithelial cell layer. The presence of cancer cells beyond the myoepithelium defines invasive breast cancer, yet the role of the myoepithelium during invasion remains unclear. We developed a 3D organotypic culture assay to model this process through lineage-specific expression of the prometastatic transcription factor Twist1. We sought to distinguish the functional role of the myoepithelium in regulating invasion and local dissemination. Myoepithelial-specific Twist1 expression induced cell-autonomous myoepithelial cell escape. Remarkably, luminal-specific Twist1 expression was rarely sufficient for escape. Time-lapse microscopy revealed that myoepithelial cells collectively restrain and reinternalize invading Twist1+ luminal cells. Barrier function correlated with myoepithelial abundance and required the expression of α-smooth muscle actin and P-cadherin. We next demonstrated that myoepithelial cells can restrain and recapture invasive cancer cells. Our data establish the concept of the myoepithelium as a dynamic barrier to luminal dissemination and implicate both smooth muscle contractility and intercellular adhesion in barrier function.
Collapse
Affiliation(s)
- Orit Katarina Sirka
- Departments of Cell Biology, Oncology, and Biomedical Engineering, Center for Cell Dynamics, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Eliah R Shamir
- Departments of Cell Biology, Oncology, and Biomedical Engineering, Center for Cell Dynamics, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Andrew J Ewald
- Departments of Cell Biology, Oncology, and Biomedical Engineering, Center for Cell Dynamics, The Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
30
|
Terabayashi T, Hanada K, Motani K, Kosako H, Yamaoka M, Kimura T, Ishizaki T. Baicalein disturbs the morphological plasticity and motility of breast adenocarcinoma cells depending on the tumor microenvironment. Genes Cells 2018; 23:466-479. [PMID: 29667279 DOI: 10.1111/gtc.12584] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 03/21/2018] [Indexed: 12/20/2022]
Abstract
During tumor invasion, cancer cells change their morphology and mode of migration based on communication with the surrounding environment. Numerous studies have indicated that paracrine interactions from non-neoplastic cells impact the migratory and invasive properties of cancer cells. Thus, these interactions are potential targets for anticancer therapies. In this study, we showed that the flavones member baicalein suppresses the motility of breast cancer cells that is promoted by paracrine interactions. First, we identified laminin-332 (LN-332) as a principle paracrine factor in conditioned medium from mammary epithelium-derived MCF10A cells that regulates the morphology and motility of breast adenocarcinoma MDA-MB-231 cells. Then, we carried out a morphology-based screen for small compounds, which showed that baicalein suppressed the morphological changes and migratory activity of MDA-MB-231 cells that were induced by conditioned medium from MCF10A cells and LN-332. We also found that baicalein caused narrower and incomplete lamellipodia formation in conditioned medium-treated MDA-MB-231 cells, although actin dynamics downstream of Rho family small GTPases were unaffected. These results suggest the importance of mammary epithelial cells in the cancer microenvironment promoting the migratory activity of breast adenocarcinoma cells and show a novel mechanism through which baicalein inhibits cancer cell motility.
Collapse
Affiliation(s)
- Takeshi Terabayashi
- Department of Pharmacology, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Katsuhiro Hanada
- Clinical Engineering Research Center, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Kou Motani
- Division of Cell Signaling, Fujii Memorial Institute of Medical Sciences, Tokushima University, Tokushima, Japan
| | - Hidetaka Kosako
- Division of Cell Signaling, Fujii Memorial Institute of Medical Sciences, Tokushima University, Tokushima, Japan
| | - Mami Yamaoka
- Department of Pharmacology, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Toshihide Kimura
- Department of Pharmacology, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Toshimasa Ishizaki
- Department of Pharmacology, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| |
Collapse
|
31
|
Gambara G, Gaebler M, Keilholz U, Regenbrecht CRA, Silvestri A. From Chemotherapy to Combined Targeted Therapeutics: In Vitro and in Vivo Models to Decipher Intra-tumor Heterogeneity. Front Pharmacol 2018; 9:77. [PMID: 29491834 PMCID: PMC5817069 DOI: 10.3389/fphar.2018.00077] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 01/23/2018] [Indexed: 12/15/2022] Open
Abstract
Recent advances in next-generation sequencing and other omics technologies capable to map cell fate provide increasing evidence on the crucial role of intra-tumor heterogeneity (ITH) for cancer progression. The different facets of ITH, from genomic to microenvironmental heterogeneity and the hierarchical cellular architecture originating from the cancer stem cell compartment, contribute to the range of tumor phenotypes. Decoding these complex data resulting from the analysis of tumor tissue complexity poses a challenge for developing novel therapeutic strategies that can counteract tumor evolution and cellular plasticity. To achieve this aim, the development of in vitro and in vivo cancer models that resemble the complexity of ITH is crucial in understanding the interplay of cells and their (micro)environment and, consequently, in testing the efficacy of new targeted treatments and novel strategies of tailoring combinations of treatments to the individual composition of the tumor. This challenging approach may be an important cornerstone in overcoming the development of pharmaco-resistances during multiple lines of treatment. In this paper, we report the latest advances in patient-derived 3D (PD3D) cell cultures and patient-derived tumor xenografts (PDX) as in vitro and in vivo models that can retain the genetic and phenotypic heterogeneity of the tumor tissue.
Collapse
Affiliation(s)
- Guido Gambara
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin, Berlin, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Manuela Gaebler
- Department of Interdisciplinary Oncology, HELIOS Klinikum Berlin-Buch GmbH, Berlin, Germany
| | - Ulrich Keilholz
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin, Berlin, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | | |
Collapse
|
32
|
Rakha EA, Miligy IM, Gorringe KL, Toss MS, Green AR, Fox SB, Schmitt FC, Tan PH, Tse GM, Badve S, Decker T, Vincent-Salomon A, Dabbs DJ, Foschini MP, Moreno F, Wentao Y, Geyer FC, Reis-Filho JS, Pinder SE, Lakhani SR, Ellis IO. Invasion in breast lesions: the role of the epithelial-stroma barrier. Histopathology 2018; 72:1075-1083. [DOI: 10.1111/his.13446] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Emad A Rakha
- Department of Histopathology; Nottingham City Hospital NHS Trust; Nottingham University; Nottingham UK
| | - Islam M Miligy
- Department of Histopathology; Nottingham City Hospital NHS Trust; Nottingham University; Nottingham UK
| | - Kylie L Gorringe
- Cancer Genomics Program; Peter MacCallum Cancer Centre; Melbourne Vic. Australia
- The Sir Peter MacCallum Department of Oncology; University of Melbourne; Melbourne Vic. Australia
| | - Michael S Toss
- Department of Histopathology; Nottingham City Hospital NHS Trust; Nottingham University; Nottingham UK
| | - Andrew R Green
- Department of Histopathology; Nottingham City Hospital NHS Trust; Nottingham University; Nottingham UK
| | - Stephen B Fox
- Pathology Department; Peter MacCallum Cancer Centre; Melbourne Vic. Australia
| | - Fernando C Schmitt
- Institute of Molecular Pathology and Immunology (IPATIMUP) and Medical Faculty; University of Porto; Porto Portugal
| | - Puay-Hoon Tan
- Department of Pathology; Singapore General Hospital; Singapore
| | - Gary M Tse
- Department of Anatomical and Cellular Pathology; Prince of Wales Hospital; Hong Kong
| | - Sunil Badve
- Departments of Pathology and Internal Medicine; Clarian Pathology Laboratory of Indiana University; Indianapolis IN USA
| | - Thomas Decker
- Breast-Screening-Pathology; Reference Centre Munster; Gerhard Domagk-Institute of Pathology; University Hospital Münster; Münster Germany
| | | | - David J Dabbs
- University of Pittsburgh Medical Centre; Pittsburgh PA USA
| | - Maria P Foschini
- Department of Biomedical and Neuromotor Sciences; Section of Anatomic Pathology at Bellaria Hospital; University of Bologna; Bologna Italy
| | - Filipa Moreno
- Anatomic Pathology Department; Centro Hospitalar do Porto; Porto Portugal
| | - Yang Wentao
- Pathology; Fudan University Shanghai Cancer Center; Shanghai China
| | - Felipe C Geyer
- Department of Pathology; Memorial Sloan Kettering Cancer Centre; New York NY USA
| | - Jorge S Reis-Filho
- Department of Pathology; Memorial Sloan Kettering Cancer Centre; New York NY USA
| | - Sarah E Pinder
- Division of Cancer Studies; King's College London; Guy's Hospital; London UK
| | - Sunil R Lakhani
- Discipline of Molecular & Cellular Pathology; Faculty of Medicine; University of Queensland; The Royal Brisbane & Women's Hospital; Brisbane QLD Australia
| | - Ian O Ellis
- Department of Histopathology; Nottingham City Hospital NHS Trust; Nottingham University; Nottingham UK
| |
Collapse
|
33
|
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
|
34
|
Duivenvoorden HM, Rautela J, Edgington‐Mitchell LE, Spurling A, Greening DW, Nowell CJ, Molloy TJ, Robbins E, Brockwell NK, Lee CS, Chen M, Holliday A, Selinger CI, Hu M, Britt KL, Stroud DA, Bogyo M, Möller A, Polyak K, Sloane BF, O'Toole SA, Parker BS. Myoepithelial cell‐specific expression of stefin A as a suppressor of early breast cancer invasion. J Pathol 2017; 243:496-509. [DOI: 10.1002/path.4990] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 08/21/2017] [Accepted: 09/18/2017] [Indexed: 12/19/2022]
Affiliation(s)
- Hendrika M Duivenvoorden
- Department of Biochemistry and Genetics La Trobe Institute for Molecular Science Melbourne VIC Australia
| | - Jai Rautela
- Department of Biochemistry and Genetics La Trobe Institute for Molecular Science Melbourne VIC Australia
- Sir Peter MacCallum Department of Oncology University of Melbourne VIC Australia
- The Walter and Eliza Hall Institute of Medical Research Melbourne VIC Australia
- Department of Medical Biology University of Melbourne VIC Australia
| | - Laura E Edgington‐Mitchell
- Department of Biochemistry and Genetics La Trobe Institute for Molecular Science Melbourne VIC Australia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences Monash University Melbourne VIC Australia
| | - Alex Spurling
- Department of Biochemistry and Genetics La Trobe Institute for Molecular Science Melbourne VIC Australia
| | - David W Greening
- Department of Biochemistry and Genetics La Trobe Institute for Molecular Science Melbourne VIC Australia
| | - Cameron J Nowell
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences Monash University Melbourne VIC Australia
| | | | - Elizabeth Robbins
- Department of Tissue Pathology and Diagnostic Oncology Royal Prince Alfred Hospital Camperdown NSW Australia
| | - Natasha K Brockwell
- Department of Biochemistry and Genetics La Trobe Institute for Molecular Science Melbourne VIC Australia
| | - Cheok Soon Lee
- Department of Tissue Pathology and Diagnostic Oncology Royal Prince Alfred Hospital Camperdown NSW Australia
- Sydney Medical School University of Sydney NSW Australia
- Cancer Pathology and Cell Biology Laboratory Ingham Institute for Applied Medical Research, and University of New South Wales NSW Australia
- Cancer Pathology, Bosch Institute University of Sydney NSW Australia
| | - Maoshan Chen
- Department of Biochemistry and Genetics La Trobe Institute for Molecular Science Melbourne VIC Australia
| | - Anne Holliday
- Department of Tissue Pathology and Diagnostic Oncology Royal Prince Alfred Hospital Camperdown NSW Australia
| | - Cristina I Selinger
- Department of Tissue Pathology and Diagnostic Oncology Royal Prince Alfred Hospital Camperdown NSW Australia
| | - Min Hu
- Department of Medical Oncology Dana‐Farber Cancer Institute, Harvard Medical School Boston Massachusetts USA
| | - Kara L Britt
- Peter MacCallum Cancer Centre Melbourne VIC Australia
| | - David A Stroud
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute Monash University Melbourne VIC Australia
| | - Matthew Bogyo
- Department of Pathology Stanford University School of Medicine California USA
| | - Andreas Möller
- Immunology Department QIMR Berghofer Medical Research Institute Brisbane QLD Australia
| | - Kornelia Polyak
- Department of Medical Oncology Dana‐Farber Cancer Institute, Harvard Medical School Boston Massachusetts USA
| | - Bonnie F Sloane
- Department of Pharmacology Wayne State University School of Medicine Detroit Michigan USA
- Barbara Ann Karmanos Cancer Institute Wayne State University School of Medicine Detroit Michigan USA
| | - Sandra A O'Toole
- Sydney Medical School University of Sydney NSW Australia
- Garvan Institute of Medical Research Darlinghurst NSW Australia
- Australian Clinical Labs Bella Vista NSW Australia
| | - Belinda S Parker
- Department of Biochemistry and Genetics La Trobe Institute for Molecular Science Melbourne VIC Australia
| |
Collapse
|
35
|
Cellular subtype may predict survival outcomes in salivary adenoid cystic carcinoma patients-a single-institution experience. Virchows Arch 2017; 472:505-512. [PMID: 29063189 DOI: 10.1007/s00428-017-2248-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 08/22/2017] [Accepted: 10/16/2017] [Indexed: 01/22/2023]
|
36
|
Ogura M, Bridgeman VL, Malanchi I. Macrophages unlock progression of breast cancer cells experiencing matrigel-segregation in transplantation models. Sci Rep 2017; 7:11028. [PMID: 28887504 PMCID: PMC5591247 DOI: 10.1038/s41598-017-11403-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 08/21/2017] [Indexed: 12/14/2022] Open
Abstract
Basement membrane matrix proteins, such as matrigel, are able to improve the efficiency of tumour transplantation. This assay represents the gold standard to measure tumour initiation potential in vivo of a limited number of cancer cells. However, in culture conditions, matrigel directly signals to cancer cells altering their phenotype. We here investigate how matrigel influences the tumour reconstitution dynamics of breast cancer cells in vivo. This is particularly relevant in the setting of limiting dilution assay where cells are transplanted in a relatively high amount of Matrigel. We show that matrigel initially induces a normalized growth of transplanted MMTV-PyMT breast tumours cells. This occurs in the context of a matrigel-segregation effect where cancer cells are transiently isolated from host tissue. We identify macrophages as gatekeepers of the cancer-host cell interaction: depriving transplants from macrophages locked cancer cells in this isolated environment where they fail to form tumours despite retaining their intrinsic tumorigenic potential. This is a decisive proof of concept that cancer cells' malignant behaviour can be dominated by their microenvironment. Moreover, considering that diverse breast cancer cells are differently subjected to a segregation effect, this needs to be considered when comparing tumour initiation potential of different cancer cells.
Collapse
Affiliation(s)
- Misa Ogura
- Tumour Host Interaction Lab, The Francis Crick Institute, 1 Midland Rd, NW1 1AT, London, UK
| | - Victoria L Bridgeman
- Tumour Host Interaction Lab, The Francis Crick Institute, 1 Midland Rd, NW1 1AT, London, UK
| | - Ilaria Malanchi
- Tumour Host Interaction Lab, The Francis Crick Institute, 1 Midland Rd, NW1 1AT, London, UK.
| |
Collapse
|
37
|
Insulin-like Growth Factor II Messenger RNA-binding Protein 3 in Salivary Gland Tumors. Appl Immunohistochem Mol Morphol 2017; 24:422-6. [PMID: 26258757 DOI: 10.1097/pai.0000000000000205] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Insulin-like growth factor II mRNA-binding protein 3 (IMP3) is strongly expressed in malignant tumors and has been associated with their aggressive behavior. The aim of this study was to evaluate the presence of IMP3 in a series of salivary gland tumors. The sample consisted of 9 pleomorphic adenomas (PA), 14 adenoid cystic carcinomas (ACC), and 13 mucoepidermoid carcinomas (MEC) that were investigated by immunohistochemical technique. All cases of PA and MEC were positive for IMP3 particularly in the cytoplasm. PA showed 4 cases as high expression and 6 as low expression. MEC showed 10 cases as low expression and 3 as high expression. For ACC, 4 cases were high expression, whereas 6 cases were low expression. No significant difference was observed between tumors (P>0.05, Fisher's test) when both scores of IMP3 were compared. This study showed that MEC seems to be more sensitive to IMP3 than ACC and provided an insight into this protein in salivary gland tumors. Furthermore, although IMP3 is not a specific diagnostic marker to distinguish the tumors studied, it seems to mediate cell adhesion and migration in these tumors. Further studies should be performed to better understand the IMP3 biology in salivary gland tumors.
Collapse
|
38
|
Lo PK, Zhang Y, Yao Y, Wolfson B, Yu J, Han SY, Duru N, Zhou Q. Tumor-associated myoepithelial cells promote the invasive progression of ductal carcinoma in situ through activation of TGFβ signaling. J Biol Chem 2017; 292:11466-11484. [PMID: 28512126 DOI: 10.1074/jbc.m117.775080] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 05/12/2017] [Indexed: 12/18/2022] Open
Abstract
The normal myoepithelium has a tumor-suppressing nature and inhibits the progression of ductal carcinoma in situ (DCIS) into invasive ductal carcinoma (IDC). Conversely, a growing number of studies have shown that tumor-associated myoepithelial cells have a tumor-promoting effect. Moreover, the exact role of tumor-associated myoepithelial cells in the DCIS-to-IDC development remains undefined. To address this, we explored the role of tumor-associated myoepithelial cells in the DCIS-to-IDC progression. We developed a direct coculture system to study the cell-cell interactions between DCIS cells and tumor-associated myoepithelial cells. Coculture studies indicated that tumor-associated myoepithelial cells promoted the invasive progression of a DCIS cell model in vitro, and mechanistic studies revealed that the interaction with DCIS cells stimulated tumor-associated myoepithelial cells to secrete TGFβ1, which subsequently contributed to activating the TGFβ/Smads pathway in DCIS cells. We noted that activation of the TGFβ signaling pathway promoted the epithelial-mesenchymal transition, basal-like phenotypes, stemness, and invasiveness of DCIS cells. Importantly, xenograft studies further demonstrated that tumor-associated myoepithelial cells enhanced the DCIS-to-IDC progression in vivo Furthermore, we found that TGFβ-mediated induction of oncogenic miR-10b-5p expression and down-regulation of RB1CC1, a miR-10b-5p-targeted tumor-suppressor gene, contributed to the invasive progression of DCIS. Our findings provide the first experimental evidence to directly support the paradigm that altered DCIS-associated myoepithelial cells promote the invasive progression of DCIS into IDC via TGFβ signaling activation.
Collapse
Affiliation(s)
- Pang-Kuo Lo
- From the Department of Biochemistry and Molecular Biology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland 21201 and
| | - Yongshu Zhang
- From the Department of Biochemistry and Molecular Biology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland 21201 and
| | - Yuan Yao
- From the Department of Biochemistry and Molecular Biology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland 21201 and
| | - Benjamin Wolfson
- From the Department of Biochemistry and Molecular Biology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland 21201 and
| | - Justine Yu
- From the Department of Biochemistry and Molecular Biology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland 21201 and
| | - Shu-Yan Han
- From the Department of Biochemistry and Molecular Biology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland 21201 and.,the Key Laboratory of Carcinogenesis and Translational Research, Department of Integration of Chinese and Western Medicine, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Nadire Duru
- From the Department of Biochemistry and Molecular Biology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland 21201 and
| | - Qun Zhou
- From the Department of Biochemistry and Molecular Biology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland 21201 and
| |
Collapse
|
39
|
Sameni M, Cavallo-Medved D, Franco OE, Chalasani A, Ji K, Aggarwal N, Anbalagan A, Chen X, Mattingly RR, Hayward SW, Sloane BF. Pathomimetic avatars reveal divergent roles of microenvironment in invasive transition of ductal carcinoma in situ. Breast Cancer Res 2017; 19:56. [PMID: 28506312 PMCID: PMC5433063 DOI: 10.1186/s13058-017-0847-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 04/25/2017] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND The breast tumor microenvironment regulates progression of ductal carcinoma in situ (DCIS) to invasive ductal carcinoma (IDC). However, it is unclear how interactions between breast epithelial and stromal cells can drive this progression and whether there are reliable microenvironmental biomarkers to predict transition of DCIS to IDC. METHODS We used xenograft mouse models and a 3D pathomimetic model termed mammary architecture and microenvironment engineering (MAME) to study the interplay between human breast myoepithelial cells (MEPs) and cancer-associated fibroblasts (CAFs) on DCIS progression. RESULTS Our results show that MEPs suppress tumor formation by DCIS cells in vivo even in the presence of CAFs. In the in vitro MAME model, MEPs reduce the size of 3D DCIS structures and their degradation of extracellular matrix. We further show that the tumor-suppressive effects of MEPs on DCIS are linked to inhibition of urokinase plasminogen activator (uPA)/urokinase plasminogen activator receptor (uPAR)-mediated proteolysis by plasminogen activator inhibitor 1 (PAI-1) and that they can lessen the tumor-promoting effects of CAFs by attenuating interleukin 6 (IL-6) signaling pathways. CONCLUSIONS Our studies using MAME are, to our knowledge, the first to demonstrate a divergent interplay between MEPs and CAFs within the DCIS tumor microenvironment. We show that the tumor-suppressive actions of MEPs are mediated by PAI-1, uPA and its receptor, uPAR, and are sustained even in the presence of the CAFs, which themselves enhance DCIS tumorigenesis via IL-6 signaling. Identifying tumor microenvironmental regulators of DCIS progression will be critical for defining a robust and predictive molecular signature for clinical use.
Collapse
Affiliation(s)
- Mansoureh Sameni
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201 USA
| | - Dora Cavallo-Medved
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201 USA
- Department of Biological Sciences, University of Windsor, Windsor, ON N9B 3P4 Canada
| | - Omar E. Franco
- Department of Surgery, NorthShore University HealthSystem Research Institute, Evanston, IL 60201 USA
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, TN 37232 USA
| | - Anita Chalasani
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201 USA
| | - Kyungmin Ji
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201 USA
| | - Neha Aggarwal
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201 USA
| | - Arulselvi Anbalagan
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201 USA
| | - Xuequn Chen
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201 USA
| | - Raymond R. Mattingly
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201 USA
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201 USA
| | - Simon W. Hayward
- Department of Surgery, NorthShore University HealthSystem Research Institute, Evanston, IL 60201 USA
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, TN 37232 USA
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37232 USA
| | - Bonnie F. Sloane
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201 USA
- Department of Biological Sciences, University of Windsor, Windsor, ON N9B 3P4 Canada
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201 USA
| |
Collapse
|
40
|
Sarper M, Allen MD, Gomm J, Haywood L, Decock J, Thirkettle S, Ustaoglu A, Sarker SJ, Marshall J, Edwards DR, Jones JL. Loss of MMP-8 in ductal carcinoma in situ (DCIS)-associated myoepithelial cells contributes to tumour promotion through altered adhesive and proteolytic function. Breast Cancer Res 2017; 19:33. [PMID: 28330493 PMCID: PMC5363009 DOI: 10.1186/s13058-017-0822-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 03/02/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Normal myoepithelial cells (MECs) play an important tumour-suppressor role in the breast but display an altered phenotype in ductal carcinoma in situ (DCIS), gaining tumour-promoter functions. Matrix metalloproteinase-8 (MMP-8) is expressed by normal MECs but is lost in DCIS. This study investigated the function of MMP-8 in MECs and the impact of its loss in DCIS. METHODS Primary normal and DCIS-associated MECs, and normal (N-1089) and DCIS-modified myoepithelial (β6-1089) cell lines, were used to assess MMP-8 expression and function. β6-1089 lacking MMP-8 were transfected with MMP-8 WT and catalytically inactive MMP-8 EA, and MMP-8 in N-1089 MEC was knocked down with siRNA. The effect on adhesion and migration to extracellular matrix (ECM), localisation of α6β4 integrin to hemidesmosomes (HD), TGF-β signalling and gelatinase activity was measured. The effect of altering MEC MMP-8 expression on tumour cell invasion was investigated in 2D and 3D organotypic models. RESULTS Assessment of primary cells and MEC lines confirmed expression of MMP-8 in normal MEC and its loss in DCIS-MEC. Over-expression of MMP-8 WT but not MMP-8 EA in β6-1089 cells increased adhesion to ECM proteins and reduced migration. Conversely, knock-down of MMP-8 in N-1089 reduced adhesion and increased migration. Expression of MMP-8 WT in β6-1089 led to greater localisation of α6β4 to HD and reduced retraction fibre formation, this being reversed by MMP-8 knock-down in N-1089. Over-expression of MMP-8 WT reduced TGF-β signalling and gelatinolytic activity. MMP-8 knock-down enhanced TGF-β signalling and gelatinolytic activity, which was reversed by blocking MMP-9 by knock-down or an inhibitor. MMP-8 WT but not MMP-8 EA over-expression in β6-1089 reduced breast cancer cell invasion in 2D and 3D invasion assays, while MMP-8 knock-down in N-1089 enhanced cancer cell invasion. Staining of breast cancer cases for MMP-8 revealed a statistically significant loss of MMP-8 expression in DCIS with invasion versus pure DCIS (p = 0.001). CONCLUSIONS These data indicate MMP-8 is a vital component of the myoepithelial tumour-suppressor function. It restores MEC interaction with the matrix, opposes TGF-β signalling and MMP-9 proteolysis, which contributes to inhibition of tumour cell invasion. Assessment of MMP-8 expression may help to determine risk of DCIS progression.
Collapse
Affiliation(s)
- Muge Sarper
- Translational Cancer Discovery Team, CRUK Cancer Therapeutics Unit, Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, UK
| | - Michael D Allen
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.
| | - Jenny Gomm
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Linda Haywood
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Julie Decock
- Cancer Research Centre, Qatar Biomedical Research Institute, Qatar Foundation, Doha, Qatar
| | - Sally Thirkettle
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Ahsen Ustaoglu
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Shah-Jalal Sarker
- Centre for Experimental Cancer Medicine, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - John Marshall
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Dylan R Edwards
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - J Louise Jones
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.
| |
Collapse
|
41
|
Simmons A, Burrage PM, Nicolau DV, Lakhani SR, Burrage K. Environmental factors in breast cancer invasion: a mathematical modelling review. Pathology 2017; 49:172-180. [PMID: 28081961 DOI: 10.1016/j.pathol.2016.11.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 11/07/2016] [Accepted: 11/13/2016] [Indexed: 12/17/2022]
Abstract
This review presents a brief overview of breast cancer, focussing on its heterogeneity and the role of mathematical modelling and simulation in teasing apart the underlying biophysical processes. Following a brief overview of the main known pathophysiological features of ductal carcinoma, attention is paid to differential equation-based models (both deterministic and stochastic), agent-based modelling, multi-scale modelling, lattice-based models and image-driven modelling. A number of vignettes are presented where these modelling approaches have elucidated novel aspects of breast cancer dynamics, and we conclude by offering some perspectives on the role mathematical modelling can play in understanding breast cancer development, invasion and treatment therapies.
Collapse
Affiliation(s)
- Alex Simmons
- School of Mathematical Sciences, and ARC Centre of Excellence for Mathematical and Statistical Frontiers, Queensland University of Technology, Gardens Point, Brisbane, Qld, Australia
| | - Pamela M Burrage
- School of Mathematical Sciences, and ARC Centre of Excellence for Mathematical and Statistical Frontiers, Queensland University of Technology, Gardens Point, Brisbane, Qld, Australia
| | - Dan V Nicolau
- School of Mathematical Sciences, and ARC Centre of Excellence for Mathematical and Statistical Frontiers, Queensland University of Technology, Gardens Point, Brisbane, Qld, Australia; Mathematical Institute, University of Oxford, Oxford, United Kingdom; Molecular Sense Ltd, Oxford, United Kingdom
| | - Sunil R Lakhani
- The University of Queensland, Centre for Clinical Research and School of Medicine and Pathology Queensland, The Royal Brisbane and Women's Hospital, Brisbane, Qld, Australia
| | - Kevin Burrage
- School of Mathematical Sciences, and ARC Centre of Excellence for Mathematical and Statistical Frontiers, Queensland University of Technology, Gardens Point, Brisbane, Qld, Australia; Department of Computer Science, University of Oxford, United Kingdom.
| |
Collapse
|
42
|
|
43
|
Yeong J, Thike AA, Tan PH, Iqbal J. Identifying progression predictors of breast ductal carcinoma in situ. J Clin Pathol 2016; 70:102-108. [PMID: 27864452 DOI: 10.1136/jclinpath-2016-204154] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 10/07/2016] [Indexed: 01/08/2023]
Abstract
Ductal carcinoma in situ (DCIS) refers to neoplastic epithelial cells proliferating within the mammary ducts of the breast, which have not breached the basement membrane nor invaded surrounding tissues. Traditional thinking holds that DCIS represents an early step in a linear progression towards invasive ductal carcinoma (IDC). However, as only approximately half of DCIS cases progress to IDC, important questions around the key determinants of malignant progression need to be answered. Recent studies have revealed that molecular differences between DCIS and IDC cells are not found at the genomic level; instead, altered patterns of gene expression and post-translational regulation lead to distinct transcriptomic and proteomic profiles. Therefore, understanding malignant progression will require a different approach that takes into account the diverse tumour cell extrinsic factors driving changes in tumour cell gene expression necessary for the invasive phenotype. Here, we review the roles of the tumour stroma (including mesenchymal cells, immune cells and the extracellular matrix) and myoepithelial cells in malignant progression and make a case for a more integrated approach to the study and assessment of DCIS and its progression, or lack thereof, to invasive disease.
Collapse
Affiliation(s)
- Joe Yeong
- Division of Pathology, Singapore General Hospital, Singapore, Singapore.,Singapore Immunology Network (SIgN), Agency of Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Aye Aye Thike
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Puay Hoon Tan
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Jabed Iqbal
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| |
Collapse
|
44
|
HER2 induced EMT and tumorigenicity in breast epithelial progenitor cells is inhibited by coexpression of EGFR. Oncogene 2015; 35:4244-55. [PMID: 26686087 PMCID: PMC4981873 DOI: 10.1038/onc.2015.489] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 11/11/2015] [Accepted: 11/14/2015] [Indexed: 02/06/2023]
Abstract
The members of the epidermal growth factor receptor (EGFR) kinase family are important players in breast morphogenesis and cancer. EGFR2/HER2 and EGFR expression have a prognostic value in certain subtypes of breast cancer such as HER2-amplified, basal-like and luminal type B. Many clinically approved small molecular inhibitors and monoclonal antibodies have been designed to target HER2, EGFR or both. There is, however, still limited knowledge on how the two receptors are expressed in normal breast epithelium, what effects they have on cellular differentiation and how they participate in neoplastic transformation. D492 is a breast epithelial cell line with stem cell properties that can undergo epithelial to mesenchyme transition (EMT), generate luminal- and myoepithelial cells and form complex branching structures in three-dimensional (3D) culture. Here, we show that overexpression of HER2 in D492 (D492HER2) resulted in EMT, loss of contact growth inhibition and increased oncogenic potential in vivo. HER2 overexpression, furthermore, inhibited endogenous EGFR expression. Re-introducing EGFR in D492HER2 (D492HER2/EGFR) partially reversed the mesenchymal state of the cells, as an epithelial phenotype reappeared both in 3D cultures and in vivo. The D492HER2/EGFR xenografts grow slower than the D492HER2 tumors, while overexpression of EGFR alone (D492EGFR) was not oncogenic in vivo. Consistent with the EGFR-mediated epithelial phenotype, overexpression of EGFR drove the cells toward a myoepithelial phenotype in 3D culture. The effect of two clinically approved anti-HER2 and EGFR therapies, trastuzumab and cetuximab, was tested alone and in combination on D492HER2 xenografts. While trastuzumab had a growth inhibitory effect compared with untreated control, the effect of cetuximab was limited. When administered in combination, the growth inhibitory effect of trastuzumab was less pronounced. Collectively, our data indicate that in HER2-overexpressing D492 cells, EGFR can behave as a tumor suppressor, by pushing the cells towards epithelial differentiation.
Collapse
|
45
|
Russell TD, Jindal S, Agunbiade S, Gao D, Troxell M, Borges VF, Schedin P. Myoepithelial cell differentiation markers in ductal carcinoma in situ progression. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:3076-89. [PMID: 26343330 PMCID: PMC4630168 DOI: 10.1016/j.ajpath.2015.07.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Revised: 07/14/2015] [Accepted: 07/21/2015] [Indexed: 11/20/2022]
Abstract
We describe a preclinical model that investigates progression of early-stage ductal carcinoma in situ (DCIS) and report that compromised myoepithelial cell differentiation occurs before transition to invasive disease. Human breast cancer MCF10DCIS.com cells were delivered into the mouse mammary teat by intraductal injection in the absence of surgical manipulations and accompanying wound-healing confounders. DCIS-like lesions developed throughout the mammary ducts with full representation of human DCIS histologic patterns. Tumor cells were incorporated into the normal mammary epithelium, developed ductal intraepithelial neoplasia and DCIS, and progressed to invasive carcinoma, suggesting the model provides a rigorous approach to study early stages of breast cancer progression. Mammary glands were evaluated for myoepithelium integrity with immunohistochemical assays. Progressive loss of the myoepithelial cell differentiation markers p63, calponin, and α-smooth muscle actin was observed in the mouse myoepithelium surrounding DCIS-involved ducts. p63 loss was an early indicator, calponin loss intermediate, and α-smooth muscle actin a later indicator of compromised myoepithelium. Loss of myoepithelial calponin was specifically associated with gain of the basal marker p63 in adjacent tumor cells. In single time point biopsies obtained from 16 women diagnosed with pure DCIS, a similar loss in myoepithelial cell markers was observed. These results suggest that further research is warranted into the role of myoepithelial cell p63 and calponin expression on DCIS progression to invasive disease.
Collapse
Affiliation(s)
- Tanya D Russell
- Division of Medical Oncology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Sonali Jindal
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Samiat Agunbiade
- Division of Medical Oncology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Dexiang Gao
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Megan Troxell
- Department of Pathology, Oregon Health & Science University, Portland, Oregon; Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Virginia F Borges
- Division of Medical Oncology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; University of Colorado Cancer Center, Aurora, Colorado
| | - Pepper Schedin
- Division of Medical Oncology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon; Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon.
| |
Collapse
|
46
|
Sánchez-Céspedes R, Millán Y, Guil-Luna S, Reymundo C, Espinosa de Los Monteros A, Martín de Las Mulas J. Myoepithelial cells in canine mammary tumours. Vet J 2015; 207:45-52. [PMID: 26639832 DOI: 10.1016/j.tvjl.2015.10.035] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 10/07/2015] [Accepted: 10/10/2015] [Indexed: 02/05/2023]
Abstract
Mammary tumours are the most common neoplasms of female dogs. Compared to mammary tumours of humans and cats, myoepithelial (ME) cell involvement is common in canine mammary tumours (CMT) of any subtype. Since ME cell involvement in CMT influences both histogenetic tumour classification and prognosis, correct identification of ME cells is important. This review describes immunohistochemical methods for identification of canine mammary ME cells used in vivo. In addition, phenotypic and genotypic methods to isolate ME cells for in vitro studies to analyse tumour-suppressor protein production and gene expression are discussed. The contribution of ME cells to both histogenetic classifications and the prognosis of CMT is compared with other species and the potential use of ME cells as a method to identify carcinoma in situ is discussed.
Collapse
Affiliation(s)
| | - Yolanda Millán
- Department of Comparative Pathology, University of Córdoba, 14014 Córdoba, Spain
| | - Silvia Guil-Luna
- Department of Comparative Pathology, University of Córdoba, 14014 Córdoba, Spain
| | - Carlos Reymundo
- Department of Pathology, University of Córdoba, 14071 Córdoba, Spain
| | - Antonio Espinosa de Los Monteros
- Unit of Histology and Animal Pathology, Institute for Animal Health, Veterinary School, University of Las Palmas de Gran Canaria, 35413 Las Palmas, Spain
| | | |
Collapse
|
47
|
Krasnapolski MA, Lodillinsky C, Bal De Kier Joffé E, Eiján AM. Hypoxia-induced nitric oxide release by luminal cells stimulates proliferation and uPA secretion of myoepithelial cells in a bicellular murine mammary tumor. J Cancer Res Clin Oncol 2015; 141:1727-38. [PMID: 25687381 DOI: 10.1007/s00432-015-1934-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 02/05/2015] [Indexed: 12/28/2022]
Abstract
INTRODUCTION LM38 murine mammary adenocarcinoma model is formed by LM38-LP (myoepithelial and luminal), LM38-HP (luminal) and LM38-D2 (myoepithelial) cell lines. In a previous work, we had shown that LM38-HP and LM38-D2 cell lines are less malignant than the bicellular LM38-LP cell line. PURPOSE To study the role of nitric oxide (NO) as one of the mediators of functional interactions between malignant luminal and myoepithelial cells. METHODS AND RESULTS Using immunohistochemistry, in vivo iNOS expression was only detected in the luminal cells of bicellular LM38-LP and most cells of LM38-HP tumors. In cobalt-induced pseudohypoxia, LM38-LP and LM38-HP cell lines significantly increased HIF-1α and iNOS expression (Western blotting) and therefore NO production (Griess method). This increase was inhibited by the iNOS inhibitor 1400 W. On the other side, NO was not detectable in LM38-D2 cells either in basal or in pseudohypoxia. In addition, pseudohypoxia increased urokinase-type plasminogen activator (uPA) secretion by LM38-LP and LM38-HP cells and migration in the LM38-LP cell line, without modulating these properties in LM38-D2 cells (radial caseinolysis). The NO donor DETA/NONOate (500 μM) was able to increase uPA secretion and in vitro growth of LM38-D2. In agreement, 1400 W prevented in vivo growth of the myoepithelial LM38-D2 cells. CONCLUSIONS Hypoxia leads to an enhanced NO production by the luminal component, through HIF-1α and iNOS, which can stimulate myoepithelial cell proliferation and uPA secretion. In these new conditions, myoepithelial cells might act as an invasive forefront generating gaps that could help luminal cells to escape from the primary tumor.
Collapse
|
48
|
Giussani M, Merlino G, Cappelletti V, Tagliabue E, Daidone MG. Tumor-extracellular matrix interactions: Identification of tools associated with breast cancer progression. Semin Cancer Biol 2015; 35:3-10. [PMID: 26416466 DOI: 10.1016/j.semcancer.2015.09.012] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 09/23/2015] [Indexed: 12/18/2022]
Abstract
Several evidences support the concept that cancer development and progression are not entirely cancer cell-autonomous processes, but may be influenced, and possibly driven, by cross-talk between cancer cells and the surrounding microenvironment in which, besides immune cells, stromal cells and extracellular matrix (ECM) play a major role in regulating distinct biologic processes. Stroma and ECM-related signatures proved to influence breast cancer progression, and to contribute to the identification of tumor phenotypes resistant to cytotoxic and hormonal treatments. The possible clinical implications of the interplay between tumor cells and the microenvironment, with special reference to ECM remodelling, will be discussed in this review.
Collapse
Affiliation(s)
- Marta Giussani
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G.A. Amadeo, 42, 20133 Milan, Italy.
| | - Giuseppe Merlino
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G.A. Amadeo, 42, 20133 Milan, Italy.
| | - Vera Cappelletti
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G.A. Amadeo, 42, 20133 Milan, Italy.
| | - Elda Tagliabue
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G.A. Amadeo, 42, 20133 Milan, Italy.
| | - Maria Grazia Daidone
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G.A. Amadeo, 42, 20133 Milan, Italy.
| |
Collapse
|
49
|
Soysal SD, Tzankov A, Muenst SE. Role of the Tumor Microenvironment in Breast Cancer. Pathobiology 2015; 82:142-52. [DOI: 10.1159/000430499] [Citation(s) in RCA: 213] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
50
|
Acasigua GA, Warner KA, Nör F, Helman J, Pearson AT, Fossati AC, Wang S, Nör JE. BH3-mimetic small molecule inhibits the growth and recurrence of adenoid cystic carcinoma. Oral Oncol 2015; 51:839-47. [PMID: 26121939 DOI: 10.1016/j.oraloncology.2015.06.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 06/12/2015] [Accepted: 06/13/2015] [Indexed: 11/20/2022]
Abstract
OBJECTIVES To evaluate the anti-tumor effect of BM-1197, a new potent and highly specific small molecule inhibitor of Bcl-2/Bcl-xL, in preclinical models of human adenoid cystic carcinoma (ACC). METHODS Low passage primary human adenoid cystic carcinoma cells (UM-HACC-2A,-2B,-5,-6) and patient-derived xenograft (PDX) models (UM-PDX-HACC) were developed from surgical specimens obtained from 4 patients. The effect of BM-1197 on cell viability and cell cycle were evaluated in vitro using this panel of low passage ACC cells. The effect of BM-1197 on tumor growth, recurrence and tumor cell apoptosis in vivo was evaluated with the PDX model of ACC (UM-PDX-HACC-5). RESULTS Exposure of low passage primary human ACC cells to BM-1197 mediated an IC50 of 0.92-2.82 μM. This correlated with an increase in the fraction of apoptotic cells (p<0.0001) and an increase in caspase-3 activity (p<0.0001), but no noticeable differences in cell cycle (p>0.05). In vivo, BM-1197 inhibited tumor growth (p=0.0256) and induced tumor cell apoptosis (p=0.0165) without causing significant systemic toxicities, as determined by mouse weight over time. Surprisingly, weekly BM-1197 decreased the incidence of tumor recurrence (p=0.0297), as determined by Kaplan-Meier analysis. CONCLUSION These data demonstrated that single agent BM-1197 induces apoptosis and inhibits tumor growth in preclinical models of adenoid cystic carcinoma. Notably, single agent BM-1197 inhibited tumor recurrence, which is considered a major clinical challenge in the clinical management of adenoid cystic carcinoma. Collectively, these results suggest that patients with adenoid cystic carcinoma might benefit from therapy with a BH3-mimetic small molecule.
Collapse
Affiliation(s)
- Gerson A Acasigua
- Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109-1078, United States; Department of Morphological Sciences, Federal University of Rio Grande do Sul, Brazil
| | - Kristy A Warner
- Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109-1078, United States
| | - Felipe Nör
- Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109-1078, United States; Department of Morphological Sciences, Federal University of Rio Grande do Sul, Brazil
| | - Joseph Helman
- Department of Oral and Maxillofacial Surgery, University of Michigan School of Dentistry, United States; Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, United States
| | - Alexander T Pearson
- Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109-1078, United States; Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, United States
| | - Anna C Fossati
- Department of Morphological Sciences, Federal University of Rio Grande do Sul, Brazil
| | - Shaomeng Wang
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, United States; Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, United States
| | - Jacques E Nör
- Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109-1078, United States; Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, United States; Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI, United States; Department of Otolaryngology, University of Michigan School of Medicine, Ann Arbor, MI, United States.
| |
Collapse
|