1
|
Moragas N, Fernandez-Nogueira P, Recalde-Percaz L, Inman JL, López-Plana A, Bergholtz H, Noguera-Castells A, Del Burgo PJ, Chen X, Sorlie T, Gascón P, Bragado P, Bissell M, Carbó N, Fuster G. The SEMA3F-NRP1/NRP2 axis is a key factor in the acquisition of invasive traits in in situ breast ductal carcinoma. Breast Cancer Res 2024; 26:122. [PMID: 39138514 PMCID: PMC11320849 DOI: 10.1186/s13058-024-01871-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 07/12/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND A better understanding of ductal carcinoma in situ (DCIS) is urgently needed to identify these preinvasive lesions as distinct clinical entities. Semaphorin 3F (SEMA3F) is a soluble axonal guidance molecule, and its coreceptors Neuropilin 1 (NRP1) and NRP2 are strongly expressed in invasive epithelial BC cells. METHODS We utilized two cell line models to represent the progression from a healthy state to the mild-aggressive or ductal carcinoma in situ (DCIS) stage and, ultimately, to invasive cell lines. Additionally, we employed in vivo models and conducted analyses on patient databases to ensure the translational relevance of our results. RESULTS We revealed SEMA3F as a promoter of invasion during the DCIS-to-invasive ductal carcinoma transition in breast cancer (BC) through the action of NRP1 and NRP2. In epithelial cells, SEMA3F activates epithelialmesenchymal transition, whereas it promotes extracellular matrix degradation and basal membrane and myoepithelial cell layer breakdown. CONCLUSIONS Together with our patient database data, these proof-of-concept results reveal new SEMA3F-mediated mechanisms occurring in the most common preinvasive BC lesion, DCIS, and represent potent and direct activation of its transition to invasion. Moreover, and of clinical and therapeutic relevance, the effects of SEMA3F can be blocked directly through its coreceptors, thus preventing invasion and keeping DCIS lesions in the preinvasive state.
Collapse
MESH Headings
- Humans
- Neuropilin-1/metabolism
- Neuropilin-1/genetics
- Female
- Breast Neoplasms/pathology
- Breast Neoplasms/metabolism
- Breast Neoplasms/genetics
- Neuropilin-2/metabolism
- Neuropilin-2/genetics
- Neoplasm Invasiveness
- Carcinoma, Intraductal, Noninfiltrating/metabolism
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Carcinoma, Intraductal, Noninfiltrating/genetics
- Cell Line, Tumor
- Nerve Tissue Proteins/metabolism
- Nerve Tissue Proteins/genetics
- Epithelial-Mesenchymal Transition/genetics
- Animals
- Membrane Proteins/metabolism
- Membrane Proteins/genetics
- Mice
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/genetics
- Gene Expression Regulation, Neoplastic
- Signal Transduction
Collapse
Affiliation(s)
- Núria Moragas
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona (UB), 08028, Barcelona, Spain
- Institute of Biomedicine of the Universitat de Barcelona (IBUB), Barcelona, Spain
| | - Patricia Fernandez-Nogueira
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona (UB), 08028, Barcelona, Spain
- Institute of Biomedicine of the Universitat de Barcelona (IBUB), Barcelona, Spain
- Department of Biomedicine, School of Medicine, Universitat de Barcelona (UB), 08036, Barcelona, Spain
| | - Leire Recalde-Percaz
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona (UB), 08028, Barcelona, Spain
- Institute of Biomedicine of the Universitat de Barcelona (IBUB), Barcelona, Spain
| | - Jamie L Inman
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Rd., Berkeley, CA, 94720, USA
| | - Anna López-Plana
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona (UB), 08028, Barcelona, Spain
- Institute of Biomedicine of the Universitat de Barcelona (IBUB), Barcelona, Spain
| | - Helga Bergholtz
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, 0450, Oslo, Norway
| | - Aleix Noguera-Castells
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona (UB), 08028, Barcelona, Spain
- Institute of Biomedicine of the Universitat de Barcelona (IBUB), Barcelona, Spain
- Cancer Epigenetics Group, Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Catalonia, Spain
- Centro de Investigacion Biomedica en Red Cancer (CIBERONC), Madrid, Spain
- Department of Biosciences, Faculty of Science, Technology and Engineering, University of Vic - Central University of Catalonia (UVic-UCC), Vic, Barcelona, Catalonia, Spain
| | - Pedro J Del Burgo
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona (UB), 08028, Barcelona, Spain
| | - Xieng Chen
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona (UB), 08028, Barcelona, Spain
| | - Therese Sorlie
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, 0450, Oslo, Norway
| | - Pere Gascón
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona (UB), 08028, Barcelona, Spain
| | - Paloma Bragado
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Universidad Complutense de Madrid, Health Research Institute of the Hospital Clínico San Carlos, 28040, Madrid, Spain
| | - Mina Bissell
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Rd., Berkeley, CA, 94720, USA
| | - Neus Carbó
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona (UB), 08028, Barcelona, Spain
- Institute of Biomedicine of the Universitat de Barcelona (IBUB), Barcelona, Spain
| | - Gemma Fuster
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona (UB), 08028, Barcelona, Spain.
- Institute of Biomedicine of the Universitat de Barcelona (IBUB), Barcelona, Spain.
- Tissue Repair and Regeneration Laboratory (TR2Lab), Institute of Research and Innovation of Life Sciences and Health, Catalunya Central (IRIS-CC), UVIC-UCC, Vic, Spain.
| |
Collapse
|
2
|
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
|
3
|
Wang J, Li B, Luo M, Huang J, Zhang K, Zheng S, Zhang S, Zhou J. Progression from ductal carcinoma in situ to invasive breast cancer: molecular features and clinical significance. Signal Transduct Target Ther 2024; 9:83. [PMID: 38570490 PMCID: PMC10991592 DOI: 10.1038/s41392-024-01779-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 02/14/2024] [Accepted: 02/26/2024] [Indexed: 04/05/2024] Open
Abstract
Ductal carcinoma in situ (DCIS) represents pre-invasive breast carcinoma. In untreated cases, 25-60% DCIS progress to invasive ductal carcinoma (IDC). The challenge lies in distinguishing between non-progressive and progressive DCIS, often resulting in over- or under-treatment in many cases. With increasing screen-detected DCIS in these years, the nature of DCIS has aroused worldwide attention. A deeper understanding of the biological nature of DCIS and the molecular journey of the DCIS-IDC transition is crucial for more effective clinical management. Here, we reviewed the key signaling pathways in breast cancer that may contribute to DCIS initiation and progression. We also explored the molecular features of DCIS and IDC, shedding light on the progression of DCIS through both inherent changes within tumor cells and alterations in the tumor microenvironment. In addition, valuable research tools utilized in studying DCIS including preclinical models and newer advanced technologies such as single-cell sequencing, spatial transcriptomics and artificial intelligence, have been systematically summarized. Further, we thoroughly discussed the clinical advancements in DCIS and IDC, including prognostic biomarkers and clinical managements, with the aim of facilitating more personalized treatment strategies in the future. Research on DCIS has already yielded significant insights into breast carcinogenesis and will continue to pave the way for practical clinical applications.
Collapse
Affiliation(s)
- Jing Wang
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China
| | - Baizhou Li
- Department of Pathology, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Meng Luo
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China
- Department of Plastic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jia Huang
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China
| | - Kun Zhang
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shu Zheng
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China
| | - Suzhan Zhang
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China.
| | - Jiaojiao Zhou
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
4
|
Behbod F, Chen JH, Thompson A. Human Ductal Carcinoma In Situ: Advances and Future Perspectives. Cold Spring Harb Perspect Med 2023; 13:a041319. [PMID: 36781223 PMCID: PMC10547390 DOI: 10.1101/cshperspect.a041319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
Due to widespread adoption of screening mammography, there has been a significant increase in new diagnoses of ductal carcinoma in situ (DCIS). However, DCIS outcomes remain unclear. A large fraction of human DCIS (>50%) may not need the multimodality treatment options currently offered to all DCIS patients. More importantly, while we may be overtreating many, we cannot identify those most at risk of invasion or metastasis following a DCIS diagnosis. This review summarizes the studies that have furthered our understanding of DCIS pathology and mechanisms of invasive progression by using advanced technologies including spatial genomics, transcriptomics, and multiplex proteomics. This review also highlights a need for rethinking DCIS with a more focused view on epithelial states and programs and their cross talk with the microenvironment.
Collapse
Affiliation(s)
- Fariba Behbod
- Department of Pathology and Laboratory Medicine, MS 3045, The University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Jennifer H Chen
- Michael E. Debakey Department of Surgery, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Alastair Thompson
- Section of Breast Surgery, Baylor College of Medicine, Co-Director, Lester and Sue Smith Breast Center, Dan L Duncan Comprehensive Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
5
|
Gibson SV, Madzharova E, Tan AC, Allen MD, Keller UAD, Louise Jones J, Carter EP, Grose RP. ADAMTS3 restricts cancer invasion in models of early breast cancer progression through enhanced fibronectin degradation. Matrix Biol 2023; 121:74-89. [PMID: 37336268 DOI: 10.1016/j.matbio.2023.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/16/2023] [Accepted: 06/16/2023] [Indexed: 06/21/2023]
Abstract
Proteases have long been associated with cancer progression, due to their ability to facilitate invasion upon matrix remodelling. However, proteases are not simply degraders of the matrix, but also play fundamental roles in modulating cellular behaviour through the proteolytic processing of specific substrates. Indeed, proteases can elicit both pro- and anti- tumorigenic effects depending on context. Using a heterocellular spheroid model of breast cancer progression, we demonstrate the repressive function of myoepithelial ADAMTS3, with its loss directing myoepithelial-led invasion of luminal cells through a physiologically relevant matrix. Degradomic analysis, using terminal amine isotopic labelling of substrates (TAILS), combined with functional assays, implicate ADAMTS3 as a mediator of fibronectin degradation. We show further that loss of ADAMTS3 enhances levels of fibronectin in the microenvironment, promoting invasion through canonical integrin α5β1 activation. Our data highlight a tumour suppressive role for ADAMTS3 in early stage breast cancer, and contribute to the growing evidence that proteases can restrain cancer progression.
Collapse
Affiliation(s)
- Shayin V Gibson
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Elizabeta Madzharova
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Amandine C Tan
- Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, SE5 8AF, UK
| | - Michael D Allen
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Ulrich Auf dem Keller
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - 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; Department of Life Sciences, University of Bath, Claverton Down, Bath BA2 7AY, UK.
| | - Richard P Grose
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK.
| |
Collapse
|
6
|
Hutten SJ, de Bruijn R, Lutz C, Badoux M, Eijkman T, Chao X, Ciwinska M, Sheinman M, Messal H, Herencia-Ropero A, Kristel P, Mulder L, van der Waal R, Sanders J, Almekinders MM, Llop-Guevara A, Davies HR, van Haren MJ, Martin NI, Behbod F, Nik-Zainal S, Serra V, van Rheenen J, Lips EH, Wessels LFA, Wesseling J, Scheele CLGJ, Jonkers J. A living biobank of patient-derived ductal carcinoma in situ mouse-intraductal xenografts identifies risk factors for invasive progression. Cancer Cell 2023; 41:986-1002.e9. [PMID: 37116492 PMCID: PMC10171335 DOI: 10.1016/j.ccell.2023.04.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 02/21/2023] [Accepted: 04/04/2023] [Indexed: 04/30/2023]
Abstract
Ductal carcinoma in situ (DCIS) is a non-obligate precursor of invasive breast cancer (IBC). Due to a lack of biomarkers able to distinguish high- from low-risk cases, DCIS is treated similar to early IBC even though the minority of untreated cases eventually become invasive. Here, we characterized 115 patient-derived mouse-intraductal (MIND) DCIS models reflecting the full spectrum of DCIS observed in patients. Utilizing the possibility to follow the natural progression of DCIS combined with omics and imaging data, we reveal multiple prognostic factors for high-risk DCIS including high grade, HER2 amplification, expansive 3D growth, and high burden of copy number aberrations. In addition, sequential transplantation of xenografts showed minimal phenotypic and genotypic changes over time, indicating that invasive behavior is an intrinsic phenotype of DCIS and supporting a multiclonal evolution model. Moreover, this study provides a collection of 19 distributable DCIS-MIND models spanning all molecular subtypes.
Collapse
Affiliation(s)
- Stefan J Hutten
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands; Oncode Institute, Amsterdam, the Netherlands
| | - Roebi de Bruijn
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands; Oncode Institute, Amsterdam, the Netherlands; Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands
| | - Catrin Lutz
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands; Oncode Institute, Amsterdam, the Netherlands
| | - Madelon Badoux
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands; Oncode Institute, Amsterdam, the Netherlands
| | - Timo Eijkman
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands; Oncode Institute, Amsterdam, the Netherlands
| | - Xue Chao
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands; Oncode Institute, Amsterdam, the Netherlands
| | - Marta Ciwinska
- Center for Cancer Biology, VIB, Department of Oncology, KU Leuven, 3000 Leuven, Belgium
| | - Michael Sheinman
- Oncode Institute, Amsterdam, the Netherlands; Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands
| | - Hendrik Messal
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands; Oncode Institute, Amsterdam, the Netherlands
| | - Andrea Herencia-Ropero
- Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain; Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona, Barcelona, Spain
| | - Petra Kristel
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands
| | - Lennart Mulder
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands
| | - Rens van der Waal
- Core Facility Molecular Pathology & Biobanking, The Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands
| | - Joyce Sanders
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands
| | - Mathilde M Almekinders
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands
| | - Alba Llop-Guevara
- Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Helen R Davies
- Academic Department of Medical Genetics, School of Clinical Medicine, University of Cambridge, CB2 0QQ Cambridge, UK; Early Cancer Institute, University of Cambridge, CB2 0XZ Cambridge, UK
| | - Matthijs J van Haren
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2302 BH Leiden, the Netherlands
| | - Nathaniel I Martin
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2302 BH Leiden, the Netherlands
| | - Fariba Behbod
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Serena Nik-Zainal
- Academic Department of Medical Genetics, School of Clinical Medicine, University of Cambridge, CB2 0QQ Cambridge, UK; Early Cancer Institute, University of Cambridge, CB2 0XZ Cambridge, UK
| | - Violeta Serra
- Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Jacco van Rheenen
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands; Oncode Institute, Amsterdam, the Netherlands
| | - Esther H Lips
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands
| | - Lodewyk F A Wessels
- Oncode Institute, Amsterdam, the Netherlands; Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands
| | - Jelle Wesseling
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands; Division of Diagnostic Oncology, Netherlands Cancer Institute - Antonie van Leeuwenhoek Hospital, 1066 CX Amsterdam, the Netherlands; Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Colinda L G J Scheele
- Center for Cancer Biology, VIB, Department of Oncology, KU Leuven, 3000 Leuven, Belgium
| | - Jos Jonkers
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands; Oncode Institute, Amsterdam, the Netherlands.
| |
Collapse
|
7
|
Bernhardt SM, Mitchell E, Stamnes S, Hoffmann RJ, Calhoun A, Klug A, Russell TD, Pennock ND, Walker JM, Schedin P. Isogenic Mammary Models of Intraductal Carcinoma Reveal Progression to Invasiveness in the Absence of a Non-Obligatory In Situ Stage. Cancers (Basel) 2023; 15:2257. [PMID: 37190184 PMCID: PMC10136757 DOI: 10.3390/cancers15082257] [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: 03/13/2023] [Revised: 04/04/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
In breast cancer, progression to invasive ductal carcinoma (IDC) involves interactions between immune, myoepithelial, and tumor cells. Development of IDC can proceed through ductal carcinoma in situ (DCIS), a non-obligate, non-invasive stage, or IDC can develop without evidence of DCIS and these cases associate with poorer prognosis. Tractable, immune-competent mouse models are needed to help delineate distinct mechanisms of local tumor cell invasion and prognostic implications. To address these gaps, we delivered murine mammary carcinoma cell lines directly into the main mammary lactiferous duct of immune-competent mice. Using two strains of immune-competent mice (BALB/c, C57BL/6), one immune-compromised (severe combined immunodeficiency; SCID) C57BL/6 strain, and six different murine mammary cancer cell lines (D2.OR, D2A1, 4T1, EMT6, EO771, Py230), we found early loss of ductal myoepithelial cell differentiation markers p63, α-smooth muscle actin, and calponin, and rapid formation of IDC in the absence of DCIS. Rapid IDC formation also occurred in the absence of adaptive immunity. Combined, these studies demonstrate that loss of myoepithelial barrier function does not require an intact immune system, and suggest that these isogenic murine models may prove a useful tool to study IDC in the absence of a non-obligatory DCIS stage-an under-investigated subset of poor prognostic human breast cancer.
Collapse
Affiliation(s)
- Sarah M. Bernhardt
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Elizabeth Mitchell
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Stephanie Stamnes
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Reuben J. Hoffmann
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Andrea Calhoun
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Alex Klug
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Tanya D. Russell
- Center for Advancing Professional Excellence, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Nathan D. Pennock
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Radiation Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Joshua M. Walker
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Radiation Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Pepper Schedin
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Young Women’s Breast Cancer Translational Program, University of Colorado Cancer Center, Aurora, CO 80045, USA
| |
Collapse
|
8
|
Neagu AN, Whitham D, Seymour L, Haaker N, Pelkey I, Darie CC. Proteomics-Based Identification of Dysregulated Proteins and Biomarker Discovery in Invasive Ductal Carcinoma, the Most Common Breast Cancer Subtype. Proteomes 2023; 11:13. [PMID: 37092454 PMCID: PMC10123686 DOI: 10.3390/proteomes11020013] [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: 01/19/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 04/05/2023] Open
Abstract
Invasive ductal carcinoma (IDC) is the most common histological subtype of malignant breast cancer (BC), and accounts for 70-80% of all invasive BCs. IDC demonstrates great heterogeneity in clinical and histopathological characteristics, prognoses, treatment strategies, gene expressions, and proteomic profiles. Significant proteomic determinants of the progression from intraductal pre-invasive malignant lesions of the breast, which characterize a ductal carcinoma in situ (DCIS), to IDC, are still poorly identified, validated, and clinically applied. In the era of "6P" medicine, it remains a great challenge to determine which patients should be over-treated versus which need to be actively monitored without aggressive treatment. The major difficulties for designating DCIS to IDC progression may be solved by understanding the integrated genomic, transcriptomic, and proteomic bases of invasion. In this review, we showed that multiple proteomics-based techniques, such as LC-MS/MS, MALDI-ToF MS, SELDI-ToF-MS, MALDI-ToF/ToF MS, MALDI-MSI or MasSpec Pen, applied to in-tissue, off-tissue, BC cell lines and liquid biopsies, improve the diagnosis of IDC, as well as its prognosis and treatment monitoring. Classic proteomics strategies that allow the identification of dysregulated protein expressions, biological processes, and interrelated pathway analyses based on aberrant protein-protein interaction (PPI) networks have been improved to perform non-invasive/minimally invasive biomarker detection of early-stage IDC. Thus, in modern surgical oncology, highly sensitive, rapid, and accurate MS-based detection has been coupled with "proteome point sampling" methods that allow for proteomic profiling by in vivo "proteome point characterization", or by minimal tissue removal, for ex vivo accurate differentiation and delimitation of IDC. For the detection of low-molecular-weight proteins and protein fragments in bodily fluids, LC-MS/MS and MALDI-MS techniques may be coupled to enrich and capture methods which allow for the identification of early-stage IDC protein biomarkers that were previously invisible for MS-based techniques. Moreover, the detection and characterization of protein isoforms, including posttranslational modifications of proteins (PTMs), is also essential to emphasize specific molecular mechanisms, and to assure the early-stage detection of IDC of the breast.
Collapse
Affiliation(s)
- Anca-Narcisa Neagu
- Laboratory of Animal Histology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iasi, Carol I bvd. No. 20A, 700505 Iasi, Romania
| | - Danielle Whitham
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| | - Logan Seymour
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| | - Norman Haaker
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| | - Isabella Pelkey
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| | - Costel C. Darie
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| |
Collapse
|
9
|
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: 7] [Impact Index Per Article: 3.5] [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
|
10
|
Chibly AM, Patel VN, Aure MH, Pasquale MC, Martin GE, Ghannam M, Andrade J, Denegre NG, Simpson C, Goldstein DP, Liu FF, Lombaert IMA, Hoffman MP. Neurotrophin signaling is a central mechanism of salivary dysfunction after irradiation that disrupts myoepithelial cells. NPJ Regen Med 2023; 8:17. [PMID: 36966175 PMCID: PMC10039923 DOI: 10.1038/s41536-023-00290-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 02/27/2023] [Indexed: 03/27/2023] Open
Abstract
The mechanisms that prevent regeneration of irradiated (IR) salivary glands remain elusive. Bulk RNAseq of IR versus non-IR human salivary glands showed that neurotrophin signaling is highly disrupted post-radiation. Neurotrophin receptors (NTRs) were significantly upregulated in myoepithelial cells (MECs) post-IR, and single cell RNAseq revealed that MECs pericytes, and duct cells are the main sources of neurotrophin ligands. Using two ex vivo models, we show that nerve growth factor (NGF) induces expression of MEC genes during development, and upregulation of NTRs in adult MECs is associated with stress-induced plasticity and morphological abnormalities in IR human glands. As MECs are epithelial progenitors after gland damage and are required for proper acinar cell contraction and secretion, we propose that MEC-specific upregulation of NTRs post-IR disrupts MEC differentiation and potentially impedes the ability of the gland to regenerate.
Collapse
Affiliation(s)
- Alejandro M Chibly
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Vaishali N Patel
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Marit H Aure
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Mary C Pasquale
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Gemma E Martin
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Mousa Ghannam
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Julianne Andrade
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Noah G Denegre
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Colleen Simpson
- Department of Otolaryngology-Head & Neck Surgery, Princess Margaret Cancer Center, Toronto, ON, M5G 2C4, Canada
| | - David P Goldstein
- Department of Otolaryngology-Head & Neck Surgery, Princess Margaret Cancer Center, Toronto, ON, M5G 2C4, Canada
| | - Fei-Fei Liu
- Department of Radiation Oncology, Princess Margaret Cancer Center, Toronto, ON, M5G 2M9, Canada
| | - Isabelle M A Lombaert
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
- Department of Biologic and Material Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI, 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Matthew P Hoffman
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
11
|
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: 9] [Impact Index Per Article: 3.0] [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
|
12
|
Rebbeck CA, Xian J, Bornelöv S, Geradts J, Hobeika A, Geiger H, Alvarez JF, Rozhkova E, Nicholls A, Robine N, Lyerly HK, Hannon GJ. Gene expression signatures of individual ductal carcinoma in situ lesions identify processes and biomarkers associated with progression towards invasive ductal carcinoma. Nat Commun 2022; 13:3399. [PMID: 35697697 PMCID: PMC9192778 DOI: 10.1038/s41467-022-30573-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 05/06/2022] [Indexed: 12/27/2022] Open
Abstract
Ductal carcinoma in situ (DCIS) is considered a non-invasive precursor to breast cancer, and although associated with an increased risk of developing invasive disease, many women with DCIS will never progress beyond their in situ diagnosis. The path from normal duct to invasive ductal carcinoma (IDC) is not well understood, and efforts to do so are hampered by the substantial heterogeneity that exists between patients, and even within patients. Here we show gene expression analysis from > 2,000 individually micro-dissected ductal lesions representing 145 patients. Combining all samples into one continuous trajectory we show there is a progressive loss in basal layer integrity heading towards IDC, coupled with two epithelial to mesenchymal transitions, one early and a second coinciding with the convergence of DCIS and IDC expression profiles. We identify early processes and potential biomarkers, including CAMK2N1, MNX1, ADCY5, HOXC11 and ANKRD22, whose reduced expression is associated with the progression of DCIS to invasive breast cancer.
Collapse
Affiliation(s)
- Clare A Rebbeck
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.
| | - Jian Xian
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Susanne Bornelöv
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Joseph Geradts
- Department of Pathology & Laboratory Medicine, East Carolina University Brody School of Medicine, Greenville, NC, USA
| | - Amy Hobeika
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | | | - Jose Franco Alvarez
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Elena Rozhkova
- Department of Dermatology, Boston University School of Medicine, Boston, MA, USA
| | - Ashley Nicholls
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | | | - Herbert K Lyerly
- Department of Surgery, Duke University Medical Center, Durham, NC, USA.
| | - Gregory J Hannon
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|
13
|
Hophan SL, Odnokoz O, Liu H, Luo Y, Khan S, Gradishar W, Zhou Z, Badve S, Torres MA, Wan Y. Ductal Carcinoma In Situ of Breast: From Molecular Etiology to Therapeutic Management. Endocrinology 2022; 163:bqac027. [PMID: 35245349 PMCID: PMC8962444 DOI: 10.1210/endocr/bqac027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Indexed: 11/19/2022]
Abstract
Ductal carcinoma in situ (DCIS) makes up a majority of noninvasive breast cancer cases. DCIS is a neoplastic proliferation of epithelial cells within the ductal structure of the breast. Currently, there is little known about the progression of DCIS to invasive ductal carcinoma (IDC), or the molecular etiology behind each DCIS lesion or grade. The DCIS lesions can be heterogeneous in morphology, genetics, cellular biology, and clinical behavior, posing challenges to our understanding of the molecular mechanisms by which approximately half of all DCIS lesions progress to an invasive status. New strategies that pinpoint molecular mechanisms are necessary to overcome this gap in understanding, which is a barrier to more targeted therapy. In this review, we will discuss the etiological factors associated with DCIS, as well as the complexity of each nuclear grade lesion. Moreover, we will discuss the possible molecular features that lead to progression of DCIS to IDC. We will highlight current therapeutic management and areas for improvement.
Collapse
Affiliation(s)
- Shelby Lynn Hophan
- Department of Obstetrics and Gynecology, Department of Pharmacology, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Olena Odnokoz
- Department of Pharmacology and Chemical Biology, Winship Cancer Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Huiping Liu
- Department of Pharmacology, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yuan Luo
- Department of Preventive Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Seema Khan
- Department of Surgery, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - William Gradishar
- Department of Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Zhuan Zhou
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sunil Badve
- Department of Pathology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Mylin A Torres
- Department of Hematology and Oncology, Winship Cancer Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Yong Wan
- Department of Pharmacology and Chemical Biology, Winship Cancer Center, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Hematology and Oncology, Winship Cancer Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
14
|
Wilson GM, Dinh P, Pathmanathan N, Graham JD. Ductal Carcinoma in Situ: Molecular Changes Accompanying Disease Progression. J Mammary Gland Biol Neoplasia 2022; 27:101-131. [PMID: 35567670 PMCID: PMC9135892 DOI: 10.1007/s10911-022-09517-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/13/2022] [Indexed: 10/26/2022] Open
Abstract
Ductal carcinoma in situ (DCIS) is a non-obligate precursor of invasive ductal carcinoma (IDC), whereby if left untreated, approximately 12% of patients develop invasive disease. The current standard of care is surgical removal of the lesion, to prevent potential progression, and radiotherapy to reduce risk of recurrence. There is substantial overtreatment of DCIS patients, considering not all DCIS lesions progress to invasive disease. Hence, there is a critical imperative to better predict which DCIS lesions are destined for poor outcome and which are not, allowing for tailored treatment. Active surveillance is currently being trialed as an alternative management practice, but this approach relies on accurately identifying cases that are at low risk of progression to invasive disease. Two DCIS-specific genomic profiling assays that attempt to distinguish low and high-risk patients have emerged, but imperfections in risk stratification coupled with a high price tag warrant the continued search for more robust and accessible prognostic biomarkers. This search has largely turned researchers toward the tumor microenvironment. Recent evidence suggests that a spectrum of cell types within the DCIS microenvironment are genetically and phenotypically altered compared to normal tissue and play critical roles in disease progression. Uncovering the molecular mechanisms contributing to DCIS progression has provided optimism for the search for well-validated prognostic biomarkers that can accurately predict the risk for a patient developing IDC. The discovery of such markers would modernize DCIS management and allow tailored treatment plans. This review will summarize the current literature regarding DCIS diagnosis, treatment, and pathology.
Collapse
Affiliation(s)
- Gemma M Wilson
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia
| | - Phuong Dinh
- Westmead Breast Cancer Institute, Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Nirmala Pathmanathan
- Westmead Breast Cancer Institute, Westmead Hospital, Westmead, NSW, 2145, Australia
| | - J Dinny Graham
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia.
- Westmead Breast Cancer Institute, Westmead Hospital, Westmead, NSW, 2145, Australia.
| |
Collapse
|
15
|
The Utility of Myoepithelial Cell Layer Identification in Adnexal Carcinomas. Am J Dermatopathol 2022; 44:155-162. [PMID: 35171883 DOI: 10.1097/dad.0000000000001844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
ABSTRACT The distinction of metastatic carcinomas to the skin (MCS) from cutaneous adnexal carcinomas can pose a significant diagnostic challenge. The differentiation between (MCS) from a primary cutaneous adnexal tumor is one of the most difficult tasks in the field of dermatopathology, and immunohistochemistry has only been partially helpful in solving this problem. In routine diagnostic surgical pathology, it is essential to identify the myoepithelial cell layer by immunohistochemistry to distinguish between an in situ and invasive breast carcinomas and when establishing the presence of microinvasion. The purpose of this study was to evaluate the role of myoepithelial cell layer expression in difficult cases of cutaneous adnexal carcinomas in which histologically it was challenging to separate them from MCS. We studied 38 adnexal carcinomas and evaluated them for myoepithelial markers to confirm the primary nature of the neoplasm. The used markers to search for myoepithelial cell layer retention included calponin, p63, and smooth muscle actin. Of the 38 cases, we found that 13 cases showed myoepithelial layer retention, confirming the primary cutaneous origin of the neoplastic process. The results of our study suggest that the presence of an identifiable retention of the myoepithelial cell layer in adnexal carcinomas could be a useful adjunct observation in the diagnosis of primary adnexal carcinomas, especially in the clinical setting of a questionable primary adnexal versus metastatic neoplasm.
Collapse
|
16
|
Sofía M, Sebastián R, Emanuel C, Branham MT, Marzese DM, Matthew S, De Blas G, Rodolfo A, Michael L, María R. When left does not seem right: epigenetic and bioelectric differences between left- and right-sided breast cancer. Mol Med 2022; 28:15. [PMID: 35123413 PMCID: PMC8817536 DOI: 10.1186/s10020-022-00440-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 01/18/2022] [Indexed: 01/22/2023] Open
Abstract
Background During embryogenesis lateral symmetry is broken, giving rise to Left/Right (L/R) breast tissues with distinct identity. L/R-sided breast tumors exhibit consistently-biased incidence, gene expression, and DNA methylation. We postulate that a differential L/R tumor-microenvironment crosstalk generates different tumorigenesis mechanisms. Methods We performed in-silico analyses on breast tumors of public datasets, developed xenografted tumors, and conditioned MDA-MB-231 cells with L/R mammary extracts. Results We found L/R differential DNA methylation involved in embryogenic and neuron-like functions. Focusing on ion-channels, we discovered significant L/R epigenetic and bioelectric differences. Specifically, L-sided cells presented increased methylation of hyperpolarizing ion channel genes and increased Ca2+ concentration and depolarized membrane potential, compared to R-ones. Functional consequences were associated with increased proliferation in left tumors, assessed by KI67 expression and mitotic count. Conclusions Our findings reveal considerable L/R asymmetry in cancer processes, and suggest specific L/R epigenetic and bioelectric differences as future targets for cancer therapeutic approaches in the breast and many other paired organs. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-022-00440-5.
Collapse
|
17
|
Hong Y, Limback D, Elsarraj HS, Harper H, Haines H, Hansford H, Ricci M, Kaufman C, Wedlock E, Xu M, Zhang J, May L, Cusick T, Inciardi M, Redick M, Gatewood J, Winblad O, Aripoli A, Huppe A, Balanoff C, Wagner JL, Amin AL, Larson KE, Ricci L, Tawfik O, Razek H, Meierotto RO, Madan R, Godwin AK, Thompson J, Hilsenbeck SG, Futreal A, Thompson A, Hwang ES, Fan F, Behbod F. Mouse-INtraDuctal (MIND): an in vivo model for studying the underlying mechanisms of DCIS malignancy. J Pathol 2022; 256:186-201. [PMID: 34714554 PMCID: PMC8738143 DOI: 10.1002/path.5820] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/05/2021] [Accepted: 10/25/2021] [Indexed: 11/24/2022]
Abstract
Due to widespread adoption of screening mammography, there has been a significant increase in new diagnoses of ductal carcinoma in situ (DCIS). However, DCIS prognosis remains unclear. To address this gap, we developed an in vivo model, Mouse-INtraDuctal (MIND), in which patient-derived DCIS epithelial cells are injected intraductally and allowed to progress naturally in mice. Similar to human DCIS, the cancer cells formed in situ lesions inside the mouse mammary ducts and mimicked all histologic subtypes including micropapillary, papillary, cribriform, solid, and comedo. Among 37 patient samples injected into 202 xenografts, at median duration of 9 months, 20 samples (54%) injected into 95 xenografts showed in vivo invasive progression, while 17 (46%) samples injected into 107 xenografts remained non-invasive. Among the 20 samples that showed invasive progression, nine samples injected into 54 xenografts exhibited a mixed pattern in which some xenografts showed invasive progression while others remained non-invasive. Among the clinically relevant biomarkers, only elevated progesterone receptor expression in patient DCIS and the extent of in vivo growth in xenografts predicted an invasive outcome. The Tempus XT assay was used on 16 patient DCIS formalin-fixed, paraffin-embedded sections including eight DCISs that showed invasive progression, five DCISs that remained non-invasive, and three DCISs that showed a mixed pattern in the xenografts. Analysis of the frequency of cancer-related pathogenic mutations among the groups showed no significant differences (KW: p > 0.05). There were also no differences in the frequency of high, moderate, or low severity mutations (KW; p > 0.05). These results suggest that genetic changes in the DCIS are not the primary driver for the development of invasive disease. © 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
MESH Headings
- Animals
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma, Intraductal, Noninfiltrating/genetics
- Carcinoma, Intraductal, Noninfiltrating/metabolism
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Cell Movement
- Cell Proliferation
- Disease Progression
- Epithelial Cells/metabolism
- Epithelial Cells/pathology
- Epithelial Cells/transplantation
- Female
- Heterografts
- Humans
- Mice, Inbred NOD
- Mice, SCID
- Mutation
- Neoplasm Invasiveness
- Neoplasm Transplantation
- Receptors, Progesterone/metabolism
- Time Factors
- Mice
Collapse
Affiliation(s)
- Yan Hong
- Department of Pathology and Laboratory MedicineThe University of Kansas Medical CenterKansas CityKSUSA
| | - Darlene Limback
- Department of Pathology and Laboratory MedicineThe University of Kansas Medical CenterKansas CityKSUSA
| | - Hanan S Elsarraj
- Department of Pathology and Laboratory MedicineThe University of Kansas Medical CenterKansas CityKSUSA
| | - Haleigh Harper
- University of Kansas School of MedicineThe University of Kansas Medical CenterKansas CityKSUSA
| | - Haley Haines
- Department of Pathology and Laboratory MedicineThe University of Kansas Medical CenterKansas CityKSUSA
| | - Hayley Hansford
- Department of Pathology and Laboratory MedicineThe University of Kansas Medical CenterKansas CityKSUSA
| | - Michael Ricci
- Department of Pathology and Laboratory MedicineThe University of Kansas Medical CenterKansas CityKSUSA
| | - Carolyn Kaufman
- University of Kansas School of MedicineThe University of Kansas Medical CenterKansas CityKSUSA
| | - Emily Wedlock
- Department of Pathology and Laboratory MedicineThe University of Kansas Medical CenterKansas CityKSUSA
| | - Mingchu Xu
- Department of Genomic MedicineThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Jianhua Zhang
- Department of Genomic MedicineThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Lisa May
- Department of RadiologyThe University of Kansas School of Medicine‐WichitaWichitaKSUSA
| | - Therese Cusick
- Department of SurgeryThe University of Kansas School of Medicine‐WichitaWichitaKSUSA
| | - Marc Inciardi
- Department of RadiologyThe University of Kansas Medical CenterKansas CityKSUSA
| | - Mark Redick
- Department of RadiologyThe University of Kansas Medical CenterKansas CityKSUSA
| | - Jason Gatewood
- Department of RadiologyThe University of Kansas Medical CenterKansas CityKSUSA
| | - Onalisa Winblad
- Department of RadiologyThe University of Kansas Medical CenterKansas CityKSUSA
| | - Allison Aripoli
- Department of RadiologyThe University of Kansas Medical CenterKansas CityKSUSA
| | - Ashley Huppe
- Department of RadiologyThe University of Kansas Medical CenterKansas CityKSUSA
| | - Christa Balanoff
- Department of General Surgery, Breast Surgical Oncology DivisionThe University of Kansas Medical CenterKansas CityKSUSA
| | - Jamie L Wagner
- Department of General Surgery, Breast Surgical Oncology DivisionThe University of Kansas Medical CenterKansas CityKSUSA
| | - Amanda L Amin
- Department of General Surgery, Breast Surgical Oncology DivisionThe University of Kansas Medical CenterKansas CityKSUSA
| | - Kelsey E Larson
- Department of General Surgery, Breast Surgical Oncology DivisionThe University of Kansas Medical CenterKansas CityKSUSA
| | - Lawrence Ricci
- Department of RadiologyTruman Medical CenterKansas CityMOUSA
| | - Ossama Tawfik
- Department of Pathology, St Luke's Health System of Kansas CityMAWD Pathology GroupKansas CityMOUSA
| | | | - Ruby O Meierotto
- Breast RadiologySaint Luke's Cancer Institute, Saint Luke's Health SystemKansas CityMOUSA
| | - Rashna Madan
- Department of Pathology and Laboratory MedicineThe University of Kansas Medical CenterKansas CityKSUSA
| | - Andrew K Godwin
- Department of Pathology and Laboratory MedicineThe University of Kansas Medical CenterKansas CityKSUSA
| | - Jeffrey Thompson
- Department of BiostatisticsThe University of Kansas Medical CenterKansas CityKSUSA
| | - Susan G Hilsenbeck
- Lester and Sue Smith Breast Center, Biostatistics and Informatics Shared Resources, Duncan Cancer CenterBaylor College of MedicineHoustonTXUSA
| | - Andy Futreal
- Department of Genomic Medicine, Division of Cancer MedicineThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Alastair Thompson
- Section of Breast SurgeryBaylor College of Medicine, Lester and Sue Smith Breast Center, Dan L Duncan Comprehensive Cancer CenterHoustonTXUSA
| | | | - Fang Fan
- Department of PathologyCity of Hope Medical CenterDuarteCAUSA
| | - Fariba Behbod
- Department of Pathology and Laboratory MedicineThe University of Kansas Medical CenterKansas CityKSUSA
| | | |
Collapse
|
18
|
Tumour-infiltrating lymphocytes add prognostic information for patients with low-risk DCIS: findings from the SweDCIS randomised radiotherapy trial. Eur J Cancer 2022; 168:128-137. [DOI: 10.1016/j.ejca.2022.01.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/25/2021] [Accepted: 01/09/2022] [Indexed: 12/21/2022]
|
19
|
Lee HJ, Myung JK, Kim HS, Lee DH, Go HS, Choi JH, Koh HM, Lee SJ, Jang B. Expression of LGR5 in mammary myoepithelial cells and in triple-negative breast cancers. Sci Rep 2021; 11:17750. [PMID: 34493772 PMCID: PMC8423726 DOI: 10.1038/s41598-021-97351-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/24/2021] [Indexed: 11/25/2022] Open
Abstract
Lineage tracing in mice indicates that LGR5 is an adult stem cell marker in multiple organs, such as the intestine, stomach, hair follicles, ovary, and mammary glands. Despite many studies exploring the presence of LGR5 cells in human tissues, little is known about its expression profile in either human mammary tissue or pathological lesions. In this study we aim to investigate LGR5 expression in normal, benign, and malignant lesions of the human breast using RNA in situ hybridization. LGR5 expression has not been observed in normal lactiferous ducts and terminal duct lobular units, whereas LGR5-positive cells have been specifically observed in the basal myoepithelium of ducts in the regenerative tissues, ductal carcinoma in situ, and in ducts surrounded by invasive cancer cells. These findings suggest LGR5 marks facultative stem cells that are involved in post injury regeneration instead of homeostatic stem cells. LGR5 positivity was found in 3% (9 of 278 cases) of invasive breast cancers (BC), and it showed positive associations with higher histologic grades (P = 0.001) and T stages (P < 0.001), while having negative correlations with estrogen receptor (P < 0.001) and progesterone receptor (P < 0.001) expression. Remarkably, all LGR5-positive BC, except one, belong to triple-negative BC (TNBC), representing 24% (9 of 38 cases) of all of them. LGR5 histoscores have no correlations with EGFR, CK5/6, Ki-67, or P53 expression. Additionally, no β-catenin nuclear localization was observed in LGR5-positive BC, indicating that canonical Wnt pathway activation is less likely involved in LGR5 expression in BC. Our results demonstrate that LGR5 expression is induced in regenerative conditions in the myoepithelium of human mammary ducts and that its expression is only observed in TNBC subtype among all invasive BC. Further studies regarding the functional and prognostic impact of LGR5 in TNBC are warranted.
Collapse
Affiliation(s)
- Hyun Ju Lee
- Department of Pathology, Soonchunhyang University College of Medicine and Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Jae Kyung Myung
- Department of Pathology, Hanyang University College of Medicine, Seoul, Korea
| | - Hye Sung Kim
- Department of Pathology, Jeju National University School of Medicine, Jeju, South Korea
| | - Dong Hui Lee
- Department of Pathology, Jeju National University School of Medicine and Jeju National University Hospital, Aran 13 gil 15, Jeju city, Jeju, 63241, Korea
| | - Hyun Su Go
- Department of Pathology, Jeju National University School of Medicine, Jeju, South Korea
| | - Jae Hyuck Choi
- Department of Surgery, Jeju National University School of Medicine and Jeju National University Hospital, Jeju, South Korea
| | - Hyun Min Koh
- Department of Pathology, Gyeongsang National University Changwon Hospital, Changwon, South Korea
| | - Su-Jae Lee
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, South Korea
| | - Bogun Jang
- Department of Pathology, Jeju National University School of Medicine and Jeju National University Hospital, Aran 13 gil 15, Jeju city, Jeju, 63241, Korea.
| |
Collapse
|
20
|
Juppet Q, De Martino F, Marcandalli E, Weigert M, Burri O, Unser M, Brisken C, Sage D. Deep Learning Enables Individual Xenograft Cell Classification in Histological Images by Analysis of Contextual Features. J Mammary Gland Biol Neoplasia 2021; 26:101-112. [PMID: 33999331 PMCID: PMC8236058 DOI: 10.1007/s10911-021-09485-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 04/05/2021] [Indexed: 02/06/2023] Open
Abstract
Patient-Derived Xenografts (PDXs) are the preclinical models which best recapitulate inter- and intra-patient complexity of human breast malignancies, and are also emerging as useful tools to study the normal breast epithelium. However, data analysis generated with such models is often confounded by the presence of host cells and can give rise to data misinterpretation. For instance, it is important to discriminate between xenografted and host cells in histological sections prior to performing immunostainings. We developed Single Cell Classifier (SCC), a data-driven deep learning-based computational tool that provides an innovative approach for automated cell species discrimination based on a multi-step process entailing nuclei segmentation and single cell classification. We show that human and murine cell contextual features, more than cell-intrinsic ones, can be exploited to discriminate between cell species in both normal and malignant tissues, yielding up to 96% classification accuracy. SCC will facilitate the interpretation of H&E- and DAPI-stained histological sections of xenografted human-in-mouse tissues and it is open to new in-house built models for further applications. SCC is released as an open-source plugin in ImageJ/Fiji available at the following link: https://github.com/Biomedical-Imaging-Group/SingleCellClassifier .
Collapse
Affiliation(s)
- Quentin Juppet
- Biomedical Imaging Group, School of Engineering, Ecole Polytechnique Fédéralé de Lausanne (EPFL), Lausanne, Switzerland
- EPFL Center for Imaging, Ecole Polytechnique Fédéralé de Lausanne (EPFL), Lausanne, Switzerland
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédéralé de Lausanne (EPFL), Lausanne, Switzerland
| | - Fabio De Martino
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédéralé de Lausanne (EPFL), Lausanne, Switzerland
| | - Elodie Marcandalli
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédéralé de Lausanne (EPFL), Lausanne, Switzerland
| | - Martin Weigert
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédéralé de Lausanne (EPFL), Lausanne, Switzerland
| | - Olivier Burri
- BioImaging & Optics Platform, Ecole Polytechnique Fédéralé de Lausanne (EPFL), Lausanne, Switzerland
| | - Michael Unser
- Biomedical Imaging Group, School of Engineering, Ecole Polytechnique Fédéralé de Lausanne (EPFL), Lausanne, Switzerland
| | - Cathrin Brisken
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédéralé de Lausanne (EPFL), Lausanne, Switzerland.
| | - Daniel Sage
- Biomedical Imaging Group, School of Engineering, Ecole Polytechnique Fédéralé de Lausanne (EPFL), Lausanne, Switzerland.
- EPFL Center for Imaging, Ecole Polytechnique Fédéralé de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
21
|
D’Abbronzo G, Franco R. The changing role of the pathologist in the era of targeted therapy in personalized medicine. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2021. [DOI: 10.1080/23808993.2021.1923400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Giuseppe D’Abbronzo
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Renato Franco
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| |
Collapse
|
22
|
Deckwirth V, Rajakylä EK, Cattavarayane S, Acheva A, Schaible N, Krishnan R, Valle-Delgado JJ, Österberg M, Björkenheim P, Sukura A, Tojkander S. Cytokeratin 5 determines maturation of the mammary myoepithelium. iScience 2021; 24:102413. [PMID: 34007958 PMCID: PMC8111680 DOI: 10.1016/j.isci.2021.102413] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 12/06/2020] [Accepted: 04/06/2021] [Indexed: 12/29/2022] Open
Abstract
At invasion, transformed mammary epithelial cells expand into the stroma through a disrupted myoepithelial (ME) cell layer and basement membrane (BM). The intact ME cell layer has thus been suggested to act as a barrier against invasion. Here, we investigate the mechanisms behind the disruption of ME cell layer. We show that the expression of basal/ME proteins CK5, CK14, and α-SMA altered along increasing grade of malignancy, and their loss affected the maintenance of organotypic 3D mammary architecture. Furthermore, our data suggests that loss of CK5 prior to invasive stage causes decreased levels of Zinc finger protein SNAI2 (SLUG), a key regulator of the mammary epithelial cell lineage determination. Consequently, a differentiation bias toward luminal epithelial cell type was detected with loss of mature, α-SMA-expressing ME cells and reduced deposition of basement membrane protein laminin-5. Therefore, our data discloses the central role of CK5 in mammary epithelial differentiation and maintenance of normal ME layer.
Collapse
Affiliation(s)
- Vivi Deckwirth
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Agnes Sjöberginkatu 2, Helsinki 00014, Finland
| | - Eeva Kaisa Rajakylä
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Agnes Sjöberginkatu 2, Helsinki 00014, Finland
| | - Sandhanakrishnan Cattavarayane
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Agnes Sjöberginkatu 2, Helsinki 00014, Finland
| | - Anna Acheva
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Agnes Sjöberginkatu 2, Helsinki 00014, Finland
| | - Niccole Schaible
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ramaswamy Krishnan
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Juan José Valle-Delgado
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, Espoo 00076, Finland
| | - Monika Österberg
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, Espoo 00076, Finland
| | - Pia Björkenheim
- Veterinary Teaching Hospital, University of Helsinki, Helsinki 00014, Finland
| | - Antti Sukura
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Agnes Sjöberginkatu 2, Helsinki 00014, Finland
| | - Sari Tojkander
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Agnes Sjöberginkatu 2, Helsinki 00014, Finland
| |
Collapse
|
23
|
Rodrigues MA, Caldeira-Brant AL, Gomes DA, Silveira TL, Chiarini-Garcia H, Cassali GD. Characterization of neoplastic cells outlining the cystic space of invasive micropapillary carcinoma of the canine mammary gland. BMC Vet Res 2021; 17:130. [PMID: 33761962 PMCID: PMC7992814 DOI: 10.1186/s12917-021-02807-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 02/19/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Invasive micropapillary carcinoma (IMPC) is a rare malignant breast tumor and a variant form of invasive ductal carcinoma that is an aggressive neoplasm of the human breast and canine mammary gland. The importance of the tumor microenvironment in cancer development has gradually been recognized, but little is known about the cell types outlining the cystic space of canine IMPC. This study aimed to characterize the neoplastic cells outlining the cystic space of IMPC. RESULTS Immunohistochemistry (IHC), immunofluorescence (IF), superresolution and transmission electron microscopy (TEM) were used to assess the cell types in the cystic areas of IMPCs. Cells expressing the mesenchymal markers alpha-smooth muscle actin (αSMA), Vimentin, and S100A4 outlined the cystic space of IMPC. Furthermore, loss of epithelial cell polarity in IMPC was shown by the localization of MUC1 at the stroma-facing surface. This protein modulates lumen formation and inhibits the cell-stroma interaction. Immunohistochemical and IF staining for the myoepithelial cell marker p63 were negative in IMPC samples. Furthermore, associated with peculiar morphology, such as thin cytoplasmic extensions outlining cystic spaces, was observed under TEM. These observations suggested cells with characteristics of myoepithelial-like cells. CONCLUSIONS The cells outlining the cystic space of IMPC in the canine mammary gland were characterized using IHC, IF and TEM. The presence of cells expressing αSMA, Vimentin, and S100A4 in the IMPC stroma suggested a role for tumor-associated fibroblasts in the IMPC microenvironment. The reversal of cell polarity revealed by the limited basal localization of MUC1 may be an important factor contributing to the invasiveness of IMPC. For the first time, the cystic space of canine mammary gland IMPC was shown to be delimited by myoepithelial-like cells that had lost p63 expression. These findings may enhance our understanding of the cellular microenvironment of invasive tumors to improve cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Michele A Rodrigues
- Department of General Pathology, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, Minas Gerais, CEP: 31270-901, Brazil
| | - Andre L Caldeira-Brant
- Department of Morphology, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, Minas Gerais, CEP: 31270-901, Brazil
| | - Dawidson A Gomes
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, Minas Gerais, CEP: 31270-901, Brazil
| | - Tatiany L Silveira
- Department of General Pathology, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, Minas Gerais, CEP: 31270-901, Brazil
| | - Hélio Chiarini-Garcia
- Department of Morphology, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, Minas Gerais, CEP: 31270-901, Brazil
| | - Geovanni D Cassali
- Department of General Pathology, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, Minas Gerais, CEP: 31270-901, Brazil.
| |
Collapse
|
24
|
Parigoris E, Lee S, Mertz D, Turner M, Liu AY, Sentosa J, Djomehri S, Chang HC, Luker K, Luker G, Kleer CG, Takayama S. Cancer Cell Invasion of Mammary Organoids with Basal-In Phenotype. Adv Healthc Mater 2021; 10:e2000810. [PMID: 32583612 PMCID: PMC7759600 DOI: 10.1002/adhm.202000810] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Indexed: 01/08/2023]
Abstract
This paper describes mammary organoids with a basal-in phenotype where the basement membrane is located on the interior surface of the organoid. A key materials consideration to induce this basal-in phenotype is the use of a minimal gel scaffold that the epithelial cells self-assemble around and encapsulate. When MDA-MB-231 breast cancer cells are co-cultured with epithelial cells from day 0 under these conditions, cells self-organize into patterns with distinct cancer cell populations both inside and at the periphery of the epithelial organoid. In another type of experiment, the robust formation of the basement membrane on the epithelial organoid interior enables convenient studies of MDA-MB-231 invasion in a tumor progression-relevant direction relative to epithelial cell-basement membrane positioning. That is, the study of cancer invasion through the epithelium first, followed by the basement membrane to the basal side, is realized in an experimentally convenient manner where the cancer cells are simply seeded on the outside of preformed organoids, and their invasion into the organoid is monitored. Interestingly, invasion is more prominent when tumor cells are added to day 7 organoids with less developed basement membranes compared to day 16 organoids with more defined ones.
Collapse
Affiliation(s)
- Eric Parigoris
- Wallace H. Coulter Department of Biomedical Engineering and The Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, 30332, USA
| | - Soojung Lee
- Wallace H. Coulter Department of Biomedical Engineering and The Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, 30332, USA
| | - David Mertz
- Wallace H. Coulter Department of Biomedical Engineering and The Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, 30332, USA
| | - Madeleine Turner
- Wallace H. Coulter Department of Biomedical Engineering and The Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, 30332, USA
| | - Amy Y Liu
- Wallace H. Coulter Department of Biomedical Engineering and The Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, 30332, USA
| | - Jason Sentosa
- Wallace H. Coulter Department of Biomedical Engineering and The Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, 30332, USA
| | - Sabra Djomehri
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Hao Chen Chang
- Wallace H. Coulter Department of Biomedical Engineering and The Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, 30332, USA
| | - Kathryn Luker
- Departments of Radiology and Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI, 48109, USA
| | - Gary Luker
- Departments of Radiology and Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI, 48109, USA
| | - Celina G Kleer
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Shuichi Takayama
- Wallace H. Coulter Department of Biomedical Engineering and The Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, 30332, USA
| |
Collapse
|
25
|
Guo R, Tian Y, Zhu M, Huang Y, Qiang L, Jin X, Yang J. [Expression of thymidylate synthase in salivary adenoid myoepithelial cells and its clinical significance]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:469-474. [PMID: 32895123 DOI: 10.12122/j.issn.1673-4254.2020.04.04] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To evaluate the expression of thymidylate synthase (TS) in myoepithelial cells (MECs) of salivary adenoid tissues and explore its clinical significance. METHODS Immunohistochemical staining EnVision method was used to detect the expression of TS, P63, Calponin, CK5/6 and S-100 in 32 salivary gland specimens, including 10 non-neoplastic and salivary inflammation specimens, 11 mixed tumor specimens, 5 basal cell carcinoma specimens and 6 adenoid cyst carcinoma specimens. The specificity and sensitivity of TS as a specific molecular marker of salivary muscle epithelial cells were evaluated in comparison with P63, Calponin, CK5/6 and S-100. RESULTS The expression pattern of TS in all the salivary gland tissue specimens was identical with that of p63. TS and P63 both showed strong immunohistochemical expressions in MECs of salivary adenoid tissue specimens. Calponin, CK5/6, and S-100 showed cytoplasmic/membranous expressions in the MECs. In addition, TS exhibited weak or moderate cytoplasmic expression in a few salivary gland epithelial cells, cancer cells and scattered stromal cells, with negative expression in the cell nuclei. The expression of TS in the MECs of all the salivary adenoid specimens was highly consistent with those of P63, Calponin, CK5/6 and S-100 (P>0.05) Except for CK5/6 expression in Salivary inflammation and Salivary gland specimens. Kappa>0.75. The specificity and sensitivity of TS as a molecular marker of MECs were both 100%. CONCLUSIONS TS is a new specific marker of MECs for differential diagnosis of salivary gland tumors.
Collapse
Affiliation(s)
- Rui Guo
- Deaprtment of Pathology, Second Hospital of Xi'an Jiaotong University College of Medicine, Xi'an 710004, China
| | - Yi Tian
- Deaprtment of Pathology, Second Hospital of Xi'an Jiaotong University College of Medicine, Xi'an 710004, China
| | - Mingming Zhu
- Department of Dermatology, Third Affiliated Hospital of Xinxiang Medical College, Xinxiang 453003, China
| | - Ying Huang
- Deaprtment of Pathology, Second Hospital of Xi'an Jiaotong University College of Medicine, Xi'an 710004, China
| | - Lei Qiang
- Deaprtment of Pathology, Second Hospital of Xi'an Jiaotong University College of Medicine, Xi'an 710004, China
| | - Xueyuan Jin
- Deaprtment of Pathology, Second Hospital of Xi'an Jiaotong University College of Medicine, Xi'an 710004, China
| | - Jun Yang
- Deaprtment of Pathology, Second Hospital of Xi'an Jiaotong University College of Medicine, Xi'an 710004, China
| |
Collapse
|
26
|
Mitchell E, Jindal S, Chan T, Narasimhan J, Sivagnanam S, Gray E, Chang YH, Weinmann S, Schedin P. Loss of myoepithelial calponin-1 characterizes high-risk ductal carcinoma in situ cases, which are further stratified by T cell composition. Mol Carcinog 2020; 59:701-712. [PMID: 32134153 PMCID: PMC7317523 DOI: 10.1002/mc.23171] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/14/2020] [Accepted: 02/15/2020] [Indexed: 12/15/2022]
Abstract
A hallmark of ductal carcinoma in situ (DCIS) progression is a loss of the surrounding ductal myoepithelium. However, whether compromise in myoepithelial differentiation, rather than overt cellular loss, can be used to predict the risk of DCIS progression is unknown. Here we address this question utilizing pure and mixed DCIS cases (N = 30) as surrogates for DCIS at low and high risk for progression, respectively. We used multiplex immunohistochemical staining to evaluate the relationship between myoepithelial cell differentiation and lymphoid immune cell types associated with poor prognostic DCIS. Our results show that myoepithelial calponin-1 discriminates between pure and mixed DCIS lesions better than histological subtype, presence of necrosis, or nuclear grade. Additionally, focal loss of myoepithelial cells associated with increased PD-1+CD8+ T cells, which suggests a link between the myoepithelium and immune surveillance. To identify associations between calponin-1 expression and immune response, we performed unsupervised hierarchical clustering of myoepithelial and immune cell biomarkers on 219 DCIS lesions from 30 cases. Notably, the majority of pure (low-risk) DCIS lesions clustered in a high calponin-1, T cell low group, whereas the majority of mixed (high-risk) DCIS lesions clustered in a low calponin-1, T cell high group, specifically with CD8+ and PD-1+CD8+ T cells. However, a subset of pure DCIS lesions had a similar calponin-1 and immune signature as the majority of mixed DCIS lesions, which have low calponin-1 and T cell enrichment-raising the possibility that these pure DCIS lesions might be at a high risk for progression.
Collapse
Affiliation(s)
- Elizabeth Mitchell
- Department of Cell, Developmental, and Cancer BiologyOregon Health and Science UniversityPortlandOregon
| | - Sonali Jindal
- Department of Cell, Developmental, and Cancer BiologyOregon Health and Science UniversityPortlandOregon
- Cancer Prevention and Control, Knight Cancer InstituteOregon Health and Science UniversityPortlandOregon
| | - Tiffany Chan
- Department of Cell, Developmental, and Cancer BiologyOregon Health and Science UniversityPortlandOregon
| | - Jayasri Narasimhan
- Department of Cell, Developmental, and Cancer BiologyOregon Health and Science UniversityPortlandOregon
| | - Shamilene Sivagnanam
- Computational Biology Program, Department of Cell, Developmental, and Cancer BiologyOregon Health and Science UniversityPortlandOregon
| | - Elliot Gray
- Department of Biomedical Engineering, Oregon Center for Spatial Systems BiomedicineOregon Health and Science UniversityPortlandOregon
| | - Young Hwan Chang
- Department of Biomedical Engineering, Oregon Center for Spatial Systems BiomedicineOregon Health and Science UniversityPortlandOregon
| | - Sheila Weinmann
- Center for Health ResearchKaiser Permanente NorthwestPortlandOregon
| | - Pepper Schedin
- Department of Cell, Developmental, and Cancer BiologyOregon Health and Science UniversityPortlandOregon
- Cancer Prevention and Control, Knight Cancer InstituteOregon Health and Science UniversityPortlandOregon
| |
Collapse
|
27
|
Suh J, Kim DH, Lee YH, Jang JH, Surh YJ. Fibroblast growth factor-2, derived from cancer-associated fibroblasts, stimulates growth and progression of human breast cancer cells via FGFR1 signaling. Mol Carcinog 2020; 59:1028-1040. [PMID: 32557854 DOI: 10.1002/mc.23233] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/22/2020] [Accepted: 05/27/2020] [Indexed: 12/29/2022]
Abstract
Cancer-associated fibroblasts (CAFs) constitute a major compartment of the tumor microenvironment. In the present study, we investigated the role for CAFs in breast cancer progression and underlying molecular mechanisms. Human breast cancer MDA-MB-231 cells treated with the CAF-conditioned media manifested a more proliferative phenotype, as evidenced by enhanced messenger RNA (mRNA) expression of Cyclin D1, c-Myc, and proliferating cell nuclear antigen. Analysis of data from The Cancer Genome Atlas revealed that fibroblast growth factor-2 (FGF2) expression was well correlated with the presence of CAFs. We noticed that the mRNA level of FGF2 in CAFs was higher than that in normal fibroblasts. FGF2 exerts its biological effects through interaction with FGF receptor 1 (FGFR1). In the breast cancer tissue array, 42% estrogen receptor-negative patients coexpressed FGF2 and FGFR1, whereas only 19% estrogen receptor-positive patients exhibited coexpression. CAF-stimulated MDA-MB-231 cell migration and invasiveness were abolished when FGF2-neutralizing antibody was added to the conditioned media of CAFs. In a xenograft mouse model, coinjection of MDA-MB-231 cells with activated fibroblasts expressing FGF2 dramatically enhanced tumor growth, and this was abrogated by silencing of FGFR1 in cancer cells. In addition, treatment of MDA-MB-231 cells with FGF2 enhanced expression of Cyclin D1, a key molecule involved in cell cycle progression. FGF2-induced cell migration and upregulation of Cyclin D1 were abolished by siRNA-mediated FGFR1 silencing. Taken together, the above findings suggest that CAFs promote growth, migration and invasion of MDA-MB-231 cells via the paracrine FGF2-FGFR1 loop in the breast tumor microenvironment.
Collapse
Affiliation(s)
- Jinyoung Suh
- Tumor Microenvironment Global Core Research Center and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
| | - Do-Hee Kim
- Department of Chemistry, College of Convergence and Integrated Science, Kyonggi University, Suwon, Gyeonggi-do, South Korea
| | - Yeon-Hwa Lee
- Tumor Microenvironment Global Core Research Center and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
| | - Jeong-Hoon Jang
- Tumor Microenvironment Global Core Research Center and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
| | - Young-Joon Surh
- Tumor Microenvironment Global Core Research Center and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea.,Cancer Research Institute, Seoul National University, Seoul, South Korea
| |
Collapse
|
28
|
Shan NL, Minden A, Furmanski P, Bak MJ, Cai L, Wernyj R, Sargsyan D, Cheng D, Wu R, Kuo HCD, Li SN, Fang M, Maehr H, Kong AN, Suh N. Analysis of the Transcriptome: Regulation of Cancer Stemness in Breast Ductal Carcinoma In Situ by Vitamin D Compounds. Cancer Prev Res (Phila) 2020; 13:673-686. [PMID: 32467291 DOI: 10.1158/1940-6207.capr-19-0566] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 04/01/2020] [Accepted: 05/20/2020] [Indexed: 12/21/2022]
Abstract
Ductal carcinoma in situ (DCIS), which accounts for one out of every five new breast cancer diagnoses, will progress to potentially lethal invasive ductal carcinoma (IDC) in about 50% of cases. Vitamin D compounds have been shown to inhibit progression to IDC in the MCF10DCIS model. This inhibition appears to involve a reduction in the cancer stem cell-like population in MCF10DCIS tumors. To identify genes that are involved in the vitamin D effects, a global transcriptomic analysis was undertaken of MCF10DCIS cells grown in mammosphere cultures, in which cancer stem-like cells grow preferentially and produce colonies by self-renewal and maturation, in the presence and absence of 1α25(OH)2D3 and a vitamin D analog, BXL0124. Using next-generation RNA-sequencing, we found that vitamin D compounds downregulated genes involved in maintenance of breast cancer stem-like cells (e.g., GDF15), epithelial-mesenchymal transition, invasion, and metastasis (e.g., LCN2 and S100A4), and chemoresistance (e.g., NGFR, PPP1R1B, and AGR2), while upregulating genes associated with a basal-like phenotype (e.g., KRT6A and KRT5) and negative regulators of breast tumorigenesis (e.g., EMP1). Gene methylation status was analyzed to determine whether the changes in expression induced by vitamin D compounds occurred via this mechanism. Ingenuity pathway analysis was performed to identify upstream regulators and downstream signaling pathway genes differentially regulated by vitamin D, including TP63 and vitamin D receptor -mediated canonical pathways in particular. This study provides a global profiling of changes in the gene signature of DCIS regulated by vitamin D compounds and possible targets for chemoprevention of DCIS progression to IDC in patients.
Collapse
Affiliation(s)
- Naing Lin Shan
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Audrey Minden
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey.,Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Philip Furmanski
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey.,Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Min Ji Bak
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Li Cai
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey.,Department of Biomedical Engineering, School of Engineering, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Roman Wernyj
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Davit Sargsyan
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - David Cheng
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Renyi Wu
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Hsiao-Chen D Kuo
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Shanyi N Li
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Mingzhu Fang
- Environmental and Occupational Health Sciences Institute and School of Public Health, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Hubert Maehr
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Ah-Ng Kong
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey.,Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Nanjoo Suh
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey. .,Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| |
Collapse
|
29
|
Wang L, Xu C, Liu X, Yang Y, Cao L, Xiang G, Liu F, Wang S, Liu J, Meng Q, Jiao J, Niu Y. TGF-β1 stimulates epithelial-mesenchymal transition and cancer-associated myoepithelial cell during the progression from in situ to invasive breast cancer. Cancer Cell Int 2019; 19:343. [PMID: 31889895 PMCID: PMC6923856 DOI: 10.1186/s12935-019-1068-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 12/12/2019] [Indexed: 12/28/2022] Open
Abstract
Background The progression of ductal carcinoma in situ (DCIS) into invasive ductal carcinoma (IDC) is prevented by normal breast myoepithelial cells. Studies have suggested that EMT-associated genes were enriched in IDC in contrast to DCIS. This paper explored the relationship and potential mechanism between myoepithelial cells and EMT-associated genes in facilitating the transformation from DCIS to breast cancer. Methods EMT markers and myoepithelial phenotypic markers in IDC, DCIS, and healthy breast tissue were characterized using immunohistochemical assay. Both in vivo and in vitro models were created to mimic the various cell–cell interactions in the development of invasive breast cancer. Results We found that EMT markers were more abundant in invasive carcinomas than DCIS and adjacent normal breast tissue. Meanwhile, TGF-β1 regulated the morphology of MCF-7 (epithelial cells substitute) migration and EMT markers during the transformation from DCIS to invasive breast cancer. Additionally, TGF-β1 also regulated invasion, migration and cytokines secretion of MDA-MB-231 (myoepithelial cells substitute) and epithelial cells when co-cultured with MCF-7 both in vitro and in vivo. Conclusions In conclusion, these findings demonstrated that both EMT phenotypes and cancer-associated myoepithelial cells may have an impact on the development of invasive breast cancer.
Collapse
Affiliation(s)
- Li Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060 China.,2The Second Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
| | - Cong Xu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060 China.,3Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
| | - Xia Liu
- 5Department of Oncology, General Hospital of Tianjin Medical University, 154 Anshan Road, Heping District, Tianjin, 300052 China
| | - Yang Yang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060 China.,3Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
| | - Lu Cao
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060 China.,3Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
| | - Guomin Xiang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060 China.,3Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
| | - Fang Liu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060 China.,3Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
| | - Shuling Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060 China.,4Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
| | - Jing Liu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060 China.,3Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
| | - Qingxiang Meng
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060 China.,3Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
| | - Jiao Jiao
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060 China.,3Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
| | - Yun Niu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060 China.,3Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
| |
Collapse
|
30
|
Steenbrugge J, Vander Elst N, Demeyere K, De Wever O, Sanders NN, Van Den Broeck W, Dirix L, Van Laere S, Meyer E. Comparative Profiling of Metastatic 4T1- vs. Non-metastatic Py230-Based Mammary Tumors in an Intraductal Model for Triple-Negative Breast Cancer. Front Immunol 2019; 10:2928. [PMID: 31921184 PMCID: PMC6927949 DOI: 10.3389/fimmu.2019.02928] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 11/28/2019] [Indexed: 12/30/2022] Open
Abstract
The transition of ductal carcinoma in situ (DCIS) to invasive carcinoma (IC) in breast cancer can be faithfully reproduced by the intraductal mouse model. Envisaging to use this model for therapeutic testing, we aimed to in-depth characterize the tumor immunity associated with the differential progression of two types of intraductal tumors. More specifically, we focused on triple-negative breast cancer (TNBC) and intraductally inoculated luciferase-expressing metastatic 4T1 and locally invasive Py230 cells in lactating mammary glands of syngeneic BALB/c and C57BL/6 female mice, respectively. Although the aggressive 4T1 cells rapidly formed solid tumors, Py230 tumors eventually grew to a similar size through enhanced proliferation. Yet, ductal tumor cell breakthrough and metastasis occurred earlier in the 4T1- compared to the Py230-based intraductal model and was associated with high expression of matrix metalloproteinase (MMP)-9, vascular endothelial growth factor (VEGF), chitinase 3-like 1 (CHI3L1) and lipocalin 2 (LCN2) as well as an increased influx of immune cells (mainly macrophages, neutrophils and T-cells). Moreover, activated cytotoxic T-cells, B-cells and programmed death-1 (PD-1)-positive cells were more prominent in the 4T1-based intraductal model in line with enhanced pro-inflammatory cytokine and gene expression profiles. Py230-based tumors showed a more immunosuppressed anti-inflammatory profile with a high amount of regulatory T-cells, which may account for the decreased T-cell activation but increased proliferation compared to the 4T1-based tumors. Taken together, our results highlight the differential immunological aspects of aggressive metastatic and non-aggressive intraductal progression of 4T1- vs. Py230-based tumors, providing a base for future studies to explore therapy using these intraductal TNBC models.
Collapse
Affiliation(s)
- Jonas Steenbrugge
- Laboratory of Biochemistry, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.,Translational Cancer Research Unit Antwerp, Center for Oncological Research, General Hospital Sint-Augustinus, Wilrijk, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| | - Niels Vander Elst
- Laboratory of Biochemistry, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Kristel Demeyere
- Laboratory of Biochemistry, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Olivier De Wever
- Cancer Research Institute Ghent, Ghent, Belgium.,Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Niek N Sanders
- Cancer Research Institute Ghent, Ghent, Belgium.,Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Wim Van Den Broeck
- Department of Morphology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Luc Dirix
- Translational Cancer Research Unit Antwerp, Center for Oncological Research, General Hospital Sint-Augustinus, Wilrijk, Belgium
| | - Steven Van Laere
- Translational Cancer Research Unit Antwerp, Center for Oncological Research, General Hospital Sint-Augustinus, Wilrijk, Belgium
| | - Evelyne Meyer
- Laboratory of Biochemistry, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| |
Collapse
|
31
|
Ding L, Su Y, Fassl A, Hinohara K, Qiu X, Harper NW, Huh SJ, Bloushtain-Qimron N, Jovanović B, Ekram M, Zi X, Hines WC, Alečković M, Gil Del Alcazar C, Caulfield RJ, Bonal DM, Nguyen QD, Merino VF, Choudhury S, Ethington G, Panos L, Grant M, Herlihy W, Au A, Rosson GD, Argani P, Richardson AL, Dillon D, Allred DC, Babski K, Kim EMH, McDonnell CH, Wagner J, Rowberry R, Bobolis K, Kleer CG, Hwang ES, Blum JL, Cristea S, Sicinski P, Fan R, Long HW, Sukumar S, Park SY, Garber JE, Bissell M, Yao J, Polyak K. Perturbed myoepithelial cell differentiation in BRCA mutation carriers and in ductal carcinoma in situ. Nat Commun 2019; 10:4182. [PMID: 31519911 PMCID: PMC6744561 DOI: 10.1038/s41467-019-12125-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 08/21/2019] [Indexed: 12/24/2022] Open
Abstract
Myoepithelial cells play key roles in normal mammary gland development and in limiting pre-invasive to invasive breast tumor progression, yet their differentiation and perturbation in ductal carcinoma in situ (DCIS) are poorly understood. Here, we investigated myoepithelial cells in normal breast tissues of BRCA1 and BRCA2 germline mutation carriers and in non-carrier controls, and in sporadic DCIS. We found that in the normal breast of non-carriers, myoepithelial cells frequently co-express the p63 and TCF7 transcription factors and that p63 and TCF7 show overlapping chromatin peaks associated with differentiated myoepithelium-specific genes. In contrast, in normal breast tissues of BRCA1 mutation carriers the frequency of p63+TCF7+ myoepithelial cells is significantly decreased and p63 and TCF7 chromatin peaks do not overlap. These myoepithelial perturbations in normal breast tissues of BRCA1 germline mutation carriers may play a role in their higher risk of breast cancer. The fraction of p63+TCF7+ myoepithelial cells is also significantly decreased in DCIS, which may be associated with invasive progression.
Collapse
Affiliation(s)
- Lina Ding
- Department of Medical Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Ying Su
- Department of Medical Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
- Deciphera Pharmaceuticals, Waltham, MA, USA
| | - Anne Fassl
- Department of Cancer Biology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Kunihiko Hinohara
- Department of Medical Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
- Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Xintao Qiu
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Nicholas W Harper
- Department of Medical Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
| | - Sung Jin Huh
- Department of Medical Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
- ImmunoGen, Inc, Waltham, MA, USA
| | - Noga Bloushtain-Qimron
- Department of Medical Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- EMEA Site Intelligence and Activation, Tel Aviv, Israel
| | - Bojana Jovanović
- Department of Medical Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Muhammad Ekram
- Department of Medical Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
- WuXi NextCODE, Cambridge, MA, USA
| | - Xiaoyuan Zi
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
- Second Military Medical University, Shanghai, 200433, P.R. China
| | - William C Hines
- Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
- University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Maša Alečković
- Department of Medical Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Carlos Gil Del Alcazar
- Department of Medical Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Ryan J Caulfield
- Lurie Family Imaging Center, Center for Biomedical Imaging in Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
| | - Dennis M Bonal
- Lurie Family Imaging Center, Center for Biomedical Imaging in Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
| | - Quang-De Nguyen
- Lurie Family Imaging Center, Center for Biomedical Imaging in Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
| | - Vanessa F Merino
- Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Sibgat Choudhury
- Department of Medical Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
- Metamark Genetics Inc, Worcester, MA, USA
| | | | - Laura Panos
- Baylor-Charles A. Sammons Cancer Center, Dallas, TX, 75246, USA
| | - Michael Grant
- Baylor-Charles A. Sammons Cancer Center, Dallas, TX, 75246, USA
| | - William Herlihy
- Baylor-Charles A. Sammons Cancer Center, Dallas, TX, 75246, USA
| | - Alfred Au
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, 94143, USA
| | - Gedge D Rosson
- Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Pedram Argani
- Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Andrea L Richardson
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Pathology, Harvard Medical School, Boston, MA, 02115, USA
- Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Deborah Dillon
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Pathology, Harvard Medical School, Boston, MA, 02115, USA
| | - D Craig Allred
- Department of Pathology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Kirsten Babski
- Sutter Roseville Medical Center, Roseville, CA, 95661, USA
| | - Elizabeth Min Hui Kim
- Sutter Roseville Medical Center, Roseville, CA, 95661, USA
- Cancer Treatment Centers of America, Atlanta, GA, USA
| | | | - Jon Wagner
- Sutter Roseville Medical Center, Roseville, CA, 95661, USA
| | - Ron Rowberry
- Sutter Roseville Medical Center, Roseville, CA, 95661, USA
| | | | - Celina G Kleer
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - E Shelley Hwang
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, 94143, USA
- Duke University, Durham, NC, USA
| | - Joanne L Blum
- Baylor-Charles A. Sammons Cancer Center, Dallas, TX, 75246, USA
| | - Simona Cristea
- Department of Data Science, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- Department of Biostatistics, Harvard T. H. Chan School of Public Health Boston, Boston, MA, 02215, USA
- Department of Stem Cell and Regenerative Biology, Harvard University Cambridge, Cambridge, MA, 02138, USA
| | - Piotr Sicinski
- Department of Cancer Biology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | - Henry W Long
- Department of Medical Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Saraswati Sukumar
- Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - So Yeon Park
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Judy E Garber
- Department of Medical Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Mina Bissell
- Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Jun Yao
- MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA.
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA.
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA.
| |
Collapse
|
32
|
Jeong J, Kim W, Hens J, Dann P, Schedin P, Friedman PA, Wysolmerski JJ. NHERF1 Is Required for Localization of PMCA2 and Suppression of Early Involution in the Female Lactating Mammary Gland. Endocrinology 2019; 160:1797-1810. [PMID: 31087002 PMCID: PMC6619491 DOI: 10.1210/en.2019-00230] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 05/07/2019] [Indexed: 12/11/2022]
Abstract
Prior studies have demonstrated that the calcium pump, plasma membrane calcium ATPase 2 (PMCA2), mediates calcium transport into milk and prevents mammary epithelial cell death during lactation. PMCA2 also regulates cell proliferation and cell death in breast cancer cells, in part by maintaining the receptor tyrosine kinase ErbB2/HER2 within specialized plasma membrane domains. Furthermore, the regulation of PMCA2 membrane localization and activity in breast cancer cells requires its interaction with the PDZ domain-containing scaffolding molecule sodium-hydrogen exchanger regulatory factor (NHERF) 1. In this study, we asked whether NHERF1 also interacts with PMCA2 in normal mammary epithelial cells during lactation. Our results demonstrate that NHERF1 expression is upregulated during lactation and that it interacts with PMCA2 at the apical membrane of secretory luminal epithelial cells. Similar to PMCA2, NHERF1 expression is rapidly reduced by milk stasis after weaning. Examining lactating NHERF1 knockout (KO) mice showed that NHERF1 contributes to the proper apical location of PMCA2, for proper apical-basal polarity in luminal epithelial cells, and that it participates in the suppression of Stat3 activation and the prevention of premature mammary gland involution. Additionally, we found that PMCA2 also interacts with the closely related scaffolding molecule, NHERF2, at the apical membrane, which likely maintains PMCA2 at the plasma membrane of mammary epithelial cells in lactating NHERF1KO mice. Based on these data, we conclude that, during lactation, NHERF1 is required for the proper expression and apical localization of PMCA2, which, in turn, contributes to preventing the premature activation of Stat3 and the lysosome-mediated cell death pathway that usually occur only early in mammary involution.
Collapse
Affiliation(s)
- Jaekwang Jeong
- Section of Endocrinology and Metabolism, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Wonnam Kim
- Division of Pharmacology, College of Korean Medicine, Semyung University, Jecheon, Republic of Korea
| | - Julie Hens
- Section of Endocrinology and Metabolism, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Pamela Dann
- Section of Endocrinology and Metabolism, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Pepper Schedin
- Department of Cell, Developmental, and Cancer Biology, Oregon Health and Science University, Portland, Oregon
| | - Peter A Friedman
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - John J Wysolmerski
- Section of Endocrinology and Metabolism, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
- Correspondence: John J. Wysolmerski, MD, Section of Endocrinology and Metabolism, Department of Internal Medicine, TAC S123a, Yale University School of Medicine, 333 Cedar Street, FMT 102, Box 208020, New Haven, Connecticut 06520. E-mail:
| |
Collapse
|
33
|
Tsuji Y, Kuramochi M, Izawa T, Akiyoshi H, Yamate J, Kuwamura M. Hepatic Myoepithelial Carcinoma in a Dog: Immunohistochemical Comparison With Other Canine Hepatic Carcinomas. Vet Pathol 2019; 56:889-894. [PMID: 31170892 DOI: 10.1177/0300985819854439] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
An 11-year-old female miniature Dachshund dog presented with a solid, soft, gray mass on the hepatic lateral left lobe. Histologically, the mass consisted of neoplastic proliferation of cells with round nuclei and eosinophilic and vacuolated cytoplasm arranged in alveolar, trabecular, and solid patterns. Immunohistochemically, the neoplastic cells were positive for pancytokeratin (CK AE1/AE3), CK5, CK14, vimentin, Sox9, and myoepithelial markers (α-smooth muscle actin, p63, and calponin). The morphological and immunohistochemical findings indicated a diagnosis of myoepithelial carcinoma. We conducted immunohistochemical studies on other representative canine hepatic tumors. Although the myoepithelial phenotype was not observed in the hepatocellular carcinoma, some tumor cells in cholangiocarcinoma showed immunohistochemical features of myoepithelium, suggesting that some neoplastic cells in cholangiocarcinoma may have the potential to differentiate into myoepithelial cells. To our knowledge, this is the first report in veterinary medicine of a hepatic carcinoma with a myoepithelial phenotype.
Collapse
Affiliation(s)
- Yuka Tsuji
- Laboratory of Veterinary Pathology, Osaka Prefecture University, Izumisano, Osaka, Japan
| | - Mizuki Kuramochi
- Laboratory of Veterinary Pathology, Osaka Prefecture University, Izumisano, Osaka, Japan
| | - Takeshi Izawa
- Laboratory of Veterinary Pathology, Osaka Prefecture University, Izumisano, Osaka, Japan
| | - Hideo Akiyoshi
- Veterinary Surgery, Osaka Prefecture University, Izumisano, Osaka, Japan
| | - Jyoji Yamate
- Laboratory of Veterinary Pathology, Osaka Prefecture University, Izumisano, Osaka, Japan
| | - Mitsuru Kuwamura
- Laboratory of Veterinary Pathology, Osaka Prefecture University, Izumisano, Osaka, Japan
| |
Collapse
|
34
|
Atiya HI, Dvorkin-Gheva A, Hassell J, Patel S, Parker RL, Hartstone-Rose A, Hodge J, Fan D, Ramsdell AF. Intraductal Adaptation of the 4T1 Mouse Model of Breast Cancer Reveals Effects of the Epithelial Microenvironment on Tumor Progression and Metastasis. Anticancer Res 2019; 39:2277-2287. [PMID: 31092419 DOI: 10.21873/anticanres.13344] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 04/02/2019] [Accepted: 04/05/2019] [Indexed: 11/10/2022]
Abstract
BACKGROUND Low success rates in oncology drug development are prompting re-evaluation of preclinical models, including orthotopic tumor engraftment. In breast cancer models, tumor cells are typically injected into mouse mammary fat pads (MFP). However, this approach bypasses the epithelial microenvironment, potentially altering tumor properties in ways that affect translational application. MATERIALS AND METHODS Tumors were generated by mammary intraductal (MIND) engraftment of 4T1 carcinoma cells. Growth, histopathology, and molecular features were quantified. RESULTS Despite growth similar to that of 4T1 MFP tumors, 4T1 MIND tumors exhibit distinct histopathology and increased metastasis. Furthermore, >6,000 transcripts were found to be uniquely up-regulated in 4T1 MIND tumor cells, including genes that drive several cancer hallmarks, in addition to two known therapeutic targets that were not up-regulated in 4T1 MFP tumor cells. CONCLUSION Engraftment into the epithelial microenvironment generates tumors that more closely recapitulate the complexity of malignancy, suggesting that intraductal adaptation of orthotopic mammary models may be an important step towards improving outcomes in preclinical drug screening and development.
Collapse
Affiliation(s)
- Huda I Atiya
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC, U.S.A
| | - Anna Dvorkin-Gheva
- Department of Biochemistry and Biomedical Science, McMaster University, Hamilton, ON, Canada
| | - John Hassell
- Department of Biochemistry and Biomedical Science, McMaster University, Hamilton, ON, Canada
| | - Shrusti Patel
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC, U.S.A
| | - Rachel L Parker
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC, U.S.A
| | - Adam Hartstone-Rose
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, U.S.A
| | - Johnie Hodge
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC, U.S.A
| | - Daping Fan
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC, U.S.A
| | - Ann F Ramsdell
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC, U.S.A. .,Program in Women's and Gender Studies, College of Arts and Sciences, University of South Carolina, Columbia, SC, U.S.A.,Department of Regenerative Medicine and Cell Biology, and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, U.S.A
| |
Collapse
|
35
|
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
|
36
|
Sinha VC, Piwnica-Worms H. Intratumoral Heterogeneity in Ductal Carcinoma In Situ: Chaos and Consequence. J Mammary Gland Biol Neoplasia 2018; 23:191-205. [PMID: 30194658 PMCID: PMC6934090 DOI: 10.1007/s10911-018-9410-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/30/2018] [Indexed: 02/06/2023] Open
Abstract
Ductal carcinoma in situ (DCIS) is a non-invasive proliferative growth in the breast that serves as a non-obligate precursor to invasive ductal carcinoma. The widespread adoption of screening mammography has led to a steep increase in the detection of DCIS, which now comprises approximately 20% of new breast cancer diagnoses in the United States. Interestingly, the intratumoral heterogeneity (ITH) that has been observed in invasive breast cancers may have been established early in tumorigenesis, given the vast and varied ITH that has been detected in DCIS. This review will discuss the intratumoral heterogeneity of DCIS, focusing on the phenotypic and genomic heterogeneity of tumor cells, as well as the compositional heterogeneity of the tumor microenvironment. In addition, we will assess the spatial heterogeneity that is now being appreciated in these lesions, and summarize new approaches to evaluate heterogeneity of tumor and stromal cells in the context of their spatial organization. Importantly, we will discuss how a growing understanding of ITH has led to a more holistic appreciation of the complex biology of DCIS, specifically its evolution and natural history. Finally, we will consider ways in which our knowledge of DCIS ITH might be translated in the future to guide clinical care for DCIS patients.
Collapse
Affiliation(s)
- Vidya C Sinha
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Houston, TX, 77030, USA
| | - Helen Piwnica-Worms
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Houston, TX, 77030, USA.
| |
Collapse
|
37
|
Behbod F, Gomes AM, Machado HL. Modeling Human Ductal Carcinoma In Situ in the Mouse. J Mammary Gland Biol Neoplasia 2018; 23:269-278. [PMID: 30145750 PMCID: PMC6244883 DOI: 10.1007/s10911-018-9408-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 08/22/2018] [Indexed: 12/13/2022] Open
Abstract
Breast cancer development is a multi-step process in which genetic and molecular heterogeneity occurs at multiple stages. Ductal carcinoma arises from pre-invasive lesions such as atypical ductal hyperplasia (ADH) and ductal carcinoma in situ (DCIS), which progress to invasive and metastatic cancer. The feasibility of obtaining tissue samples from all stages of progression from the same patient is low, and thus molecular studies dissecting the mechanisms that mediate the transition from pre-invasive DCIS to invasive carcinoma have been hampered. In the past 25 years, numerous mouse models have been developed that partly recapitulate the histological and biological properties of early stage lesions. In this review, we discuss in vivo model systems of breast cancer progression from syngeneic mouse models to human xenografts, with particular focus on how accurately these models mimic human disease.
Collapse
Affiliation(s)
- Fariba Behbod
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Angelica M Gomes
- Department of Biochemistry and Molecular Biology, Tulane Cancer Center, Tulane University School of Medicine, 1430 Tulane Ave, #8543, New Orleans, LA, USA
| | - Heather L Machado
- Department of Biochemistry and Molecular Biology, Tulane Cancer Center, Tulane University School of Medicine, 1430 Tulane Ave, #8543, New Orleans, LA, USA.
| |
Collapse
|
38
|
Steenbrugge J, Breyne K, Demeyere K, De Wever O, Sanders NN, Van Den Broeck W, Colpaert C, Vermeulen P, Van Laere S, Meyer E. Anti-inflammatory signaling by mammary tumor cells mediates prometastatic macrophage polarization in an innovative intraductal mouse model for triple-negative breast cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:191. [PMID: 30111338 PMCID: PMC6094904 DOI: 10.1186/s13046-018-0860-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 07/31/2018] [Indexed: 12/11/2022]
Abstract
Background Murine breast cancer models relying on intraductal tumor cell inoculations are attractive because they allow the study of breast cancer from early ductal carcinoma in situ to metastasis. Using a fully immunocompetent 4T1-based intraductal model for triple-negative breast cancer (TNBC) we aimed to investigate the immunological responses that guide such intraductal tumor progression, focusing on the prominent role of macrophages. Methods Intraductal inoculations were performed in lactating female mice with luciferase-expressing 4T1 mammary tumor cells either with or without additional RAW264.7 macrophages, mimicking basal versus increased macrophage-tumor cell interactions in the ductal environment. Imaging of 4T1-derived luminescence was used to monitor primary tumor growth and metastases. Tumor proliferation, hypoxia, disruption of the ductal architecture and tumor immune populations were determined immunohistochemically. M1- (pro-inflammatory) and M2-related (anti-inflammatory) cytokine levels were determined by Luminex assays and ELISA to investigate the activation state of the macrophage inoculum. Levels of the metastatic proteins matrix metalloproteinase 9 (MMP-9) and vascular endothelial growth factor (VEGF) as well as of the immune-related disease biomarkers chitinase 3-like 1 (CHI3L1) and lipocalin 2 (LCN2) were measured by ELISA to evaluate disease progression at the protein level. Results Mice intraductally co-injected with macrophages showed severe splenomegaly with faster ductal breakthrough of tumor cells and increased metastases in axillary lymph nodes and lungs. These mice showed higher M1-related cytokines in the early disease stages (at 1 to 3 weeks post-inoculation) due to the pro-inflammatory nature of RAW264.7 macrophages with increased Ly6G-positive neutrophils and decreased anti-inflammatory macrophages in the tumor microenvironment. However, upon metastasis (at 5 weeks post-inoculation), a prominent increase in M2-related cytokine levels was detected and established a tumor microenvironment with similar immune populations and cytokine responses as in mice which received only 4T1 tumor cells. The observed tumor-associated immune responses and the increased metastasis were associated with significantly induced local and systemic levels of MMP-9, VEGF, CHI3L1 and LCN2. Conclusions The current experimental study with an innovative immunocompetent intraductal model for TNBC pinpoints towards a metastasis-supporting M1 to M2 macrophage polarization in the mammary ducts mediated by 4T1-derived signaling. We propose to explore this process as immunotherapeutic target. Electronic supplementary material The online version of this article (10.1186/s13046-018-0860-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jonas Steenbrugge
- Laboratory of Biochemistry, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium. .,Translational Cancer Research Unit Antwerp, Center for Oncological Research, General Hospital Sint-Augustinus, Wilrijk, Belgium. .,Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| | - Koen Breyne
- Laboratory of Biochemistry, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium.,Present address: Department of Pharmacological and Physiological Sciences, The University of Chicago, Chicago, IL, USA
| | - Kristel Demeyere
- Laboratory of Biochemistry, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Olivier De Wever
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.,Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Ghent, Belgium
| | - Niek N Sanders
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.,Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Wim Van Den Broeck
- Department of Morphology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Cecile Colpaert
- Department of Pathology, General Hospital Sint-Augustinus, Wilrijk, Belgium
| | - Peter Vermeulen
- Translational Cancer Research Unit Antwerp, Center for Oncological Research, General Hospital Sint-Augustinus, Wilrijk, Belgium
| | - Steven Van Laere
- Translational Cancer Research Unit Antwerp, Center for Oncological Research, General Hospital Sint-Augustinus, Wilrijk, Belgium
| | - Evelyne Meyer
- Laboratory of Biochemistry, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|
39
|
Özdemir BC, Sflomos G, Brisken C. The challenges of modeling hormone receptor-positive breast cancer in mice. Endocr Relat Cancer 2018; 25:R319-R330. [PMID: 29563191 DOI: 10.1530/erc-18-0063] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 03/21/2018] [Indexed: 12/26/2022]
Abstract
Estrogen receptor-positive (ER+) tumors account for 70-80% of all breast cancer (BC) cases and are characterized by estrogen dependency for their growth. Endocrine therapies using estrogen receptor antagonists or aromatase inhibitors represent a key component of the standard of care for these tumors. The occurrence of de novo or acquired resistance to estrogen withdrawal represents an important clinical problem, impacting on patient survival. In addition, despite an initially favorable outcome, a part of ER+ BC patients present with disease recurrence locally or at distant sites years or even decades after apparent remission. In vivo models that closely mimic human disease are urgently needed to study the biology of these tumors, investigate the molecular mechanisms underlying endocrine resistance and identify patients at risk of recurrence. Despite the similarities in the overall hormonal regulation of mammary gland development between mice and humans, the majority of the mammary carcinomas occurring in genetically engineered mouse models (GEMMs) are ER negative and most xenograft models are based on few ER+ cancer cell lines. We recently showed that the microenvironment is critical for ER+ cancer cells and discuss in this review the potential of intraductal xenograft model for basic and preclinical research.
Collapse
Affiliation(s)
- Berna C Özdemir
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
- International Cancer Prevention Institute, Epalinges, Switzerland
| | - George Sflomos
- ISREC - Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Cathrin Brisken
- International Cancer Prevention Institute, Epalinges, Switzerland
- ISREC - Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
40
|
Gray E, Mitchell E, Jindal S, Schedin P, Chang YH. A METHOD FOR QUANTIFICATION OF CALPONIN EXPRESSION IN MYOEPITHELIAL CELLS IN IMMUNOHISTOCHEMICAL IMAGES OF DUCTAL CARCINOMA IN SITU. PROCEEDINGS. IEEE INTERNATIONAL SYMPOSIUM ON BIOMEDICAL IMAGING 2018; 2018:796-799. [PMID: 30364524 PMCID: PMC6196724 DOI: 10.1109/isbi.2018.8363692] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Ductal carcinoma in situ (DCIS) is breast cancer confined within mammary ducts, surrounded by an intact myoepithelial cell layer that prevents local invasion. A DCIS diagnosis confers increased lifetime risk of developing invasive breast cancer (IBC) and results in surgical excision with radiation, and possibly endocrine- or chemo-therapy. DCIS is known to be over treated, with associated co-morbidities. Biomarkers are needed that delineate patients at low risk of DCIS progression from patients requiring more aggressive treatment. Investigating the role of myoepithelial cell differentiation in barrier function is anticipated to provide insight into DCIS progression and delineate between low and high risk lesions. Here, we develop a high throughput technique to assess loss of myoepithelial differentiation markers. This method facilitates automated analysis of a clinically relevant histopathologic feature, as demonstrated by a high correlation with pathologist annotation (r = 0.959), and further, contributes analytical foundations to a multiplexed immunohistochemistry (IHC) approach.
Collapse
Affiliation(s)
- Elliot Gray
- Department of Biomedical Engineering and Computational Biology Program
| | - Elizabeth Mitchell
- Department of Cell, Developmental and Cancer Biology Oregon Health & Science University
| | - Sonali Jindal
- Department of Cell, Developmental and Cancer Biology Oregon Health & Science University
| | - Pepper Schedin
- Department of Cell, Developmental and Cancer Biology Oregon Health & Science University
| | - Young Hwan Chang
- Department of Biomedical Engineering and Computational Biology Program
| |
Collapse
|
41
|
Bresson L, Faraldo MM, Di-Cicco A, Quintanilla M, Glukhova MA, Deugnier MA. Podoplanin regulates mammary stem cell function and tumorigenesis by potentiating Wnt/β-catenin signaling. Development 2018; 145:dev.160382. [PMID: 29361573 DOI: 10.1242/dev.160382] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 01/15/2018] [Indexed: 12/28/2022]
Abstract
Stem cells (SCs) drive mammary development, giving rise postnatally to an epithelial bilayer composed of luminal and basal myoepithelial cells. Dysregulation of SCs is thought to be at the origin of certain breast cancers; however, the molecular identity of SCs and the factors regulating their function remain poorly defined. We identified the transmembrane protein podoplanin (Pdpn) as a specific marker of the basal compartment, including multipotent SCs, and found Pdpn localized at the basal-luminal interface. Embryonic deletion of Pdpn targeted to basal cells diminished basal and luminal SC activity and affected the expression of several Wnt/β-catenin signaling components in basal cells. Moreover, Pdpn loss attenuated mammary tumor formation in a mouse model of β-catenin-induced breast cancer, limiting tumor-initiating cell expansion and promoting molecular features associated with mesenchymal-to-epithelial cell transition. In line with the loss-of-function data, we demonstrated that mechanistically Pdpn enhances Wnt/β-catenin signaling in mammary basal cells. Overall, this study uncovers a role for Pdpn in mammary SC function and, importantly, identifies Pdpn as a new regulator of Wnt/β-catenin signaling, a key pathway in mammary development and tumorigenesis.
Collapse
Affiliation(s)
- Laura Bresson
- Institut Curie, PSL Research University, CNRS, UMR144, Paris, F-75248, France.,Université Paris Sud, Université Paris-Saclay, F-91405, Orsay, France.,Sorbonne Universités, UPMC Univ Paris 06, F-75005, Paris, France
| | - Marisa M Faraldo
- Institut Curie, PSL Research University, CNRS, UMR144, Paris, F-75248, France.,INSERM, Paris, F-75013, France
| | - Amandine Di-Cicco
- Institut Curie, PSL Research University, CNRS, UMR144, Paris, F-75248, France
| | - Miguel Quintanilla
- Instituto de Investigaciones Biomedicas Alberto Sols, CSIC-UAM, Madrid, Spain
| | - Marina A Glukhova
- Institut Curie, PSL Research University, CNRS, UMR144, Paris, F-75248, France.,INSERM, Paris, F-75013, France
| | - Marie-Ange Deugnier
- Institut Curie, PSL Research University, CNRS, UMR144, Paris, F-75248, France .,INSERM, Paris, F-75013, France
| |
Collapse
|
42
|
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
|
43
|
Al Nemer AM. Histologic factors predicting invasion in patients with ductal carcinoma in situ (DCIS) in the preoperative core biopsy. Pathol Res Pract 2017; 213:429-434. [DOI: 10.1016/j.prp.2017.02.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 02/12/2017] [Accepted: 02/21/2017] [Indexed: 10/20/2022]
|
44
|
Tucker H, Beaudry K, Parsons C, Ellis S, Akers R. Impaired mammary development in tamoxifen-treated prepubertal heifers is associated with altered development and morphology of myoepithelial cells. J Dairy Sci 2016; 99:10093-10101. [DOI: 10.3168/jds.2016-11550] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 08/18/2016] [Indexed: 01/15/2023]
|
45
|
Steenbrugge J, Breyne K, Denies S, Dekimpe M, Demeyere K, De Wever O, Vermeulen P, Van Laere S, Sanders NN, Meyer E. Comparison of the Adipose and Luminal Mammary Gland Compartment as Orthotopic Inoculation Sites in a 4T1-Based Immunocompetent Preclinical Model for Triple-Negative Breast Cancer. J Mammary Gland Biol Neoplasia 2016; 21:113-122. [PMID: 27714576 DOI: 10.1007/s10911-016-9362-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 09/15/2016] [Indexed: 10/20/2022] Open
Abstract
Breast tumorigenesis is classically studied in mice by inoculating tumor cells in the fat pad, the adipose compartment of the mammary gland. Alternatively, the mammary ducts, which constitute the luminal mammary gland compartment, also provide a suitable inoculation site to induce breast cancer in murine models. The microenvironments in these compartments influence tumor cell progression, yet this effect has not been investigated in an immunocompetent context. Here, we compared both mammary gland compartments as distinct inoculation sites, taking into account the immunological aspect by inoculating 4T1 tumor cells in immunocompetent mice. Following tumor cell inoculation in the adipose compartment of non-pretreated/naive, hormonally pretreated/naive and non-pretreated/lactating mice, the primary tumors developed similarly. However, a slower onset of primary tumor growth was found after inoculations in the luminal compartment of non-pretreated/lactating mice. Despite this difference in tumor development rate, metastasis to the liver and lungs was equally observed and was accompanied by lymphatic spreading of tumor cells and progressive splenomegaly with both inoculation types. Chitinase 3-like 1 (CHI3L1) and lipocalin 2 (LCN2) served as innovative biomarkers for disease progression showing increased levels in primary tumors and sera of the non-pretreated/lactating inoculation groups. A slower increase in circulating CHI3L1 but not LCN2 levels, was observed after inoculations in the luminal compartment which corroborated the slower tumor development at this inoculation site. Our results highlight the critical impact of different mammary gland compartments on tumor development in syngeneic murine models and support the use of novel tumor progression biomarkers in an immune-competent environment.
Collapse
Affiliation(s)
- Jonas Steenbrugge
- Laboratory of Biochemistry, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.
- Translational Cancer Research Unit Antwerp, Center for Oncological Research, General Hospital Sint-Augustinus, Wilrijk, Belgium.
| | - Koen Breyne
- Laboratory of Biochemistry, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Sofie Denies
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Melissa Dekimpe
- Laboratory of Biochemistry, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Kristel Demeyere
- Laboratory of Biochemistry, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Olivier De Wever
- Department of Radiation Oncology and Experimental Cancer Research, Laboratory of Experimental Cancer Research, Ghent University, Ghent, Belgium
| | - Peter Vermeulen
- Translational Cancer Research Unit Antwerp, Center for Oncological Research, General Hospital Sint-Augustinus, Wilrijk, Belgium
| | - Steven Van Laere
- Translational Cancer Research Unit Antwerp, Center for Oncological Research, General Hospital Sint-Augustinus, Wilrijk, Belgium
| | - Niek N Sanders
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Evelyne Meyer
- Laboratory of Biochemistry, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
46
|
Wang Y, Jindal S, Martel M, Wu Y, Schedin P, Troxell M. Myoepithelial cells in lobular carcinoma in situ: distribution and immunophenotype. Hum Pathol 2016; 55:126-34. [PMID: 27195907 DOI: 10.1016/j.humpath.2016.05.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 04/15/2016] [Accepted: 05/05/2016] [Indexed: 12/18/2022]
Abstract
Myoepithelial cells have important physical and paracrine roles in breast tissue development, maintenance, and tumor suppression. Recent molecular and immunohistochemical studies have demonstrated phenotypic alterations in ductal carcinoma in situ-associated myoepithelial cells. Although the relationship of lobular carcinoma in situ (LCIS) and myoepithelial cells was described in 1980, further characterization of LCIS-associated myoepithelial cells is lacking. We stained 27 breast specimens harboring abundant LCIS with antibodies to smooth muscle myosin heavy chain, smooth muscle actin, and calponin. Dual stains for E-cadherin/smooth muscle myosin heavy chain and CK7/p63 were also performed. In each case, the intensity and distribution of staining in LCIS-associated myoepithelial cells were compared with normal breast tissue on the same slide. In 78% of the cases, LCIS-associated myoepithelial cells demonstrated decreased staining intensity for one or more myoepithelial markers. The normal localization of myoepithelial cells (flat against the basement membrane, pattern N) was seen in 96% of LCIS, yet 85% of cases had areas with myoepithelial cell cytoplasm oriented perpendicular to the basement membrane (pattern P), and in 30% of cases, myoepithelial cells appeared focally admixed with LCIS cells (pattern C). This study characterizes detailed architectural and immunophenotypic alterations of LCIS-associated myoepithelial cells. The finding of variably diminished staining favors application of several myoepithelial immunostains in clinical practice. The interaction of LCIS with myoepithelial cells, especially in light of the perpendicular and central architectural arrangements, deserves further mechanistic investigation.
Collapse
Affiliation(s)
- Ying Wang
- Department of Pathology, Oregon Health & Science University, Portland, OR 97239
| | - Sonali Jindal
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239
| | - Maritza Martel
- Department of Pathology, Providence Health and Services, Portland, OR 97213
| | - Yaping Wu
- Department of Pathology, Providence Health and Services, Portland, OR 97213
| | - Pepper Schedin
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239; Oregon Health & Science University, Knight Cancer Institute, Portland, OR 97239
| | - Megan Troxell
- Department of Pathology, Oregon Health & Science University, Portland, OR 97239; Oregon Health & Science University, Knight Cancer Institute, Portland, OR 97239.
| |
Collapse
|
47
|
Pang JMB, Gorringe KL, Fox SB. Reply to the Baader-Meinhof phenomenon in ductal carcinoma in situof the breast. Histopathology 2016; 69:523-4. [DOI: 10.1111/his.12968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
48
|
Jacob T, Gray JW, Troxell M, Vu TQ. Multiplexed imaging reveals heterogeneity of PI3K/MAPK network signaling in breast lesions of known PIK3CA genotype. Breast Cancer Res Treat 2016; 159:575-83. [PMID: 27581127 DOI: 10.1007/s10549-016-3962-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 08/25/2016] [Indexed: 11/29/2022]
Abstract
PURPOSE Activating genetic changes in the phosphatidylinositol-3-kinase (PI3K) signaling pathway are found in over half of invasive breast cancers (IBCs). Previously, we discovered numerous hotspot PIK3CA mutations in proliferative breast lesions. Here, we investigate the spatial nature of PI3K pathway signaling and its relationship with PI3K genotype in breast lesions. METHODS We identified PI3K phosphosignaling network signatures in columnar cell change (CCL), usual ductal hyperplasia (UDH), ductal carcinoma in situ (DCIS), and IBC in 26 lesions of known PIK3CA genotype from 10 human breast specimens using a hyperspectral-based multiplexed tissue imaging platform (MTIP) to simultaneously quantitate PI3K/MAPK pathway targets (pAKT473, pAKT308, pPRAS40, pS6, and pERK) in FFPE tissue, with single-cell resolution. RESULTS We found that breast lesional epithelia contained spatially heterogeneous patterns of PI3K pathway phosphoprotein signatures, even within microscopic areas of CCL, UDH, DCIS, and IBC. Most lesions contained 3-12 unique phosphoprotein signatures within the same microscopic field. The dominant phosphoprotein signature for each lesion was not well correlated with lesion genotype or lesion histology, yet samples from the same patient tended to group together. Further, 5 UDH/CCL lesions across different patients had a common phosphosignature at the epithelial-stromal interface (possible myoepithelial cells) that was distinct from both the adjacent lesional epithelium and distinct from adjacent stroma. CONCLUSION We present the first spatial mapping of PI3K phosphoprotein networks in proliferative breast lesions and demonstrate complex PI3K signaling heterogeneity that defies simple correlation between PIK3CA genotype and phosphosignal pattern.
Collapse
Affiliation(s)
- Thomas Jacob
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, 97201, USA
| | - Joe W Gray
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, 97201, USA.,OHSU Center for Spatial Systems Bioscience, Portland, OR, 97201, USA.,The Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Megan Troxell
- The Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 97239, USA.,Department of Pathology, Oregon Health & Science University, Portland, OR, 97239, USA.,Department of Pathology, Stanford University Medical Center, Stanford, CA, 94305, USA
| | - Tania Q Vu
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, 97201, USA. .,OHSU Center for Spatial Systems Bioscience, Portland, OR, 97201, USA. .,The Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 97239, USA.
| |
Collapse
|
49
|
The extracellular matrix in breast cancer predicts prognosis through composition, splicing, and crosslinking. Exp Cell Res 2015; 343:73-81. [PMID: 26597760 DOI: 10.1016/j.yexcr.2015.11.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 11/11/2015] [Indexed: 12/19/2022]
Abstract
The extracellular matrix in the healthy breast has an important tumor suppressive role, whereas the abnormal ECM in tumors can promote aggressiveness, and has been linked to breast cancer relapse, survival and resistance to chemotherapy. This review article gives an overview of the elements of the ECM which have been linked to prognosis of breast cancers, including changes in ECM protein composition, splicing, and microstructure.
Collapse
|