201
|
Yu T, Di G. Role of tumor microenvironment in triple-negative breast cancer and its prognostic significance. Chin J Cancer Res 2017; 29:237-252. [PMID: 28729775 PMCID: PMC5497211 DOI: 10.21147/j.issn.1000-9604.2017.03.10] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Breast cancer has been shown to live in the tumor microenvironment, which consists of not only breast cancer cells themselves but also a significant amount of pathophysiologically altered surrounding stroma and cells. Diverse components of the breast cancer microenvironment, such as suppressive immune cells, re-programmed fibroblast cells, altered extracellular matrix (ECM) and certain soluble factors, synergistically impede an effective anti-tumor response and promote breast cancer progression and metastasis. Among these components, stromal cells in the breast cancer microenvironment are characterized by molecular alterations and aberrant signaling pathways, whereas the ECM features biochemical and biomechanical changes. However, triple-negative breast cancer (TNBC), the most aggressive subtype of this disease that lacks effective therapies available for other subtypes, is considered to feature a unique microenvironment distinct from that of other subtypes, especially compared to Luminal A subtype. Because these changes are now considered to significantly impact breast cancer development and progression, these unique alterations may serve as promising prognostic factors of clinical outcome or potential therapeutic targets for the treatment of TNBC. In this review, we focus on the composition of the TNBC microenvironment, concomitant distinct biological alteration, specific interplay between various cell types and TNBC cells, and the prognostic implications of these findings.
Collapse
Affiliation(s)
- Tianjian Yu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Genhong Di
- Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
202
|
Breast Cancer Microenvironment and the Metastatic Process. Breast Cancer 2017. [DOI: 10.1007/978-3-319-48848-6_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
203
|
Tumour Heterogeneity: The Key Advantages of Single-Cell Analysis. Int J Mol Sci 2016; 17:ijms17122142. [PMID: 27999407 PMCID: PMC5187942 DOI: 10.3390/ijms17122142] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 12/12/2016] [Accepted: 12/13/2016] [Indexed: 01/06/2023] Open
Abstract
Tumour heterogeneity refers to the fact that different tumour cells can show distinct morphological and phenotypic profiles, including cellular morphology, gene expression, metabolism, motility, proliferation and metastatic potential. This phenomenon occurs both between tumours (inter-tumour heterogeneity) and within tumours (intra-tumour heterogeneity), and it is caused by genetic and non-genetic factors. The heterogeneity of cancer cells introduces significant challenges in using molecular prognostic markers as well as for classifying patients that might benefit from specific therapies. Thus, research efforts for characterizing heterogeneity would be useful for a better understanding of the causes and progression of disease. It has been suggested that the study of heterogeneity within Circulating Tumour Cells (CTCs) could also reflect the full spectrum of mutations of the disease more accurately than a single biopsy of a primary or metastatic tumour. In previous years, many high throughput methodologies have raised for the study of heterogeneity at different levels (i.e., RNA, DNA, protein and epigenetic events). The aim of the current review is to stress clinical implications of tumour heterogeneity, as well as current available methodologies for their study, paying specific attention to those able to assess heterogeneity at the single cell level.
Collapse
|
204
|
Das S. Identification and targeting of microRNAs modulating acquired chemotherapy resistance in Triple negative breast cancer (TNBC): A better strategy to combat chemoresistance. Med Hypotheses 2016; 96:5-8. [DOI: 10.1016/j.mehy.2016.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 09/14/2016] [Indexed: 01/09/2023]
|
205
|
Targeting the angio-proteostasis network: Combining the forces against cancer. Pharmacol Ther 2016; 167:1-12. [DOI: 10.1016/j.pharmthera.2016.07.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 07/14/2016] [Indexed: 01/24/2023]
|
206
|
Screening and analysis of breast cancer genes regulated by the human mammary microenvironment in a humanized mouse model. Oncol Lett 2016; 12:5261-5268. [PMID: 28101242 DOI: 10.3892/ol.2016.5310] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 03/09/2016] [Indexed: 01/02/2023] Open
Abstract
Tumor microenvironments play critical regulatory roles in tumor growth. Although mouse cancer models have contributed to the understanding of human tumor biology, the effectiveness of mouse cancer models is limited by the inability of the models to accurately present humanized tumor microenvironments. Previously, a humanized breast cancer model in severe combined immunodeficiency mice was established, in which human breast cancer tissue was implanted subcutaneously, followed by injection of human breast cancer cells. It was demonstrated that breast cancer cells showed improved growth in the human mammary microenvironment compared with a conventional subcutaneous mouse model. In the present study, the novel mouse model and microarray technology was used to analyze changes in the expression of genes in breast cancer cells that are regulated by the human mammary microenvironment. Humanized breast and conventional subcutaneous mouse models were established, and orthotopic tumor cells were obtained from orthotopic tumor masses by primary culture. An expression microarray using Illumina HumanHT-12 v4 Expression BeadChip and database analyses were performed to investigate changes in gene expression between tumors from each microenvironment. A total of 94 genes were differentially expressed between the primary cells cultured from the humanized and conventional mouse models. Significant upregulation of genes that promote cell proliferation and metastasis or inhibit apoptosis, such as SH3-domain binding protein 5 (BTK-associated), sodium/chloride cotransporter 3 and periostin, osteoblast specific factor, and genes that promote angiogenesis, such as KIAA1618, was also noted. Other genes that restrain cell proliferation and accelerate cell apoptosis, including tripartite motif containing TRIM36 and NES1, were downregulated. The present results revealed differences in various aspects of tumor growth and metabolism between the two model groups and indicated the functional changes specific to the human mammary microenvironment.
Collapse
|
207
|
Breast Cancer Cell Colonization of the Human Bone Marrow Adipose Tissue Niche. Neoplasia 2016; 17:849-861. [PMID: 26696367 PMCID: PMC4688564 DOI: 10.1016/j.neo.2015.11.005] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Revised: 11/05/2015] [Accepted: 11/10/2015] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND/OBJECTIVES Bone is a preferred site of breast cancer metastasis, suggesting the presence of tissue-specific features that attract and promote the outgrowth of breast cancer cells. We sought to identify parameters of human bone tissue associated with breast cancer cell osteotropism and colonization in the metastatic niche. METHODS Migration and colonization patterns of MDA-MB-231-fLuc-EGFP (luciferase-enhanced green fluorescence protein) and MCF-7-fLuc-EGFP breast cancer cells were studied in co-culture with cancellous bone tissue fragments isolated from 14 hip arthroplasties. Breast cancer cell migration into tissues and toward tissue-conditioned medium was measured in Transwell migration chambers using bioluminescence imaging and analyzed as a function of secreted factors measured by multiplex immunoassay. Patterns of breast cancer cell colonization were evaluated with fluorescence microscopy and immunohistochemistry. RESULTS Enhanced MDA-MB-231-fLuc-EGFP breast cancer cell migration to bone-conditioned versus control medium was observed in 12/14 specimens (P = .0014) and correlated significantly with increasing levels of the adipokines/cytokines leptin (P = .006) and IL-1β (P = .001) in univariate and multivariate regression analyses. Fluorescence microscopy and immunohistochemistry of fragments underscored the extreme adiposity of adult human bone tissues and revealed extensive breast cancer cell colonization within the marrow adipose tissue compartment. CONCLUSIONS Our results show that breast cancer cells migrate to human bone tissue-conditioned medium in association with increasing levels of leptin and IL-1β, and colonize the bone marrow adipose tissue compartment of cultured fragments. Bone marrow adipose tissue and its molecular signals may be important but understudied components of the breast cancer metastatic niche.
Collapse
|
208
|
Morgan MM, Johnson BP, Livingston MK, Schuler LA, Alarid ET, Sung KE, Beebe DJ. Personalized in vitro cancer models to predict therapeutic response: Challenges and a framework for improvement. Pharmacol Ther 2016; 165:79-92. [PMID: 27218886 PMCID: PMC5439438 DOI: 10.1016/j.pharmthera.2016.05.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Personalized cancer therapy focuses on characterizing the relevant phenotypes of the patient, as well as the patient's tumor, to predict the most effective cancer therapy. Historically, these methods have not proven predictive in regards to predicting therapeutic response. Emerging culture platforms are designed to better recapitulate the in vivo environment, thus, there is renewed interest in integrating patient samples into in vitro cancer models to assess therapeutic response. Successful examples of translating in vitro response to clinical relevance are limited due to issues with patient sample acquisition, variability and culture. We will review traditional and emerging in vitro models for personalized medicine, focusing on the technologies, microenvironmental components, and readouts utilized. We will then offer our perspective on how to apply a framework derived from toxicology and ecology towards designing improved personalized in vitro models of cancer. The framework serves as a tool for identifying optimal readouts and culture conditions, thus maximizing the information gained from each patient sample.
Collapse
Affiliation(s)
- Molly M Morgan
- Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Brian P Johnson
- Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Megan K Livingston
- Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Linda A Schuler
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Elaine T Alarid
- Department of Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | - Kyung E Sung
- Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States.
| | - David J Beebe
- Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States; Department of Oncology, University of Wisconsin-Madison, Madison, WI, United States.
| |
Collapse
|
209
|
Wang R, Chu GCY, Mrdenovic S, Annamalai AA, Hendifar AE, Nissen NN, Tomlinson JS, Lewis M, Palanisamy N, Tseng HR, Posadas EM, Freeman MR, Pandol SJ, Zhau HE, Chung LWK. Cultured circulating tumor cells and their derived xenografts for personalized oncology. Asian J Urol 2016; 3:240-253. [PMID: 29264192 PMCID: PMC5730836 DOI: 10.1016/j.ajur.2016.08.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 08/16/2016] [Indexed: 02/07/2023] Open
Abstract
Recent cancer research has demonstrated the existence of circulating tumor cells (CTCs) in cancer patient's blood. Once identified, CTC biomarkers will be invaluable tools for clinical diagnosis, prognosis and treatment. In this review, we propose ex vivo culture as a rational strategy for large scale amplification of the limited numbers of CTCs from a patient sample, to derive enough CTCs for accurate and reproducible characterization of the biophysical, biochemical, gene expressional and behavioral properties of the harvested cells. Because of tumor cell heterogeneity, it is important to amplify all the CTCs in a blood sample for a comprehensive understanding of their role in cancer metastasis. By analyzing critical steps and technical issues in ex vivo CTC culture, we developed a cost-effective and reproducible protocol directly culturing whole peripheral blood mononuclear cells, relying on an assumed survival advantage in CTCs and CTC-like cells over the normal cells to amplify this specified cluster of cancer cells.
Collapse
Affiliation(s)
- Ruoxiang Wang
- Uro-Oncology Research, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Gina C Y Chu
- Uro-Oncology Research, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stefan Mrdenovic
- Uro-Oncology Research, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Alagappan A Annamalai
- Uro-Oncology Research, Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Andrew E Hendifar
- Uro-Oncology Research, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Nicholas N Nissen
- Uro-Oncology Research, Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - James S Tomlinson
- Department of Surgery, West Los Angeles VA Hospital, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, USA
| | - Michael Lewis
- Department of Pathology, West Los Angeles VA Hospital, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, USA
| | | | - Hsian-Rong Tseng
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, USA
| | - Edwin M Posadas
- Uro-Oncology Research, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Michael R Freeman
- Uro-Oncology Research, Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stephen J Pandol
- Uro-Oncology Research, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Haiyen E Zhau
- Uro-Oncology Research, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Leland W K Chung
- Uro-Oncology Research, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Uro-Oncology Research, Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
210
|
Tumor-associated macrophages in human breast cancer parenchyma negatively correlate with lymphatic metastasis after neoadjuvant chemotherapy. Immunobiology 2016; 222:101-109. [PMID: 27510849 DOI: 10.1016/j.imbio.2016.08.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 08/04/2016] [Indexed: 12/27/2022]
Abstract
Breast cancer is the leading cause of cancer death in women worldwide with high morbidity and mortality. Tumor-associated macrophages (TAM) are major innate immune cells in the tumor microenvironment controlling primary tumor growth and metastasis. Neoadjuvant chemotherapy (NACT) is a conventional pre-operative treatment for breast cancer. In the present study we examined the distribution of TAM in five distinct intratumoral morphological compartments of human breast cancer and their correlation with clinical parameters after NACT. Our data indicated that CD68+ but not stabilin-1+ TAM in areas with parenchymal elements negatively correlate with lymphatic metastasis after NACT. However, in cases where lymphatic metastases were detected (28 out of 50 analyzed samples) both amount of CD68+ and stabilin-1+ macrophages in the areas with coarse fibrous stroma directly correlated with the number of positive lymph nodes. In patients with complete response to the preoperative NACT the average score of CD68 expression in the areas with coarse fibrous stroma was lower compared with cases of a partial response and stable disease. We concluded that function of TAM after NACT depends on their intratumoral localization and local tumor microenvironment which plays an important role in polarization of macrophages towards tumor-suppressive or tumor-supportive types.
Collapse
|
211
|
Zhu W, Harvey S, Macura KJ, Euhus DM, Artemov D. Invasive Breast Cancer Preferably and Predominantly Occurs at the Interface Between Fibroglandular and Adipose Tissue. Clin Breast Cancer 2016; 17:e11-e18. [PMID: 27568102 DOI: 10.1016/j.clbc.2016.07.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Accepted: 07/20/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND Increasing evidence suggests adipocyte involvement in malignant breast tumor invasive front or margin. The aim of this study was to evaluate the location of invasive breast tumors in relation to fibroglandular and adipose tissue by dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI). PATIENTS AND METHODS Pretreatment breast DCE-MRI images of 294 patients with biopsy-proven invasive breast cancer from 2008 to 2014 were studied. Invasive breast tumors were visualized as enhanced lesions in the postcontrast subtraction images. Positive identification of biopsy-confirmed invasive breast tumors on DCE-MRI images was achieved by correlation of findings from breast MRI and pathology reports. Tumor location in relation to fibroglandular and adipose tissue was investigated using precontrast T1-weighted MRI images. RESULTS Of 294 patients, 291 had DCE-MRI discernable invasive breast tumors located at the interface between fibroglandular and adipose tissues, regardless of the tumor size, type, receptor status, or breast composition. CONCLUSION Invasive breast cancer preferably and predominantly occurs adjacent to breast adipose tissue.
Collapse
Affiliation(s)
- Wenlian Zhu
- Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD.
| | - Susan Harvey
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Katarzyna J Macura
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - David M Euhus
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Dmitri Artemov
- Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
212
|
Vallerand D, Massonnet G, Kébir F, Gentien D, Maciorowski Z, De la Grange P, Sigal-Zafrani B, Richardson M, Humbert S, Thuleau A, Assayag F, de Plater L, Nicolas A, Scholl S, Marangoni E, Weigand S, Roman-Roman S, Savina A, Decaudin D. Characterization of Breast Cancer Preclinical Models Reveals a Specific Pattern of Macrophage Polarization. PLoS One 2016; 11:e0157670. [PMID: 27388901 PMCID: PMC4936680 DOI: 10.1371/journal.pone.0157670] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 06/02/2016] [Indexed: 12/23/2022] Open
Abstract
Drug discovery efforts have focused on the tumor microenvironment in recent years. However, few studies have characterized the stroma component in patient-derived xenografts (PDXs) and genetically engineered mouse models (GEMs). In this study, we characterized the stroma in various models of breast cancer tumors in mice. We performed transcriptomic and flow cytometry analyses on murine populations for a series of 25 PDXs and the two most commonly used GEMs (MMTV-PyMT and MMTV-erBb2). We sorted macrophages from five models. We then profiled gene expression in these cells, which were also subjected to flow cytometry for phenotypic characterization. Hematopoietic cell composition, mostly macrophages and granulocytes, differed between tumors. Macrophages had a specific polarization phenotype related to their M1/M2 classification and associated with the expression of genes involved in the recruitment, invasion and metastasis processes. The heterogeneity of the stroma component of the models studied suggests that tumor cells modify their microenvironment to satisfy their needs. Our observations suggest that such models are of relevance for preclinical studies.
Collapse
Affiliation(s)
- David Vallerand
- Translational Research Department, Laboratory of Preclinical Investigation, Institut Curie, PSL University, Paris, France
- Institut Roche, Boulogne-Billancourt, France
| | - Gérald Massonnet
- Translational Research Department, Laboratory of Preclinical Investigation, Institut Curie, PSL University, Paris, France
| | - Fatima Kébir
- Department of Pathology, Institut Curie, Paris, France
| | - David Gentien
- Platform of Molecular Biology Facilities, Institut Curie, PSL University, Paris, France
| | - Zofia Maciorowski
- Flow Cytometry Core Facility, Institut Curie, PSL University, Paris, France
| | | | - Brigitte Sigal-Zafrani
- Department of Pathology, Institut Curie, Paris, France
- Inserm, U830, Institut Curie, PSL University, Paris, France
| | | | - Sandrine Humbert
- CNRS UMR3306, INSERM U1005, Institut Curie, PSL University, Orsay, France
| | - Aurélie Thuleau
- Translational Research Department, Laboratory of Preclinical Investigation, Institut Curie, PSL University, Paris, France
| | - Franck Assayag
- Translational Research Department, Laboratory of Preclinical Investigation, Institut Curie, PSL University, Paris, France
| | - Ludmilla de Plater
- Translational Research Department, Laboratory of Preclinical Investigation, Institut Curie, PSL University, Paris, France
| | - André Nicolas
- Department of Pathology, Institut Curie, Paris, France
| | - Suzy Scholl
- Department of Medical Oncology, Institut Curie, Institut Curie, Paris, France
| | - Elisabetta Marangoni
- Translational Research Department, Laboratory of Preclinical Investigation, Institut Curie, PSL University, Paris, France
| | | | - Sergio Roman-Roman
- Translational Research Department, Institut Curie, PSL University, Paris, France
| | | | - Didier Decaudin
- Translational Research Department, Laboratory of Preclinical Investigation, Institut Curie, PSL University, Paris, France
- Department of Medical Oncology, Institut Curie, Institut Curie, Paris, France
- Translational Research Department, Institut Curie, PSL University, Paris, France
- * E-mail:
| |
Collapse
|
213
|
Brandan ME, Cruz-Bastida JP, Rosado-Méndez IM, Villaseñor-Navarro Y, Pérez-Ponce H, Galván HA, Trujillo-Zamudio FE, Sánchez-Suárez P, Benítez-Bribiesca L. Clinical study of contrast-enhanced digital mammography and the evaluation of blood and lymphatic microvessel density. Br J Radiol 2016; 89:20160232. [PMID: 27376457 DOI: 10.1259/bjr.20160232] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE To correlate image parameters in contrast-enhanced digital mammography (CEDM) with blood and lymphatic microvessel density (MVD). METHODS 18 Breast Imaging-Reporting and Data System (BI-RADS)-4 to BI-RADS-5 patients were subjected to CEDM. Craniocaudal views were acquired, two views (low and high energy) before iodine contrast medium (CM) injection and four views (high energy) 1-5 min afterwards. Processing included registration and two subtraction modalities, traditional single-energy temporal (high-energy) and "dual-energy temporal with a matrix", proposed to improve lesion conspicuity. Images were calibrated into iodine thickness, and iodine uptake, contrast, time-intensity and time-contrast kinetic curves were quantified. Image indicators were compared with MVD evaluated by anti-CD105 and anti-podoplanin (D2-40) immunohistochemistry. RESULTS 11 lesions were cancerous and 7 were benign. CEDM subtraction strongly increased conspicuity of lesions enhanced by iodine uptake. A strong correlation was observed between lymphatic vessels and blood vessels; all benign lesions had <30 blood microvessels per field, and all cancers had more than this value. MVD showed no correlation with iodine uptake, nor with contrast. The most frequent curve was early uptake followed by plateau for uptake and contrast in benign and malignant lesions. The positive-predictive value of uptake dynamics was 73% and that of contrast was 64%. CONCLUSION CEDM increased lesion visibility and showed additional features compared with conventional mammography. Lack of correlation between image parameters and MVD is probably due to tumour tissue heterogeneity, mammography projective nature and/or dependence of extracellular iodine irrigation on tissue composition. ADVANCES IN KNOWLEDGE Quantitative analysis of CEDM images was performed. Image parameters and MVD showed no correlation. Probably, this is indication of the complex dependence of CM perfusion on tumour microenvironment.
Collapse
Affiliation(s)
- María-Ester Brandan
- 1 Instituto de Física, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Juan P Cruz-Bastida
- 1 Instituto de Física, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Iván M Rosado-Méndez
- 1 Instituto de Física, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | - Héctor Pérez-Ponce
- 1 Instituto de Física, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | | | | | | |
Collapse
|
214
|
Abstract
Communication between the diverse assortment of cells that constitute the tumor microenvironment plays an important role in tumor development. Using a p53-null mouse model, Zhang and colleagues describe a novel feedback loop involving breast cancer stem cells and their progeny mediated by WNT2, CXCL12, and IL6.
Collapse
Affiliation(s)
- Michael D Brooks
- Comprehensive Cancer Center, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Max S Wicha
- Comprehensive Cancer Center, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
215
|
Yang J, Qian S, Cai X, Lu W, Hu C, Sun X, Yang Y, Yu Q, Gao SP, Cao P. Chikusetsusaponin IVa Butyl Ester (CS-IVa-Be), a Novel IL6R Antagonist, Inhibits IL6/STAT3 Signaling Pathway and Induces Cancer Cell Apoptosis. Mol Cancer Ther 2016; 15:1190-200. [PMID: 26929249 DOI: 10.1158/1535-7163.mct-15-0551] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 02/24/2016] [Indexed: 01/10/2023]
Abstract
The activation of IL6/STAT3 signaling is associated with the pathogenesis of many cancers. Agents that suppress IL6/STAT3 signaling have cancer-therapeutic potential. In this study, we found that chikusetsusaponin IVa butyl ester (CS-IVa-Be), a triterpenoid saponin extracted from Acanthopanas gracilistylus W.W.Smith, induced cancer cell apoptosis. CS-IVa-Be inhibited constitutive and IL6-induced STAT3 activation, repressed STAT3 DNA-binding activity, STAT3 nuclear translocation, IL6-induced STAT3 luciferase reporter activity, IL6-induced STAT3-regulated antiapoptosis gene expression in MDA-MB-231 cells, and IL6-induced TF-1 cell proliferation. Surprisingly, CS-IVa-Be inhibited IL6 family cytokines rather than other cytokines induced STAT3 activation. Further studies indicated that CS-IVa-Be is an antagonist of IL6 receptor via directly binding to the IL6Rα with a Kd of 663 ± 74 nmol/L and the GP130 (IL6Rβ) with a Kd of 1,660 ± 243 nmol/L, interfering with the binding of IL6 to IL6R (IL6Rα and GP130) in vitro and in cancer cells. The inhibitory effect of CS-IVa-Be on the IL6-IL6Rα-GP130 interaction was relatively specific as CS-IVa-Be showed higher affinity to IL6Rα than to LIFR (Kd: 4,910 ± 1,240 nmol/L) and LeptinR (Kd: 4,990 ± 915 nmol/L). We next demonstrated that CS-IVa-Be not only directly induced cancer cell apoptosis but also sensitized MDA-MB-231 cells to TRAIL-induced apoptosis via upregulating DR5. Our findings suggest that CS-IVa-Be as a novel IL6R antagonist inhibits IL6/STAT3 signaling pathway and sensitizes the MDA-MB-231 cells to TRAIL-induced cell death. Mol Cancer Ther; 15(6); 1190-200. ©2016 AACR.
Collapse
Affiliation(s)
- Jie Yang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Shihui Qian
- Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Xueting Cai
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Wuguang Lu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Chunping Hu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Xiaoyan Sun
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Yang Yang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Qiang Yu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - S Paul Gao
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Peng Cao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China.
| |
Collapse
|
216
|
Lo PK, Wolfson B, Zhou Q. Cancer stem cells and early stage basal-like breast cancer. World J Obstet Gynecol 2016; 5:150-161. [PMID: 28239564 PMCID: PMC5321620 DOI: 10.5317/wjog.v5.i2.150] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 10/21/2015] [Accepted: 01/22/2016] [Indexed: 02/05/2023] Open
Abstract
Ductal carcinoma in situ (DCIS) is a category of early stage, non-invasive breast tumor defined by the intraductal proliferation of malignant breast epithelial cells. DCIS is a heterogeneous disease composed of multiple molecular subtypes including luminal, HER2 and basal-like types, which are characterized by immunohistochemical analyses and gene expression profiling. Following surgical and radiation therapies, patients with luminal-type, estrogen receptor-positive DCIS breast tumors can benefit from adjuvant endocrine-based treatment. However, there are no available targeted therapies for patients with basal-like DCIS (BL-DCIS) tumors due to their frequent lack of endocrine receptors and HER2 amplification, rendering them potentially susceptible to recurrence. Moreover, multiple lines of evidence suggest that DCIS is a non-obligate precursor of invasive breast carcinoma. This raises the possibility that targeting precursor BL-DCIS is a promising strategy to prevent BL-DCIS patients from the development of invasive basal-like breast cancer. An accumulating body of evidence demonstrates the existence of cancer stem-like cells (CSCs) in BL-DCIS, which potentially determine the features of BL-DCIS and their ability to progress into invasive cancer. This review encompasses the current knowledge in regard to the characteristics of BL-DCIS, identification of CSCs, and their biological properties in BL-DCIS. We summarize recently discovered relevant molecular signaling alterations that promote the generation of CSCs in BL-DCIS and the progression of BL-DCIS to invasive breast cancer, as well as the influence of the tissue microenvironment on CSCs and the invasive transition. Finally, we discuss the translational implications of these findings for the prognosis and prevention of BL-DCIS relapse and progression.
Collapse
|
217
|
Goliwas KF, Miller LM, Marshall LE, Berry JL, Frost AR. Preparation and Analysis of In Vitro Three Dimensional Breast Carcinoma Surrogates. J Vis Exp 2016. [PMID: 27214165 DOI: 10.3791/54004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Three dimensional (3D) culture is a more physiologically relevant method to model cell behavior in vitro than two dimensional culture. Carcinomas, including breast carcinomas, are complex 3D tissues composed of cancer epithelial cells and stromal components, including fibroblasts and extracellular matrix (ECM). Yet most in vitro models of breast carcinoma consist only of cancer epithelial cells, omitting the stroma and, therefore, the 3D architecture of a tumor in vivo. Appropriate 3D modeling of carcinoma is important for accurate understanding of tumor biology, behavior, and response to therapy. However, the duration of culture and volume of 3D models is limited by the availability of oxygen and nutrients within the culture. Herein, we demonstrate a method in which breast carcinoma epithelial cells and stromal fibroblasts are incorporated into ECM to generate a 3D breast cancer surrogate that includes stroma and can be cultured as a solid 3D structure or by using a perfusion bioreactor system to deliver oxygen and nutrients. Following setup and an initial growth period, surrogates can be used for preclinical drug testing. Alternatively, the cellular and matrix components of the surrogate can be modified to address a variety of biological questions. After culture, surrogates are fixed and processed to paraffin, in a manner similar to the handling of clinical breast carcinoma specimens, for evaluation of parameters of interest. The evaluation of one such parameter, the density of cells present, is explained, where ImageJ and CellProfiler image analysis software systems are applied to photomicrographs of histologic sections of surrogates to quantify the number of nucleated cells per area. This can be used as an indicator of the change in cell number over time or the change in cell number resulting from varying growth conditions and treatments.
Collapse
Affiliation(s)
- Kayla F Goliwas
- Department of Pathology, University of Alabama at Birmingham
| | - Lindsay M Miller
- Department of Biomedical Engineering, University of Alabama at Birmingham
| | - Lauren E Marshall
- Department of Biomedical Engineering, University of Alabama at Birmingham
| | - Joel L Berry
- Department of Biomedical Engineering, University of Alabama at Birmingham
| | - Andra R Frost
- Department of Pathology, University of Alabama at Birmingham;
| |
Collapse
|
218
|
Abstract
Metastasis is the underlying cause of death for the majority of breast cancer patients. Despite significant advances in recent years in basic research and clinical development, therapies that specifically target metastatic breast cancer remain inadequate, and represents the single greatest obstacle to reducing mortality of late-stage breast cancer. Recent efforts have leveraged genomic analysis of breast cancer and molecular dissection of tumor-stromal cross-talk to uncover a number of promising candidates for targeted treatment of metastatic breast cancer. Rational combinations of therapeutic agents targeting tumor-intrinsic properties and microenvironmental components provide a promising strategy to develop precision treatments with higher specificity and less toxicity. In this review, we discuss the emerging therapeutic targets in breast cancer metastasis, from tumor-intrinsic pathways to those that involve the host tissue components, including the immune system.
Collapse
Affiliation(s)
- Zhuo Li
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, United States
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, United States.
| |
Collapse
|
219
|
Wu X, Chen G, Qiu J, Lu J, Zhu W, Chen J, Zhuo S, Yan J. Visualization of basement membranes in normal breast and breast cancer tissues using multiphoton microscopy. Oncol Lett 2016; 11:3785-3789. [PMID: 27313695 DOI: 10.3892/ol.2016.4472] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 03/18/2016] [Indexed: 12/14/2022] Open
Abstract
Since basement membranes represent a critical barrier during breast cancer progression, timely imaging of these signposts is essential for early diagnosis of breast cancer. A label-free method using multiphoton microscopy (MPM) based on two-photon excited fluorescence signals and second harmonic generation signals for analyzing the morphology of basement membrane in normal and cancerous breast tissues is likely to enable a better understanding of the pathophysiology of breast cancer and facilitate improved clinical management and treatment of this disease. The aim of this study was to determine whether MPM has the potential for label-free assessment of the morphology of basement membrane in normal and cancerous breast tissues. A total of 60 tissue section samples (comprising 30 fresh breast cancer specimens and 30 normal breast tissues) were first imaged (fresh, unfixed and unstained) with MPM and are then processed for routine hematoxylin and eosin (H&E) histopathology. Comparisons were made between MPM imaging and gold standard sections for each specimen stained with H&E. Simply by visualizing morphological features appearing on multiphoton images, cancerous lesions may be readily identified by the loss of basement membrane and tumor cells characterized by irregular size and shape, enlarged nuclei and increased nuclear-cytoplasmic ratio. These results suggest that MPM has potential as a label-free method of imaging the morphology of basement membranes and cell features to effectively distinguish between normal and cancerous breast tissues.
Collapse
Affiliation(s)
- Xiufeng Wu
- Department of Surgery, Fujian Provincial Tumor Hospital, Teaching Hospital of Fujian Medical University, Fuzhou, Fujian 350014, P.R. China; Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Gang Chen
- Department of Pathology, Fujian Provincial Tumor Hospital, Teaching Hospital of Fujian Medical University, Fuzhou, Fujian 350014, P.R. China
| | - Jingting Qiu
- Institute of Laser and Optoelectronics Technology, Fujian Provincial Key Laboratory for Photonics Technology, Key Laboratory of Optoelectronic Science and Technology for Medicine of the Ministry of Education, Fujian Normal University, Fuzhou, Fujian 350007, P.R. China
| | - Jianping Lu
- Department of Pathology, Fujian Provincial Tumor Hospital, Teaching Hospital of Fujian Medical University, Fuzhou, Fujian 350014, P.R. China
| | - Weifeng Zhu
- Department of Pathology, Fujian Provincial Tumor Hospital, Teaching Hospital of Fujian Medical University, Fuzhou, Fujian 350014, P.R. China
| | - Jianxin Chen
- Institute of Laser and Optoelectronics Technology, Fujian Provincial Key Laboratory for Photonics Technology, Key Laboratory of Optoelectronic Science and Technology for Medicine of the Ministry of Education, Fujian Normal University, Fuzhou, Fujian 350007, P.R. China
| | - Shuangmu Zhuo
- Institute of Laser and Optoelectronics Technology, Fujian Provincial Key Laboratory for Photonics Technology, Key Laboratory of Optoelectronic Science and Technology for Medicine of the Ministry of Education, Fujian Normal University, Fuzhou, Fujian 350007, P.R. China
| | - Jun Yan
- Department of Surgery, Fujian Provincial Tumor Hospital, Teaching Hospital of Fujian Medical University, Fuzhou, Fujian 350014, P.R. China; Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
220
|
Osteocytic connexin hemichannels suppress breast cancer growth and bone metastasis. Oncogene 2016; 35:5597-5607. [PMID: 27041582 PMCID: PMC5050050 DOI: 10.1038/onc.2016.101] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 01/13/2016] [Accepted: 01/25/2016] [Indexed: 12/15/2022]
Abstract
Although the skeleton is one of predominant sites for breast cancer metastasis, why breast cancer cells often become dormant after homing to bone is not well understood. Here, we reported an intrinsic self-defense mechanism of bone cells against breast cancer cells: a critical role of connexin (Cx) 43 hemichannels in osteocytes in the suppression of breast cancer bone metastasis. Cx43 hemichannels allow passage of small molecules between the intracellular and extracellular environments. The treatment of bisphosphonate drugs, either alendronate (ALN) or zoledronic acid (ZOL), opened Cx43 hemichannels in osteocytes. Conditioned media (CM) collected from MLO-Y4 osteocyte cells treated with bisphosphonates inhibited the anchorage-independent growth, migration and invasion of MDA-MB-231 human breast cancer cells and Py8119 mouse mammary carcinoma cells and this inhibitory effect was attenuated with Cx43(E2), a specific hemichannel blocking antibody. The opening of osteocytic Cx43 hemichannels by mechanical stimulation had similar inhibitory effects on breast cancer cells and this inhibition was attenuated by Cx43(E2) antibody as well. These inhibitory effects on cancer cells were mediated by ATP released from osteocyte Cx43 hemichannels. Furthermore, both Cx43 osteocyte-specific knockout mice and osteocyte-specific Δ130–136 transgenic mice with impaired Cx43 gap junctions and hemichannels showed significantly increased tumor growth and attenuated the inhibitory effect of ZOL. However, R76W transgenic mice with functional hemichannels but not gap junctions in osteocytes did not display a significant difference. Together, our studies establish the specific inhibitory role of osteocytic Cx43 hemichannels, and exploiting the activity of this channel could serve as a de novo therapeutic strategy.
Collapse
|
221
|
Lian C, Ruan L, Shang D, Wu Y, Lu Y, Lü P, Yang Y, Wei Y, Dong X, Ren D, Chen K, Liu H, Tu Z. Heparin-Binding Epidermal Growth Factor-Like Growth Factor as a Potent Target for Breast Cancer Therapy. Cancer Biother Radiopharm 2016; 31:85-90. [DOI: 10.1089/cbr.2015.1956] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Caixia Lian
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Lingling Ruan
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Dongsheng Shang
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Yanfang Wu
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Yongjin Lu
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Peng Lü
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Yuhua Yang
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Yajun Wei
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Xiaojing Dong
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Dewan Ren
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Keping Chen
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Hanqing Liu
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Zhigang Tu
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| |
Collapse
|
222
|
Consolino L, Longo DL, Dastrù W, Cutrin JC, Dettori D, Lanzardo S, Oliviero S, Cavallo F, Aime S. Functional imaging of the angiogenic switch in a transgenic mouse model of human breast cancer by dynamic contrast enhanced magnetic resonance imaging. Int J Cancer 2016; 139:404-13. [PMID: 26941084 DOI: 10.1002/ijc.30073] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 02/24/2016] [Indexed: 01/16/2023]
Abstract
Tumour progression depends on several sequential events that include the microenvironment remodelling processes and the switch to the angiogenic phenotype, leading to new blood vessels recruitment. Non-invasive imaging techniques allow the monitoring of functional alterations in tumour vascularity and cellularity. The aim of this work was to detect functional changes in vascularisation and cellularity through Dynamic Contrast Enhanced (DCE) and Diffusion Weighted (DW) Magnetic Resonance Imaging (MRI) modalities during breast cancer initiation and progression of a transgenic mouse model (BALB-neuT mice). Histological examination showed that BALB-neuT mammary glands undergo a slow neoplastic progression from simple hyperplasia to invasive carcinoma, still preserving normal parts of mammary glands. DCE-MRI results highlighted marked functional changes in terms of vessel permeability (K(trans) , volume transfer constant) and vascularisation (vp , vascular volume fraction) in BALB-neuT hyperplastic mammary glands if compared to BALB/c ones. When breast tissue progressed from simple to atypical hyperplasia, a strong increase in DCE-MRI biomarkers was observed in BALB-neuT in comparison to BALB/c mice (K(trans) = 5.3 ± 0.7E-4 and 3.1 ± 0.5E-4; vp = 7.4 ± 0.8E-2 and 4.7 ± 0.6E-2 for BALB-neuT and BALB/c, respectively) that remained constant during the successive steps of the neoplastic transformation. Consistent with DCE-MRI observations, microvessel counting revealed a significant increase in tumour vessels. Our study showed that DCE-MRI estimates can accurately detect the angiogenic switch at early step of breast cancer carcinogenesis. These results support the view that this imaging approach is an excellent tool to characterize microvasculature changes, despite only small portions of the mammary glands developed neoplastic lesions in a transgenic mouse model.
Collapse
Affiliation(s)
- Lorena Consolino
- Department of Molecular Biotechnology and Health Sciences, University of Torino, via Nizza 52, Torino, 10126, Italy
| | - Dario Livio Longo
- (CNR) c/o Molecular Biotechnologies Center, Istituto di Biostrutture e Bioimmagini, via Nizza 52, Torino, 10126, Italy.,Molecular Imaging Center, University of Torino, via Nizza 52, Torino, 10126, Italy
| | - Walter Dastrù
- Department of Molecular Biotechnology and Health Sciences, University of Torino, via Nizza 52, Torino, 10126, Italy.,Molecular Imaging Center, University of Torino, via Nizza 52, Torino, 10126, Italy
| | - Juan Carlos Cutrin
- Department of Molecular Biotechnology and Health Sciences, University of Torino, via Nizza 52, Torino, 10126, Italy
| | - Daniela Dettori
- Human Genetics Foundation (HuGeF), via Nizza 52, Torino, 10126, Italy
| | - Stefania Lanzardo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, via Nizza 52, Torino, 10126, Italy
| | | | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, via Nizza 52, Torino, 10126, Italy
| | - Silvio Aime
- Department of Molecular Biotechnology and Health Sciences, University of Torino, via Nizza 52, Torino, 10126, Italy.,Molecular Imaging Center, University of Torino, via Nizza 52, Torino, 10126, Italy
| |
Collapse
|
223
|
Tong H, Ke JQ, Jiang FZ, Wang XJ, Wang FY, Li YR, Lu W, Wan XP. Tumor-associated macrophage-derived CXCL8 could induce ERα suppression via HOXB13 in endometrial cancer. Cancer Lett 2016; 376:127-36. [PMID: 27018308 DOI: 10.1016/j.canlet.2016.03.036] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 03/21/2016] [Accepted: 03/21/2016] [Indexed: 01/05/2023]
Abstract
PURPOSE To elucidate the role of tumor-associated macrophage (TAM) in the loss of ERα in endometrial cancer (EC) and the underlying mechanism. MATERIALS AND METHODS Tissue microarrays and immunohistochemistry assays were performed using endometrial cancer tissue along with coculture, immunofluorescence, invasion assays and ChIP-qPCR using a human endometrial cancer cell line. RESULTS Compared with normal tissue, an increased number of TAM was found in EC tissue (34.0 ± 2.6 vs. 8.3 ± 1.1, respectively; p < 0.001), which may downregulate ERα (27.4%, p < 0.05 for HEC-1A and 16.9%, p < 0.05 for Ishikawa) and promote EC cell invasion (1.8-fold, p < 0.001 for HEC-1A and 2.0-fold, p < 0.001 for Ishikawa). Furthermore, we found that TAM-derived CXCL8 mediated the loss of ERα and cancer invasion via HOXB13. HOXB13 was highly expressed in the ERα-negative subtype (r = -0.204, p = 0.002) and low expression of ESR1 was associated with a poor prognosis for EC patients (log-rank p < 0.05). CONCLUSION TAM-secreted CXCL8 downregulated the ERα expression of EC cells via HOXB13, which may be associated with cancer invasion, metastasis and poor prognosis.
Collapse
Affiliation(s)
- Huan Tong
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie-Qi Ke
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fei-Zhou Jiang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Jun Wang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fang-Yuan Wang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi-Ran Li
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen Lu
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiao-Ping Wan
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
224
|
Marcus H, Attar-Schneider O, Dabbah M, Zismanov V, Tartakover-Matalon S, Lishner M, Drucker L. Mesenchymal stem cells secretomes' affect multiple myeloma translation initiation. Cell Signal 2016; 28:620-30. [PMID: 26976208 DOI: 10.1016/j.cellsig.2016.03.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/01/2016] [Accepted: 03/08/2016] [Indexed: 12/29/2022]
Abstract
Bone marrow mesenchymal stem cells' (BM-MSCs) role in multiple myeloma (MM) pathogenesis is recognized. Recently, we have published that co-culture of MM cell lines with BM-MSCs results in mutual modulation of phenotype and proteome (via translation initiation (TI) factors eIF4E/eIF4GI) and that there are differences between normal donor BM-MSCs (ND-MSCs) and MM BM-MSCs (MM-MSCs) in this crosstalk. Here, we aimed to assess the involvement of soluble BM-MSCs' (ND, MM) components, more easily targeted, in manipulation of MM cell lines phenotype and TI with specific focus on microvesicles (MVs) capable of transferring critical biological material. We applied ND and MM-MSCs 72h secretomes to MM cell lines (U266 and ARP-1) for 12-72h and then assayed the cells' (viability, cell count, cell death, proliferation, cell cycle, autophagy) and TI (factors: eIF4E, teIF4GI; regulators: mTOR, MNK1/2, 4EBP; targets: cyclin D1, NFκB, SMAD5, cMyc, HIF1α). Furthermore, we dissected the secretome into >100kDa and <100kDa fractions and repeated the experiments. Finally, MVs were isolated from the ND and MM-MSCs secretomes and applied to MM cell lines. Phenotype and TI were assessed. Secretomes of BM-MSCs (ND, MM) significantly stimulated MM cell lines' TI, autophagy and proliferation. The dissected secretome yielded different effects on MM cell lines phenotype and TI according to fraction (>100kDa- repressed; <100kDa- stimulated) but with no association to source (ND, MM). Finally, in analyses of MVs extracted from BM-MSCs (ND, MM) we witnessed differences in accordance with source: ND-MSCs MVs inhibited proliferation, autophagy and TI whereas MM-MSCs MVs stimulated them. These observations highlight the very complex communication between MM and BM-MSCs and underscore its significance to major processes in the malignant cells. Studies into the influential MVs cargo are underway and expected to uncover targetable signals in the regulation of the TI/proliferation/autophagy cascade.
Collapse
Affiliation(s)
- H Marcus
- Oncogenetic Laboratory, Tel Aviv University, Tel Aviv, Israel; Sackler faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - O Attar-Schneider
- Oncogenetic Laboratory, Tel Aviv University, Tel Aviv, Israel; Sackler faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - M Dabbah
- Oncogenetic Laboratory, Tel Aviv University, Tel Aviv, Israel; Sackler faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - V Zismanov
- Oncogenetic Laboratory, Tel Aviv University, Tel Aviv, Israel; Sackler faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - S Tartakover-Matalon
- Oncogenetic Laboratory, Tel Aviv University, Tel Aviv, Israel; Sackler faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - M Lishner
- Oncogenetic Laboratory, Tel Aviv University, Tel Aviv, Israel; Internal Medicine Department, Meir Medical Center, Kfar Saba, Tel Aviv University, Tel Aviv, Israel; Sackler faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - L Drucker
- Oncogenetic Laboratory, Tel Aviv University, Tel Aviv, Israel; Sackler faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
225
|
Ko P, Kim D, You E, Jung J, Oh S, Kim J, Lee KH, Rhee S. Extracellular Matrix Rigidity-dependent Sphingosine-1-phosphate Secretion Regulates Metastatic Cancer Cell Invasion and Adhesion. Sci Rep 2016; 6:21564. [PMID: 26877098 PMCID: PMC4753492 DOI: 10.1038/srep21564] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 01/27/2016] [Indexed: 12/14/2022] Open
Abstract
Dynamic interaction between cancer cells and the surrounding microenvironment is critical for cancer progression via changes in cellular behavior including alteration of secreted molecules. However, the molecular mechanisms underlying the influence exerted by the cancer microenvironment on secretion of molecules during cancer progression remain largely unknown. In this study, we report that secretion of spingsine-1-phosphate (S1P) and its regulator, SphK1 expression is dependent of the substrate rigidity, which is critical for the balance between cancer cell invasion and adhesion. Conditioned media (CM) of MDA-MB-231, an aggressive breast cancer cell obtained from soft substrate (~0.5 kPa) induced chemo-attractive invasion, while CM obtained from stiff substrate (~2.5 kPa) increased cell adhesion instead. We found that the expression of SphK1 is upregulated in the stiff substrate, resulting in an increase in S1P levels in the CM. We also found that upregulation of SphK1 expression in the stiff substrate is dominant in metastatic cancer cells but not in primary cancer cells. These results suggest that alterations in the mechanical environment of the ECM surrounding the tumor cells actively regulate cellular properties such as secretion, which in turn, may contribute to cancer progression.
Collapse
Affiliation(s)
- Panseon Ko
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Daehwan Kim
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Eunae You
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jangho Jung
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Somi Oh
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jaehyun Kim
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Kwang-Ho Lee
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Sangmyung Rhee
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| |
Collapse
|
226
|
Mathematical modelling of phenotypic plasticity and conversion to a stem-cell state under hypoxia. Sci Rep 2016; 6:18074. [PMID: 26838463 PMCID: PMC4738268 DOI: 10.1038/srep18074] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 11/04/2015] [Indexed: 12/11/2022] Open
Abstract
Hypoxia, or oxygen deficiency, is known to be associated with breast tumour progression, resistance to conventional therapies and poor clinical prognosis. The epithelial-mesenchymal transition (EMT) is a process that confers invasive and migratory capabilities as well as stem cell properties to carcinoma cells thus promoting metastatic progression. In this work, we examined the impact of hypoxia on EMT-associated cancer stem cell (CSC) properties, by culturing transformed human mammary epithelial cells under normoxic and hypoxic conditions, and applying in silico mathematical modelling to simulate the impact of hypoxia on the acquisition of CSC attributes and the transitions between differentiated and stem-like states. Our results indicate that both the heterogeneity and the plasticity of the transformed cell population are enhanced by exposure to hypoxia, resulting in a shift towards a more stem-like population with increased EMT features. Our findings are further reinforced by gene expression analyses demonstrating the upregulation of EMT-related genes, as well as genes associated with therapy resistance, in hypoxic cells compared to normoxic counterparts. In conclusion, we demonstrate that mathematical modelling can be used to simulate the role of hypoxia as a key contributor to the plasticity and heterogeneity of transformed human mammary epithelial cells.
Collapse
|
227
|
Hima S, Sreeja S. Modulatory role of 17β-estradiol in the tumor microenvironment of thyroid cancer. IUBMB Life 2015; 68:85-96. [DOI: 10.1002/iub.1462] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 11/24/2015] [Indexed: 12/13/2022]
Affiliation(s)
- Sithul Hima
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology; Thycaud Thiruvananthapuram Kerala India
| | - Sreeharshan Sreeja
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology; Thycaud Thiruvananthapuram Kerala India
| |
Collapse
|
228
|
Goswami S, Sharma-Walia N. Osteoprotegerin secreted by inflammatory and invasive breast cancer cells induces aneuploidy, cell proliferation and angiogenesis. BMC Cancer 2015; 15:935. [PMID: 26608463 PMCID: PMC4660791 DOI: 10.1186/s12885-015-1837-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2015] [Indexed: 12/12/2022] Open
Abstract
Background Osteoprotegerin (OPG) is a glycoprotein that has multifaceted role and is associated with several cancer malignancies like that of bladder carcinoma, gastric carcinoma, prostate cancer, multiple myeloma and breast cancer. Also OPG has been associated with several organ pathologies. The widespread expression of OPG suggests that OPG may have multiple biological activities that are yet to be explored. Methods The anchorage-independent sphere cultures of the adherent cells were instrumental in our study as it provided a deeper insight into the complexity of a 3D tumor. Cytokine profiling was performed for OPG’s detection in the microenvironment. ELISA and western blotting were performed to quantify the OPG secretion and measure the protein levels respectively. OPG expression was detected in human breast cancer tissue samples by IHC. To decipher OPG’s role in tumor aggressiveness both recombinant human OPG as well as OPG rich and depleted breast cancer cell conditioned media were tested. Western blotting and MTT assay were performed to detect changes in signaling pathways and proliferation that were induced in presence of OPG. Onset of aneuploidy, in presence of OPG, was measured by cell cycle analysis and western blotting. Finally, human Breast Cancer qBiomarker Copy Number PCR Array was used to detect how OPG remarkably induced gene copy numbers for oncogenic pathway regulators. Results SUM149PT and SUM1315M02 cells secrete high levels of the cytokine OPG compared to primary human mammary epithelial cells (HMEC). High expression of OPG was also detected in human breast cancer tissue samples compared to the uninvolved tissue from the same patient. OPG induced proliferation of control HMEC spheres and triggered the onset of aneuploidy in HMEC sphere cultures. OPG induced the expression of aneuploidy related kinases Aurora-A Kinase (IAK-1), Bub1 and BubR1 probably through the receptor activator of nuclear factor kappa-B ligand (RANKL) and syndecan-1 receptors via the Erk, AKT and GSK3(3 signaling pathway. Gene copy numbers for oncogenic pathway regulators such AKT1, Aurora-A Kinase (AURKA or IAK-1), epidermal growth factor receptor (EGFR) and MYC with a reduction in the copy numbers of cyclin dependent kinase inhibitor 2A (CDKN2A), PTEN and DNA topoisomerase 2 alpha (TOP2A) were induced in presence of OPG. Conclusions These results highlight the role of OPG in reprogramming normal mammary epithelial cells to a tumorigenic state and suggest promising avenues for treating inflammatory breast cancer as well as highly invasive breast cancer with new therapeutic targets. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1837-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sudeshna Goswami
- Department of Microbiology and Immunology, H. M. Bligh Cancer Research Laboratories, Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA.
| | - Neelam Sharma-Walia
- Department of Microbiology and Immunology, H. M. Bligh Cancer Research Laboratories, Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA.
| |
Collapse
|
229
|
Ward EM, DeSantis CE, Lin CC, Kramer JL, Jemal A, Kohler B, Brawley OW, Gansler T. Cancer statistics: Breast cancer in situ. CA Cancer J Clin 2015; 65:481-95. [PMID: 26431342 DOI: 10.3322/caac.21321] [Citation(s) in RCA: 177] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 08/06/2015] [Accepted: 09/02/2015] [Indexed: 01/11/2023] Open
Abstract
An estimated 60,290 new cases of breast carcinoma in situ are expected to be diagnosed in 2015, and approximately 1 in 33 women is likely to receive an in situ breast cancer diagnosis in her lifetime. Although in situ breast cancers are relatively common, their clinical significance and optimal treatment are topics of uncertainty and concern for both patients and clinicians. In this article, the American Cancer Society provides information about occurrence and treatment patterns for the 2 major subtypes of in situ breast cancer in the United States-ductal carcinoma in situ and lobular carcinoma in situ-using data from the North American Association of Central Cancer Registries and the 13 oldest Surveillance, Epidemiology, and End Results registries. The authors also present an overview of in situ breast cancer detection, treatment, risk factors, and prevention and discuss research needs and initiatives.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Breast Neoplasms/epidemiology
- Breast Neoplasms/pathology
- Breast Neoplasms/surgery
- Carcinoma in Situ/epidemiology
- Carcinoma in Situ/pathology
- Carcinoma in Situ/surgery
- Carcinoma, Intraductal, Noninfiltrating/epidemiology
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Carcinoma, Intraductal, Noninfiltrating/surgery
- Carcinoma, Lobular/epidemiology
- Carcinoma, Lobular/pathology
- Carcinoma, Lobular/surgery
- Female
- Humans
- Incidence
- Middle Aged
- Registries
- Risk Factors
- United States/epidemiology
- Young Adult
Collapse
Affiliation(s)
- Elizabeth M Ward
- National Vice President, Intramural Research, American Cancer Society, Atlanta, GA
| | - Carol E DeSantis
- Senior Epidemiologist, Surveillance and Health Services Research, American Cancer Society, Atlanta, GA
| | - Chun Chieh Lin
- Senior Epidemiologist, Surveillance and Health Services Research, American Cancer Society, Atlanta, GA
| | - Joan L Kramer
- Assistant Professor of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA
| | - Ahmedin Jemal
- Vice President, Surveillance and Health Services Research, American Cancer Society, Atlanta, GA
| | - Betsy Kohler
- Executive Director, North American Association of Central Cancer Registries, Springfield, IL
| | - Otis W Brawley
- Chief Medical Officer, American Cancer Society, Atlanta, GA
| | - Ted Gansler
- Director of Medical Content, American Cancer Society, Atlanta, GA
| |
Collapse
|
230
|
Basu S, Combe K, Kwiatkowski F, Caldefie-Chézet F, Penault-Llorca F, Bignon YJ, Vasson MP. Cellular Expression of Cyclooxygenase, Aromatase, Adipokines, Inflammation and Cell Proliferation Markers in Breast Cancer Specimen. PLoS One 2015; 10:e0138443. [PMID: 26431176 PMCID: PMC4592217 DOI: 10.1371/journal.pone.0138443] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 08/31/2015] [Indexed: 12/15/2022] Open
Abstract
Current evidences suggest that expression of Ki67, cyclooxygenase (COX), aromatase, adipokines, prostaglandins, free radicals, β-catenin and α-SMA might be involved in breast cancer pathogenesis. The main objective of this study was to compare expression/localization of these potential compounds in breast cancer tissues with tissues collected adjacent to the tumor using immunohistochemistry and correlated with clinical pathology. The breast cancer specimens were collected from 30 women aged between 49 and 89 years who underwent breast surgery following cancer diagnosis. Expression levels of molecules by different stainings were graded as a score on a scale based upon staining intensity and proportion of positive cells/area or individually. AdipoR1, adiponectin, Ob-R, leptin, COX-1, COX-2, aromatase, PGF2α, F2-isoprostanes and α-SMA were localised on higher levels in the breast tissues adjacent to the tumor compared to tumor specimens when considering either score or staining area whereas COX-2 and AdipoR2 were found to be higher considering staining intensity and Ki67 on score level in the tumor tissue. There was no significant difference observed on β-catenin either on score nor on staining area and intensity between tissues adjacent to the tumor and tumor tissues. A positive correlation was found between COX-1 and COX-2 in the tumor tissues. In conclusion, these suggest that Ki67, COXs, aromatase, prostaglandin, free radicals, adipokines, β-catenin and α-SMA are involved in breast cancer. These further focus the need of examination of tissues adjacent to tumor, tumor itself and compare them with normal or benign breast tissues for a better understanding of breast cancer pathology and future evaluation of therapeutic benefit.
Collapse
Affiliation(s)
- Samar Basu
- Clermont Université, Université d'Auvergne, UMR 1019, Unité de Nutrition Humaine, CRNH-Auvergne, BP 10448, F-63000, Clermont-Ferrand, France
- Oxidative Stress and Inflammation, Department of Public Health and Caring Sciences, Faculty of Medicine, Uppsala University, Uppsala, Sweden
- * E-mail:
| | - Kristell Combe
- Clermont Université, Université d'Auvergne, UMR 1019, Unité de Nutrition Humaine, CRNH-Auvergne, BP 10448, F-63000, Clermont-Ferrand, France
| | | | - Florence Caldefie-Chézet
- Clermont Université, Université d'Auvergne, UMR 1019, Unité de Nutrition Humaine, CRNH-Auvergne, BP 10448, F-63000, Clermont-Ferrand, France
| | | | - Yves-Jean Bignon
- Centre Jean Perrin, Unicancer, F-63000, Clermont-Ferrand, France
| | - Marie-Paule Vasson
- Clermont Université, Université d'Auvergne, UMR 1019, Unité de Nutrition Humaine, CRNH-Auvergne, BP 10448, F-63000, Clermont-Ferrand, France
- Centre Jean Perrin, Unicancer, F-63000, Clermont-Ferrand, France
- CHU Clermont-Ferrand, Unité d’exploration nutritionnelle, F-63003, Clermont-Ferrand, France
| |
Collapse
|
231
|
Sai S, Vares G, Kim EH, Karasawa K, Wang B, Nenoi M, Horimoto Y, Hayashi M. Carbon ion beam combined with cisplatin effectively disrupts triple negative breast cancer stem-like cells in vitro. Mol Cancer 2015; 14:166. [PMID: 26338199 PMCID: PMC4560051 DOI: 10.1186/s12943-015-0429-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Accepted: 08/06/2015] [Indexed: 12/23/2022] Open
Abstract
Aims Although a relatively small proportion of all breast cancer (BC), triple negative (TN) BC is responsible for a relatively large proportion of BC deaths because of its worse clinical outcome. To investigate whether a carbon ion beam alone or in combination with cisplatin (CDDP) has a beneficial effect compared to X-rays, we target triple negative (TN) breast cancer stem-like cells (CSCs). Methods Human breast CSCs sorted from MDA-MB-231 and MDA-MB-453 cells were treated with a carbon ion beam or X-ray irradiation alone or in combination with CDDP, and then colony, spheroid and tumor formation assays, RT-PCR Array analysis, and immunofluorescence γH2AX foci assay were performed. Results The colony, spheroid formation, and tumorigenicity assays confirmed that CD44+/CD24- and ESA+/CD24- cells have CSC properties in MDA-MB-231 and MDA-MB-453 cells, respectively. The proportion of CSCs was more enriched after CDDP combination with either X-ray or carbon ion beam, however carbon ion beam combined with CDDP significantly suppressed colony and spheroid formation and more significantly inhibited cell cycle progression (sub-G1 arrest) compared to X-ray combined with CDDP or carbon ion beam alone. RT-PCR Array analysis showed that carbon ion beam combined with CDDP significantly induced apoptosis-related Cytochrome c, almost completely eliminated expression of the CSC markers CD44 and ESA, and significantly inhibited angiogenesis, and metastasis-related HIF1α and CD26 compared to carbon ion beam alone, X-ray alone, or X-ray combined with CDDP. The immunofluorescence assay showed that not only the number but also the size of γH2AX foci in CSCs were larger 24 h after carbon ion beam combined with CDDP compared to those of X-ray alone and X-ray combined with CDDP. Conclusions Carbon ion beam combined with CDDP has superior potential to kill TN breast CSCs with irreparable severe DNA damage and enhanced apoptosis. Electronic supplementary material The online version of this article (doi:10.1186/s12943-015-0429-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sei Sai
- Medical Physics Research Program, Research Center for Charged Particle Therapy, National Institute of Radiological Sciences, 4-9-1 Anagawa Inage-ku, Chiba, Chiba, 263-8555, Japan.
| | - Guillaume Vares
- Radiation Risk Reduction Research Program, Research Center for Radiation Protection, National Institute of Radiological Sciences, Chiba, Japan
| | - Eun Ho Kim
- Division of Heavy Ion Clinical Research, Korea Institute of Radiological and Medical Sciences, 215-4 Gongneung-dong, Nowon-Gu, Seoul, 139-706, South Korea
| | - Kumiko Karasawa
- Research Center Hospital for Charged Particle Therapy, National Institute of Radiological Sciences, Chiba, Japan
| | - Bing Wang
- Radiation Risk Reduction Research Program, Research Center for Radiation Protection, National Institute of Radiological Sciences, Chiba, Japan
| | - Mitsuru Nenoi
- Radiation Risk Reduction Research Program, Research Center for Radiation Protection, National Institute of Radiological Sciences, Chiba, Japan
| | - Yoshiya Horimoto
- Department of Breast Oncology, Juntendo University School of Medicine, Tokyo, Japan
| | - Mitsuhiro Hayashi
- Department of Breast Oncology, Tokyo Medical University Hachioji Medical Center, Tokyo, Japan
| |
Collapse
|
232
|
Abstract
Breast carcinoma is a heterogenous disease. Carcinomas lacking expression of estrogen, progesterone, and HER2/neu receptors by immunohistochemistry and Her2 amplification are designated as triple negative. This group of carcinomas comprises approximately 10% to 20% of all breast carcinomas and is characterized by an aggressive nature with shorter rates of disease-free and overall survival. This aggressive behavior is further compounded by the lack of available targeted therapies. Patients receive cytoxic chemotherapy regimens. Although tumors are initially sensitive to this therapy, drugs are toxic and ineffective in maintaining long-term response thereby providing limited benefit. Much effort is being spent on this group of cancers for the identification of appropriate molecular targets, an effort that is proving challenging due to the presence of marked heterogeneity, both at the morphologic and molecular levels. An understanding of the advances in this field is crucial for developing targeted therapies and tailored patient management protocols. This report summarizes the pathologic subtypes of breast cancer that are commonly of a triple-negative immunophenotype and recent molecular advances in this field.
Collapse
|
233
|
Soysal SD, Tzankov A, Muenst SE. Role of the Tumor Microenvironment in Breast Cancer. Pathobiology 2015; 82:142-52. [DOI: 10.1159/000430499] [Citation(s) in RCA: 213] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
234
|
Choi Y, Hyun E, Seo J, Blundell C, Kim HC, Lee E, Lee S, Moon A, Moon WK, Huh D. A microengineered pathophysiological model of early-stage breast cancer. LAB ON A CHIP 2015; 15:3350-7. [PMID: 26158500 PMCID: PMC4524879 DOI: 10.1039/c5lc00514k] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
A mounting body of evidence in cancer research suggests that the local microenvironment of tumor cells has a profound influence on cancer progression and metastasis. In vitro studies on the tumor microenvironment and its pharmacological modulation, however, are often hampered by the technical challenges associated with creating physiological cell culture environments that integrate cancer cells with the key components of their native niche such as neighboring cells and extracellular matrix (ECM) to mimic complex microarchitecture of cancerous tissue. Using early-stage breast cancer as a model disease, here we describe a biomimetic microengineering strategy to reconstitute three-dimensional (3D) structural organization and microenvironment of breast tumors in human cell-based in vitro models. Specifically, we developed a microsystem that enabled co-culture of breast tumor spheroids with human mammary ductal epithelial cells and mammary fibroblasts in a compartmentalized 3D microfluidic device to replicate microarchitecture of breast ductal carcinoma in situ (DCIS). We also explored the potential of this breast cancer-on-a-chip system as a drug screening platform by evaluating the efficacy and toxicity of an anticancer drug (paclitaxel). Our microengineered disease model represents the first critical step towards recapitulating pathophysiological complexity of breast cancer, and may serve as an enabling tool to systematically examine the contribution of the breast cancer microenvironment to the progression of DCIS to an invasive form of the disease.
Collapse
Affiliation(s)
- Yoonseok Choi
- Department of Radiology, Seoul National University Hospital, Seoul, 110-744, Republic of Korea
| | - Eunjeh Hyun
- Department of Biomedical Engineering, College of Medicine and Institute of Medical and Biological Engineering, Seoul National University, Seoul, 110-744, Republic of Korea
| | - Jeongyun Seo
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cassidy Blundell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hee Chan Kim
- Interdisciplinary Program, Bioengineering Major, Graduate School, Seoul National University, Seoul, 110-744, Republic of Korea
| | - Eunhee Lee
- Department of Radiology, Seoul National University Hospital, Seoul, 110-744, Republic of Korea
| | - Suhyun Lee
- Department of Radiology, Seoul National University Hospital, Seoul, 110-744, Republic of Korea
| | - Aree Moon
- College of Pharmacy, Duksung Women’s University, Seoul 132-714, Republic of Korea
| | - Woo Kyung Moon
- Department of Radiology, Seoul National University Hospital, Seoul, 110-744, Republic of Korea
- To whom correspondence should be addressed. (D. Huh), Tel: 1-215-898-5208. (W.K. Moon), Tel: +82-2-2072-3928
| | - Dongeun Huh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- To whom correspondence should be addressed. (D. Huh), Tel: 1-215-898-5208. (W.K. Moon), Tel: +82-2-2072-3928
| |
Collapse
|
235
|
Texture analysis on MR images helps predicting non-response to NAC in breast cancer. BMC Cancer 2015; 15:574. [PMID: 26243303 PMCID: PMC4526309 DOI: 10.1186/s12885-015-1563-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 07/16/2015] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND To assess the performance of a predictive model of non-response to neoadjuvant chemotherapy (NAC) in patients with breast cancer based on texture, kinetic, and BI-RADS parameters measured from dynamic MRI. METHODS Sixty-nine patients with invasive ductal carcinoma of the breast who underwent pre-treatment MRI were studied. Morphological parameters and biological markers were measured. Pathological complete response was defined as the absence of invasive and in situ cancer in breast and nodes. Pathological non-responders, partial and complete responders were identified. Dynamic imaging was performed at 1.5 T with a 3D axial T1W GRE fat-suppressed sequence. Visual texture, kinetic and BI-RADS parameters were measured in each lesion. ROC analysis and leave-one-out cross-validation were used to assess the performance of individual parameters, then the performance of multi-parametric models in predicting non-response to NAC. RESULTS A model based on four pre-NAC parameters (inverse difference moment, GLN, LRHGE, wash-in) and k-means clustering as statistical classifier identified non-responders with 84 % sensitivity. BI-RADS mass/non-mass enhancement, biological markers and histological grade did not contribute significantly to the prediction. CONCLUSION Pre-NAC texture and kinetic parameters help predicting non-benefit to NAC. Further testing including larger groups of patients with different tumor subtypes is needed to improve the generalization properties and validate the performance of the predictive model.
Collapse
|
236
|
Oh EY, Christensen SM, Ghanta S, Jeong JC, Bucur O, Glass B, Montaser-Kouhsari L, Knoblauch NW, Bertos N, Saleh SM, Haibe-Kains B, Park M, Beck AH. Extensive rewiring of epithelial-stromal co-expression networks in breast cancer. Genome Biol 2015; 16:128. [PMID: 26087699 PMCID: PMC4471934 DOI: 10.1186/s13059-015-0675-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 05/13/2015] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Epithelial-stromal crosstalk plays a critical role in invasive breast cancer pathogenesis; however, little is known on a systems level about how epithelial-stromal interactions evolve during carcinogenesis. RESULTS We develop a framework for building genome-wide epithelial-stromal co-expression networks composed of pairwise co-expression relationships between mRNA levels of genes expressed in the epithelium and stroma across a population of patients. We apply this method to laser capture micro-dissection expression profiling datasets in the setting of breast carcinogenesis. Our analysis shows that epithelial-stromal co-expression networks undergo extensive rewiring during carcinogenesis, with the emergence of distinct network hubs in normal breast, and estrogen receptor-positive and estrogen receptor-negative invasive breast cancer, and the emergence of distinct patterns of functional network enrichment. In contrast to normal breast, the strongest epithelial-stromal co-expression relationships in invasive breast cancer mostly represent self-loops, in which the same gene is co-expressed in epithelial and stromal regions. We validate this observation using an independent laser capture micro-dissection dataset and confirm that self-loop interactions are significantly increased in cancer by performing computational image analysis of epithelial and stromal protein expression using images from the Human Protein Atlas. CONCLUSIONS Epithelial-stromal co-expression network analysis represents a new approach for systems-level analyses of spatially localized transcriptomic data. The analysis provides new biological insights into the rewiring of epithelial-stromal co-expression networks and the emergence of epithelial-stromal co-expression self-loops in breast cancer. The approach may facilitate the development of new diagnostics and therapeutics targeting epithelial-stromal interactions in cancer.
Collapse
Affiliation(s)
- Eun-Yeong Oh
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, MA, 02215, USA. .,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA. .,Harvard Medical School, Boston, MA, 02215, USA.
| | - Stephen M Christensen
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, MA, 02215, USA. .,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA. .,Harvard Medical School, Boston, MA, 02215, USA.
| | - Sindhu Ghanta
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, MA, 02215, USA. .,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA. .,Harvard Medical School, Boston, MA, 02215, USA.
| | - Jong Cheol Jeong
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, MA, 02215, USA. .,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA. .,Harvard Medical School, Boston, MA, 02215, USA.
| | - Octavian Bucur
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, MA, 02215, USA. .,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA. .,Harvard Medical School, Boston, MA, 02215, USA.
| | - Benjamin Glass
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, MA, 02215, USA. .,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA. .,Harvard Medical School, Boston, MA, 02215, USA.
| | - Laleh Montaser-Kouhsari
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, MA, 02215, USA. .,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA. .,Harvard Medical School, Boston, MA, 02215, USA.
| | - Nicholas W Knoblauch
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, MA, 02215, USA. .,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA. .,Harvard Medical School, Boston, MA, 02215, USA.
| | - Nicholas Bertos
- Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada.
| | - Sadiq Mi Saleh
- Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada.
| | - Benjamin Haibe-Kains
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada. .,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, M5G 1L7, Canada.
| | - Morag Park
- Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada.
| | - Andrew H Beck
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, MA, 02215, USA. .,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA. .,Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
237
|
Elbaz M, Nasser MW, Ravi J, Wani NA, Ahirwar DK, Zhao H, Oghumu S, Satoskar AR, Shilo K, Carson WE, Ganju RK. Modulation of the tumor microenvironment and inhibition of EGF/EGFR pathway: novel anti-tumor mechanisms of Cannabidiol in breast cancer. Mol Oncol 2015; 9:906-919. [PMID: 25660577 PMCID: PMC4387115 DOI: 10.1016/j.molonc.2014.12.010] [Citation(s) in RCA: 153] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 12/08/2014] [Accepted: 12/27/2014] [Indexed: 12/12/2022] Open
Abstract
The anti-tumor role and mechanisms of Cannabidiol (CBD), a non-psychotropic cannabinoid compound, are not well studied especially in triple-negative breast cancer (TNBC). In the present study, we analyzed CBD's anti-tumorigenic activity against highly aggressive breast cancer cell lines including TNBC subtype. We show here -for the first time-that CBD significantly inhibits epidermal growth factor (EGF)-induced proliferation and chemotaxis of breast cancer cells. Further studies revealed that CBD inhibits EGF-induced activation of EGFR, ERK, AKT and NF-kB signaling pathways as well as MMP2 and MMP9 secretion. In addition, we demonstrated that CBD inhibits tumor growth and metastasis in different mouse model systems. Analysis of molecular mechanisms revealed that CBD significantly inhibits the recruitment of tumor-associated macrophages in primary tumor stroma and secondary lung metastases. Similarly, our in vitro studies showed a significant reduction in the number of migrated RAW 264.7 cells towards the conditioned medium of CBD-treated cancer cells. The conditioned medium of CBD-treated cancer cells also showed lower levels of GM-CSF and CCL3 cytokines which are important for macrophage recruitment and activation. In summary, our study shows -for the first time-that CBD inhibits breast cancer growth and metastasis through novel mechanisms by inhibiting EGF/EGFR signaling and modulating the tumor microenvironment. These results also indicate that CBD can be used as a novel therapeutic option to inhibit growth and metastasis of highly aggressive breast cancer subtypes including TNBC, which currently have limited therapeutic options and are associated with poor prognosis and low survival rates.
Collapse
Affiliation(s)
- Mohamad Elbaz
- Department of Pathology, The Ohio State University, Wexner Medical Center, 43210, USA; The Comprehensive Cancer Center, The Ohio State University, Wexner Medical Center, 43210, USA.
| | - Mohd W Nasser
- Department of Pathology, The Ohio State University, Wexner Medical Center, 43210, USA; The Comprehensive Cancer Center, The Ohio State University, Wexner Medical Center, 43210, USA.
| | - Janani Ravi
- Department of Pathology, The Ohio State University, Wexner Medical Center, 43210, USA; The Comprehensive Cancer Center, The Ohio State University, Wexner Medical Center, 43210, USA.
| | - Nissar A Wani
- Department of Pathology, The Ohio State University, Wexner Medical Center, 43210, USA; The Comprehensive Cancer Center, The Ohio State University, Wexner Medical Center, 43210, USA.
| | - Dinesh K Ahirwar
- Department of Pathology, The Ohio State University, Wexner Medical Center, 43210, USA; The Comprehensive Cancer Center, The Ohio State University, Wexner Medical Center, 43210, USA.
| | - Helong Zhao
- Department of Pathology, The Ohio State University, Wexner Medical Center, 43210, USA; The Comprehensive Cancer Center, The Ohio State University, Wexner Medical Center, 43210, USA.
| | - Steve Oghumu
- Department of Pathology, The Ohio State University, Wexner Medical Center, 43210, USA; The Comprehensive Cancer Center, The Ohio State University, Wexner Medical Center, 43210, USA.
| | - Abhay R Satoskar
- Department of Pathology, The Ohio State University, Wexner Medical Center, 43210, USA; The Comprehensive Cancer Center, The Ohio State University, Wexner Medical Center, 43210, USA.
| | - Konstantin Shilo
- Department of Pathology, The Ohio State University, Wexner Medical Center, 43210, USA; The Comprehensive Cancer Center, The Ohio State University, Wexner Medical Center, 43210, USA.
| | - William E Carson
- The Comprehensive Cancer Center, The Ohio State University, Wexner Medical Center, 43210, USA; Department of Surgery, The Ohio State University, Wexner Medical Center, 43210, USA.
| | - Ramesh K Ganju
- Department of Pathology, The Ohio State University, Wexner Medical Center, 43210, USA; The Comprehensive Cancer Center, The Ohio State University, Wexner Medical Center, 43210, USA.
| |
Collapse
|
238
|
Fibulin-3 is a novel TGF-β pathway inhibitor in the breast cancer microenvironment. Oncogene 2015; 34:5635-47. [PMID: 25823021 PMCID: PMC4589427 DOI: 10.1038/onc.2015.13] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 12/17/2014] [Accepted: 01/05/2015] [Indexed: 12/11/2022]
Abstract
TGF-β is an important regulator of breast cancer progression. However, how the breast cancer microenvironment regulates TGF-β signaling during breast cancer progression remains largely unknown. Here, we identified fibulin-3 as a secreted protein in the breast cancer microenvironment, which efficiently inhibits TGF-β signaling in both breast cancer cells and endothelial cells. Mechanistically, fibulin-3 interacts with the type I TGF-β receptor (TβRI) to block TGF-β induced complex formation of TβRI with the type II TGF-β receptor (TβRII) and subsequent downstream TGF-β signaling. Fibulin-3 expression decreases during breast cancer progression, with low fibulin-3 levels correlating with a poorer prognosis. Functionally, high fibulin-3 levels inhibited TGF-β-induced EMT, migration, invasion and endothelial permeability, while loss of fibulin-3 expression/function promoted these TGF-β-mediated effects. Further, restoring fibulin-3 expression in breast cancer cells inhibited TGF-β signaling, breast cancer cell EMT, invasion and metastasis in vivo. These studies provide a novel mechanism for how TGF-β signaling is regulated by the tumor microenvironment, and provide insight into targeting the TGF-β signaling pathway in human breast cancer patients.
Collapse
|
239
|
Nifuroxazide induces apoptosis and impairs pulmonary metastasis in breast cancer model. Cell Death Dis 2015; 6:e1701. [PMID: 25811798 PMCID: PMC4385941 DOI: 10.1038/cddis.2015.63] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 02/01/2015] [Accepted: 02/09/2015] [Indexed: 02/05/2023]
Abstract
Breast carcinoma is the most common female cancer with considerable metastatic potential. Signal transducers and activators of the transcription 3 (Stat3) signaling pathway is constitutively activated in many cancers including breast cancer and has been validated as a novel potential anticancer target. Here, we reported our finding with nifuroxazide, an antidiarrheal agent identified as a potent inhibitor of Stat3. The potency of nifuroxazide on breast cancer was assessed in vitro and in vivo. In this investigation, we found that nifuroxazide decreased the viability of three breast cancer cell lines and induced apoptosis of cancer cells in a dose-dependent manner. In addition, western blot analysis demonstrated that the occurrence of its apoptosis was associated with activation of cleaved caspases-3 and Bax, downregulation of Bcl-2. Moreover, nifuroxazide markedly blocked cancer cell migration and invasion, and the reduction of phosphorylated-Stat3Tyr705, matrix metalloproteinase (MMP) MMP-2 and MMP-9 expression were also observed. Furthermore, in our animal experiments, intraperitoneal administration of 50 mg/kg/day nifuroxazide suppressed 4T1 tumor growth and blocked formation of pulmonary metastases without detectable toxicity. Meanwhile, histological and immunohistochemical analyses revealed a decrease in Ki-67-positive cells, MMP-9-positive cells and an increase in cleaved caspase-3-positive cells upon nifuroxazide. Notably, nifuroxazide reduced the number of myeloid-derived suppressor cell in the lung. Our data indicated that nifuroxazide may potentially be a therapeutic agent for growth and metastasis of breast cancer.
Collapse
|
240
|
Templeton ZS, Bachmann MH, Alluri RV, Maloney WJ, Contag CH, King BL. Methods for culturing human femur tissue explants to study breast cancer cell colonization of the metastatic niche. J Vis Exp 2015:52656. [PMID: 25867136 PMCID: PMC4401351 DOI: 10.3791/52656] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Bone is the most common site of breast cancer metastasis. Although it is widely accepted that the microenvironment influences cancer cell behavior, little is known about breast cancer cell properties and behaviors within the native microenvironment of human bone tissue.We have developed approaches to track, quantify and modulate human breast cancer cells within the microenvironment of cultured human bone tissue fragments isolated from discarded femoral heads following total hip replacement surgeries. Using breast cancer cells engineered for luciferase and enhanced green fluorescent protein (EGFP) expression, we are able to reproducibly quantitate migration and proliferation patterns using bioluminescence imaging (BLI), track cell interactions within the bone fragments using fluorescence microscopy, and evaluate breast cells after colonization with flow cytometry. The key advantages of this model include: 1) a native, architecturally intact tissue microenvironment that includes relevant human cell types, and 2) direct access to the microenvironment, which facilitates rapid quantitative and qualitative monitoring and perturbation of breast and bone cell properties, behaviors and interactions. A primary limitation, at present, is the finite viability of the tissue fragments, which confines the window of study to short-term culture. Applications of the model system include studying the basic biology of breast cancer and other bone-seeking malignancies within the metastatic niche, and developing therapeutic strategies to effectively target breast cancer cells in bone tissues.
Collapse
Affiliation(s)
| | | | - Rajiv V Alluri
- Department of Pediatrics, Stanford University School of Medicine
| | - William J Maloney
- Department of Orthopaedic Surgery, Stanford University School of Medicine
| | | | - Bonnie L King
- Department of Pediatrics, Stanford University School of Medicine;
| |
Collapse
|
241
|
Lee SK, Choi MY, Bae SY, Lee JH, Lee HC, Kil WH, Lee JE, Kim SW, Nam SJ. Immediate postoperative inflammation is an important prognostic factor in breast cancer. Oncology 2015; 88:337-44. [PMID: 25721153 DOI: 10.1159/000368985] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 10/06/2014] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Inflammation is associated with worse outcomes in cancer. Operations induce an acute inflammatory response and could impact the clinical outcomes in breast cancer. The neutrophil-lymphocyte ratio (NLR) is a well-known indicator of inflammation. We investigated the prognostic significance of perioperative inflammation with the NLR in breast cancer. METHODS We reviewed the clinical and pathological records of women diagnosed with invasive breast carcinoma at the Samsung Medical Center between 2000 and 2010. The NLR levels in the immediate preoperative period and the postoperative periods (1 week and 1 month) were assessed. RESULTS The NLRs of a total of 3,116 breast cancer patients were examined. In the univariate analysis, the NLR in postoperative week 1, total mastectomy, the presence of lymphovascular invasion, a higher nuclear grade and pathologic TNM stage, and negative hormone receptor and subtypes were factors associated with poor disease-specific survival. The NLR in postoperative week 1 remained a significant prognostic factor in the multivariate analysis. A cutoff level of 5.2, determined by the minimum p value approach, was found to be a significant level for discriminating the impact on breast cancer-specific mortality (p = 0.0116 adjusted by the Bonferroni correction). CONCLUSIONS Immediate postoperative inflammation is an important prognostic marker in breast cancer patients.
Collapse
Affiliation(s)
- Se Kyung Lee
- Division of Breast and Endocrine Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
242
|
Sağlam Ö, Ünal ZS, Subaşı C, Ulukaya E, Karaöz E. IL-6 originated from breast cancer tissue-derived mesenchymal stromal cells may contribute to carcinogenesis. Tumour Biol 2015; 36:5667-77. [PMID: 25697898 DOI: 10.1007/s13277-015-3241-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Accepted: 02/09/2015] [Indexed: 12/21/2022] Open
Abstract
Tumor microenvironment is an important factor, which sustains and promotes the tumors by inflammatory signals. Interleukin-6 (IL-6) is known as a multifunctional cytokine, which is a major activator of the signaling pathway of Janus kinases (JAKs)/signal transducer and activator of transcription 3 (STAT3). In this study, we aimed to investigate the effect of IL-6 in the tumor microenvironment on carcinogenesis. For this purpose, healthy breast tissue-derived stromal cells (HBT-SCs) and malign breast tissue-derived stromal cells (MBT-SCs) were co-cultured with MCF-7 (human breast adenocarcinoma cell line) cells using semipermeable membranes. The cell proliferation was monitored with water-soluble tetrazolium (WST) and carboxyfluorescein succinimidyl ester (CFSE) assays. Protein levels were measured by enzyme-linked immunosorbent assay (ELISA) and Western blot hybridization, while gene expressions were measured by real-time PCR. The results demonstrated that IL-6 protein levels increased significantly in the supernatants of MBT-SCs when they were co-cultured with MCF-7 cells. In accordance with this, the expression of IL-6 was significantly higher in MBT-SCs. Additionally, the expression of STAT3 in MCF-7 cells increased slightly when they were co-cultured with MBT-SCs. Considering together, there is an important interaction between tumor microenvironment and tumor cells mediated by IL-6 signaling. Thereby, the targeting on IL-6 signaling in the treatment of cancer might effectively prevent the tumor progression.
Collapse
Affiliation(s)
- Özlem Sağlam
- Center for Stem Cell and Gene Therapies Research and Practice, Department of Stem Cell, Kocaeli University, Kocaeli, Turkey
| | | | | | | | | |
Collapse
|
243
|
Quantitative high throughput screening using a primary human three-dimensional organotypic culture predicts in vivo efficacy. Nat Commun 2015; 6:6220. [PMID: 25653139 DOI: 10.1038/ncomms7220] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 01/07/2015] [Indexed: 02/06/2023] Open
Abstract
The tumour microenvironment contributes to cancer metastasis and drug resistance. However, most high throughput screening (HTS) assays for drug discovery use cancer cells grown in monolayers. Here we show that a multilayered culture containing primary human fibroblasts, mesothelial cells and extracellular matrix can be adapted into a reliable 384- and 1,536-multi-well HTS assay that reproduces the human ovarian cancer (OvCa) metastatic microenvironment. We validate the identified inhibitors in secondary in vitro and in vivo biological assays using three OvCa cell lines: HeyA8, SKOV3ip1 and Tyk-nu. The active compounds directly inhibit at least two of the three OvCa functions: adhesion, invasion and growth. In vivo, these compounds prevent OvCa adhesion, invasion and metastasis, and improve survival in mouse models. Collectively, these data indicate that a complex three-dimensional culture of the tumour microenvironment can be adapted for quantitative HTS and may improve the disease relevance of assays used for drug screening.
Collapse
|
244
|
The reprogramming of tumor stroma by HSF1 is a potent enabler of malignancy. Cell 2015; 158:564-78. [PMID: 25083868 DOI: 10.1016/j.cell.2014.05.045] [Citation(s) in RCA: 303] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 04/03/2014] [Accepted: 05/28/2014] [Indexed: 02/06/2023]
Abstract
Stromal cells within the tumor microenvironment are essential for tumor progression and metastasis. Surprisingly little is known about the factors that drive the transcriptional reprogramming of stromal cells within tumors. We report that the transcriptional regulator heat shock factor 1 (HSF1) is frequently activated in cancer-associated fibroblasts (CAFs), where it is a potent enabler of malignancy. HSF1 drives a transcriptional program in CAFs that complements, yet is completely different from, the program it drives in adjacent cancer cells. This CAF program is uniquely structured to support malignancy in a non-cell-autonomous way. Two central stromal signaling molecules-TGF-β and SDF1-play a critical role. In early-stage breast and lung cancer, high stromal HSF1 activation is strongly associated with poor patient outcome. Thus, tumors co-opt the ancient survival functions of HSF1 to orchestrate malignancy in both cell-autonomous and non-cell-autonomous ways, with far-reaching therapeutic implications.
Collapse
|
245
|
Chen X, Qian Y, Wu S. The Warburg effect: evolving interpretations of an established concept. Free Radic Biol Med 2015; 79:253-63. [PMID: 25277420 PMCID: PMC4356994 DOI: 10.1016/j.freeradbiomed.2014.08.027] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 08/15/2014] [Accepted: 08/23/2014] [Indexed: 12/20/2022]
Abstract
Metabolic reprogramming and altered bioenergetics have emerged as hallmarks of cancer and an area of active basic and translational cancer research. Drastically upregulated glucose transport and metabolism in most cancers regardless of the oxygen supply, a phenomenon called the Warburg effect, is a major focuses of the research. Warburg speculated that cancer cells, due to defective mitochondrial oxidative phosphorylation (OXPHOS), switch to glycolysis for ATP synthesis, even in the presence of oxygen. Studies in the recent decade indicated that while glycolysis is indeed drastically upregulated in almost all cancer cells, mitochondrial respiration continues to operate normally at rates proportional to oxygen supply. There is no OXPHOS-to-glycolysis switch but rather upregulation of glycolysis. Furthermore, upregulated glycolysis appears to be for synthesis of biomass and reducing equivalents in addition to ATP production. The new finding that a significant amount of glycolytic intermediates is diverted to the pentose phosphate pathway (PPP) for production of NADPH has profound implications in how cancer cells use the Warburg effect to cope with reactive oxygen species (ROS) generation and oxidative stress, opening the door for anticancer interventions taking advantage of this. Recent findings in the Warburg effect and its relationship with ROS and oxidative stress controls will be reviewed. Cancer treatment strategies based on these new findings will be presented and discussed.
Collapse
Affiliation(s)
- Xiaozhuo Chen
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; Molecular and Cellular Biology Program, Ohio University, Athens, OH 45701, USA; Department of Biomedical Sciences, Ohio University, Athens, OH 45701, USA; Department of Chemistry and Biochemistry, Ohio University, Athens, OH 45701, USA
| | - Yanrong Qian
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; Molecular and Cellular Biology Program, Ohio University, Athens, OH 45701, USA; Department of Chemistry and Biochemistry, Ohio University, Athens, OH 45701, USA
| | - Shiyong Wu
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; Molecular and Cellular Biology Program, Ohio University, Athens, OH 45701, USA; Department of Chemistry and Biochemistry, Ohio University, Athens, OH 45701, USA.
| |
Collapse
|
246
|
Zheng MJ, Wang J, Xu L, Zha XM, Zhao Y, Ling LJ, Wang S. Human mammary microenvironment better regulates the biology of human breast cancer in humanized mouse model. Med Oncol 2015; 32:427. [PMID: 25572806 DOI: 10.1007/s12032-014-0427-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Accepted: 12/02/2014] [Indexed: 01/21/2023]
Abstract
During the past decades, many efforts have been made in mimicking the clinical progress of human cancer in mouse models. Previously, we developed a human breast tissue-derived (HB) mouse model. Theoretically, it may mimic the interactions between "species-specific" mammary microenvironment of human origin and human breast cancer cells. However, detailed evidences are absent. The present study (in vivo, cellular, and molecular experiments) was designed to explore the regulatory role of human mammary microenvironment in the progress of human breast cancer cells. Subcutaneous (SUB), mammary fat pad (MFP), and HB mouse models were developed for in vivo comparisons. Then, the orthotopic tumor masses from three different mouse models were collected for primary culture. Finally, the biology of primary cultured human breast cancer cells was compared by cellular and molecular experiments. Results of in vivo mouse models indicated that human breast cancer cells grew better in human mammary microenvironment. Cellular and molecular experiments confirmed that primary cultured human breast cancer cells from HB mouse model showed a better proliferative and anti-apoptotic biology than those from SUB to MFP mouse models. Meanwhile, primary cultured human breast cancer cells from HB mouse model also obtained the migratory and invasive biology for "species-specific" tissue metastasis to human tissues. Comprehensive analyses suggest that "species-specific" mammary microenvironment of human origin better regulates the biology of human breast cancer cells in our humanized mouse model of breast cancer, which is more consistent with the clinical progress of human breast cancer.
Collapse
Affiliation(s)
- Ming-Jie Zheng
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, 210029, Nanjing, China
| | | | | | | | | | | | | |
Collapse
|
247
|
Pan MH, Chiou YS, Chen LH, Ho CT. Breast cancer chemoprevention by dietary natural phenolic compounds: Specific epigenetic related molecular targets. Mol Nutr Food Res 2014; 59:21-35. [DOI: 10.1002/mnfr.201400515] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 10/08/2014] [Accepted: 11/03/2014] [Indexed: 12/21/2022]
Affiliation(s)
- Min-Hsiung Pan
- Institute of Food Science and Technology; National Taiwan University; Taipei Taiwan
- Department of Medical Research, China Medical University Hospital; China Medical University; Taichung Taiwan
| | - Yi-Siou Chiou
- Institute of Food Science and Technology; National Taiwan University; Taipei Taiwan
| | - Li-Hua Chen
- Institute of Food Science and Technology; National Taiwan University; Taipei Taiwan
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University; New Brunswick; NJ USA
| |
Collapse
|
248
|
Andolfi L, Bourkoula E, Migliorini E, Palma A, Pucer A, Skrap M, Scoles G, Beltrami AP, Cesselli D, Lazzarino M. Investigation of adhesion and mechanical properties of human glioma cells by single cell force spectroscopy and atomic force microscopy. PLoS One 2014; 9:e112582. [PMID: 25390644 PMCID: PMC4229222 DOI: 10.1371/journal.pone.0112582] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 10/08/2014] [Indexed: 11/18/2022] Open
Abstract
Active cell migration and invasion is a peculiar feature of glioma that makes this tumor able to rapidly infiltrate into the surrounding brain tissue. In our recent work, we identified a novel class of glioma-associated-stem cells (defined as GASC for high-grade glioma -HG- and Gasc for low-grade glioma -LG-) that, although not tumorigenic, act supporting the biological aggressiveness of glioma-initiating stem cells (defined as GSC for HG and Gsc for LG) favoring also their motility. Migrating cancer cells undergo considerable molecular and cellular changes by remodeling their cytoskeleton and cell interactions with surrounding environment. To get a better understanding about the role of the glioma-associated-stem cells in tumor progression, cell deformability and interactions between glioma-initiating stem cells and glioma-associated-stem cells were investigated. Adhesion of HG/LG-cancer cells on HG/LG-glioma-associated stem cells was studied by time-lapse microscopy, while cell deformability and cell-cell adhesion strengths were quantified by indentation measurements by atomic force microscopy and single cell force spectroscopy. Our results demonstrate that for both HG and LG glioma, cancer-initiating-stem cells are softer than glioma-associated-stem cells, in agreement with their neoplastic features. The adhesion strength of GSC on GASC appears to be significantly lower than that observed for Gsc on Gasc. Whereas, GSC spread and firmly adhere on Gasc with an adhesion strength increased as compared to that obtained on GASC. These findings highlight that the grade of glioma-associated-stem cells plays an important role in modulating cancer cell adhesion, which could affect glioma cell migration, invasion and thus cancer aggressiveness. Moreover this work provides evidence about the importance of investigating cell adhesion and elasticity for new developments in disease diagnostics and therapeutics.
Collapse
Affiliation(s)
- Laura Andolfi
- Istituto Officina dei Materiali-National Research Council, Trieste, Italy
- * E-mail:
| | - Eugenia Bourkoula
- Department of Medical and Biological Sciences, University of Udine, Udine, Italy
| | - Elisa Migliorini
- Département de Chimie Moléculaire, Ingénierie et Interactions Bio Moléculaires, Université Joseph Fourier, Grenoble, France
| | - Anita Palma
- Department of Medical and Biological Sciences, University of Udine, Udine, Italy
| | - Anja Pucer
- Department of Medical and Biological Sciences, University of Udine, Udine, Italy
| | - Miran Skrap
- Department of Medical and Biological Sciences, University of Udine, Udine, Italy
| | - Giacinto Scoles
- Department of Medical and Biological Sciences, University of Udine, Udine, Italy
| | | | - Daniela Cesselli
- Department of Medical and Biological Sciences, University of Udine, Udine, Italy
| | - Marco Lazzarino
- Istituto Officina dei Materiali-National Research Council, Trieste, Italy
- Cluster in Biomedicine, Trieste, Italy
| |
Collapse
|
249
|
Marongiu F, Serra MP, Sini M, Angius F, Laconi E. Clearance of senescent hepatocytes in a neoplastic-prone microenvironment delays the emergence of hepatocellular carcinoma. Aging (Albany NY) 2014; 6:26-34. [PMID: 24464501 PMCID: PMC3927807 DOI: 10.18632/aging.100631] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Increasing evidence indicates that carcinogenesis is dependent on the tissue context in which it occurs, implying that the latter can be a target for preventive or therapeutic strategies. We tested the possibility that re-normalizing a senescent, neoplastic-prone tissue microenvironment would exert a modulatory effect on the emergence of neoplastic disease. Rats were exposed to a protocol for the induction of hepatocellular carcinoma (HCC). Using an orthotopic and syngeneic system for cell transplantation, one group of animal was then delivered 8 million normal hepatocytes, via the portal circulation. Hepatocytes transplantation resulted in a prominent decrease in the incidence of both pre-neoplastic and neoplastic lesions. At the end of 1 year 50% of control animals presented with HCC, while no HCC were observed in the transplanted group. Extensive hepatocyte senescence was induced by the carcinogenic protocol in the host liver; however, senescent cells were largely cleared following infusion of normal hepatocytes. Furthermore, levels of Il-6 increased in rats exposed to the carcinogenic protocol, while they returned to near control values in the group receiving hepatocyte transplantation. These results support the concept that strategies aimed at normalizing a neoplastic-prone tissue landscape can modulate progression of neoplastic disease.
Collapse
Affiliation(s)
- Fabio Marongiu
- Department of Biomedical Sciences, Unit of Experimental Medicine, University of Cagliari, 09124 Cagliari, Italy
| | | | | | | | | |
Collapse
|
250
|
Sun X, Sandhu R, Figueroa JD, Gierach GL, Sherman ME, Troester MA. Benign breast tissue composition in breast cancer patients: association with risk factors, clinical variables, and gene expression. Cancer Epidemiol Biomarkers Prev 2014; 23:2810-8. [PMID: 25249325 DOI: 10.1158/1055-9965.epi-14-0507] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Breast tissue composition (epithelium, non-fatty stroma, and adipose) changes qualitatively and quantitatively throughout the lifespan, and may mediate relationships between risk factors and breast cancer initiation. We sought to identify relationships between tissue composition, risk factors, tumor characteristics, and gene expression. METHODS Participants were 146 patients from the Polish Breast Cancer Study, with data on risk factor and clinicopathological characteristics. Benign breast tissue composition was evaluated using digital image analysis of histologic sections. Whole-genome microarrays were performed on the same tissue blocks. RESULTS Mean epithelial, non-fatty stromal, and adipose proportions were 8.4% (SD = 4.9%), 27.7% (SD = 24.0%), and 64.0% (SD = 24.0%), respectively. Among women <50 years old, stroma proportion decreased and adipose proportion increased with age, with approximately 2% difference per year (P < 0.01). The variation in epithelial proportion with age was modest (0.1% per year). Higher epithelial proportion was associated with obesity (7.6% in nonobese vs. 10.1% in obese; P = 0.02) and with poorly differentiated tumors (7.8% in well/moderate vs. 9.9% in poor; P = 0.05). Gene expression signatures associated with epithelial and stromal proportion were identified and validated. Stroma-associated genes were in metabolism and stem cell maintenance pathways, whereas epithelial genes were enriched for cytokine and immune response pathways. CONCLUSIONS Breast tissue composition was associated with age, body mass index, and tumor grade, with consequences for breast gene expression. IMPACT Breast tissue morphologic factors may influence breast cancer etiology. Composition and gene expression may act as biomarkers of breast cancer risk and progression.
Collapse
Affiliation(s)
| | - Rupninder Sandhu
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jonine D Figueroa
- Hormonal and Reproductive Epidemiology Branch, Division of Cancer Epidemiology and Genetics, and
| | - Gretchen L Gierach
- Hormonal and Reproductive Epidemiology Branch, Division of Cancer Epidemiology and Genetics, and
| | - Mark E Sherman
- Hormonal and Reproductive Epidemiology Branch, Division of Cancer Epidemiology and Genetics, and Breast and Gynecologic Cancer Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland
| | - Melissa A Troester
- Department of Epidemiology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| |
Collapse
|