101
|
Kocatürk B, Versteeg HH. Orthotopic injection of breast cancer cells into the mammary fat pad of mice to study tumor growth. J Vis Exp 2015. [PMID: 25742185 DOI: 10.3791/51967] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Breast cancer growth can be studied in mice using a plethora of models. Genetic manipulation of breast cancer cells may provide insights into the functions of proteins involved in oncogenic progression or help to discover new tumor suppressors. In addition, injecting cancer cells into mice with different genotypes might provide a better understanding of the importance of the stromal compartment. Many models may be useful to investigate certain aspects of disease progression but do not recapitulate the entire cancerous process. In contrast, breast cancer cells engraftment to the mammary fat pad of mice better recapitulates the location of the disease and presence of the proper stromal compartment and therefore better mimics human cancerous disease. In this article, we describe how to implant breast cancer cells into mice orthotopically and explain how to collect tissues to analyse the tumor milieu and metastasis to distant organs. Using this model, many aspects (growth, angiogenesis, and metastasis) of cancer can be investigated simply by providing a proper environment for tumor cells to grow.
Collapse
Affiliation(s)
- Begüm Kocatürk
- Department of Hemostasis and Thrombosis, Leiden University Medical Center
| | - Henri H Versteeg
- Department of Hemostasis and Thrombosis, Leiden University Medical Center;
| |
Collapse
|
102
|
Nasser MW, Wani NA, Ahirwar DK, Powell CA, Ravi J, Elbaz M, Zhao H, Padilla L, Zhang X, Shilo K, Ostrowski M, Shapiro C, Carson WE, Ganju RK. RAGE mediates S100A7-induced breast cancer growth and metastasis by modulating the tumor microenvironment. Cancer Res 2015; 75:974-85. [PMID: 25572331 DOI: 10.1158/0008-5472.can-14-2161] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
RAGE is a multifunctional receptor implicated in diverse processes including inflammation and cancer. In this study, we report that RAGE expression is upregulated widely in aggressive triple-negative breast cancer (TNBC) cells, both in primary tumors and in lymph node metastases. In evaluating the functional contributions of RAGE in breast cancer, we found that RAGE-deficient mice displayed a reduced propensity for breast tumor growth. In an established model of lung metastasis, systemic blockade by injection of a RAGE neutralizing antibody inhibited metastasis development. Mechanistic investigations revealed that RAGE bound to the proinflammatory ligand S100A7 and mediated its ability to activate ERK, NF-κB, and cell migration. In an S100A7 transgenic mouse model of breast cancer (mS100a7a15 mice), administration of either RAGE neutralizing antibody or soluble RAGE was sufficient to inhibit tumor progression and metastasis. In this model, we found that RAGE/S100A7 conditioned the tumor microenvironment by driving the recruitment of MMP9-positive tumor-associated macrophages. Overall, our results highlight RAGE as a candidate biomarker for TNBCs, and they reveal a functional role for RAGE/S100A7 signaling in linking inflammation to aggressive breast cancer development.
Collapse
Affiliation(s)
- Mohd W Nasser
- Department of Pathology, The Ohio State Medical Center, Columbus, Ohio
| | - Nissar Ahmad Wani
- Department of Pathology, The Ohio State Medical Center, Columbus, Ohio
| | - Dinesh K Ahirwar
- Department of Pathology, The Ohio State Medical Center, Columbus, Ohio
| | | | - Janani Ravi
- Department of Pathology, The Ohio State Medical Center, Columbus, Ohio
| | - Mohamad Elbaz
- Department of Pathology, The Ohio State Medical Center, Columbus, Ohio
| | - Helong Zhao
- Department of Pathology, The Ohio State Medical Center, Columbus, Ohio
| | - Laura Padilla
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| | - Xiaoli Zhang
- Centre for Biostatics, The Ohio State Medical Center, Columbus, Ohio
| | - Konstantin Shilo
- Department of Pathology, The Ohio State Medical Center, Columbus, Ohio
| | - Michael Ostrowski
- Comprehensive Cancer Center, The Ohio State Medical Center, Columbus, Ohio
| | - Charles Shapiro
- Department of Pathology, The Ohio State Medical Center, Columbus, Ohio
| | - William E Carson
- Comprehensive Cancer Center, The Ohio State Medical Center, Columbus, Ohio. Department of Surgery, The Ohio State Medical Center, Columbus, Ohio
| | - Ramesh K Ganju
- Department of Pathology, The Ohio State Medical Center, Columbus, Ohio.
| |
Collapse
|
103
|
Ogba N, Manning NG, Bliesner BS, Ambler SK, Haughian JM, Pinto MP, Jedlicka P, Joensuu K, Heikkilä P, Horwitz KB. Luminal breast cancer metastases and tumor arousal from dormancy are promoted by direct actions of estradiol and progesterone on the malignant cells. Breast Cancer Res 2014; 16:489. [PMID: 25475897 PMCID: PMC4303198 DOI: 10.1186/s13058-014-0489-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 11/19/2014] [Indexed: 12/14/2022] Open
Abstract
Introduction Luminal, estrogen receptor-positive (ER+) breast cancers can metastasize but lie dormant for years before recurrences prove lethal. Understanding the roles of estrogen (E) or progestin (P) in development of luminal metastases or in arousal from dormancy is hindered by few preclinical models. We have developed such models. Methods Immunocompromised, ovariectomized (ovx’d) mice were intracardiac-injected with luminal or basal human breast cancer cells. Four lines were tested: luminal ER+PR+ cytokeratin 5-negative (CK5−) E3 and MCF-7 cells, basal ER−PR−CK5+ estrogen withdrawn-line 8 (EWD8) cells, and basal ER−PR−CK5− MDA-MB-231 cells. Development of micrometastases or macrometastases was quantified in ovx’d mice and in mice supplemented with E or P or both. Metastatic deposits were analyzed by immunohistochemistry for luminal, basal, and proliferation markers. Results ER−PR− cells generated macrometastases in multiple organs in the absence or presence of hormones. By contrast, ovx’d mice injected with ER+PR+ cells appeared to be metastases-free until they were supplemented with E or E+P. Furthermore, unlike parental ER+PR+CK5− cells, luminal metastases were heterogeneous, containing a significant (6% to 30%) proportion of non-proliferative ER−PR−CK5+ cells that would be chemotherapy-resistant. Additionally, because these cells lack receptors, they would also be endocrine therapy-resistant. With regard to ovx’d control mice injected with ER+PR+ cells that appeared to be metastases-free, systematic pathologic analysis of organs showed that some harbor a reservoir of dormant micrometastases that are ER+ but PR−. Such cells may also be endocrine therapy- and chemotherapy-resistant. Their emergence as macrometastases can be triggered by E or E+P restoration. Conclusions We conclude that hormones promote development of multi-organ macrometastases in luminal disease. The metastases display a disturbing heterogeneity, containing newly emergent ER−PR− subpopulations that would be resistant to endocrine therapy and chemotherapy. Similar cells are found in luminal metastases of patients. Furthermore, lack of hormones is not protective. While no overt metastases form in ovx’d mice, luminal tumor cells can seed distant organs, where they remain dormant as micrometastases and sheltered from therapies but arousable by hormone repletion. This has implications for breast cancer survivors or women with occult disease who are prescribed hormones for contraception or replacement purposes. Electronic supplementary material The online version of this article (doi:10.1186/s13058-014-0489-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ndiya Ogba
- Department of Medicine, University of Colorado Anschutz Medical Campus, 12801 E. 7th Avenue, Aurora, CO, 80045, USA.
| | - Nicole G Manning
- Department of Medicine, University of Colorado Anschutz Medical Campus, 12801 E. 7th Avenue, Aurora, CO, 80045, USA.
| | - Brian S Bliesner
- Department of Medicine, University of Colorado Anschutz Medical Campus, 12801 E. 7th Avenue, Aurora, CO, 80045, USA.
| | - S Kelly Ambler
- Department of Medicine, University of Colorado Anschutz Medical Campus, 12801 E. 7th Avenue, Aurora, CO, 80045, USA.
| | - James M Haughian
- Department of Medicine, University of Colorado Anschutz Medical Campus, 12801 E. 7th Avenue, Aurora, CO, 80045, USA.
| | - Mauricio P Pinto
- Department of Medicine, University of Colorado Anschutz Medical Campus, 12801 E. 7th Avenue, Aurora, CO, 80045, USA.
| | - Paul Jedlicka
- Department of Pathology, University of Colorado Anschutz Medical Campus, 12801 E. 7th Avenue, Aurora, CO, 80045, USA.
| | - Kristiina Joensuu
- Department of Pathology, University of Helsinki, 12801 E. 7th Avenue, Helsinki, 00014, Finland.
| | - Päivi Heikkilä
- Department of Pathology, University of Helsinki, 12801 E. 7th Avenue, Helsinki, 00014, Finland.
| | - Kathryn B Horwitz
- Department of Medicine, University of Colorado Anschutz Medical Campus, 12801 E. 7th Avenue, Aurora, CO, 80045, USA. .,Department of Pathology, University of Colorado Anschutz Medical Campus, 12801 E. 7th Avenue, Aurora, CO, 80045, USA.
| |
Collapse
|
104
|
Sueoka-Aragane N, Sato A, Kobayashi N, Ide M, Yokoo M, Nagano Y, Sueoka E, Okada S, Kimura S. Correlation between plasma DNA and tumor status in an animal model. PLoS One 2014; 9:e111881. [PMID: 25462870 PMCID: PMC4251827 DOI: 10.1371/journal.pone.0111881] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 10/02/2014] [Indexed: 02/08/2023] Open
Abstract
Overcoming metastasis is one of the most important issues with lung cancer. Since metastasis arises through complex steps, a suitable animal model is indispensable for investigation of metastasis. To establish an animal model reflecting human metastatic lung cancers, we used NOD/SCID/Jak3null (NOJ) mice, which exhibit deficiencies in NK cell activity, macrophage and dendritic cell function, and complement activation, as well as T and B cell deficiencies. After screening twenty human lung cancer cell lines through expression patterns of E-cadherin and vimentin according to epithelial mesenchymal transition features, an H1975 cell line carrying EGFR mutations, L858R and T790M, was selected for investigation. Inoculation of the cells into the dorsal flanks caused systemic metastases after one month in lymph nodes, liver, lung, and peritoneum, suggesting that metastases occurred both lymphogenically and hematogenously. We confirmed the existence of H1975 cells in metastatic lesions by detection of T790M and L858R using the mutation-biased PCR and quenching probe (MBP-QP) system previously established in our laboratory. In addition, tumor-derived plasma DNA could be detected using the MBP-QP method. The amount of tumor-derived DNA was associated with tumor volume, whereas an unrelated large amount of tumor-derived DNA was circulating in the presence of metastasis. We present a novel animal model with systemic metastasis with human lung cancer cells. The amount of tumor derived DNA would be related with tumor volume and tumor progression such as metastasis.
Collapse
Affiliation(s)
- Naoko Sueoka-Aragane
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Akemi Sato
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Naomi Kobayashi
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Masaru Ide
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Masako Yokoo
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Yumi Nagano
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Eisaburo Sueoka
- Department of Laboratory Medicine, Saga University Hospital, Saga, Japan
| | - Seiji Okada
- Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | - Shinya Kimura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| |
Collapse
|
105
|
Pettersen EO, Ebbesen P, Gieling RG, Williams KJ, Dubois L, Lambin P, Ward C, Meehan J, Kunkler IH, Langdon SP, Ree AH, Flatmark K, Lyng H, Calzada MJ, Peso LD, Landazuri MO, Görlach A, Flamm H, Kieninger J, Urban G, Weltin A, Singleton DC, Haider S, Buffa FM, Harris AL, Scozzafava A, Supuran CT, Moser I, Jobst G, Busk M, Toustrup K, Overgaard J, Alsner J, Pouyssegur J, Chiche J, Mazure N, Marchiq I, Parks S, Ahmed A, Ashcroft M, Pastorekova S, Cao Y, Rouschop KM, Wouters BG, Koritzinsky M, Mujcic H, Cojocari D. Targeting tumour hypoxia to prevent cancer metastasis. From biology, biosensing and technology to drug development: the METOXIA consortium. J Enzyme Inhib Med Chem 2014; 30:689-721. [PMID: 25347767 DOI: 10.3109/14756366.2014.966704] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 09/15/2014] [Indexed: 01/06/2023] Open
Abstract
The hypoxic areas of solid cancers represent a negative prognostic factor irrespective of which treatment modality is chosen for the patient. Still, after almost 80 years of focus on the problems created by hypoxia in solid tumours, we still largely lack methods to deal efficiently with these treatment-resistant cells. The consequences of this lack may be serious for many patients: Not only is there a negative correlation between the hypoxic fraction in tumours and the outcome of radiotherapy as well as many types of chemotherapy, a correlation has been shown between the hypoxic fraction in tumours and cancer metastasis. Thus, on a fundamental basis the great variety of problems related to hypoxia in cancer treatment has to do with the broad range of functions oxygen (and lack of oxygen) have in cells and tissues. Therefore, activation-deactivation of oxygen-regulated cascades related to metabolism or external signalling are important areas for the identification of mechanisms as potential targets for hypoxia-specific treatment. Also the chemistry related to reactive oxygen radicals (ROS) and the biological handling of ROS are part of the problem complex. The problem is further complicated by the great variety in oxygen concentrations found in tissues. For tumour hypoxia to be used as a marker for individualisation of treatment there is a need for non-invasive methods to measure oxygen routinely in patient tumours. A large-scale collaborative EU-financed project 2009-2014 denoted METOXIA has studied all the mentioned aspects of hypoxia with the aim of selecting potential targets for new hypoxia-specific therapy and develop the first stage of tests for this therapy. A new non-invasive PET-imaging method based on the 2-nitroimidazole [(18)F]-HX4 was found to be promising in a clinical trial on NSCLC patients. New preclinical models for testing of the metastatic potential of cells were developed, both in vitro (2D as well as 3D models) and in mice (orthotopic grafting). Low density quantitative real-time polymerase chain reaction (qPCR)-based assays were developed measuring multiple hypoxia-responsive markers in parallel to identify tumour hypoxia-related patterns of gene expression. As possible targets for new therapy two main regulatory cascades were prioritised: The hypoxia-inducible-factor (HIF)-regulated cascades operating at moderate to weak hypoxia (<1% O(2)), and the unfolded protein response (UPR) activated by endoplasmatic reticulum (ER) stress and operating at more severe hypoxia (<0.2%). The prioritised targets were the HIF-regulated proteins carbonic anhydrase IX (CAIX), the lactate transporter MCT4 and the PERK/eIF2α/ATF4-arm of the UPR. The METOXIA project has developed patented compounds targeting CAIX with a preclinical documented effect. Since hypoxia-specific treatments alone are not curative they will have to be combined with traditional anti-cancer therapy to eradicate the aerobic cancer cell population as well.
Collapse
|
106
|
Drews-Elger K, Iorns E, Dias A, Miller P, Ward TM, Dean S, Clarke J, Campion-Flora A, Rodrigues DN, Reis-Filho JS, Rae JM, Thomas D, Berry D, El-Ashry D, Lippman ME. Infiltrating S100A8+ myeloid cells promote metastatic spread of human breast cancer and predict poor clinical outcome. Breast Cancer Res Treat 2014; 148:41-59. [DOI: 10.1007/s10549-014-3122-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 08/30/2014] [Indexed: 01/08/2023]
|
107
|
Khotskaya YB, Goverdhan A, Shen J, Ponz-Sarvise M, Chang SS, Hsu MC, Wei Y, Xia W, Yu D, Hung MC. S6K1 promotes invasiveness of breast cancer cells in a model of metastasis of triple-negative breast cancer. Am J Transl Res 2014; 6:361-376. [PMID: 25075253 PMCID: PMC4113498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 05/15/2014] [Indexed: 06/03/2023]
Abstract
Breast cancer is the second-leading cause of oncology-related death in US women. Of all invasive breast cancers, patients with tumors lacking expression of the estrogen and progesterone hormone receptors and overexpression of human epidermal growth factor receptor 2 have the poorest clinical prognosis. These referred to as triple-negative breast cancer (TNBC) represent an aggressive form of disease that is marked by early-onset metastasis, high tumor recurrence rate, and low overall survival during the first three years post-diagnosis. In this report, we discuss a novel model of early-onset TNBC metastasis to bone and lungs, derived from MDA-MB-231 cells. Breast cancer cells injected intravenously produced rapid, osteolytic metastases in long bones and spines of athymic nude mice, with concurrent metastasis to lungs, liver, and soft tissues. From the bone metastases, we developed a highly metastatic luciferase-tagged cell line variant named MDA-231-LUC Met. In this report, we demonstrate that the Akt/mTOR/S6K1 axis is hyperactivated in these cells, leading to a dramatic increase in phosphorylation of S6 ribosomal protein at Ser235/236. Lastly, we provide evidence that inhibition of the furthest downstream kinase in the mTOR pathway, S6K1, with a highly specific inhibitor PF-4708671 inhibits cell migration, and thus may provide a potent anti-metastatic adjuvant therapy approach.
Collapse
Affiliation(s)
- Yekaterina B Khotskaya
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas
| | - Aarthi Goverdhan
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas
- The University of Texas Graduate School of Biomedical Sciences at HoustonHouston, Texas
| | - Jia Shen
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas
- The University of Texas Graduate School of Biomedical Sciences at HoustonHouston, Texas
| | - Mariano Ponz-Sarvise
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas
| | - Shih-Shin Chang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas
- The University of Texas Graduate School of Biomedical Sciences at HoustonHouston, Texas
| | - Ming-Chuan Hsu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas
| | - Yongkun Wei
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas
| | - Weiya Xia
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas
| | - Dihua Yu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas
- The University of Texas Graduate School of Biomedical Sciences at HoustonHouston, Texas
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas
- The University of Texas Graduate School of Biomedical Sciences at HoustonHouston, Texas
- Center for Molecular Medicine and Graduate Institute of Cancer Biology, China Medical UniversityTaichung 402, Taiwan
- Department of Biotechnology, Asia UniversityTaichung, Taiwan
| |
Collapse
|
108
|
Xie G, Ji A, Yuan Q, Jin Z, Yuan Y, Ren C, Guo Z, Yao Q, Yang K, Lin X, Chen L. Tumour-initiating capacity is independent of epithelial-mesenchymal transition status in breast cancer cell lines. Br J Cancer 2014; 110:2514-23. [PMID: 24755887 PMCID: PMC4021510 DOI: 10.1038/bjc.2014.153] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 02/24/2014] [Accepted: 02/26/2014] [Indexed: 12/18/2022] Open
Abstract
Background: Epithelial–mesenchymal transition (EMT) and cancer stem cells (CSCs) are considered to be crucial for cancer biology. The purpose of this study was to determine whether EMT directly led to the acquisition of tumour-initiating capacity in breast cancer cell lines. Methods: Epithelial–mesenchymal transition was induced in five breast cancer cell lines and one normal breast cell line by EMT-related cytokine stimulation. Mesenchymal–epithelial transition (MET) was induced by stably overexpressing miR-200c in three mesenchymal-like breast cancer cell lines. Molecular expression and cell function analysis were performed to evaluate the effect of EMT or MET on tumour-initiating capacity and other biological characteristics. Results: The induction of EMT did not enhance tumour-initiating capacity but, instead, conferred a CD44+/CD24−/low phenotype as well as cell proliferation, migration, and resistance to doxorubicin and radiation on breast cancer cell lines. Furthermore, MET did not lead to inhibition or loss of the tumour-initiating capacity in mesenchymal-like breast cancer cell lines, but it markedly attenuated other malignant properties, including proliferation, invasion, and resistance to therapy. Conclusions: Epithelial–mesenchymal transition does not alter tumour-initiating capacity of breast cancer cells but some other biological characteristics. Therefore, EMT and tumour-initiating capacity may not be directly linked in breast cancer cell lines.
Collapse
Affiliation(s)
- G Xie
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - A Ji
- Department of Pharmaceutical Science, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, PR China
| | - Q Yuan
- Jules Stein Eye Institute, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Z Jin
- Department of Pharmacy, Jining First People's Hospital, Jining, Shandong 272111, PR China
| | - Y Yuan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - C Ren
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Z Guo
- Breast Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Q Yao
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - K Yang
- Department of Neurosurgery, Institute of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - X Lin
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - L Chen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| |
Collapse
|
109
|
Hergueta-Redondo M, Sarrió D, Molina-Crespo Á, Megias D, Mota A, Rojo-Sebastian A, García-Sanz P, Morales S, Abril S, Cano A, Peinado H, Moreno-Bueno G. Gasdermin-B promotes invasion and metastasis in breast cancer cells. PLoS One 2014; 9:e90099. [PMID: 24675552 PMCID: PMC3967990 DOI: 10.1371/journal.pone.0090099] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 01/29/2014] [Indexed: 12/21/2022] Open
Abstract
Gasdermin B (GSDMB) belongs to the Gasdermin protein family that comprises four members (GSDMA-D). Gasdermin B expression has been detected in some tumor types such as hepatocarcinomas, gastric and cervix cancers; and its over-expression has been related to tumor progression. At least four splicing isoforms of GSDMB have been identified, which may play differential roles in cancer. However, the implication of GSDMB in carcinogenesis and tumor progression is not well understood. Here, we uncover for the first time the functional implication of GSDMB in breast cancer. Our data shows that high levels of GSDMB expression is correlated with reduced survival and increased metastasis in breast cancer patients included in an expression dataset (>1,000 cases). We demonstrate that GSDMB is upregulated in breast carcinomas compared to normal breast tissue, being the isoform 2 (GSDMB-2) the most differentially expressed. In order to evaluate the functional role of GSDMB in breast cancer two GSDMB isoforms were studied (GSDMB-1 and GSDMB-2). The overexpression of both isoforms in the MCF7 breast carcinoma cell line promotes cell motility and invasion, while its silencing in HCC1954 breast carcinoma cells decreases the migratory and invasive phenotype. Importantly, we demonstrate that both isoforms have a differential role on the activation of Rac-1 and Cdc-42 Rho-GTPases. Moreover, our data support that GSMDB-2 induces a pro-tumorigenic and pro-metastatic behavior in mouse xenograft models as compared to GSDMB-1. Finally, we observed that although both GSDMB isoforms interact in vitro with the chaperone Hsp90, only the GSDMB-2 isoform relies on this chaperone for its stability. Taken together, our results provide for the first time evidences that GSDMB-2 induces invasion, tumor progression and metastasis in MCF7 cells and that GSDMB can be considered as a new potential prognostic marker in breast cancer.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Animals
- Breast Neoplasms/genetics
- Breast Neoplasms/mortality
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/mortality
- Carcinoma, Ductal, Breast/pathology
- Cell Line, Tumor
- Cell Movement/genetics
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Datasets as Topic
- Disease Models, Animal
- Female
- Gelatin/metabolism
- Gene Expression
- Genes, Reporter
- HSP90 Heat-Shock Proteins/metabolism
- Heterografts
- Humans
- Mice
- Middle Aged
- Molecular Imaging
- Neoplasm Grading
- Neoplasm Invasiveness
- Neoplasm Metastasis
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Phenotype
- Prognosis
- Protein Binding
- Proteolysis
- rho GTP-Binding Proteins/metabolism
Collapse
Affiliation(s)
- Marta Hergueta-Redondo
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPAZ, Madrid, Spain
| | - David Sarrió
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPAZ, Madrid, Spain
| | - Ángela Molina-Crespo
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPAZ, Madrid, Spain
| | - Diego Megias
- Centro Nacional de Investigaciones Oncológicas, CNIO, Madrid, Spain
| | - Alba Mota
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPAZ, Madrid, Spain
| | | | | | - Saleta Morales
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPAZ, Madrid, Spain
| | - Sandra Abril
- Hospital MD Anderson Cancer Centre, Madrid, Spain
| | - Amparo Cano
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPAZ, Madrid, Spain
| | - Héctor Peinado
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, Cell and Developmental Biology, Weill Cornell Medical College, New York, New York, United States of America
| | - Gema Moreno-Bueno
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPAZ, Madrid, Spain
- Fundación MD Anderson Internacional, Madrid, Spain
- * E-mail:
| |
Collapse
|
110
|
Drews-Elger K, Brinkman JA, Miller P, Shah SH, Harrell JC, da Silva TG, Ao Z, Schlater A, Azzam DJ, Diehl K, Thomas D, Slingerland JM, Perou CM, Lippman ME, El-Ashry D. Primary breast tumor-derived cellular models: characterization of tumorigenic, metastatic, and cancer-associated fibroblasts in dissociated tumor (DT) cultures. Breast Cancer Res Treat 2014; 144:503-17. [DOI: 10.1007/s10549-014-2887-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 02/14/2014] [Indexed: 12/20/2022]
|
111
|
Chang EW, Gardecki J, Pitman M, Wilsterman EJ, Patel A, Tearney GJ, Iftimia N. Low coherence interferometry approach for aiding fine needle aspiration biopsies. JOURNAL OF BIOMEDICAL OPTICS 2014; 19:116005. [PMID: 25375634 PMCID: PMC4222708 DOI: 10.1117/1.jbo.19.11.116005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 10/13/2014] [Indexed: 05/06/2023]
Abstract
We present portable preclinical low-coherence interference (LCI) instrumentation for aiding fine needle aspiration biopsies featuring the second-generation LCI-based biopsy probe and an improved scoring algorithm for tissue differentiation. Our instrument and algorithm were tested on 38 mice with cultured tumor mass and we show the specificity, sensitivity, and positive predictive value of tumor detection of over 0.89, 0.88, and 0.96, respectively.
Collapse
Affiliation(s)
- Ernest W. Chang
- Physical Sciences, Inc., 20 New England Business Ctr. Drive, Andover, Massachusetts 01810, United States
| | - Joseph Gardecki
- Wellman Center for Photomedicine, Massachusetts General Hospital, 40 Blossom, Boston, Massachusetts 02114, United States
| | - Martha Pitman
- Massachusetts General Hospital, Department of Pathology, 55 Fruit Street, Boston, Massachusetts 02114, United States
| | - Eric J. Wilsterman
- Wellman Center for Photomedicine, Massachusetts General Hospital, 40 Blossom, Boston, Massachusetts 02114, United States
| | - Ankit Patel
- Physical Sciences, Inc., 20 New England Business Ctr. Drive, Andover, Massachusetts 01810, United States
| | - Guillermo J. Tearney
- Wellman Center for Photomedicine, Massachusetts General Hospital, 40 Blossom, Boston, Massachusetts 02114, United States
| | - Nicusor Iftimia
- Physical Sciences, Inc., 20 New England Business Ctr. Drive, Andover, Massachusetts 01810, United States
- Address all correspondence to: Nicusor Iftimia, E-mail:
| |
Collapse
|
112
|
Milsom CC, Lee CR, Hackl C, Man S, Kerbel RS. Differential post-surgical metastasis and survival in SCID, NOD-SCID and NOD-SCID-IL-2Rγ(null) mice with parental and subline variants of human breast cancer: implications for host defense mechanisms regulating metastasis. PLoS One 2013; 8:e71270. [PMID: 23967178 PMCID: PMC3743873 DOI: 10.1371/journal.pone.0071270] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 07/03/2013] [Indexed: 01/06/2023] Open
Abstract
We compare for the first time, the metastatic aggressiveness of the parental MDA-MB-231 breast cancer cell line and two luciferase-tagged in vivo-derived and selected pro-metastatic variants (LM2-4/luc+ and 164/8-1B/luc+) in SCID, NOD-SCID and NOD-SCID-IL-2Rγnull (NSG) mice following orthotopic implantation and primary tumour resection. The variants are known to be more aggressively metastatic in SCID mice, compared to the parental line which has limited spontaneous metastatic competence in these mice. When 2×106 cells were injected into the mammary fat pad, the growth of the resultant primary tumours was identical for the various cell lines in the three strains of mice. However, metastatic spread of all three cell lines, including the MDA-MB-231 parental cell line, was strikingly more aggressive in the highly immunocompromised NSG mice compared to both NOD-SCID and SCID mice, resulting in extensive multi-organ metastases and a significant reduction in overall survival. While these studies were facilitated by monitoring post-surgical spontaneous metastases using whole body bioluminescence imaging, we observed that the luciferase-tagged parental line showed altered growth and diminished metastatic properties compared to its untagged counterpart. Our results are the first to show that host immunity can have a profound impact on the spread of spontaneous visceral metastases and survival following resection of a primary tumour in circumstances where the growth of primary tumours is not similarly affected; as such they highlight the importance of immunity in the metastatic process, and by extension, suggest certain therapeutic strategies that may have a significant impact on reducing metastasis.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Cell Proliferation
- Cell Transformation, Neoplastic
- Female
- Gene Deletion
- Humans
- Luciferases, Firefly/genetics
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/surgery
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Neoplasm Grading
- Neoplasm Metastasis
- Receptors, Interleukin-2/deficiency
- Receptors, Interleukin-2/genetics
- Survival Analysis
Collapse
Affiliation(s)
- Chloe C. Milsom
- Department of Medical Biophysics, University of Toronto and Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- * E-mail: (RSK); (CCM)
| | - Christina R. Lee
- Department of Medical Biophysics, University of Toronto and Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Christina Hackl
- Department of Medical Biophysics, University of Toronto and Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Shan Man
- Department of Medical Biophysics, University of Toronto and Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Robert S. Kerbel
- Department of Medical Biophysics, University of Toronto and Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- * E-mail: (RSK); (CCM)
| |
Collapse
|