151
|
The Role of Tumor Microenvironment and Impact of Cancer Stem Cells on Breast Cancer Progression and Growth. SERBIAN JOURNAL OF EXPERIMENTAL AND CLINICAL RESEARCH 2019. [DOI: 10.2478/sjecr-2018-0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Breast cancer is not only a mass of genetically abnormal tissue in the breast. This is a well-organized system of a complex heterogeneous tissue. Cancer cells produce regulatory signals that stimulate stromal cells to proliferate and migrate; then, stromal elements respond to these signals by releasing components necessary for tumor development that provide structural support, vasculature, and extracellular matrices. Developing tumors can mobilize a variety of cell types from both local and distant niches via secret chemical factors derived from cancer cells themselves or neighboring cells disrupted by growing neoplasm, such as fibroblasts, immune inflammatory cells, and endothelial cells. CSCs are a group of very few cells that are tumorigenic (able to form tumors) and are defined as those cells within a tumor that can self-renew and lead to tumorigenesis. BCSCs represent a small population of cells that have stem cell characteristics and are related to breast cancer. There are different theories about the origin of BCSCs. BCSCs are responsible for breast carcinoma metastasis. Usually, there is a metastatic spread to the bones, and rarely to the lungs and liver. A phenomenon that allows BCSCs to make the transition from epithelial to mesenchymal expression and thus avoid the effect of cytotoxic agents is the epithelial-mesenchymal transition (EMT). During this process, cells change their molecular characteristics in terms of loss of epithelial characteristics taking the mesenchymal phenotype. This process plays a key role in the progression, invasion, and metastasis of breast tumors.
Collapse
|
152
|
Borlinhas F, Loução R, C Conceição R, Ferreira HA. Gamma Distribution Model in the Evaluation of Breast Cancer Through Diffusion-Weighted MRI: A Preliminary Study. J Magn Reson Imaging 2018; 50:230-238. [PMID: 30589146 DOI: 10.1002/jmri.26599] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 11/21/2018] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The gamma distribution (GD) model is based on the statistical distribution of the apparent diffusion coefficient (ADC) parameter. The GD model is expected to reflect the probability of the distribution of water molecule mobility in different regions of tissue, but also the intra- and extracellular diffusion and perfusion components (f1 , f2 , f3 fractions). PURPOSE To assess the GD model in the characterization and diagnostic performance of breast lesions. STUDY TYPE Prospective. POPULATION In all, 48 females with 24 benign and 33 malignant breast lesions. FIELD STRENGTH/SEQUENCE A diffusion-weighted sequence (b = 0-3000 s/mm2 ) with a 3 T scanner. ASSESSMENT For each group of benign, malignant, invasive, and in situ breast lesions, the ADC was obtained. Also, θ and k parameters (scale and shape of the statistic distribution, respectively), f1 , f2 , and f3 fractions were obtained from fitting the GD model to diffusion data. STATISTICAL TESTS Lesion types were compared regarding diffusion parameters using nonparametric statistics and receiver operating characteristic curve diagnostic performance. RESULTS The majority of GD parameters (k, f1 , f2 , f3 fractions) showed significant differences between benign and malignant lesions, and between in situ and invasive lesions (f1 , f2 , f3 fractions) (P ≤ 0.001). The best diagnostic performances were obtained with ADC and f1 fraction in benign vs. malignant lesions (area under curve [AUC] = 0.923 and 0.913, sensitivity = 93.9% and 81.8%, specificity = 79.2% and 91.7%, accuracy = 87.7% and 86.0%, respectively). In invasive lesions vs. in situ lesions, the best diagnostic performance was obtained with f1 fraction, which outperformed ADC results (AUC = 0.978 and 0.941, and sensitivity = 91.3% for both parameters, specificity = 100.0% and 90.0%, accuracy = 93.9% and 90.9%, respectively). DATA CONCLUSION This work shows that the GD model provides information in addition to the ADC parameter, suggesting its potential in the diagnosis of breast lesions. Level of Evidence 2: Technical Efficacy Stage 2 J. Magn. Reson. Imaging 2019;50:230-238.
Collapse
Affiliation(s)
- Filipa Borlinhas
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Ricardo Loução
- Institute of Neuroscience and Medicine (INM - 4), Forschungszentrum Jülich, Jülich, Germany
| | - Raquel C Conceição
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Hugo A Ferreira
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| |
Collapse
|
153
|
Sinha VC, Piwnica-Worms H. Intratumoral Heterogeneity in Ductal Carcinoma In Situ: Chaos and Consequence. J Mammary Gland Biol Neoplasia 2018; 23:191-205. [PMID: 30194658 PMCID: PMC6934090 DOI: 10.1007/s10911-018-9410-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/30/2018] [Indexed: 02/06/2023] Open
Abstract
Ductal carcinoma in situ (DCIS) is a non-invasive proliferative growth in the breast that serves as a non-obligate precursor to invasive ductal carcinoma. The widespread adoption of screening mammography has led to a steep increase in the detection of DCIS, which now comprises approximately 20% of new breast cancer diagnoses in the United States. Interestingly, the intratumoral heterogeneity (ITH) that has been observed in invasive breast cancers may have been established early in tumorigenesis, given the vast and varied ITH that has been detected in DCIS. This review will discuss the intratumoral heterogeneity of DCIS, focusing on the phenotypic and genomic heterogeneity of tumor cells, as well as the compositional heterogeneity of the tumor microenvironment. In addition, we will assess the spatial heterogeneity that is now being appreciated in these lesions, and summarize new approaches to evaluate heterogeneity of tumor and stromal cells in the context of their spatial organization. Importantly, we will discuss how a growing understanding of ITH has led to a more holistic appreciation of the complex biology of DCIS, specifically its evolution and natural history. Finally, we will consider ways in which our knowledge of DCIS ITH might be translated in the future to guide clinical care for DCIS patients.
Collapse
Affiliation(s)
- Vidya C Sinha
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Houston, TX, 77030, USA
| | - Helen Piwnica-Worms
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Houston, TX, 77030, USA.
| |
Collapse
|
154
|
Wang Y, Shi W, Kuss M, Mirza S, Qi D, Krasnoslobodtsev A, Zeng J, Band H, Band V, Duan B. 3D Bioprinting of Breast Cancer Models for Drug Resistance Study. ACS Biomater Sci Eng 2018; 4:4401-4411. [PMID: 33418833 DOI: 10.1021/acsbiomaterials.8b01277] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Ying Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, People’s Republic of China
| | | | | | | | - Dianjun Qi
- Department of General Practice, The First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, People’s Republic of China
| | - Alexey Krasnoslobodtsev
- Department of Physics, University of Nebraska at Omaha, 6001 Dodge Street, Omaha 68182, Nebraska, United States
| | - Jiping Zeng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, People’s Republic of China
| | | | | | | |
Collapse
|
155
|
Wang X, Jiao X, Meng Y, Chen H, Griffin N, Gao X, Shan F. Methionine enkephalin (MENK) inhibits human gastric cancer through regulating tumor associated macrophages (TAMs) and PI3K/AKT/mTOR signaling pathway inside cancer cells. Int Immunopharmacol 2018; 65:312-322. [PMID: 30343258 DOI: 10.1016/j.intimp.2018.10.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/09/2018] [Accepted: 10/15/2018] [Indexed: 02/07/2023]
Abstract
This study was to explore the effect and mechanisms of anti- human gastric cancer by MENK in vitro and in vivo. The results showed in MENK-treated xenograft tissue, the percentage of M2-type macrophages decreased while M1-type macrophages increased. MENK increased the expression of M1-related cytokine TNF-α and attenuated the expression of M2-related cytokine IL-10 expression. MENK upregulated the expression of opioid receptor (OGFr), while it inhibited HGC27 and SGC7901 cells through blocking PI3K/AKT/mTOR signal pathway in vitro and in vivo. These effects of MENK could be cancelled when OGFr was knockdown. This indicates that binding to OGFr by MENK appears to be essential for the anti- GC cells. Therefore, it is concluded that MENK might skew macrophage toward M2 phenotype from M1 phenotype within tumor and induce cells apoptosis though blocking OGFr/PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Xiaonan Wang
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Xue Jiao
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Yiming Meng
- Central Laboratory, Cancer Hospital of China Medical University, Shenyang 110042, China
| | - Hao Chen
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Noreen Griffin
- Immune Therapeutics, Inc., 37 North Orange Avenue, Suite 607, Orlando, FL 32801, USA
| | - Xinghua Gao
- Department of Dermatology, No. 1 Teaching Hospital, China Medical University, Shenyang, 110016, China
| | - Fengping Shan
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang 110122, China.
| |
Collapse
|
156
|
Althobiti M, Aleskandarany MA, Joseph C, Toss M, Mongan N, Diez-Rodriguez M, Nolan CC, Ashankyty I, Ellis IO, Green AR, Rakha EA. Heterogeneity of tumour-infiltrating lymphocytes in breast cancer and its prognostic significance. Histopathology 2018; 73:887-896. [DOI: 10.1111/his.13695] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 06/25/2018] [Indexed: 02/04/2023]
Affiliation(s)
- Maryam Althobiti
- Nottingham Breast Cancer Research Centre; Division of Cancer and Stem Cells; School of Medicine; University of Nottingham; Nottingham City Hospital; Nottingham UK
| | - Mohammed A Aleskandarany
- Nottingham Breast Cancer Research Centre; Division of Cancer and Stem Cells; School of Medicine; University of Nottingham; Nottingham City Hospital; Nottingham UK
| | - Chitra Joseph
- Nottingham Breast Cancer Research Centre; Division of Cancer and Stem Cells; School of Medicine; University of Nottingham; Nottingham City Hospital; Nottingham UK
| | - Michael Toss
- Nottingham Breast Cancer Research Centre; Division of Cancer and Stem Cells; School of Medicine; University of Nottingham; Nottingham City Hospital; Nottingham UK
| | - Nigel Mongan
- School of Veterinary Medicine and Science; University of Nottingham; Sutton Bonington UK
- Department of Pharmacology; Weill Cornell Medicine; New York NY USA
| | - Maria Diez-Rodriguez
- Nottingham Breast Cancer Research Centre; Division of Cancer and Stem Cells; School of Medicine; University of Nottingham; Nottingham City Hospital; Nottingham UK
| | - Christopher C Nolan
- Nottingham Breast Cancer Research Centre; Division of Cancer and Stem Cells; School of Medicine; University of Nottingham; Nottingham City Hospital; Nottingham UK
| | - Ibraheem Ashankyty
- Molecular Diagnostics and Personalised Therapeutics Unit; University of Ha'il; Ha'il Saudi Arabia
| | - Ian O Ellis
- Nottingham Breast Cancer Research Centre; Division of Cancer and Stem Cells; School of Medicine; University of Nottingham; Nottingham City Hospital; Nottingham UK
| | - Andrew R Green
- Nottingham Breast Cancer Research Centre; Division of Cancer and Stem Cells; School of Medicine; University of Nottingham; Nottingham City Hospital; Nottingham UK
| | - Emad A Rakha
- Nottingham Breast Cancer Research Centre; Division of Cancer and Stem Cells; School of Medicine; University of Nottingham; Nottingham City Hospital; Nottingham UK
| |
Collapse
|
157
|
Pseudopterosin Inhibits Proliferation and 3D Invasion in Triple-Negative Breast Cancer by Agonizing Glucocorticoid Receptor Alpha. Molecules 2018; 23:molecules23081992. [PMID: 30103404 PMCID: PMC6222322 DOI: 10.3390/molecules23081992] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 08/07/2018] [Accepted: 08/08/2018] [Indexed: 12/18/2022] Open
Abstract
Pseudopterosin, produced by the sea whip of the genus Antillogorgia, possesses a variety of promising biological activities, including potent anti-inflammatory effects. However, few studies examined pseudopterosin in the treatment of cancer cells and, to our knowledge, the ability to inhibit triple-negative breast cancer (TNBC) proliferation or invasion has not been explored. Thus, we evaluated the as-yet unknown mechanism of action of pseudopterosin: Pseudopterosin was able to inhibit proliferation of TNBC. Interestingly, analyzing breast cancer cell proliferation after knocking down glucocorticoid receptor α (GRα) revealed that the antiproliferative effects of pseudopterosin were significantly inhibited when GRα expression was reduced. Furthermore, pseudopterosin inhibited the invasion of MDA-MB-231 3D tumor spheroids embedded in an extracellular-like matrix. Remarkably, the knockdown of GRα in 3D tumor spheroids revealed increased ability of cells to invade the surrounding matrix. In a coculture, encompassing peripheral blood mononuclear cells (PBMC) and MDA-MB-231 cells, and the production of interleukin 6 (IL-6) and interleukin 8 (IL-8) significantly increased compared to a monoculture. Notably, pseudopterosin indicated to block cytokine elevation, representing key players in tumor progression in the coculture. Thus, our results reveal pseudopterosin treatment as a potential novel approach in TNBC therapy.
Collapse
|
158
|
The Role of Desmoplasia and Stromal Fibroblasts on Anti-cancer Drug Resistance in a Microengineered Tumor Model. Cell Mol Bioeng 2018; 11:419-433. [PMID: 31719892 DOI: 10.1007/s12195-018-0544-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 07/19/2018] [Indexed: 01/21/2023] Open
Abstract
Introduction Cancer associated fibroblasts (CAFs) are known to participate in anti-cancer drug resistance by upregulating desmoplasia and pro-survival mechanisms within the tumor microenvironment. In this regard, anti-fibrotic drugs (i.e., tranilast) have been repurposed to diminish the elastic modulus of the stromal matrix and reduce tumor growth in presence of chemotherapeutics (i.e., doxorubicin). However, the quantitative assessment on impact of these stromal targeting drugs on matrix stiffness and tumor progression is still missing in the sole presence of CAFs. Methods We developed a high-density 3D microengineered tumor model comprised of MDA-MB-231 (highly invasive breast cancer cells) embedded microwells, surrounded by CAFs encapsulated within collagen I hydrogel. To study the influence of tranilast and doxorubicin on fibrosis, we probed the matrix using atomic force microscopy (AFM) and assessed matrix protein deposition. We further studied the combinatorial influence of the drugs on cancer cell proliferation and invasion. Results Our results demonstrated that the combinatorial action of tranilast and doxorubicin significantly diminished the stiffness of the stromal matrix compared to the control. The two drugs in synergy disrupted fibronectin assembly and reduced collagen fiber density. Furthermore, the combination of these drugs, condensed tumor growth and invasion. Conclusion In this work, we utilized a 3D microengineered model to tease apart the role of tranilast and doxorubicin in the sole presence of CAFs on desmoplasia, tumor growth and invasion. Our study lay down a ground work on better understanding of the role of biomechanical properties of the matrix on anti-cancer drug efficacy in the presence of single class of stromal cells.
Collapse
|
159
|
Zhong H, Daoud A, Han J, An X, Qiao C, Duan L, Wang Y, Chen Z, Zhou J, Shang J. A Small β-Carboline Derivative "B-9-3" Modulates TGF-β Signaling Pathway Causing Tumor Regression in Vivo. Front Pharmacol 2018; 9:788. [PMID: 30079021 PMCID: PMC6063040 DOI: 10.3389/fphar.2018.00788] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 06/28/2018] [Indexed: 11/13/2022] Open
Abstract
Targeting tumor microenvironment (TME) is crucial in order to overcome the anti-cancer therapy resistance. In this study, we report the antitumor activity of a newly synthesized β-carboline derivative “B-9-3.” Here, this small molecule showed a promising antitumor activity in vivo along with an enhanced immune response as reflected by a reduction of regulatory T cells and increased CD4+/CD8+ T cells. Further, B-9-3 decreased the number of myofibroblasts not only in the tumor but also in the lung suggesting an anti-metastatic action. The reduction of myofibroblasts was associated with lower expression of epithelial-to-mesenchymal transition markers and a decrease of phosphorylated SMAD2/3 complex indicating the implication of TGF-β signaling pathway in B-9-3’s effect. The blockade of myofibroblasts induction by B-9-3 was also verified in vitro in human fibroblasts treated with TGF-β. To elucidate the mechanism of B-9-3’s action on TGF-β pathway, first, we investigated the molecular interaction between B-9-3 and TGF-β receptors using docking method. Data showed a weak interaction of B-9-3 with the ATP-binding pocket of TGFβRI but a strong one with a ternary complex formed of extracellular domains of TGFβRI, TGFβRII, and TGF-β. In addition, the role of TGFβRI and TGFβRII in B-9-3’s activity was explored in vitro. B-9-3 did not decrease any of the two receptors’ protein level and only reduced phosphorylated SMAD2/3 suggesting that its effect was more probably due to its interaction with the ternary complex rather than decreasing the expression of TGF-β receptors or interfering with their ATP-binding domains. B-9-3 is a small active molecule which acts on the TGF-β signaling pathway and improves the TME to inhibit the proliferation and the metastasis of the tumor with the potential for clinical application.
Collapse
Affiliation(s)
- Hui Zhong
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Abdelkader Daoud
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jichun Han
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiaohong An
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Caili Qiao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Lanlan Duan
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yichuan Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhenfeng Chen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin, China
| | - Jia Zhou
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jing Shang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
160
|
Qin H, Liu L, Sun S, Zhang D, Sheng J, Li B, Yang W. The impact of PI3K inhibitors on breast cancer cell and its tumor microenvironment. PeerJ 2018; 6:e5092. [PMID: 29942710 PMCID: PMC6014315 DOI: 10.7717/peerj.5092] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 06/05/2018] [Indexed: 12/13/2022] Open
Abstract
The phosphoinositide 3-kinase (PI3K) pathway shows frequent aberrant alterations and pathological activation in breast cancer cells. While PI3K inhibitors have not achieved expectant therapeutic efficacy in clinical trials, and several studies provide promising combination strategies to substantially maximize therapeutic outcomes. Besides its direct impact on regulating cancer cells survival, PI3K inhibitors are also demonstrated to have an immunomodulatory impact based on the tumor microenvironment. Inhibition of the leukocyte-enriched PI3K isoforms may break immune tolerance and restore cytotoxic T cell activity by reprogramming the tumor microenvironment. In addition, PI3K inhibitors have pleiotropic effects on tumor angiogenesis and even induce tumor vascular normalization. In this review, we discuss the mechanism of PI3K inhibitor suppression of breast cancer cells and modulation of the tumor microenvironment in order to provide further thoughts for breast cancer treatment.
Collapse
Affiliation(s)
- Hanjiao Qin
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Linlin Liu
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Shu Sun
- Affiliated Hospital of Changchun University Of Traditional Chinese Medicine, Changchun, Jilin, China
| | - Dan Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Jiyao Sheng
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Wei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
161
|
Yang D, Li Y, Xing L, Tan Y, Sun J, Zeng B, Xiang T, Tan J, Ren G, Wang Y. Utilization of adipocyte-derived lipids and enhanced intracellular trafficking of fatty acids contribute to breast cancer progression. Cell Commun Signal 2018; 16:32. [PMID: 29914512 PMCID: PMC6006729 DOI: 10.1186/s12964-018-0221-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 02/22/2018] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND To determine whether adipocyte-derived lipids could be transferred into breast cancer cells and investigate the underlying mechanisms of subsequent lipolysis and fatty acid trafficking in breast cancer cells. METHODS A Transwell co-culture system was used in which human breast cancer cells were cultured in the absence or presence of differentiated murine 3 T3-L1 adipocytes. Migration/invasion and proliferation abilities were compared between breast cancer cells that were cultivated alone and those co-cultivated with mature adipocytes. The ability of lipolysis in breast cancer cells were measured, as well as the expression of the rate-limiting lipase ATGL and fatty acid transporter FABP5. ATGL and FABP5 were then ablated to investigate their impact on the aggressiveness of breast cancer cells that were surrounded by adipocytes. Further, immunohistochemistry was performed to detect differential expression of ATGL and FABP5 in breast cancer tissue sections. RESULTS The migration and invasion abilities of cancer cells were significantly enhanced after co-culture with adipocytes, accompanied by elevated lipolysis and expression of ATGL and FABP5. Abrogation of ATGL and FABP5 sharply attenuated the malignancy of co-cultivated breast cancer cells. However, this phenomenon was not observed if a lipid emulsion was added to the culture medium to substitute for adipocytes. Furthermore, epithelial-mesenchymal transaction was induced in co-cultivated breast cancer cells. That may partially due to the stimulation of PPARβ/δ and MAPK, which was resulted from upregulation of FABP5. As evidenced by immunohistochemistry, ATGL and FABP5 also had higher expression levels at the invasive front of the breast tumor, in where the adipocytes abound, compared to the central area in tissue specimens. CONCLUSIONS Lipid originating from tumor-surrounding adipocytes could be transferred into breast cancer cells. Adipocyte-cancer cell crosstalk rather than lipids alone induced upregulation of lipases and fatty acid transport protein in cancer cells to utilize stored lipids for tumor progression. The increased expression of the key lipase ATGL and intracellular fatty acid trafficking protein FABP5 played crucial roles in this process via fueling or signaling.
Collapse
Affiliation(s)
- Dejuan Yang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yunhai Li
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Lei Xing
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yiqing Tan
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jiazheng Sun
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Beilei Zeng
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Tingxiu Xiang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jinxiang Tan
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Guosheng Ren
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China. .,Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Yuanyuan Wang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China. .,Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
162
|
Gyamfi J, Lee YH, Eom M, Choi J. Interleukin-6/STAT3 signalling regulates adipocyte induced epithelial-mesenchymal transition in breast cancer cells. Sci Rep 2018; 8:8859. [PMID: 29891854 PMCID: PMC5995871 DOI: 10.1038/s41598-018-27184-9] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 05/22/2018] [Indexed: 01/08/2023] Open
Abstract
The tumour microenvironment is a key regulators of tumour progression through the secretion of growth factors that activate epithelial-mesenchymal transition (EMT). Induction of EMT is a key step for transition from a benign state to a metastatic tumour. Adipose tissue forms a bulk portion of the breast cancer microenvironment, emerging evidence indicates the potential for adipocytes to influence tumour progression through the secretion of adipokines that can induce EMT. The molecular mechanisms underlying how adipocytes enhance breast cancer progression is largely unknown. We hypothesized that paracrine signalling by adipocytes can activate EMT and results in increased migration and invasion characteristics of breast cancer cells. We found that IL-6 secreted by adipocytes induce EMT in breast cancer cells. The effect of IL-6 expression on EMT is mediated through activation of the signal transducer and activated of transcription 3 (STAT3). Blocking of IL-6 signalling in breast cancer cells and adipocytes, decreased proliferation, migration and invasion capabilities and altered the expression of genes regulating EMT. Together, our results suggest that matured human adipocytes can enhance the aggressive behaviour of breast cancer cells and induce an EMT-phenotype through paracrine IL-6/STAT3 signalling.
Collapse
Affiliation(s)
- Jones Gyamfi
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Korea
| | - Yun-Hee Lee
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Korea
| | - Minseob Eom
- Department of Pathology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Junjeong Choi
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Korea.
| |
Collapse
|
163
|
Immune oncology, immune responsiveness and the theory of everything. J Immunother Cancer 2018; 6:50. [PMID: 29871670 PMCID: PMC5989400 DOI: 10.1186/s40425-018-0355-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 05/11/2018] [Indexed: 12/12/2022] Open
Abstract
Anti-cancer immunotherapy is encountering its own checkpoint. Responses are dramatic and long lasting but occur in a subset of tumors and are largely dependent upon the pre-existing immune contexture of individual cancers. Available data suggest that three landscapes best define the cancer microenvironment: immune-active, immune-deserted and immune-excluded. This trichotomy is observable across most solid tumors (although the frequency of each landscape varies depending on tumor tissue of origin) and is associated with cancer prognosis and response to checkpoint inhibitor therapy (CIT). Various gene signatures (e.g. Immunological Constant of Rejection - ICR and Tumor Inflammation Signature - TIS) that delineate these landscapes have been described by different groups. In an effort to explain the mechanisms of cancer immune responsiveness or resistance to CIT, several models have been proposed that are loosely associated with the three landscapes. Here, we propose a strategy to integrate compelling data from various paradigms into a “Theory of Everything”. Founded upon this unified theory, we also propose the creation of a task force led by the Society for Immunotherapy of Cancer (SITC) aimed at systematically addressing salient questions relevant to cancer immune responsiveness and immune evasion. This multidisciplinary effort will encompass aspects of genetics, tumor cell biology, and immunology that are pertinent to the understanding of this multifaceted problem.
Collapse
|
164
|
Feng Y, Spezia M, Huang S, Yuan C, Zeng Z, Zhang L, Ji X, Liu W, Huang B, Luo W, Liu B, Lei Y, Du S, Vuppalapati A, Luu HH, Haydon RC, He TC, Ren G. Breast cancer development and progression: Risk factors, cancer stem cells, signaling pathways, genomics, and molecular pathogenesis. Genes Dis 2018; 5:77-106. [PMID: 30258937 PMCID: PMC6147049 DOI: 10.1016/j.gendis.2018.05.001] [Citation(s) in RCA: 739] [Impact Index Per Article: 105.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 05/08/2018] [Indexed: 12/14/2022] Open
Abstract
As the most commonly occurring cancer in women worldwide, breast cancer poses a formidable public health challenge on a global scale. Breast cancer consists of a group of biologically and molecularly heterogeneous diseases originated from the breast. While the risk factors associated with this cancer varies with respect to other cancers, genetic predisposition, most notably mutations in BRCA1 or BRCA2 gene, is an important causative factor for this malignancy. Breast cancers can begin in different areas of the breast, such as the ducts, the lobules, or the tissue in between. Within the large group of diverse breast carcinomas, there are various denoted types of breast cancer based on their invasiveness relative to the primary tumor sites. It is important to distinguish between the various subtypes because they have different prognoses and treatment implications. As there are remarkable parallels between normal development and breast cancer progression at the molecular level, it has been postulated that breast cancer may be derived from mammary cancer stem cells. Normal breast development and mammary stem cells are regulated by several signaling pathways, such as estrogen receptors (ERs), HER2, and Wnt/β-catenin signaling pathways, which control stem cell proliferation, cell death, cell differentiation, and cell motility. Furthermore, emerging evidence indicates that epigenetic regulations and noncoding RNAs may play important roles in breast cancer development and may contribute to the heterogeneity and metastatic aspects of breast cancer, especially for triple-negative breast cancer. This review provides a comprehensive survey of the molecular, cellular and genetic aspects of breast cancer.
Collapse
Affiliation(s)
- Yixiao Feng
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, Departments of General Surgery, Clinical Laboratory Medicine, Orthopaedic Surgery, Plastic Surgery and Burn, and Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Mia Spezia
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Shifeng Huang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, Departments of General Surgery, Clinical Laboratory Medicine, Orthopaedic Surgery, Plastic Surgery and Burn, and Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Chengfu Yuan
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Biochemistry and Molecular Biology, China Three Gorges University School of Medicine, Yichang 443002, China
| | - Zongyue Zeng
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Linghuan Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Xiaojuan Ji
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Wei Liu
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, Departments of General Surgery, Clinical Laboratory Medicine, Orthopaedic Surgery, Plastic Surgery and Burn, and Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Bo Huang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
- Department of Clinical Laboratory Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Wenping Luo
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing 401147, China
| | - Bo Liu
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, Departments of General Surgery, Clinical Laboratory Medicine, Orthopaedic Surgery, Plastic Surgery and Burn, and Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Yan Lei
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, Departments of General Surgery, Clinical Laboratory Medicine, Orthopaedic Surgery, Plastic Surgery and Burn, and Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Scott Du
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Student Inquiry Research Program, Illinois Mathematics and Science Academy (IMSA), Aurora, IL 60506, USA
| | - Akhila Vuppalapati
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Student Inquiry Research Program, Illinois Mathematics and Science Academy (IMSA), Aurora, IL 60506, USA
| | - Hue H. Luu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Rex C. Haydon
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Guosheng Ren
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, Departments of General Surgery, Clinical Laboratory Medicine, Orthopaedic Surgery, Plastic Surgery and Burn, and Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
165
|
Ghosh A, Sarkar S, Banerjee S, Behbod F, Tawfik O, McGregor D, Graff S, Banerjee SK. MIND model for triple-negative breast cancer in syngeneic mice for quick and sequential progression analysis of lung metastasis. PLoS One 2018; 13:e0198143. [PMID: 29813119 PMCID: PMC5973560 DOI: 10.1371/journal.pone.0198143] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 05/14/2018] [Indexed: 12/25/2022] Open
Abstract
Mouse models of breast cancer with specific molecular subtypes (e.g., ER or HER2 positive) in an immunocompetent or an immunocompromised environment significantly contribute to our understanding of cancer biology, despite some limitations, and they give insight into targeted therapies. However, an ideal triple-negative breast cancer (TNBC) mouse model is lacking. What has been missing in the TNBC mouse model is a sequential progression of the disease in an essential native microenvironment. This notion inspired us to develop a TNBC-model in syngeneic mice using a mammary intraductal (MIND) method. To achieve this goal, Mvt-1and 4T1 TNBC mouse cell lines were injected into the mammary ducts via nipples of FVB/N mice and BALB/c wild-type immunocompetent mice, respectively. We established that the TNBC-MIND model in syngeneic mice could epitomize all breast cancer progression stages and metastasis into the lungs via lymphatic or hematogenous dissemination within four weeks. Collectively, the syngeneic mouse-TNBC-MIND model may serve as a unique platform for further investigation of the underlying mechanisms of TNBC growth and therapies.
Collapse
Affiliation(s)
- Arnab Ghosh
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri, United States of America
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Sandipto Sarkar
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri, United States of America
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Snigdha Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri, United States of America
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Fariba Behbod
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Ossama Tawfik
- Saint Luke’s Hospital of Kansas City, Kansas City, Missouri, United States of America
| | - Douglas McGregor
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri, United States of America
- Pathology Department, VA Medical Center, Kansas City, Missouri, United States of America
| | - Stephanie Graff
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri, United States of America
- Sarah Cannon Cancer Center at HCA Midwest Health, Overland Park, Kansas, United States of America
| | - Sushanta K. Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri, United States of America
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- * E-mail: ,
| |
Collapse
|
166
|
Morgan MM, Livingston MK, Warrick JW, Stanek EM, Alarid ET, Beebe DJ, Johnson BP. Mammary fibroblasts reduce apoptosis and speed estrogen-induced hyperplasia in an organotypic MCF7-derived duct model. Sci Rep 2018; 8:7139. [PMID: 29740030 PMCID: PMC5940820 DOI: 10.1038/s41598-018-25461-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 04/13/2018] [Indexed: 12/22/2022] Open
Abstract
The estrogen receptor (ER) regulates the survival and growth of breast cancer cells, but it is less clear how components of the tissue microenvironment affect ER-mediated responses. We set out to test how human mammary fibroblasts (HMFs) modulate ER signaling and downstream cellular responses. We exposed an organotypic mammary model consisting of a collagen-embedded duct structure lined with MCF7 cells to 17-β estradiol (E2), with and without HMFs in the surrounding matrix. MCF7 cells grown as ductal structures were polarized and proliferated at rates comparable to in vivo breast tissue. In both culture platforms, exposure to E2 increased ER transactivation, increased proliferation, and induced ductal hyperplasia. When the surrounding matrix contained HMFs, the onset and severity of E2-induced ductal hyperplasia was increased due to decreased apoptosis. The reduced apoptosis may be due to fibroblasts modulating ER signaling in MCF7 cells, as suggested by the increased ER transactivation and reduced ER protein in MCF7 cells grown in co-culture. These findings demonstrate the utility of organotypic platforms when studying stromal:epithelial interactions, and add to existing literature that implicate the mammary microenvironment in ER + breast cancer progression.
Collapse
Affiliation(s)
- Molly M Morgan
- Department of Biomedical Engineering, University of Wisconsin Madison, Madison, Wisconsin, USA
| | - Megan K Livingston
- Department of Biomedical Engineering, University of Wisconsin Madison, Madison, Wisconsin, USA.,Department of Chemistry, University of Wisconsin Madison, Madison, Wisconsin, USA
| | - Jay W Warrick
- Department of Biomedical Engineering, University of Wisconsin Madison, Madison, Wisconsin, USA
| | - Eli M Stanek
- Department of Biomedical Engineering, University of Wisconsin Madison, Madison, Wisconsin, USA
| | - Elaine T Alarid
- Department of Oncology, University of Wisconsin Madison, Madison, Wisconsin, USA
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin Madison, Madison, Wisconsin, USA.
| | - Brian P Johnson
- Department of Biomedical Engineering, University of Wisconsin Madison, Madison, Wisconsin, USA.
| |
Collapse
|
167
|
Zins K, Heller G, Mayerhofer M, Schreiber M, Abraham D. Differential prognostic impact of interleukin-34 mRNA expression and infiltrating immune cell composition in intrinsic breast cancer subtypes. Oncotarget 2018; 9:23126-23148. [PMID: 29796177 PMCID: PMC5955405 DOI: 10.18632/oncotarget.25226] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 04/05/2018] [Indexed: 12/14/2022] Open
Abstract
Interleukin-34 (IL-34) is a ligand for the CSF-1R and has also two additional receptors, PTPRZ1 and syndecan-1. IL-34 plays a role in innate immunity, inflammation, and cancer. However, the role of IL-34 in breast cancer is still ill-defined. We analyzed IL-34 mRNA expression in breast cancer cell lines and breast cancer patients and applied established computational approaches (CIBERSORT, ESTIMATE, TIMER, TCIA), to analyze gene expression data from The Cancer Genome Atlas (TCGA). Expression of IL-34 was associated with a favorable prognosis in luminal and HER2 but not basal breast cancer patients. Gene expression of CSF-1 and CSF-1R was strongly associated with myeloid cell infiltration, while we found no or only weak correlations between IL-34, PTPRZ1, syndecan-1 and myeloid cells. In vitro experiments showed that tyrosine phosphorylation of CSF-1R, ERK, and FAK and cell migration are differentially regulated by IL-34 and CSF-1 in breast cancer cell lines. Collectively, our data suggest that correlation of IL-34 gene expression with survival is dependent on the molecular breast cancer subtype. Furthermore, IL-34 is not associated with myeloid cell infiltration and directly regulates breast cancer cell migration and signaling.
Collapse
Affiliation(s)
- Karin Zins
- Division of Cell and Developmental Biology, Center for Anatomy and Cell Biology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Gerwin Heller
- Department of Medicine I, Clinical Division of Oncology, Medical University of Vienna, A-1090 Vienna, Austria.,Comprehensive Cancer Center Vienna, A-1090 Vienna, Austria
| | - Mathias Mayerhofer
- Division of Cell and Developmental Biology, Center for Anatomy and Cell Biology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Martin Schreiber
- Department of Obstetrics and Gynecology, Medical University of Vienna, A-1090 Vienna, Austria.,Comprehensive Cancer Center Vienna, A-1090 Vienna, Austria
| | - Dietmar Abraham
- Division of Cell and Developmental Biology, Center for Anatomy and Cell Biology, Medical University of Vienna, A-1090 Vienna, Austria.,Comprehensive Cancer Center Vienna, A-1090 Vienna, Austria
| |
Collapse
|
168
|
Logun M, Zhao W, Mao L, Karumbaiah L. Microfluidics in Malignant Glioma Research and Precision Medicine. ADVANCED BIOSYSTEMS 2018; 2:1700221. [PMID: 29780878 PMCID: PMC5959050 DOI: 10.1002/adbi.201700221] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Indexed: 01/09/2023]
Abstract
Glioblastoma multiforme (GBM) is an aggressive form of brain cancer that has no effective treatments and a prognosis of only 12-15 months. Microfluidic technologies deliver microscale control of fluids and cells, and have aided cancer therapy as point-of-care devices for the diagnosis of breast and prostate cancers. However, a few microfluidic devices are developed to study malignant glioma. The ability of these platforms to accurately replicate the complex microenvironmental and extracellular conditions prevailing in the brain and facilitate the measurement of biological phenomena with high resolution and in a high-throughput manner could prove useful for studying glioma progression. These attributes, coupled with their relatively simple fabrication process, make them attractive for use as point-of-care diagnostic devices for detection and treatment of GBM. Here, the current issues that plague GBM research and treatment, as well as the current state of the art in glioma detection and therapy, are reviewed. Finally, opportunities are identified for implementing microfluidic technologies into research and diagnostics to facilitate the rapid detection and better therapeutic targeting of GBM.
Collapse
Affiliation(s)
- Meghan Logun
- Regenerative Bioscience Center, ADS Complex, University of Georgia, 425 River Road, Athens, GA 30602-2771, USA
| | - Wujun Zhao
- Department of Chemistry, University of Georgia, Athens, GA 30602-2771, USA
| | - Leidong Mao
- School of Electrical and Computer Engineering, College of Engineering, University of Georgia, Athens, GA 30602-2771, USA
| | - Lohitash Karumbaiah
- Regenerative Bioscience Center, ADS Complex, University of Georgia, 425 River Road, Athens, GA 30602-2771, USA
| |
Collapse
|
169
|
Kumari A, Silakari O, Singh RK. Recent advances in colony stimulating factor-1 receptor/c-FMS as an emerging target for various therapeutic implications. Biomed Pharmacother 2018; 103:662-679. [PMID: 29679908 DOI: 10.1016/j.biopha.2018.04.046] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 04/06/2018] [Accepted: 04/06/2018] [Indexed: 12/25/2022] Open
Abstract
Colony stimulating factor-1 (CSF-1) is one of the most common proinflammatory cytokine responsible for various inflammatory disorders. It has a remarkable role in the development and progression of osteoarthritis, cancer and other autoimmune disease conditions. The CSF-1 acts by binding to the receptor, called colony stimulating factor-1 receptor (CSF-1R) also known as c-FMS resulting in the cascade of signalling pathway causing cell proliferation and differentiation. Interleukin-34 (IL-34), recently identified as another ligand for CSF-IR, is a cytokine protein. Both, CSF-1 and IL-34, although two distinct cytokines, follow the similar signalling pathway on binding to the same receptor, CSF-1R. Like CSF-1, IL-34 promotes the differentiation and survival of monocyte, macrophages and osteoclasts. This CSF-1R/c-FMS is over expressed in many cancers and on tumour associated macrophages, consequently, have been exploited as a drug target for promising treatment for cancer and inflammatory diseases. Some CSF-1R/c-FMS inhibitors such as ABT-869, Imatinib, AG013736, JNJ-40346527, PLX3397, DCC-3014 and Ki20227 have been successfully used in these disease conditions. Many c-FMS inhibitors have been the candidates of clinical trials, but suffer from some side effects like cardiotoxicity, vomiting, swollen eyes, diarrhoea, etc. If selectivity of cFMS inhibition is achieved successfully, side effects can be overruled and this approach may become a novel therapy for treatment of various therapeutic interventions. Thus, successful targeting of c-FMS may result in multifunctional therapy. With this background of information, the present review focuses on the recent developments in the area of CSF-1R/c-FMS inhibitors with emphasis on crystal structure, mechanism of action and various therapeutic implications in which c-FMS plays a pivotal role. The review on structure activity relationship of various compounds acting as the inhibitors of c-FMS which gives the selection criteria for the development of novel molecules is also being presented.
Collapse
Affiliation(s)
- Archana Kumari
- Rayat-Bahra Institute of Pharmacy, Dist. Hoshiarpur, 146104, Punjab, India
| | - Om Silakari
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, 147002, Patiala, India
| | - Rajesh K Singh
- Department of Pharmaceutical Chemistry, Shivalik College of Pharmacy, Nangal, Dist. Rupnagar, 140126, Punjab, India.
| |
Collapse
|
170
|
Zielinska HA, Holly JMP, Bahl A, Perks CM. Inhibition of FASN and ERα signalling during hyperglycaemia-induced matrix-specific EMT promotes breast cancer cell invasion via a caveolin-1-dependent mechanism. Cancer Lett 2018; 419:187-202. [PMID: 29331414 PMCID: PMC5832758 DOI: 10.1016/j.canlet.2018.01.028] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 12/25/2017] [Accepted: 01/08/2018] [Indexed: 12/11/2022]
Abstract
Since disturbed metabolic conditions such as obesity and diabetes can be critical determinants of breast cancer progression and therapeutic failure, we aimed to determine the mechanism responsible for their pro-oncogenic effects. Using non-invasive, epithelial-like ERα-positive MCF-7 and T47D human breast cancer cells we found that hyperglycaemia induced epithelial to mesenchymal transition (EMT), a key programme responsible for the development of metastatic disease. This was demonstrated by loss of the epithelial marker E-cadherin together with increases in mesenchymal markers such as vimentin, fibronectin and the transcription factor SLUG, together with an enhancement of cell growth and invasion. These phenotypic changes were only observed with cells grown on fibronectin and not with those plated on collagen. Analyzing metabolic parameters, we found that hyperglycaemia-induced, matrix-specific EMT promoted the Warburg effect by upregulating glucose uptake, lactate release and specific glycolytic enzymes and transporters. We showed that silencing of fatty acid synthase (FASN) and the downstream ERα, which we showed previously to mediate hyperglycaemia-induced chemoresistance in these cells, resulted in suppression of cell growth: however, this also resulted in a dramatic enhancement of cell invasion and SLUG mRNA levels via a novel caveolin-1-dependent mechanism.
Collapse
Affiliation(s)
- H A Zielinska
- IGFs & Metabolic Endocrinology Group, School of Clinical Sciences, University of Bristol, Learning and Research Building, Southmead Hospital, Bristol BS10 5NB, UK.
| | - J M P Holly
- IGFs & Metabolic Endocrinology Group, School of Clinical Sciences, University of Bristol, Learning and Research Building, Southmead Hospital, Bristol BS10 5NB, UK
| | - A Bahl
- Department of Clinical Oncology, Bristol Haematology and Oncology Centre, University Hospitals Bristol, Bristol, UK
| | - C M Perks
- IGFs & Metabolic Endocrinology Group, School of Clinical Sciences, University of Bristol, Learning and Research Building, Southmead Hospital, Bristol BS10 5NB, UK
| |
Collapse
|
171
|
Kuol N, Stojanovska L, Apostolopoulos V, Nurgali K. Role of the Nervous System in Tumor Angiogenesis. CANCER MICROENVIRONMENT 2018; 11:1-11. [PMID: 29502307 DOI: 10.1007/s12307-018-0207-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 02/19/2018] [Indexed: 12/12/2022]
Abstract
The development of cancer involves an intricate process, wherein many identified and unidentified factors play a role. Tumor angiogenesis, growth of new blood vessels, is one of the major prerequisites for tumor growth as tumor cells rely on adequate oxygen and nutrient supply as well as the removal of waste products. Growth factors including VEGF orchestrate the development of angiogenesis. In addition, nervous system via the release of neurotransmitters contributes to tumor angiogenesis. The nervous system governs functional activities of many organs, and, as tumors are not independent organs within an organism, this system is integrally involved in tumor growth and progression via regulating tumor angiogenesis. Various neurotransmitters have been reported to play an important role in tumor angiogenesis.
Collapse
Affiliation(s)
- Nyanbol Kuol
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, Australia
| | - Lily Stojanovska
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, Australia
| | - Vasso Apostolopoulos
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, Australia
| | - Kulmira Nurgali
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, Australia. .,Department of Medicine Western Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Regenerative Medicine and Stem Cells Program, Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, Australia.
| |
Collapse
|
172
|
IMP1, an mRNA binding protein that reduces the metastatic potential of breast cancer in a mouse model. Oncotarget 2018; 7:72662-72671. [PMID: 27655671 PMCID: PMC5341935 DOI: 10.18632/oncotarget.12083] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 09/02/2016] [Indexed: 12/22/2022] Open
Abstract
Cells that are able to localize β-actin mRNA efficiently have decreased metastatic potential. Invasive carcinoma cells derived from primary mammary tumors have reduced levels of an RNA binding protein IMP1/ZBP1/IGF2BP1, required for β-actin mRNA localization. We showed previously that in human breast carcinoma cells in vitro, this protein suppresses invasion. In this work we examined whether its re-expression can suppress breast cancer metastasis in a breast cancer mouse model. We developed a mouse conditionally expressing IMP1-GFP (hereinafter referred to as the IMP1 transgene) specifically in the mammary gland of a PYMT breast cancer mouse. We found that mice conditionally expressing the IMP1 transgene showed little or no metastases to the lungs from the primary tumor in contrast to PYMT mice not expressing IMP1, which uniformly develop metastases at an early stage.
Collapse
|
173
|
Nakasone ES, Hurvitz SA, McCann KE. Harnessing the immune system in the battle against breast cancer. Drugs Context 2018; 7:212520. [PMID: 29456568 PMCID: PMC5810622 DOI: 10.7573/dic.212520] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/16/2018] [Accepted: 01/17/2018] [Indexed: 12/14/2022] Open
Abstract
Breast cancer is the most prevalent malignancy in women and the second most common cause of cancer-related death worldwide. Despite major innovations in early detection and advanced therapeutics, up to 30% of women with node-negative breast cancer and 70% of women with node-positive breast cancer will develop recurrence. The recognition that breast tumors are infiltrated by a complex array of immune cells that influence their development, progression, and metastasis, as well as their responsiveness to systemic therapies has sparked major interest in the development of immunotherapies. In fact, not only the native host immune system can be altered to promote potent antitumor response, but also its components can be manipulated to generate effective therapeutic strategies. We present here a review of the major approaches to immunotherapy in breast cancers, both successes and failures, as well as new therapies on the horizon.
Collapse
Affiliation(s)
- Elizabeth S Nakasone
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Sara A Hurvitz
- Division of Hematology/Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Kelly E McCann
- Division of Hematology/Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
174
|
Gyamfi J, Eom M, Koo JS, Choi J. Multifaceted Roles of Interleukin-6 in Adipocyte-Breast Cancer Cell Interaction. Transl Oncol 2018; 11:275-285. [PMID: 29413760 PMCID: PMC5884177 DOI: 10.1016/j.tranon.2017.12.009] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 12/18/2017] [Accepted: 12/18/2017] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is the most common malignancy in women worldwide, with a developmental process spanning decades. The malignant cells recruit a variety of cells including fibroblasts, endothelial cells, immune cells, and adipocytes, creating the tumor microenvironment. The tumor microenvironment has emerged as active participants in breast cancer progression and response to treatment through autocrine and paracrine interaction with the malignant cells. Adipose tissue is abundant in the breast cancer microenvironment; interactions with cancer cells create cancer-associated adipocytes which produce a variety of adipokines that influence breast cancer initiation, metastasis, angiogenesis, and cachexia. Interleukin (IL)-6 has emerged as key compound significantly produced by breast cancer cells and adipocytes, with the potential of inducing proliferation, epithelial-mesenchymal phenotype, stem cell phenotype, angiogenesis, cachexia, and therapeutic resistance in breast cancer cells. Our aim is to present a brief knowledge of IL-6’s role in breast cancer. This review summarizes our current understanding of the breast microenvironment, with emphasis on adipocytes as key players in breast cancer tumorigenesis. The effects of key adipocytes such as leptin, adipokines, TGF-b, and IL-6 are discussed. Finally, we discuss the role of IL-6 in various aspects of cancer progression.
Collapse
Affiliation(s)
- Jones Gyamfi
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Korea
| | - Minseob Eom
- Department of Pathology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Ja-Seung Koo
- Department of Pathology, Yonsei University College of Medicine.
| | - Junjeong Choi
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Korea.
| |
Collapse
|
175
|
Mueller C, Haymond A, Davis JB, Williams A, Espina V. Protein biomarkers for subtyping breast cancer and implications for future research. Expert Rev Proteomics 2018; 15:131-152. [PMID: 29271260 PMCID: PMC6104835 DOI: 10.1080/14789450.2018.1421071] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Breast cancer subtypes are currently defined by a combination of morphologic, genomic, and proteomic characteristics. These subtypes provide a molecular portrait of the tumor that aids diagnosis, prognosis, and treatment escalation/de-escalation options. Gene expression signatures describing intrinsic breast cancer subtypes for predicting risk of recurrence have been rapidly adopted in the clinic. Despite the use of subtype classifications, many patients develop drug resistance, breast cancer recurrence, or therapy failure. Areas covered: This review provides a summary of immunohistochemistry, reverse phase protein array, mass spectrometry, and integrative studies that are revealing differences in biological functions within and between breast cancer subtypes. We conclude with a discussion of rigor and reproducibility for proteomic-based biomarker discovery. Expert commentary: Innovations in proteomics, including implementation of assay guidelines and standards, are facilitating refinement of breast cancer subtypes. Proteomic and phosphoproteomic information distinguish biologically functional subtypes, are predictive of recurrence, and indicate likelihood of drug resistance. Actionable, activated signal transduction pathways can now be quantified and characterized. Proteomic biomarker validation in large, well-designed studies should become a public health priority to capitalize on the wealth of information gleaned from the proteome.
Collapse
Affiliation(s)
- Claudius Mueller
- a Center for Applied Proteomics and Molecular Medicine , George Mason University , Manassas , VA , USA
| | - Amanda Haymond
- a Center for Applied Proteomics and Molecular Medicine , George Mason University , Manassas , VA , USA
| | - Justin B Davis
- a Center for Applied Proteomics and Molecular Medicine , George Mason University , Manassas , VA , USA
| | - Alexa Williams
- a Center for Applied Proteomics and Molecular Medicine , George Mason University , Manassas , VA , USA
| | - Virginia Espina
- a Center for Applied Proteomics and Molecular Medicine , George Mason University , Manassas , VA , USA
| |
Collapse
|
176
|
Todorović-Raković N, Radulovic M, Vujasinović T, Milovanović J, Nikolić-Vukosavljević D. The time-dependent prognostic value of intratumoral cytokine expression profiles in a natural course of primary breast cancer with a long-term follow-up. Cytokine 2018; 102:12-17. [PMID: 29245048 DOI: 10.1016/j.cyto.2017.11.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 11/27/2017] [Accepted: 11/29/2017] [Indexed: 01/05/2023]
Abstract
Despite the increasing evidence for the importance of immunity in breast cancer, the contradictory role of inflammation has not been thoroughly researched. In this study, we investigate the prognostic value of intratumoral inflammation as evaluated by cytokine mRNA levels. Intratumoral mRNA was measured for IL1β, IL6, IL8, IL10 and IL17A, using Taqman quantitative PCR. By the AUC criteria, none of the cytokines associated with metastasis outcome over the entire follow-up period. However, separation of the follow-up period has revealed a time-dependent and robust prognostic association of IL β. It discriminated between patients with and without metastasis relapse by AUCs of 0.21 and 0.82 during the early and late follow-up of 0-7 and 7-14 years, respectively. Interestingly, the prognostic effect by IL1β shifted during follow-up from good prognosis in the first seven years to bad prognosis thereafter. By the less stringent criteria of Cox regression analysis, other cytokines also significantly associated positively or negatively with metastasis outcome. IL17A associated with good prognosis in the first 7 years of follow up while IL6 associated with poor and IL10 with good prognosis from 7 to 14 years. The revealed time-dependent prognostic effects of cytokine mRNA levels are intriguing and may reflect valuable biological information which should be considered in breast cancer immunotherapy research.
Collapse
Affiliation(s)
- Nataša Todorović-Raković
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia.
| | - Marko Radulovic
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Tijana Vujasinović
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Jelena Milovanović
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia
| | | |
Collapse
|
177
|
Kuol N, Stojanovska L, Apostolopoulos V, Nurgali K. Crosstalk between cancer and the neuro-immune system. J Neuroimmunol 2018; 315:15-23. [DOI: 10.1016/j.jneuroim.2017.12.016] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 12/18/2017] [Accepted: 12/18/2017] [Indexed: 02/07/2023]
|
178
|
Reznik E, Luna A, Aksoy BA, Liu EM, La K, Ostrovnaya I, Creighton CJ, Hakimi AA, Sander C. A Landscape of Metabolic Variation across Tumor Types. Cell Syst 2018; 6:301-313.e3. [PMID: 29396322 DOI: 10.1016/j.cels.2017.12.014] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 08/21/2017] [Accepted: 12/17/2017] [Indexed: 12/29/2022]
Abstract
Tumor metabolism is reorganized to support proliferation in the face of growth-related stress. Unlike the widespread profiling of changes to metabolic enzyme levels in cancer, comparatively less attention has been paid to the substrates/products of enzyme-catalyzed reactions, small-molecule metabolites. We developed an informatic pipeline to concurrently analyze metabolomics data from over 900 tissue samples spanning seven cancer types, revealing extensive heterogeneity in metabolic changes relative to normal tissue across cancers of different tissues of origin. Despite this heterogeneity, a number of metabolites were recurrently differentially abundant across many cancers, such as lactate and acyl-carnitine species. Through joint analysis of metabolomic data alongside clinical features of patient samples, we also identified a small number of metabolites, including several polyamines and kynurenine, which were associated with aggressive tumors across several tumor types. Our findings offer a glimpse onto common patterns of metabolic reprogramming across cancers, and the work serves as a large-scale resource accessible via a web application (http://www.sanderlab.org/pancanmet).
Collapse
Affiliation(s)
- Ed Reznik
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| | - Augustin Luna
- cBio Center, Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Bülent Arman Aksoy
- Genetics and Genomics Department, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Eric Minwei Liu
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Konnor La
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, NY 10065, USA; Tri-Institutional Training Program in Computational Biology & Medicine, New York, NY 10065, USA
| | - Irina Ostrovnaya
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Chad J Creighton
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - A Ari Hakimi
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Chris Sander
- cBio Center, Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Computational Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
179
|
Kuol N, Stojanovska L, Apostolopoulos V, Nurgali K. Role of the nervous system in cancer metastasis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:5. [PMID: 29334991 PMCID: PMC5769535 DOI: 10.1186/s13046-018-0674-x] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 12/30/2017] [Indexed: 12/20/2022]
Abstract
Cancer remains as one of the leading cause of death worldwide. The development of cancer involves an intricate process, wherein many identified and unidentified factors play a role. Although most studies have focused on the genetic abnormalities which initiate and promote cancer, there is overwhelming evidence that tumors interact within their environment by direct cell-to-cell contact and with signaling molecules, suggesting that cancer cells can influence their microenvironment and bidirectionally communicate with other systems. However, only in recent years the role of the nervous system has been recognized as a major contributor to cancer development and metastasis. The nervous system governs functional activities of many organs, and, as tumors are not independent organs within an organism, this system is integrally involved in tumor growth and progression.
Collapse
Affiliation(s)
- Nyanbol Kuol
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, Australia
| | - Lily Stojanovska
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, Australia
| | - Vasso Apostolopoulos
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, Australia
| | - Kulmira Nurgali
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, Australia. .,Department of Medicine, Western Health, The University of Melbourne, Regenerative Medicine and Stem Cells Program, AIMSS, Melbourne, Australia.
| |
Collapse
|
180
|
He Y, Yu D, Zhu L, Zhong S, Zhao J, Tang J. miR-149 in Human Cancer: A Systemic Review. J Cancer 2018; 9:375-388. [PMID: 29344284 PMCID: PMC5771345 DOI: 10.7150/jca.21044] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 11/21/2017] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that regulate post-transcriptional gene expression via binding to the 3'-untranslated region (3'-UTR) of targeted mRNAs. They are reported to play important roles in tumorigenesis and progression of various cancers. Among them, miR-149 was confirmed to be aberrantly regulated in various tumors. In this review, we provide a complex overview of miR-149, particularly summarize the critical roles of it in cancers and expect to lay the foundation for future works on this important microRNA.
Collapse
Affiliation(s)
- Yunjie He
- The First Clinical School of Nanjing Medical University, Nanjing 210029, P.R. China
| | - Dandan Yu
- Center of Clinical Laboratory, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Baiziting 42, Nanjing 210009, P.R. China
| | - Lingping Zhu
- The First Clinical School of Nanjing Medical University, Nanjing 210029, P.R. China
| | - Shanliang Zhong
- Center of Clinical Laboratory, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Baiziting 42, Nanjing 210009, P.R. China
| | - Jianhua Zhao
- Center of Clinical Laboratory, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Baiziting 42, Nanjing 210009, P.R. China
| | - Jinhai Tang
- The First Clinical School of Nanjing Medical University, Nanjing 210029, P.R. China.,Department of General Surgery, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, P.R. China
| |
Collapse
|
181
|
Zeng AQ, Yu Y, Yao YQ, Yang FF, Liao M, Song LJ, Li YL, Yu Y, Li YJ, Deng YL, Yang SP, Zeng CJ, Liu P, Xie YM, Yang JL, Zhang YW, Ye TH, Wei YQ. Betulinic acid impairs metastasis and reduces immunosuppressive cells in breast cancer models. Oncotarget 2017; 9:3794-3804. [PMID: 29423083 PMCID: PMC5790500 DOI: 10.18632/oncotarget.23376] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 11/27/2017] [Indexed: 02/05/2023] Open
Abstract
Breast cancer is the most common female cancer with considerable metastatic potential, explaining the need for new candidates that inhibit tumor metastasis. In our study, betulinic acid (BA), a kind of pentacyclic triterpenoid compound derived from birch trees, was evaluated for its anti-metastasis activity in vitro and in vivo. BA decreased the viability of three breast cancer cell lines and markedly impaired cell migration and invasion. In addition, BA could inhibit the activation of stat3 and FAK which resulted in a reduction of matrix metalloproteinases (MMPs), and increase of the MMPs inhibitor (TIMP-2) expression. Moreover, in our animal experiment, intraperitoneal administration of 10 mg/kg/day BA suppressed 4T1 tumor growth and blocked formation of pulmonary metastases without obvious side effects. Furthermore, histological and immunohistochemical analyses showed a decrease in MMP-9 positive cells, MMP-2 positive cells and Ki-67 positive cells and an increase in cleaved caspase-3 positive cells upon BA administration. Notably, BA reduced the number of myeloid-derived suppressor cells (MDSCs) in the lungs and tumors. Interestingly, in our caudal vein model, BA also obviously suppressed 4T1 tumor pulmonary metastases. These findings suggested that BA might be a potential agent for inhibiting the growth and metastasis of breast cancer.
Collapse
Affiliation(s)
- An-Qi Zeng
- Laboratory of Liver Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Yan Yu
- Laboratory of Liver Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Yu-Qin Yao
- Laboratory of Liver Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Fang-Fang Yang
- Laboratory of Liver Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Mengya Liao
- Sichuan Nursing Vocational College, Chengdu 610100, China
| | - Lin-Jiang Song
- Laboratory of Liver Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Ya-Li Li
- Laboratory of Liver Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Yang Yu
- Department of Peritoneal Cancer Surgery, Beijing Shijitan Hospital Affiliated to the Capital Medical University, Beijing 100038, China
| | - Yu-Jue Li
- Laboratory of Liver Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Yuan-Le Deng
- Laboratory of Liver Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Shu-Ping Yang
- Laboratory of Liver Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Chen-Juan Zeng
- Laboratory of Liver Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.,Sichuan Scientist Biotechnology Co., Ltd, Chengdu 610041, China
| | - Ping Liu
- Department of Gynecology and Obstetrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second Hospital, Sichuan University, Chengdu 610041, China
| | - Yong-Mei Xie
- Laboratory of Liver Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Jin-Liang Yang
- Laboratory of Liver Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Yi-Wen Zhang
- Laboratory of Liver Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Ting-Hong Ye
- Laboratory of Liver Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Yu-Quan Wei
- Laboratory of Liver Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| |
Collapse
|
182
|
Singh R, Dagar P, Pal S, Basu B, Shankar BS. Significant alterations of the novel 15 gene signature identified from macrophage-tumor interactions in breast cancer. Biochim Biophys Acta Gen Subj 2017; 1862:669-683. [PMID: 29248526 DOI: 10.1016/j.bbagen.2017.12.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 11/09/2017] [Accepted: 12/11/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Tumor microenvironment is composed of a largely altered extracellular matrix with different cell types. The complex interplay between macrophages and tumor cells through several soluble factors and signaling is an important factor in breast cancer progression. METHODS We have extended our earlier studies on monocyte and macrophage conditioned medium (MϕCM) and have carried out proteomic analysis to identify its constituents as well as validation. The 8-gene signature identified through macrophage-breast cancer cell interactions was queried in cBioportal for bioinformatic analyses. RESULTS Proteomic analysis (MALDI-TOF and LC-MS/MS) revealed integrin and matrix metalloproteinases in MϕCM which activated TGF-β1, IL-6, TGF- βRII and EGFR as well as its downstream STAT and SMAD signaling in breast cancer cells. Neutralization of pro-inflammatory cytokines (TNF-α. Il-1β, IL-6) abrogated the MϕCM induced migration but invasion to lesser extent. The 8- gene signature identified by macrophage-tumor interactions (TNF-α, IL-1β, IL-6, MMP1, MMP9, TGF-β1, TGF-βRII, EGFR) significantly co-occurred with TP53 mutation, WTAPP1 deletion and SLC12A5 amplification along with differential expression of PSAT1 and ESR1 at the mRNA level and TPD52and PRKCD at the protein level in TCGA (cBioportal). Together these genes form a novel 15 gene signature which is altered in 63.6% of TCGA (1105 samples) data and was associated with high risk and poor survival (p<0.05) in many breast cancer datasets (SurvExpress). CONCLUSIONS These results highlight the importance of macrophage signaling in breast cancer and the prognostic role of the15-gene signature. GENERAL SIGNIFICANCE Our study may facilitate novel prognostic markers based on tumor-macrophage interaction.
Collapse
Affiliation(s)
- Rajshri Singh
- Radiation Biology & Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Mumbai 400 085, India; HomiBhabha National Institute, Anushaktinagar, Mumbai 400 094, India
| | - Priya Dagar
- Radiation Biology & Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Mumbai 400 085, India
| | - Shyama Pal
- Radiation Biology & Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Mumbai 400 085, India
| | - Bhakti Basu
- HomiBhabha National Institute, Anushaktinagar, Mumbai 400 094, India; Molecular Biology Division, Bhabha Atomic Research Centre, Mumbai 400085, India
| | - Bhavani S Shankar
- Radiation Biology & Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Mumbai 400 085, India; HomiBhabha National Institute, Anushaktinagar, Mumbai 400 094, India.
| |
Collapse
|
183
|
Cancer immune resistance: can theories converge? Emerg Top Life Sci 2017; 1:411-419. [PMID: 33525800 PMCID: PMC7289003 DOI: 10.1042/etls20170060] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 11/07/2017] [Accepted: 11/07/2017] [Indexed: 12/13/2022]
Abstract
Immune oncology (IO) is challenged to expand its usefulness to a broader range of cancers. A second generation of IO agents acting beyond the realm of Checkpoint Inhibitor Therapy (CIT) is sought with the intent of turning immune-resistant cancers into appealing IO targets. The published literature proposes a profusion of models to explain cancer immune resistance to CIT that largely outnumber the immune landscapes and corresponding resistance mechanisms. In spite of the complex and contradicting models suggested to explain refractoriness to CIT, the identification of prevailing mechanisms and their targeting may not be as daunting as it at first appears. Here, we suggest that cancer cells go through a conserved evolutionary bottleneck facing a Two-Option Choice to evade recognition by the immune competent host: they can either adopt a clean oncogenic process devoid of immunogenic stimuli (immune-silent tumors) or display an entropic biology prone to immune recognition (immune-active tumors) but resilient to rejection thanks to the recruitment of compensatory immune suppressive processes. Strategies aimed at enhancing the effectiveness of CIT will be different according to the immune landscape targeted.
Collapse
|
184
|
He Y, Deng F, Yang S, Wang D, Chen X, Zhong S, Zhao J, Tang J. Exosomal microRNA: a novel biomarker for breast cancer. Biomark Med 2017; 12:177-188. [PMID: 29151358 DOI: 10.2217/bmm-2017-0305] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Breast cancer is the most common invasive malignancy among females worldwide. microRNAs (miRNAs) are small noncoding RNAs that regulate post-transcriptional gene expression by binding to the 3'-untranslated regions of targeted mRNAs. Recently, exosomes have been recognized to play critical roles in breast cancer. miRNAs, as the most important inclusions in exosomes, are stable in the blood and other body fluids, making them ideal candidate biomarkers. In this review, we provide a complex overview of exosomal miRNAs in breast cancer. Particularly, we summarized their critical roles in tumorigenesis and tumor progression, expecting to lay the foundation for future studies using these potential biomarkers.
Collapse
Affiliation(s)
- Yunjie He
- The First Clinical School of Nanjing Medical University, Nanjing 210029, P.R. China
| | - Fei Deng
- The First Clinical School of Nanjing Medical University, Nanjing 210029, P.R. China
| | - Sujin Yang
- The First Clinical School of Nanjing Medical University, Nanjing 210029, P.R. China
| | - Dandan Wang
- Department of General Surgery, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, P.R. China
| | - Xiu Chen
- Department of General Surgery, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, P.R. China
| | - Shanliang Zhong
- China Center of Clinical Laboratory, Nanjing Medical University Affiliated Cancer Hospital, Cancer Institute of Jiangsu Province, Baiziting 42, Nanjing 210009, P.R. China
| | - Jianhua Zhao
- China Center of Clinical Laboratory, Nanjing Medical University Affiliated Cancer Hospital, Cancer Institute of Jiangsu Province, Baiziting 42, Nanjing 210009, P.R. China
| | - Jinhai Tang
- The First Clinical School of Nanjing Medical University, Nanjing 210029, P.R. China.,Department of General Surgery, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, P.R. China
| |
Collapse
|
185
|
Runa F, Hamalian S, Meade K, Shisgal P, Gray PC, Kelber JA. Tumor microenvironment heterogeneity: challenges and opportunities. ACTA ACUST UNITED AC 2017; 3:218-229. [PMID: 29430386 DOI: 10.1007/s40610-017-0073-7] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The tumor microenvironment (TME) has been recognized as an integral component of malignancies in breast and prostate tissues, contributing in confounding ways to tumor progression, metastasis, therapy resistance and disease recurrence. Major components of the TME are immune cells, fibroblasts, pericytes, endothelial cells, mesenchymal stroma/stem cells (MSCs), and extracellular matrix (ECM) components. Herein, we discuss the molecular and cellular heterogeneity within the TME and how this presents unique challenges and opportunities for treating breast and prostate cancers.
Collapse
Affiliation(s)
- F Runa
- Department of Biology, California State University, Northridge, CA
| | - S Hamalian
- Department of Biology, California State University, Northridge, CA
| | - K Meade
- Department of Biology, California State University, Northridge, CA
| | - P Shisgal
- Department of Biology, California State University, Northridge, CA
| | - P C Gray
- The Salk Institute for Biological Studies, La Jolla, CA
| | - J A Kelber
- Department of Biology, California State University, Northridge, CA
| |
Collapse
|
186
|
Thompson C, Keck K, Hielscher A. Isolation of Intact, Whole Mouse Mammary Glands for Analysis of Extracellular Matrix Expression and Gland Morphology. J Vis Exp 2017. [PMID: 29155742 DOI: 10.3791/56512] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The goal of this procedure was to harvest the #4 abdominal mammary glands from female nulliparous mice in order to assess ECM expression and ductal architecture. Here, a small pocket below the skin was created using Mayo scissors, allowing separation of the glands within the subcutaneous tissue from the underlying peritoneum. Visualization of the glands was aided by the use of 3.5x-R surgical micro loupes. The pelt was inverted and pinned back allowing identification of the intact mammary fat pads. Each of the #4 abdominal glands was bluntly dissected by sliding the scalpel blade laterally between the subcutaneous layer and the glands. Immediately post-harvest, glands were placed in 10% neutral buffered formalin for subsequent tissue processing. Excision of the entire gland is advantageous because it primarily eliminates the risk of excluding important tissue-wide interactions between ductal epithelial cells and other microenvironmental cellular populations that could be missed in a partial biopsy. One drawback of the methodology is the use of serial sections from fixed tissues which limits analyses of ductal morphogenesis and protein expression to discrete locations within the gland. As such, changes in ductal architecture and protein expression in 3 dimensions (3D) is not readily obtainable. Overall, the technique is applicable to studies requiring whole intact murine mammary glands for downstream investigations such as developmental ductal morphogenesis or breast cancer.
Collapse
Affiliation(s)
- Christopher Thompson
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center
| | | | | |
Collapse
|
187
|
Chiang CF, Chao TT, Su YF, Hsu CC, Chien CY, Chiu KC, Shiah SG, Lee CH, Liu SY, Shieh YS. Metformin-treated cancer cells modulate macrophage polarization through AMPK-NF-κB signaling. Oncotarget 2017; 8:20706-20718. [PMID: 28157701 PMCID: PMC5400538 DOI: 10.18632/oncotarget.14982] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 01/24/2017] [Indexed: 01/09/2023] Open
Abstract
Accumulating evidence is indicating metformin to possess the potential ability in preventing tumor development and suppressing cancer growth. However, the exact mechanism of its antitumorigenic effects is still not clear. We found that metformin suppressed the ability of cancer to skew macrophage toward M2 phenotype. Metformin treated cancer cells increased macrophage expression of M1-related cytokines IL-12 and TNF-α and attenuated M2-related cytokines IL-8, IL-10, and TGF-β expression. Furthermore, metformin treated cancer cells displayed inhibited secretion of IL-4, IL-10 and IL-13; cytokines important for inducing M2 macrophages. Conversely, M1 inducing cytokine IFN-γ was upper-regulated in cancer cells. Additionally, through increasing AMPK and p65 phosphorylation, metformin treatment activated AMPK-NF-κB signaling of cancer cells that participate in regulating M1 and M2 inducing cytokines expression. Moreover, Compound C, an AMPK inhibitor, significantly increased IL-4, IL-10, and IL-13 expression while BAY-117082, an NF-κB inhibitor, decreased expression. In metformin-treated tumor tissue, the percentage of M2-like macrophages decreased while M1-like macrophages increased. These findings suggest that metformin activates cancer AMPK-NF-κB signaling, a pathway involved in regulating M1/M2 expression and inducing genes for macrophage polarization to anti-tumor phenotype.
Collapse
Affiliation(s)
- Chi-Fu Chiang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Ting-Ting Chao
- Medical Research Center, Cardinal Tien Hospital, School of Medicine, New Taipei City, Taiwan
| | - Yu-Fu Su
- Department of Radiation Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chia-Chen Hsu
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Chu-Yen Chien
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Kuo-Chou Chiu
- Department of Dentistry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shine-Gwo Shiah
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Chien-Hsing Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shyun-Yeu Liu
- Department of Oral and Maxillofacial Surgery, Chi Mei Medical Center, Tainan, Taiwan
| | - Yi-Shing Shieh
- Department of Dentistry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
188
|
Yang F, Aubele M, Walch A, Gross E, Napieralski R, Zhao S, Ahmed N, Kiechle M, Reuning U, Dorn J, Sweep F, Magdolen V, Schmitt M. Tissue kallikrein-related peptidase 4 (KLK4), a novel biomarker in triple-negative breast cancer. Biol Chem 2017; 398:1151-1164. [DOI: 10.1515/hsz-2017-0122] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 04/12/2017] [Indexed: 12/22/2022]
Abstract
AbstractTriple-negative breast cancer (TNBC), lacking the steroid hormone receptors ER and PR and the oncoprotein HER2, is characterized by its aggressive pattern and insensitivity to endocrine and HER2-directed therapy. Human kallikrein-related peptidases KLK1-15 provide a rich source of serine protease-type biomarkers associated with tumor growth and cancer progression for a variety of malignant diseases. In this study, recombinant KLK4 protein was generated and affinity-purified KLK4-directed polyclonal antibody pAb587 established to allow localization of KLK4 protein expression in tumor cell lines and archived formalin-fixed, paraffin-embedded TNBC tumor tissue specimens. For this, KLK4 protein expression was assessed by immunohistochemistry in primary tumor tissue sections (tissue microarrays) of 188 TNBC patients, mainly treated with anthracycline- or CMF-based polychemotherapy. KLK4 protein is localized in the cytoplasm of tumor and stroma cells. In this patient cohort, elevated stroma cell KLK4 expression, but not tumor cell KLK4 expression, is predictive for poor disease-free survival by univariate analysis (hazard ratio: 2.26,p=0.001) and multivariable analysis (hazard ratio: 2.12,p<0.01). Likewise, univariate analysis revealed a trend for statistical significance of elevated KLK4 stroma cell expression for overall survival of TNBC patients as well.
Collapse
|
189
|
Wada M, Canals D, Adada M, Coant N, Salama MF, Helke KL, Arthur JS, Shroyer KR, Kitatani K, Obeid LM, Hannun YA. P38 delta MAPK promotes breast cancer progression and lung metastasis by enhancing cell proliferation and cell detachment. Oncogene 2017; 36:6649-6657. [PMID: 28783172 PMCID: PMC5746050 DOI: 10.1038/onc.2017.274] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 06/30/2017] [Accepted: 07/03/2017] [Indexed: 12/18/2022]
Abstract
The protein p38 mitogen-activated protein kinase delta isoform (p38δ) is a poorly studied member of the MAPK family. Data analysis from The Cancer Genome Atlas (TCGA) database revealed that p38δ is highly expressed in all types of human breast cancers. Using a human breast cancer tissue array, we confirmed elevation in cancer tissue. The breast cancer mouse model, MMTV-PyMT (PyMT), developed breast tumors with lung metastasis; however, mice deleted in p38δ (PyMT/p38δ−/−) exhibited delayed primary tumor formation and highly reduced lung metastatic burden. At the cellular level, we demonstrate that targeting of p38δ in breast cancer cells, MCF-7 and MDA-MB-231 resulted in a reduced rate of cell proliferation. Additionally, cells lacking p38δ also displayed an increased cell-matrix adhesion and reduced cell detachment. This effect on cell adhesion was molecularly supported by the regulation of the focal adhesion kinase (FAK) by p38δ in the human breast cell lines. These studies define a previously unappreciated role for p38δ in breast cancer development and evolution by regulating tumor growth and altering metastatic properties.
Collapse
Affiliation(s)
- M Wada
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - D Canals
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - M Adada
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - N Coant
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - M F Salama
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Faculty of Veterinary Medicine, Department of Biochemistry, Mansoura University, Mansoura, Egypt
| | - K L Helke
- Department of Comparative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - J S Arthur
- MRC Protein Phosphorylation Unit, School of Life Sciences, University of Dundee, Dundee, UK
| | - K R Shroyer
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | - K Kitatani
- Tohoku Medical Megabank Organization and Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - L M Obeid
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA.,Northport VA Medical Center, Northport, NY, USA
| | - Y A Hannun
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
190
|
Jin K, Pandey NB, Popel AS. Crosstalk between stromal components and tumor cells of TNBC via secreted factors enhances tumor growth and metastasis. Oncotarget 2017; 8:60210-60222. [PMID: 28947965 PMCID: PMC5601133 DOI: 10.18632/oncotarget.19417] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 07/11/2017] [Indexed: 12/12/2022] Open
Abstract
Triple negative breast cancer (TNBC) as a metastatic disease is currently incurable. Reliable and reproducible methods for testing drugs against metastasis are not available. Stromal cells may play a critical role in tumor progression and metastasis. In this study, we determined that fibroblasts and macrophages secreted IL-8 upon induction by tumor cell-conditioned media (TCM) from MDA-MB-231 cancer cells. Our data showed that the proliferation of MDA-MB-231 cells co-cultured with fibroblasts or macrophages was enhanced compared to the monoculture. Furthermore, TNBC cell migration, a key step in tumor metastasis, was promoted by conditioned media (CM) from TCM-induced fibroblasts or macrophages. Knockdown of the IL-8 receptor CXCR2 by CRISPR-Cas9 reduces MDA-MB-231 cell proliferation and migration compared to wild type. In a mouse xenograft tumor model, the growth of MDA-MB-231-CXCR2−/− tumor was significantly decreased compared to the growth of tumors from wild-type cells. In addition, the incidence of thoracic metastasis of MDA-MB-231-CXCR2−/− tumors was reduced compared to wild type. We found that the auto- and paracrine loop exists between TNBC cells and stroma, which results in enhanced IL-8 secretion from the stromal components. Significantly, inhibition of the IL-8 signaling pathway by reparixin, an inhibitor of the IL-8 receptor, CXCR1/2, reduced MDA-MB-231 tumor growth and metastasis. Taken together, these findings implicate IL-8 signaling as a critical event in TNBC tumor growth and metastasis via crosstalk with stromal components.
Collapse
Affiliation(s)
- Kideok Jin
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Niranjan B Pandey
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
191
|
Bahrami A, Hassanian SM, Khazaei M, Hasanzadeh M, Shahidsales S, Maftouh M, Ferns GA, Avan A. The Therapeutic Potential of Targeting Tumor Microenvironment in Breast Cancer: Rational Strategies and Recent Progress. J Cell Biochem 2017; 119:111-122. [DOI: 10.1002/jcb.26183] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 06/01/2017] [Indexed: 01/05/2023]
Affiliation(s)
- Afsane Bahrami
- Department of Modern Sciences and Technologies, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
- Metabolic Syndrome Research CenterMashhad University of Medical SciencesMashhadIran
- Student Research Committee, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research CenterMashhad University of Medical SciencesMashhadIran
- Department of Medical Biochemistry, School of MedicineMashhad University of Medical SciencesMashhadIran
| | - Majid Khazaei
- Department of Physiology, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Malihe Hasanzadeh
- Department of Gynecology Oncology, Woman Health Research CenterMashhad University of Medical SciencesMashhadIran
| | | | - Mina Maftouh
- Metabolic Syndrome Research CenterMashhad University of Medical SciencesMashhadIran
| | - Gordon A. Ferns
- Brighton & Sussex Medical SchoolDivision of Medical EducationFalmer, BrightonSussex BN1 9PH, UK
| | - Amir Avan
- Metabolic Syndrome Research CenterMashhad University of Medical SciencesMashhadIran
- Cancer Research CenterMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
192
|
Reduction in Migratory Phenotype in a Metastasized Breast Cancer Cell Line via Downregulation of S100A4 and GRM3. Sci Rep 2017; 7:3459. [PMID: 28615627 PMCID: PMC5471271 DOI: 10.1038/s41598-017-03811-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 05/04/2017] [Indexed: 11/13/2022] Open
Abstract
To investigate phenotypic and genotypic alterations before and after bone metastasis, we conducted genome-wide mRNA profiling and DNA exon sequencing of two cell lines (TMD and BMD) derived from a mouse xenograft model. TMD cells were harvested from the mammary fat pad after transfecting MDA-MB-231 breast cancer cells, while BMD cells were isolated from the metastasized bone. Compared to BMD cells, TMD cells exhibited higher cellular motility. In contrast, BMD cells formed a spheroid with a smoother and more circular surface when co-cultured with osteoblasts. In characterizing mRNA expression using principal component analysis, S100 calcium-binding protein A4 (S100A4) was aligned to a principal axis associated with metastasis. Partial silencing of S100A4 suppressed migratory capabilities of TMD cells, while Paclitaxel decreased the S100A4 level and reduced TMD’s cellular motility. DNA mutation analysis revealed that the glutamate metabotropic receptor 3 (GRM3) gene gained a premature stop codon in BMD cells, and silencing GRM3 in TMD cells altered their spheroid shape closer to that of BMD cells. Collectively, this study demonstrates that metastasized cells are less migratory due in part to the post-metastatic downregulation of S100A4 and GRM3. Targeting S100A4 and GRM3 may help prevent bone metastasis.
Collapse
|
193
|
Homeotic and Embryonic Gene Expression in Breast Adipose Tissue and in Adipose Tissues Used as Donor Sites in Plastic Surgery. Plast Reconstr Surg 2017; 139:685e-692e. [PMID: 28234838 DOI: 10.1097/prs.0000000000003070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Autologous fat grafting has become an essential procedure in breast reconstructive surgery. However, molecular knowledge of different adipose donor sites remains inadequate. Tissue regeneration studies have shown that it is essential to match the Hox code of transplanted cells and host tissues to achieve correct repair. This study aims to provide a better molecular understanding of adipose tissue. METHODS Over the course of 1 year, the authors prospectively included 15 patients and studied seven adipose areas: chin, breast, arm, abdomen, thigh, hip, and knee. The first step consisted of the surgical harvesting of adipose tissue. RNA was then extracted and converted into cDNA to study gene expression levels of 10 targeted genes by real-time polymerase chain reaction. RESULTS Forty samples from Caucasian women with a mean age of 48 years were studied. The expression of PAX3, a marker of neuroectodermal origin, was significantly higher in the breast, with a decreasing gradient from the upper to lower areas of the body. An inverse gradient was found for the expression of HOXC10. This expression profile was statistically significant for the areas of the thigh and knee compared with the breast (p < 0.0083). CONCLUSIONS Breast fat may have a specific embryologic origin compared with the knee and thigh. The reinjection of adipocytes from the infraumbilical area leads to the transfer of cells highly expressing HOXC10. This study raises questions about the safety of this procedure, and future studies will be required to examine molecular modifications of adipose cells transferred to a heterotopic location. CLINICAL QUESTION/LEVEL OF EVIDENCE Therapeutic, V.
Collapse
|
194
|
Strömvall K, Thysell E, Halin Bergström S, Bergh A. Aggressive rat prostate tumors reprogram the benign parts of the prostate and regional lymph nodes prior to metastasis. PLoS One 2017; 12:e0176679. [PMID: 28472073 PMCID: PMC5417597 DOI: 10.1371/journal.pone.0176679] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 04/14/2017] [Indexed: 12/14/2022] Open
Abstract
In order to grow and spread tumors need to interact with adjacent tissues. We therefore hypothesized that small but aggressive prostate cancers influence the rest of the prostate and regional lymph nodes differently than tumors that are more indolent. Poorly metastatic (Dunning AT1) or highly metastatic (Dunning MLL) rat prostate tumor cells were injected into the ventral prostate lobe of immunocompetent rats. After 10 days—when the tumors occupied about 30% of the prostate lobe and lymph node metastases were undetectable—the global gene expression in tumors, benign parts of the prostate, and regional iliac lymph nodes were examined to define tumor-induced changes related to preparation for future metastasis. The tumors induced profound effects on the gene expression profiles in the benign parts of the prostate and these were strikingly different in the two tumor models. Gene ontology enrichment analysis suggested that tumors with high metastatic capacity were more successful than less metastatic tumors in inducing tumor-promoting changes and suppressing anti-tumor immune responses in the entire prostate. Some of these differences such as altered angiogenesis, nerve density, accumulation of T-cells and macrophages were verified by immunohistochemistry. Gene expression alterations in the regional lymph nodes suggested decreased quantity and activation of immune cells in MLL-lymph nodes that were also verified by immunostaining. In summary, even when small highly metastatic prostate tumors can affect the entire tumor-bearing organ and pre-metastatic lymph nodes differently than less metastatic tumors. When the kinetics of these extratumoral influences (by us named TINT = tumor instructed normal tissue) are more precisely defined they could potentially be used as markers of disease aggressiveness and become therapeutic targets.
Collapse
Affiliation(s)
- Kerstin Strömvall
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
- * E-mail:
| | - Elin Thysell
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | | | - Anders Bergh
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| |
Collapse
|
195
|
Pickup MW, Owens P, Moses HL. TGF-β, Bone Morphogenetic Protein, and Activin Signaling and the Tumor Microenvironment. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a022285. [PMID: 28062564 DOI: 10.1101/cshperspect.a022285] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The cellular and noncellular components surrounding the tumor cells influence many aspects of tumor progression. Transforming growth factor β (TGF-β), bone morphogenetic proteins (BMPs), and activins have been shown to regulate the phenotype and functions of the microenvironment and are attractive targets to attenuate protumorigenic microenvironmental changes. Given the pleiotropic nature of the cytokines involved, a full understanding of their effects on numerous cell types in many contexts is necessary for proper clinical intervention. In this review, we will explore the various effects of TGF-β, BMP, and activin signaling on stromal phenotypes known to associate with cancer progression. We will summarize these findings in the context of their tumor suppressive or promoting effects, as well as the molecular changes that these cytokines induce to influence stromal phenotypes.
Collapse
Affiliation(s)
- Michael W Pickup
- Department of Cancer Biology and Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee 37232
| | - Philip Owens
- Department of Cancer Biology and Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee 37232
| | - Harold L Moses
- Department of Cancer Biology and Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee 37232
| |
Collapse
|
196
|
Singh M, Venkata Krishnan H, Ranganathan S, Kiesel B, Beumer JH, Sreekumar S, Sant S. Controlled Three-Dimensional Tumor Microenvironments Recapitulate Phenotypic Features and Differential Drug Response in Early vs Advanced Stage Breast Cancer. ACS Biomater Sci Eng 2017; 4:421-431. [DOI: 10.1021/acsbiomaterials.7b00081] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
| | | | | | - Brian Kiesel
- University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania 15232, United States
| | - Jan Hendrik Beumer
- University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania 15232, United States
- Division
of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, United States
| | - Sreeja Sreekumar
- Department
of Pharmacology and Chemical Biology, Women’s Cancer Research
Center, Magee-Women’s Research Institute, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania 15232, United States
| | - Shilpa Sant
- University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania 15232, United States
| |
Collapse
|
197
|
Abstract
Advances in mammography have sparked an exponential increase in the detection of early-stage breast lesions, most commonly ductal carcinoma in situ (DCIS). More than 50% of DCIS lesions are benign and will remain indolent, never progressing to invasive cancers. However, the factors that promote DCIS invasion remain poorly understood. Here, we show that SMARCE1 is required for the invasive progression of DCIS and other early-stage tumors. We show that SMARCE1 drives invasion by regulating the expression of secreted proteases that degrade basement membrane, an ECM barrier surrounding all epithelial tissues. In functional studies, SMARCE1 promotes invasion of in situ cancers growing within primary human mammary tissues and is also required for metastasis in vivo. Mechanistically, SMARCE1 drives invasion by forming a SWI/SNF-independent complex with the transcription factor ILF3. In patients diagnosed with early-stage cancers, SMARCE1 expression is a strong predictor of eventual relapse and metastasis. Collectively, these findings establish SMARCE1 as a key driver of invasive progression in early-stage tumors.
Collapse
|
198
|
Impact of Different Analytic Approaches on the Analysis of the Breast Fibroglandular Tissue Using Diffusion Weighted Imaging. BIOMED RESEARCH INTERNATIONAL 2017; 2017:1094354. [PMID: 28349054 PMCID: PMC5352872 DOI: 10.1155/2017/1094354] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 02/15/2017] [Indexed: 12/27/2022]
Abstract
Purpose. This study investigated the impact of the different region of interest (ROI) approaches on measurement of apparent diffusion coefficient (ADC) values in the breast firbroglandular tissue (FT). Methods. Breast MR images of 38 women diagnosed with unilateral breast cancer were studied. Percent density (PD) and ADC were measured from the contralateral normal breast. Four different ROIs were used for ADC measurement. The measured PD and ADC were correlated. Results. Among the four ROIs, the manually placed small ROI on FT gave the highest mean ADC (ADC = 1839 ± 343 [×10−6 mm2/s]), while measurement from the whole breast gave the lowest mean ADC (ADC = 933 ± 383 [×10−6 mm2/s]). The ADC measured from the whole breast was highly correlated with PD with r = 0.95. In slice-to-slice comparison, the central slices with more FT had higher ADC values than the peripheral slices did, presumably due to less partial volume effect from fat. Conclusions. Our results indicated that the measured ADC heavily depends on the composition of breast tissue contained in the ROI used for the ADC measurements. Women with low breast density showing lower ADC values were most likely due to the partial volume effect of fatty tissues.
Collapse
|
199
|
Alsehli H, Gari M, Abuzinadah M, Abuzenadah A. The emerging importance of high content screening for future therapeutics. J Microsc Ultrastruct 2017. [DOI: 10.1016/j.jmau.2017.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
200
|
Abou-Shousha S, Moaaz M, Sheta M, Motawea MA. An Approach to Breast Cancer Immunotherapy: The Apoptotic Activity of Recombinant Anti-Interleukin-6 Monoclonal Antibodies in Intact Tumour Microenvironment of Breast Carcinoma. Scand J Immunol 2017; 83:427-37. [PMID: 26971879 DOI: 10.1111/sji.12426] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 03/03/2016] [Indexed: 12/11/2022]
Abstract
Current work is one of our comprehensive preclinical studies, a new approach to breast cancer (BC) immunotherapy through induction of tumour cell apoptosis. Tumour growth is not just a result of uncontrolled cell proliferation but also of reduced apoptosis. High levels of interleukin-6 (IL-6) are associated with metastatic BC and correlated with poor survival as it promotes growth of tumour-initiating cells during early tumorigenesis protecting these cells from apoptosis. Therefore, this study aims at investigating the potential of anti-IL-6 monoclonal antibodies to suppress IL-6 proliferative/anti-apoptotic activities in intact tumour microenvironment of BC. Fresh sterile tumour and normal breast tissue specimens were taken from 50 female Egyptian patients with BC undergoing radical mastectomy. A unique tissue culture system designed to provide cells of each intact tumour/normal tissue sample with its proper microenvironment either supplemented or not with anti-IL-6 monoclonal antibodies. To evaluate the apoptotic activity of anti-IL-6 as a novel candidate for BC treatment strategy, we compared its effects with those obtained using tumour necrosis-related apoptosis-inducing ligand TRAIL as an established apoptotic agent. Our results revealed that levels of either anti-IL-6- or TRAIL-induced apoptosis in the tumour or normal tissue cultures were significantly higher than those in their corresponding untreated ones (P < 0.001). No statistically significant differences have been found between apoptosis levels induced by anti-IL-6 monoclonal antibodies and those induced by TRAIL. Recombinant anti-IL-6 monoclonal antibodies could represent a novel effective element of immunotherapeutic treatment strategy for BC. The selectivity and anti-apoptotic potential of anti-IL-6 is highly hopeful in IL-6- abundant BC tumour microenvironment.
Collapse
Affiliation(s)
- S Abou-Shousha
- Department of Immunology and Allergy, Medical research Institute, Alexandria University, Alexandria, Egypt
| | - M Moaaz
- Department of Immunology and Allergy, Medical research Institute, Alexandria University, Alexandria, Egypt
| | - M Sheta
- Department of pathology, Medical research Institute, Alexandria University, Alexandria, Egypt
| | - M A Motawea
- Department of Surgery, Medical research Institute, Alexandria University, Alexandria, Egypt
| |
Collapse
|