1
|
Shi Q, Huang F, Wang Y, Liu H, Deng H, Chen YG. HER2 phosphorylation induced by TGF-β promotes mammary morphogenesis and breast cancer progression. J Cell Biol 2024; 223:e202307138. [PMID: 38407425 PMCID: PMC10896696 DOI: 10.1083/jcb.202307138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/15/2023] [Accepted: 01/16/2024] [Indexed: 02/27/2024] Open
Abstract
Transforming growth factor β (TGF-β) and HER2 signaling collaborate to promote breast cancer progression. However, their molecular interplay is largely unclear. TGF-β can activate mitogen-activated protein kinase (MAPK) and AKT, but the underlying mechanism is not fully understood. In this study, we report that TGF-β enhances HER2 activation, leading to the activation of MAPK and AKT. This process depends on the TGF-β type I receptor TβRI kinase activity. TβRI phosphorylates HER2 at Ser779, promoting Y1248 phosphorylation and HER2 activation. Mice with HER2 S779A mutation display impaired mammary morphogenesis, reduced ductal elongation, and branching. Furthermore, wild-type HER2, but not S779A mutant, promotes TGF-β-induced epithelial-mesenchymal transition, cell migration, and lung metastasis of breast cells. Increased HER2 S779 phosphorylation is observed in human breast cancers and positively correlated with the activation of HER2, MAPK, and AKT. Our findings demonstrate the crucial role of TGF-β-induced S779 phosphorylation in HER2 activation, mammary gland development, and the pro-oncogenic function of TGF-β in breast cancer progression.
Collapse
Affiliation(s)
- Qiaoni Shi
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Fei Huang
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yalong Wang
- Guangzhou National Laboratory, Guangzhou, China
| | - Huidong Liu
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, China
| | - Ye-Guang Chen
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
- Guangzhou National Laboratory, Guangzhou, China
- School of Basic Medicine, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
2
|
He Y, Goyette MA, Chapelle J, Boufaied N, Al Rahbani J, Schonewolff M, Danek EI, Muller WJ, Labbé DP, Côté JF, Lamarche-Vane N. CdGAP is a talin-binding protein and a target of TGF-β signaling that promotes HER2-positive breast cancer growth and metastasis. Cell Rep 2023; 42:112936. [PMID: 37552602 DOI: 10.1016/j.celrep.2023.112936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/10/2023] [Accepted: 07/20/2023] [Indexed: 08/10/2023] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) plays a crucial role in metastasis, which is the leading cause of death in breast cancer patients. Here, we show that Cdc42 GTPase-activating protein (CdGAP) promotes tumor formation and metastasis to lungs in the HER2-positive (HER2+) murine breast cancer model. CdGAP facilitates intravasation, extravasation, and growth at metastatic sites. CdGAP depletion in HER2+ murine primary tumors mediates crosstalk with a Dlc1-RhoA pathway and is associated with a transforming growth factor β (TGF-β)-induced EMT transcriptional signature. CdGAP is positively regulated by TGF-β signaling during EMT and interacts with the adaptor talin to modulate focal adhesion dynamics and integrin activation. Moreover, HER2+ breast cancer patients with high CdGAP mRNA expression combined with a high TGF-β-EMT signature are more likely to present lymph node invasion. Our results suggest CdGAP as a candidate therapeutic target for HER2+ metastatic breast cancer by inhibiting TGF-β and integrin/talin signaling pathways.
Collapse
Affiliation(s)
- Yi He
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada; Department of Anatomy and Cell Biology, McGill University, Montréal, QC H3A 0C7, Canada
| | - Marie-Anne Goyette
- Institut de Recherches Cliniques de Montréal, Université de Montréal, Montréal, QC H2W 1R7, Canada
| | - Jennifer Chapelle
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada; Department of Anatomy and Cell Biology, McGill University, Montréal, QC H3A 0C7, Canada
| | - Nadia Boufaied
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada
| | - Jalal Al Rahbani
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada; Department of Anatomy and Cell Biology, McGill University, Montréal, QC H3A 0C7, Canada
| | - Maribel Schonewolff
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada; Department of Anatomy and Cell Biology, McGill University, Montréal, QC H3A 0C7, Canada
| | - Eric I Danek
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada; Department of Anatomy and Cell Biology, McGill University, Montréal, QC H3A 0C7, Canada
| | - William J Muller
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montréal, QC H3A 1A3, Canada
| | - David P Labbé
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada; Department of Anatomy and Cell Biology, McGill University, Montréal, QC H3A 0C7, Canada; Division of Urology, Department of Surgery, McGill University, Montréal, QC H4A 3J1, Canada
| | - Jean-François Côté
- Department of Anatomy and Cell Biology, McGill University, Montréal, QC H3A 0C7, Canada; Institut de Recherches Cliniques de Montréal, Université de Montréal, Montréal, QC H2W 1R7, Canada
| | - Nathalie Lamarche-Vane
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada; Department of Anatomy and Cell Biology, McGill University, Montréal, QC H3A 0C7, Canada.
| |
Collapse
|
3
|
Luo F, Huang Y, Li Y, Zhao X, Xie Y, Zhang Q, Mei J, Liu X. A narrative review of the relationship between TGF-β signaling and gynecological malignant tumor. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1601. [PMID: 34790807 PMCID: PMC8576662 DOI: 10.21037/atm-21-4879] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 10/14/2021] [Indexed: 12/24/2022]
Abstract
Objective This paper reviews the association between transforming growth factor-β (TGF-β) and its receptor and tumor, focusing on gynecological malignant tumors. we hope to provide more methods to help increase the potential of TGF-β signaling targeted treatment of specific cancers. Background The occurrence of a malignant tumor is a complex process of multi-step, multi-gene regulation, and its progression is affected by various components of the tumor cells and/or tumor microenvironment. The occurrence of gynecological diseases not only affect women's health, but also bring some troubles to their normal life. Especially when gynecological malignant tumors occur, the situation is more serious, which will endanger the lives of patients. Due to differences in environmental and economic conditions, not all women have access to assistance and treatment specifically meeting their needs. TGF-β is a multi-potent growth factor that maintains homeostasis in mammals by inhibiting cell growth and promoting apoptosis in vivo. TGF-β signaling is fundamental to inflammatory disease and favors the emergence of tumors, and it also plays an important role in immunosuppression in the tumor microenvironment. In the early stages of the tumor, TGF-β acts as a tumor inhibitor, whereas in advanced tumors, mutations or deletion of the TGF-β signaling core component initiate neogenesis. Methods Literatures about TGF-β and gynecological malignant tumor were extensively reviewed to analyze and discuss. Conclusions We discussed the role of TGF-β signaling in different types of gynecological tumor cells, thus demonstrating that targeted TGF-β signaling may be an effective tumor treatment strategy.
Collapse
Affiliation(s)
- Fangyuan Luo
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, China.,Department of Obstetrics and Gynecology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Yu Huang
- Department of Obstetrics and Gynecology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Yilin Li
- Department of Obstetrics and Gynecology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Xiaolan Zhao
- Department of Obstetrics and Gynecology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Yao Xie
- Department of Obstetrics and Gynecology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Qianwen Zhang
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Jie Mei
- Department of Obstetrics and Gynecology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Xinghui Liu
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Ruiz TFR, Taboga SR, Leonel ECR. Molecular mechanisms of mammary gland remodeling: A review of the homeostatic versus bisphenol a disrupted microenvironment. Reprod Toxicol 2021; 105:1-16. [PMID: 34343637 DOI: 10.1016/j.reprotox.2021.07.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/26/2021] [Accepted: 07/29/2021] [Indexed: 12/30/2022]
Abstract
Mammary gland (MG) undergoes critical points of structural changes throughout a woman's life. During the perinatal and pubertal stages, MG develops through growth and differentiation to establish a pre-mature feature. If pregnancy and lactation occur, the epithelial compartment branches and differentiates to create a specialized structure for milk secretion and nurturing of the newborn. However, the ultimate MG modification consists of a regression process aiming to reestablish the smaller and less energy demanding structure until another production cycle happens. The unraveling of these fascinating physiologic cycles has helped the scientific community elucidate aspects of molecular regulation of proliferative and apoptotic events and remodeling of the stromal compartment. However, greater understanding of the hormonal pathways involved in MG developmental stages led to concern that endocrine disruptors such as bisphenol A (BPA), may influence these specific development/involution stages, called "windows of susceptibility". Since it is used in the manufacture of polycarbonate plastics and epoxy resins, BPA is a ubiquitous chemical present in human everyday life, exerting an estrogenic effect. Thus, descriptions of its deleterious effects on the MG, especially in terms of serum hormone concentrations, hormonal receptor expression, molecular pathways, and epigenetic alterations, have been widely published. Therefore, allied to a didactic description of the main physiological mechanisms involved in different critical points of MG development, the current review provides a summary of key mechanisms by which the endocrine disruptor BPA impacts MG homeostasis at different windows of susceptibility, causing short- and long-term effects.
Collapse
Affiliation(s)
- Thalles Fernando Rocha Ruiz
- São Paulo State University (Unesp), Department of Biology, Institute of Biosciences, Humanities and Exact Sciences, São José Do Rio Preto, Brazil.
| | - Sebastião Roberto Taboga
- São Paulo State University (Unesp), Department of Biology, Institute of Biosciences, Humanities and Exact Sciences, São José Do Rio Preto, Brazil.
| | - Ellen Cristina Rivas Leonel
- São Paulo State University (Unesp), Department of Biology, Institute of Biosciences, Humanities and Exact Sciences, São José Do Rio Preto, Brazil; Federal University of Goiás (UFG), Department of Histology, Embryology and Cell Biology, Institute of Biological Sciences, Goiânia, Brazil.
| |
Collapse
|
5
|
Fakhlaei R, Selamat J, Khatib A, Razis AFA, Sukor R, Ahmad S, Babadi AA. The Toxic Impact of Honey Adulteration: A Review. Foods 2020; 9:E1538. [PMID: 33114468 PMCID: PMC7692231 DOI: 10.3390/foods9111538] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/10/2020] [Accepted: 09/11/2020] [Indexed: 12/11/2022] Open
Abstract
Honey is characterized as a natural and raw foodstuff that can be consumed not only as a sweetener but also as medicine due to its therapeutic impact on human health. It is prone to adulterants caused by humans that manipulate the quality of honey. Although honey consumption has remarkably increased in the last few years all around the world, the safety of honey is not assessed and monitored regularly. Since the number of consumers of honey adulteration have increased in recent years, their trust and interest in this valuable product has decreased. Honey adulterants are any substances that are added to the pure honey. In this regard, this paper provides a comprehensive and critical review of the different types of adulteration, common sugar adulterants and detection methods, and draws a clear perspective toward the impact of honey adulteration on human health. Adulteration increases the consumer's blood sugar, which can cause diabetes, abdominal weight gain, and obesity, raise the level of blood lipids and can cause high blood pressure. The most common organ affected by honey adulterants is the liver followed by the kidney, heart, and brain, as shown in several in vivo research designs.
Collapse
Affiliation(s)
- Rafieh Fakhlaei
- Food Safety and Food Integrity (FOSFI), Institute of Tropical Agriculture and Food Security, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
| | - Jinap Selamat
- Food Safety and Food Integrity (FOSFI), Institute of Tropical Agriculture and Food Security, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (A.F.A.R.); (R.S.)
| | - Alfi Khatib
- Pharmacognosy Research Group, Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy, International Islamic University Malaysia, Kuantan 25200, Pahang Darul Makmur, Malaysia;
- Faculty of Pharmacy, Airlangga University, Surabaya 60155, Indonesia
| | - Ahmad Faizal Abdull Razis
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (A.F.A.R.); (R.S.)
- Natural Medicines and Products Research Laboratory, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Rashidah Sukor
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (A.F.A.R.); (R.S.)
| | - Syahida Ahmad
- Department of Biochemistry, Faculty of Biotechnology & Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
| | - Arman Amani Babadi
- School of Energy and Power Engineering, Jiangsu University, Zhenjiang 212013, China;
| |
Collapse
|
6
|
Dai Y, Wu Z, Lang C, Zhang X, He S, Yang Q, Guo W, Lai Y, Du H, Peng X, Ren D. Copy number gain of ZEB1 mediates a double-negative feedback loop with miR-33a-5p that regulates EMT and bone metastasis of prostate cancer dependent on TGF-β signaling. Am J Cancer Res 2019; 9:6063-6079. [PMID: 31534537 PMCID: PMC6735523 DOI: 10.7150/thno.36735] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/28/2019] [Indexed: 01/17/2023] Open
Abstract
Background: The reciprocal repressive loop between ZEB1 and miRNAs has been extensively reported to play an important role in tumor progression and metastasis of various human tumor types. The aim of this study was to elucidate the role and the underlying mechanism of the double-negative feedback loop between ZEB1and miR-33a-5p in bone metastasis of prostate cancer (PCa). Methods: miR-33a-5p expression was examined in 40 bone metastatic and 165 non-bone metastatic PCa tissues by real-time PCR. Statistical analysis was performed to evaluate the clinical correlation between miR-33a-5p expression and clinicopathological characteristics, and overall and bone metastasis-free survival in PCa patients. The biological roles of miR-33a-5p in bone metastasis of PCa were investigated both by EMT and the Transwell assay in vitro, and by a mouse model of left cardiac ventricle inoculation in vivo. siRNA library, real-time PCR and chromatin immunoprecipitation (ChIP) were used to identify the underlying mechanism responsible for the decreased expression of miR-33a-5p in PCa. Bioinformatics analysis, Western blotting and luciferase reporter analysis were employed to examine the relationship between miR-33a-5p and its potential targets. Clinical correlation of miR-33a-5p with its targets was examined in human PCa tissues and primary PCa cells. Results: miR-33a-5p expression was downregulated in PCa tissues with bone metastasis and bone-derived cells, and low expression of miR-33a-5p strongly and positively correlated with advanced clinicopathological characteristics, and shorter overall and bone metastasis-free survival in PCa patients. Upregulating miR-33a-5p inhibited, while silencing miR-33a-5p promoted EMT, invasion and migration of PCa cells. Importantly, upregulating miR-33a-5p significantly repressed bone metastasis of PC-3 cells in vivo. Our results further revealed that recurrent ZEB1 upregulation induced by copy number gains transcriptionally inhibited miR-33a-5p expression, contributing to the reduced expression of miR-33a-5p in bone metastatic PCa tissues. In turn, miR-33a-5p formed a double negative feedback loop with ZEB1 in target-independent manner, which was dependent on TGF-β signaling. Finally, the clinical negative correlations of miR-33a-5p with ZEB1 expression and TGF-β signaling activity were demonstrated in PCa tissues and primary PCa cells. Conclusion: Our findings elucidated that copy number gains of ZEB1-triggered a TGF-β signaling-dependent miR-33a-5p-mediated negative feedback loop was highly relevant to the bone metastasis of PCa.
Collapse
|
7
|
Lee J. 3,3′-Diindolylmethane Inhibits TNF-α- and TGF-β-Induced Epithelial–Mesenchymal Transition in Breast Cancer Cells. Nutr Cancer 2019; 71:992-1006. [DOI: 10.1080/01635581.2019.1577979] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Joomin Lee
- Department of Food and Nutrition, Chosun University, Gwangju, Korea
| |
Collapse
|
8
|
Di L, Liu LJ, Yan YM, Fu R, Li Y, Xu Y, Cheng YX, Wu ZQ. Discovery of a natural small-molecule compound that suppresses tumor EMT, stemness and metastasis by inhibiting TGFβ/BMP signaling in triple-negative breast cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:134. [PMID: 30898152 PMCID: PMC6429712 DOI: 10.1186/s13046-019-1130-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/06/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND The transforming growth factor β (TGFβ) and bone morphogenetic protein (BMP) signaling pathways are both constitutively activated in triple-negative breast cancer (TNBC). We are interested in isolating the naturally-derived small-molecule inhibitor that could simultaneously targeting TGFβ/BMP pathways and further studying its anti-proliferative/-metastatic effects as well as the underlying mechanisms in multiple tumor models. METHODS Multiple in vitro cell-based assays are used to examine the compound's inhibitory efficacy on TNBC cell growth, stemness, epithelial-mesenchymal transition (EMT), invasion and migration by targeting TGFβ/BMP signaling pathways. Transgenic breast cancer mouse model (MMTV-PyMT), subcutaneous xenograft and bone metastasis models are used to examine ZL170's effects on TNBC growth and metastasis potentials in vivo. RESULTS ZL170 dose-dependently inhibits cell proliferation, EMT, stemness, invasion and migration in vitro via specifically targeting canonical TGFβ/BMP-SMADs pathways in TNBC cells. The compound significantly hinders osteolytic bone metastasis and xenograft tumor growth without inflicting toxicity on vital organs of tumor-bearing nude mice. ZL170 strongly inhibits primary tumor growth and lung metastases in MMTV-PyMT transgenic mice. ZL170-treated tumors exhibit impaired TGFβ/BMP signaling pathways in both epithelial and stromal compartments, thereby creating a suppressive tumor microenvironment characterized by reduced extracellular matrix deposition and decreased infiltration of stromal cells. CONCLUSIONS ZL170 inhibits tumor EMT, stemness and metastasis and could be further developed as a potent anti-metastatic agent used in combination with cytotoxic drugs for treatment of TNBC and other advanced metastatic cancers.
Collapse
Affiliation(s)
- Lei Di
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Collaborative Innovation Center for Gannan Oil-Tea Camellia Industrial Development, Gannan Medical University, Ganzhou, China
| | - Li-Juan Liu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Collaborative Innovation Center for Gannan Oil-Tea Camellia Industrial Development, Gannan Medical University, Ganzhou, China
| | - Yong-Ming Yan
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, School of Pharmaceutical Sciences, Shenzhen University Health Science Center, 3688 Nanhai Ave, Shenzhen, 518060, China
| | - Rong Fu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Collaborative Innovation Center for Gannan Oil-Tea Camellia Industrial Development, Gannan Medical University, Ganzhou, China
| | - Yi Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Collaborative Innovation Center for Gannan Oil-Tea Camellia Industrial Development, Gannan Medical University, Ganzhou, China
| | - Ying Xu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Collaborative Innovation Center for Gannan Oil-Tea Camellia Industrial Development, Gannan Medical University, Ganzhou, China
| | - Yong-Xian Cheng
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, School of Pharmaceutical Sciences, Shenzhen University Health Science Center, 3688 Nanhai Ave, Shenzhen, 518060, China.
| | - Zhao-Qiu Wu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Collaborative Innovation Center for Gannan Oil-Tea Camellia Industrial Development, Gannan Medical University, Ganzhou, China.
| |
Collapse
|
9
|
Yeo HL, Fan T, Lin R, Yu J, Liao G, Chen ES, Ho M, Lin W, Chen K, Chen C, Hung J, Wu J, Chang N, Chang MD, Yu J, Yu AL. Sialylation of vasorin by ST3Gal1 facilitates TGF-β1-mediated tumor angiogenesis and progression. Int J Cancer 2019; 144:1996-2007. [PMID: 30252131 PMCID: PMC6590135 DOI: 10.1002/ijc.31891] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 07/24/2018] [Accepted: 08/30/2018] [Indexed: 12/15/2022]
Abstract
ST3Gal1 is a key sialyltransferase which adds α2,3‐linked sialic acid to substrates and generates core 1 O‐glycan structure. Upregulation of ST3Gal1 has been associated with worse prognosis of breast cancer patients. However, the protein substrates of ST3Gal1 implicated in tumor progression remain elusive. In our study, we demonstrated that ST3GAL1‐silencing significantly reduced tumor growth along with a notable decrease in vascularity of MCF7 xenograft tumors. We identified vasorin (VASN) which was shown to bind TGF‐β1, as a potential candidate that links ST3Gal1 to angiogenesis. LC‐MS/MS analysis of VASN secreted from MCF7, revealed that more than 80% of its O‐glycans are sialyl‐3T and disialyl‐T. ST3GAL1‐silencing or desialylation of VASN by neuraminidase enhanced its binding to TGF‐β1 by 2‐ to 3‐fold and thereby dampening TGF‐β1 signaling and angiogenesis, as indicated by impaired tube formation of HUVECs, suppressed angiogenesis gene expression and reduced activation of Smad2 and Smad3 in HUVEC cells. Examination of 114 fresh primary breast cancer and their adjacent normal tissues showed that the expression levels of ST3Gal1 and TGFB1 were high in tumor part and the expression of two genes was positively correlated. Kaplan Meier survival analysis showed a significantly shorter relapse‐free survival for those with lower expression VASN, notably, the combination of low VASN with high ST3GAL1 yielded even higher risk of recurrence (p = 0.025, HR = 2.967, 95% CI = 1.14–7.67). Since TGF‐β1 is known to transcriptionally activate ST3Gal1, our findings illustrated a feedback regulatory loop in which TGF‐β1 upregulates ST3Gal1 to circumvent the negative impact of VASN. What's new? The addition of sialic acid to glycoproteins is dysregulated in many cancers, and enhanced expression of one key enzyme, the sialyltransferase ST3Gal1, is associated with poor prognosis. Here, the authors identified the membrane protein vasorin as a new ST3Gal1 substrate and connect it with TGF‐β1‐induced signaling and angiogenesis in breast cancer. As silencing of ST3Gal1 dampened TGF‐β1 signaling and suppressed angiogenesis, development of ST3Gal1 inhibitors might be clinically useful to improve the prognosis of breast cancer patients.
Collapse
Affiliation(s)
- Hui Ling Yeo
- Institute of Stem Cell and Translational Cancer ResearchChang Gung Memorial Hospital at Linkou and Chang Gung UniversityTaoyuanTaiwan
- Institute of Molecular and Cellular BiologyNational Tsing Hua UniversityHsinchuTaiwan
- Chemical Biology and Molecular Biophysics ProgramTaiwan International Graduate Program Academia SinicaTaipeiTaiwan
| | - Tan‐Chi Fan
- Institute of Stem Cell and Translational Cancer ResearchChang Gung Memorial Hospital at Linkou and Chang Gung UniversityTaoyuanTaiwan
| | - Ruey‐Jen Lin
- Institute of Stem Cell and Translational Cancer ResearchChang Gung Memorial Hospital at Linkou and Chang Gung UniversityTaoyuanTaiwan
| | - Jyh‐Cherng Yu
- General Surgery, Department of SurgeryTri‐Service General Hospital, National Defense Medical CenterTaipeiTaiwan
| | - Guo‐Shiou Liao
- General Surgery, Department of SurgeryTri‐Service General Hospital, National Defense Medical CenterTaipeiTaiwan
| | - Eric Sheng‐Wen Chen
- Institute of Stem Cell and Translational Cancer ResearchChang Gung Memorial Hospital at Linkou and Chang Gung UniversityTaoyuanTaiwan
| | - Ming‐Yi Ho
- Institute of Stem Cell and Translational Cancer ResearchChang Gung Memorial Hospital at Linkou and Chang Gung UniversityTaoyuanTaiwan
| | - Wen‐Der Lin
- Institute of Stem Cell and Translational Cancer ResearchChang Gung Memorial Hospital at Linkou and Chang Gung UniversityTaoyuanTaiwan
| | - Kowa Chen
- Institute of Stem Cell and Translational Cancer ResearchChang Gung Memorial Hospital at Linkou and Chang Gung UniversityTaoyuanTaiwan
| | | | - Jung‐Tung Hung
- Institute of Stem Cell and Translational Cancer ResearchChang Gung Memorial Hospital at Linkou and Chang Gung UniversityTaoyuanTaiwan
| | - Jen‐Chine Wu
- Institute of Stem Cell and Translational Cancer ResearchChang Gung Memorial Hospital at Linkou and Chang Gung UniversityTaoyuanTaiwan
| | - Nai‐Chuan Chang
- Institute of Stem Cell and Translational Cancer ResearchChang Gung Memorial Hospital at Linkou and Chang Gung UniversityTaoyuanTaiwan
| | | | - John Yu
- Institute of Stem Cell and Translational Cancer ResearchChang Gung Memorial Hospital at Linkou and Chang Gung UniversityTaoyuanTaiwan
| | - Alice Lin‐Tsing Yu
- Institute of Stem Cell and Translational Cancer ResearchChang Gung Memorial Hospital at Linkou and Chang Gung UniversityTaoyuanTaiwan
- Genomics Research CenterAcademia SinicaTaipeiTaiwan
- Department of Pediatrics/Hematology OncologyUniversity of CaliforniaSan DiegoCAUSA
| |
Collapse
|
10
|
Du J, Yu Y, Zhan J, Zhang H. Targeted Therapies Against Growth Factor Signaling in Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1026:125-146. [PMID: 29282682 DOI: 10.1007/978-981-10-6020-5_6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Breast cancer is the most prevalent female malignancy throughout the world. Conventional treatment strategies for breast cancer consist of chemotherapy, radiation, surgery, chemoradiation, hormone therapy, and targeted therapies. Among them, targeted therapies show advantages to reduce cost and toxicity for being possible for individualized treatments based on the intrinsic subtypes of breast cancer. With deeper understanding of key signaling pathways concerning tumor growth and survival, growth factor-controlled signaling pathways are frequently dysregulated in the development and progression of breast cancer. Thus, targeted therapies against growth factor-mediated signaling pathways have been shown to have promising efficacy in both preclinical animal models and human clinical trials. In this chapter, we will briefly introduce inhibitors and monoclonal antibodies that target the main growth factor-modulated scenarios including epidermal growth factor receptor (EGFR), transforming growth factor beta (TGF-β), insulin-like growth factor 1 receptor (IGF1R), and fibroblast growth factor receptor (FGFR) signaling pathways in breast cancer therapy.
Collapse
Affiliation(s)
- Juan Du
- Department of Anatomy, Histology and Embryology, Laboratory of Molecular Cell Biology and Tumor Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yu Yu
- Department of Anatomy, Histology and Embryology, Laboratory of Molecular Cell Biology and Tumor Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jun Zhan
- Department of Anatomy, Histology and Embryology, Laboratory of Molecular Cell Biology and Tumor Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Hongquan Zhang
- Department of Anatomy, Histology and Embryology, Laboratory of Molecular Cell Biology and Tumor Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.
| |
Collapse
|
11
|
Flister MJ, Bergom C. Genetic Modifiers of the Breast Tumor Microenvironment. Trends Cancer 2018; 4:429-444. [PMID: 29860987 DOI: 10.1016/j.trecan.2018.04.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 04/13/2018] [Accepted: 04/16/2018] [Indexed: 02/06/2023]
Abstract
Multiple nonmalignant cell types in the tumor microenvironment (TME) impact breast cancer risk, metastasis, and response to therapy, yet most heritable mechanisms that influence TME cell function and breast cancer outcomes are largely unknown. Breast cancer risk is ∼30% heritable and >170 genetic loci have been associated with breast cancer traits. However, the majority of candidate genes have poorly defined mechanistic roles in breast cancer biology. Research indicates that breast cancer risk modifiers directly impact cancer cells, yet it is equally plausible that some modifier alleles impact the nonmalignant TME. The objective of this review is to examine the list of current breast cancer candidate genes that may modify breast cancer risk and outcome through the TME.
Collapse
Affiliation(s)
- Michael J Flister
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Carmen Bergom
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
12
|
Gulubova M, Aleksandrova E, Vlaykova T. Promoter polymorphisms in TGFB1
and IL10
genes influence tumor dendritic cells infiltration, development and prognosis of colorectal cancer. J Gene Med 2018; 20:e3005. [DOI: 10.1002/jgm.3005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 12/08/2017] [Accepted: 12/27/2017] [Indexed: 12/14/2022] Open
Affiliation(s)
- Maya Gulubova
- Department of General and Clinical Pathology, Forensic Medicine and Deontology, Medical Faculty; Trakia University; Stara Zagora Bulgaria
| | - Elina Aleksandrova
- Department of General and Clinical Pathology, Forensic Medicine and Deontology, Medical Faculty; Trakia University; Stara Zagora Bulgaria
- Department of Medical Chemistry and Biochemistry, Medical Faculty; Trakia University; Stara Zagora Bulgaria
| | - Tatyana Vlaykova
- Department of Medical Chemistry and Biochemistry, Medical Faculty; Trakia University; Stara Zagora Bulgaria
| |
Collapse
|
13
|
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: 43] [Impact Index Per Article: 6.1] [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
|
14
|
Miret N, Pontillo C, Ventura C, Carozzo A, Chiappini F, Kleiman de Pisarev D, Fernández N, Cocca C, Randi A. Hexachlorobenzene modulates the crosstalk between the aryl hydrocarbon receptor and transforming growth factor-β1 signaling, enhancing human breast cancer cell migration and invasion. Toxicology 2016; 366-367:20-31. [PMID: 27519288 DOI: 10.1016/j.tox.2016.08.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 08/03/2016] [Accepted: 08/08/2016] [Indexed: 12/26/2022]
Abstract
Given the number of women affected by breast cancer, considerable interest has been raised in understanding the relationships between environmental chemicals and disease onset. Hexachlorobenzene (HCB) is a dioxin-like compound that is widely distributed in the environment and is a weak ligand of the aryl hydrocarbon receptor (AhR). We previously demonstrated that HCB acts as an endocrine disruptor capable of stimulating cell proliferation, migration, invasion, and metastasis in different breast cancer models. In addition, increasing evidence indicates that transforming growth factor-β1 (TGF-β1) can contribute to tumor maintenance and progression. In this context, this work investigated the effect of HCB (0.005, 0.05, 0.5, and 5μM) on TGF-β1 signaling and AhR/TGF-β1 crosstalk in the human breast cancer cell line MDA-MB-231 and analyzed whether TGF-β1 pathways are involved in HCB-induced cell migration and invasion. RT-qPCR results indicated that HCB reduces AhR mRNA expression through TGF-β1 signaling but enhances TGF-β1 mRNA levels involving AhR signaling. Western blot analysis demonstrated that HCB could increase TGF-β1 protein levels and activation, as well as Smad3, JNK, and p38 phosphorylation. In addition, low and high doses of HCB were determined to exert differential effects on AhR protein levels, localization, and activation, with a high dose (5μM) inducing AhR nuclear translocation and AhR-dependent CYP1A1 expression. These findings also revealed that c-Src and AhR are involved in HCB-mediated activation of Smad3. HCB enhances cell migration (scratch motility assay) and invasion (Transwell assay) through the Smad, JNK, and p38 pathways, while ERK1/2 is only involved in HCB-induced cell migration. These results demonstrate that HCB modulates the crosstalk between AhR and TGF-β1 and consequently exacerbates a pro-migratory phenotype in MDA-MB-231 cells, which contributes to a high degree of malignancy. Taken together, our findings help to characterize the molecular mechanism underlying the effects of HCB on breast cancer progression.
Collapse
Affiliation(s)
- Noelia Miret
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Laboratorio de Efectos Biológicos de Contaminantes Ambientales, Paraguay 2155, 5° piso, (CP1121), Buenos Aires, Argentina.
| | - Carolina Pontillo
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Laboratorio de Efectos Biológicos de Contaminantes Ambientales, Paraguay 2155, 5° piso, (CP1121), Buenos Aires, Argentina.
| | - Clara Ventura
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Físico-Matemática, Laboratorio de Radioisótopos, Junín 954, subsuelo (CP1113), Buenos Aires, Argentina.
| | - Alejandro Carozzo
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, ININFA-CONICET, Laboratorio de Farmacología Molecular, Junín 954, PB, (CP1113), Buenos Aires, Argentina.
| | - Florencia Chiappini
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Laboratorio de Efectos Biológicos de Contaminantes Ambientales, Paraguay 2155, 5° piso, (CP1121), Buenos Aires, Argentina.
| | - Diana Kleiman de Pisarev
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Laboratorio de Efectos Biológicos de Contaminantes Ambientales, Paraguay 2155, 5° piso, (CP1121), Buenos Aires, Argentina.
| | - Natalia Fernández
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, ININFA-CONICET, Laboratorio de Farmacología Molecular, Junín 954, PB, (CP1113), Buenos Aires, Argentina.
| | - Claudia Cocca
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Físico-Matemática, Laboratorio de Radioisótopos, Junín 954, subsuelo (CP1113), Buenos Aires, Argentina.
| | - Andrea Randi
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Laboratorio de Efectos Biológicos de Contaminantes Ambientales, Paraguay 2155, 5° piso, (CP1121), Buenos Aires, Argentina.
| |
Collapse
|
15
|
Wang H, Lawson JD, Scorah N, Kamran R, Hixon MS, Atienza J, Dougan DR, Sabat M. Design, synthesis and optimization of novel Alk5 (activin-like kinase 5) inhibitors. Bioorg Med Chem Lett 2016; 26:4334-9. [PMID: 27460209 DOI: 10.1016/j.bmcl.2016.07.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 07/13/2016] [Accepted: 07/14/2016] [Indexed: 10/21/2022]
Abstract
Using SBDD, a series of 4-amino-7-azaindoles were discovered as a novel class of Alk5 inhibitors that are potent in both Alk5 enzymatic and cellular assays. Subsequently a ring cyclization strategy was utilized to improve ADME properties leading to the discovery of a series of 1H-imidazo[4,5-c]pyridin-2(3H)-one drug like Alk5 inhibitors.
Collapse
Affiliation(s)
- Haixia Wang
- Takeda California, 10410 Science Center Drive, San Diego, CA 92121, United States
| | - J David Lawson
- Takeda California, 10410 Science Center Drive, San Diego, CA 92121, United States
| | - Nick Scorah
- Takeda California, 10410 Science Center Drive, San Diego, CA 92121, United States
| | - Ruhi Kamran
- Takeda California, 10410 Science Center Drive, San Diego, CA 92121, United States
| | - Mark S Hixon
- Takeda California, 10410 Science Center Drive, San Diego, CA 92121, United States
| | - Joy Atienza
- Takeda California, 10410 Science Center Drive, San Diego, CA 92121, United States
| | - Douglas R Dougan
- Takeda California, 10410 Science Center Drive, San Diego, CA 92121, United States
| | - Mark Sabat
- Takeda California, 10410 Science Center Drive, San Diego, CA 92121, United States
| |
Collapse
|
16
|
Gao Y, Vincent DF, Davis AJ, Sansom OJ, Bartholin L, Li Q. Constitutively active transforming growth factor β receptor 1 in the mouse ovary promotes tumorigenesis. Oncotarget 2016; 7:40904-40918. [PMID: 27344183 PMCID: PMC5173031 DOI: 10.18632/oncotarget.10149] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 06/06/2016] [Indexed: 12/11/2022] Open
Abstract
Despite the well-established tumor suppressive role of TGFβ proteins, depletion of key TGFβ signaling components in the mouse ovary does not induce a growth advantage. To define the role of TGFβ signaling in ovarian tumorigenesis, we created a mouse model expressing a constitutively active TGFβ receptor 1 (TGFBR1) in ovarian somatic cells using conditional gain-of-function approach. Remarkably, these mice developed ovarian sex cord-stromal tumors with complete penetrance, leading to reproductive failure and mortality. The tumors expressed multiple granulosa cell markers and caused elevated serum inhibin and estradiol levels, reminiscent of granulosa cell tumors. Consistent with the tumorigenic effect, overactivation of TGFBR1 altered tumor microenvironment by promoting angiogenesis and enhanced ovarian cell proliferation, accompanied by impaired cell differentiation and dysregulated expression of critical genes in ovarian function. By further exploiting complementary genetic models, we substantiated our finding that constitutively active TGFBR1 is a potent oncogenic switch in mouse granulosa cells. In summary, overactivation of TGFBR1 drives gonadal tumor development. The TGFBR1 constitutively active mouse model phenocopies a number of morphological, hormonal, and molecular features of human granulosa cell tumors and are potentially valuable for preclinical testing of targeted therapies to treat granulosa cell tumors, a class of poorly defined ovarian malignancies.
Collapse
Affiliation(s)
- Yang Gao
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - David F. Vincent
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Anna Jane Davis
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Owen J. Sansom
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Laurent Bartholin
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Lyon, France
| | - Qinglei Li
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| |
Collapse
|
17
|
Hu L, Liu J, Li Z, Wang C, Nawshad A. Transforming growth factor-β1 activates ΔNp63/c-Myc to promote oral squamous cell carcinoma. Oral Surg Oral Med Oral Pathol Oral Radiol 2016; 122:460-482.e4. [PMID: 27567435 DOI: 10.1016/j.oooo.2016.05.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 05/19/2016] [Accepted: 05/24/2016] [Indexed: 01/21/2023]
Abstract
OBJECTIVE During the development of oral squamous cell carcinoma (OSCC), the transformed epithelial cells undergo increased proliferation resulting in tumor growth and invasion. Interestingly, throughout all phases of differentiation and progression to OSCC, transforming growth factor-β1 (TGF)-β1 induces cell cycle arrest or apoptosis; however, the role of TGF-β1 in promoting cancer cell proliferation has not been explored in detail. The purpose of this study was to identify the effect of TGF-β1 on OSCC cell proliferation. STUDY DESIGN Using both human OSCC samples and cell lines (UMSCC38 and UMSCC11B), we assessed protein, mRNA, gene expression, and protein-DNA interactions during OSCC progression. RESULTS Our results showed that TGF-β1 increased OSCC cell proliferation by upregulating the expression of ΔNp63 and c-Myc oncogenes. Although the basal OSCC cell proliferation is sustained by activating ΔNp63, increased induction of c-Myc causes unregulated OSCC cell proliferation. Following induction of the cell cycle by ΔNp63 and c-Myc, cancer cells that halt c-Myc activity undergo epithelial mesenchymal transition or invasion while those that continue to express ΔNp63/c-Myc undergo unlimited progression through the cell cycle. CONCLUSIONS OSCC proliferation is manifested by the induction of c-Myc in response to TGF-β1 signaling, which is essential for OSCC growth. Our data highlight the potential role of TGF-β1 in the induction of cancer progression and invasion of OSCC.
Collapse
Affiliation(s)
- Lihua Hu
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE, USA; Shandong Provincial Key Laboratory of Oral Biomedicine, Department of Orthodontics, School of Stomatology, Shandong University, Jinan, Shandong, P.R. China
| | - Jingpeng Liu
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE, USA
| | - Zhi Li
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE, USA
| | - Chunling Wang
- Shandong Provincial Key Laboratory of Oral Biomedicine, Department of Orthodontics, School of Stomatology, Shandong University, Jinan, Shandong, P.R. China
| | - Ali Nawshad
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE, USA.
| |
Collapse
|
18
|
Abstract
Metastatic spread of cancer cells from the primary tumors to distant vital organs, such as lung, liver, brain, and bone, is responsible for the majority of cancer-related deaths. Development of metastatic lesions is critically dependent on the interaction of tumor cells with the stromal microenvironment. As a multifunctional paracrine signaling factor that is abundantly produced by both tumor and stromal cells, TGFβ has been well established as an important mediator of tumor-stromal interaction during cancer metastasis. Imaging the in vivo dynamic of TGFβ signaling activity during cancer metastasis is critical for understanding the pathogenesis of the disease, and for the development of effective anti-metastasis treatments. In this chapter, I describe several xenograft methods to introduce human breast cancer cells into nude mice in order to generate spontaneous and experimental metastases, as well as the luciferase-based bioluminescence imaging method for quantitative imaging analysis of TGFβ signaling in tumor cells during metastasis.
Collapse
|
19
|
Kong FF, Zhu YL, Yuan HH, Wang JY, Zhao M, Gong XD, Liu F, Zhang WY, Wang CR, Jiang B. FOXM1 regulated by ERK pathway mediates TGF-β1-induced EMT in NSCLC. Oncol Res 2015; 22:29-37. [PMID: 25700356 PMCID: PMC7592790 DOI: 10.3727/096504014x14078436004987] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
FOXM1, a member of the Forkhead transcriptional family, plays an important role in the EMT process, and transforming growth factor-β1 (TGF-β1) has been identified as the most potent factor that can independently induce EMT in various types of cancer cells. Here we examine the important role of FOXM1 in TGF-β1-induced EMT and investigate the mechanism underlying the relationship between TGF-β1 and FOXM1. Lentivirus-mediated transfection was used to stably upregulate the expression of FOXM1, and a small interfering RNA (siRNA) was introduced to silence the expression of FOXM1. Transwell and wound-healing assays were then performed to assess the invasion and motility potential of non-small cell lung cancer (NSCLC) cells. The NSCLC cell lines exhibited EMT characteristics, including an elongated fibroblastoid shape, induced expression of EMT marker proteins, and increased migratory and invasive potential after induction with TGF-β1. The overexpression of FOXM1 enhanced TGF-β1-induced EMT in NSCLC cells. Knockdown of FOXM1 reversed TGF-β1-induced EMT in NSCLC cell lines but had no effect on the phosphorylation level of ERK. Additionally, U0126, an ERK signaling inhibitor, exerted a reversible effect on TGF-β1-induced EMT and inhibited FOXM1 expression. FOXM1 regulated by the ERK pathway can mediate TGF-β1-induced EMT in NSCLC and is a potential target for the treatment of NSCLC.
Collapse
Affiliation(s)
- Fei-Fei Kong
- Department of Oncology, Shanghai 3rd Peoples Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - You-Long Zhu
- Department of General Surgery, Shanghai 3rd Peoples Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Hai-Hua Yuan
- Department of Oncology, Shanghai 3rd Peoples Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Jiong-Yi Wang
- Department of Oncology, Shanghai 3rd Peoples Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Mei Zhao
- Department of Oncology, Shanghai 3rd Peoples Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Xiao-Di Gong
- Department of Oncology, Shanghai 3rd Peoples Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Feng Liu
- Department of Oncology, Shanghai 3rd Peoples Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Wen-Ying Zhang
- Department of Oncology, Shanghai 3rd Peoples Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Cong-Rong Wang
- Metabolic Disease Bio-Bank, Shanghai Jiao Tong University Affiliated Sixth Peoples Hospital, Shanghai Diabetes Institute,Shanghai Key Laboratory of Diabetes MellitusShanghaiChina
| | - Bin Jiang
- Department of Oncology, Shanghai 3rd Peoples Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
20
|
Wang SS, Jiang J, Liang XH, Tang YL. Links between cancer stem cells and epithelial-mesenchymal transition. Onco Targets Ther 2015; 8:2973-80. [PMID: 26527883 PMCID: PMC4621173 DOI: 10.2147/ott.s91863] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The epithelial-mesenchymal transition (EMT) has been reported to be an important program that is often activated during the process of cancer invasion and metastasis. Cancer stem cells (CSCs) that can initiate and maintain cancer are also involved in invasion and metastasis of cancer. Recently, insights into the molecular mechanisms and functional features of mesenchymal cells have been greatly colored by findings that some of them have been endowed with the self-renewal trait associated with normal tissue stem cells and CSCs. Among cancer cells experiencing EMT, only some of the most competent CSCs will succeed in planting in another organ. In this paper, we review the molecular mechanism behind the link of EMT and CSCs in cancer progression.
Collapse
Affiliation(s)
- Sha-Sha Wang
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, People's Republic of China ; Department of Oral and Maxillofacial Surgery, Sichuan University, Chengdu, People's Republic of China
| | - Jian Jiang
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, People's Republic of China ; Department of Oral and Maxillofacial Surgery, Sichuan University, Chengdu, People's Republic of China
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, People's Republic of China ; Department of Oral and Maxillofacial Surgery, Sichuan University, Chengdu, People's Republic of China
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, People's Republic of China ; Department of Oral Pathology, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
21
|
Herbertz S, Sawyer JS, Stauber AJ, Gueorguieva I, Driscoll KE, Estrem ST, Cleverly AL, Desaiah D, Guba SC, Benhadji KA, Slapak CA, Lahn MM. Clinical development of galunisertib (LY2157299 monohydrate), a small molecule inhibitor of transforming growth factor-beta signaling pathway. Drug Des Devel Ther 2015; 9:4479-99. [PMID: 26309397 PMCID: PMC4539082 DOI: 10.2147/dddt.s86621] [Citation(s) in RCA: 248] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Transforming growth factor-beta (TGF-β) signaling regulates a wide range of biological processes. TGF-β plays an important role in tumorigenesis and contributes to the hallmarks of cancer, including tumor proliferation, invasion and metastasis, inflammation, angiogenesis, and escape of immune surveillance. There are several pharmacological approaches to block TGF-β signaling, such as monoclonal antibodies, vaccines, antisense oligonucleotides, and small molecule inhibitors. Galunisertib (LY2157299 monohydrate) is an oral small molecule inhibitor of the TGF-β receptor I kinase that specifically downregulates the phosphorylation of SMAD2, abrogating activation of the canonical pathway. Furthermore, galunisertib has antitumor activity in tumor-bearing animal models such as breast, colon, lung cancers, and hepatocellular carcinoma. Continuous long-term exposure to galunisertib caused cardiac toxicities in animals requiring adoption of a pharmacokinetic/pharmacodynamic-based dosing strategy to allow further development. The use of such a pharmacokinetic/pharmacodynamic model defined a therapeutic window with an appropriate safety profile that enabled the clinical investigation of galunisertib. These efforts resulted in an intermittent dosing regimen (14 days on/14 days off, on a 28-day cycle) of galunisertib for all ongoing trials. Galunisertib is being investigated either as monotherapy or in combination with standard antitumor regimens (including nivolumab) in patients with cancer with high unmet medical needs such as glioblastoma, pancreatic cancer, and hepatocellular carcinoma. The present review summarizes the past and current experiences with different pharmacological treatments that enabled galunisertib to be investigated in patients.
Collapse
Affiliation(s)
| | - J Scott Sawyer
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Anja J Stauber
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | | | - Kyla E Driscoll
- Lilly Research Laboratories, Eli Lilly and Company, New York, NY, USA
| | - Shawn T Estrem
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Ann L Cleverly
- Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, UK
| | - Durisala Desaiah
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Susan C Guba
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Karim A Benhadji
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | | | - Michael M Lahn
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| |
Collapse
|
22
|
Bill R, Christofori G. The relevance of EMT in breast cancer metastasis: Correlation or causality? FEBS Lett 2015; 589:1577-87. [DOI: 10.1016/j.febslet.2015.05.002] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 05/06/2015] [Accepted: 05/06/2015] [Indexed: 12/22/2022]
|
23
|
Woods LT, Camden JM, El-Sayed FG, Khalafalla MG, Petris MJ, Erb L, Weisman GA. Increased Expression of TGF-β Signaling Components in a Mouse Model of Fibrosis Induced by Submandibular Gland Duct Ligation. PLoS One 2015; 10:e0123641. [PMID: 25955532 PMCID: PMC4425516 DOI: 10.1371/journal.pone.0123641] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 02/21/2015] [Indexed: 02/06/2023] Open
Abstract
Transforming growth factor-β (TGF-β) is a multi-functional cytokine with a well-described role in the regulation of tissue fibrosis and regeneration in the liver, kidney and lung. Submandibular gland (SMG) duct ligation and subsequent deligation in rodents is a classical model for studying salivary gland damage and regeneration. While previous studies suggest that TGF-β may contribute to salivary gland fibrosis, the expression of TGF-β signaling components has not been investigated in relation to mouse SMG duct ligation-induced fibrosis and regeneration following ductal deligation. Following a 7 day SMG duct ligation, TGF-β1 and TGF-β3 were significantly upregulated in the SMG, as were TGF-β receptor 1 and downstream Smad family transcription factors in salivary acinar cells, but not in ductal cells. In acinar cells, duct ligation also led to upregulation of snail, a Smad-activated E-cadherin repressor and regulator of epithelial-mesenchymal transition, whereas in ductal cells upregulation of E-cadherin was observed while snail expression was unchanged. Upregulation of these TGF-β signaling components correlated with upregulation of fibrosis markers collagen 1 and fibronectin, responses that were inhibited by administration of the TGF-β receptor 1 inhibitors SB431542 or GW788388. After SMG regeneration following a 28 day duct deligation, TGF-β signaling components and epithelial-mesenchymal transition markers returned to levels similar to non-ligated controls. The results from this study indicate that increased TGF-β signaling contributes to duct ligation-induced changes in salivary epithelium that correlate with glandular fibrosis. Furthermore, the reversibility of enhanced TGF-β signaling in acinar cells of duct-ligated mouse SMG after deligation indicates that this is an ideal model for studying TGF-β signaling mechanisms in salivary epithelium as well as mechanisms of fibrosis initiation and their resolution.
Collapse
Affiliation(s)
- Lucas T. Woods
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
| | - Jean M. Camden
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
| | - Farid G. El-Sayed
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
| | - Mahmoud G. Khalafalla
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
| | - Michael J. Petris
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
- Department of Nutritional Sciences and Exercise Physiology, University of Missouri, Columbia, Missouri, United States of America
| | - Laurie Erb
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
| | - Gary A. Weisman
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
- * E-mail:
| |
Collapse
|
24
|
Novitskiy SV, Forrester E, Pickup MW, Gorska AE, Chytil A, Aakre M, Polosukhina D, Owens P, Yusupova DR, Zhao Z, Ye F, Shyr Y, Moses HL. Attenuated transforming growth factor beta signaling promotes metastasis in a model of HER2 mammary carcinogenesis. Breast Cancer Res 2014; 16:425. [PMID: 25280532 PMCID: PMC4303109 DOI: 10.1186/s13058-014-0425-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 08/05/2014] [Indexed: 01/08/2023] Open
Abstract
Introduction Transforming growth factor beta (TGFβ) plays a major role in the regulation of tumor initiation, progression, and metastasis. It is depended on the type II TGFβ receptor (TβRII) for signaling. Previously, we have shown that deletion of TβRII in mammary epithelial of MMTV-PyMT mice results in shortened tumor latency and increased lung metastases. However, active TGFβ signaling increased the number of circulating tumor cells and metastases in MMTV-Neu mice. In the current study, we describe a newly discovered connection between attenuated TGFβ signaling and human epidermal growth factor receptor 2 (HER2) signaling in mammary tumor progression. Methods All studies were performed on MMTV-Neu mice with and without dominant-negative TβRII (DNIIR) in mammary epithelium. Mammary tumors were analyzed by flow cytometry, immunohistochemistry, and immunofluorescence staining. The levels of secreted proteins were measured by enzyme-linked immunosorbent assay. Whole-lung mount staining was used to quantitate lung metastasis. The Cancer Genome Atlas (TCGA) datasets were used to determine the relevance of our findings to human breast cancer. Results Attenuated TGFβ signaling led to a delay tumor onset, but increased the number of metastases in MMTVNeu/DNIIR mice. The DNIIR tumors were characterized by increased vasculogenesis, vessel leakage, and increased expression of vascular endothelial growth factor (VEGF). During DNIIR tumor progression, both the levels of CXCL1/5 and the number of CD11b+Gr1+ cells and T cells decreased. Analysis of TCGA datasets demonstrated a significant negative correlation between TGFBR2 and VEGF genes expression. Higher VEGFA expression correlated with shorter distant metastasis-free survival only in HER2+ patients with no differences in HER2-, estrogen receptor +/- or progesterone receptor +/- breast cancer patients. Conclusion Our studies provide insights into a novel mechanism by which epithelial TGFβ signaling modulates the tumor microenvironment, and by which it is involved in lung metastasis in HER2+ breast cancer patients. The effects of pharmacological targeting of the TGFβ pathway in vivo during tumor progression remain controversial. The targeting of TGFβ signaling should be a viable option, but because VEGF has a protumorigenic effect on HER2+ tumors, the targeting of this protein could be considered when it is associated with attenuated TGFβ signaling. Electronic supplementary material The online version of this article (doi:10.1186/s13058-014-0425-7) contains supplementary material, which is available to authorized users.
Collapse
|
25
|
Moore KM, Thomas GJ, Duffy SW, Warwick J, Gabe R, Chou P, Ellis IO, Green AR, Haider S, Brouilette K, Saha A, Vallath S, Bowen R, Chelala C, Eccles D, Tapper WJ, Thompson AM, Quinlan P, Jordan L, Gillett C, Brentnall A, Violette S, Weinreb PH, Kendrew J, Barry ST, Hart IR, Jones JL, Marshall JF. Therapeutic targeting of integrin αvβ6 in breast cancer. J Natl Cancer Inst 2014; 106:dju169. [PMID: 24974129 PMCID: PMC4151855 DOI: 10.1093/jnci/dju169] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 05/06/2014] [Accepted: 05/13/2014] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Integrin αvβ6 promotes migration, invasion, and survival of cancer cells; however, the relevance and role of αvβ6 has yet to be elucidated in breast cancer. METHODS Protein expression of integrin subunit beta6 (β6) was measured in breast cancers by immunohistochemistry (n > 2000) and ITGB6 mRNA expression measured in the Molecular Taxonomy of Breast Cancer International Consortium dataset. Overall survival was assessed using Kaplan Meier curves, and bioinformatics statistical analyses were performed (Cox proportional hazards model, Wald test, and Chi-square test of association). Using antibody (264RAD) blockade and siRNA knockdown of β6 in breast cell lines, the role of αvβ6 in Human Epidermal Growth Factor Receptor 2 (HER2) biology (expression, proliferation, invasion, growth in vivo) was assessed by flow cytometry, MTT, Transwell invasion, proximity ligation assay, and xenografts (n ≥ 3), respectively. A student's t-test was used for two variables; three-plus variables used one-way analysis of variance with Bonferroni's Multiple Comparison Test. Xenograft growth was analyzed using linear mixed model analysis, followed by Wald testing and survival, analyzed using the Log-Rank test. All statistical tests were two sided. RESULTS High expression of either the mRNA or protein for the integrin subunit β6 was associated with very poor survival (HR = 1.60, 95% CI = 1.19 to 2.15, P = .002) and increased metastases to distant sites. Co-expression of β6 and HER2 was associated with worse prognosis (HR = 1.97, 95% CI = 1.16 to 3.35, P = .01). Monotherapy with 264RAD or trastuzumab slowed growth of MCF-7/HER2-18 and BT-474 xenografts similarly (P < .001), but combining 264RAD with trastuzumab effectively stopped tumor growth, even in trastuzumab-resistant MCF-7/HER2-18 xenografts. CONCLUSIONS Targeting αvβ6 with 264RAD alone or in combination with trastuzumab may provide a novel therapy for treating high-risk and trastuzumab-resistant breast cancer patients.
Collapse
Affiliation(s)
- Kate M Moore
- Affiliations of authors: Centre for Tumour Biology (KMM, GJT, KB, AS, SV, RB, IRH, JLJ, JFM), Cancer Screening Evaluation Group (SWD, JW, RG, PC), and Molecular Oncology and Imaging (SH, CC), John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, UK; Department of Histopathology, Molecular Medical Sciences, Nottingham City Hospital NHS Trust, Nottingham, UK (IOE, ARG); Cancer Sciences Division, Southampton General Hospital, Southampton, UK (GJT, DE, WJT); Department of Surgery (AMT, PQ) and Department of Pathology (LJ), Ninewells Hospital and Medical School, Dundee, UK; Hedley Atkins Breast Pathology Laboratory, Guy's Hospital, London, UK (CG); Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventative Medicine, Queen Mary University of London, London, UK (AB); Biogen Idec, Cambridge, MA (SV, PHW); Oncology iMED, AstraZeneca, Macclesfield, UK (JK, STB)
| | - Gareth J Thomas
- Affiliations of authors: Centre for Tumour Biology (KMM, GJT, KB, AS, SV, RB, IRH, JLJ, JFM), Cancer Screening Evaluation Group (SWD, JW, RG, PC), and Molecular Oncology and Imaging (SH, CC), John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, UK; Department of Histopathology, Molecular Medical Sciences, Nottingham City Hospital NHS Trust, Nottingham, UK (IOE, ARG); Cancer Sciences Division, Southampton General Hospital, Southampton, UK (GJT, DE, WJT); Department of Surgery (AMT, PQ) and Department of Pathology (LJ), Ninewells Hospital and Medical School, Dundee, UK; Hedley Atkins Breast Pathology Laboratory, Guy's Hospital, London, UK (CG); Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventative Medicine, Queen Mary University of London, London, UK (AB); Biogen Idec, Cambridge, MA (SV, PHW); Oncology iMED, AstraZeneca, Macclesfield, UK (JK, STB)
| | - Stephen W Duffy
- Affiliations of authors: Centre for Tumour Biology (KMM, GJT, KB, AS, SV, RB, IRH, JLJ, JFM), Cancer Screening Evaluation Group (SWD, JW, RG, PC), and Molecular Oncology and Imaging (SH, CC), John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, UK; Department of Histopathology, Molecular Medical Sciences, Nottingham City Hospital NHS Trust, Nottingham, UK (IOE, ARG); Cancer Sciences Division, Southampton General Hospital, Southampton, UK (GJT, DE, WJT); Department of Surgery (AMT, PQ) and Department of Pathology (LJ), Ninewells Hospital and Medical School, Dundee, UK; Hedley Atkins Breast Pathology Laboratory, Guy's Hospital, London, UK (CG); Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventative Medicine, Queen Mary University of London, London, UK (AB); Biogen Idec, Cambridge, MA (SV, PHW); Oncology iMED, AstraZeneca, Macclesfield, UK (JK, STB)
| | - Jane Warwick
- Affiliations of authors: Centre for Tumour Biology (KMM, GJT, KB, AS, SV, RB, IRH, JLJ, JFM), Cancer Screening Evaluation Group (SWD, JW, RG, PC), and Molecular Oncology and Imaging (SH, CC), John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, UK; Department of Histopathology, Molecular Medical Sciences, Nottingham City Hospital NHS Trust, Nottingham, UK (IOE, ARG); Cancer Sciences Division, Southampton General Hospital, Southampton, UK (GJT, DE, WJT); Department of Surgery (AMT, PQ) and Department of Pathology (LJ), Ninewells Hospital and Medical School, Dundee, UK; Hedley Atkins Breast Pathology Laboratory, Guy's Hospital, London, UK (CG); Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventative Medicine, Queen Mary University of London, London, UK (AB); Biogen Idec, Cambridge, MA (SV, PHW); Oncology iMED, AstraZeneca, Macclesfield, UK (JK, STB)
| | - Rhian Gabe
- Affiliations of authors: Centre for Tumour Biology (KMM, GJT, KB, AS, SV, RB, IRH, JLJ, JFM), Cancer Screening Evaluation Group (SWD, JW, RG, PC), and Molecular Oncology and Imaging (SH, CC), John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, UK; Department of Histopathology, Molecular Medical Sciences, Nottingham City Hospital NHS Trust, Nottingham, UK (IOE, ARG); Cancer Sciences Division, Southampton General Hospital, Southampton, UK (GJT, DE, WJT); Department of Surgery (AMT, PQ) and Department of Pathology (LJ), Ninewells Hospital and Medical School, Dundee, UK; Hedley Atkins Breast Pathology Laboratory, Guy's Hospital, London, UK (CG); Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventative Medicine, Queen Mary University of London, London, UK (AB); Biogen Idec, Cambridge, MA (SV, PHW); Oncology iMED, AstraZeneca, Macclesfield, UK (JK, STB)
| | - Patrick Chou
- Affiliations of authors: Centre for Tumour Biology (KMM, GJT, KB, AS, SV, RB, IRH, JLJ, JFM), Cancer Screening Evaluation Group (SWD, JW, RG, PC), and Molecular Oncology and Imaging (SH, CC), John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, UK; Department of Histopathology, Molecular Medical Sciences, Nottingham City Hospital NHS Trust, Nottingham, UK (IOE, ARG); Cancer Sciences Division, Southampton General Hospital, Southampton, UK (GJT, DE, WJT); Department of Surgery (AMT, PQ) and Department of Pathology (LJ), Ninewells Hospital and Medical School, Dundee, UK; Hedley Atkins Breast Pathology Laboratory, Guy's Hospital, London, UK (CG); Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventative Medicine, Queen Mary University of London, London, UK (AB); Biogen Idec, Cambridge, MA (SV, PHW); Oncology iMED, AstraZeneca, Macclesfield, UK (JK, STB)
| | - Ian O Ellis
- Affiliations of authors: Centre for Tumour Biology (KMM, GJT, KB, AS, SV, RB, IRH, JLJ, JFM), Cancer Screening Evaluation Group (SWD, JW, RG, PC), and Molecular Oncology and Imaging (SH, CC), John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, UK; Department of Histopathology, Molecular Medical Sciences, Nottingham City Hospital NHS Trust, Nottingham, UK (IOE, ARG); Cancer Sciences Division, Southampton General Hospital, Southampton, UK (GJT, DE, WJT); Department of Surgery (AMT, PQ) and Department of Pathology (LJ), Ninewells Hospital and Medical School, Dundee, UK; Hedley Atkins Breast Pathology Laboratory, Guy's Hospital, London, UK (CG); Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventative Medicine, Queen Mary University of London, London, UK (AB); Biogen Idec, Cambridge, MA (SV, PHW); Oncology iMED, AstraZeneca, Macclesfield, UK (JK, STB)
| | - Andrew R Green
- Affiliations of authors: Centre for Tumour Biology (KMM, GJT, KB, AS, SV, RB, IRH, JLJ, JFM), Cancer Screening Evaluation Group (SWD, JW, RG, PC), and Molecular Oncology and Imaging (SH, CC), John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, UK; Department of Histopathology, Molecular Medical Sciences, Nottingham City Hospital NHS Trust, Nottingham, UK (IOE, ARG); Cancer Sciences Division, Southampton General Hospital, Southampton, UK (GJT, DE, WJT); Department of Surgery (AMT, PQ) and Department of Pathology (LJ), Ninewells Hospital and Medical School, Dundee, UK; Hedley Atkins Breast Pathology Laboratory, Guy's Hospital, London, UK (CG); Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventative Medicine, Queen Mary University of London, London, UK (AB); Biogen Idec, Cambridge, MA (SV, PHW); Oncology iMED, AstraZeneca, Macclesfield, UK (JK, STB)
| | - Syed Haider
- Affiliations of authors: Centre for Tumour Biology (KMM, GJT, KB, AS, SV, RB, IRH, JLJ, JFM), Cancer Screening Evaluation Group (SWD, JW, RG, PC), and Molecular Oncology and Imaging (SH, CC), John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, UK; Department of Histopathology, Molecular Medical Sciences, Nottingham City Hospital NHS Trust, Nottingham, UK (IOE, ARG); Cancer Sciences Division, Southampton General Hospital, Southampton, UK (GJT, DE, WJT); Department of Surgery (AMT, PQ) and Department of Pathology (LJ), Ninewells Hospital and Medical School, Dundee, UK; Hedley Atkins Breast Pathology Laboratory, Guy's Hospital, London, UK (CG); Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventative Medicine, Queen Mary University of London, London, UK (AB); Biogen Idec, Cambridge, MA (SV, PHW); Oncology iMED, AstraZeneca, Macclesfield, UK (JK, STB)
| | - Kellie Brouilette
- Affiliations of authors: Centre for Tumour Biology (KMM, GJT, KB, AS, SV, RB, IRH, JLJ, JFM), Cancer Screening Evaluation Group (SWD, JW, RG, PC), and Molecular Oncology and Imaging (SH, CC), John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, UK; Department of Histopathology, Molecular Medical Sciences, Nottingham City Hospital NHS Trust, Nottingham, UK (IOE, ARG); Cancer Sciences Division, Southampton General Hospital, Southampton, UK (GJT, DE, WJT); Department of Surgery (AMT, PQ) and Department of Pathology (LJ), Ninewells Hospital and Medical School, Dundee, UK; Hedley Atkins Breast Pathology Laboratory, Guy's Hospital, London, UK (CG); Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventative Medicine, Queen Mary University of London, London, UK (AB); Biogen Idec, Cambridge, MA (SV, PHW); Oncology iMED, AstraZeneca, Macclesfield, UK (JK, STB)
| | - Antonio Saha
- Affiliations of authors: Centre for Tumour Biology (KMM, GJT, KB, AS, SV, RB, IRH, JLJ, JFM), Cancer Screening Evaluation Group (SWD, JW, RG, PC), and Molecular Oncology and Imaging (SH, CC), John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, UK; Department of Histopathology, Molecular Medical Sciences, Nottingham City Hospital NHS Trust, Nottingham, UK (IOE, ARG); Cancer Sciences Division, Southampton General Hospital, Southampton, UK (GJT, DE, WJT); Department of Surgery (AMT, PQ) and Department of Pathology (LJ), Ninewells Hospital and Medical School, Dundee, UK; Hedley Atkins Breast Pathology Laboratory, Guy's Hospital, London, UK (CG); Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventative Medicine, Queen Mary University of London, London, UK (AB); Biogen Idec, Cambridge, MA (SV, PHW); Oncology iMED, AstraZeneca, Macclesfield, UK (JK, STB)
| | - Sabari Vallath
- Affiliations of authors: Centre for Tumour Biology (KMM, GJT, KB, AS, SV, RB, IRH, JLJ, JFM), Cancer Screening Evaluation Group (SWD, JW, RG, PC), and Molecular Oncology and Imaging (SH, CC), John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, UK; Department of Histopathology, Molecular Medical Sciences, Nottingham City Hospital NHS Trust, Nottingham, UK (IOE, ARG); Cancer Sciences Division, Southampton General Hospital, Southampton, UK (GJT, DE, WJT); Department of Surgery (AMT, PQ) and Department of Pathology (LJ), Ninewells Hospital and Medical School, Dundee, UK; Hedley Atkins Breast Pathology Laboratory, Guy's Hospital, London, UK (CG); Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventative Medicine, Queen Mary University of London, London, UK (AB); Biogen Idec, Cambridge, MA (SV, PHW); Oncology iMED, AstraZeneca, Macclesfield, UK (JK, STB)
| | - Rebecca Bowen
- Affiliations of authors: Centre for Tumour Biology (KMM, GJT, KB, AS, SV, RB, IRH, JLJ, JFM), Cancer Screening Evaluation Group (SWD, JW, RG, PC), and Molecular Oncology and Imaging (SH, CC), John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, UK; Department of Histopathology, Molecular Medical Sciences, Nottingham City Hospital NHS Trust, Nottingham, UK (IOE, ARG); Cancer Sciences Division, Southampton General Hospital, Southampton, UK (GJT, DE, WJT); Department of Surgery (AMT, PQ) and Department of Pathology (LJ), Ninewells Hospital and Medical School, Dundee, UK; Hedley Atkins Breast Pathology Laboratory, Guy's Hospital, London, UK (CG); Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventative Medicine, Queen Mary University of London, London, UK (AB); Biogen Idec, Cambridge, MA (SV, PHW); Oncology iMED, AstraZeneca, Macclesfield, UK (JK, STB)
| | - Claude Chelala
- Affiliations of authors: Centre for Tumour Biology (KMM, GJT, KB, AS, SV, RB, IRH, JLJ, JFM), Cancer Screening Evaluation Group (SWD, JW, RG, PC), and Molecular Oncology and Imaging (SH, CC), John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, UK; Department of Histopathology, Molecular Medical Sciences, Nottingham City Hospital NHS Trust, Nottingham, UK (IOE, ARG); Cancer Sciences Division, Southampton General Hospital, Southampton, UK (GJT, DE, WJT); Department of Surgery (AMT, PQ) and Department of Pathology (LJ), Ninewells Hospital and Medical School, Dundee, UK; Hedley Atkins Breast Pathology Laboratory, Guy's Hospital, London, UK (CG); Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventative Medicine, Queen Mary University of London, London, UK (AB); Biogen Idec, Cambridge, MA (SV, PHW); Oncology iMED, AstraZeneca, Macclesfield, UK (JK, STB)
| | - Diana Eccles
- Affiliations of authors: Centre for Tumour Biology (KMM, GJT, KB, AS, SV, RB, IRH, JLJ, JFM), Cancer Screening Evaluation Group (SWD, JW, RG, PC), and Molecular Oncology and Imaging (SH, CC), John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, UK; Department of Histopathology, Molecular Medical Sciences, Nottingham City Hospital NHS Trust, Nottingham, UK (IOE, ARG); Cancer Sciences Division, Southampton General Hospital, Southampton, UK (GJT, DE, WJT); Department of Surgery (AMT, PQ) and Department of Pathology (LJ), Ninewells Hospital and Medical School, Dundee, UK; Hedley Atkins Breast Pathology Laboratory, Guy's Hospital, London, UK (CG); Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventative Medicine, Queen Mary University of London, London, UK (AB); Biogen Idec, Cambridge, MA (SV, PHW); Oncology iMED, AstraZeneca, Macclesfield, UK (JK, STB)
| | - William J Tapper
- Affiliations of authors: Centre for Tumour Biology (KMM, GJT, KB, AS, SV, RB, IRH, JLJ, JFM), Cancer Screening Evaluation Group (SWD, JW, RG, PC), and Molecular Oncology and Imaging (SH, CC), John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, UK; Department of Histopathology, Molecular Medical Sciences, Nottingham City Hospital NHS Trust, Nottingham, UK (IOE, ARG); Cancer Sciences Division, Southampton General Hospital, Southampton, UK (GJT, DE, WJT); Department of Surgery (AMT, PQ) and Department of Pathology (LJ), Ninewells Hospital and Medical School, Dundee, UK; Hedley Atkins Breast Pathology Laboratory, Guy's Hospital, London, UK (CG); Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventative Medicine, Queen Mary University of London, London, UK (AB); Biogen Idec, Cambridge, MA (SV, PHW); Oncology iMED, AstraZeneca, Macclesfield, UK (JK, STB)
| | - Alastair M Thompson
- Affiliations of authors: Centre for Tumour Biology (KMM, GJT, KB, AS, SV, RB, IRH, JLJ, JFM), Cancer Screening Evaluation Group (SWD, JW, RG, PC), and Molecular Oncology and Imaging (SH, CC), John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, UK; Department of Histopathology, Molecular Medical Sciences, Nottingham City Hospital NHS Trust, Nottingham, UK (IOE, ARG); Cancer Sciences Division, Southampton General Hospital, Southampton, UK (GJT, DE, WJT); Department of Surgery (AMT, PQ) and Department of Pathology (LJ), Ninewells Hospital and Medical School, Dundee, UK; Hedley Atkins Breast Pathology Laboratory, Guy's Hospital, London, UK (CG); Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventative Medicine, Queen Mary University of London, London, UK (AB); Biogen Idec, Cambridge, MA (SV, PHW); Oncology iMED, AstraZeneca, Macclesfield, UK (JK, STB)
| | - Phillip Quinlan
- Affiliations of authors: Centre for Tumour Biology (KMM, GJT, KB, AS, SV, RB, IRH, JLJ, JFM), Cancer Screening Evaluation Group (SWD, JW, RG, PC), and Molecular Oncology and Imaging (SH, CC), John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, UK; Department of Histopathology, Molecular Medical Sciences, Nottingham City Hospital NHS Trust, Nottingham, UK (IOE, ARG); Cancer Sciences Division, Southampton General Hospital, Southampton, UK (GJT, DE, WJT); Department of Surgery (AMT, PQ) and Department of Pathology (LJ), Ninewells Hospital and Medical School, Dundee, UK; Hedley Atkins Breast Pathology Laboratory, Guy's Hospital, London, UK (CG); Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventative Medicine, Queen Mary University of London, London, UK (AB); Biogen Idec, Cambridge, MA (SV, PHW); Oncology iMED, AstraZeneca, Macclesfield, UK (JK, STB)
| | - Lee Jordan
- Affiliations of authors: Centre for Tumour Biology (KMM, GJT, KB, AS, SV, RB, IRH, JLJ, JFM), Cancer Screening Evaluation Group (SWD, JW, RG, PC), and Molecular Oncology and Imaging (SH, CC), John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, UK; Department of Histopathology, Molecular Medical Sciences, Nottingham City Hospital NHS Trust, Nottingham, UK (IOE, ARG); Cancer Sciences Division, Southampton General Hospital, Southampton, UK (GJT, DE, WJT); Department of Surgery (AMT, PQ) and Department of Pathology (LJ), Ninewells Hospital and Medical School, Dundee, UK; Hedley Atkins Breast Pathology Laboratory, Guy's Hospital, London, UK (CG); Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventative Medicine, Queen Mary University of London, London, UK (AB); Biogen Idec, Cambridge, MA (SV, PHW); Oncology iMED, AstraZeneca, Macclesfield, UK (JK, STB)
| | - Cheryl Gillett
- Affiliations of authors: Centre for Tumour Biology (KMM, GJT, KB, AS, SV, RB, IRH, JLJ, JFM), Cancer Screening Evaluation Group (SWD, JW, RG, PC), and Molecular Oncology and Imaging (SH, CC), John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, UK; Department of Histopathology, Molecular Medical Sciences, Nottingham City Hospital NHS Trust, Nottingham, UK (IOE, ARG); Cancer Sciences Division, Southampton General Hospital, Southampton, UK (GJT, DE, WJT); Department of Surgery (AMT, PQ) and Department of Pathology (LJ), Ninewells Hospital and Medical School, Dundee, UK; Hedley Atkins Breast Pathology Laboratory, Guy's Hospital, London, UK (CG); Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventative Medicine, Queen Mary University of London, London, UK (AB); Biogen Idec, Cambridge, MA (SV, PHW); Oncology iMED, AstraZeneca, Macclesfield, UK (JK, STB)
| | - Adam Brentnall
- Affiliations of authors: Centre for Tumour Biology (KMM, GJT, KB, AS, SV, RB, IRH, JLJ, JFM), Cancer Screening Evaluation Group (SWD, JW, RG, PC), and Molecular Oncology and Imaging (SH, CC), John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, UK; Department of Histopathology, Molecular Medical Sciences, Nottingham City Hospital NHS Trust, Nottingham, UK (IOE, ARG); Cancer Sciences Division, Southampton General Hospital, Southampton, UK (GJT, DE, WJT); Department of Surgery (AMT, PQ) and Department of Pathology (LJ), Ninewells Hospital and Medical School, Dundee, UK; Hedley Atkins Breast Pathology Laboratory, Guy's Hospital, London, UK (CG); Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventative Medicine, Queen Mary University of London, London, UK (AB); Biogen Idec, Cambridge, MA (SV, PHW); Oncology iMED, AstraZeneca, Macclesfield, UK (JK, STB)
| | - Shelia Violette
- Affiliations of authors: Centre for Tumour Biology (KMM, GJT, KB, AS, SV, RB, IRH, JLJ, JFM), Cancer Screening Evaluation Group (SWD, JW, RG, PC), and Molecular Oncology and Imaging (SH, CC), John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, UK; Department of Histopathology, Molecular Medical Sciences, Nottingham City Hospital NHS Trust, Nottingham, UK (IOE, ARG); Cancer Sciences Division, Southampton General Hospital, Southampton, UK (GJT, DE, WJT); Department of Surgery (AMT, PQ) and Department of Pathology (LJ), Ninewells Hospital and Medical School, Dundee, UK; Hedley Atkins Breast Pathology Laboratory, Guy's Hospital, London, UK (CG); Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventative Medicine, Queen Mary University of London, London, UK (AB); Biogen Idec, Cambridge, MA (SV, PHW); Oncology iMED, AstraZeneca, Macclesfield, UK (JK, STB)
| | - Paul H Weinreb
- Affiliations of authors: Centre for Tumour Biology (KMM, GJT, KB, AS, SV, RB, IRH, JLJ, JFM), Cancer Screening Evaluation Group (SWD, JW, RG, PC), and Molecular Oncology and Imaging (SH, CC), John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, UK; Department of Histopathology, Molecular Medical Sciences, Nottingham City Hospital NHS Trust, Nottingham, UK (IOE, ARG); Cancer Sciences Division, Southampton General Hospital, Southampton, UK (GJT, DE, WJT); Department of Surgery (AMT, PQ) and Department of Pathology (LJ), Ninewells Hospital and Medical School, Dundee, UK; Hedley Atkins Breast Pathology Laboratory, Guy's Hospital, London, UK (CG); Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventative Medicine, Queen Mary University of London, London, UK (AB); Biogen Idec, Cambridge, MA (SV, PHW); Oncology iMED, AstraZeneca, Macclesfield, UK (JK, STB)
| | - Jane Kendrew
- Affiliations of authors: Centre for Tumour Biology (KMM, GJT, KB, AS, SV, RB, IRH, JLJ, JFM), Cancer Screening Evaluation Group (SWD, JW, RG, PC), and Molecular Oncology and Imaging (SH, CC), John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, UK; Department of Histopathology, Molecular Medical Sciences, Nottingham City Hospital NHS Trust, Nottingham, UK (IOE, ARG); Cancer Sciences Division, Southampton General Hospital, Southampton, UK (GJT, DE, WJT); Department of Surgery (AMT, PQ) and Department of Pathology (LJ), Ninewells Hospital and Medical School, Dundee, UK; Hedley Atkins Breast Pathology Laboratory, Guy's Hospital, London, UK (CG); Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventative Medicine, Queen Mary University of London, London, UK (AB); Biogen Idec, Cambridge, MA (SV, PHW); Oncology iMED, AstraZeneca, Macclesfield, UK (JK, STB)
| | - Simon T Barry
- Affiliations of authors: Centre for Tumour Biology (KMM, GJT, KB, AS, SV, RB, IRH, JLJ, JFM), Cancer Screening Evaluation Group (SWD, JW, RG, PC), and Molecular Oncology and Imaging (SH, CC), John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, UK; Department of Histopathology, Molecular Medical Sciences, Nottingham City Hospital NHS Trust, Nottingham, UK (IOE, ARG); Cancer Sciences Division, Southampton General Hospital, Southampton, UK (GJT, DE, WJT); Department of Surgery (AMT, PQ) and Department of Pathology (LJ), Ninewells Hospital and Medical School, Dundee, UK; Hedley Atkins Breast Pathology Laboratory, Guy's Hospital, London, UK (CG); Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventative Medicine, Queen Mary University of London, London, UK (AB); Biogen Idec, Cambridge, MA (SV, PHW); Oncology iMED, AstraZeneca, Macclesfield, UK (JK, STB)
| | - Ian R Hart
- Affiliations of authors: Centre for Tumour Biology (KMM, GJT, KB, AS, SV, RB, IRH, JLJ, JFM), Cancer Screening Evaluation Group (SWD, JW, RG, PC), and Molecular Oncology and Imaging (SH, CC), John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, UK; Department of Histopathology, Molecular Medical Sciences, Nottingham City Hospital NHS Trust, Nottingham, UK (IOE, ARG); Cancer Sciences Division, Southampton General Hospital, Southampton, UK (GJT, DE, WJT); Department of Surgery (AMT, PQ) and Department of Pathology (LJ), Ninewells Hospital and Medical School, Dundee, UK; Hedley Atkins Breast Pathology Laboratory, Guy's Hospital, London, UK (CG); Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventative Medicine, Queen Mary University of London, London, UK (AB); Biogen Idec, Cambridge, MA (SV, PHW); Oncology iMED, AstraZeneca, Macclesfield, UK (JK, STB)
| | - J Louise Jones
- Affiliations of authors: Centre for Tumour Biology (KMM, GJT, KB, AS, SV, RB, IRH, JLJ, JFM), Cancer Screening Evaluation Group (SWD, JW, RG, PC), and Molecular Oncology and Imaging (SH, CC), John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, UK; Department of Histopathology, Molecular Medical Sciences, Nottingham City Hospital NHS Trust, Nottingham, UK (IOE, ARG); Cancer Sciences Division, Southampton General Hospital, Southampton, UK (GJT, DE, WJT); Department of Surgery (AMT, PQ) and Department of Pathology (LJ), Ninewells Hospital and Medical School, Dundee, UK; Hedley Atkins Breast Pathology Laboratory, Guy's Hospital, London, UK (CG); Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventative Medicine, Queen Mary University of London, London, UK (AB); Biogen Idec, Cambridge, MA (SV, PHW); Oncology iMED, AstraZeneca, Macclesfield, UK (JK, STB)
| | - John F Marshall
- Affiliations of authors: Centre for Tumour Biology (KMM, GJT, KB, AS, SV, RB, IRH, JLJ, JFM), Cancer Screening Evaluation Group (SWD, JW, RG, PC), and Molecular Oncology and Imaging (SH, CC), John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, UK; Department of Histopathology, Molecular Medical Sciences, Nottingham City Hospital NHS Trust, Nottingham, UK (IOE, ARG); Cancer Sciences Division, Southampton General Hospital, Southampton, UK (GJT, DE, WJT); Department of Surgery (AMT, PQ) and Department of Pathology (LJ), Ninewells Hospital and Medical School, Dundee, UK; Hedley Atkins Breast Pathology Laboratory, Guy's Hospital, London, UK (CG); Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventative Medicine, Queen Mary University of London, London, UK (AB); Biogen Idec, Cambridge, MA (SV, PHW); Oncology iMED, AstraZeneca, Macclesfield, UK (JK, STB).
| |
Collapse
|
26
|
Mao Y, Poschke I, Kiessling R. Tumour-induced immune suppression: role of inflammatory mediators released by myelomonocytic cells. J Intern Med 2014; 276:154-70. [PMID: 24597954 DOI: 10.1111/joim.12229] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tumour-induced immune dysfunction is a serious challenge to immunotherapy for cancer, and intact adaptive and innate cellular immunity is key to its success. Myelomonocytic cells have a central role in this immune suppression, and tumour-associated macrophages, eosinophils, neutrophils and myeloid-derived suppressor cells have all been shown to be of major importance. These myelomonocytic cells secrete a broad repertoire of inflammatory mediators providing them with powerful tools to inhibit tumour-reactive T cells and natural killer cells; free oxygen radicals including reactive oxygen species and NO, arginase, indoleamine 2,3-dioxygenase, prostaglandins, the pro-inflammatory heterodimer S100A8/9 and cytokines, such as granulocyte-macrophage colony-stimulating factor and transforming growth factor-β, have proven particularly potent in suppressing antitumour cellular immunity. Determining which of these factors prevail in individual cancer patients and designing methods aimed at neutralization or inhibition of their effects on target tissues have the potential to greatly enhance the clinical efficacy of immunotherapy.
Collapse
Affiliation(s)
- Y Mao
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | | | | |
Collapse
|
27
|
Di HS, Wang LG, Wang GL, Zhou L, Yang YY. The Signaling Mechanism of TGF-β1 Induced Bovine Mammary Epithelial Cell Apoptosis. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2014; 25:304-10. [PMID: 25049567 PMCID: PMC4092962 DOI: 10.5713/ajas.2011.11251] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 11/13/2011] [Accepted: 11/02/2011] [Indexed: 01/19/2023]
Abstract
The present study showed that Transforming growth factor beta 1 (TGF-β1) can induce apoptosis of bovine mammary epithelial cells. This apoptosis was also observed with phosphorylation of Smad2/3 within 0.5–2 h. Afterwards the signal transferred into the nucleus. Moreover, intracellular free Ca2+ concentration was significantly elevated as well as Caspase-3 activated and DNA lysised, thereby inducing the programmed cell death. This signaling pathway of TGF-β1 was blocked by SB-431542 (10−2 μM) via inhibiting ALK-5 kinase activity, which thus reversed the anti-proliferation and apoptosis effect of TGF-β1 in mammary epithelial cells. These results indicated that TGF-β1 induced apoptosis of bovine mammary epithelial cells through the ALK-5-Smad2/3 pathway, which plays an important role in inhibiting survival of mammary epithelial cells. Moreover, intracellular Ca2+ also played a critical role in TGF-β1-induced cell apoptosis.
Collapse
Affiliation(s)
- He-Shuang Di
- Department of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China ; Jiangsu Animal Husbandry and Veterinary College, Taizhou 225300, China
| | - Li-Gang Wang
- Jiangsu Animal Husbandry and Veterinary College, Taizhou 225300, China
| | - Gen-Lin Wang
- Department of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Lei Zhou
- Department of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yuan-Yuan Yang
- Department of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| |
Collapse
|
28
|
Bernemann C, Hülsewig C, Ruckert C, Schäfer S, Blümel L, Hempel G, Götte M, Greve B, Barth PJ, Kiesel L, Liedtke C. Influence of secreted frizzled receptor protein 1 (SFRP1) on neoadjuvant chemotherapy in triple negative breast cancer does not rely on WNT signaling. Mol Cancer 2014; 13:174. [PMID: 25033833 PMCID: PMC4110378 DOI: 10.1186/1476-4598-13-174] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 07/09/2014] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Triple negative breast cancer (TNBC) is characterized by lack of expression of both estrogen and progesterone receptor as well as lack of overexpression or amplification of HER2. Despite an increased probability of response to chemotherapy, many patients resistant to current chemotherapy regimens suffer from a worse prognosis compared to other breast cancer subtypes. However, molecular determinants of response to chemotherapy specific to TNBC remain largely unknown. Thus, there is a high demand for biomarkers potentially stratifying triple negative breast cancer patients for neoadjuvant chemotherapies or alternative therapies. METHODS In order to identify genes correlating with both the triple negative breast cancer subtype as well as response to neoadjuvant chemotherapy we employed publicly available gene expression profiles of patients, which had received neoadjuvant chemotherapy. Analysis of tissue microarrays as well as breast cancer cell lines revealed correlation to the triple negative breast cancer subtype. Subsequently, effects of siRNA-mediated knockdown on response to standard chemotherapeutic agents as well as radiation therapy were analyzed. Additionally, we evaluated the molecular mechanisms by which SFRP1 alters the carcinogenic properties of breast cancer cells. RESULTS SFRP1 was identified as being significantly overexpressed in TNBC compared to other breast cancer subtypes. Additionally, SFRP1 expression is significantly correlated with an increased probability of positive response to neoadjuvant chemotherapy. Knockdown of SFRP1 in triple negative breast cancer cells renders the cells more resistant to standard chemotherapy. Moreover, tumorigenic properties of the cells are modified by knockdown, as shown by both migration or invasion capacity as well reduced apoptotic events. Surprisingly, we found that these effects do not rely on Wnt signaling. Furthermore, we show that pro-apoptotic as well as migratory pathways are differentially regulated after SFRP1 knockdown. CONCLUSION We could firstly show that SFRP1 strongly correlates with the triple negative breast cancer subtype and secondly, that SFRP1 might be used as a marker stratifying patients to positively respond to neoadjuvant chemotherapy. The mechanisms by which tumor suppressor SFRP1 influences carcinogenic properties of cancer cells do not rely on Wnt signaling, thereby demonstrating the complexity of tumor associated signaling pathways.
Collapse
Affiliation(s)
- Christof Bernemann
- Translational Tumor Biology Group, Albert-Schweitzer Campus 1 A1, 48149, Münster, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Zarzynska JM. Two faces of TGF-beta1 in breast cancer. Mediators Inflamm 2014; 2014:141747. [PMID: 24891760 PMCID: PMC4033515 DOI: 10.1155/2014/141747] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 04/16/2014] [Accepted: 04/24/2014] [Indexed: 12/14/2022] Open
Abstract
Breast cancer (BC) is potentially life-threatening malignancy that still causes high mortality among women. Scientific research in this field is focused on deeper understanding of pathogenesis and progressing of BC, in order to develop relevant diagnosis and improve therapeutic treatment. Multifunctional cytokine TGF- β 1 is one of many factors that have a direct influence on BC pathophysiology. Expression of TGF- β 1, induction of canonical and noncanonical signaling pathways, and mutations in genes encoding TGF- β 1 and its receptors are correlated with oncogenic activity of this cytokine. In early stages of BC this cytokine inhibits epithelial cell cycle progression and promotes apoptosis, showing tumor suppressive effects. However, in late stages, TGF- β 1 is linked with increased tumor progression, higher cell motility, cancer invasiveness, and metastasis. It is also involved in cancer microenvironment modification and promotion of epithelial to mesenchymal transition (EMT). This review summarizes the current knowledge on the phenomenon called "TGF- β 1 paradox", showing that better understanding of TGF- β 1 functions can be a step towards development of new therapeutic approaches. According to current knowledge several drugs against TGF- β 1 have been developed and are either in nonclinical or in early stages of clinical investigation.
Collapse
Affiliation(s)
- Joanna Magdalena Zarzynska
- Department of Food Hygiene and Public Health, Faculty of Veterinary Medicine, WULS-SGGW, Nowoursynowska 159, 02-776 Warsaw, Poland
| |
Collapse
|
30
|
Principe DR, Doll JA, Bauer J, Jung B, Munshi HG, Bartholin L, Pasche B, Lee C, Grippo PJ. TGF-β: duality of function between tumor prevention and carcinogenesis. J Natl Cancer Inst 2014; 106:djt369. [PMID: 24511106 DOI: 10.1093/jnci/djt369] [Citation(s) in RCA: 396] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Several mechanisms underlying tumor progression have remained elusive, particularly in relation to transforming growth factor beta (TGF-β). Although TGF-β initially inhibits epithelial growth, it appears to promote the progression of advanced tumors. Defects in normal TGF-β pathways partially explain this paradox, which can lead to a cascade of downstream events that drive multiple oncogenic pathways, manifesting as several key features of tumorigenesis (uncontrolled proliferation, loss of apoptosis, epithelial-to-mesenchymal transition, sustained angiogenesis, evasion of immune surveillance, and metastasis). Understanding the mechanisms of TGF-β dysregulation will likely reveal novel points of convergence between TGF-β and other pathways that can be specifically targeted for therapy.
Collapse
Affiliation(s)
- Daniel R Principe
- Affiliations of authors: Department of Medicine, Division of Gastroenterology (DRP, JB, BJ) and Division of Hematology/Oncology (HGM), Department of Surgery, Division of GI Surgical Oncology (DRP, PJG), and Department of Urology (CL), Northwestern University Feinberg School of Medicine, Chicago, IL; Department of Biomedical Engineering. McCormick School of Engineering, Northwestern University, Evanston, IL (DRP); Department of Biomedical Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI (JAD); UMR INSERM U1052, CNRS 5286, Université Lyon 1, Centre de Recherche en Cancérologie de Lyon, Lyon, France (LB); Division of Hematology/Oncology, Department of Medicine, University of Alabama-Birmingham, Birmingham, AL (BP); Department of Pathology and Laboratory Medicine, University of California-Irvine, Irvine, CA (CL)
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Brix DM, Clemmensen KKB, Kallunki T. When Good Turns Bad: Regulation of Invasion and Metastasis by ErbB2 Receptor Tyrosine Kinase. Cells 2014; 3:53-78. [PMID: 24709902 PMCID: PMC3980748 DOI: 10.3390/cells3010053] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 01/14/2014] [Accepted: 01/20/2014] [Indexed: 12/18/2022] Open
Abstract
Overexpression and activation of ErbB2 receptor tyrosine kinase in breast cancer is strongly linked to an aggressive disease with high potential for invasion and metastasis. In addition to inducing very aggressive, metastatic cancer, ErbB2 activation mediates processes such as increased cancer cell proliferation and survival and is needed for normal physiological activities, such as heart function and development of the nervous system. How does ErbB2 activation make cancer cells invasive and when? Comprehensive understanding of the cellular mechanisms leading to ErbB2-induced malignant processes is necessary for answering these questions. Here we present current knowledge about the invasion-promoting function of ErbB2 and the mechanisms involved in it. Obtaining detailed information about the "bad" behavior of ErbB2 can facilitate development of novel treatments against ErbB2-positive cancers.
Collapse
Affiliation(s)
- Ditte Marie Brix
- Unit of Cell Death and Metabolism, Danish Cancer Society Research Center, Danish Cancer Society, Strandboulevarden 49, DK-2100 Copenhagen, Denmark.
| | - Knut Kristoffer Bundgaard Clemmensen
- Unit of Cell Death and Metabolism, Danish Cancer Society Research Center, Danish Cancer Society, Strandboulevarden 49, DK-2100 Copenhagen, Denmark.
| | - Tuula Kallunki
- Unit of Cell Death and Metabolism, Danish Cancer Society Research Center, Danish Cancer Society, Strandboulevarden 49, DK-2100 Copenhagen, Denmark.
| |
Collapse
|
32
|
Abstract
The influence of the microenvironment on tumour progression is becoming clearer. In this Review we address the role of an essential signalling pathway, that of transforming growth factor-β, in the regulation of components of the tumour microenvironment and how this contributes to tumour progression.
Collapse
Affiliation(s)
- Michael Pickup
- Vanderbilt University Medical Center, Vanderbilt-Ingram Comprehensive Cancer Center, Medicine and Pathology, Cancer Biology, 2220 Pierce Avenue, 691 Preston Research Building, Nashville, Tennessee 37232, USA
| | | | | |
Collapse
|
33
|
Wang YQ, Li YM, Li X, Liu T, Liu XK, Zhang JQ, Guo JW, Guo LY, Qiao L. Hypermethylation of TGF-β1 gene promoter in gastric cancer. World J Gastroenterol 2013; 19:5557-5564. [PMID: 24023501 PMCID: PMC3761111 DOI: 10.3748/wjg.v19.i33.5557] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 06/11/2013] [Accepted: 07/19/2013] [Indexed: 02/06/2023] Open
Abstract
AIM: To examine transforming growth factor-β1 (TGF-β1) promoter methylation in gastric cancer and to determine if Helicobacter pylori (H. pylori) or interleukin (IL)-1β could induce TGF-β1 hypermethylation in vitro.
METHODS: We examined the frequency and extent of TGF-β1 promoter methylation using methylation-specific PCR in the gastric tissues from 47 gastric cancer patients and 39 non-gastric cancer subjects. H. pylori infection was confirmed by a positive result from either a serological test, histological analysis or C13 urea breath test. GES-1 and MKN-45 cells co-cultured with H. pylori or treated with IL-1β for 12, 24 and 48 h in vitro tested the effects of H. pylori or IL-1β on TGF-β1.
RESULTS: Twenty-four/forty-seven (51%) cases of gastric cancer (GC) tissues showed TGF-β1 promoter methylation, 15/47 (31.9%) cases of matched non-cancerous gastric mucosa tissues from the GC patients, and 11/39 (28%) case of the normal gastric mucosa tissues from non-GC subjects showed TGF-β1 promoter methylation (51% vs 28%, P < 0.05). Significantly higher levels of methylation of TGF-β1 were found in the tumor tissues than in non-tumor tissues from GC patients (0.24 ± 0.06 vs 0.17 ± 0.04, P < 0.05) and normal gastric tissues from non-GC subjects (0.24 ± 0.06 vs 0.15 ± 0.03, P < 0.05). TGF-β1 methylation was found in 48.3% of H. pylori-positive gastric mucosal tissues whereas only 23.1% of H. pylori-negative gastric mucosal tissues showed TGF-β1 methylation (48.3% vs 23.1%, P < 0.05). IL-1β appeared to induce a dose-dependent methylation of TGF-β1 and the strongest methylation was observed in GES-1 cells treated with 2.5 ng/mL of IL-1β for 48 h. Further studies showed that pre-treatment of GES-1 cells with 20 ng/mL IL-1RA for 1 h could partially abolish the effect of IL-1β on TGF-β1 methylation. Infection of GES-1 cells by H. pylori was not found to induce significant TGF-β1 promoter methylation.
CONCLUSION: Our data revealed that TGF-β1 promoter is methylated in GC patients. IL-1β may be an important mediator for H. pylori induced gene methylation during GC development.
Collapse
|
34
|
López-Díaz FJ, Gascard P, Balakrishnan SK, Zhao J, Del Rincon SV, Spruck C, Tlsty TD, Emerson BM. Coordinate transcriptional and translational repression of p53 by TGF-β1 impairs the stress response. Mol Cell 2013; 50:552-64. [PMID: 23706820 DOI: 10.1016/j.molcel.2013.04.029] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 03/25/2013] [Accepted: 04/24/2013] [Indexed: 10/26/2022]
Abstract
Cellular stress results in profound changes in RNA and protein synthesis. How cells integrate this intrinsic, p53-centered program with extracellular signals is largely unknown. We demonstrate that TGF-β1 signaling interferes with the stress response through coordinate transcriptional and translational repression of p53 levels, which reduces p53-activated transcription, and apoptosis in precancerous cells. Mechanistically, E2F-4 binds constitutively to the TP53 gene and induces transcription. TGF-β1-activated Smads are recruited to a composite Smad/E2F-4 element by an E2F-4/p107 complex that switches to a Smad corepressor, which represses TP53 transcription. TGF-β1 also causes dissociation of ribosomal protein RPL26 and elongation factor eEF1A from p53 mRNA, thereby reducing p53 mRNA association with polyribosomes and p53 translation. TGF-β1 signaling is dominant over stress-induced transcription and translation of p53 and prevents stress-imposed downregulation of Smad proteins. Thus, crosstalk between the TGF-β and p53 pathways defines a major node of regulation in the cellular stress response, enhancing drug resistance.
Collapse
Affiliation(s)
- Fernando J López-Díaz
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Ren J, Wang R, Huang G, Song H, Chen Y, Chen L. sFRP1 inhibits epithelial-mesenchymal transition in A549 human lung adenocarcinoma cell line. Cancer Biother Radiopharm 2013; 28:565-71. [PMID: 23802127 DOI: 10.1089/cbr.2012.1453] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) plays an important role in tumor metastasis of human nonsmall cell lung cancer (NSCLC). The Wnt pathway is identified as a key regulator of normal tissue development, and its aberrant activation contributes to the process of EMT. The secreted frizzled-related protein 1 (sFRP1), a Wnt-signaling antagonist, is downregulated in many tumors, including lung cancer. However, the role of sFRP1 in EMT and tumor metastasis remains unclear. In this study, we found that sFRP1 was dramatically downregulated in transforming growth factor β1 (TGF-β1)-induced EMT in the A549 human lung cancer cell line. Restoration of sFRP1 could inhibit the TGF-β1-induced EMT phenotype and tumor metastasis of the A549 cell line both in vitro and in vivo through inhibition of the Wnt pathway. Furthermore, FH535, a reversible Wnt-signaling inhibitor, exerted a similar effect on the TGF-β1-induced EMT phenotype. These results indicate that sFRP1, an endogenous antagonist of the Wnt pathway, inhibits TGF-β1-induced EMT, and might be a potential biomarker for the treatment of NSCLC.
Collapse
Affiliation(s)
- Jin Ren
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, China
| | | | | | | | | | | |
Collapse
|
36
|
Mu X, Lin S, Yang J, Chen C, Chen Y, Herzig MC, Washburn K, Halff GA, Walter CA, Sun B, Sun LZ. TGF-β signaling is often attenuated during hepatotumorigenesis, but is retained for the malignancy of hepatocellular carcinoma cells. PLoS One 2013; 8:e63436. [PMID: 23704908 PMCID: PMC3660330 DOI: 10.1371/journal.pone.0063436] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 03/29/2013] [Indexed: 01/07/2023] Open
Abstract
The role of transforming growth factor-beta (TGF-β) signaling in hepatocarcinogenesis remains controversial. We aimed to reveal TGF-β signaling status in human and murine tissues of hepatocellular carcinoma (HCC) and the mechanisms that mediate TGF-β’s role in regulating HCC malignancy. Here, TGF-β pathway component expression and activation in human and murine HCC tissues were measured with quantitative RT-PCR and Western blotting assays. The role of TGF-β receptor and Smad signaling in the growth and survival of several HCC cell lines was determined with several in vitro and in vivo approaches. We found that TGF-β receptor II (TβRII) expression was downregulated in two different HCC patient cohorts. Consistently, Smad3 phosphorylation was also downregulated in HCC tissues in comparison to that in adjacent normal tissues. Interestingly, many HCC cell lines were sensitive to TGF-β and growth-inhibited by exogenous TGF-β. However, stable knockdown of TβRII inhibited cell growth on plastic and in soft agar, and induced apoptosis resulting in suppressed subcutaneous tumor growth and metastatic potential in vivo. Furthermore, knockdown of Smad4 also led to a significant inhibition of growth on plastic and in soft agar with concomitant increase of apoptosis, PTEN expression, and reduced nuclear accumulation of linker region-phosphorylated Smad3. Taken together, TGF-β signaling pathway plays a dichotomous role in hepatocellular carcinogenesis. It appears to suppress HCC development, but is retained for HCC cell survival and malignancy. Furthermore, Smad4 can mediate both growth inhibitory activity induced by exogenous TGF-β and the survival activity induced by autocrine TGF-β revealing a delicate selection of the two opposing activities of TGF-β during HCC evolution.
Collapse
Affiliation(s)
- Xiaoxin Mu
- Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Shu Lin
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Junhua Yang
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Chen Chen
- Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yun Chen
- Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Maryanne C. Herzig
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Kenneth Washburn
- Transplant Center, University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Glenn A. Halff
- Transplant Center, University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Christi A. Walter
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, Texas, United States of America
- Cancer Therapy and Cancer Center, University of Texas Health Science Center, San Antonio, Texas, United States of America
- South Texas Veteran’s Health Care System, Audie Murphy Hospital, San Antonio, Texas, United States of America
| | - Beicheng Sun
- Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- * E-mail: ) (L-ZS; (LS) (BS)
| | - Lu-Zhe Sun
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, Texas, United States of America
- Cancer Therapy and Cancer Center, University of Texas Health Science Center, San Antonio, Texas, United States of America
- * E-mail: ) (L-ZS; (LS) (BS)
| |
Collapse
|
37
|
Suen KM, Lin CC, George R, Melo FA, Biggs ER, Ahmed Z, Drake MN, Arur S, Arold ST, Ladbury JE. Interaction with Shc prevents aberrant Erk activation in the absence of extracellular stimuli. Nat Struct Mol Biol 2013; 20:620-7. [PMID: 23584453 PMCID: PMC4059177 DOI: 10.1038/nsmb.2557] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Accepted: 03/07/2013] [Indexed: 01/08/2023]
Abstract
Control mechanisms that prevent aberrant signaling are necessary to maintain cellular homeostasis. We describe a new mechanism by which the adaptor protein Shc directly binds the MAP kinase Erk, thus preventing its activation in the absence of extracellular stimuli. The Shc-Erk complex restricts Erk nuclear translocation, restraining Erk-dependent transcription of genes, including those responsible for oncogenic growth. The complex forms through unique binding sites on both the Shc PTB domain and the N-terminal lobe of Erk. Upon receptor tyrosine kinase stimulation, a conformational change within Shc-induced through interaction with the phosphorylated receptor-releases Erk, allowing it to fulfill its role in signaling. Thus, in addition to its established role in promoting MAP kinase signaling in stimulated cells, Shc negatively regulates Erk activation in the absence of growth factors and thus could be considered a tumor suppressor in human cells.
Collapse
Affiliation(s)
- Kin Man Suen
- Department of Biochemistry and Molecular Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Chiechi A, Waning DL, Stayrook KR, Buijs JT, Guise TA, Mohammad KS. Role of TGF- β in breast cancer bone metastases. ACTA ACUST UNITED AC 2013; 4:15-30. [PMID: 24558636 PMCID: PMC3928102 DOI: 10.4236/abb.2013.410a4003] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Breast cancer is the most prevalent cancer among females worldwide leading to approximately 350,000 deaths each year. It has long been known that cancers preferentially metastasize to particular organs, and bone metastases occur in ~70% of patients with advanced breast cancer. Breast cancer bone metastases are predominantly osteolytic and accompanied by increased fracture risk, pain, nerve compression and hypercalcemia, causing severe morbidity. In the bone matrix, transforming growth factor-β (TGF-β) is one of the most abundant growth factors, which is released in active form upon tumor-induced osteoclastic bone resorption. TGF-β, in turn, stimulates bone metastatic tumor cells to secrete factors that further drive osteolytic bone destruction adjacent to the tumor. Thus, TGF-β is a crucial factor responsible for driving the feed-forward vicious cycle of cancer growth in bone. Moreover, TGF-β activates epithelial-to-mesenchymal transition, increases tumor cell invasiveness and angiogenesis and induces immunosuppression. Blocking the TGF-β signaling pathway to interrupt this vicious cycle between breast cancer and bone offers a promising target for therapeutic intervention to decrease skeletal metastasis. This review will describe the role of TGF-β in breast cancer and bone metastasis, and pre-clinical and clinical data will be evaluated for the potential use of TGF-β inhibitors in clinical practice to treat breast cancer bone metastases.
Collapse
Affiliation(s)
- Antonella Chiechi
- Division of Endocrinology, Department of Internal Medicine, Indiana University, Indianapolis, USA
| | - David L Waning
- Division of Endocrinology, Department of Internal Medicine, Indiana University, Indianapolis, USA
| | - Keith R Stayrook
- Division of Endocrinology, Department of Internal Medicine, Indiana University, Indianapolis, USA
| | - Jeroen T Buijs
- Division of Endocrinology, Department of Internal Medicine, Indiana University, Indianapolis, USA ; Department of Urology, Medical Center, Leiden University, Leiden, The Netherlands
| | - Theresa A Guise
- Division of Endocrinology, Department of Internal Medicine, Indiana University, Indianapolis, USA
| | - Khalid S Mohammad
- Division of Endocrinology, Department of Internal Medicine, Indiana University, Indianapolis, USA
| |
Collapse
|
39
|
Park J, Scherer PE. Adipocyte-derived endotrophin promotes malignant tumor progression. J Clin Invest 2012; 122:4243-56. [PMID: 23041627 DOI: 10.1172/jci63930] [Citation(s) in RCA: 249] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Accepted: 08/02/2012] [Indexed: 12/21/2022] Open
Abstract
Adipocytes represent a major cell type in the mammary tumor microenvironment and are important for tumor growth. Collagen VI (COL6) is highly expressed in adipose tissue, upregulated in the obese state, and enriched in breast cancer lesions and is a stimulator of mammary tumor growth. Here, we have described a cleavage product of the COL6α3 chain, endotrophin (ETP), which serves as the major mediator of the COL6-mediated tumor effects. ETP augmented fibrosis, angiogenesis, and inflammation through recruitment of macrophages and endothelial cells. Moreover, ETP expression was associated with aggressive mammary tumor growth and high metastatic growth. These effects were partially mediated through enhanced TGF-β signaling, which contributes to tissue fibrosis and epithelial-mesenchymal transition (EMT) of tumor cells. Our results highlight the crucial role of ETP as an obesity-associated factor that promotes tumor growth in the context of adipocyte interactions with tumor and stromal cells.
Collapse
Affiliation(s)
- Jiyoung Park
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390-8549, USA
| | | |
Collapse
|
40
|
Boudreau HE, Casterline BW, Rada B, Korzeniowska A, Leto TL. Nox4 involvement in TGF-beta and SMAD3-driven induction of the epithelial-to-mesenchymal transition and migration of breast epithelial cells. Free Radic Biol Med 2012; 53:1489-99. [PMID: 22728268 PMCID: PMC3448829 DOI: 10.1016/j.freeradbiomed.2012.06.016] [Citation(s) in RCA: 163] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 06/11/2012] [Accepted: 06/12/2012] [Indexed: 12/11/2022]
Abstract
The epithelial-to-mesenchymal transition (EMT) is the development of increased cell plasticity that occurs normally during wound healing and embryonic development and can be coopted for cancer invasion and metastasis. TGF-beta induces EMT but the mechanism is unclear. Our studies suggest that Nox4, a member of the NADPH oxidase (Nox) family, is a source of reactive oxygen species (ROS) affecting cell migration and fibronectin expression, an EMT marker, in normal and metastatic breast epithelial cells. We found that TGF-beta induces Nox4 expression (mRNA and protein) and ROS generation in normal (MCF10A) and metastatic (MDA-MB-231) human breast epithelial cells. Conversely, cells expressing a dominant-negative form of Nox4 or Nox4-targeted shRNA showed significantly lower ROS production on TGF-beta treatment. Expression of a constitutively active TGF-beta receptor type I significantly increased Nox4 promoter activity, mRNA and protein expression, and ROS generation. Nox4 transcriptional regulation by TGF-beta was SMAD3 dependent based on the effect of constitutively active SMAD3 increasing Nox4 promoter activity, whereas dominant-negative SMAD3 or SIS3, a SMAD3-specific inhibitor, had the opposite effect. Furthermore, Nox4 knockdown, dominant-negative Nox4 or SMAD3, or SIS3 blunted TGF-beta induced wound healing and cell migration, whereas cell proliferation was not affected. Our experiments further indicate that Nox4 plays a role in TGF-beta regulation of fibronectin mRNA expression, based on the effects of dominant-negative Nox4 in reducing fibronectin mRNA in TGF-beta-treated MDA-MB-231and MCF10A cells. Collectively, these data indicate that Nox4 contributes to NADPH oxidase-dependent ROS production that may be critical for the progression of the EMT in breast epithelial cells, and thereby has therapeutic implications.
Collapse
Affiliation(s)
- Howard E. Boudreau
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Benjamin W. Casterline
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Balazs Rada
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Agnieszka Korzeniowska
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Thomas L. Leto
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| |
Collapse
|
41
|
Matise LA, Palmer TD, Ashby WJ, Nashabi A, Chytil A, Aakre M, Pickup MW, Gorska AE, Zijlstra A, Moses HL. Lack of transforming growth factor-β signaling promotes collective cancer cell invasion through tumor-stromal crosstalk. Breast Cancer Res 2012; 14:R98. [PMID: 22748014 PMCID: PMC3680921 DOI: 10.1186/bcr3217] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Accepted: 07/02/2012] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Transforming growth factor beta (TGF-β) has a dual role during tumor progression, initially as a suppressor and then as a promoter. Epithelial TGF-β signaling regulates fibroblast recruitment and activation. Concurrently, TGF-β signaling in stromal fibroblasts suppresses tumorigenesis in adjacent epithelia, while its ablation potentiates tumor formation. Much is known about the contribution of TGF-β signaling to tumorigenesis, yet the role of TGF-β in epithelial-stromal migration during tumor progression is poorly understood. We hypothesize that TGF-β is a critical regulator of tumor-stromal interactions that promote mammary tumor cell migration and invasion. METHODS Fluorescently labeled murine mammary carcinoma cells, isolated from either MMTV-PyVmT transforming growth factor-beta receptor II knockout (TβRII KO) or TβRIIfl/fl control mice, were combined with mammary fibroblasts and xenografted onto the chicken embryo chorioallantoic membrane. These combinatorial xenografts were used as a model to study epithelial-stromal crosstalk. Intravital imaging of migration was monitored ex ovo, and metastasis was investigated in ovo. Epithelial RNA from in ovo tumors was isolated by laser capture microdissection and analyzed to identify gene expression changes in response to TGF-β signaling loss. RESULTS Intravital microscopy of xenografts revealed that mammary fibroblasts promoted two migratory phenotypes dependent on epithelial TGF-β signaling: single cell/strand migration or collective migration. At epithelial-stromal boundaries, single cell/strand migration of TβRIIfl/fl carcinoma cells was characterized by expression of α-smooth muscle actin and vimentin, while collective migration of TβRII KO carcinoma cells was identified by E-cadherin+/p120+/β-catenin+ clusters. TβRII KO tumors also exhibited a twofold greater metastasis than TβRIIfl/fl tumors, attributed to enhanced extravasation ability. In TβRII KO tumor epithelium compared with TβRIIfl/fl epithelium, Igfbp4 and Tspan13 expression was upregulated while Col1α2, Bmp7, Gng11, Vcan, Tmeff1, and Dsc2 expression was downregulated. Immunoblotting and quantitative PCR analyses on cultured cells validated these targets and correlated Tmeff1 expression with disease progression of TGF-β-insensitive mammary cancer. CONCLUSION Fibroblast-stimulated carcinoma cells utilize TGF-β signaling to drive single cell/strand migration but migrate collectively in the absence of TGF-β signaling. These migration patterns involve the signaling regulation of several epithelial-to-mesenchymal transition pathways. Our findings concerning TGF-β signaling in epithelial-stromal interactions are important in identifying migratory mechanisms that can be targeted as recourse for breast cancer treatment.
Collapse
|
42
|
Luo Y, Tsuchiya KD, Il Park D, Fausel R, Kanngurn S, Welcsh P, Dzieciatkowski S, Wang J, Grady WM. RET is a potential tumor suppressor gene in colorectal cancer. Oncogene 2012; 32:2037-47. [PMID: 22751117 PMCID: PMC3465636 DOI: 10.1038/onc.2012.225] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Cancer arises as the consequence of mutations and epigenetic alterations that activate oncogenes and inactivate tumor suppressor genes. Through a genome-wide screen for methylated genes in colon neoplasms, we identified aberrantly methylated RET in colorectal cancer. RET, a transmembrane receptor tyrosine kinase and a receptor for the glial cell-derived neurotrophic factor family ligands, was one of the first oncogenes to be identified, and has been shown to be an oncogene in thyroid cancer and pheochromocytoma. However, unexpectedly, we found RET is methylated in 27% of colon adenomas and in 63% of colorectal cancers, and now provide evidence that RET has tumor suppressor activity in colon cancer. The aberrant methylation of RET correlates with decreased RET expression, whereas the restoration of RET in colorectal cancer cell lines results in apoptosis. Furthermore, in support of a tumor suppressor function of RET, mutant RET has also been found in primary colorectal cancer. We now show that these mutations inactivate RET, which is consistent with RET being a tumor suppressor gene in the colon. These findings suggest that the aberrant methylation of RET and the mutational inactivation of RET promote colorectal cancer formation, and that RET can serve as a tumor suppressor gene in the colon. Moreover, the increased frequency of methylated RET in colon cancers compared with adenomas suggests RET inactivation is involved in the progression of colon adenomas to cancer.
Collapse
Affiliation(s)
- Y Luo
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
USP4 is regulated by AKT phosphorylation and directly deubiquitylates TGF-β type I receptor. Nat Cell Biol 2012; 14:717-26. [PMID: 22706160 DOI: 10.1038/ncb2522] [Citation(s) in RCA: 248] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Accepted: 05/11/2012] [Indexed: 12/13/2022]
Abstract
The stability and membrane localization of the transforming growth factor-β (TGF-β) type I receptor (TβRI) determines the levels of TGF-β signalling. TβRI is targeted for ubiquitylation-mediated degradation by the SMAD7-SMURF2 complex. Here we performed a genome-wide gain-of-function screen and identified ubiquitin-specific protease (USP) 4 as a strong inducer of TGF-β signalling. USP4 was found to directly interact with TβRI and act as a deubiquitylating enzyme, thereby controlling TβRI levels at the plasma membrane. Depletion of USP4 mitigates TGF-β-induced epithelial to mesenchymal transition and metastasis. Importantly, AKT (also known as protein kinase B), which has been associated with poor prognosis in breast cancer, directly associates with and phosphorylates USP4. AKT-mediated phosphorylation relocates nuclear USP4 to the cytoplasm and membrane and is required for maintaining its protein stability. Moreover, AKT-induced breast cancer cell migration was inhibited by USP4 depletion and TβRI kinase inhibition. Our results uncover USP4 as an important determinant for crosstalk between TGF-β and AKT signalling pathways.
Collapse
|
44
|
Nuclear translocation of type I transforming growth factor β receptor confers a novel function in RNA processing. Mol Cell Biol 2012; 32:2183-95. [PMID: 22473997 DOI: 10.1128/mcb.00320-12] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Signaling of transforming growth factor β (TGF-β) is redirected in cancer to promote malignancy, but how TGF-β function is altered in a transformed cell is not fully understood. We investigated TGF-β signaling by profiling proteins that differentially bound to type I TGF-β receptor (TβRI) in nontransformed, HER2-transformed, and HER2-negative breast cancer cells using immunoprecipitation followed by protein identification. Interestingly, several nuclear proteins implicated in posttranscriptional RNA processing were uniquely identified in the TβRI coprecipitates from HER2-transformed cells. Ligand-inducible nuclear translocation of TβRI was observed only in transformed cells, and the translocation required importin β1, nucleolin, and Smad2/3. This trafficking was dependent on the high Ran GTPase activity resulting from oncogenic transformation. In the nucleus, TβRI associated with purine-rich RNA sequences in a synergistic manner with the RNA-binding factor hnRNP A1. We further found that nuclear translocation of TβRI specifically induced epidermal growth factor receptor (EGFR) transcript isoform c, which encodes a soluble EGFR protein, through alternative splicing or 3'-end processing. Our study confirms a cancer-specific nuclear translocation of TβRI and demonstrates its potential function in regulating nuclear RNA processing, as well as a novel gain-of-function mechanism of TGF-β signaling in cancer.
Collapse
|
45
|
Zhu Q, Luo K. SnoN in regulation of embryonic development and tissue morphogenesis. FEBS Lett 2012; 586:1971-6. [PMID: 22710172 DOI: 10.1016/j.febslet.2012.03.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 03/03/2012] [Accepted: 03/05/2012] [Indexed: 01/15/2023]
Abstract
SnoN (Ski-novel protein) plays an important role in embryonic development, tumorigenesis and aging. Past studies largely focused on its roles in tumorigenesis. Recent studies of its expression patterns and functions in mouse models and mammalian cells have revealed that SnoN interacts with multiple signaling molecules at different cellular levels to modulate the activities of several signaling pathways in a tissue context and developmental stage dependent manner. These studies suggest that SnoN may have broad functions in the embryonic development and tissue morphogenesis.
Collapse
Affiliation(s)
- Qingwei Zhu
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | | |
Collapse
|
46
|
A novel gain-of-function mutation of TGF-β receptor II promotes cancer progression via delayed receptor internalization in oral squamous cell carcinoma. Cancer Lett 2012; 315:161-9. [DOI: 10.1016/j.canlet.2011.09.036] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Revised: 09/27/2011] [Accepted: 09/27/2011] [Indexed: 12/11/2022]
|
47
|
Thiolloy S, Edwards JR, Fingleton B, Rifkin DB, Matrisian LM, Lynch CC. An osteoblast-derived proteinase controls tumor cell survival via TGF-beta activation in the bone microenvironment. PLoS One 2012; 7:e29862. [PMID: 22238668 PMCID: PMC3251607 DOI: 10.1371/journal.pone.0029862] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Accepted: 12/05/2011] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Breast to bone metastases frequently induce a "vicious cycle" in which osteoclast mediated bone resorption and proteolysis results in the release of bone matrix sequestered factors that drive tumor growth. While osteoclasts express numerous proteinases, analysis of human breast to bone metastases unexpectedly revealed that bone forming osteoblasts were consistently positive for the proteinase, MMP-2. Given the role of MMP-2 in extracellular matrix degradation and growth factor/cytokine processing, we tested whether osteoblast derived MMP-2 contributed to the vicious cycle of tumor progression in the bone microenvironment. METHODOLOGY/PRINCIPAL FINDINGS To test our hypothesis, we utilized murine models of the osteolytic tumor-bone microenvironment in immunocompetent wild type and MMP-2 null mice. In longitudinal studies, we found that host MMP-2 significantly contributed to tumor progression in bone by protecting against apoptosis and promoting cancer cell survival (caspase-3; immunohistochemistry). Our data also indicate that host MMP-2 contributes to tumor induced osteolysis (μCT, histomorphometry). Further ex vivo/in vitro experiments with wild type and MMP-2 null osteoclast and osteoblast cultures identified that 1) the absence of MMP-2 did not have a deleterious effect on osteoclast function (cd11B isolation, osteoclast differentiation, transwell migration and dentin resorption assay); and 2) that osteoblast derived MMP-2 promoted tumor survival by regulating the bioavailability of TGFβ, a factor critical for cell-cell communication in the bone (ELISA, immunoblot assay, clonal and soft agar assays). CONCLUSION/SIGNIFICANCE Collectively, these studies identify a novel "mini-vicious cycle" between the osteoblast and metastatic cancer cells that is key for initial tumor survival in the bone microenvironment. In conclusion, the findings of our study suggest that the targeted inhibition of MMP-2 and/or TGFβ would be beneficial for the treatment of bone metastases.
Collapse
Affiliation(s)
- Sophie Thiolloy
- Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - James R. Edwards
- Nuffield Orthopaedic Centre, University of Oxford, Oxford, United Kingdom
| | - Barbara Fingleton
- Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Daniel B. Rifkin
- Department of Cell Biology, New York University School of Medicine, New York, New York, United States of America
| | - Lynn M. Matrisian
- Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Conor C. Lynch
- Tumor Biology Department, H. Lee Moffitt Cancer Center, Tampa, Florida, United States of America
| |
Collapse
|
48
|
Buijs JT, Stayrook KR, Guise TA. TGF-β in the Bone Microenvironment: Role in Breast Cancer Metastases. CANCER MICROENVIRONMENT : OFFICIAL JOURNAL OF THE INTERNATIONAL CANCER MICROENVIRONMENT SOCIETY 2011; 4:261-81. [PMID: 21748439 PMCID: PMC3234330 DOI: 10.1007/s12307-011-0075-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Accepted: 06/28/2011] [Indexed: 01/29/2023]
Abstract
Breast cancer is the most prevalent cancer among females worldwide. It has long been known that cancers preferentially metastasize to particular organs, and bone metastases occur in ∼70% of patients with advanced breast cancer. Breast cancer bone metastases are predominantly osteolytic and accompanied by bone destruction, bone fractures, pain, and hypercalcemia, causing severe morbidity and hospitalization. In the bone matrix, transforming growth factor-β (TGF-β) is one of the most abundant growth factors, which is released in active form upon tumor-induced osteoclastic bone resorption. TGF-β, in turn, stimulates bone metastatic cells to secrete factors that further drive osteolytic destruction of the bone adjacent to the tumor, categorizing TGF-β as a crucial factor responsible for driving the feed-forward vicious cycle of cancer growth in bone. Moreover, TGF-β activates epithelial-to-mesenchymal transition, increases tumor cell invasiveness and angiogenesis and induces immunosuppression. Blocking the TGF-β signaling pathway to interrupt this vicious cycle between breast cancer and bone offers a promising target for therapeutic intervention to decrease skeletal metastasis. This review will describe the role of TGF-β in breast cancer and bone metastasis, and pre-clinical and clinical data will be evaluated for the potential use of TGF-β inhibitors in clinical practice to treat breast cancer bone metastases.
Collapse
Affiliation(s)
- Jeroen T. Buijs
- Department of Medicine, Division of Endocrinology, Indiana University School of Medicine, 980 West Walnut Street, Walther Hall R3, #C132, Indianapolis, IN USA
| | - Keith R. Stayrook
- Department of Medicine, Division of Endocrinology, Indiana University School of Medicine, 980 West Walnut Street, Walther Hall R3, #C132, Indianapolis, IN USA
| | - Theresa A. Guise
- Department of Medicine, Division of Endocrinology, Indiana University School of Medicine, 980 West Walnut Street, Walther Hall R3, #C132, Indianapolis, IN USA
| |
Collapse
|
49
|
Park CY, Kim DK, Sheen YY. EW-7203, a novel small molecule inhibitor of transforming growth factor-β (TGF-β) type I receptor/activin receptor-like kinase-5, blocks TGF-β1-mediated epithelial-to-mesenchymal transition in mammary epithelial cells. Cancer Sci 2011; 102:1889-96. [PMID: 21707864 PMCID: PMC11158462 DOI: 10.1111/j.1349-7006.2011.02014.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Recently, small molecule inhibitors of transforming growth factorβ (TGF-β) type I receptor kinase ⁄ activin receptor-like kinase-5 (ALK5) have been developed to target TGF-β signalling as a therapeutic strategy for combating cancer. In the present study, the authors examined a novel small molecule inhibitor of ALK5, 3-((5- ([1,2,4]triazolo[1,5-a]pyridin-6-yl)-4-(6-methylpyridin-2-yl)thiazol-2-ylamino)methyl)benzonitrile (EW-7203) in breast cancer cells to determine if it has potential for cancer treatment. The inhibitory effects of EW-7203 on TGF-β-induced Smad signalling and epithelial- to-mesenchymal transition (EMT) were investigated in mammary epithelial cells using luciferase reporter assays, immunoblotting, confocal microscopy and wound healing assays. In addition, the suppressive effects of EW-7203 on mammary cancer metastasis to the lung were examined using a Balb ⁄ c xenograft model system. The novel ALK5 inhibitor, EW-7203, inhibited the TGF-β1-stimulated transcriptional activation of p3TP-Lux and pCA-GA₁₂- Luc. In addition, EW-7203 decreased phosphorylated Smad2 levels and the nuclear translocation of Smad2 was increased by TGF-β1. In addition, EW-7203 inhibited TGF-β1-induced EMT and wound healing of NMuMG cells. Furthermore, in xenografted Balb ⁄ c mice, EW-7203 inhibited metastasis to the lung from breast tumors. The novel ALK5 inhibitor, EW-7203, efficiently inhibited TGF-β1-induced Smad signalling, EMT and breast tumor metastasis to the lung in vivo, demonstrating that EW-7203 has therapeutic potential for breast cancer metastasis to the lung.
Collapse
Affiliation(s)
- Chul-Yong Park
- College of Pharmacy, Ewha Womans University, Seodaemun-gu, Seoul, Korea
| | | | | |
Collapse
|
50
|
Borczuk AC, Sole M, Lu P, Chen J, Wilgus ML, Friedman RA, Albelda SM, Powell CA. Progression of human bronchioloalveolar carcinoma to invasive adenocarcinoma is modeled in a transgenic mouse model of K-ras-induced lung cancer by loss of the TGF-β type II receptor. Cancer Res 2011; 71:6665-75. [PMID: 21911454 DOI: 10.1158/0008-5472.can-11-1590] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Clinical investigations have suggested that repression of the TGF-β type II receptor (TβRII) may be an important step in progression of lung adenocarcinoma from an indolent in situ state to a frank invasive carcinoma. To test this hypothesis, we compared the effects of deleting the murine homolog of this receptor (Tgfbr2) in a mouse model of mutant K-ras-induced lung carcinoma, which normally induces the formation of multifocal tumors of low invasive potential. In this model, loss of Tgfbr2 induced a highly invasive phenotype associated with lymph node metastasis and reduced survival. Tumor-associated stromal cells displayed an immunosuppressive profile marked by increased numbers of B and T cells. Moreover, tumor stromal cell profiling revealed a developmental TGF-β response profile that associated with a collagenized extracellular matrix and increased invasion of TGF-β nonresponsive tumor cells. Together, these results suggest that our KrasTgfbr2(-/-) mouse model of invasive lung carcinoma mirrors the genomic response and clinical progression of human lung adenocarcinoma, recapitulating changes in lung stromal pathways that occur in the tumor microenvironment during malignant progression in this disease.
Collapse
Affiliation(s)
- Alain C Borczuk
- Department of Pathology, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | | | | | | | | | | | | | | |
Collapse
|