101
|
Ying Z, Tian H, Li Y, Lian R, Li W, Wu S, Zhang HZ, Wu J, Liu L, Song J, Guan H, Cai J, Zhu X, Li J, Li M. CCT6A suppresses SMAD2 and promotes prometastatic TGF-β signaling. J Clin Invest 2017; 127:1725-1740. [PMID: 28375158 DOI: 10.1172/jci90439] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 02/02/2017] [Indexed: 12/21/2022] Open
Abstract
Paradoxically, during early tumor development in many cancer types, TGF-β acts as a tumor suppressor, whereas in the advanced stages of these cancers, increased TGF-β expression is linked to high metastasis and poor prognosis. These findings suggest that unidentified mechanisms may function to rewire TGF-β signaling toward its prometastatic role in cancer cells. Our current study using non-small-cell lung carcinoma (NSCLC) cell lines, animal models, and clinical specimens demonstrates that suppression of SMAD2, with SMAD3 function intact, switches TGF-β-induced transcriptional responses to a prometastatic state. Importantly, we identified chaperonin containing TCP1 subunit 6A (CCT6A) as an inhibitor and direct binding protein of SMAD2 and found that CCT6A suppresses SMAD2 function in NSCLC cells and promotes metastasis. Furthermore, selective inhibition of SMAD3 or CCT6A efficiently suppresses TGF-β-mediated metastasis. Our findings provide a mechanism that directs TGF-β signaling toward its prometastatic arm and may contribute to the development of therapeutic strategies targeting TGF-β for NSCLC.
Collapse
|
102
|
Varadaraj A, Jenkins LM, Singh P, Chanda A, Snider J, Lee NY, Amsalem-Zafran AR, Ehrlich M, Henis YI, Mythreye K. TGF-β triggers rapid fibrillogenesis via a novel TβRII-dependent fibronectin-trafficking mechanism. Mol Biol Cell 2017; 28:1195-1207. [PMID: 28298487 PMCID: PMC5415016 DOI: 10.1091/mbc.e16-08-0601] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 02/22/2017] [Accepted: 02/27/2017] [Indexed: 02/02/2023] Open
Abstract
There is increased recycling of soluble fibronectin from the cell surface for fibrillogenesis. This recycling is regulated by TGF-β in a transcription- and SMAD-independent manner via specific TβRII and integrin α5β1 interactions. The recycling of fibronectin is Rab11 dependent and is required for TGF-β–induced cell migration. Fibronectin (FN) is a critical regulator of extracellular matrix (ECM) remodeling through its availability and stepwise polymerization for fibrillogenesis. Availability of FN is regulated by its synthesis and turnover, and fibrillogenesis is a multistep, integrin-dependent process essential for cell migration, proliferation, and tissue function. Transforming growth factor β (TGF-β) is an established regulator of ECM remodeling via transcriptional control of ECM proteins. Here we show that TGF-β, through increased FN trafficking in a transcription- and SMAD-independent manner, is a direct and rapid inducer of the fibrillogenesis required for TGF-β–induced cell migration. Whereas TGF-β signaling is dispensable for rapid fibrillogenesis, stable interactions between the cytoplasmic domain of the type II TGF-β receptor (TβRII) and the FN receptor (α5β1 integrin) are required. We find that, in response to TGF-β, cell surface–internalized FN is not degraded by the lysosome but instead undergoes recycling and incorporation into fibrils, a process dependent on TβRII. These findings are the first to show direct use of trafficked and recycled FN for fibrillogenesis, with a striking role for TGF-β in this process. Given the significant physiological consequences associated with FN availability and polymerization, our findings provide new insights into the regulation of fibrillogenesis for cellular homeostasis.
Collapse
Affiliation(s)
- Archana Varadaraj
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208
| | - Laura M Jenkins
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208
| | - Priyanka Singh
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208
| | - Anindya Chanda
- Department of Environmental Health Sciences, University of South Carolina, Columbia, SC 29201
| | - John Snider
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208
| | - N Y Lee
- Division of Pharmacology, College of Pharmacy, Ohio State University, Columbus, OH 43210
| | | | - Marcelo Ehrlich
- Department of Cell Research and Immunology, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yoav I Henis
- Department of Neurobiology, Tel Aviv University, Tel Aviv 69978, Israel
| | - Karthikeyan Mythreye
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208 .,Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208
| |
Collapse
|
103
|
Wei G, Xu Y, Peng T, Yan J, Wang Z, Sun Z. Sanguinarine exhibits antitumor activity via up-regulation of Fas-associated factor 1 in non-small cell lung cancer. J Biochem Mol Toxicol 2017; 31. [PMID: 28296008 DOI: 10.1002/jbt.21914] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 02/06/2017] [Accepted: 02/09/2017] [Indexed: 01/04/2023]
Affiliation(s)
- Guangxia Wei
- Department of Cardiothoracic Surgery, Huangshi Central Hospital; Affiliated Hospital of Hubei Polytechnic University; Edong Healthcare Group Huangshi 435000 People's Republic of China
| | - Yahuan Xu
- Department of Cardiothoracic Surgery, Huangshi Central Hospital; Affiliated Hospital of Hubei Polytechnic University; Edong Healthcare Group Huangshi 435000 People's Republic of China
| | - Tao Peng
- Department of Cardiothoracic Surgery, Huangshi Central Hospital; Affiliated Hospital of Hubei Polytechnic University; Edong Healthcare Group Huangshi 435000 People's Republic of China
| | - Jie Yan
- Department of Cardiothoracic Surgery, Huangshi Central Hospital; Affiliated Hospital of Hubei Polytechnic University; Edong Healthcare Group Huangshi 435000 People's Republic of China
| | - Zhengjun Wang
- Department of Cardiothoracic Surgery, Huangshi Central Hospital; Affiliated Hospital of Hubei Polytechnic University; Edong Healthcare Group Huangshi 435000 People's Republic of China
| | - Zhanwen Sun
- Department of Cardiothoracic Surgery, Huangshi Central Hospital; Affiliated Hospital of Hubei Polytechnic University; Edong Healthcare Group Huangshi 435000 People's Republic of China
| |
Collapse
|
104
|
Yang W, Wang JG, Xu J, Zhou D, Ren K, Hou C, Chen L, Liu X. HCRP1 inhibits TGF-β induced epithelial-mesenchymal transition in hepatocellular carcinoma. Int J Oncol 2017; 50:1233-1240. [DOI: 10.3892/ijo.2017.3903] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 02/07/2017] [Indexed: 11/06/2022] Open
|
105
|
Downregulation of microRNA-145 promotes epithelial-mesenchymal transition via regulating Snail in osteosarcoma. Cancer Gene Ther 2017; 24:83-88. [PMID: 28186090 DOI: 10.1038/cgt.2017.1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 11/21/2016] [Accepted: 01/05/2017] [Indexed: 12/12/2022]
Abstract
Metastasis is the principal cause of cancer death and occurs through multiple, complex processes. Epithelial to mesenchymal transition (EMT) is an important process during embryonic development and has also been hypothesized to exhibit a significant role in cancer cell invasion and metastasis. MicroRNAs (miRNAs) are a class of widespread noncoding RNAs. In recent years, many studies have shown that miRNAs could influence the signaling pathways and downstream events that define EMT on a molecular level. However, the exact role and mechanisms of miR-145 in EMT of osteosarcoma (OS) was unknown. In the present study, miR-145 was downregulated in OS tissues and cell lines and it was shown that miR-145 expression was closely correlated with advanced tumor progression in patients of OS. In addition, miR-145 upregulation by miR-145 agomir significantly inhibited MG63 cells invasion and migration ability. MiR-145 was reported to be able to inhibit EMT in cancers. Following the examination of changes in cell epithelial and mesenchymal markers, it was found that upregulation of miR-145 strongly reversed EMT in MG63 cells. Meanwhile, the expression of Snail, a strong E-cadherin transcription repressor was also attenuated by miR-145 agomir. Furthermore, the decreased EMT and invasion and metastasis caused by miR-145 agomir could be restored by Snail siRNA. In conclusion, the results demonstrated that miR-145 could mediate EMT by targeting Snail and miR-145 might be a novel EMT regulating transcription factor that involved in the progression of OS. The specific drugs targeting miR-145-mediated EMT process might be new promising cancer therapies.
Collapse
|
106
|
Liu S, Liu L, Ye W, Ye D, Wang T, Guo W, Liao Y, Xu D, Song H, Zhang L, Zhu H, Deng J, Zhang Z. High Vimentin Expression Associated with Lymph Node Metastasis and Predicated a Poor Prognosis in Oral Squamous Cell Carcinoma. Sci Rep 2016; 6:38834. [PMID: 27966589 PMCID: PMC5155220 DOI: 10.1038/srep38834] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 11/14/2016] [Indexed: 12/20/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a common public health problem worldwide with poor prognosis, which is largely due to lymph node metastasis and recurrence. Identification of specific molecular markers of OSCC with lymph node metastasis would be very important for early and specific diagnosis. In this study, we screened for the potential prognosis markers via unbiased transcriptomic microarray analysis in paired two OSCC cell lines, a lymph node metastatic HN12 cell line and a low metastatic parental HN4 cell line. The results showed that vimentin, with 87-fold increase of expression, was on the top of all upregulated genes in metastatic HN12 cells compared to non-metastatic HN4 cells. Treatment of non-metastatic HN4 cells with TGF-β1 induced epithelial to mesenchymal transition (EMT), with increased vimentin expression as well as enhanced migration activity. Consistently, knockdown of vimentin via siRNA resulted in suppressed invasion and migration activities of HN12 cells, suggesting an essential role of vimentin in EMT-related functions of OSCC cells. Finally, immunohistochemical (IHC) staining analysis showed that high vimentin expression was strongly associated with high lymph node metastases (p < 0.05), and poor overall survival (p < 0.05) in OSCC patients. Thus, high vimentin expression is strongly associated with increased metastatic potential, and may serve as a prediction marker for poor prognosis in OSCC patients.
Collapse
Affiliation(s)
- Shuli Liu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Stomatology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liu Liu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Stomatology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weimin Ye
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Stomatology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dongxia Ye
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Stomatology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tong Wang
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenzheng Guo
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yueling Liao
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dongliang Xu
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongyong Song
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Zhang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Stomatology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hanguang Zhu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Stomatology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiong Deng
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Translation Medicine Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhiyuan Zhang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Stomatology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
107
|
Robinson NJ, Schiemann WP. Means to the ends: The role of telomeres and telomere processing machinery in metastasis. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1866:320-329. [PMID: 27768860 PMCID: PMC5138103 DOI: 10.1016/j.bbcan.2016.10.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 10/12/2016] [Accepted: 10/15/2016] [Indexed: 12/29/2022]
Abstract
Despite significant clinical advancements, cancer remains a leading cause of mortality throughout the world due largely to the process of metastasis and the dissemination of cancer cells from their primary tumor of origin to distant secondary sites. The clinical burden imposed by metastasis is further compounded by a paucity of information regarding the factors that mediate metastatic progression. Linear chromosomes are capped by structures known as telomeres, which dictate cellular lifespan in humans by shortening progressively during successive cell divisions. Although telomere shortening occurs in nearly all somatic cells, telomeres may be elongated via two seemingly disjoint pathways: (i) telomerase-mediated extension, and (ii) homologous recombination-based alternative lengthening of telomeres (ALT). Both telomerase and ALT are activated in various human cancers, with more recent evidence implicating both pathways as potential mediators of metastasis. Here we review the known roles of telomere homeostasis in metastasis and posit a mechanism whereby metastatic activity is determined by a dynamic fluctuation between ALT and telomerase, as opposed to the mere activation of a generic telomere elongation program. Additionally, the pleiotropic nature of the telomere processing machinery makes it an attractive therapeutic target for metastasis, and as such, we also explore the therapeutic implications of our proposed mechanism.
Collapse
Affiliation(s)
- Nathaniel J Robinson
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - William P Schiemann
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
108
|
Shanmugasundaram K, Block K. Renal Carcinogenesis, Tumor Heterogeneity, and Reactive Oxygen Species: Tactics Evolved. Antioxid Redox Signal 2016; 25:685-701. [PMID: 27287984 PMCID: PMC5069729 DOI: 10.1089/ars.2015.6569] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 06/07/2016] [Accepted: 06/10/2016] [Indexed: 12/13/2022]
Abstract
SIGNIFICANCE The number of kidney cancers is growing 3-5% each year due to unknown etiologies. Intra- and inter-tumor mediators increase oxidative stress and drive tumor heterogeneity. Recent Advances: Technology advancement in state-of-the-art instrumentation and methodologies allows researchers to detect and characterize global landscaping modifications in genes, proteins, and pathophysiology patterns at the single-cell level. CRITICAL ISSUES We postulate that the sources of reactive oxygen species (ROS) and their activation within subcellular compartments will change over a timeline of tumor evolvement and contribute to tumor heterogeneity. Therefore, the complexity of intracellular changes within a tumor and ROS-induced tumor heterogeneity coupled to the advancement of detecting these events globally are limited at the level of data collection, organization, and interpretation using software algorithms and bioinformatics. FUTURE DIRECTIONS Integrative and collaborative research, combining the power of numbers with careful experimental design, protocol development, and data interpretation, will translate cancer biology and therapeutics to a heightened level or leave the abundant raw data as stagnant and underutilized. Antioxid. Redox Signal. 25, 685-701.
Collapse
Affiliation(s)
| | - Karen Block
- Department of Medicine, University of Texas Health Science Center, San Antonio, Texas
- South Texas Veterans Health Care System, Audie L. Murphy Memorial Hospital Division, San Antonio, Texas
| |
Collapse
|
109
|
Ling G, Ji Q, Ye W, Ma D, Wang Y. Epithelial-mesenchymal transition regulated by p38/MAPK signaling pathways participates in vasculogenic mimicry formation in SHG44 cells transfected with TGF-β cDNA loaded lentivirus in vitro and in vivo. Int J Oncol 2016; 49:2387-2398. [PMID: 27748800 DOI: 10.3892/ijo.2016.3724] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 10/04/2016] [Indexed: 12/13/2022] Open
Abstract
TGF-β-induced epithelial-mesenchymal transition (EMT) plays an important role in tumor progression. We assessed whether the TGF-β-induced EMT contributed to vasculogenic mimicry (VM) formation in glioma, we established an SHG44 cell line stably transfected with TGF-β cDNA loaded lentivirus. SB203580 was employed to inhibit the TGF-β-induced EMT. The results showed that the VM forming ability of cells could be improved by TGF-β over-expression. The migration and invasion capabilities of cells were also enhanced due to EMT. SB203580 was able to weaken cell migration, invasion and VM forming abilities via blocking p38/MAPK signaling pathways, but it had tiny influence on MMP/LAMC2 chain. Consequently, we concluded that EMT inhibition via p38/MAPK signaling pathways would partly impair TGF-β-induced VM formation in glioma.
Collapse
Affiliation(s)
- Gengqiang Ling
- Department of Neurosurgery, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, P.R. China
| | - Qiao Ji
- Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Heilongjiang, P.R. China
| | - Wei Ye
- Department of Neurosurgery, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, P.R. China
| | - Dongying Ma
- Department of Neurosurgery, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, P.R. China
| | - Yuena Wang
- Department of Neurosurgery, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, P.R. China
| |
Collapse
|
110
|
Goyal S, Nangia-Makker P, Farhana L, Yu Y, Majumdar APN. Racial disparity in colorectal cancer: Gut microbiome and cancer stem cells. World J Stem Cells 2016; 8:279-287. [PMID: 27679684 PMCID: PMC5031889 DOI: 10.4252/wjsc.v8.i9.279] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 06/28/2016] [Accepted: 07/22/2016] [Indexed: 02/06/2023] Open
Abstract
Over the past two decades there has been remarkable progress in cancer diagnosis, treatment and screening. The basic mechanisms leading to pathogenesis of various types of cancers are also understood better and some patients, if diagnosed at a particular stage go on to lead a normal pre-diagnosis life. Despite these achievements, racial disparity in some cancers remains a mystery. The higher incidence, aggressiveness and mortality of breast, prostate and colorectal cancers (CRCs) in African-Americans as compared to Caucasian-Americans are now well documented. The polyp-carcinoma sequence in CRC and easy access to colonic epithelia or colonic epithelial cells through colonoscopy/colonic effluent provides the opportunity to study colonic stem cells early in course of natural history of the disease. With the advent of metagenomic sequencing, uncultivable organisms can now be identified in stool and their numbers correlated with the effects on colonic epithelia. It would be expected that these techniques would revolutionize our understanding of the racial disparity in CRC and pave a way for the same in other cancers as well. Unfortunately, this has not happened. Our understanding of the underlying factors responsible in African-Americans for higher incidence and mortality from colorectal carcinoma remains minimal. In this review, we aim to summarize the available data on role of microbiome and cancer stem cells in racial disparity in CRC. This will provide a platform for further research on this topic.
Collapse
|
111
|
Hahn JM, McFarland KL, Combs KA, Supp DM. Partial epithelial-mesenchymal transition in keloid scars: regulation of keloid keratinocyte gene expression by transforming growth factor-β1. BURNS & TRAUMA 2016; 4:30. [PMID: 27574697 PMCID: PMC4994224 DOI: 10.1186/s41038-016-0055-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 07/04/2016] [Indexed: 12/17/2022]
Abstract
Background Keloids are an extreme form of abnormal scarring that result from a pathological fibroproliferative wound healing process. The molecular mechanisms driving keloid pathology remain incompletely understood, hindering development of targeted, effective therapies. Recent studies in our laboratory demonstrated that keloid keratinocytes exhibit adhesion abnormalities and display a transcriptional signature reminiscent of cells undergoing epithelial-mesenchymal transition (EMT), suggesting a role for EMT in keloid pathology. In the current study, we further define the EMT-like phenotype of keloid scars and investigate regulation of EMT-related genes in keloid. Methods Primary keratinocytes from keloid scar and normal skin were cultured in the presence or absence of transforming growth factor beta 1 (TGF-β1) +/− inhibitors of TGF-β1 and downstream signaling pathways. Gene expression was measured using quantitative polymerase chain reaction. Migration was analyzed using an in vitro wound healing assay. Proteins in keloid scar and normal skin sections were localized by immunohistochemistry. Statistical analyses utilized SigmaPlot (SyStat Software, San Jose, CA) or SAS® (SAS Institute, Cary, NC). Results In keloid and normal keratinocytes, TGF-β1 regulated expression of EMT-related genes, including hyaluronan synthase 2, vimentin, cadherin-11, wingless-type MMTV integration site family, member 5A, frizzled 7, ADAM metallopeptidase domain 19, and interleukin-6. Inhibition of canonical TGF-β1 signaling in keloid keratinocytes significantly inhibited expression of these genes, and TGF-β1 stimulation of normal keratinocytes increased their expression. The inhibition of the extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathway or the p38 mitogen-activated protein kinase pathway attenuated TGF-β1-induced expression of subsets of these genes. Migration of keloid keratinocytes, previously shown to be increased compared with normal keratinocytes, was significantly reduced by inhibition of TGF-β1 or ERK1/2 signaling. Biomarkers of EMT, including reduced E-cadherin and increased active β-catenin, were observed in keloid epidermis in vivo. However, evidence of basement membrane breakdown in keloid scar was not observed. Conclusions The results suggest that keloid keratinocytes exist in an EMT-like metastable state, similar to activated keratinocytes in healing wounds. The EMT-like gene expression pattern of keloid keratinocytes is regulated by canonical and non-canonical TGF-β1 signaling pathways. Therefore, interventions targeting TGF-β1-regulated EMT-like gene expression in keloid keratinocytes may serve to suppress keloid scarring. Electronic supplementary material The online version of this article (doi:10.1186/s41038-016-0055-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jennifer M Hahn
- Research Department, Shriners Hospitals for Children - Cincinnati, Cincinnati, OH USA
| | - Kevin L McFarland
- Research Department, Shriners Hospitals for Children - Cincinnati, Cincinnati, OH USA
| | - Kelly A Combs
- Research Department, Shriners Hospitals for Children - Cincinnati, Cincinnati, OH USA
| | - Dorothy M Supp
- Research Department, Shriners Hospitals for Children - Cincinnati, Cincinnati, OH USA ; Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH USA
| |
Collapse
|
112
|
Gooding AJ, Schiemann WP. Harnessing protein kinase A activation to induce mesenchymal-epithelial programs to eliminate chemoresistant, tumor-initiating breast cancer cells. Transl Cancer Res 2016; 5:S226-S232. [PMID: 28680830 PMCID: PMC5495186 DOI: 10.21037/tcr.2016.08.09] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Alex J Gooding
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106
| | - William P Schiemann
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106
| |
Collapse
|
113
|
Fontelles CC, Guido LN, Rosim MP, Andrade FDO, Jin L, Inchauspe J, Pires VC, de Castro IA, Hilakivi-Clarke L, de Assis S, Ong TP. Paternal programming of breast cancer risk in daughters in a rat model: opposing effects of animal- and plant-based high-fat diets. Breast Cancer Res 2016; 18:71. [PMID: 27456846 PMCID: PMC4960664 DOI: 10.1186/s13058-016-0729-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 06/17/2016] [Indexed: 12/20/2022] Open
Abstract
Background Although males contribute half of the embryo’s genome, only recently has interest begun to be directed toward the potential impact of paternal experiences on the health of offspring. While there is evidence that paternal malnutrition may increase offspring susceptibility to metabolic diseases, the influence of paternal factors on a daughter’s breast cancer risk has been examined in few studies. Methods Male Sprague-Dawley rats were fed, before and during puberty, either a lard-based (high in saturated fats) or a corn oil-based (high in n-6 polyunsaturated fats) high-fat diet (60 % of fat-derived energy). Control animals were fed an AIN-93G control diet (16 % of fat-derived energy). Their 50-day-old female offspring fed only a commercial diet were subjected to the classical model of mammary carcinogenesis based on 7,12-dimethylbenz[a]anthracene initiation, and mammary tumor development was evaluated. Sperm cells and mammary gland tissue were subjected to cellular and molecular analysis. Results Compared with female offspring of control diet-fed male rats, offspring of lard-fed male rats did not differ in tumor latency, growth, or multiplicity. However, female offspring of lard-fed male rats had increased elongation of the mammary epithelial tree, number of terminal end buds, and tumor incidence compared with both female offspring of control diet-fed and corn oil-fed male rats. Compared with female offspring of control diet-fed male rats, female offspring of corn oil-fed male rats showed decreased tumor growth but no difference regarding tumor incidence, latency, or multiplicity. Additionally, female offspring of corn oil-fed male rats had longer tumor latency as well as decreased tumor growth and multiplicity compared with female offspring of lard-fed male rats. Paternal consumption of animal- or plant-based high-fat diets elicited opposing effects, with lard rich in saturated fatty acids increasing breast cancer risk in offspring and corn oil rich in n-6 polyunsaturated fatty acids decreasing it. These effects could be linked to alterations in microRNA expression in fathers’ sperm and their daughters’ mammary glands, and to modifications in breast cancer-related protein expression in this tissue. Conclusions Our findings highlight the importance of paternal nutrition in affecting future generations’ risk of developing breast cancer. Electronic supplementary material The online version of this article (doi:10.1186/s13058-016-0729-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Camile Castilho Fontelles
- Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, Avenida Professor Lineu Prestes 580, Bloco 14, São Paulo, SP, 05508-000, Brazil
| | - Luiza Nicolosi Guido
- Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, Avenida Professor Lineu Prestes 580, Bloco 14, São Paulo, SP, 05508-000, Brazil
| | - Mariana Papaléo Rosim
- Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, Avenida Professor Lineu Prestes 580, Bloco 14, São Paulo, SP, 05508-000, Brazil
| | - Fábia de Oliveira Andrade
- Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, Avenida Professor Lineu Prestes 580, Bloco 14, São Paulo, SP, 05508-000, Brazil
| | - Lu Jin
- Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, 20007, USA
| | - Jessica Inchauspe
- Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, 20007, USA
| | - Vanessa Cardoso Pires
- Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, Avenida Professor Lineu Prestes 580, Bloco 14, São Paulo, SP, 05508-000, Brazil
| | - Inar Alves de Castro
- Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, Avenida Professor Lineu Prestes 580, Bloco 14, São Paulo, SP, 05508-000, Brazil
| | | | - Sonia de Assis
- Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, 20007, USA
| | - Thomas Prates Ong
- Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, Avenida Professor Lineu Prestes 580, Bloco 14, São Paulo, SP, 05508-000, Brazil. .,Food Research Center (FoRC), São Paulo, 05508-000, Brazil.
| |
Collapse
|
114
|
Yan Q, Zhang W, Wu Y, Wu M, Zhang M, Shi X, Zhao J, Nan Q, Chen Y, Wang L, Cheng T, Li J, Bai Y, Liu S, Wang J. KLF8 promotes tumorigenesis, invasion and metastasis of colorectal cancer cells by transcriptional activation of FHL2. Oncotarget 2016; 6:25402-17. [PMID: 26320172 PMCID: PMC4694840 DOI: 10.18632/oncotarget.4517] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 07/03/2015] [Indexed: 12/23/2022] Open
Abstract
The transcription factor Krüppel-like factor (KLF)8 plays an important role in the formation of several human tumors, including colorectal cancer. We recently identified four-and-a-half LIM protein 2 (FHL2) as a critical inducer of the epithelial-to-mesenchymal transition (EMT) and invasion. However, the molecular mechanism by which KLF8 affects FHL2-mediated tumor proliferation, EMT and metastasis remains unknown. Here, we showed that KLF8 overexpression promoted EMT and metastatic phenotypes. KLF8 expression was stimulated by transforming growth factor (TGF)-β1. Moreover, KLF8 acted as a potential EMT inducer by stimulating vimentin expression and inducing a loss of E-cadherin in stable KLF8-transfected cells. KLF8 overexpression induced a strong increase in FHL2 expression, and a positive correlation between the expression patterns of KLF8 and FHL2 was observed in CRC cells. Promoter reporter and chromatin immunoprecipitation (ChIP) assays demonstrated that KLF8 directly bound to and activated the human FHL2 gene promoter. However, siRNA-mediated repression of FHL2 in KLF8-overexpressing cells reversed the EMT and the proliferative and metastatic phenotypes. In vivo, KLF8 promoted FHL2-mediated proliferation and metastasis via orthotopic implantation. Taken together, this work identified KLF8-induced FHL2 activation as a novel and critical signaling mechanism underlying human breast/colorectal cancer invasion and metastasis.
Collapse
Affiliation(s)
- Qingqing Yan
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wenjing Zhang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yao Wu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Meiyan Wu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mengnan Zhang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xinpeng Shi
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jinjun Zhao
- Department of Rheumatism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qingzhen Nan
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ye Chen
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Long Wang
- Division of Vascular Interventional Radiology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tianming Cheng
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiachu Li
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yang Bai
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Side Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jide Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
115
|
Brown WS, Tan L, Smith A, Gray NS, Wendt MK. Covalent Targeting of Fibroblast Growth Factor Receptor Inhibits Metastatic Breast Cancer. Mol Cancer Ther 2016; 15:2096-106. [PMID: 27371729 DOI: 10.1158/1535-7163.mct-16-0136] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 05/31/2016] [Indexed: 12/14/2022]
Abstract
Therapeutic targeting of late-stage breast cancer is limited by an inadequate understanding of how tumor cell signaling evolves during metastatic progression and by the currently available small molecule inhibitors capable of targeting these processes. Herein, we demonstrate that both β3 integrin and fibroblast growth factor receptor-1 (FGFR1) are part of an epithelial-mesenchymal transition (EMT) program that is required to facilitate metastatic outgrowth in response to fibroblast growth factor-2 (FGF2). Mechanistically, β3 integrin physically disrupts an interaction between FGFR1 and E-cadherin, leading to a dramatic redistribution of FGFR1 subcellular localization, enhanced FGF2 signaling and increased three-dimensional (3D) outgrowth of metastatic breast cancer cells. This ability of β3 integrin to drive FGFR signaling requires the enzymatic activity of focal adhesion kinase (FAK). Consistent with these mechanistic data, we demonstrate that FGFR, β3 integrin, and FAK constitute a molecular signature capable of predicting decreased survival of patients with the basal-like subtype of breast cancer. Importantly, covalent targeting of a conserved cysteine in the P-loop of FGFR1-4 with our newly developed small molecule, FIIN-4, more effectively blocks 3D metastatic outgrowth as compared with currently available FGFR inhibitors. In vivo application of FIIN-4 potently inhibited the growth of metastatic, patient-derived breast cancer xenografts and murine-derived metastases growing within the pulmonary microenvironment. Overall, the current studies demonstrate that FGFR1 works in concert with other EMT effector molecules to drive aberrant downstream signaling, and that these events can be effectively targeted using our novel therapeutics for the treatment of the most aggressive forms of breast cancer. Mol Cancer Ther; 15(9); 2096-106. ©2016 AACR.
Collapse
Affiliation(s)
- Wells S Brown
- Purdue University Center for Cancer Research, Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana
| | - Li Tan
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Andrew Smith
- Purdue University Center for Cancer Research, Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana
| | - Nathanael S Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Michael K Wendt
- Purdue University Center for Cancer Research, Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana.
| |
Collapse
|
116
|
Ueda S, Saeki T, Takeuchi H, Shigekawa T, Yamane T, Kuji I, Osaki A. In vivo imaging of eribulin-induced reoxygenation in advanced breast cancer patients: a comparison to bevacizumab. Br J Cancer 2016; 114:1212-8. [PMID: 27140309 PMCID: PMC4891505 DOI: 10.1038/bjc.2016.122] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 03/31/2016] [Accepted: 04/07/2016] [Indexed: 11/24/2022] Open
Abstract
Background: Eribulin mesylate (eribulin) is a first-in-class halichondrin B-based microtubule dynamics inhibitor. To compare the anti-angiogenic activity of eribulin to that of bevacizumab, we compared tumour vessel remodelling and reoxygenation between the two agents. Methods: Patients with advanced breast cancer with stage III/IV were eligible for the study. Patients were assigned to receive either eribulin or single-agent bevacizumab. Tissue concentrations of oxyhaemoglobin (O2Hb) and deoxyhaemoglobin (HHb), and oxygen saturation (SO2) of breast tumours before and day 7 after the first infusion were repeatedly measured using diffuse optical spectroscopic imaging (DOSI). A pair of blood samples was collected for multiplex biomarker studies. Results: Baseline DOSI measurement of all 29 patients (eribulin, n=14 and bevacizumab, n=15) revealed significantly higher tumour concentrations of O2Hb and HHb than that in the normal breast tissue. After eribulin treatment, DOSI revealed a significant decrease in HHb concentration and increased SO2 during the observation period. This trend was not observed for bevacizumab. Instead, bevacizumab significantly decreased the concentration of O2Hb. The multiplex biomarker study revealed that both eribulin and bevacizumab decreased plasma concentrations of VEGF and bFGF, but only eribulin treatment suppressed the plasma concentration of TGF-β1. Conclusions: Eribulin, but not bevacizumab, treatment increased tumour SO2. Suppression of TGF-β1 by eribulin could have a favourable anti-angiogenic effect. Our results suggest that differences in vascular remodelling between these two agents may account for their different effects on tumour reoxygenation.
Collapse
Affiliation(s)
- Shigeto Ueda
- Department of Breast Oncology, International Medical Center, Saitama Medical University, 1371-1 Yamane, Hidaka, Saitama 350-1298, Japan
| | - Toshiaki Saeki
- Department of Breast Oncology, International Medical Center, Saitama Medical University, 1371-1 Yamane, Hidaka, Saitama 350-1298, Japan
| | - Hideki Takeuchi
- Department of Breast Oncology, International Medical Center, Saitama Medical University, 1371-1 Yamane, Hidaka, Saitama 350-1298, Japan
| | - Takashi Shigekawa
- Department of Breast Oncology, International Medical Center, Saitama Medical University, 1371-1 Yamane, Hidaka, Saitama 350-1298, Japan
| | - Tomohiko Yamane
- Department of Nuclear Medicine, International Medical Center, Saitama Medical University, 1371-1 Yamane, Hidaka, Saitama 350-1298, Japan
| | - Ichiei Kuji
- Department of Nuclear Medicine, International Medical Center, Saitama Medical University, 1371-1 Yamane, Hidaka, Saitama 350-1298, Japan
| | - Akihiko Osaki
- Department of Breast Oncology, International Medical Center, Saitama Medical University, 1371-1 Yamane, Hidaka, Saitama 350-1298, Japan
| |
Collapse
|
117
|
Rateitschak K, Kaderali L, Wolkenhauer O, Jaster R. Autocrine TGF-β/ZEB/microRNA-200 signal transduction drives epithelial-mesenchymal transition: Kinetic models predict minimal drug dose to inhibit metastasis. Cell Signal 2016; 28:861-70. [PMID: 27000495 DOI: 10.1016/j.cellsig.2016.03.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 02/28/2016] [Accepted: 03/07/2016] [Indexed: 02/07/2023]
Abstract
The epithelial-mesenchymal transition (EMT) is the crucial step that cancer cells must pass before they can undergo metastasis. The transition requires the activity of complex functional networks that downregulate properties of the epithelial phenotype and upregulate characteristics of the mesenchymal phenotype. The networks frequently include reciprocal repressions between transcription factors (TFs) driving the EMT and microRNAs (miRs) inducing the reverse process, termed mesenchymal-epithelial transition (MET). In this work we develop four kinetic models that are based on experimental data and hypotheses describing how autocrine transforming growth factor-β (TGF-β) signal transduction induces and maintains an EMT by upregulating the TFs ZEB1 and ZEB2 which repress the expression of the miR-200b/c family members. After successful model calibration we validate our models by predicting requirements for the maintenance of the mesenchymal steady state which agree with experimental data. Finally, we apply our validated kinetic models for the design of experiments in cancer therapy. We demonstrate how steady state properties of the kinetic models, combined with data from tumor-derived cell lines of individual patients, can predict the minimal amount of an inhibitor to induce a MET.
Collapse
Affiliation(s)
- Katja Rateitschak
- Department of Systems Biology and Bioinformatics, University of Rostock, 18051 Rostock, Germany; Institute for Bioinformatics, University Medicine Greifswald, 17475 Greifswald, Germany.
| | - Lars Kaderali
- Institute for Bioinformatics, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Olaf Wolkenhauer
- Department of Systems Biology and Bioinformatics, University of Rostock, 18051 Rostock, Germany; Stellenbosch Institute for Advanced Study (STIAS), Wallenberg Research Center at Stellenbosch University, Marais Street, Stellenbosch 7600, South Africa
| | - Robert Jaster
- Department of Medicine II, Division of Gastroenterology, Rostock University Medical Center, 18057 Rostock, Germany
| |
Collapse
|
118
|
Majumder S, Bhowal A, Basu S, Mukherjee P, Chatterji U, Sengupta S. Deregulated E2F5/p38/SMAD3 Circuitry Reinforces the Pro-Tumorigenic Switch of TGFβ Signaling in Prostate Cancer. J Cell Physiol 2016; 231:2482-92. [PMID: 26919443 DOI: 10.1002/jcp.25361] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 02/23/2016] [Indexed: 12/13/2022]
Affiliation(s)
- Subhadipa Majumder
- Department of Biochemistry; University of Calcutta; Kolkata West Bengal India
| | - Ankur Bhowal
- Department of Zoology; University of Calcutta; Kolkata West Bengal India
| | - Sanmitra Basu
- Department of Biochemistry; University of Calcutta; Kolkata West Bengal India
| | - Pritha Mukherjee
- Department of Zoology; University of Calcutta; Kolkata West Bengal India
| | - Urmi Chatterji
- Department of Zoology; University of Calcutta; Kolkata West Bengal India
| | | |
Collapse
|
119
|
Runa F, Adamian Y, Kelber JA. Ascending the PEAK1 toward targeting TGFβ during cancer progression: Recent advances and future perspectives. CANCER CELL & MICROENVIRONMENT 2016; 3:e1162. [PMID: 29392163 PMCID: PMC5790177 DOI: 10.14800/ccm.1162] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cancer is the second leading cause of death in the United States. Mortality in patients with solid, epithelial-derived tumors strongly correlates with disease stage and the systemic metastatic load. In such cancers, notable morphological and molecular changes have been attributed to cells as they pass through a continuum of epithelial-mesenchymal transition (EMT) states and many of these changes are essential for metastasis. While cancer metastasis is a complex cascade that is regulated by cell-autonomous and microenvironmental influences, it is well-accepted that understanding and controlling metastatic disease is a viable method for increasing patient survival. In the past 5 years, the novel non-receptor tyrosine kinase PEAK1 has surfaced as a central regulator of tumor progression and metastasis in the context of solid, epithelial cancers. Here, we review this literature with a special focus on our recent work demonstrating that PEAK1 mediates non-canonical pro-tumorigenic TGFβ signaling and is an intracellular control point between tumor cells and their extracellular microenvironment. We conclude with a brief discussion of potential applications derived from our current understanding of PEAK1 biology.
Collapse
Affiliation(s)
- Farhana Runa
- Department of Biology, California State University, Northridge, CA, USA
| | - Yvess Adamian
- Department of Biology, California State University, Northridge, CA, USA
| | | |
Collapse
|
120
|
Stark T, Livas L, Kyprianou N. Inflammation in prostate cancer progression and therapeutic targeting. Transl Androl Urol 2016; 4:455-63. [PMID: 26816843 PMCID: PMC4708587 DOI: 10.3978/j.issn.2223-4683.2015.04.12] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Chronic inflammation contributes to the onset and progression of human cancer, via modifications in the tumor microenvironment by remodeling the extracellular matrix (ECM) and initiating epithelial mesenchymal transition (EMT). At the biological level, chronically inflamed cells release cytokines that are functionally dictating a constitutively active stroma, promoting tumor growth and metastasis. In prostate cancer, inflammation correlates with increased development of “risk factor” lesions or proliferative inflammatory atrophy (PIA). Chronic inflammation in benign prostate biopsy specimens can be associated with high-grade prostate tumors in adjacent areas. In this article, we discuss the current understanding of the incidence of inflammation in prostate cancer progression and the significance of the process in therapeutic targeting of specific inflammatory signaling pathways and critical effectors during tumor progression. Further understanding of the process of chronic inflammation in prostate tumor progression to metastasis will enable development and optimization of novel therapeutic modalities for the treatment of high-risk patients with advanced disease.
Collapse
Affiliation(s)
- Timothy Stark
- 1 Department of Urology, 2 Department of Molecular Biochemistry, 3 Department of Pathology, 4 The Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Lydia Livas
- 1 Department of Urology, 2 Department of Molecular Biochemistry, 3 Department of Pathology, 4 The Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Natasha Kyprianou
- 1 Department of Urology, 2 Department of Molecular Biochemistry, 3 Department of Pathology, 4 The Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| |
Collapse
|
121
|
Valcourt U, Carthy J, Okita Y, Alcaraz L, Kato M, Thuault S, Bartholin L, Moustakas A. Analysis of Epithelial-Mesenchymal Transition Induced by Transforming Growth Factor β. Methods Mol Biol 2016; 1344:147-81. [PMID: 26520123 DOI: 10.1007/978-1-4939-2966-5_9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In recent years, the importance of the cell biological process of epithelial-mesenchymal transition (EMT) has been established via an exponentially growing number of reports. EMT has been documented during embryonic development, tissue fibrosis, and cancer progression in vitro, in animal models in vivo and in human specimens. EMT relates to many molecular and cellular alterations that occur when epithelial cells undergo a switch in differentiation that generates mesenchymal-like cells with newly acquired migratory and invasive properties. In addition, EMT relates to a nuclear reprogramming similar to the one occurring in the generation of induced pluripotent stem cells. Via such a process, EMT is gradually established to promote the generation and maintenance of adult tissue stem cells which under disease states such as cancer, are known as cancer stem cells. EMT is induced by developmental growth factors, oncogenes, radiation, and hypoxia. A prominent growth factor that causes EMT is transforming growth factor β (TGF-β).A series of molecular and cellular techniques can be applied to define and characterize the state of EMT in diverse biological samples. These methods range from DNA and RNA-based techniques that measure the expression of key EMT regulators and markers of epithelial or mesenchymal differentiation to functional assays of cell mobility, invasiveness and in vitro stemness. This chapter focuses on EMT induced by TGF-β and provides authoritative protocols and relevant reagents and citations of key publications aiming at assisting newcomers that enter this prolific area of biomedical sciences, and offering a useful reference tool to pioneers and aficionados of the field.
Collapse
Affiliation(s)
- Ulrich Valcourt
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France.,CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France.,Université de Lyon, 69000, Lyon, France.,Université Lyon 1, 69000, Lyon, France.,Centre Léon Bérard, 69000, Lyon, France
| | - Jonathon Carthy
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, 751 24, Uppsala, Sweden
| | - Yukari Okita
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, 751 24, Uppsala, Sweden.,Department of Experimental Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Lindsay Alcaraz
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France.,CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France.,Université de Lyon, 69000, Lyon, France.,Université Lyon 1, 69000, Lyon, France.,Centre Léon Bérard, 69000, Lyon, France
| | - Mitsuyasu Kato
- Department of Experimental Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Sylvie Thuault
- INSERM UMR 911 CRO2, Faculty of Pharmacy, Marseille, France
| | - Laurent Bartholin
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France.,CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France.,Université de Lyon, 69000, Lyon, France.,Université Lyon 1, 69000, Lyon, France.,Centre Léon Bérard, 69000, Lyon, France
| | - Aristidis Moustakas
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, 751 24, Uppsala, Sweden. .,Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Box 582, 751 23, Uppsala, Sweden.
| |
Collapse
|
122
|
Sha K, Yeh S, Chang C, Nastiuk KL, Krolewski JJ. TNF signaling mediates an enzalutamide-induced metastatic phenotype of prostate cancer and microenvironment cell co-cultures. Oncotarget 2015; 6:25726-40. [PMID: 26327448 PMCID: PMC4694862 DOI: 10.18632/oncotarget.4535] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 07/17/2015] [Indexed: 12/25/2022] Open
Abstract
The dramatic responses tumors display to targeted therapies are limited by acquired or pre-existing mechanisms of therapy resistance. We recently discovered that androgen receptor blockade by the anti-androgen enzalutamide paradoxically enhanced metastasis and that these pro-metastatic effects were mediated by the chemoattractant CCL2. CCL2 is regulated by TNF, which is negatively regulated by androgen signaling. Thus, we asked if TNF mediates the pro-metastatic effects of enzalutamide. We found that androgen withdrawal or enzalutamide induced TNF mRNA and protein secretion in castration resistant prostate cancer (C4-2) cells, but not in macrophage-like (THP1) or myofibroblast-like (WPMY1) cells. Androgen deprivation therapy (ADT) induced autocrine CCL2 expression in C4-2 (as well as a murine CRPC cell line), while exogenous TNF induced CCL2 in THP1 and WPMY1. TNF was most potent in myofibroblast cultures, suggesting ADT induces CCL2 via paracrine interactions within the tumor microenvironment. A soluble TNF receptor (etanercept) blocked enzalutamide-induced CCL2 protein secretion and mRNA, implying dependence on secreted TNF. A small molecule inhibitor of CCR2 (the CCL2 receptor) significantly reduced TNF induced migration, while etanercept inhibited enzalutamide-induced migration and invasion of C4-2. Analysis of human prostate cancers suggests that a TNF-CCL2 paracrine loop is induced in response to ADT and might account for some forms of prostate cancer therapy resistance.
Collapse
MESH Headings
- Androgen Antagonists/pharmacology
- Animals
- Antineoplastic Agents, Hormonal/pharmacology
- Benzamides
- Cell Line, Tumor
- Cell Movement/drug effects
- Chemokine CCL2/genetics
- Chemokine CCL2/metabolism
- Coculture Techniques
- Dose-Response Relationship, Drug
- Drug Resistance, Neoplasm
- Gene Expression Regulation, Neoplastic
- Humans
- Male
- Mice
- Myofibroblasts/drug effects
- Myofibroblasts/metabolism
- Myofibroblasts/pathology
- Neoplasm Invasiveness
- Neoplasm Metastasis
- Nitriles
- Paracrine Communication/drug effects
- Phenylthiohydantoin/analogs & derivatives
- Phenylthiohydantoin/pharmacology
- Prostatic Neoplasms, Castration-Resistant/drug therapy
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/metabolism
- Prostatic Neoplasms, Castration-Resistant/pathology
- Receptors, CCR2/antagonists & inhibitors
- Receptors, CCR2/metabolism
- Signal Transduction/drug effects
- Time Factors
- Tumor Microenvironment/drug effects
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/metabolism
- Tumor Necrosis Factor-alpha/pharmacology
- Up-Regulation
Collapse
Affiliation(s)
- Kai Sha
- Department of Pathology and Laboratory Medicine, University of Rochester, School of Medicine and Dentistry; Rochester, NY 14642, USA
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY 14263
| | - Shuyuan Yeh
- Department of Pathology and Laboratory Medicine, University of Rochester, School of Medicine and Dentistry; Rochester, NY 14642, USA
- Department of Urology, University of Rochester, School of Medicine and Dentistry; Rochester, NY 14642, USA
| | - Chawnshang Chang
- Department of Pathology and Laboratory Medicine, University of Rochester, School of Medicine and Dentistry; Rochester, NY 14642, USA
- Department of Urology, University of Rochester, School of Medicine and Dentistry; Rochester, NY 14642, USA
- Department of Radiation Oncology, University of Rochester, School of Medicine and Dentistry; Rochester, NY 14642, USA
- Wilmot Cancer Institute, University of Rochester, School of Medicine and Dentistry; Rochester, NY 14642, USA
| | - Kent L. Nastiuk
- Department of Pathology and Laboratory Medicine, University of Rochester, School of Medicine and Dentistry; Rochester, NY 14642, USA
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY 14263
| | - John J. Krolewski
- Department of Pathology and Laboratory Medicine, University of Rochester, School of Medicine and Dentistry; Rochester, NY 14642, USA
- Wilmot Cancer Institute, University of Rochester, School of Medicine and Dentistry; Rochester, NY 14642, USA
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY 14263
- Center for Personalized Medicine, Roswell Park Cancer Institute, Buffalo, NY 14263
| |
Collapse
|
123
|
O'Leary K, Shia A, Cavicchioli F, Haley V, Comino A, Merlano M, Mauri F, Walter K, Lackner M, Wischnewsky MB, Crook T, Lo Nigro C, Schmid P. Identification of Endoglin as an epigenetically regulated tumour-suppressor gene in lung cancer. Br J Cancer 2015; 113:970-8. [PMID: 26325105 PMCID: PMC4578092 DOI: 10.1038/bjc.2015.302] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 07/17/2015] [Accepted: 07/29/2015] [Indexed: 12/25/2022] Open
Abstract
Background: The transforming growth factor-beta (TGF- β) pathway has been implicated in proliferation, migration and invasion of various cancers. Endoglin is a TGF-β accessory receptor that modulates signalling. We identified Endoglin as an epigenetically silenced tumour-suppressor gene in lung cancer by means of a genome-wide screening approach, then sought to characterise its effect on lung cancer progression. Methods: Methylation microarray and RNA sequencing were carried out on lung cancer cell lines. Epigenetic silencing of Endoglin was confirmed by methylation and expression analyses. An expression vector and a 20-gene expression panel were used to evaluate Endoglin function. Pyrosequencing was carried out on two independent cohorts comprising 112 and 202 NSCLC cases, respectively, and the impact of Endoglin methylation on overall survival (OS) was evaluated. Results: Methylation in the promoter region resulted in silencing of Endoglin, which could be reactivated by demethylation. Increased invasion coupled with altered EMT marker expression was observed in cell lines with an epithelial-like, but not those with a mesenchymal-like, profile when Endoglin was absent. Methylation was associated with decreased OS in stage I but not in stages II–III disease. Conclusions: We show that Endoglin is a common target of epigenetic silencing in lung cancer. We reveal a link between Endoglin silencing and EMT progression that might be associated with decreased survival in stage I disease.
Collapse
Affiliation(s)
- K O'Leary
- Brighton and Sussex Medical School, University of Sussex, Brighton BN1 9RY, UK
| | - A Shia
- Brighton and Sussex Medical School, University of Sussex, Brighton BN1 9RY, UK.,Barts Cancer Institute, Queen Mary University of London, Old Anatomy Building, Charterhouse Square, London EC1M 6BQ, UK
| | - F Cavicchioli
- Brighton and Sussex Medical School, University of Sussex, Brighton BN1 9RY, UK
| | - V Haley
- Brighton and Sussex Medical School, University of Sussex, Brighton BN1 9RY, UK
| | - A Comino
- Pathology Department, S. Croce General Hospital, via Coppino 26, 12100, Cuneo, Italy
| | - M Merlano
- Medical Oncology, Oncology Department, S. Croce General Hospital, via Carle 25, 12100, Cuneo, Italy
| | - F Mauri
- Department of Histopathology, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK
| | - K Walter
- Oncology Biomarker Development, Genentech, Inc., 550 Grandview Boulevard, South San Francisco, CA 94080, USA
| | - M Lackner
- Oncology Biomarker Development, Genentech, Inc., 550 Grandview Boulevard, South San Francisco, CA 94080, USA
| | - M B Wischnewsky
- eScience Lab, Department of Biomathematics, University of Bremen, Bremen 28359, Germany
| | - T Crook
- Division of Cancer Research, Medical Research Institute, Jacqui Wood Cancer Centre, University of Dundee, Ninewells Hospital And Medical School, Dundee DD1 9SY, UK
| | - C Lo Nigro
- Laboratory of Cancer Genetics and Translational Oncology, Oncology Department, S. Croce Genreal Hospital, via Carle 25, Cuneo 12100, Italy
| | - P Schmid
- Brighton and Sussex Medical School, University of Sussex, Brighton BN1 9RY, UK.,Barts Cancer Institute, Queen Mary University of London, Old Anatomy Building, Charterhouse Square, London EC1M 6BQ, UK
| |
Collapse
|
124
|
Agajanian M, Runa F, Kelber JA. Identification of a PEAK1/ZEB1 signaling axis during TGFβ/fibronectin-induced EMT in breast cancer. Biochem Biophys Res Commun 2015; 465:606-12. [DOI: 10.1016/j.bbrc.2015.08.071] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 08/16/2015] [Indexed: 10/23/2022]
|
125
|
Xia X, Zhang K, Cen G, Jiang T, Cao J, Huang K, Huang C, Zhao Q, Qiu Z. MicroRNA-301a-3p promotes pancreatic cancer progression via negative regulation of SMAD4. Oncotarget 2015; 6:21046-63. [PMID: 26019136 PMCID: PMC4673249 DOI: 10.18632/oncotarget.4124] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 05/02/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Aim to determine the clinicopathological and prognostic role of miR-301a-3p in pancreatic ductal adenocarcinoma(PDAC), to investigate the biological mechanism of miR-301a-3p in vitro and in vivo. METHODS By tissue microarray analysis, we studied miR-301a-3p expression in PDAC patients and its clinicopathological correlations as well as prognostic significance. qRT-PCR was used to test miR-301a-3p expression in PDAC tissues and cell lines. Functional experiments including in vitro and in vivo were performed. RESULTS Significantly higher expression of miR-301a-3p were found in PDAC patients with lymph node metastasis and advanced pathological stages and identified as an independent prognostic factor for worse survival. In PDAC samples and cell lines, miR-301a-3p was significantly up-regulated compared with matched non-tumor tissues and normal pancreatic ductal cells, respectively. Overexpression of miR-301a-3p enhanced PDAC cells colony, invasion and migration abilities in vitro as well as tumorigenicity in vivo. Furthermore, SMAD4 was identified as a target gene of miR-301a-3p by cell as well as mice xenograft experiments. In PDAC tissue microarray, a significantly inverse correlation between miR-301a-3p ISH scores and SMAD4 IHC scores were observed in both tumor and corresponding non-tumor tissues. CONCLUSIONS MiR-301a-3p functions as a novel oncogene in PDAC and the oncogenic activity may involve its inhibition of the target gene SMAD4.
Collapse
Affiliation(s)
- Xiang Xia
- Department of General Surgery, Shanghai Jiaotong University Affiliated First People's Hospital, Shanghai, China
| | - Kundong Zhang
- Department of General Surgery, Shanghai Jiaotong University Affiliated First People's Hospital, Shanghai, China
| | - Gang Cen
- Department of General Surgery, Shanghai Jiaotong University Affiliated First People's Hospital, Shanghai, China
| | - Tao Jiang
- Department of General Surgery, Shanghai Jiaotong University Affiliated First People's Hospital, Shanghai, China
| | - Jun Cao
- Department of General Surgery, Shanghai Jiaotong University Affiliated First People's Hospital, Shanghai, China
| | - Kejian Huang
- Department of General Surgery, Shanghai Jiaotong University Affiliated First People's Hospital, Shanghai, China
| | - Chen Huang
- Department of General Surgery, Shanghai Jiaotong University Affiliated First People's Hospital, Shanghai, China
| | - Qian Zhao
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis and National Ministry of Education, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhengjun Qiu
- Department of General Surgery, Shanghai Jiaotong University Affiliated First People's Hospital, Shanghai, China
| |
Collapse
|
126
|
Agajanian M, Campeau A, Hoover M, Hou A, Brambilla D, Kim SL, Klemke RL, Kelber JA. PEAK1 Acts as a Molecular Switch to Regulate Context-Dependent TGFβ Responses in Breast Cancer. PLoS One 2015; 10:e0135748. [PMID: 26267863 PMCID: PMC4533969 DOI: 10.1371/journal.pone.0135748] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 07/25/2015] [Indexed: 12/21/2022] Open
Abstract
Transforming Growth Factor β (TGFβ) has dual functions as both a tumor suppressor and a promoter of cancer progression within the tumor microenvironment, but the molecular mechanisms by which TGFβ signaling switches between these outcomes and the contexts in which this switch occurs remain to be fully elucidated. We previously identified PEAK1 as a new non-receptor tyrosine kinase that associates with the cytoskeleton, and facilitates signaling of HER2/Src complexes. We also showed PEAK1 functions downstream of KRas to promote tumor growth, metastasis and therapy resistance using preclinical in vivo models of human tumor progression. In the current study, we analyzed PEAK1 expression in human breast cancer samples and found PEAK1 levels correlate with mesenchymal gene expression, poor cellular differentiation and disease relapse. At the cellular level, we also observed that PEAK1 expression was highest in mesenchymal breast cancer cells, correlated with migration potential and increased in response to TGFβ-induced epithelial-mesenchymal transition (EMT). Thus, we sought to evaluate the role of PEAK1 in the switching of TGFβ from a tumor suppressing to tumor promoting factor. Notably, we discovered that high PEAK1 expression causes TGFβ to lose its anti-proliferative effects, and potentiates TGFβ-induced proliferation, EMT, cell migration and tumor metastasis in a fibronectin-dependent fashion. In the presence of fibronectin, PEAK1 caused a switching of TGFβ signaling from its canonical Smad2/3 pathway to non-canonical Src and MAPK signaling. This report is the first to provide evidence that PEAK1 mediates signaling cross talk between TGFβ receptors and integrin/Src/MAPK pathways and that PEAK1 is an important molecular regulator of TGFβ-induced tumor progression and metastasis in breast cancer. Finally, PEAK1 overexpression/upregulation cooperates with TGFβ to reduce breast cancer sensitivity to Src kinase inhibition. These findings provide a rational basis to develop therapeutic agents to target PEAK1 expression/function or upstream/downstream pathways to abrogate breast cancer progression.
Collapse
Affiliation(s)
- Megan Agajanian
- Department of Biology, California State University Northridge, Northridge, CA 91330, United States of America
| | - Anaamika Campeau
- Department of Biology, California State University Northridge, Northridge, CA 91330, United States of America
| | - Malachia Hoover
- Department of Biology, California State University Northridge, Northridge, CA 91330, United States of America
| | - Alexander Hou
- Department of Biology, California State University Northridge, Northridge, CA 91330, United States of America
- Department of Biology, Georgetown University, Washington, DC 20057, United States of America
| | - Daniel Brambilla
- Department of Biology, California State University Northridge, Northridge, CA 91330, United States of America
| | - Sa La Kim
- Department of Biology, California State University Northridge, Northridge, CA 91330, United States of America
| | - Richard L. Klemke
- Department of Pathology & Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, United States of America
| | - Jonathan A. Kelber
- Department of Biology, California State University Northridge, Northridge, CA 91330, United States of America
- * E-mail:
| |
Collapse
|
127
|
Cichon MA, Radisky DC. Extracellular matrix as a contextual determinant of transforming growth factor-β signaling in epithelial-mesenchymal transition and in cancer. Cell Adh Migr 2015; 8:588-94. [PMID: 25482625 PMCID: PMC4594483 DOI: 10.4161/19336918.2014.972788] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Extracellular matrix (ECM) provides both structural support and contextual information to cells within tissues and organs. The combination of biochemical and biomechanical signals from the ECM modulates responses to extracellular signals toward differentiation, proliferation, or apoptosis; alterations in the ECM are necessary for development and remodeling processes, but aberrations in the composition and organization of ECM are associated with disease pathology and can predispose to development of cancer. The primary cell surface sensors of the ECM are the integrins, which provide the physical connection between the ECM and the cytoskeleton and also convey biochemical information about the composition of the ECM. Transforming growth factor-β (TGF-β) is an extracellular signaling molecule that is a powerful controller of a variety of cellular functions, and that has been found to induce very different outcomes according to cell type and cellular context. It is becoming clear that ECM-mediated signaling through integrins is reciprocally influenced by TGF-β: integrin expression, activation, and responses are affected by cellular exposure to TGF-β, and TGF-β activation and cellular responses are in turn controlled by signaling from the ECM through integrins. Epithelial-mesenchymal transition (EMT), a physiological process that is activated by TGF-β in normal development and in cancer, is also affected by the composition and structure of the ECM. Here, we will outline how signaling from the ECM controls the contextual response to TGF-β, and how this response is selectively modulated during disease, with an emphasis on recent findings, current challenges, and future opportunities.
Collapse
|
128
|
Winkler I, Wilczynska B, Bojarska-Junak A, Gogacz M, Adamiak A, Postawski K, Darmochwal-Kolarz D, Rechberger T, Tabarkiewicz J. Regulatory T lymphocytes and transforming growth factor beta in epithelial ovarian tumors-prognostic significance. J Ovarian Res 2015; 8:39. [PMID: 26077607 PMCID: PMC4513978 DOI: 10.1186/s13048-015-0164-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 06/01/2015] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Regulatory T lymphocytes (Treg) are characterized by the presence of CD4+ surface antigen. Today the transcription factor FOXP3 is considered to be the most specific marker of Treg cells. The aim of the study was to estimate the percentage of Treg in peripheral blood and the tissue of the epithelial ovarian tumor and blood serum TGF-beta concentrations and relationships between them. Moreover, the aim of the study was to answer the question whether the percentage of Treg lymphocytes affects the time of survival in patients with ovarian cancer. METHODS The patients were divided into four groups, depending on the histopathological examination result: I--a group without any pathology within the ovaries (C; n = 20), II--a group with benign tumors (B; n = 25), III - with borderline tumors (BR; n = 11), IV--a group with cancer of the ovary (M; n = 24). The percentage of Treg lymphocytes in peripheral blood and the tissue was assessed using the flow cytometry method. TGF-beta cytokine concentration was estimated with the ELISA immunoenzymatic test. Statistical analysis of the results was conducted using the computer program Statistica 10.0PL (StatSoft, Inc). RESULTS No significant differences were found in percentages of Treg lymphocytes in peripheral blood between individual groups of patients (p = 0.11). However, we observed marked differences in the tissue of malignant and non-malignant tumors between individual groups of patients (p = 0.003). The analysis with the post hoc test revealed significantly higher TGF-beta concentration in the group of women with malignant tumors. Moreover, no relationship was found between TGF-beta concentration and the percentage of Treg cells in peripheral blood and tumors of the ovary. No correlation was found between the percentage of Treg lymphocytes in peripheral blood (p = 0.4) and the tissue of ovarian tumors (p = 0.3) and the time of survival of patients with ovarian cancer. CONCLUSIONS The recruitment of Treg lymphocytes toward the tumor is one of the mechanisms of escape of neoplasm from the response of the immune system. The percentage of Treg lymphocytes in peripheral blood and the neoplastic tissue does not influence the time of survival of patients with ovarian cancer.
Collapse
Affiliation(s)
- Izabela Winkler
- II Department of Surgical Gynaecology, Medical University in Lublin, Jaczewski Street, 20-954, Lublin, Poland.
| | - Barbara Wilczynska
- Department of Clinical Immunology, Medical University in Lublin, Chodźki 4a Street, 20-093, Lublin, Poland. .,Department of Paediatric Endocrinology and Diabetology with Endocrine-Metabolic Laboratory, Chodźki 2 Street, 20-093, Lublin, Poland.
| | - Agnieszka Bojarska-Junak
- Department of Clinical Immunology, Medical University in Lublin, Chodźki 4a Street, 20-093, Lublin, Poland.
| | - Marek Gogacz
- II Department of Surgical Gynaecology, Medical University in Lublin, Jaczewski Street, 20-954, Lublin, Poland.
| | - Aneta Adamiak
- II Department of Surgical Gynaecology, Medical University in Lublin, Jaczewski Street, 20-954, Lublin, Poland.
| | - Krzysztof Postawski
- II Department of Surgical Gynaecology, Medical University in Lublin, Jaczewski Street, 20-954, Lublin, Poland.
| | - Dorota Darmochwal-Kolarz
- Department of Obstetrics and Perinatology, Medical University of Lublin, Jaczewski Street, 20-954, Lublin, Poland. .,Centre for Innovative Research in Medical and Natural Sciences, Medical Faculty of University of Rzeszów, Warzywna Street, 35-959, Rzeszów, Poland.
| | - Tomasz Rechberger
- II Department of Surgical Gynaecology, Medical University in Lublin, Jaczewski Street, 20-954, Lublin, Poland.
| | - Jacek Tabarkiewicz
- Centre for Innovative Research in Medical and Natural Sciences, Medical Faculty of University of Rzeszów, Warzywna Street, 35-959, Rzeszów, Poland.
| |
Collapse
|
129
|
Li Q, Hou L, Ding G, Li Y, Wang J, Qian B, Sun J, Wang Q. KDM6B induces epithelial-mesenchymal transition and enhances clear cell renal cell carcinoma metastasis through the activation of SLUG. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:6334-6344. [PMID: 26261509 PMCID: PMC4525843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 05/17/2015] [Indexed: 06/04/2023]
Abstract
Clear cell renal cell carcinoma (ccRCC) is one of the most common kidney cancers; epithelial-mesenchymal transition (EMT) is associated with carcinoma invasion and metastasis. There have been several studies about the molecular regulation of EMT, but the relationship between histone demethylase and EMT is little known. Here, we reported KDM6B has high expression level in ccRCC and is positively correlated with poor ccRCC prognosis. KDM6B, also known as JMJD3, is a histone demethylase, can remove repressive histone H3K27me3 marks from chromatin, thereby activating gene expression. We found that the knockdown of KDM6B could inhibit ccRCC tumorigenesis in vitro; furthermore, KDM6B could induce EMT in ccRCC cells by activating the expression of master transcription factor SLUG. ChIP assays revealed that KDM6B stimulated SLUG expression by demethylate histone H3K27me3. The knockdown of KDM6B strongly inhibited ccRCC cell invasion in vitro, while the overexpression of KDM6B shown the opposite trend. Meanwhile, our analysis of the ccRCC tissue found that KDM6B expression was significantly corresponded with lymph node metastasis. Together, our data provide a novel epigenetic mechanism regulating tumor cell invasion and EMT, and provide a biomolecule for ccRCC diagnosis and prognosis.
Collapse
Affiliation(s)
- Qiang Li
- Department of Urology, The First Affiliated Hospital of Medical College, Shihezi UniversityShihezi 832008, China
| | - Liejun Hou
- Department of Urology, The Affiliated Hospital of Medical College, Ningbo UniversityNingbo, China
| | - Guofu Ding
- Department of Urology, The First Affiliated Hospital of Medical College, Shihezi UniversityShihezi 832008, China
| | - Yinglong Li
- Department of Urology, The First Affiliated Hospital of Medical College, Shihezi UniversityShihezi 832008, China
| | - Jiangping Wang
- Department of Urology, The First Affiliated Hospital of Medical College, Shihezi UniversityShihezi 832008, China
| | - Biao Qian
- Department of Urology, The First Affiliated Hospital of Medical College, Shihezi UniversityShihezi 832008, China
| | - Jianguo Sun
- Institute of Physiology and Psychology, School of Basic Medical Sciences, Shanghai Jiao Tong UniversityShanghai, China
| | - Qinzhang Wang
- Department of Urology, The First Affiliated Hospital of Medical College, Shihezi UniversityShihezi 832008, China
| |
Collapse
|
130
|
Xia YY, Yin L, Jiang N, Guo WJ, Tian H, Jiang XS, Wu J, Chen M, Wu JZ, He X. Downregulating HMGA2 attenuates epithelial-mesenchymal transition-induced invasion and migration in nasopharyngeal cancer cells. Biochem Biophys Res Commun 2015; 463:357-63. [PMID: 26025649 DOI: 10.1016/j.bbrc.2015.05.068] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 05/17/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Epithelial-mesenchymal transition (EMT) is associated with invasion and metastasis of cancer cells. High-mobility group AT-hook 2 (HMGA2) has been found to play a critical role in EMT in a number of malignant tumors. However, whether HMGA2 regulates the EMT in human nasopharyngeal carcinoma (NPC) is unclear. OBJECTIVE The aim of this study was to investigate the effect and mechanism of HMGA2 in inducing invasion and migration in NPC. METHODS In NPC tissues samples, the association of HMGA2 mRNA expression with clinicopathological characteristics were estimated by real-time quantitative RT-PCR(qRT-PCR). In vitro, following the silencing of HMGA2 in CNE-1 and CNE-2 cell lines, the viability and metastatic ability were analyzed using Cell Counting Kit-8 (CCK8), colony formation assay, and transwell assay. EMT and transforming growth factor-beta (TGFβ)/Smad3 signaling pathway-related protein expression changes were evaluated using western blot. RESULTS HMGA2 was upregulated in NPC cell lines and clinical specimens (P < 0.01), and HMGA2 expression correlated significantly with metastasis (P = 0.02) and disease-free survival of NPC (hazard ratio: 3.52; 95% confidence interval: 1.34-7.79; P = 0.01). In addition, following in vitro knockdown of HMGA2, the aggressiveness of cells was markedly inhibited, Vimentin and Snail level was downregulated and E-cadherin expression was upregulated. Moreover, the expression of key proteins TGFβRII and p-Smad3 of the TGFβ/Smad3 signaling pathway was inhibited by the downregulation of HMGA2. CONCLUSION HMGA2 might maintain EMT-induced invasion and migration through the TGFβ/Smad3 signaling pathway in NPC cell lines.
Collapse
Affiliation(s)
- You-You Xia
- Department of Radiation Oncology, Lianyungang First People's Hospital, Lianyungang, Jiangsu, China; The Fourth Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Li Yin
- Department of Radiation Oncology, The Affiliated Jiangsu Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ning Jiang
- Department of Radiation Oncology, The Affiliated Jiangsu Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wen-Jie Guo
- Department of Radiation Oncology, The Affiliated Jiangsu Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hao Tian
- The Fourth Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xue-Song Jiang
- Department of Radiation Oncology, The Affiliated Jiangsu Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jing Wu
- The Fourth Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Meng Chen
- The Fourth Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jian-Zhong Wu
- Research Center of Clinical Oncology, The Affiliated Jiangsu Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xia He
- Department of Radiation Oncology, The Affiliated Jiangsu Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
131
|
Krušlin B, Ulamec M, Tomas D. Prostate cancer stroma: an important factor in cancer growth and progression. Bosn J Basic Med Sci 2015; 15:1-8. [PMID: 26042506 PMCID: PMC4469930 DOI: 10.17305/bjbms.2015.449] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 05/04/2015] [Accepted: 05/04/2015] [Indexed: 12/30/2022] Open
Abstract
Reactive stromal changes that occur in different human cancers might play a role in local tumor spreading and progression. Studies done on various human cancers have shown activated stromal cell phenotypes, modified extracellular matrix (ECM) composition, and increased microvessel density. Furthermore, they exhibit biological markers consistent with stroma at the site of wound repair. In prostate cancer, stroma is composed of fibroblasts, myofibroblasts, endothelial cells and immune cells. Predominant cells in the tumorous stroma are, however, fibroblasts/myofibroblasts. They are responsible for the synthesis, deposition and remodeling of the ECM. Epithelial tumorous cells, in interaction with stromal cells and with the help of various molecules of ECM, create a microenvironment suitable for cancer cell proliferation, movement, and differentiation. In this review, we discussed the role of different stromal components in prostate cancer as well as their potential prognostic and therapeutic significance.
Collapse
Affiliation(s)
- Božo Krušlin
- Department of pathology, Sestre milosrdnice University Hospital, Zagreb.
| | | | | |
Collapse
|
132
|
Zhe C, Yu F, Tian J, Zheng S. Trps1 regulates biliary epithelial-mesenchymal transition and has roles during biliary fibrosis in liver grafts: a preliminary study. PLoS One 2015; 10:e0123233. [PMID: 25886207 PMCID: PMC4401436 DOI: 10.1371/journal.pone.0123233] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 03/01/2015] [Indexed: 11/18/2022] Open
Abstract
Objective To investigate the role(s) of Trps1 in non-anastomotic biliary stricture (NABS) following liver transplantation. Methods Immunohistochemical and histological techniques were used to detect Trps1, E-cadherin, CK19, vimentin, α-SMA, and collagen deposition. Human intrahepatic biliary epithelial cells (HIBECs) were infected with a Trps1 adenovirus, or transfected with Trps1 short-interfering RNAs (siRNAs). Reverse transcription polymerase chain reaction (RT-PCR) assays and western blotting were used to determine expression levels of epithelial and mesenchymal markers, and Trps1 in HIBECs. Results Expression of Trps1 and epithelial markers was down-regulated or absent in NABS liver samples. Mesenchymal markers were seen in biliary epithelial cells (BECs), with collagen deposited around the bile duct. Trps1 expression positively correlated with epithelial markers. Expression of epithelial marker mRNAs and proteins in HIBECs decreased with prolonged cold preservation (CP), while mesenchymal marker expression increased. A 12-h CP period led to increased Trps1 mRNA and protein levels. Expression of E-cadherin was increased in HIBECs following Trps1 adenovirus infection and CP/reperfusion injury (CPRI), with vimentin expression levels reduced and CPRI-mediated epithelial-mesenchymal transition (EMT) inhibited. Transfection of HIBECs with Trps1 siRNAs in conjunction with CPRI revealed that E-cadherin expression was decreased, vimentin expression was increased, and CPRI-mediated EMT was promoted. Conclusion Trps1 is involved in NABS pathogenesis following liver transplantation and negatively correlates with BEC EMT and biliary fibrosis in liver grafts. Trps1 demonstrates antagonistic effects that could reverse EMT.
Collapse
Affiliation(s)
- Cheng Zhe
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, No. 29 Gaotanyan Road, Shapingba District, Chongqing, 400038, China
| | - Fan Yu
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, No. 29 Gaotanyan Road, Shapingba District, Chongqing, 400038, China
| | - Ju Tian
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, No. 29 Gaotanyan Road, Shapingba District, Chongqing, 400038, China
| | - Shuguo Zheng
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, No. 29 Gaotanyan Road, Shapingba District, Chongqing, 400038, China
- * E-mail:
| |
Collapse
|
133
|
Thacker PC, Karunagaran D. Curcumin and emodin down-regulate TGF-β signaling pathway in human cervical cancer cells. PLoS One 2015; 10:e0120045. [PMID: 25786122 PMCID: PMC4365016 DOI: 10.1371/journal.pone.0120045] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 02/02/2015] [Indexed: 12/20/2022] Open
Abstract
Cervical cancer is the major cause of cancer related deaths in women, especially in developing countries and Human Papilloma Virus infection in conjunction with multiple deregulated signaling pathways leads to cervical carcinogenesis. TGF-β signaling in later stages of cancer is known to induce epithelial to mesenchymal transition promoting tumor growth. Phytochemicals, curcumin and emodin, are effective as chemopreventive and chemotherapeutic compounds against several cancers including cervical cancer. The main objective of this work was to study the effect of curcumin and emodin on TGF-β signaling pathway and its functional relevance to growth, migration and invasion in two cervical cancer cell lines, SiHa and HeLa. Since TGF-β and Wnt/β-catenin signaling pathways are known to cross talk having common downstream targets, we analyzed the effect of TGF-β on β-catenin (an important player in Wnt/β-catenin signaling) and also studied whether curcumin and emodin modulate them. We observed that curcumin and emodin effectively down regulate TGF-β signaling pathway by decreasing the expression of TGF-β Receptor II, P-Smad3 and Smad4, and also counterbalance the tumorigenic effects of TGF-β by inhibiting the TGF-β-induced migration and invasion. Expression of downstream effectors of TGF-β signaling pathway, cyclinD1, p21 and Pin1, was inhibited along with the down regulation of key mesenchymal markers (Snail and Slug) upon curcumin and emodin treatment. Curcumin and emodin were also found to synergistically inhibit cell population and migration in SiHa and HeLa cells. Moreover, we found that TGF-β activates Wnt/β-catenin signaling pathway in HeLa cells, and curcumin and emodin down regulate the pathway by inhibiting β-catenin. Taken together our data provide a mechanistic basis for the use of curcumin and emodin in the treatment of cervical cancer.
Collapse
Affiliation(s)
- Pooja Chandrakant Thacker
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
| | - Devarajan Karunagaran
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
- * E-mail:
| |
Collapse
|
134
|
Cheng CW, Hsiao JR, Fan CC, Lo YK, Tzen CY, Wu LW, Fang WY, Cheng AJ, Chen CH, Chang IS, Jiang SS, Chang JY, Lee AYL. Loss of GDF10/BMP3b as a prognostic marker collaborates with TGFBR3 to enhance chemotherapy resistance and epithelial-mesenchymal transition in oral squamous cell carcinoma. Mol Carcinog 2015; 55:499-513. [PMID: 25728212 DOI: 10.1002/mc.22297] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 12/18/2014] [Accepted: 01/14/2015] [Indexed: 12/11/2022]
Abstract
Growth differentiation factor-10 (GDF10), commonly referred as BMP3b, is a member of the transforming growth factor-β (TGF-β) superfamily. GDF10/BMP3b has been considered as a tumor suppressor, however, little is known about the molecular mechanism of its roles in tumor suppression in oral cancer. Clinical significance of GDF10 downregulation in oral squamous cell carcinoma (OSCC) was evaluated using three independent cohorts of OSCC patients. The molecular mechanisms of GDF10 in the suppression of cell survival, cell migration/invasion and epithelial-mesenchymal transition (EMT) were investigated by using oral cancer cell lines. The present study shows that GDF10 is downregulated during oral carcinogenesis, and GDF10 expression is also an independent risk factor for overall survival of OSCC patients. Overexpression of GDF10 attenuates cell proliferation, transformation, migration/invasion, and EMT. GDF10-inhibited EMT is mediated by ERK signaling but not by typical TGF-β signaling. In addition, overexpression of GDF10 promotes DNA damage-induced apoptosis and sensitizes the response to all-trans retinoic acid (ATRA) and camptothecin (CPT). Intriguingly, the expression of GDF10 is induced by type III TGF-β receptor (TGFBR3) through TGF-β-SMAD2/3 signaling. Our findings suggest that TGFBR3 is an upstream activator of GDF10 expression and they share the same signaling to inhibit EMT and migration/invasion. These results support that GDF10 acts as a hinge to collaborate with TGFBR3 in the transition of EMT-MET program. Taken together, we illustrated the clinical significance and the molecular mechanisms of tumor-suppressive GDF10 in OSCC.
Collapse
Affiliation(s)
- Chieh-Wen Cheng
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Miaoli, Taiwan
| | - Jenn-Ren Hsiao
- Department of Otolaryngology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chi-Chen Fan
- Department of Physiology, Mackay Memorial Hospital, Taipei, Taiwan.,Department of Medical Laboratory Science and Biotechnology, Yuanpei University, Hsinchu, Taiwan
| | - Yu-Kang Lo
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Miaoli, Taiwan
| | - Chi-Yuan Tzen
- Department of Pathology, Mackay Memorial Hospital, Taipei, Taiwan
| | - Li-Wha Wu
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wei-Yu Fang
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ann-Joy Cheng
- Department of Medical Biotechnology, Chang Gung University, Taoyuan, Taiwan
| | - Chung-Hsing Chen
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Miaoli, Taiwan
| | - I-Shou Chang
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Miaoli, Taiwan
| | - Shih Sheng Jiang
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Miaoli, Taiwan
| | - Jang-Yang Chang
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Miaoli, Taiwan.,Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Alan Yueh-Luen Lee
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Miaoli, Taiwan.,Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| |
Collapse
|
135
|
Thien A, Prentzell MT, Holzwarth B, Kläsener K, Kuper I, Boehlke C, Sonntag AG, Ruf S, Maerz L, Nitschke R, Grellscheid SN, Reth M, Walz G, Baumeister R, Neumann-Haefelin E, Thedieck K. TSC1 activates TGF-β-Smad2/3 signaling in growth arrest and epithelial-to-mesenchymal transition. Dev Cell 2015; 32:617-30. [PMID: 25727005 DOI: 10.1016/j.devcel.2015.01.026] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 12/19/2014] [Accepted: 01/22/2015] [Indexed: 11/27/2022]
Abstract
The tuberous sclerosis proteins TSC1 and TSC2 are key integrators of growth factor signaling. They suppress cell growth and proliferation by acting in a heteromeric complex to inhibit the mammalian target of rapamycin complex 1 (mTORC1). In this study, we identify TSC1 as a component of the transforming growth factor β (TGF-β)-Smad2/3 pathway. Here, TSC1 functions independently of TSC2. TSC1 interacts with the TGF-β receptor complex and Smad2/3 and is required for their association with one another. TSC1 regulates TGF-β-induced Smad2/3 phosphorylation and target gene expression and controls TGF-β-induced growth arrest and epithelial-to-mesenchymal transition (EMT). Hyperactive Akt specifically activates TSC1-dependent cytostatic Smad signaling to induce growth arrest. Thus, TSC1 couples Akt activity to TGF-β-Smad2/3 signaling. This has implications for cancer treatments targeting phosphoinositide 3-kinases and Akt because they may impair tumor-suppressive cytostatic TGF-β signaling by inhibiting Akt- and TSC1-dependent Smad activation.
Collapse
Affiliation(s)
- Antje Thien
- Bioinformatics and Molecular Genetics (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Renal Division, University Hospital Freiburg, 79106 Freiburg, Germany
| | - Mirja Tamara Prentzell
- Bioinformatics and Molecular Genetics (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, the Netherlands
| | - Birgit Holzwarth
- Bioinformatics and Molecular Genetics (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
| | - Kathrin Kläsener
- Molecular Immunology (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Molecular Immunology, Max-Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
| | - Ineke Kuper
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, the Netherlands; Department for Neuroscience, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
| | | | - Annika G Sonntag
- Bioinformatics and Molecular Genetics (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
| | - Stefanie Ruf
- Bioinformatics and Molecular Genetics (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, the Netherlands; BIOSS Centre for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Research Training Group (RTG) 1104, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
| | - Lars Maerz
- Bioinformatics and Molecular Genetics (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
| | - Roland Nitschke
- BIOSS Centre for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Center for Biological Systems Analysis (ZBSA), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
| | | | - Michael Reth
- Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Molecular Immunology (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Molecular Immunology, Max-Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
| | - Gerd Walz
- Renal Division, University Hospital Freiburg, 79106 Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Center for Biological Systems Analysis (ZBSA), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
| | - Ralf Baumeister
- Bioinformatics and Molecular Genetics (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Research Training Group (RTG) 1104, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Center for Biological Systems Analysis (ZBSA), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; ZBMZ Centre for Biochemistry and Molecular Cell Research (Faculty of Medicine), Albert-Ludwigs-University Freiburg, 79106 Freiburg, Germany
| | | | - Kathrin Thedieck
- Bioinformatics and Molecular Genetics (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, the Netherlands; BIOSS Centre for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Department for Neuroscience, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany.
| |
Collapse
|
136
|
Ludwig N, Kim YJ, Mueller SC, Backes C, Werner TV, Galata V, Sartorius E, Bohle RM, Keller A, Meese E. Posttranscriptional deregulation of signaling pathways in meningioma subtypes by differential expression of miRNAs. Neuro Oncol 2015; 17:1250-60. [PMID: 25681310 DOI: 10.1093/neuonc/nov014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 01/16/2015] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Micro (mi)RNAs are key regulators of gene expression and offer themselves as biomarkers for cancer development and progression. Meningioma is one of the most frequent primary intracranial tumors. As of yet, there are limited data on the role of miRNAs in meningioma of different histological subtypes and the affected signaling pathways. METHODS In this study, we compared expression of 1205 miRNAs in different meningioma grades and histological subtypes using microarrays and independently validated deregulation of selected miRNAs with quantitative real-time PCR. Clinical utility of a subset of miRNAs as biomarkers for World Health Organization (WHO) grade II meningioma based on quantitative real-time data was tested. Potential targets of deregulated miRNAs were discovered with an in silico analysis. RESULTS We identified 13 miRNAs deregulated between different subtypes of benign meningiomas, and 52 miRNAs deregulated in anaplastic meningioma compared with benign meningiomas. Known and putative target genes of deregulated miRNAs include genes involved in epithelial-to-mesenchymal transition for benign meningiomas, and Wnt, transforming growth factor-β, and vascular endothelial growth factor signaling for higher-grade meningiomas. Furthermore, a 4-miRNA signature (miR-222, -34a*, -136, and -497) shows promise as a biomarker differentiating WHO grade II from grade I meningiomas with an area under the curve of 0.75. CONCLUSIONS Our data provide novel insights into the contribution of miRNAs to the phenotypic spectrum in benign meningiomas. By deregulating translation of genes belonging to signaling pathways known to be important for meningioma genesis and progression, miRNAs provide a second in line amplification of growth promoting cellular signals. MiRNAs as biomarkers for diagnosis of aggressive meningiomas might prove useful and should be explored further in a prospective manner.
Collapse
Affiliation(s)
- Nicole Ludwig
- Department of Human Genetics Medical School, Saarland University, Homburg/Saar, Germany (N.L., S.C.M., C.B., T.V.W., V.G., E.M.); Institute of Pathology Medical School, Saarland University, Homburg/Saar, Germany (Y.-J.K., E.S., R.M.B.); Chair for Clinical Bioinformatics, Saarland University, University Hospital, Saarbrücken, Germany (S.C.M., C.B., V.G., A.K.)
| | - Yoo-Jin Kim
- Department of Human Genetics Medical School, Saarland University, Homburg/Saar, Germany (N.L., S.C.M., C.B., T.V.W., V.G., E.M.); Institute of Pathology Medical School, Saarland University, Homburg/Saar, Germany (Y.-J.K., E.S., R.M.B.); Chair for Clinical Bioinformatics, Saarland University, University Hospital, Saarbrücken, Germany (S.C.M., C.B., V.G., A.K.)
| | - Sabine C Mueller
- Department of Human Genetics Medical School, Saarland University, Homburg/Saar, Germany (N.L., S.C.M., C.B., T.V.W., V.G., E.M.); Institute of Pathology Medical School, Saarland University, Homburg/Saar, Germany (Y.-J.K., E.S., R.M.B.); Chair for Clinical Bioinformatics, Saarland University, University Hospital, Saarbrücken, Germany (S.C.M., C.B., V.G., A.K.)
| | - Christina Backes
- Department of Human Genetics Medical School, Saarland University, Homburg/Saar, Germany (N.L., S.C.M., C.B., T.V.W., V.G., E.M.); Institute of Pathology Medical School, Saarland University, Homburg/Saar, Germany (Y.-J.K., E.S., R.M.B.); Chair for Clinical Bioinformatics, Saarland University, University Hospital, Saarbrücken, Germany (S.C.M., C.B., V.G., A.K.)
| | - Tamara V Werner
- Department of Human Genetics Medical School, Saarland University, Homburg/Saar, Germany (N.L., S.C.M., C.B., T.V.W., V.G., E.M.); Institute of Pathology Medical School, Saarland University, Homburg/Saar, Germany (Y.-J.K., E.S., R.M.B.); Chair for Clinical Bioinformatics, Saarland University, University Hospital, Saarbrücken, Germany (S.C.M., C.B., V.G., A.K.)
| | - Valentina Galata
- Department of Human Genetics Medical School, Saarland University, Homburg/Saar, Germany (N.L., S.C.M., C.B., T.V.W., V.G., E.M.); Institute of Pathology Medical School, Saarland University, Homburg/Saar, Germany (Y.-J.K., E.S., R.M.B.); Chair for Clinical Bioinformatics, Saarland University, University Hospital, Saarbrücken, Germany (S.C.M., C.B., V.G., A.K.)
| | - Elke Sartorius
- Department of Human Genetics Medical School, Saarland University, Homburg/Saar, Germany (N.L., S.C.M., C.B., T.V.W., V.G., E.M.); Institute of Pathology Medical School, Saarland University, Homburg/Saar, Germany (Y.-J.K., E.S., R.M.B.); Chair for Clinical Bioinformatics, Saarland University, University Hospital, Saarbrücken, Germany (S.C.M., C.B., V.G., A.K.)
| | - Rainer M Bohle
- Department of Human Genetics Medical School, Saarland University, Homburg/Saar, Germany (N.L., S.C.M., C.B., T.V.W., V.G., E.M.); Institute of Pathology Medical School, Saarland University, Homburg/Saar, Germany (Y.-J.K., E.S., R.M.B.); Chair for Clinical Bioinformatics, Saarland University, University Hospital, Saarbrücken, Germany (S.C.M., C.B., V.G., A.K.)
| | - Andreas Keller
- Department of Human Genetics Medical School, Saarland University, Homburg/Saar, Germany (N.L., S.C.M., C.B., T.V.W., V.G., E.M.); Institute of Pathology Medical School, Saarland University, Homburg/Saar, Germany (Y.-J.K., E.S., R.M.B.); Chair for Clinical Bioinformatics, Saarland University, University Hospital, Saarbrücken, Germany (S.C.M., C.B., V.G., A.K.)
| | - Eckart Meese
- Department of Human Genetics Medical School, Saarland University, Homburg/Saar, Germany (N.L., S.C.M., C.B., T.V.W., V.G., E.M.); Institute of Pathology Medical School, Saarland University, Homburg/Saar, Germany (Y.-J.K., E.S., R.M.B.); Chair for Clinical Bioinformatics, Saarland University, University Hospital, Saarbrücken, Germany (S.C.M., C.B., V.G., A.K.)
| |
Collapse
|
137
|
Pettersson F, Del Rincon SV, Emond A, Huor B, Ngan E, Ng J, Dobocan MC, Siegel PM, Miller WH. Genetic and pharmacologic inhibition of eIF4E reduces breast cancer cell migration, invasion, and metastasis. Cancer Res 2015; 75:1102-12. [PMID: 25608710 DOI: 10.1158/0008-5472.can-14-1996] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The translation initiation factor eIF4E is an oncogene that is commonly overexpressed in primary breast cancers and metastases. In this article, we report that a pharmacologic inhibitor of eIF4E function, ribavirin, safely and potently suppresses breast tumor formation. Ribavirin administration blocked the growth of primary breast tumors in several murine models and reduced the development of lung metastases in an invasive model. Mechanistically, eIF4E silencing or blockade reduced the invasiveness and metastatic capability of breast cancer cells in a manner associated with decreased activity of matrix metalloproteinase (MMP)-3 and MMP-9. Furthermore, eIF4E silencing or ribavirin treatment suppressed features of epithelial-to-mesenchymal transition, a process crucial for metastasis. Our findings offer a preclinical rationale to explore broadening the clinical evaluation of ribavirin, currently being tested in patients with eIF4E-overexpressing leukemia, as a strategy to treat solid tumors such as metastatic breast cancer.
Collapse
Affiliation(s)
- Filippa Pettersson
- Lady Davis Institute for Medical Research, Segal Cancer Centre, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Sonia V Del Rincon
- Lady Davis Institute for Medical Research, Segal Cancer Centre, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Audrey Emond
- Lady Davis Institute for Medical Research, Segal Cancer Centre, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Bonnie Huor
- Lady Davis Institute for Medical Research, Segal Cancer Centre, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Elaine Ngan
- Goodman Cancer Centre, McGill University, Montreal, Quebec, Canada
| | - Jonathan Ng
- Lady Davis Institute for Medical Research, Segal Cancer Centre, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Monica C Dobocan
- Lady Davis Institute for Medical Research, Segal Cancer Centre, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Peter M Siegel
- Goodman Cancer Centre, McGill University, Montreal, Quebec, Canada
| | - Wilson H Miller
- Lady Davis Institute for Medical Research, Segal Cancer Centre, Jewish General Hospital, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
138
|
Gonçalves BF, Campos SGPD, Costa CF, Scarano WR, Góes RM, Taboga SR. Key participants of the tumor microenvironment of the prostate: an approach of the structural dynamic of cellular elements and extracellular matrix components during epithelial-stromal transition. Acta Histochem 2015; 117:4-13. [PMID: 25466989 DOI: 10.1016/j.acthis.2014.10.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 10/23/2014] [Accepted: 10/24/2014] [Indexed: 01/03/2023]
Abstract
Cancer is a multistep process that begins with the transformation of normal epithelial cells and continues with tumor growth, stromal invasion and metastasis. The remodeling of the peritumoral environment is decisive for the onset of tumor invasiveness. This event is dependent on epithelial-stromal interactions, degradation of extracellular matrix components and reorganization of fibrillar components. Our research group has studied in a new proposed rodent model the participation of cellular and molecular components in the prostate microenvironment that contributes to cancer progression. Our group adopted the gerbil Meriones unguiculatus as an alternative experimental model for prostate cancer study. This model has presented significant responses to hormonal treatments and to development of spontaneous and induced neoplasias. The data obtained indicate reorganization of type I collagen fibers and reticular fibers, synthesis of new components such as tenascin and proteoglycans, degradation of basement membrane components and elastic fibers and increased expression of metalloproteinases. Fibroblasts that border the region, apparently participate in the stromal reaction. The roles of each of these events, as well as some signaling molecules, participants of neoplastic progression and factors that promote genetic reprogramming during epithelial-stromal transition are also discussed.
Collapse
|
139
|
Wamsley JJ, Kumar M, Allison DF, Clift SH, Holzknecht CM, Szymura SJ, Hoang SA, Xu X, Moskaluk CA, Jones DR, Bekiranov S, Mayo MW. Activin upregulation by NF-κB is required to maintain mesenchymal features of cancer stem-like cells in non-small cell lung cancer. Cancer Res 2014; 75:426-35. [PMID: 25432175 DOI: 10.1158/0008-5472.can-13-2702] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Soluble growth factors and cytokines within the tumor microenvironment aid in the induction of the epithelial-to-mesenchymal transition (EMT). Although EMT promotes the development of cancer-initiating cells (CIC), cellular mechanisms by which cancer cells maintain mesenchymal phenotypes remain poorly understood. Work presented here indicates that induction of EMT stimulates non-small cell lung cancer (NSCLC) to secrete soluble factors that function in an autocrine fashion. Using gene expression profiling of all annotated and predicted secreted gene products, we find that NF-κB activity is required to upregulate INHBA/Activin, a morphogen in the TGFβ superfamily. INHBA is capable of inducing and maintaining mesenchymal phenotypes, including the expression of EMT master-switch regulators and self-renewal factors that sustain CIC phenotypes and promote lung metastasis. Our work demonstrates that INHBA mRNA and protein expression are commonly elevated in primary human NSCLC and provide evidence that INHBA is a critical autocrine factor that maintains mesenchymal properties of CICs to promote metastasis in NSCLC.
Collapse
Affiliation(s)
- J Jacob Wamsley
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia
| | - Manish Kumar
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia
| | - David F Allison
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia
| | - Sheena H Clift
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia
| | - Caitlyn M Holzknecht
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia
| | - Szymon J Szymura
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia
| | - Stephen A Hoang
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia
| | - Xiaojiang Xu
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia
| | | | - David R Jones
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia. Department of Thoracic Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Stefan Bekiranov
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia
| | - Marty W Mayo
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia.
| |
Collapse
|
140
|
Nagura M, Matsumura N, Baba T, Murakami R, Kharma B, Hamanishi J, Yamaguchi K, Abiko K, Koshiyama M, Mandai M, Murata T, Murphy SK, Konishi I. Invasion of uterine cervical squamous cell carcinoma cells is facilitated by locoregional interaction with cancer-associated fibroblasts via activating transforming growth factor-beta. Gynecol Oncol 2014; 136:104-11. [PMID: 25434636 DOI: 10.1016/j.ygyno.2014.11.075] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 11/12/2014] [Accepted: 11/21/2014] [Indexed: 02/01/2023]
Abstract
OBJECTIVE Local invasion is a common pattern of spread in uterine cervical squamous cell carcinoma (CSCC). Although transforming growth factor-beta (TGF-β) facilitates invasion of various types of cancer cells, the role of the TGF-β pathway in CSCC is unclear. In this study, we analyzed the role of TGF-β signaling in the progression of CSCC. METHODS Immunohistochemistry was used to examine the expression of TGF-β pathway molecules in 67 CSCC samples with clinicopathological data. Activation of the TGF-β pathway was investigated following co-culture of CSCC cells and cervical cancer-associated fibroblasts (CCAFs). RESULTS Clinicopathological analysis of CSCC samples revealed that prominent expression of TGF-β receptor-2 was more frequent in CSCC with lymphovascular space invasion (LVSI) than without LVSI (p < 0.01). Lymph node metastasis was more frequent in cases in which phosphorylated SMAD3 (pSMAD3) was localized exclusively at the boundary of tumor clusters (n = 9, p < 0.05). Recombinant TGF-β1 increased pSMAD3 expression and enhanced cellular invasion (p < 0.005) in CSCC cells, which was attenuated by an inhibitor of the TGF-β receptor (p < 0.005). Enhanced pSMAD3 expression and invasion was also observed when conditioned media from CSCC cells co-cultured with CCAFs were administered. Luciferase assays showed that this medium contained a large amount of active TGF-β. Along with TGF-β activation, thrombospondin-1 was upregulated in both CSCC cells and CCAFs, while thrombospondin-1 silencing in either CSCC cells or CCAFs repressed the activity of TGF-β. Thrombospondin-1 was prominently expressed in cases with pSMAD3 boundary staining (p < 0.05). CONCLUSIONS These results suggest that interaction between CSCC cells and surrounding CCAFs activates TGF-β via thrombospondin-1 secretion to facilitate CSCC invasion.
Collapse
Affiliation(s)
- Michikazu Nagura
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Japan
| | - Noriomi Matsumura
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Japan
| | - Tsukasa Baba
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Japan.
| | - Ryusuke Murakami
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Japan
| | - Budiman Kharma
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Japan
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Japan
| | - Ken Yamaguchi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Japan
| | - Kaoru Abiko
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Japan
| | - Masafumi Koshiyama
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Japan
| | - Masaki Mandai
- Department of Obstetrics and Gynecology, Kinki University School of Medicine, Japan
| | - Takuya Murata
- Department of Obstetrics and Gynecology, Kawasaki Medical University, Japan
| | - Susan K Murphy
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, USA
| | - Ikuo Konishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Japan
| |
Collapse
|
141
|
Yue Z, Feng W, Xiangke L, Liuxing W, Qingxia F, Jianbo G. WAVE3 promotes epithelial-mesenchymal transition of gastric cancer through upregulation of Snail. Cancer Gene Ther 2014; 21:499-506. [PMID: 25378074 DOI: 10.1038/cgt.2014.52] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 09/14/2014] [Accepted: 09/15/2014] [Indexed: 12/28/2022]
Abstract
WAVE3, an actin cytoskeleton remodeling protein overexpressed in many kinds of cancers, has been associated with a lot of metastatic diseases. However, the role and mechanisms of the high expression of WAVE3 in human gastric cancer has not been fully elucidated. Here we demonstrated that WAVE3 was expressed in all six kinds of gastric-cancer cell lines: BGC-823, SGC-7901, AGS, MGC803, MKN28 and MKN45. Furthermore, a correlation was found between aggressiveness of these cell lines and expression of WAVE3. Next, we investigated the role of WAVE3 in SGC-7901 cells and found that upregulating WAVE3 could promote the migration, invasion and proliferation of SGC-7901 cells in vitro. It has been reported that WAVE3 could induce cancer invasion and metastasis by participating epithelial-mesenchymal transition (EMT). However, the mechanisms are not entirely clear. In this study we showed that elevated WAVE3 levels could induce EMT in SGC-7901 cells by dampening the expression of E-cadherin while increasing the expression of vimentin. Elevated WAVE3 levels could also improve the expression of transcription factor Snail. In addition, downregulating Snail could particularly reduce EMT and the metastasis, invasion and proliferation activity in SGC-7901 cells elevated by overexpression of WAVE3. Taken together, we demonstrated that WAVE3 promoted gastric-cancer-cells migration and invasion by taking part in EMT via upregulation of Snail. WAVE3 could be a useful target for gastric-cancer prevention and therapy.
Collapse
Affiliation(s)
- Z Yue
- Department of Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PRC
| | - W Feng
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PRC
| | - L Xiangke
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PRC
| | - W Liuxing
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PRC
| | - F Qingxia
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PRC
| | - G Jianbo
- Department of Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PRC
| |
Collapse
|
142
|
Neuzillet C, Tijeras-Raballand A, Cohen R, Cros J, Faivre S, Raymond E, de Gramont A. Targeting the TGFβ pathway for cancer therapy. Pharmacol Ther 2014; 147:22-31. [PMID: 25444759 DOI: 10.1016/j.pharmthera.2014.11.001] [Citation(s) in RCA: 471] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 09/25/2014] [Indexed: 02/07/2023]
Abstract
The TGFβ signaling pathway has pleiotropic functions regulating cell growth, differentiation, apoptosis, motility and invasion, extracellular matrix production, angiogenesis, and immune response. TGFβ signaling deregulation is frequent in tumors and has crucial roles in tumor initiation, development and metastasis. TGFβ signaling inhibition is an emerging strategy for cancer therapy. The role of the TGFβ pathway as a tumor-promoter or suppressor at the cancer cell level is still a matter of debate, due to its differential effects at the early and late stages of carcinogenesis. In contrast, at the microenvironment level, the TGFβ pathway contributes to generate a favorable microenvironment for tumor growth and metastasis throughout all the steps of carcinogenesis. Then, targeting the TGFβ pathway in cancer may be considered primarily as a microenvironment-targeted strategy. In this review, we focus on the TGFβ pathway as a target for cancer therapy. In the first part, we provide a comprehensive overview of the roles played by this pathway and its deregulation in cancer, at the cancer cell and microenvironment levels. We go on to describe the preclinical and clinical results of pharmacological strategies to target the TGFβ pathway, with a highlight on the effects on tumor microenvironment. We then explore the perspectives to optimize TGFβ inhibition therapy in different tumor settings.
Collapse
Affiliation(s)
- Cindy Neuzillet
- INSERM U728 & U773 and Department of Medical Oncology, Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), 100 boulevard du Général Leclerc, 92110 Clichy, France
| | | | - Romain Cohen
- AAREC Filia Research, Translational Department, 1 place Paul Verlaine, 92100 Boulogne-Billancourt, France
| | - Jérôme Cros
- Department of Pathology, Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), 100 boulevard du Général Leclerc, 92110 Clichy, France
| | - Sandrine Faivre
- INSERM U728 & U773 and Department of Medical Oncology, Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), 100 boulevard du Général Leclerc, 92110 Clichy, France
| | - Eric Raymond
- New Drug Evaluation Laboratory, Centre of Experimental Therapeutics and Medical Oncology, Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV) Lausanne, Switzerland
| | - Armand de Gramont
- New Drug Evaluation Laboratory, Centre of Experimental Therapeutics and Medical Oncology, Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV) Lausanne, Switzerland.
| |
Collapse
|
143
|
Loss of BRMS1 promotes a mesenchymal phenotype through NF-κB-dependent regulation of Twist1. Mol Cell Biol 2014; 35:303-17. [PMID: 25368381 DOI: 10.1128/mcb.00869-14] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Breast cancer metastasis suppressor 1 (BRMS1) is downregulated in non-small cell lung cancer (NSCLC), and its reduction correlates with disease progression. Herein, we investigate the mechanisms through which loss of the BRMS1 gene contributes to epithelial-to-mesenchymal transition (EMT). Using a short hairpin RNA (shRNA) system, we show that loss of BRMS1 promotes basal and transforming growth factor beta-induced EMT in NSCLC cells. NSCLC cells expressing BRMS1 shRNAs (BRMS1 knockdown [BRMS1(KD)]) display mesenchymal characteristics, including enhanced cell migration and differential regulation of the EMT markers. Mesenchymal phenotypes observed in BRMS1(KD) cells are dependent on RelA/p65, the transcriptionally active subunit of nuclear factor kappa B (NF-κB). In addition, chromatin immunoprecipitation analysis demonstrates that loss of BRMS1 increases Twist1 promoter occupancy of RelA/p65 K310-a key histone modification associated with increased transcription. Knockdown of Twist1 results in reversal of BRMS1(KD)-mediated EMT phenotypic changes. Moreover, in our animal model, BRMS1(KD)/Twist1(KD) double knockdown cells were less efficient in establishing lung tumors than BRMS1(KD) cells. Collectively, this study demonstrates that loss of BRMS1 promotes malignant phenotypes that are dependent on NF-κB-dependent regulation of Twist1. These observations offer fresh insight into the mechanisms through which BRMS1 regulates the development of metastases in NSCLC.
Collapse
|
144
|
Brown WS, Wendt MK. Integrin-mediated resistance to epidermal growth factor receptor-targeted therapy: an inflammatory situation. Breast Cancer Res 2014; 16:448. [PMID: 25255930 PMCID: PMC4728772 DOI: 10.1186/s13058-014-0448-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Targeting the function of epidermal growth factor receptor (EGFR) has failed as an effective clinical option for breast cancer. Understanding the drivers of inherent resistance has been a challenge. One possible mechanism is the acquisition of stem-like properties through the process of epithelial-mesenchymal transition. A recent study by Seguin and colleagues adds to our understanding of this process by demonstrating a functional role for unligated αvβ3 integrin in mediating a stem-like phenotype and facilitating resistance to EGFR-targeted therapy via enhanced downstream coupling to a KRAS:RalB:NF-κB pathway. Importantly, the identified mechanism may reveal a possible strategy for sensitizing breast cancer cells to EGFR-targeted therapies.
Collapse
|
145
|
Ponnusamy MP, Seshacharyulu P, Lakshmanan I, Vaz AP, Chugh S, Batra SK. Emerging role of mucins in epithelial to mesenchymal transition. Curr Cancer Drug Targets 2014; 13:945-56. [PMID: 24168188 DOI: 10.2174/15680096113136660100] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Revised: 07/21/2013] [Accepted: 09/07/2013] [Indexed: 12/13/2022]
Abstract
Epithelial to mesenchymal transition (EMT) is an important and complex phenomenon that determines the aggressiveness of cancer cells. The morphological transformation of cancerous cells is accompanied by various cellular processes such as alterations in cell-cell adhesion, cell matrix degradation, down regulation of epithelial marker Ecadherin and upregulation of mesenchymal markers N-cadherin and Vimentin. Besides these markers several other important tumor antigens/mucins are also involved in the EMT process. Mainly high molecular weight glycoproteins such as mucin molecules (MUC1, MUC4 and MUC16) play a major role in the cellular transformation and signaling alteration in EMT process. In addition to these factors, EMT may be an essential process triggering the emergence or expansion of the CSC population, which slowly results in the initiation of tumor at metastatic sites. Furthermore, mucins have been demonstrated to be involved in the EMT process and also in the enrichment of cancer stem cell population. Mucin mediated EMT is very complex since the key components of tumor microenvironment are also regulating mucin molecules. In this review, we have discussed all the aforementioned factors and their mechanistic involvement for EMT process.
Collapse
Affiliation(s)
| | | | | | | | | | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, 68198-5870, USA.
| |
Collapse
|
146
|
Ru NY, Wu J, Chen ZN, Bian H. HAb18G/CD147 is involved in TGF-β-induced epithelial-mesenchymal transition and hepatocellular carcinoma invasion. Cell Biol Int 2014; 39:44-51. [PMID: 25044444 DOI: 10.1002/cbin.10341] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 06/09/2014] [Indexed: 01/07/2023]
Abstract
Epithelial-mesenchymal transition (EMT) induced by the transforming growth factor beta (TGF-β) is involved in hepatocarcinogenesis and hepatocellular carcinoma (HCC) metastasis. HAb18G/CD147, a member of the immunoglobulin family, plays an important role in tumor invasion and metastasis. HAb18G/CD147 promotes EMT of hepatocytes through TGF-β signaling and is transcriptionally regulated by Slug. We investigated the role of HAb18G/CD147 in TGF-β-induced EMT in HCC invasion. Two human HCC cell lines, SMMC-7721 and HepG2, were used to determine the role of HAb18G/CD147 in EMT. Upregulation of HAb18G/CD147 induced by the high doses of TGF-β1 in SMMC-7721 (5 ng/mL) and HepG2 cells (10 ng/mL) (P < 0.05). CD147 upregulation was coupled with upregulation of Snail1 and Slug. CD147 knockout significantly decreased the expression of N-cadherin and vimentin, and colony formation ability of SMMC-7721 cells. TGF-β1 enhanced the migration capacity of SMMC-7721 cells, which was markedly attenuated by CD147 knockdown. Thus, HAb18G/CD147 is involved in TGF-β-induced EMT and HCC invasion.
Collapse
Affiliation(s)
- Ning-Yu Ru
- Cell Engineering Research Center and Department of Cell Biology, State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, China; Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, 710032, China
| | | | | | | |
Collapse
|
147
|
Miura Y, Hagiwara N, Radisky DC, Hirai Y. CCAAT/enhancer binding protein beta (C/EBPβ) isoform balance as a regulator of epithelial-mesenchymal transition in mouse mammary epithelial cells. Exp Cell Res 2014; 327:146-55. [PMID: 24881817 DOI: 10.1016/j.yexcr.2014.05.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 05/03/2014] [Accepted: 05/21/2014] [Indexed: 12/22/2022]
Abstract
Activation of the epithelial-mesenchymal transition (EMT) program promotes cell invasion and metastasis, and is reversed through mesenchymal-epithelial transition (MET) after formation of distant metastases. Here, we show that an imbalance of gene products encoded by the transcriptional factor C/EBPβ, LAP (liver-enriched activating protein) and LIP (liver-enriched inhibitory protein), can regulate both EMT- and MET-like phenotypic changes in mouse mammary epithelial cells. By using tetracycline repressive LIP expression constructs, we found that SCp2 cells, a clonal epithelial line of COMMA1-D cells, expressed EMT markers, lost the ability to undergo alveolar-like morphogenesis in 3D Matrigel, and acquired properties of benign adenoma cells. Conversely, we found that inducible expression of LAP in SCg6 cells, a clonal fibroblastic line of COMMA1-D cells, began to express epithelial keratins with suppression of proliferation. The overexpression of the C/EBPβ gene products in these COMMA1-D derivatives was suppressed by long-term cultivation on tissue culture plastic, but gene expression was maintained in cells grown on Matrigel or exposed to proteasome inhibitors. Thus, imbalances of C/EBPβ gene products in mouse mammary epithelial cells, which are affected by contact with basement membrane, are defined as a potential regulator of metastatic potential.
Collapse
Affiliation(s)
- Yuka Miura
- Department of Bioscience, Graduate School of Science and Technology, Kwansei Gakuin University, Hyogo, 2-1 Gakuen, Sanda 669-1337 Japan
| | - Natsumi Hagiwara
- Department of Bioscience, Graduate School of Science and Technology, Kwansei Gakuin University, Hyogo, 2-1 Gakuen, Sanda 669-1337 Japan
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32225 USA
| | - Yohei Hirai
- Department of Bioscience, Graduate School of Science and Technology, Kwansei Gakuin University, Hyogo, 2-1 Gakuen, Sanda 669-1337 Japan.
| |
Collapse
|
148
|
Jonckheere N, Skrypek N, Van Seuningen I. Mucins and tumor resistance to chemotherapeutic drugs. Biochim Biophys Acta Rev Cancer 2014; 1846:142-51. [PMID: 24785432 DOI: 10.1016/j.bbcan.2014.04.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 04/22/2014] [Accepted: 04/23/2014] [Indexed: 12/30/2022]
Abstract
Epithelial cancer patients not considered eligible for surgical resection frequently benefit from chemotherapy. Chemotherapy is the treatment of cancer with one or combination of cytotoxic or cytostatic drugs. Recent advances in chemotherapy allowed a great number of cancer patients to receive treatment with significant results. Unfortunately, resistance to chemotherapeutic drug treatment is a major challenge for clinicians in the majority of epithelial cancers because it is responsible for the inefficiency of therapies. Mucins belong to a heterogeneous group of large O-glycoproteins that can be either secreted or membrane-bound. Implications of mucins have been described in relation to cancer cell behavior and cell signaling pathways associated with epithelial tumorigenesis. Because of the frequent alteration of the pattern of mucin expression in cancers as well as their structural and functional characteristics, mucins are thought to also be involved in response to therapies. In this report, we review the roles of mucins in chemoresistance and the associated underlying molecular mechanisms (physical barrier, resistance to apoptosis, drug metabolism, cell stemness, epithelial-mesenchymal transition) and discuss the therapeutic tools/strategies and/or prognosis biomarkers for personalized chemotherapy that could be proposed from these studies.
Collapse
Affiliation(s)
- Nicolas Jonckheere
- Inserm, UMR837, Jean Pierre Aubert Research Center, Team #5 "Mucins, Epithelial Differentiation and Carcinogenesis", rue Polonovski, 59045 Lille Cedex, France; Université Lille Nord de France, Lille, France; Centre Hospitalier Régional et Universitaire de Lille, Place de Verdun, 59037 Lille Cedex, France.
| | - Nicolas Skrypek
- Inserm, UMR837, Jean Pierre Aubert Research Center, Team #5 "Mucins, Epithelial Differentiation and Carcinogenesis", rue Polonovski, 59045 Lille Cedex, France; Université Lille Nord de France, Lille, France; Centre Hospitalier Régional et Universitaire de Lille, Place de Verdun, 59037 Lille Cedex, France
| | - Isabelle Van Seuningen
- Inserm, UMR837, Jean Pierre Aubert Research Center, Team #5 "Mucins, Epithelial Differentiation and Carcinogenesis", rue Polonovski, 59045 Lille Cedex, France; Université Lille Nord de France, Lille, France; Centre Hospitalier Régional et Universitaire de Lille, Place de Verdun, 59037 Lille Cedex, France
| |
Collapse
|
149
|
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
|
150
|
HiJAK'd Signaling; the STAT3 Paradox in Senescence and Cancer Progression. Cancers (Basel) 2014; 6:741-55. [PMID: 24675570 PMCID: PMC4074801 DOI: 10.3390/cancers6020741] [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: 02/08/2014] [Revised: 03/08/2014] [Accepted: 03/11/2014] [Indexed: 12/11/2022] Open
Abstract
Clinical and epidemiological data have associated chronic inflammation with cancer progression. Most tumors show evidence of infiltrating immune and inflammatory cells, and chronic inflammatory disorders are known to increase the overall risk of cancer development. While immune cells are often observed in early hyperplastic lesions in vivo, there remains debate over whether these immune cells and the cytokines they produce in the developing hyperplastic microenvironment act to inhibit or facilitate tumor development. The interleukin-6 (IL-6) family of cytokines, which includes IL-6 and oncostatin M (OSM), among others (LIF, CT-1, CNTF, and CLC), are secreted by immune cells, stromal cells, and epithelial cells, and regulate diverse biological processes. Each of the IL-6 family cytokines signals through a distinct receptor complex, yet each receptor complex uses a shared gp130 subunit, which is critical for signal transduction following cytokine binding. Activation of gp130 results in the activation of Signal Transducer and Activator of Transcription 3 (STAT3), and the Mitogen-Activated Protein Kinase (MAPK) and Phosphatidylinositol 3-Kinase (PI3K) signaling cascades. Tumor suppressive signaling can often be observed in normal cells following prolonged STAT3 activation. However, there is mounting evidence that the IL-6 family cytokines can contribute to later stages of tumor progression in many ways. Here we will review how the microenvironmental IL-6 family cytokine OSM influences each stage of the transformation process. We discuss the intrinsic adaptations a developing cancer cell must make in order to tolerate and circumvent OSM-mediated growth suppression, as well as the OSM effectors that are hijacked during tumor expansion and metastasis. We propose that combining current therapies with new ones that suppress the signals generated from the tumor microenvironment will significantly impact an oncologist’s ability to treat cancer.
Collapse
|