1
|
Wei C, Mao A, Liu Y, Zhang Q, Pan G, Liu W, Liu J. Proteomics Analysis of Polyphyllin D-Treated Triple-Negative Breast Cancer Cells Reveal the Anticancer Mechanisms of Polyphyllin D. Appl Biochem Biotechnol 2024; 196:3148-3161. [PMID: 37624509 PMCID: PMC11166742 DOI: 10.1007/s12010-023-04679-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2023] [Indexed: 08/26/2023]
Abstract
Polyphyllin D (PD), one of the important steroid saponins in traditional medicinal herb Paris polyphylla, has been demonstrated to have anticancer activity both in vitro and in vivo. However, the mechanisms through which PD exerts its anticancer effects in triple-negative breast cancer (TNBC) remain unclear. Our study was presented to evaluate the anticancer effect and the potential mechanisms of PD in two TNBC cell lines, BT-549 and MDA-MB-231. Through comprehensively comparing the liquid chromatography-tandem mass spectrometry (LC-MS/MS) data of PD-treated and untreated BT-549 and MDA-MB-231 cells, we found that PD could induce apoptosis of TNBC cells by activating oxidative phosphorylation pathway in BT-549 cells, as well as inhibiting spliceosome function alteration in MDA-MB-231 cells. These results suggested that the mechanisms underlying the pro-apoptotic effect of PD on TNBC may be cell type-specificity-dependent. Moreover, we found that nodal modulator 2/3 (NOMO2/3) were downregulated both in PD-treated BT-549 and MDA-MB-231 cells, suggesting that NOMO2/3 may be the potential target of PD. Verification experiments revealed that PD deceased NOMO2/3 expression at protein level, rather than mRNA level. Whether NOMO2/3 are the upstream modulators of oxidative phosphorylation pathway and spliceosome needs further validation. In conclusion, a comprehensive proteomics study was performed on PD-treated or untreated TNBC cells, revealing the anticancer mechanisms of PD.
Collapse
Affiliation(s)
- Chuanchao Wei
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, 201100, China
| | - Anwei Mao
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, 201100, China
| | - Yongzhi Liu
- Department of General Surgery, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Zhejiang, China
| | - Qing Zhang
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, 201100, China
| | - Gaofeng Pan
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, 201100, China
| | - Weiyan Liu
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, 201100, China
| | - Jiazhe Liu
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, 201100, China.
| |
Collapse
|
2
|
Untiveros G, Dezi L, Gillette M, Sidor J, Strizzi L. Normal Skin Cells Increase Aggressiveness of Cutaneous Melanoma by Promoting Epithelial-to-Mesenchymal Transition via Nodal and Wnt Activity. Int J Mol Sci 2021; 22:11719. [PMID: 34769150 PMCID: PMC8583838 DOI: 10.3390/ijms222111719] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/20/2021] [Accepted: 10/26/2021] [Indexed: 01/17/2023] Open
Abstract
Melanoma is a lethal form of skin cancer triggered by genetic and environmental factors. Excision of early-stage, poorly aggressive melanoma often leads to a successful outcome; however, left undiagnosed these lesions can progress to metastatic disease. This research investigates whether the exposure of poorly aggressive melanoma to certain normal skin cells can explain how non-metastatic melanoma becomes more aggressive while still confined to the skin. To this end, we used a serial co-culture approach to sequentially expose cells from two different, poorly aggressive human melanoma cell lines against normal cells of the skin beginning with normal melanocytes, then epidermal keratinocytes, and finally dermal fibroblasts. Protein extraction of melanoma cells occurred at each step of the co-culture sequence for western blot (WB) analysis. In addition, morphological and functional changes were assessed to detect differences between the serially co-cultured melanoma cells and non-co-cultured cells. Results show that the co-cultured melanoma cells assumed a more mesenchymal morphology and displayed a significant increase in proliferation and invasiveness compared to control or reference cells. WB analysis of protein from the co-cultured melanoma cells showed increased expression of Snail and decreased levels of E-cadherin suggesting that epithelial-to-mesenchymal transition (EMT) is occurring in these co-cultured cells. Additional WB analysis showed increased levels of Nodal protein and signaling and signs of increased Wnt activity in the co-cultured melanoma cells compared to reference cells. These data suggest that interaction between poorly aggressive melanoma cells with normal cells of the skin may regulate the transition from localized, poorly aggressive melanoma to invasive, metastatic disease via Nodal and/or Wnt induced EMT.
Collapse
Affiliation(s)
- Gustavo Untiveros
- Department of Pathology, College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA;
| | - Lindsay Dezi
- Department of Biomedical Sciences, College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA;
| | - Megan Gillette
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA; (M.G.); (J.S.)
| | - Julia Sidor
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA; (M.G.); (J.S.)
| | - Luigi Strizzi
- Department of Pathology, College of Graduate Studies, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA
| |
Collapse
|
3
|
Boustan A, Mosaffa F, Jahangiri R, Heidarian-Miri H, Dahmardeh-Ghalehno A, Jamialahmadi K. Role of SALL4 and Nodal in the prognosis and tamoxifen resistance of estrogen receptor-positive breast cancer. MOLECULAR BIOLOGY RESEARCH COMMUNICATIONS 2021; 10:109-119. [PMID: 34476264 PMCID: PMC8340312 DOI: 10.22099/mbrc.2021.39878.1597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Despite the discovery of a number of different mechanisms underlying tamoxifen resistance, its molecular pathway is not completely clear. The upregulation of SALL4 and Nodal has been reported in breast cancer. Nevertheless, their role in tamoxifen resistance has not been investigated. In the present study, we compared Nodal and SALL4 expression in 72 tamoxifen sensitive (TAMS) and tamoxifen-resistant (TAMR) patients. Afterward, the correlation of expression data with clinicopathological features and survival of patients was studied. Results showed that both SALL4 and Nodal were significantly upregulated in TAMR compared to TAMS patients. Besides, there was a positive association between Nodal and SALL4 expression. Furthermore, we evaluated their correlation with the expression of Oct4, Nanog and Sox2 stemness markers. The results demonstrated that in most tissue samples there was a positive correlation between Nodal and SALL4 expression with these stemness markers. Besides, the overexpression of SALL4 and Nodal significantly correlated with the N stage. Moreover, the overexpression of SALL4 was associated with extracapsular invasion and lymphatic invasion. High level expressions of SALL4 and Nodal had a significant association with worse disease-free survival (DFS) rates. In addition, increased level of Nodal expression provides a superior predictor factor for DFS. The multivariate Cox regression analysis also revealed that for DFS, perineural invasion (PNI) was independently an unfavorable prognostic value. These findings suggest that the high expression of SALL4 and Nodal could contribute to tamoxifen resistance and worse survival rates in tamoxifen-treated ER+ breast cancer patients.
Collapse
Affiliation(s)
- Arad Boustan
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Mosaffa
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Rosa Jahangiri
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Heidarian-Miri
- Department of Epidemiology, School of Health, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Asefeh Dahmardeh-Ghalehno
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khadijeh Jamialahmadi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
4
|
Catara G, Spano D. Combinatorial Strategies to Target Molecular and Signaling Pathways to Disarm Cancer Stem Cells. Front Oncol 2021; 11:689131. [PMID: 34381714 PMCID: PMC8352560 DOI: 10.3389/fonc.2021.689131] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/01/2021] [Indexed: 12/14/2022] Open
Abstract
Cancer is an urgent public health issue with a very huge number of cases all over the world expected to increase by 2040. Despite improved diagnosis and therapeutic protocols, it remains the main leading cause of death in the world. Cancer stem cells (CSCs) constitute a tumor subpopulation defined by ability to self-renewal and to generate the heterogeneous and differentiated cell lineages that form the tumor bulk. These cells represent a major concern in cancer treatment due to resistance to conventional protocols of radiotherapy, chemotherapy and molecular targeted therapy. In fact, although partial or complete tumor regression can be achieved in patients, these responses are often followed by cancer relapse due to the expansion of CSCs population. The aberrant activation of developmental and oncogenic signaling pathways plays a relevant role in promoting CSCs therapy resistance. Although several targeted approaches relying on monotherapy have been developed to affect these pathways, they have shown limited efficacy. Therefore, an urgent need to design alternative combinatorial strategies to replace conventional regimens exists. This review summarizes the preclinical studies which provide a proof of concept of therapeutic efficacy of combinatorial approaches targeting the CSCs.
Collapse
Affiliation(s)
- Giuliana Catara
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Daniela Spano
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| |
Collapse
|
5
|
Wong JYK, Mukherjee R, Miao J, Bilyk O, Triana V, Miskolzie M, Henninot A, Dwyer JJ, Kharchenko S, Iampolska A, Volochnyuk DM, Lin YS, Postovit LM, Derda R. Genetically-encoded discovery of proteolytically stable bicyclic inhibitors for morphogen NODAL. Chem Sci 2021; 12:9694-9703. [PMID: 34349940 PMCID: PMC8294009 DOI: 10.1039/d1sc01916c] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 05/25/2021] [Indexed: 12/19/2022] Open
Abstract
In this manuscript, we developed a two-fold symmetric linchpin (TSL) that converts readily available phage-displayed peptides libraries made of 20 common amino acids to genetically-encoded libraries of bicyclic peptides displayed on phage. TSL combines an aldehyde-reactive group and two thiol-reactive groups; it bridges two side chains of cysteine [C] with an N-terminal aldehyde group derived from the N-terminal serine [S], yielding a novel bicyclic topology that lacks a free N-terminus. Phage display libraries of SX1CX2X3X4X5X6X7C sequences, where X is any amino acid but Cys, were converted to a library of bicyclic TSL-[S]X1[C]X2X3X4X5X6X7[C] peptides in 45 ± 15% yield. Using this library and protein morphogen NODAL as a target, we discovered bicyclic macrocycles that specifically antagonize NODAL-induced signaling in cancer cells. At a 10 μM concentration, two discovered bicyclic peptides completely suppressed NODAL-induced phosphorylation of SMAD2 in P19 embryonic carcinoma cells. The TSL-[S]Y[C]KRAHKN[C] bicycle inhibited NODAL-induced proliferation of NODAL-TYK-nu ovarian carcinoma cells with apparent IC50 of 1 μM. The same bicycle at 10 μM concentration did not affect the growth of the control TYK-nu cells. TSL-bicycles remained stable over the course of the 72 hour-long assays in a serum-rich cell-culture medium. We further observed general stability in mouse serum and in a mixture of proteases (Pronase™) for 21 diverse bicyclic macrocycles of different ring sizes, amino acid sequences, and cross-linker geometries. TSL-constrained peptides to expand the previously reported repertoire of phage-displayed bicyclic architectures formed by cross-linking Cys side chains. We anticipate that it will aid the discovery of proteolytically stable bicyclic inhibitors for a variety of protein targets.
Collapse
Affiliation(s)
- Jeffrey Y-K Wong
- Department of Chemistry, University of Alberta Edmonton AB T6G 2G2 Canada
| | - Raja Mukherjee
- Department of Chemistry, University of Alberta Edmonton AB T6G 2G2 Canada
| | - Jiayuan Miao
- Department of Chemistry, Tufts University Medford MA 02155 USA
| | - Olena Bilyk
- Department of Experimental Oncology, University of Alberta Edmonton AB T6G 2G2 Canada
| | - Vivian Triana
- Department of Chemistry, University of Alberta Edmonton AB T6G 2G2 Canada
| | - Mark Miskolzie
- Department of Chemistry, University of Alberta Edmonton AB T6G 2G2 Canada
| | | | - John J Dwyer
- Ferring Research Institute San Diego California 92121 USA
| | | | - Anna Iampolska
- Enamine Ltd. Chervonotkatska Street 78 Kyiv 02094 Ukraine
| | | | - Yu-Shan Lin
- Department of Chemistry, Tufts University Medford MA 02155 USA
| | - Lynne-Marie Postovit
- Department of Experimental Oncology, University of Alberta Edmonton AB T6G 2G2 Canada
| | - Ratmir Derda
- Department of Chemistry, University of Alberta Edmonton AB T6G 2G2 Canada
| |
Collapse
|
6
|
Dieters-Castator D, Dantonio PM, Piaseczny M, Zhang G, Liu J, Kuljanin M, Sherman S, Jewer M, Quesnel K, Kang EY, Köbel M, Siegers GM, Leask A, Hess D, Lajoie G, Postovit LM. Embryonic protein NODAL regulates the breast tumor microenvironment by reprogramming cancer-derived secretomes. Neoplasia 2021; 23:375-390. [PMID: 33784590 PMCID: PMC8041663 DOI: 10.1016/j.neo.2021.02.004] [Citation(s) in RCA: 5] [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: 08/21/2020] [Revised: 02/23/2021] [Accepted: 02/26/2021] [Indexed: 02/07/2023]
Abstract
The tumor microenvironment (TME) is an important mediator of breast cancer progression. Cancer-associated fibroblasts constitute a major component of the TME and may originate from tissue-associated fibroblasts or infiltrating mesenchymal stromal cells (MSCs). The mechanisms by which cancer cells activate fibroblasts and recruit MSCs to the TME are largely unknown, but likely include deposition of a pro-tumorigenic secretome. The secreted embryonic protein NODAL is clinically associated with breast cancer stage and promotes tumor growth, metastasis, and vascularization. Herein, we show that NODAL expression correlates with the presence of activated fibroblasts in human triple-negative breast cancers and that it directly induces Cancer-associated fibroblasts phenotypes. We further show that NODAL reprograms cancer cell secretomes by simultaneously altering levels of chemokines (e.g., CXCL1), cytokines (e.g., IL-6) and growth factors (e.g., PDGFRA), leading to alterations in MSC chemotaxis. We therefore demonstrate a hitherto unappreciated mechanism underlying the dynamic regulation of the TME.
Collapse
Affiliation(s)
| | - Paola M Dantonio
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Matt Piaseczny
- Department of Anatomy and Cell Biology, Western University, London, ON, Canada
| | - Guihua Zhang
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Jiahui Liu
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Miljan Kuljanin
- Robarts Research Institute, London, ON, Canada; Department of Biochemistry, Western University, London, ON, Canada
| | - Stephen Sherman
- Robarts Research Institute, London, ON, Canada; Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Michael Jewer
- Department of Anatomy and Cell Biology, Western University, London, ON, Canada; Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Katherine Quesnel
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Eun Young Kang
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
| | - Martin Köbel
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
| | | | - Andrew Leask
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - David Hess
- Robarts Research Institute, London, ON, Canada; Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Gilles Lajoie
- Department of Biochemistry, Western University, London, ON, Canada
| | - Lynne-Marie Postovit
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada; Department of Oncology, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
7
|
Wang X, Liu S, Cao H, Li X, Rong Y, Liu G, Du H, Shen H. Increasing Embryonic Morphogen Nodal Expression Suggests Malignant Transformation in Colorectal Lesions and as a Potential Marker for CMS4 Subtype of Colorectal Cancer. Pathol Oncol Res 2021; 27:587029. [PMID: 34257534 PMCID: PMC8262187 DOI: 10.3389/pore.2021.587029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 01/22/2021] [Indexed: 12/02/2022]
Abstract
Nodal, an embryonic morphogen in TGF-β family, is related with tumorigenicity and progression in various tumors including colorectal cancer (CRC). However, the difference of Nodal expression between CRC and colorectal polyps has not yet been investigated. Besides, whether Nodal can be used as a marker for consensus molecular subtype classification-4 (CMS4) of CRC is also worth studying. We analyzed Nodal expression in patients of CRC (161), high-grade intraepithelial neoplasia (HGIN, 28) and five types of colorectal polyps (116). The Nodal expression difference among groups and the association between Nodal expression and clinicopathological features were analyzed. Two categories logistic regression model was used to predict the odds ratio (OR) of risk factors for high tumor-stroma percentage (TSP), and ROC curve was used to assess the diagnostic value of Nodal in predicting high TSP in CRC. We found that Nodal expression was significantly elevated in CRC and HGIN (p < 0.0001). The increased expression of Nodal was related with high TSP, mismatch repair-proficient (pMMR) status, lymph node metastasis and advanced AJCC stage (p < 0.05). Besides, Nodal expression was the only risk factor for high TSP (OR = 6.94; p < 0.001), and ROC curve demonstrated that Nodal expression was able to efficiently distinguish high and low TSP. In conclusion, different expression of Nodal between CRC/HGIN and benign lesions is suggestive of a promoting role for Nodal in colorectal tumor progression. Besides, Nodal might also be used as a potential marker for CMS4 subtype of CRC.
Collapse
Affiliation(s)
- Xiaopai Wang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Department of Pathology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Shousheng Liu
- Department of General Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Huijiao Cao
- Department of General Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiubo Li
- Department of Pathology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yuming Rong
- Department of General Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Guorong Liu
- Department of Pathology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Hong Du
- Department of Pathology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Hong Shen
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
8
|
Zhu X, Xue D, Liu J, Dong F, Li Y, Liu Y. Nodal is involved in chemoresistance of renal cell carcinoma cells via regulation of ABCB1. J Cancer 2021; 12:2041-2049. [PMID: 33754002 PMCID: PMC7974526 DOI: 10.7150/jca.52092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 12/26/2020] [Indexed: 11/05/2022] Open
Abstract
Renal cell carcinoma (RCC) is the third most frequent malignancy within urological oncology. Understanding mechanisms of chemoresistance in RCC cell is important for therapy and drug development. We established cisplatin (CDDP) resistant RCC cells by treating cells with increasing concentrations of CDDP. Nodal, an important embryonic morphogen, was increased in RCC/CDDP cells. Targeted inhibition of Nodal via its siRNA or neutralization antibody restored sensitivity of RCC resistant cells to CDDP treatment. It was due to that si-Nodal can decrease expression of P-glycoprotein (P-gp, encoded by ABCB1), one important ATP-binding cassette (ABC) membrane transporter for drug efflux. si-Nodal can decrease the transcription and promoter activity of ABCB1. Mechanistically, si-Nodal can decrease the phosphorylation of p65, which can bind to the promoter of ABCB1 and then trigger its transcription. Further, CDDP treatment decreased the expression of Nodal in culture medium of RCC cells. Collectively, we found that Nodal can regulate chemoresistance of RCC cells via regulating transcription of ABCB1.
Collapse
Affiliation(s)
- Xingwang Zhu
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Dongwei Xue
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Jia Liu
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Fengming Dong
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Yongzhi Li
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Yili Liu
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| |
Collapse
|
9
|
Daraghma H, Untiveros G, Raskind A, Iaccarino E, Sandomenico A, Ruvo M, Arnouk H, Ciancio MJ, Cuevas-Nunez M, Strizzi L. The role of Nodal and Cripto-1 in human oral squamous cell carcinoma. Oral Dis 2020; 27:1137-1147. [PMID: 32916013 DOI: 10.1111/odi.13640] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 08/05/2020] [Accepted: 08/31/2020] [Indexed: 12/16/2022]
Abstract
Oral squamous cell carcinoma (OSCC) is a common epithelial malignancy of the oral cavity. Nodal and Cripto-1 (CR-1) are important developmental morphogens expressed in several adult cancers and are associated with disease progression. Whether Nodal and CR-1 are simultaneously expressed in the same tumor and how this affects cancer biology are unclear. We investigate the expression and potential role of both Nodal and CR-1 in human OSCC. Immunohistochemistry results show that Nodal and CR-1 are both expressed in the same human OSCC sample and that intensity of Nodal staining is correlated with advanced-stage disease. However, this was not observed with CR-1 staining. Western blot analysis of lysates from two human OSCC line experiments shows expression of CR-1 and Nodal, and their respective signaling molecules, Src and ERK1/2. Treatment of SCC25 and SCC15 cells with both Nodal and CR-1 inhibitors simultaneously resulted in reduced cell viability and reduced levels of P-Src and P-ERK1/2. Further investigation showed that the combination treatment with both Nodal and CR-1 inhibitors was capable of reducing invasiveness of SCC25 cells. Our results show a possible role for Nodal/CR-1 function during progression of human OSCC and that targeting both proteins simultaneously may have therapeutic potential.
Collapse
Affiliation(s)
- Hussein Daraghma
- Department of Pathology, College of Graduate Studies, Midwestern University, Downers Grove, IL, USA.,Biomedical Sciences Program, College of Graduate Studies, Midwestern University, Downers Grove, IL, USA
| | - Gustavo Untiveros
- Department of Pathology, College of Graduate Studies, Midwestern University, Downers Grove, IL, USA
| | - Aleksandr Raskind
- Department of Pathology, College of Graduate Studies, Midwestern University, Downers Grove, IL, USA
| | - Emanuela Iaccarino
- Institute of Biostructure and Bioimaging, National Research Council (IBB-CNR), Naples, Italy
| | - Annamaria Sandomenico
- Institute of Biostructure and Bioimaging, National Research Council (IBB-CNR), Naples, Italy
| | - Menotti Ruvo
- Institute of Biostructure and Bioimaging, National Research Council (IBB-CNR), Naples, Italy
| | - Hilal Arnouk
- Department of Pathology, College of Graduate Studies, Midwestern University, Downers Grove, IL, USA
| | - Mae J Ciancio
- Biomedical Sciences Program, College of Graduate Studies, Midwestern University, Downers Grove, IL, USA
| | - Maria Cuevas-Nunez
- College of Dental Medicine Illinois, Midwestern University, Downers Grove, IL, USA
| | - Luigi Strizzi
- Department of Pathology, College of Graduate Studies, Midwestern University, Downers Grove, IL, USA
| |
Collapse
|
10
|
Siegers GM, Dutta I, Kang EY, Huang J, Köbel M, Postovit LM. Aberrantly Expressed Embryonic Protein NODAL Alters Breast Cancer Cell Susceptibility to γδ T Cell Cytotoxicity. Front Immunol 2020; 11:1287. [PMID: 32636849 PMCID: PMC7319087 DOI: 10.3389/fimmu.2020.01287] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 05/21/2020] [Indexed: 01/18/2023] Open
Abstract
Gamma delta (γδ) T cells kill transformed cells, and increased circulating γδ T cells levels correlate with improved outcome in cancer patients; however, their function within the breast tumor microenvironment (TME) remains controversial. As tumors progress, they begin to express stem-cell associated proteins, concomitant with the emergence of therapy resistant metastatic disease. For example, invasive breast cancers often secrete the embryonic morphogen, NODAL. NODAL has been shown to promote angiogenesis, therapy resistance and metastasis in breast cancers. However, to date, little is known about how this secreted protein may interact with cells in the TME. Herein we explore how NODAL in the TME may influence γδ T cell function. We have assessed the proximity of γδ T cells to NODAL in a cohort of triple negative breast tumors. In all cases in which γδ T cells could be identified in these tumors, γδ T cells were found in close proximity to NODAL-expressing tumor cells. Migration of γδ and αβ T cells was similar toward MDA-MB-231 cells in which NODAL had been knocked down (shN) and MDA-MB-231 scrambled control cells (shC). Furthermore, Vδ1 γδ T cells did not migrate preferentially toward conditioned medium from these cell lines. While 24-h exposure to NODAL did not impact CD69, PD-1, or T cell antigen receptor (TCR) expression on γδ T cells, long term exposure resulted in decreased Vδ2 TCR expression. Maturation of γδ T cells was not significantly influenced by NODAL stimulation. While neither short- nor long-term NODAL stimulation impacted the ability of γδ T cells to kill MCF-7 breast cancer cells, the absence of NODAL resulted in greater sensitivity of targets to γδ T cell cytotoxicity, while overexpression of NODAL conferred resistance. This appeared to be at least in part due to an inverse correlation between NODAL and surface MICA/B expression on breast cancer target lines. As such, it appears that NODAL may play a role in strategies employed by breast cancer cells to evade γδ T cell targeting, and this should be considered in the development of safe and effective γδ T cell immunotherapies.
Collapse
Affiliation(s)
| | - Indrani Dutta
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Eun Young Kang
- Department of Pathology and Laboratory Medicine, Foothills Medical Centre, University of Calgary, Calgary, AB, Canada
| | - Jing Huang
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Martin Köbel
- Department of Pathology and Laboratory Medicine, Foothills Medical Centre, University of Calgary, Calgary, AB, Canada
| | - Lynne-Marie Postovit
- Department of Oncology, University of Alberta, Edmonton, AB, Canada.,Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| |
Collapse
|
11
|
Rodrigues Sousa E, Zoni E, Karkampouna S, La Manna F, Gray PC, De Menna M, Kruithof-de Julio M. A Multidisciplinary Review of the Roles of Cripto in the Scientific Literature Through a Bibliometric Analysis of its Biological Roles. Cancers (Basel) 2020; 12:cancers12061480. [PMID: 32517087 PMCID: PMC7352664 DOI: 10.3390/cancers12061480] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/01/2020] [Accepted: 06/02/2020] [Indexed: 12/21/2022] Open
Abstract
Cripto is a small glycosylphosphatidylinisitol (GPI)-anchored and secreted oncofetal protein that plays important roles in regulating normal physiological processes, including stem cell differentiation, embryonal development, and tissue growth and remodeling, as well as pathological processes such as tumor initiation and progression. Cripto functions as a co-receptor for TGF-β ligands such as Nodal, GDF1, and GDF3. Soluble and secreted forms of Cripto also exhibit growth factor-like activity and activate SRC/MAPK/PI3K/AKT pathways. Glucose-Regulated Protein 78 kDa (GRP78) binds Cripto at the cell surface and has been shown to be required for Cripto signaling via both TGF-β and SRC/MAPK/PI3K/AKT pathways. To provide a comprehensive overview of the scientific literature related to Cripto, we performed, for the first time, a bibliometric analysis of the biological roles of Cripto as reported in the scientific literature covering the last 10 years. We present different fields of knowledge in comprehensive areas of research on Cripto, ranging from basic to translational research, using a keyword-driven approach. Our ultimate aim is to aid the scientific community in conducting targeted research by identifying areas where research has been conducted so far and, perhaps more importantly, where critical knowledge is still missing.
Collapse
Affiliation(s)
- Elisa Rodrigues Sousa
- Department for Biomedical Research, Urology Research Laboratory, University of Bern, 3008 Bern, Switzerland; (E.R.S.); (E.Z.); (S.K.); (F.L.M.); (M.D.M.)
| | - Eugenio Zoni
- Department for Biomedical Research, Urology Research Laboratory, University of Bern, 3008 Bern, Switzerland; (E.R.S.); (E.Z.); (S.K.); (F.L.M.); (M.D.M.)
- Department of Urology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| | - Sofia Karkampouna
- Department for Biomedical Research, Urology Research Laboratory, University of Bern, 3008 Bern, Switzerland; (E.R.S.); (E.Z.); (S.K.); (F.L.M.); (M.D.M.)
| | - Federico La Manna
- Department for Biomedical Research, Urology Research Laboratory, University of Bern, 3008 Bern, Switzerland; (E.R.S.); (E.Z.); (S.K.); (F.L.M.); (M.D.M.)
- Department of Urology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | | | - Marta De Menna
- Department for Biomedical Research, Urology Research Laboratory, University of Bern, 3008 Bern, Switzerland; (E.R.S.); (E.Z.); (S.K.); (F.L.M.); (M.D.M.)
| | - Marianna Kruithof-de Julio
- Department for Biomedical Research, Urology Research Laboratory, University of Bern, 3008 Bern, Switzerland; (E.R.S.); (E.Z.); (S.K.); (F.L.M.); (M.D.M.)
- Department of Urology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Correspondence:
| |
Collapse
|
12
|
Jewer M, Lee L, Leibovitch M, Zhang G, Liu J, Findlay SD, Vincent KM, Tandoc K, Dieters-Castator D, Quail DF, Dutta I, Coatham M, Xu Z, Puri A, Guan BJ, Hatzoglou M, Brumwell A, Uniacke J, Patsis C, Koromilas A, Schueler J, Siegers GM, Topisirovic I, Postovit LM. Translational control of breast cancer plasticity. Nat Commun 2020; 11:2498. [PMID: 32427827 PMCID: PMC7237473 DOI: 10.1038/s41467-020-16352-z] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 04/27/2020] [Indexed: 12/19/2022] Open
Abstract
Plasticity of neoplasia, whereby cancer cells attain stem-cell-like properties, is required for disease progression and represents a major therapeutic challenge. We report that in breast cancer cells NANOG, SNAIL and NODAL transcripts manifest multiple isoforms characterized by different 5' Untranslated Regions (5'UTRs), whereby translation of a subset of these isoforms is stimulated under hypoxia. The accumulation of the corresponding proteins induces plasticity and "fate-switching" toward stem cell-like phenotypes. Mechanistically, we observe that mTOR inhibitors and chemotherapeutics induce translational activation of a subset of NANOG, SNAIL and NODAL mRNA isoforms akin to hypoxia, engendering stem-cell-like phenotypes. These effects are overcome with drugs that antagonize translational reprogramming caused by eIF2α phosphorylation (e.g. ISRIB), suggesting that the Integrated Stress Response drives breast cancer plasticity. Collectively, our findings reveal a mechanism of induction of plasticity of breast cancer cells and provide a molecular basis for therapeutic strategies aimed at overcoming drug resistance and abrogating metastasis.
Collapse
Affiliation(s)
- Michael Jewer
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Laura Lee
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Matthew Leibovitch
- Lady Davis Institute, Departments of Oncology and Biochemistry, Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Guihua Zhang
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Jiahui Liu
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Scott D Findlay
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Krista M Vincent
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Kristofferson Tandoc
- Lady Davis Institute, Departments of Oncology and Biochemistry, Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Dylan Dieters-Castator
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Daniela F Quail
- Goodman Cancer Center, McGill University, Montreal, QC, Canada
| | - Indrani Dutta
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | | | - Zhihua Xu
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Aakshi Puri
- Lady Davis Institute, Departments of Oncology and Biochemistry, Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Bo-Jhih Guan
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - Maria Hatzoglou
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - Andrea Brumwell
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - James Uniacke
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Christos Patsis
- Lady Davis Institute, Departments of Oncology and Biochemistry, Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Antonis Koromilas
- Lady Davis Institute, Departments of Oncology and Biochemistry, Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Julia Schueler
- Charles River Discovery Research Services Germany, Freiburg, Germany
| | | | - Ivan Topisirovic
- Lady Davis Institute, Departments of Oncology and Biochemistry, Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Lynne-Marie Postovit
- Department of Oncology, University of Alberta, Edmonton, AB, Canada.
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
13
|
Harpelunde Poulsen K, Nielsen JE, Grønkær Toft B, Joensen UN, Rasmussen LJ, Blomberg Jensen M, Mitchell RT, Juul A, Rajpert-De Meyts E, Jørgensen A. Influence of Nodal signalling on pluripotency factor expression, tumour cell proliferation and cisplatin-sensitivity in testicular germ cell tumours. BMC Cancer 2020; 20:349. [PMID: 32326899 PMCID: PMC7181506 DOI: 10.1186/s12885-020-06820-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/02/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Testicular germ cell tumours (TGCTs) are characterised by an overall high cisplatin-sensitivity which has been linked to their continued expression of pluripotency factors. Recently, the Nodal signalling pathway has been implicated in the regulation of pluripotency factor expression in fetal germ cells, and the pathway could therefore also be involved in regulating expression of pluripotency factors in malignant germ cells, and hence cisplatin-sensitivity in TGCTs. METHODS We used in vitro culture of the TGCT-derived cell line NTera2, ex vivo tissue culture of primary TGCT specimens and xenografting of NTera2 cells into nude mice in order to investigate the consequences of manipulating Nodal and Activin signalling on pluripotency factor expression, apoptosis, proliferation and cisplatin-sensitivity. RESULTS The Nodal signalling factors were markedly expressed concomitantly with the pluripotency factor OCT4 in GCNIS cells, seminomas and embryonal carcinomas. Despite this, inhibition of Nodal and Activin signalling either alone or simultaneously did not affect proliferation or apoptosis in malignant germ cells in vitro or ex vivo. Interestingly, inhibition of Nodal signalling in vitro reduced the expression of pluripotency factors and Nodal pathway genes, while stimulation of the pathway increased their expression. However, cisplatin-sensitivity was not affected following pharmacological inhibition of Nodal/Activin signalling or siRNA-mediated knockdown of the obligate co-receptor CRIPTO in NTera2 cells in vitro or in a xenograft model. CONCLUSION Our findings suggest that the Nodal signalling pathway may be involved in regulating pluripotency factor expression in malignant germ cells, but manipulation of the pathway does not appear to affect cisplatin-sensitivity or tumour cell proliferation.
Collapse
Affiliation(s)
- K Harpelunde Poulsen
- Department of Growth and Reproduction, Copenhagen University Hospital (Rigshospitalet), Blegdamsvej 9, DK-2100, Copenhagen, Denmark.,International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Blegdamsvej 9, Copenhagen, Denmark
| | - J E Nielsen
- Department of Growth and Reproduction, Copenhagen University Hospital (Rigshospitalet), Blegdamsvej 9, DK-2100, Copenhagen, Denmark.,International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Blegdamsvej 9, Copenhagen, Denmark
| | - B Grønkær Toft
- Pathology Department, Copenhagen University Hospital (Rigshospitalet), Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - U N Joensen
- Department of Urology, Copenhagen University Hospital (Rigshospitalet), Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - L J Rasmussen
- Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - M Blomberg Jensen
- Department of Growth and Reproduction, Copenhagen University Hospital (Rigshospitalet), Blegdamsvej 9, DK-2100, Copenhagen, Denmark
| | - R T Mitchell
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - A Juul
- Department of Growth and Reproduction, Copenhagen University Hospital (Rigshospitalet), Blegdamsvej 9, DK-2100, Copenhagen, Denmark.,International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Blegdamsvej 9, Copenhagen, Denmark
| | - E Rajpert-De Meyts
- Department of Growth and Reproduction, Copenhagen University Hospital (Rigshospitalet), Blegdamsvej 9, DK-2100, Copenhagen, Denmark.,International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Blegdamsvej 9, Copenhagen, Denmark
| | - A Jørgensen
- Department of Growth and Reproduction, Copenhagen University Hospital (Rigshospitalet), Blegdamsvej 9, DK-2100, Copenhagen, Denmark. .,International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Blegdamsvej 9, Copenhagen, Denmark.
| |
Collapse
|
14
|
Sandomenico A, Ruvo M. Targeting Nodal and Cripto-1: Perspectives Inside Dual Potential Theranostic Cancer Biomarkers. Curr Med Chem 2019; 26:1994-2050. [PMID: 30207211 DOI: 10.2174/0929867325666180912104707] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 07/13/2018] [Accepted: 07/17/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND Elucidating the mechanisms of recurrence of embryonic signaling pathways in tumorigenesis has led to the discovery of onco-fetal players which have physiological roles during normal development but result aberrantly re-activated in tumors. In this context, Nodal and Cripto-1 are recognized as onco-developmental factors, which are absent in normal tissues but are overexpressed in several solid tumors where they can serve as theranostic agents. OBJECTIVE To collect, review and discuss the most relevant papers related to the involvement of Nodal and Cripto-1 in the development, progression, recurrence and metastasis of several tumors where they are over-expressed, with a particular attention to their occurrence on the surface of the corresponding sub-populations of cancer stem cells (CSC). RESULTS We have gathered, rationalized and discussed the most interesting findings extracted from some 370 papers related to the involvement of Cripto-1 and Nodal in all tumor types where they have been detected. Data demonstrate the clear connection between Nodal and Cripto-1 presence and their multiple oncogenic activities across different tumors. We have also reviewed and highlighted the potential of targeting Nodal, Cripto-1 and the complexes that they form on the surface of tumor cells, especially of CSC, as an innovative approach to detect and suppress tumors with molecules that block one or more mechanisms that they regulate. CONCLUSION Overall, Nodal and Cripto-1 represent two innovative and effective biomarkers for developing potential theranostic anti-tumor agents that target normal as well as CSC subpopulations and overcome both pharmacological resistance and tumor relapse.
Collapse
Affiliation(s)
- Annamaria Sandomenico
- Istituto di Biostrutture e Bioimmagini, Consiglio Nazionale delle Ricerche (IBB-CNR), via Mezzocannone, 16, 80134, Napoli, Italy
| | - Menotti Ruvo
- Istituto di Biostrutture e Bioimmagini, Consiglio Nazionale delle Ricerche (IBB-CNR), via Mezzocannone, 16, 80134, Napoli, Italy
| |
Collapse
|
15
|
Guo Q, Li VZ, Nichol JN, Huang F, Yang W, Preston SEJ, Talat Z, Lefrère H, Yu H, Zhang G, Basik M, Gonçalves C, Zhan Y, Plourde D, Su J, Torres J, Marques M, Habyan SA, Bijian K, Amant F, Witcher M, Behbod F, McCaffrey L, Alaoui-Jamali M, Giannakopoulos NV, Brackstone M, Postovit LM, Del Rincón SV, Miller WH. MNK1/NODAL Signaling Promotes Invasive Progression of Breast Ductal Carcinoma In Situ. Cancer Res 2019; 79:1646-1657. [PMID: 30659022 PMCID: PMC6513674 DOI: 10.1158/0008-5472.can-18-1602] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 11/02/2018] [Accepted: 01/10/2019] [Indexed: 12/11/2022]
Abstract
The mechanisms by which breast cancers progress from relatively indolent ductal carcinoma in situ (DCIS) to invasive ductal carcinoma (IDC) are not well understood. However, this process is critical to the acquisition of metastatic potential. MAPK-interacting serine/threonine-protein kinase 1 (MNK1) signaling can promote cell invasion. NODAL, a morphogen essential for embryogenic patterning, is often reexpressed in breast cancer. Here we describe a MNK1/NODAL signaling axis that promotes DCIS progression to IDC. We generated MNK1 knockout (KO) or constitutively active MNK1 (caMNK1)-expressing human MCF-10A-derived DCIS cell lines, which were orthotopically injected into the mammary glands of mice. Loss of MNK1 repressed NODAL expression, inhibited DCIS to IDC conversion, and decreased tumor relapse and metastasis. Conversely, caMNK1 induced NODAL expression and promoted IDC. The MNK1/NODAL axis promoted cancer stem cell properties and invasion in vitro. The MNK1/2 inhibitor SEL201 blocked DCIS progression to invasive disease in vivo. In clinical samples, IDC and DCIS with microinvasion expressed higher levels of phospho-MNK1 and NODAL versus low-grade (invasion-free) DCIS. Cumulatively, our data support further development of MNK1 inhibitors as therapeutics for preventing invasive disease. SIGNIFICANCE: These findings provide new mechanistic insight into progression of ductal carcinoma and support clinical application of MNK1 inhibitors to delay progression of indolent ductal carcinoma in situ to invasive ductal carcinoma.
Collapse
Affiliation(s)
- Qianyu Guo
- Division of Experimental Medicine, McGill University, Montréal, Québec, Canada
- Department of Oncology, Segal Cancer Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Vivian Z Li
- Department of Oncology, Segal Cancer Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Jessica N Nichol
- Department of Oncology, Segal Cancer Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Fan Huang
- Division of Experimental Medicine, McGill University, Montréal, Québec, Canada
- Department of Oncology, Segal Cancer Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - William Yang
- Division of Experimental Medicine, McGill University, Montréal, Québec, Canada
- Department of Oncology, Segal Cancer Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Samuel E J Preston
- Division of Experimental Medicine, McGill University, Montréal, Québec, Canada
- Department of Oncology, Segal Cancer Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Zahra Talat
- Department of Oncology, Segal Cancer Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Hanne Lefrère
- Department of Oncology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Henry Yu
- Division of Experimental Medicine, McGill University, Montréal, Québec, Canada
- Department of Oncology, Segal Cancer Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Guihua Zhang
- Cancer Research Institute of Northern Alberta, Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Mark Basik
- Division of Experimental Medicine, McGill University, Montréal, Québec, Canada
- Department of Oncology, Segal Cancer Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Christophe Gonçalves
- Department of Oncology, Segal Cancer Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Yao Zhan
- Division of Experimental Medicine, McGill University, Montréal, Québec, Canada
- Department of Oncology, Segal Cancer Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Dany Plourde
- Department of Oncology, Segal Cancer Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Jie Su
- Department of Oncology, Segal Cancer Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Jose Torres
- Division of Experimental Medicine, McGill University, Montréal, Québec, Canada
- Department of Oncology, Segal Cancer Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Maud Marques
- Department of Oncology, Segal Cancer Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Sara Al Habyan
- Goodman Cancer Centre, McGill University, Montréal, Québec, Canada
| | - Krikor Bijian
- Department of Oncology, Segal Cancer Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Frédéric Amant
- Department of Oncology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Michael Witcher
- Division of Experimental Medicine, McGill University, Montréal, Québec, Canada
- Department of Oncology, Segal Cancer Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Fariba Behbod
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Centre, Kansas City, Kansas
| | - Luke McCaffrey
- Division of Experimental Medicine, McGill University, Montréal, Québec, Canada
- Goodman Cancer Centre, McGill University, Montréal, Québec, Canada
| | - Moulay Alaoui-Jamali
- Division of Experimental Medicine, McGill University, Montréal, Québec, Canada
- Department of Oncology, Segal Cancer Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Nadia V Giannakopoulos
- Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Muriel Brackstone
- Departments of Surgery and Oncology, Western University, London, Ontario, Canada
| | - Lynne-Marie Postovit
- Cancer Research Institute of Northern Alberta, Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
- Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Sonia V Del Rincón
- Division of Experimental Medicine, McGill University, Montréal, Québec, Canada.
- Department of Oncology, Segal Cancer Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Wilson H Miller
- Division of Experimental Medicine, McGill University, Montréal, Québec, Canada.
- Department of Oncology, Segal Cancer Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Rossy Cancer Network, McGill University, Montréal, Québec, Canada
| |
Collapse
|
16
|
The Stem Cell Phenotype of Aggressive Breast Cancer Cells. Cancers (Basel) 2019; 11:cancers11030340. [PMID: 30857267 PMCID: PMC6468512 DOI: 10.3390/cancers11030340] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 02/25/2019] [Accepted: 03/04/2019] [Indexed: 11/16/2022] Open
Abstract
Aggressive cancer cells are characterized by their capacity to proliferate indefinitely and to propagate a heterogeneous tumor comprised of subpopulations with varying degrees of metastatic propensity and drug resistance properties. Particularly daunting is the challenge we face in the field of oncology of effectively targeting heterogeneous tumor cells expressing a variety of markers, especially those associated with a stem cell phenotype. This dilemma is especially relevant in breast cancer, where therapy is based on traditional classification schemes, including histological criteria, differentiation status, and classical receptor markers. However, not all patients respond in a similar manner to standard-of-care therapy, thereby necessitating the need to identify and evaluate novel biomarkers associated with the difficult-to-target stem cell phenotype and drug resistance. Findings related to the convergence of embryonic and tumorigenic signaling pathways have identified the embryonic morphogen Nodal as a promising new oncofetal target that is reactivated only in aggressive cancers, but not in normal tissues. The work presented in this paper confirms previous studies demonstrating the importance of Nodal as a cancer stem cell molecule associated with aggressive breast cancer, and advances the field by providing new findings showing that Nodal is not targeted by standard-of-care therapy in breast cancer patients. Most noteworthy is the linkage found between Nodal expression and the drug resistance marker ATP-binding cassette member 1 (ABCA1), which may provide new insights into developing combinatorial approaches to overcome drug resistance and disease recurrence.
Collapse
|
17
|
Chen W, Jiang T, Mao H, Gao R, Gao X, He Y, Zhang H, Chen Q. Nodal Promotes the Migration and Invasion of Bladder Cancer Cells via Regulation of Snail. J Cancer 2019; 10:1511-1519. [PMID: 31031861 PMCID: PMC6485227 DOI: 10.7150/jca.29205] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 01/04/2019] [Indexed: 01/11/2023] Open
Abstract
Urinary bladder cancer is one of commonly diagnosed malignancies worldwide, especially in males. Understanding the mechanisms of advanced metastasis in bladder cell is important for therapy and drug development. Nodal, an important embryonic morphogen, has been reported to modulate tumorigenesis. We found that the expression of Nodal was upregulated in bladder cancer cells and tissues as compared to their corresponding controls. Knockdown of Nodal can suppress the migration, invasion, and epithelial-to-mesenchymal transition (EMT) of bladder cancer cells. Nodal can positively regulate the expression of Snail, one powerful EMT transcription factors, in bladder cancer cells. Overexpression of Snail can attenuate the si-Nodal suppressed cell migration and invasion. Nodal can increase the transcription and protein stability of Snail in bladder cancer cells. YY1, which can be activated by Nodal, is responsible for Nodal induced transcription of Snail. ATM, which can stabilize Snail by phosphorylation on Serine-100, was involved in Nodal upregulated protein stability of Snail. Collectively, our data showed that Nodal can trigger the malignancy of bladder cancer cells via increasing the transcription and protein stability of Snail. It indicated that Nodal might be a potential therapeutic target for bladder cancer treatment.
Collapse
Affiliation(s)
- Wenwei Chen
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Tao Jiang
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Houping Mao
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Rui Gao
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Xingjian Gao
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Yanfeng He
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Hua Zhang
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Qin Chen
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| |
Collapse
|
18
|
Seftor EA, Margaryan NV, Seftor REB, Hendrix MJC. Heterogeneity of Melanoma with Stem Cell Properties. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1139:105-114. [PMID: 31134497 DOI: 10.1007/978-3-030-14366-4_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Metastatic melanoma continues to present a significant challenge-with a cure rate of less than 10% and a median survival of 6-9 months. Despite noteworthy advances in the field, the heterogeneity of melanoma tumors, comprised of cell subpopulations expressing a cancer stem cell (CSC) phenotype concomitant with drug resistance markers presents a formidable challenge in the design of current therapies. Particularly vexing is the ability of distinct subpopulations of melanoma cells to resist standard-of-care treatments, resulting in relapse and progression to metastasis. Recent studies have provided new information and insights into the expression and function of CSC markers associated with the aggressive melanoma phenotype, such as the embryonic morphogen Nodal and CD133, together with a drug resistance marker ABCA1. This chapter highlights major findings that demonstrate the promise of targeting Nodal as a viable option to pursue in combination with standard-of-care therapy. In recognizing that aggressive melanoma tumors utilize multiple mechanisms to survive, we must consider a more strategic approach to effectively target heterogeneity, tumor cell plasticity, and functional adaptation and resistance to current therapies-to eliminate relapse, disease progression, and metastasis.
Collapse
Affiliation(s)
- Elisabeth A Seftor
- Department of Biochemistry and Cancer Institute, West Virginia University Health Sciences Center, One Medical Center Drive, Morgantown, WV, USA
| | - Naira V Margaryan
- Department of Biochemistry and Cancer Institute, West Virginia University Health Sciences Center, One Medical Center Drive, Morgantown, WV, USA
| | - Richard E B Seftor
- Department of Biochemistry and Cancer Institute, West Virginia University Health Sciences Center, One Medical Center Drive, Morgantown, WV, USA
| | - Mary J C Hendrix
- Department of Biology, Shepherd University, Shepherdstown, WV, USA.
| |
Collapse
|
19
|
Kalyan A, Carneiro BA, Chandra S, Kaplan J, Chae YK, Matsangou M, Hendrix MJC, Giles F. Nodal Signaling as a Developmental Therapeutics Target in Oncology. Mol Cancer Ther 2018; 16:787-792. [PMID: 28468864 DOI: 10.1158/1535-7163.mct-16-0215] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 11/22/2016] [Accepted: 12/05/2016] [Indexed: 11/16/2022]
Abstract
The tumor microenvironment is a vital feature of oncogenesis and tumor progression. There are several parallels between cancer cells and early developmental stem cells, including their plasticity and signaling mechanisms. In early fetal development, Nodal is expressed for endodermal and mesodermal differentiation. This expression has been shown reemerge in the setting of epithelial cancers, such as breast and melanoma. High Nodal expression correlates to an aggressive tumor grade in these malignancies. Nodal signal begins with its interaction with its coreceptor, Cripto-1, leading to activation of Smad2/Smad3 and ultimately downstream transcription and translation. Lefty is the natural inhibitor of Nodal and controls Nodal signaling during fetal development. However, cancer cells lack the presence of Lefty, thus leading to uncontrolled tumor growth. Given this understanding, inhibition of the Nodal pathway offers a new novel therapeutic target in oncology. Mol Cancer Ther; 16(5); 787-92. ©2017 AACR.
Collapse
Affiliation(s)
- Aparna Kalyan
- Developmental Therapeutics Program, Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Olson Pavilion, Chicago, Illinois. .,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Benedito A Carneiro
- Developmental Therapeutics Program, Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Olson Pavilion, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Sunandana Chandra
- Developmental Therapeutics Program, Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Olson Pavilion, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Jason Kaplan
- Developmental Therapeutics Program, Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Olson Pavilion, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Young Kwang Chae
- Developmental Therapeutics Program, Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Olson Pavilion, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Maria Matsangou
- Developmental Therapeutics Program, Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Olson Pavilion, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Mary J C Hendrix
- Developmental Therapeutics Program, Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Olson Pavilion, Chicago, Illinois.,Cancer Biology and Epigenomics Program, Stanley Manne Children's Research Institute, Anne and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Francis Giles
- Developmental Therapeutics Program, Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Olson Pavilion, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| |
Collapse
|
20
|
Gong W, Sun B, Zhao X, Zhang D, Sun J, Liu T, Gu Q, Dong X, Liu F, Wang Y, Lin X, Li Y. Nodal signaling promotes vasculogenic mimicry formation in breast cancer via the Smad2/3 pathway. Oncotarget 2018; 7:70152-70167. [PMID: 27659524 PMCID: PMC5342542 DOI: 10.18632/oncotarget.12161] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 09/14/2016] [Indexed: 01/06/2023] Open
Abstract
Vasculogenic mimicry (VM) is a nonangiogenesis-dependent pathway that promotes tumor growth and disease progression. Nodal signaling has several vital roles in both embryo development and cancer progression. However, the effects of Nodal signaling on VM formation in breast cancer and its underlying mechanisms are ill-defined. We analyzed the relationship between Nodal signaling and VM formation in one hundred human breast cancer cases and the results showed that the expression of Nodal was significantly correlated with VM formation, tumor metastasis, differentiation grade, TNM stage and poor prognosis. Furthermore, up-regulation of Nodal expression promoted VM formation of breast cancer cells in vitro and in vivo. Knockdown of Nodal expression restrained VM formation. In addition, Nodal induced EMT and up-regulated the expression of Slug, Snail and c-Myc. We found that blocking the Smad2/3 pathway by administering SB431542 inhibited VM formation in breast cancer cell lines and xenografts. Taken together, Nodal signaling through the Smad2/3 pathway up-regulated Slug, Snail and c-Myc to induce EMT, thereby promoting VM formation. Our study suggests that the Nodal signaling pathway may serve as a therapeutic target to inhibit VM formation and improve prognosis in breast cancer patients.
Collapse
Affiliation(s)
- Wenchen Gong
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China
| | - Baocun Sun
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China.,Department of Pathology, Tianjin General Hospital, Tianjin Medical University, Tianjin, 300052, China.,Department of Pathology, Tianjin Cancer Hospital, Tianjin Medical University, Tianjin, 300060, China
| | - Xiulan Zhao
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China.,Department of Pathology, Tianjin General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Danfang Zhang
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China.,Department of Pathology, Tianjin General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Junying Sun
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China
| | - Tieju Liu
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China.,Department of Pathology, Tianjin General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Qiang Gu
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China.,Department of Pathology, Tianjin General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Xueyi Dong
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China
| | - Fang Liu
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China
| | - Yong Wang
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China
| | - Xian Lin
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China
| | - Yanlei Li
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China
| |
Collapse
|
21
|
Calvanese L, Focà A, Sandomenico A, Focà G, Caporale A, Doti N, Iaccarino E, Leonardi A, D'Auria G, Ruvo M, Falcigno L. Structural insights into the interaction of a monoclonal antibody and Nodal peptides by STD-NMR spectroscopy. Bioorg Med Chem 2017; 25:6589-6596. [PMID: 29113739 DOI: 10.1016/j.bmc.2017.10.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 10/05/2017] [Accepted: 10/26/2017] [Indexed: 12/31/2022]
Abstract
Nodal is a growth factor expressed during early embryonic development, but reactivated in several advanced-stage cancers. Targeting of Nodal signaling, which occurs via the binding to Cripto-1 co-receptor, results in inhibition of cell aggressiveness and reduced tumor growth. The Nodal binding region to Cripto-1 was identified and targeted with a high affinity monoclonal antibody (3D1). By STD-NMR technique, we investigated the interaction of Nodal fragments with 3D1 with the aim to elucidate at atomic level the interaction surface. Data indicate with high accuracy the antibody-antigen contact atoms and confirm the information previously obtained by immune-enzymatic methods. Main residues contacted by 3D1 are P46, V47, E49 and E50, which belong to the Nodal loop involved in the interaction with the co-receptor.
Collapse
Affiliation(s)
- Luisa Calvanese
- CIRPeB, University of Naples Federico II, via Mezzocannone, 16, 80134 Napoli, Italy
| | - Annalia Focà
- Institute of Biostructures and Bioimaging, National Research Council, IBB-CNR, via Mezzocannone, 16, 80134 Napoli, Italy
| | - Annamaria Sandomenico
- CIRPeB, University of Naples Federico II, via Mezzocannone, 16, 80134 Napoli, Italy; Institute of Biostructures and Bioimaging, National Research Council, IBB-CNR, via Mezzocannone, 16, 80134 Napoli, Italy
| | - Giuseppina Focà
- Institute of Biostructures and Bioimaging, National Research Council, IBB-CNR, via Mezzocannone, 16, 80134 Napoli, Italy
| | - Andrea Caporale
- CIRPeB, University of Naples Federico II, via Mezzocannone, 16, 80134 Napoli, Italy
| | - Nunzianna Doti
- Institute of Biostructures and Bioimaging, National Research Council, IBB-CNR, via Mezzocannone, 16, 80134 Napoli, Italy
| | - Emanuela Iaccarino
- DISTABIF, Università degli Studi della Campania "Lugi Vanvitelli", via Vivaldi, 43, 80100 Caserta, Italy
| | - Antonio Leonardi
- Dept. Medicina Molecolare e Biotecnologie Mediche, Università Federico II di Napoli, Naples, Italy
| | - Gabriella D'Auria
- CIRPeB, University of Naples Federico II, via Mezzocannone, 16, 80134 Napoli, Italy; Institute of Biostructures and Bioimaging, National Research Council, IBB-CNR, via Mezzocannone, 16, 80134 Napoli, Italy; Dept. of Pharmacy, University of Naples Federico II, via Mezzocannone, 16, 80134 Napoli, Italy
| | - Menotti Ruvo
- CIRPeB, University of Naples Federico II, via Mezzocannone, 16, 80134 Napoli, Italy; Institute of Biostructures and Bioimaging, National Research Council, IBB-CNR, via Mezzocannone, 16, 80134 Napoli, Italy.
| | - Lucia Falcigno
- CIRPeB, University of Naples Federico II, via Mezzocannone, 16, 80134 Napoli, Italy; Institute of Biostructures and Bioimaging, National Research Council, IBB-CNR, via Mezzocannone, 16, 80134 Napoli, Italy; Dept. of Pharmacy, University of Naples Federico II, via Mezzocannone, 16, 80134 Napoli, Italy.
| |
Collapse
|
22
|
Margaryan NV, Seftor EA, Seftor RE, Hendrix MJ. Targeting the Stem Cell Properties of Adult Breast Cancer Cells: Using Combinatorial Strategies to Overcome Drug Resistance. CURRENT MOLECULAR BIOLOGY REPORTS 2017; 3:159-164. [PMID: 29152453 PMCID: PMC5687579 DOI: 10.1007/s40610-017-0067-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE OF REVIEW Cancer is a major public health problem worldwide. In aggressive cancers, which are heterogeneous in nature, there exists a paucity of targetable molecules that can be used to predict outcome and response to therapy in patients, especially those in the high risk category with a propensity to relapse following chemotherapy. This review addresses the challenges pertinent to treating aggressive cancer cells with inherent stem cell properties, with a special focus on triple-negative breast cancer (TNBC). RECENT FINDINGS Plasticity underlies the cancer stem cell (CSC) phenotype in aggressive cancers like TNBC. Progenitors and CSCs implement similar signaling pathways to sustain growth, and the convergence of embryonic and tumorigenic signaling pathways has led to the discovery of novel oncofetal targets, rigorously regulated during normal development, but aberrantly reactivated in aggressive forms of cancer. SUMMARY Translational studies have shown that Nodal, an embryonic morphogen, is reactivated in aggressive cancers, but not in normal tissues, and underlies tumor growth, invasion, metastasis and drug resistance. Front-line therapies do not inhibit Nodal, but when a combinatorial approach is used with an agent such as doxorubicin followed by anti-Nodal antibody therapy, significant decreases in cell growth and viability occur. These findings are of special interest in the development of new therapeutic interventions that target the stem cell properties of cancer cells to overcome drug resistance and metastasis.
Collapse
Affiliation(s)
- Naira V. Margaryan
- Department of Biochemistry, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26506 USA
- Cancer Institute, West Virginia University, Morgantown, WV 26506 USA
- Department of Biology, Shepherd University, Shepherdstown, WV 25443 USA
| | - Elisabeth A. Seftor
- Department of Biochemistry, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26506 USA
- Cancer Institute, West Virginia University, Morgantown, WV 26506 USA
- Department of Biology, Shepherd University, Shepherdstown, WV 25443 USA
| | - Richard E.B. Seftor
- Department of Biochemistry, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26506 USA
- Cancer Institute, West Virginia University, Morgantown, WV 26506 USA
- Department of Biology, Shepherd University, Shepherdstown, WV 25443 USA
| | - Mary J.C. Hendrix
- Department of Internal Medicine, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26506 USA
- Cancer Institute, West Virginia University, Morgantown, WV 26506 USA
- Department of Biology, Shepherd University, Shepherdstown, WV 25443 USA
| |
Collapse
|
23
|
Bodenstine TM, Chandler GS, Reed DW, Margaryan NV, Gilgur A, Atkinson J, Ahmed N, Hyser M, Seftor EA, Strizzi L, Hendrix MJC. Nodal expression in triple-negative breast cancer: Cellular effects of its inhibition following doxorubicin treatment. Cell Cycle 2017; 15:1295-302. [PMID: 27007464 DOI: 10.1080/15384101.2016.1160981] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Triple-negative breast cancer (TNBC) represents an aggressive cancer subtype characterized by the lack of expression of estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2). The independence of TNBC from these growth promoting factors eliminates the efficacy of therapies which specifically target them, and limits TNBC patients to traditional systemic neo/adjuvant chemotherapy. To better understand the growth advantage of TNBC - in the absence of ER, PR and HER2, we focused on the embryonic morphogen Nodal (associated with the cancer stem cell phenotype), which is re-expressed in aggressive breast cancers. Most notably, our previous data demonstrated that inhibition of Nodal signaling in breast cancer cells reduces their tumorigenic capacity. Furthermore, inhibiting Nodal in other cancers has resulted in improved effects of chemotherapy, although the mechanisms for this remain unknown. Thus, we hypothesized that targeting Nodal in TNBC cells in combination with conventional chemotherapy may improve efficacy and represent a potential new strategy. Our preliminary data demonstrate that Nodal is highly expressed in TNBC when compared to invasive hormone receptor positive samples. Treatment of Nodal expressing TNBC cell lines with a neutralizing anti-Nodal antibody reduces the viability of cells that had previously survived treatment with the anthracycline doxorubicin. We show that inhibiting Nodal may alter response mechanisms employed by cancer cells undergoing DNA damage. These data suggest that development of therapies which target Nodal in TNBC may lead to additional treatment options in conjunction with chemotherapy regimens - by altering signaling pathways critical to cellular survival.
Collapse
Affiliation(s)
- Thomas M Bodenstine
- a Cancer Biology and Epigenomics Program at the Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago , Chicago , IL , USA
| | - Grace S Chandler
- a Cancer Biology and Epigenomics Program at the Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago , Chicago , IL , USA
| | - David W Reed
- a Cancer Biology and Epigenomics Program at the Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago , Chicago , IL , USA
| | - Naira V Margaryan
- a Cancer Biology and Epigenomics Program at the Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago , Chicago , IL , USA
| | - Alina Gilgur
- a Cancer Biology and Epigenomics Program at the Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago , Chicago , IL , USA
| | | | - Nida Ahmed
- b Presence Saint Francis Hospital , Evanston , IL , USA
| | - Matthew Hyser
- b Presence Saint Francis Hospital , Evanston , IL , USA
| | - Elisabeth A Seftor
- a Cancer Biology and Epigenomics Program at the Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago , Chicago , IL , USA
| | - Luigi Strizzi
- a Cancer Biology and Epigenomics Program at the Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago , Chicago , IL , USA.,c Department of Pathology , Northwestern University Feinberg School of Medicine , Chicago , IL , USA
| | - Mary J C Hendrix
- a Cancer Biology and Epigenomics Program at the Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago , Chicago , IL , USA.,d Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine , Chicago , IL , USA
| |
Collapse
|
24
|
Hendrix MJ, Kandela I, Mazar AP, Seftor EA, Seftor RE, Margaryan NV, Strizzi L, Murphy GF, Long GV, Scolyer RA. Targeting melanoma with front-line therapy does not abrogate Nodal-expressing tumor cells. J Transl Med 2017; 97:176-186. [PMID: 27775691 DOI: 10.1038/labinvest.2016.107] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 08/24/2016] [Accepted: 09/06/2016] [Indexed: 01/12/2023] Open
Abstract
Metastatic melanoma is a highly aggressive skin cancer with a poor prognosis. It is the leading cause of skin cancer deaths with a median overall survival for advanced-stage metastatic disease of <6 months. Despite advances in the field with conventional and targeted therapies, the heterogeneity of melanoma poses the greatest ongoing challenge, ultimately leading to relapse and progression to a more drug-resistant tumor in most patients. Particularly noteworthy are recent findings, indicating that these therapies exert selective pressure on tumors resulting in the activation of pathways associated with cancer stem cells that are unresponsive to current therapy. Our previous studies have shown how Nodal, an embryonic morphogen of the transforming growth factor-beta superfamily, is one of these critical factors that is reactivated in aggressive melanoma and resistant to conventional chemotherapy, such as dacarbazine. In the current study, we sought to determine whether BRAF inhibitor (BRAFi) therapy targeted Nodal-expressing tumor cells in uniquely matched unresectable stage III and IV melanoma patient samples before and after therapy that preceded their eventual death due to disease. The results demonstrate that BRAFi treatment failed to affect Nodal levels in melanoma tissues. Accompanying experiments in soft agar and in nude mice showed the advantage of using combinatorial treatment with BRAFi plus anti-Nodal monoclonal antibody to suppress tumor growth and metastasis. These data provide a promising new approach using front-line therapy combined with targeting a cancer stem cell-associated molecule-producing a more efficacious response than monotherapy.
Collapse
Affiliation(s)
- Mary Jc Hendrix
- Department of Biology, Shepherd University, Shepherdstown, WV, USA.,Program in Cancer Biology and Epigenomics, Stanley Manne Children's Research Institute at Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Robert C. Byrd Health Sciences Center, West Virginia University Cancer Institute, West Virginia University, Morgantown, WV, USA
| | - Irawati Kandela
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Evanston, IL, USA
| | - Andrew P Mazar
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Evanston, IL, USA
| | - Elisabeth A Seftor
- Program in Cancer Biology and Epigenomics, Stanley Manne Children's Research Institute at Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Robert C. Byrd Health Sciences Center, West Virginia University Cancer Institute, West Virginia University, Morgantown, WV, USA
| | - Richard Eb Seftor
- Program in Cancer Biology and Epigenomics, Stanley Manne Children's Research Institute at Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Robert C. Byrd Health Sciences Center, West Virginia University Cancer Institute, West Virginia University, Morgantown, WV, USA
| | - Naira V Margaryan
- Program in Cancer Biology and Epigenomics, Stanley Manne Children's Research Institute at Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Robert C. Byrd Health Sciences Center, West Virginia University Cancer Institute, West Virginia University, Morgantown, WV, USA
| | - Luigi Strizzi
- Program in Cancer Biology and Epigenomics, Stanley Manne Children's Research Institute at Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Pathology, Midwestern University, Downers Grove, IL, USA
| | - George F Murphy
- Department of Pathology, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Georgina V Long
- Melanoma Institute Australia and Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia and Sydney Medical School, The University of Sydney, Sydney, NSW, Australia.,Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| |
Collapse
|
25
|
Bodenstine TM, Chandler GS, Seftor REB, Seftor EA, Hendrix MJC. Plasticity underlies tumor progression: role of Nodal signaling. Cancer Metastasis Rev 2016; 35:21-39. [PMID: 26951550 DOI: 10.1007/s10555-016-9605-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The transforming growth factor beta (TGFβ) superfamily member Nodal is an established regulator of early embryonic development, with primary roles in endoderm induction, left-right asymmetry, and primitive streak formation. Nodal signals through TGFβ family receptors at the plasma membrane and induces signaling cascades leading to diverse transcriptional regulation. While conceptually simple, the regulation of Nodal and its molecular effects are profoundly complex and context dependent. Pioneering work by developmental biologists has characterized the signaling pathways, regulatory components, and provided detailed insight into the mechanisms by which Nodal mediates changes at the cellular and organismal levels. Nodal is also an important factor in maintaining pluripotency of embryonic stem cells through regulation of core transcriptional programs. Collectively, this work has led to an appreciation for Nodal as a powerful morphogen capable of orchestrating multiple cellular phenotypes. Although Nodal is not active in most adult tissues, its reexpression and signaling have been linked to multiple types of human cancer, and Nodal has emerged as a driver of tumor growth and cellular plasticity. In vitro and in vivo experimental evidence has demonstrated that inhibition of Nodal signaling reduces cancer cell aggressive characteristics, while clinical data have established associations with Nodal expression and patient outcomes. As a result, there is great interest in the potential targeting of Nodal activity in a therapeutic setting for cancer patients that may provide new avenues for suppressing tumor growth and metastasis. In this review, we evaluate our current understanding of the complexities of Nodal function in cancer and highlight recent experimental evidence that sheds light on the therapeutic potential of its inhibition.
Collapse
Affiliation(s)
- Thomas M Bodenstine
- Stanley Manne Children's Research Institute, Cancer Biology and Epigenomics Program, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 225 E. Chicago Avenue, Box 222, Chicago, IL, 60611, USA
| | - Grace S Chandler
- Stanley Manne Children's Research Institute, Cancer Biology and Epigenomics Program, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 225 E. Chicago Avenue, Box 222, Chicago, IL, 60611, USA
| | - Richard E B Seftor
- Stanley Manne Children's Research Institute, Cancer Biology and Epigenomics Program, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 225 E. Chicago Avenue, Box 222, Chicago, IL, 60611, USA
| | - Elisabeth A Seftor
- Stanley Manne Children's Research Institute, Cancer Biology and Epigenomics Program, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 225 E. Chicago Avenue, Box 222, Chicago, IL, 60611, USA
| | - Mary J C Hendrix
- Stanley Manne Children's Research Institute, Cancer Biology and Epigenomics Program, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 225 E. Chicago Avenue, Box 222, Chicago, IL, 60611, USA.
| |
Collapse
|
26
|
Qi YF, Wu L, Li ZQ, Wu ML, Wang HF, Chan KY, Lu LL, Cai SH, Wang HS, Du J. Nodal signaling modulates the expression of Oct-4 via nuclear translocation of β-catenin in lung and prostate cancer cells. Arch Biochem Biophys 2016; 608:34-41. [PMID: 27592306 DOI: 10.1016/j.abb.2016.07.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 07/02/2016] [Accepted: 07/04/2016] [Indexed: 11/16/2022]
Abstract
Nodal is a member of transforming growth factor beta (TGF-β) superfamily. Nodal promotes the self-renewal of human cancer stem cells (CSCs) and triggers carcinogenesis of human cancers via an autocrine manner through Smad2/3 pathway. In our study, generation of Nodal-overexpressed cancer cells was constructed, and the effect of Nodal on the stem cell marker Oct-4 was evaluated by overexpression or blocked Nodal/ALKs signaling pathway in non-small cell lung cancer cells A549 and prostate cancer cells PC3. Functionally, Nodal also increased the proliferation via the β-catenin nuclear translocation. This increase was attributed to GSK-3β dephosphorylating, and activin receptor-like kinase 4/7 (ALK4/7) played a major role in human cancer cells. Our study provides a positive understanding of Nodal function in cancer cells and suggests a potential novel target for clinical therapeutic research.
Collapse
Affiliation(s)
- Yi-Fei Qi
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, No. 132 Waihuandong Road, University Town, Guangzhou 510006, China
| | - Long Wu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jinan University, Guangzhou 510632, China
| | - Zi-Qian Li
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, No. 132 Waihuandong Road, University Town, Guangzhou 510006, China
| | - Meng-Ling Wu
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, No. 132 Waihuandong Road, University Town, Guangzhou 510006, China
| | - Hai-Fang Wang
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, No. 132 Waihuandong Road, University Town, Guangzhou 510006, China
| | - Ka-Ying Chan
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, No. 132 Waihuandong Road, University Town, Guangzhou 510006, China
| | - Lin-Lin Lu
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, No. 132 Waihuandong Road, University Town, Guangzhou 510006, China
| | - Shao-Hui Cai
- Department of Pharmacology, School of Pharmaceutical Sciences, Jinan University, Guangzhou 510632, China
| | - Hong-Sheng Wang
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, No. 132 Waihuandong Road, University Town, Guangzhou 510006, China.
| | - Jun Du
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, No. 132 Waihuandong Road, University Town, Guangzhou 510006, China.
| |
Collapse
|
27
|
Oliveira-Nunes MC, Assad Kahn S, de Oliveira Barbeitas AL, E Spohr TCLDS, Dubois LGF, Ventura Matioszek GM, Querido W, Campanati L, de Brito Neto JM, Lima FRS, Moura-Neto V, Carneiro K. The availability of the embryonic TGF-β protein Nodal is dynamically regulated during glioblastoma multiforme tumorigenesis. Cancer Cell Int 2016; 16:46. [PMID: 27330409 PMCID: PMC4912793 DOI: 10.1186/s12935-016-0324-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 06/07/2016] [Indexed: 12/19/2022] Open
Abstract
Background Glioblastoma (GBM) is the most common primary brain tumor presenting self-renewing cancer stem cells. The role of these cells on the development of the tumors has been proposed to recapitulate programs from embryogenesis. Recently, the embryonic transforming growth factor-β (TGF-β) protein Nodal has been shown to be reactivated upon tumor development; however, its availability in GBM cells has not been addressed so far. In this study, we investigated by an original approach the mechanisms that dynamically control both intra and extracellular Nodal availability during GBM tumorigenesis. Methods We characterized the dynamics of Nodal availability in both stem and more differentiated GBM cells through morphological analysis, immunofluorescence of Nodal protein and of early (EEA1 and Rab5) and late (Rab7 and Rab11) endocytic markers and Western Blot. Tukey’s test was used to analyze the prevalent correlation of Nodal with different endocytic markers inside specific differentiation states, and Sidak’s multiple comparisons test was used to compare the prevalence of Nodal/endocytic markers co-localization between two differentiation states of GBM cells. Paired t test was used to analyze the abundance of Nodal protein, in extra and intracellular media. Results The cytoplasmic distribution of Nodal was dynamically regulated and strongly correlated with the differentiation status of GBM cells. While Nodal-positive vesicle-like particles were symmetrically distributed in GBM stem cells (GBMsc), they presented asymmetric perinuclear localization in more differentiated GBM cells (mdGBM). Strikingly, when subjected to dedifferentiation, the distribution of Nodal in mdGBM shifted to a symmetric pattern. Moreover, the availability of both intracellular and secreted Nodal were downregulated upon GBMsc differentiation, with cells becoming elongated, negative for Nodal and positive for Nestin. Interestingly, the co-localization of Nodal with endosomal vesicles also depended on the differentiation status of the cells, with Nodal seen more packed in EEA1/Rab5 + vesicles in GBMsc and more in Rab7/11 + vesicles in mdGBM. Conclusions Our results show for the first time that Nodal availability relates to GBM cell differentiation status and that it is dynamically regulated by an endocytic pathway during GBM tumorigenesis, shedding new light on molecular pathways that might emerge as putative targets for Nodal signaling in GBM therapy. Electronic supplementary material The online version of this article (doi:10.1186/s12935-016-0324-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Maria Cecília Oliveira-Nunes
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho 373, F2-01, Rio de Janeiro, Rio de Janeiro 21941-902 Brazil
| | - Suzana Assad Kahn
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, 265 Campus Drive, Stanford, California 94305 USA ; Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho 373, F1-20, Rio de Janeiro, Rio de Janeiro 21941-902 Brazil
| | - Ana Luiza de Oliveira Barbeitas
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho 373, F2-01, Rio de Janeiro, Rio de Janeiro 21941-902 Brazil
| | - Tania Cristina Leite de Sampaio E Spohr
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho 373, F1-20, Rio de Janeiro, Rio de Janeiro 21941-902 Brazil ; Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Rua do Rezende, 156, Rio de Janeiro, Rio de Janeiro 20231-092 Brazil
| | - Luiz Gustavo Feijó Dubois
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho 373, F1-20, Rio de Janeiro, Rio de Janeiro 21941-902 Brazil ; Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Rua do Rezende, 156, Rio de Janeiro, Rio de Janeiro 20231-092 Brazil
| | - Grasiella Maria Ventura Matioszek
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho 373, B1-29, Rio de Janeiro, Rio de Janeiro 21941-902 Brazil
| | - William Querido
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho 373, F2-30, Rio de Janeiro, Rio de Janeiro 21941-902 Brazil
| | - Loraine Campanati
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho 373, F1-20, Rio de Janeiro, Rio de Janeiro 21941-902 Brazil
| | - José Marques de Brito Neto
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho 373, F2-01, Rio de Janeiro, Rio de Janeiro 21941-902 Brazil
| | - Flavia Regina Souza Lima
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho 373, F1-20, Rio de Janeiro, Rio de Janeiro 21941-902 Brazil
| | - Vivaldo Moura-Neto
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho 373, F1-20, Rio de Janeiro, Rio de Janeiro 21941-902 Brazil ; Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Rua do Rezende, 156, Rio de Janeiro, Rio de Janeiro 20231-092 Brazil
| | - Katia Carneiro
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho 373, F2-01, Rio de Janeiro, Rio de Janeiro 21941-902 Brazil
| |
Collapse
|
28
|
Hendrix MJC, Seftor EA, Seftor REB, Chao JT, Chien DS, Chu YW. Tumor cell vascular mimicry: Novel targeting opportunity in melanoma. Pharmacol Ther 2016; 159:83-92. [PMID: 26808163 PMCID: PMC4779708 DOI: 10.1016/j.pharmthera.2016.01.006] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In 1999, the American Journal of Pathology published an article, entitled "Vascular channel formation by human melanoma cells in vivo and in vitro: vasculogenic mimicry" by Maniotis and colleagues, which ignited a spirited debate for several years and earned the journal's distinction of a "citation classic" (Maniotis et al., 1999). Tumor cell vasculogenic mimicry (VM), also known as vascular mimicry, describes the plasticity of aggressive cancer cells forming de novo vascular networks and is associated with the malignant phenotype and poor clinical outcome. The tumor cells capable of VM share the commonality of a stem cell-like, transendothelial phenotype, which may be induced by hypoxia. Since its introduction as a novel paradigm for melanoma tumor perfusion, many studies have contributed new findings illuminating the underlying molecular pathways supporting VM in a variety of tumors, including carcinomas, sarcomas, glioblastomas, astrocytomas, and melanomas. Of special significance is the lack of effectiveness of angiogenesis inhibitors on tumor cell VM, suggesting a selective resistance by this phenotype to conventional therapy. Facilitating the functional plasticity of tumor cell VM are key proteins associated with vascular, stem cell, extracellular matrix, and hypoxia-related signaling pathways--each deserving serious consideration as potential therapeutic targets and diagnostic indicators of the aggressive, metastatic phenotype. This review highlights seminal findings pertinent to VM, including the effects of a novel, small molecular compound, CVM-1118, currently under clinical development to target VM, and illuminates important molecular pathways involved in the suppression of this plastic, aggressive phenotype, using melanoma as a model.
Collapse
Affiliation(s)
- Mary J C Hendrix
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60614, United States; Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States.
| | - Elisabeth A Seftor
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60614, United States
| | - Richard E B Seftor
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60614, United States; Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | | | | | | |
Collapse
|
29
|
Galvão A, Skarzynski D, Ferreira-Dias G. Nodal Promotes Functional Luteolysis via Down-Regulation of Progesterone and Prostaglandins E2 and Promotion of PGF2α Synthetic Pathways in Mare Corpus Luteum. Endocrinology 2016; 157:858-71. [PMID: 26653568 DOI: 10.1210/en.2015-1362] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
In the present work, we investigated the role of Nodal, an embryonic morphogen from the TGFβ superfamily in corpus luteum (CL) secretory activity using cells isolated from equine CL as a model. Expression pattern of Nodal and its receptors activin receptor A type IIB (ACVR2B), activin receptor-like kinase (Alk)-7, and Alk4, as well as the Nodal physiological role, demonstrate the involvement of this pathway in functional luteolysis. Nodal and its receptors were immune localized in small and large luteal cells and endothelial cells, except ACVR2B, which was not detected in the endothelium. Nodal mRNA in situ hybridization confirmed its transcription in steroidogenic and endothelial cells. Expression analysis of the aforementioned factors evidenced that Nodal and Alk7 proteins peaked at the mid-CL (P < .01), the time of luteolysis initiation, whereas Alk4 and ACVR2B proteins increased from mid- to late CL (P < .05). The Nodal treatment of luteal cells decreased progesterone and prostaglandin (PG) E2 concentrations in culture media (P < .05) as well as mRNA and protein of secretory enzymes steroidogenic acute regulatory protein, cholesterol side-chain cleavage enzyme, cytosolic PGE2 synthase, and microsomal PGE2 synthase-1 (P < .05). Conversely, PGF2α secretion and gene expression of PG-endoperoxidase synthase 2 and PGF2α synthase were increased after Nodal treatment (P < .05). Mid-CL cells cultured with PGF2α had increased Nodal protein expression (P < .05) and phosphorylated mothers against decapentaplegic-3 phosphorylation (P < .05). Finally, the supportive interaction between Nodal and PGF2α on luteolysis was shown to its greatest extent because both factors together more significantly inhibited progesterone (P < .05) and promoted PGF2α (P < .05) synthesis than Nodal or PGF2α alone. Our results neatly pinpoint the sites of action of the Nodal signaling pathway toward functional luteolysis in the mare.
Collapse
Affiliation(s)
- António Galvão
- Institute of Animal Reproduction and Food Research (A.G., D.S.), Polish Academy of Sciences, 10-748 Olsztyn, Poland; Faculty of Veterinary Medicine (A.G., G.F.-D.), Centre for Interdisciplinary Research in Animal Health, University of Lisbon, 1300-477 Lisbon, Portugal
| | - Dariusz Skarzynski
- Institute of Animal Reproduction and Food Research (A.G., D.S.), Polish Academy of Sciences, 10-748 Olsztyn, Poland; Faculty of Veterinary Medicine (A.G., G.F.-D.), Centre for Interdisciplinary Research in Animal Health, University of Lisbon, 1300-477 Lisbon, Portugal
| | - Graça Ferreira-Dias
- Institute of Animal Reproduction and Food Research (A.G., D.S.), Polish Academy of Sciences, 10-748 Olsztyn, Poland; Faculty of Veterinary Medicine (A.G., G.F.-D.), Centre for Interdisciplinary Research in Animal Health, University of Lisbon, 1300-477 Lisbon, Portugal
| |
Collapse
|
30
|
Fan Y, Li D, Qian J, Liu Y, Feng H, Li D. Increased expression of FERM domain-containing 4A protein is closely associated with the development of rectal cancer. Exp Ther Med 2015; 11:421-426. [PMID: 26893625 PMCID: PMC4734186 DOI: 10.3892/etm.2015.2933] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Accepted: 11/03/2015] [Indexed: 01/25/2023] Open
Abstract
The aim of the present study was to detect the expression levels of FERM domain-containing 4A (FRMD4A) in rectal cancer tissues and peripheral blood and to investigate the correlation between FRMD4A and cancer development. A total of 78 consecutive patients were enrolled in this study. Thirty healthy individuals were used as the control group. The expression of FRMD4A in rectal cancer and the corresponding normal adjacent tissues was detected by immunohistochemistry and western blotting. The expression of FRMD4A mRNA in peripheral blood was detected by reverse transcription-quantitative polymerase chain reaction. The expression of FRMD4A in rectal cancer tissues was found to be negatively correlated with the degree of differentiation, depth of invasion and Dukes' stage. A negative correlation was identified between FRMD4A and epithelial cadherin expression. The expression of FRMD4A in the peripheral blood of patients with rectal cancer was significantly increased compared with that in the control group (P<0.05). Expression of FRMD4A in the peripheral blood in the patients with lymph node metastasis was significantly increased compared with that in the patients without lymph node metastasis (P<0.05). These results indicate that the expression of FRMD4A is significantly increased in rectal cancer tissues and the peripheral blood of patients with rectal cancer, and the expression levels of FRMD4A are closely associated with differentiation, invasion of rectal cancer and Dukes' stage. In conclusion, the findings of the present study suggest that FRMD4A may be used as a target for the diagnosis and treatment of rectal cancer.
Collapse
Affiliation(s)
- Yongtian Fan
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Dechuan Li
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Jun Qian
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Yong Liu
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Haiyang Feng
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Dechuan Li
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| |
Collapse
|
31
|
Duan W, Li R, Ma J, Lei J, Xu Q, Jiang Z, Nan L, Li X, Wang Z, Huo X, Han L, Wu Z, Wu E, Ma Q. Overexpression of Nodal induces a metastatic phenotype in pancreatic cancer cells via the Smad2/3 pathway. Oncotarget 2015; 6:1490-506. [PMID: 25557170 PMCID: PMC4359309 DOI: 10.18632/oncotarget.2686] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 11/02/2014] [Indexed: 01/05/2023] Open
Abstract
Metastasis is the major cause for the high mortality rate of pancreatic cancer. Human embryonic stem cell (hESC) associated genes frequently correlate with malignant disease progression. Recent studies have demonstrated that the embryonic protein Nodal, which plays a critical role during embryonic development, is re-expressed in several types of tumors and promotes cancers progression. However, little is known about the role of Nodal in pancreatic cancer. Here, we show that Nodal expression is upregulated in human pancreatic cancer tissues. Moreover, Nodal expression levels correlate well with the grade of pancreatic cancer differentiation. In addition, we present clear evidence that Nodal induces signal transduction through the Smad2/3-dependent pathway in vitro. Furthermore, we show that Nodal promotes pancreatic cancer cell migration and invasion, induces epithelial-mesenchymal transition (EMT) and enhances the expression of matrix metalloproteinase-2 (MMP2) and CXC chemokine receptor 4 (CXCR4). Using an in vivo liver metastasis model of pancreatic cancer, we observed that blocking Nodal signaling activity with the small-molecule inhibitor SB431542 decreases the number and size of liver metastases. Taken together, our results suggest that Nodal overexpression induces a metastatic phenotype in pancreatic cancer cells, and that targeting Nodal signaling may be a promising therapeutic strategy for pancreatic cancer.
Collapse
Affiliation(s)
- Wanxing Duan
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China
| | - Rong Li
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China.,Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jiguang Ma
- Department of Oncology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China
| | - Jianjun Lei
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China
| | - Qinhong Xu
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China
| | - Zhengdong Jiang
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China
| | - Ligang Nan
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xuqi Li
- Department of General Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiongwei Huo
- Department of General Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China
| | - Liang Han
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China
| | - Zheng Wu
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China
| | - Erxi Wu
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58105, USA
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
32
|
Ning F, Wang HF, Guo Q, Liu ZC, Li ZQ, Du J. Expression and significance of Nodal in human cancers: a meta-analysis. Int J Clin Exp Med 2015; 8:20227-20235. [PMID: 26884935 PMCID: PMC4723780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 10/27/2015] [Indexed: 06/05/2023]
Abstract
Cancer is a global and growing problem. Nodal, which has been showed to be involved in occurrence and development of cancers, is an important embryonic morphogen. The aim of this study was to evaluate the significance of Nodal expression in human cancers based on the published related articles. Online databases were searched to retrieve relevant articles published between 2000 and 2015. The odds ratio (OR) with its 95% confident intervals (CI) were employed to calculate the strength of significance. Finally, a total of 11 articles were screened out, including 801 cancer patients and 372 healthy controls. Nine kinds of cancers were contained, and Nodal was detected in 56.7% of all participants (665/1173). Overall, our result found that Nodal was highly expressed in cancer patients than that in healthy controls, indicating that Nodal expression was significantly associated with cancers progression (OR=21.72, 95% CI=9.94-47.46, P<0.00001). Subgroup analysis showed that Nodal expression was significantly corrected with high WHO grade of human cancers (III+IV versus I+II: OR=2.46, 95% CI=1.63-3.71, P<0.00001). This significant relationship was also found in tumor size, differentiation degree, not observed in gender, age and lymphatic metastasis status of patients with all studied cancers in this meta-analysis. In conclusion, our results demonstrated that Nodal might be implicated in cancer progression, suggesting that it was a potential biomarker and therapeutic target for cancers.
Collapse
Affiliation(s)
- Fen Ning
- Guangzhou Institute of Pediatrics, Department of Obstetrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical UniversityGuangzhou 510623, Guangdong, China
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen UniversityGuangzhou 510006, Guangdong, China
| | - Hai-Fang Wang
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen UniversityGuangzhou 510006, Guangdong, China
| | - Qiang Guo
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen UniversityGuangzhou 510006, Guangdong, China
- School of Pharmaceutical Sciences, Wenzhou Medical UniversityWenzhou 325035, Zhejiang, China
| | - Zong-Cai Liu
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen UniversityGuangzhou 510006, Guangdong, China
- Laboratory of Endocrinology and Metabolism, Guangzhou Women and Children’s Medical Center, Guangzhou Medical UniversityGuangzhou 510623, Guangdong, China
| | - Zi-Qian Li
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen UniversityGuangzhou 510006, Guangdong, China
| | - Jun Du
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen UniversityGuangzhou 510006, Guangdong, China
| |
Collapse
|
33
|
Kok DEG, Dhonukshe-Rutten RAM, Lute C, Heil SG, Uitterlinden AG, van der Velde N, van Meurs JBJ, van Schoor NM, Hooiveld GJEJ, de Groot LCPGM, Kampman E, Steegenga WT. The effects of long-term daily folic acid and vitamin B12 supplementation on genome-wide DNA methylation in elderly subjects. Clin Epigenetics 2015; 7:121. [PMID: 26568774 PMCID: PMC4644301 DOI: 10.1186/s13148-015-0154-5] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 11/04/2015] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Folate and its synthetic form folic acid function as donor of one-carbon units and have been, together with other B-vitamins, implicated in programming of epigenetic processes such as DNA methylation during early development. To what extent regulation of DNA methylation can be altered via B-vitamins later in life, and how this relates to health and disease, is not exactly known. The aim of this study was to identify effects of long-term supplementation with folic acid and vitamin B12 on genome-wide DNA methylation in elderly subjects. This project was part of a randomized, placebo-controlled trial on effects of supplemental intake of folic acid and vitamin B12 on bone fracture incidence (B-vitamins for the PRevention Of Osteoporotic Fractures (B-PROOF) study). Participants with mildly elevated homocysteine levels, aged 65-75 years, were randomly assigned to take 400 μg folic acid and 500 μg vitamin B12 per day or a placebo during an intervention period of 2 years. DNA was isolated from buffy coats, collected before and after intervention, and genome-wide DNA methylation was determined in 87 participants (n = 44 folic acid/vitamin B12, n = 43 placebo) using the Infinium HumanMethylation450 BeadChip. RESULTS After intervention with folic acid and vitamin B12, 162 (versus 14 in the placebo group) of the 431,312 positions were differentially methylated as compared to baseline. Comparisons of the DNA methylation changes in the participants receiving folic acid and vitamin B12 versus placebo revealed one single differentially methylated position (cg19380919) with a borderline statistical significance. However, based on the analyses of differentially methylated regions (DMRs) consisting of multiple positions, we identified 6 regions that differed statistically significantly between the intervention and placebo group. Pronounced changes were found for regions in the DIRAS3, ARMC8, and NODAL genes, implicated in carcinogenesis and early embryonic development. Furthermore, serum levels of folate and vitamin B12 or plasma homocysteine were related to DNA methylation of 173, 425, and 11 regions, respectively. Interestingly, for several members of the developmental HOX genes, DNA methylation was related to serum levels of folate. CONCLUSIONS Long-term supplementation with folic acid and vitamin B12 in elderly subjects resulted in effects on DNA methylation of several genes, among which genes implicated in developmental processes.
Collapse
Affiliation(s)
- Dieuwertje E G Kok
- Division of Human Nutrition, Wageningen University, PO Box 8129, 6700 EV Wageningen, The Netherlands
| | | | - Carolien Lute
- Division of Human Nutrition, Wageningen University, PO Box 8129, 6700 EV Wageningen, The Netherlands
| | - Sandra G Heil
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - André G Uitterlinden
- Genetic Laboratory Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands ; Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Nathalie van der Velde
- Genetic Laboratory Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands ; Department of Internal Medicine, Section of Geriatrics, Academic Medical Center, Amsterdam, The Netherlands
| | - Joyce B J van Meurs
- Genetic Laboratory Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Natasja M van Schoor
- Department of Epidemiology and Biostatistics, EMGO Institute for Health and Care Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Guido J E J Hooiveld
- Division of Human Nutrition, Wageningen University, PO Box 8129, 6700 EV Wageningen, The Netherlands
| | - Lisette C P G M de Groot
- Division of Human Nutrition, Wageningen University, PO Box 8129, 6700 EV Wageningen, The Netherlands
| | - Ellen Kampman
- Division of Human Nutrition, Wageningen University, PO Box 8129, 6700 EV Wageningen, The Netherlands
| | - Wilma T Steegenga
- Division of Human Nutrition, Wageningen University, PO Box 8129, 6700 EV Wageningen, The Netherlands
| |
Collapse
|
34
|
Strizzi L, Sandomenico A, Margaryan NV, Focà A, Sanguigno L, Bodenstine TM, Chandler GS, Reed DW, Gilgur A, Seftor EA, Seftor RE, Khalkhali-Ellis Z, Leonardi A, Ruvo M, Hendrix MJ. Effects of a novel Nodal-targeting monoclonal antibody in melanoma. Oncotarget 2015; 6:34071-86. [PMID: 26460952 PMCID: PMC4741437 DOI: 10.18632/oncotarget.6049] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 09/24/2015] [Indexed: 12/31/2022] Open
Abstract
Nodal is highly expressed in various human malignancies, thus supporting the rationale for exploring Nodal as a therapeutic target. Here, we describe the effects of a novel monoclonal antibody (mAb), 3D1, raised against human Nodal. In vitro treatment of C8161 human melanoma cells with 3D1 mAb shows reductions in anchorage-independent growth and vasculogenic network formation. 3D1 treated cells also show decreases of Nodal and downstream signaling molecules, P-Smad2 and P-ERK and of P-H3 and CyclinB1, with an increase in p27. Similar effects were previously reported in human breast cancer cells where Nodal expression was generally down-regulated; following 3D1 mAb treatment, both Nodal and P-H3 levels are reduced. Noteworthy is the reduced growth of human melanoma xenografts in Nude mice treated with 3D1 mAb, where immunostaining of representative tumor sections show diminished P-Smad2 expression. Similar effects both in vitro and in vivo were observed in 3D1 treated A375SM melanoma cells harboring the active BRAF(V600E) mutation compared to treatments with IgG control or a BRAF inhibitor, dabrafenib. Finally, we describe a 3D1-based ELISA for the detection of Nodal in serum samples from cancer patients. These data suggest the potential of 3D1 mAb for selecting and targeting Nodal expressing cancers.
Collapse
Affiliation(s)
- Luigi Strizzi
- Cancer Biology and Epigenomics Program, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Annamaria Sandomenico
- Istituto di Biostrutture e Bioimmagini del CNR and CIRPeB, Università Federico II di Napoli, Naples, Italy
| | - Naira V. Margaryan
- Cancer Biology and Epigenomics Program, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Annalia Focà
- Istituto di Biostrutture e Bioimmagini del CNR and CIRPeB, Università Federico II di Napoli, Naples, Italy
| | - Luca Sanguigno
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II di Napoli, Naples, Italy
| | - Thomas M. Bodenstine
- Cancer Biology and Epigenomics Program, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Grace S. Chandler
- Cancer Biology and Epigenomics Program, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - David W. Reed
- Cancer Biology and Epigenomics Program, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Alina Gilgur
- Cancer Biology and Epigenomics Program, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Elisabeth A. Seftor
- Cancer Biology and Epigenomics Program, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Richard E.B. Seftor
- Cancer Biology and Epigenomics Program, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Zhila Khalkhali-Ellis
- Cancer Biology and Epigenomics Program, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Antonio Leonardi
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II di Napoli, Naples, Italy
| | - Menotti Ruvo
- Istituto di Biostrutture e Bioimmagini del CNR and CIRPeB, Università Federico II di Napoli, Naples, Italy
| | - Mary J.C. Hendrix
- Cancer Biology and Epigenomics Program, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
35
|
New Anti-Nodal Monoclonal Antibodies Targeting the Nodal Pre-Helix Loop Involved in Cripto-1 Binding. Int J Mol Sci 2015; 16:21342-62. [PMID: 26370966 PMCID: PMC4613256 DOI: 10.3390/ijms160921342] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 08/21/2015] [Accepted: 08/27/2015] [Indexed: 12/25/2022] Open
Abstract
Nodal is a potent embryonic morphogen belonging to the TGF-β superfamily. Typically, it also binds to the ALK4/ActRIIB receptor complex in the presence of the co-receptor Cripto-1. Nodal expression is physiologically restricted to embryonic tissues and human embryonic stem cells, is absent in normal cells but re-emerges in several human cancers, including melanoma, breast, and colon cancer. Our aim was to obtain mAbs able to recognize Nodal on a major CBR (Cripto-Binding-Region) site and to block the Cripto-1-mediated signalling. To achieve this, antibodies were raised against hNodal(44-67) and mAbs generated by the hybridoma technology. We have selected one mAb, named 3D1, which strongly associates with full-length rhNodal (KD 1.4 nM) and recognizes the endogenous protein in a panel of human melanoma cell lines by western blot and FACS analyses. 3D1 inhibits the Nodal-Cripto-1 binding and blocks Smad2/3 phosphorylation. Data suggest that inhibition of the Nodal-Cripto-1 axis is a valid therapeutic approach against melanoma and 3D1 is a promising and interesting agent for blocking Nodal-Cripto mediated tumor development. These findings increase the interest for Nodal as both a diagnostic and prognostic marker and as a potential new target for therapeutic intervention.
Collapse
|
36
|
Hardy KM, Strizzi L, Margaryan NV, Gupta K, Murphy GF, Scolyer RA, Hendrix MJC. Targeting nodal in conjunction with dacarbazine induces synergistic anticancer effects in metastatic melanoma. Mol Cancer Res 2015; 13:670-80. [PMID: 25767211 DOI: 10.1158/1541-7786.mcr-14-0077] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 01/05/2015] [Indexed: 12/11/2022]
Abstract
UNLABELLED Metastatic melanoma is a highly aggressive skin cancer with a poor prognosis. Despite a complete response in fewer than 5% of patients, the chemotherapeutic agent dacarbazine (DTIC) remains the reference drug after almost 40 years. More recently, FDA-approved drugs have shown promise but patient outcome remains modest, predominantly due to drug resistance. As such, combinatorial targeting has received increased attention, and will advance with the identification of new molecular targets. One attractive target for improving melanoma therapy is the growth factor Nodal, whose normal expression is largely restricted to embryonic development, but is reactivated in metastatic melanoma. In this study, we sought to determine how Nodal-positive human melanoma cells respond to DTIC treatment and to ascertain whether targeting Nodal in combination with DTIC would be more effective than monotherapy. A single treatment with DTIC inhibited cell growth but did not induce apoptosis. Rather than reducing Nodal expression, DTIC increased the size of the Nodal-positive subpopulation, an observation coincident with increased cellular invasion. Importantly, clinical tissue specimens from patients with melanomas refractory to DTIC therapy stained positive for Nodal expression, both in pre- and post-DTIC tumors, underscoring the value of targeting Nodal. In vitro, anti-Nodal antibodies alone had some adverse effects on proliferation and apoptosis, but combining DTIC treatment with anti-Nodal antibodies decreased cell growth and increased apoptosis synergistically, at concentrations incapable of producing meaningful effects as monotherapy. IMPLICATIONS Targeting Nodal in combination with DTIC therapy holds promise for the treatment of metastatic melanoma.
Collapse
Affiliation(s)
- Katharine M Hardy
- Program in Cancer Biology and Epigenomics, Stanley Manne Children's Research Institute at Ann and Robert H. Lurie Children's Hospital of Chicago, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Luigi Strizzi
- Program in Cancer Biology and Epigenomics, Stanley Manne Children's Research Institute at Ann and Robert H. Lurie Children's Hospital of Chicago, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Naira V Margaryan
- Program in Cancer Biology and Epigenomics, Stanley Manne Children's Research Institute at Ann and Robert H. Lurie Children's Hospital of Chicago, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Kanika Gupta
- Program in Cancer Biology and Epigenomics, Stanley Manne Children's Research Institute at Ann and Robert H. Lurie Children's Hospital of Chicago, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois. Howard Hughes Medical Institute NU Bioscientist Program, Weinberg College of Arts and Sciences, Northwestern University, Evanston, Illinois
| | - George F Murphy
- Department of Pathology, Harvard Medical School, Brigham & Women's Hospital, Boston, Massachusetts
| | - Richard A Scolyer
- Melanoma Institute Australia; Sydney Medical School, The University of Sydney; and Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Mary J C Hendrix
- Program in Cancer Biology and Epigenomics, Stanley Manne Children's Research Institute at Ann and Robert H. Lurie Children's Hospital of Chicago, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
37
|
Kong B, Wang W, Esposito I, Friess H, Michalski CW, Kleeff J. Increased expression of Nodal correlates with reduced patient survival in pancreatic cancer. Pancreatology 2015; 15:156-61. [PMID: 25708930 DOI: 10.1016/j.pan.2015.02.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 01/12/2015] [Accepted: 02/01/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND Nodal (nodal growth differentiation factor) and its inhibitor Lefty (left right determination factor), which are ligands of the TGF (transforming growth factor) β superfamily, are responsible for the determination of left-right asymmetry in vertebrates. Nodal/Lefty signaling has been suggested to play a role in the development of metastatic melanoma and breast cancer. However, it remains unclear whether this pathway is also involved in human pancreatic ductal adenocarcinoma (PDAC). METHODS Pancreatic cancer patient specimens with clinical data (n = 54) were used to investigate the clinical significance of Nodal-Lefty signaling. A set of in vitro assays were carried out in a human pancreatic cancer cell line (Colo-357) to assess the functional relevance of Nodal-Lefty signaling. RESULTS Nodal was absent in the human normal pancreas, while Lefty was present in islet cells. Though Nodal and Lefty expression were found in cancer cells at various expression levels, the cancer-associated tubular complexes were particularly positive for Lefty. Survival analysis revealed that high expression of Nodal correlated with reduced patient survival (median survival 17.8 vs 33.0 months, p = 0.013). Cultured pancreatic cancer cell lines expressed Nodal and Lefty at different levels. In vitro functional assays revealed that treatment with human recombinant Nodal inhibited cell growth and increased invasion of Colo-357 pancreatic cancer cells whereas no effect was found upon treatment with recombinant Lefty. CONCLUSION Nodal-Lefty signaling might be involved in the pathogenesis of PDAC as Nodal expression marks a subtype of PDAC with unfavorable prognosis.
Collapse
Affiliation(s)
- Bo Kong
- Department of Surgery, Technische Universität München, Munich, Germany
| | - Weibin Wang
- Department of Surgery, Technische Universität München, Munich, Germany; Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Irene Esposito
- Institute of Pathology, Technische Universität München, Munich, Germany
| | - Helmut Friess
- Department of Surgery, Technische Universität München, Munich, Germany
| | | | - Jörg Kleeff
- Department of Surgery, Technische Universität München, Munich, Germany.
| |
Collapse
|
38
|
Human Cerberus prevents nodal-receptor binding, inhibits nodal signaling, and suppresses nodal-mediated phenotypes. PLoS One 2015; 10:e0114954. [PMID: 25603319 PMCID: PMC4300205 DOI: 10.1371/journal.pone.0114954] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 11/15/2014] [Indexed: 02/06/2023] Open
Abstract
The Transforming Growth Factor-ß (TGFß) family ligand Nodal is an essential embryonic morphogen that is associated with progression of breast and other cancers. It has therefore been suggested that Nodal inhibitors could be used to treat breast cancers where Nodal plays a defined role. As secreted antagonists, such as Cerberus, tightly regulate Nodal signaling during embryonic development, we undertook to produce human Cerberus, characterize its biochemical activities, and determine its effect on human breast cancer cells. Using quantitative methods, we investigated the mechanism of Nodal signaling, we evaluated binding of human Cerberus to Nodal and other TGFß family ligands, and we characterized the mechanism of Nodal inhibition by Cerberus. Using cancer cell assays, we examined the ability of Cerberus to suppress aggressive breast cancer cell phenotypes. We found that human Cerberus binds Nodal with high affinity and specificity, blocks binding of Nodal to its signaling partners, and inhibits Nodal signaling. Moreover, we showed that Cerberus profoundly suppresses migration, invasion, and colony forming ability of Nodal expressing and Nodal supplemented breast cancer cells. Taken together, our studies provide mechanistic insights into Nodal signaling and Nodal inhibition with Cerberus and highlight the potential value of Cerberus as anti-Nodal therapeutic.
Collapse
|
39
|
Calvanese L, Sandomenico A, Caporale A, Focà A, Focà G, D'Auria G, Falcigno L, Ruvo M. Conformational features and binding affinities to Cripto, ALK7 and ALK4 of Nodal synthetic fragments. J Pept Sci 2015; 21:283-93. [PMID: 25588905 DOI: 10.1002/psc.2733] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 12/03/2014] [Accepted: 12/03/2014] [Indexed: 12/31/2022]
Abstract
Nodal, a member of the TGF-β superfamily, is a potent embryonic morphogen also implicated in tumor progression. As for other TGF-βs, it triggers the signaling functions through the interaction with the extracellular domains of type I and type II serine/threonine kinase receptors and with the co-receptor Cripto. Recently, we reported the molecular models of Nodal in complex with its type I receptors (ALK4 and ALK7) as well as with Cripto, as obtained by homology modeling and docking simulations. From such models, potential binding epitopes have been identified. To validate such hypotheses, a series of mutated Nodal fragments have been synthesized. These peptide analogs encompass residues 44-67 of the Nodal protein, corresponding to the pre-helix loop and the H3 helix, and reproduce the wild-type sequence or bear some modifications to evaluate the hot-spot role of modified residues in the receptor binding. Here, we show the structural characterization in solution by CD and NMR of the Nodal peptides and the measurement of binding affinity toward Cripto by surface plasmon resonance. Data collected by both conformational analyses and binding measurements suggest a role for Y58 of Nodal in the recognition with Cripto and confirm that previously reported for E49 and E50. Surface plasmon resonance binding assays with recombinant proteins show that Nodal interacts in vitro also with ALK7 and ALK4 and preliminary data, generated using the Nodal synthetic fragments, suggest that Y58 of Nodal may also be involved in the recognition with these protein partners.
Collapse
Affiliation(s)
- Luisa Calvanese
- CIRPeB, University of Naples Federico II, via Mezzocannone, 16, 80134, Napoli, Italy
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Arai D, Hayakawa K, Ohgane J, Hirosawa M, Nakao Y, Tanaka S, Shiota K. An epigenetic regulatory element of the Nodal gene in the mouse and human genomes. Mech Dev 2014; 136:143-54. [PMID: 25528267 DOI: 10.1016/j.mod.2014.12.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 12/12/2014] [Accepted: 12/15/2014] [Indexed: 01/28/2023]
Abstract
Nodal signaling plays critical roles during embryonic development. The Nodal gene is not expressed in adult tissues but is frequently activated in cancer cells, contributing to progression toward malignancy. Although several regulatory elements of the Nodal gene have been identified, the epigenetic mechanisms by which Nodal expression is regulated over the long term remain unclear. We found a region exhibiting dynamic changes in DNA methylation at approximately -3.0 kb to -0.4 kb upstream from the transcriptional start site (TSS) that we termed the epigenetic regulatory element (ERE). The ERE was unmethylated in mouse embryonic stem cells (mESCs) but became increasingly methylated in differentiated cells and tissues, concomitant with the downregulation of Nodal mRNA expression. In vitro reporter assays identified an Oct3/4 binding motif within the ERE, indicating that the ERE is responsible for the activation of Nodal in mESCs. Furthermore, the ERE was a target of differentiation-associated Polycomb silencing, and the chromatin condensed when mESCs differentiated to embryoid bodies (EBs). Pharmacological inhibition of PRC2 led to the reactivation of Nodal expression in EBs and mouse embryonic fibroblasts (MEFs). The ERE was also targeted by PRC2 in normal human cells. In NODAL-expressing human cancer cells, accumulation of EZH2 and trimethylation of H3K27 at the ERE were diminished. In conclusion, Nodal is epigenetically controlled through the ERE in the mouse embryo and human cells.
Collapse
Affiliation(s)
- Daisuke Arai
- Laboratory of Cellular Biochemistry, Department of Animal Resource Sciences/Veterinary Medical Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; Laboratory of Chemical Biology, Department of Chemistry and Biochemistry, School of Advanced Science and Engineering, Waseda University, 3-4-1 Ohkubo, Shinjuku-ku, Tokyo 169-8555, Japan
| | - Koji Hayakawa
- Laboratory of Cellular Biochemistry, Department of Animal Resource Sciences/Veterinary Medical Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Jun Ohgane
- Laboratory of Genomic Function Engineering, Department of Life Sciences, School of Agriculture, Meiji University, 1-1-1 Higashi-mita, Tama-ku, Kawasaki 214-8571, Japan
| | - Mitsuko Hirosawa
- Laboratory of Cellular Biochemistry, Department of Animal Resource Sciences/Veterinary Medical Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Yoichi Nakao
- Laboratory of Chemical Biology, Department of Chemistry and Biochemistry, School of Advanced Science and Engineering, Waseda University, 3-4-1 Ohkubo, Shinjuku-ku, Tokyo 169-8555, Japan
| | - Satoshi Tanaka
- Laboratory of Cellular Biochemistry, Department of Animal Resource Sciences/Veterinary Medical Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Kunio Shiota
- Laboratory of Cellular Biochemistry, Department of Animal Resource Sciences/Veterinary Medical Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan.
| |
Collapse
|
41
|
Lefty inhibits glioma growth by suppressing Nodal-activated Smad and ERK1/2 pathways. J Neurol Sci 2014; 347:137-42. [DOI: 10.1016/j.jns.2014.09.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 09/13/2014] [Accepted: 09/15/2014] [Indexed: 01/01/2023]
|
42
|
Khalkhali-Ellis Z, Kirschmann DA, Seftor EA, Gilgur A, Bodenstine TM, Hinck AP, Hendrix MJC. Divergence(s) in nodal signaling between aggressive melanoma and embryonic stem cells. Int J Cancer 2014; 136:E242-51. [PMID: 25204799 DOI: 10.1002/ijc.29198] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 08/04/2014] [Accepted: 08/12/2014] [Indexed: 12/21/2022]
Abstract
The significant role of the embryonic morphogen Nodal in maintaining the pluripotency of embryonic stem cells is well documented. Interestingly, the recent discovery of Nodal's re-expression in several aggressive and metastatic cancers has highlighted its critical role in self renewal and maintenance of the stem cell-like characteristics of tumor cells, such as melanoma. However, the key TGFβ/Nodal signaling component(s) governing Nodal's effects in metastatic melanoma remain mostly unknown. By employing receptor profiling at the mRNA and protein level(s), we made the novel discovery that embryonic stem cells and metastatic melanoma cells share a similar repertoire of Type I serine/threonine kinase receptors, but diverge in their Type II receptor expression. Ligand:receptor crosslinking and native gel binding assays indicate that metastatic melanoma cells employ the heterodimeric TGFβ receptor I/TGFβ receptor II (TGFβRI/TGFβRII) for signal transduction, whereas embryonic stem cells use the Activin receptors I and II (ACTRI/ACTRII). This unexpected receptor usage by tumor cells was tested by: neutralizing antibody to block its function; and transfecting the dominant negative receptor to compete with the endogenous receptor for ligand binding. Furthermore, a direct biological role for TGFβRII was found to underlie vasculogenic mimicry (VM), an endothelial phenotype contributing to vascular perfusion and associated with the functional plasticity of aggressive melanoma. Collectively, these findings reveal the divergence in Nodal signaling between embryonic stem cells and metastatic melanoma that can impact new therapeutic strategies targeting the re-emergence of embryonic pathways.
Collapse
Affiliation(s)
- Zhila Khalkhali-Ellis
- Cancer Biology and Epigenomics Program, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL; Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL
| | | | | | | | | | | | | |
Collapse
|
43
|
Kirsammer G, Strizzi L, Margaryan NV, Gilgur A, Hyser M, Atkinson J, Kirschmann DA, Seftor EA, Hendrix MJC. Nodal signaling promotes a tumorigenic phenotype in human breast cancer. Semin Cancer Biol 2014; 29:40-50. [PMID: 25073112 DOI: 10.1016/j.semcancer.2014.07.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 07/17/2014] [Indexed: 11/19/2022]
Abstract
The Ras-ERK pathway is deregulated in approximately a third of human cancers, particularly those of epithelial origin. In aggressive, triple-negative, basal-like breast cancers, most tumors display increased MEK and ERK phosphorylation and exhibit a gene expression profile characteristic of Kras or EGFR mutant tumors; however, Ras family genetic mutations are uncommon in triple-negative breast cancer and EGFR mutations account for only a subset of these tumors. Therefore, the upstream events that activate MAPK signaling and promote tumor aggression in triple-negative breast cancers remain poorly defined. We have previously shown that a secreted TGF-β family signaling ligand, Nodal, is expressed in breast cancer in correlation with disease progression. Here we highlight key findings demonstrating that Nodal is required in aggressive human breast cancer cells to activate ERK signaling and downstream tumorigenic phenotypes both in vitro and in vivo. Experimental knockdown of Nodal signaling downregulates ERK activity, resulting in loss of c-myc, upregulation of p27, G1 cell cycle arrest, increased apoptosis and decreased tumorigenicity. The data suggest that ERK activation by Nodal signaling regulates c-myc and p27 proteins post-translationally and that this cascade is essential for aggressive breast tumor behavior in vivo. As the MAPK pathway is an important target for treating triple-negative breast cancers, upstream Nodal signaling may represent a promising target for breast cancer diagnosis and combined therapies aimed at blocking ERK pathway activation.
Collapse
Affiliation(s)
- Gina Kirsammer
- Cancer Biology and Epigenomics Program, Ann and Robert H. Lurie Children's Hospital of Chicago Research Center, 2430 N Halsted St., Chicago, IL 60614, United States
| | - Luigi Strizzi
- Cancer Biology and Epigenomics Program, Ann and Robert H. Lurie Children's Hospital of Chicago Research Center, 2430 N Halsted St., Chicago, IL 60614, United States; Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States
| | - Naira V Margaryan
- Cancer Biology and Epigenomics Program, Ann and Robert H. Lurie Children's Hospital of Chicago Research Center, 2430 N Halsted St., Chicago, IL 60614, United States
| | - Alina Gilgur
- Cancer Biology and Epigenomics Program, Ann and Robert H. Lurie Children's Hospital of Chicago Research Center, 2430 N Halsted St., Chicago, IL 60614, United States
| | - Matthew Hyser
- Presence Saint Francis Hospital, 355 Ridge Ave, Evanston, IL 60202, United States
| | - Janis Atkinson
- Presence Saint Francis Hospital, 355 Ridge Ave, Evanston, IL 60202, United States
| | - Dawn A Kirschmann
- Cancer Biology and Epigenomics Program, Ann and Robert H. Lurie Children's Hospital of Chicago Research Center, 2430 N Halsted St., Chicago, IL 60614, United States
| | - Elisabeth A Seftor
- Cancer Biology and Epigenomics Program, Ann and Robert H. Lurie Children's Hospital of Chicago Research Center, 2430 N Halsted St., Chicago, IL 60614, United States
| | - Mary J C Hendrix
- Cancer Biology and Epigenomics Program, Ann and Robert H. Lurie Children's Hospital of Chicago Research Center, 2430 N Halsted St., Chicago, IL 60614, United States; Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States.
| |
Collapse
|
44
|
Seftor EA, Seftor REB, Weldon D, Kirsammer GT, Margaryan NV, Gilgur A, Hendrix MJC. Melanoma tumor cell heterogeneity: a molecular approach to study subpopulations expressing the embryonic morphogen nodal. Semin Oncol 2014; 41:259-266. [PMID: 24787297 DOI: 10.1053/j.seminoncol.2014.02.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
As the frequency of melanoma increases, current treatment strategies are struggling to significantly impact patient survival. One of the critical issues in designing efficient therapies is understanding the composition of heterogeneous melanoma tumors in order to target cancer stem cells (CSCs) and drug-resistant subpopulations. In this review, we summarize recent findings pertinent to the reemergence of the embryonic Nodal signaling pathway in melanoma and its significance as a prognostic biomarker and therapeutic target. In addition, we offer a novel molecular approach to studying the functional relevance of Nodal-expressing subpopulations and their CSC phenotype.
Collapse
Affiliation(s)
- Elisabeth A Seftor
- Cancer Biology and Epigenomics Program, Ann and Robert H. Lurie Children's Hospital of Chicago Research Center, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine
| | - Richard E B Seftor
- Cancer Biology and Epigenomics Program, Ann and Robert H. Lurie Children's Hospital of Chicago Research Center, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine
| | | | - Gina T Kirsammer
- Cancer Biology and Epigenomics Program, Ann and Robert H. Lurie Children's Hospital of Chicago Research Center, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine
| | - Naira V Margaryan
- Cancer Biology and Epigenomics Program, Ann and Robert H. Lurie Children's Hospital of Chicago Research Center, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine
| | - Alina Gilgur
- Cancer Biology and Epigenomics Program, Ann and Robert H. Lurie Children's Hospital of Chicago Research Center, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine
| | - Mary J C Hendrix
- Cancer Biology and Epigenomics Program, Ann and Robert H. Lurie Children's Hospital of Chicago Research Center, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine
| |
Collapse
|
45
|
Age-Dependent Association between Protein Expression of the Embryonic Stem Cell Marker Cripto-1 and Survival of Glioblastoma Patients. Transl Oncol 2013; 6:732-41. [PMID: 24466376 DOI: 10.1593/tlo.13427] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 08/13/2013] [Accepted: 08/18/2013] [Indexed: 12/26/2022] Open
Abstract
Exploring the re-emergence of embryonic signaling pathways may reveal important information for cancer biology. Nodal is a transforming growth factor-β (TGF-β)-related morphogen that plays a critical role during embryonic development. Nodal signaling is regulated by the Cripto-1 co-receptor and another TGF-β member, Lefty. Although these molecules are poorly detected in differentiated tissues, they have been found in different human cancers. Poor prognosis of glioblastomas justifies the search for novel signaling pathways that can be exploited as potential therapeutic targets. Because our intracranial glioblastoma rat xenograft model has revealed importance of gene ontology categories related to development and differentiation, we hypothesized that increased activity of Nodal signaling could be found in glioblastomas. We examined the gene expressions of Nodal, Cripto-1, and Lefty in microarrays of invasive and angiogenic xenograft samples developed from four patients with glioblastoma. Protein expression was evaluated by immunohistochemistry in 199 primary glioblastomas, and expression levels were analyzed for detection of correlations with available clinical information. Gene expression of Nodal, Lefty, and Cripto-1 was detected in the glioblastoma xenografts. Most patient samples showed significant levels of Cripto-1 detected by immunohistochemistry, whereas only weak to moderate levels were detected for Nodal and Lefty. Most importantly, the higher Cripto-1 scores were associated with shorter survival in a subset of younger patients. These findings suggest for the first time that Cripto-1, an important molecule in developmental biology, may represent a novel prognostic marker and therapeutic target in categories of younger patients with glioblastoma.
Collapse
|
46
|
Wielscher M, Liou W, Pulverer W, Singer CF, Rappaport-Fuerhauser C, Kandioler D, Egger G, Weinhäusel A. Cytosine 5-Hydroxymethylation of the LZTS1 Gene Is Reduced in Breast Cancer. Transl Oncol 2013; 6:715-21. [PMID: 24466374 PMCID: PMC3890706 DOI: 10.1593/tlo.13523] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 09/06/2013] [Accepted: 09/09/2013] [Indexed: 02/07/2023] Open
Abstract
Change of DNA cytosine methylation (5mC) is an early event in the development of cancer, and the recent discovery of a 5-hydroxymethylated form (5hmC) of cytosine suggests a regulatory epigenetic role that might be different from 5-methylcytosine. Here, we aimed at elucidating the role of 5hmC in breast cancer. To interrogate the 5hmC levels of the leucine zipper, putative tumor suppressor 1 (LZTS1) gene in detail, we analyzed 75 primary breast cancer tissue samples from initial diagnosis and 12 normal breast tissue samples derived from healthy persons. Samples were subjected to 5hmC glucosyltransferase treatment followed by restriction digestion and segment-specific amplification of 11 polymerase chain reaction products. Nine of the 11 5'LZTS1 fragments showed significantly lower (fold change of 1.61-6.01, P < .05) 5hmC content in primary breast cancer tissue compared to normal breast tissue samples. No significant differences were observed for 5mC DNA methylation. Furthermore, both LZTS1 and TET1 mRNA expressions were significantly reduced in tumor samples (n = 75, P < .001, Student's t test), which correlated significantly with 5hmC levels in samples. 5hmC levels in breast cancer tissues were associated with unfavorable histopathologic parameters such as lymph node involvement (P < .05, Student's t test). A decrease of 5hmC levels of LZTS1, a classic tumor suppressor gene known to influence metastasis in breast cancer progression, is correlated to down-regulation of LZTS1 mRNA expression in breast cancer and might epigenetically enhance carcinogenesis. The study provides support for the novel hypothesis that suggests a strong influence of 5hmC on mRNA expression. Finally, one may also consider 5hmC as a new biomarker.
Collapse
Affiliation(s)
- Matthias Wielscher
- Molecular Diagnostics Unit, Health and Environment Department, Austrian Institute of Technology, Vienna, Austria
| | - Willy Liou
- Molecular Diagnostics Unit, Health and Environment Department, Austrian Institute of Technology, Vienna, Austria
| | - Walter Pulverer
- Molecular Diagnostics Unit, Health and Environment Department, Austrian Institute of Technology, Vienna, Austria
| | - Christian F Singer
- Department of Obstetrics and Gynecology and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | | | | | - Gerda Egger
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Andreas Weinhäusel
- Molecular Diagnostics Unit, Health and Environment Department, Austrian Institute of Technology, Vienna, Austria
| |
Collapse
|
47
|
Wang XF, Wang HS, Zhang F, Guo Q, Wang H, Wang KF, Zhang G, Bu XZ, Cai SH, Du J. Nodal promotes the generation of M2-like macrophages and downregulates the expression of IL-12. Eur J Immunol 2013; 44:173-83. [DOI: 10.1002/eji.201343535] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 09/06/2013] [Accepted: 09/24/2013] [Indexed: 01/08/2023]
Affiliation(s)
- Xian-Feng Wang
- Department of Microbial and Biochemical Pharmacy; School of Pharmaceutical Sciences, Sun Yat-sen University; Guangzhou P.R. China
| | - Hong-Sheng Wang
- Department of Microbial and Biochemical Pharmacy; School of Pharmaceutical Sciences, Sun Yat-sen University; Guangzhou P.R. China
| | - Fan Zhang
- Department of Microbial and Biochemical Pharmacy; School of Pharmaceutical Sciences, Sun Yat-sen University; Guangzhou P.R. China
| | - Qiang Guo
- Department of Microbial and Biochemical Pharmacy; School of Pharmaceutical Sciences, Sun Yat-sen University; Guangzhou P.R. China
| | - Hao Wang
- Department of Microbial and Biochemical Pharmacy; School of Pharmaceutical Sciences, Sun Yat-sen University; Guangzhou P.R. China
| | - Ke-Fang Wang
- Department of Obstetrics and Gynecology; Beijing Anzhen Hospital, Capital Medical University; Beijing P.R. China
| | - Ge Zhang
- Department of Microbial and Biochemical Pharmacy; School of Pharmaceutical Sciences, Sun Yat-sen University; Guangzhou P.R. China
| | - Xian-zhang Bu
- Department of Microbial and Biochemical Pharmacy; School of Pharmaceutical Sciences, Sun Yat-sen University; Guangzhou P.R. China
| | - Shao-Hui Cai
- Department of Pharmacology; School of Pharmaceutical Sciences, Jinan University; Guangzhou P.R. China
| | - Jun Du
- Department of Microbial and Biochemical Pharmacy; School of Pharmaceutical Sciences, Sun Yat-sen University; Guangzhou P.R. China
| |
Collapse
|
48
|
Wang Y, Jiang Y, Tian T, Hori Y, Wada N, Ikeda JI, Morii E. Inhibitory effect of Nodal on the expression of aldehyde dehydrogenase 1 in endometrioid adenocarcinoma of uterus. Biochem Biophys Res Commun 2013; 440:731-6. [PMID: 24120944 DOI: 10.1016/j.bbrc.2013.09.139] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 09/30/2013] [Indexed: 01/16/2023]
Abstract
Cancers consist of heterogeneous populations. Recently, it has been demonstrated that cells with tumorigenic potential are limited to a small population, called cancer-initiating cells (CICs). Aldehyde dehydrogenase 1 (ALDH1) is one of the markers of CICs. We previously reported that ALDH1-high cases of uterine endometrioid adenocarcinoma showed poor prognosis, and ALDH1-high population of endometrioid adenocarcinoma cell line was more tumorigenic, resistant to anti-cancer drugs, and invasive than ALDH1-low population. Here, the regulatory signaling for ALDH1 was examined. The inhibition of TGF-β signaling increased ALDH1-high population. Among TGF-β family members, Nodal expression and ALDH1 expression levels were mutually exclusive. Immunohistochemical analysis on clinical samples revealed Nodal-high tumor cells to be ALDH-low and vise versa, suggesting that Nodal may inhibit ALDH1 expression via stimulating TGF-β signaling in uterine endometrioid adenocarcinoma. In fact, the addition of Nodal to endometrioid adenocarcinoma cell line reduced ALDH1-high population. Although ALDH1 mRNA level was not affected, the amount of ALDH1 protein appeared to be reduce by Nodal through ubiquitine-proteasome pathway. The regulation of TGF-β signaling might be a novel therapeutic target of CICs in endometrioid adenocarcinoma.
Collapse
Affiliation(s)
- Yi Wang
- Department of Pathology, Osaka University Graduate School of Medicine, Yamada-Oka 2-2, Suita 565-0871, Japan
| | | | | | | | | | | | | |
Collapse
|
49
|
Lai JH, Jan HJ, Liu LW, Lee CC, Wang SG, Hueng DY, Cheng YY, Lee HM, Ma HI. Nodal regulates energy metabolism in glioma cells by inducing expression of hypoxia-inducible factor 1α. Neuro Oncol 2013; 15:1330-41. [PMID: 23911596 DOI: 10.1093/neuonc/not086] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND A shift in glucose metabolism from oxidative phosphorylation to anaerobic glycolysis is the biochemical hallmark of malignant cancer cells. METHODS In the present study, we demonstrated that Nodal stimulated the expression of glycolytic enzymes and decreased reliance on mitochondrial oxidative phosphorylation in human glioma cancer cells. The shift in glucose metabolism was mediated by induction of the hypoxia-inducible factor (HIF). RESULTS Nodal protein expression was shown to be correlated with expression levels of glucose transporter (Glut)-1, hexokinase (HK)-II, pyruvate dehydrogenase kinase (PDK)-1, the phosphorylation level of pyruvate dehydrogenase (PDH), glucose uptake, and lactate accumulation in human glioma cells. These effects were inversely correlated with mitochondrial oxygen consumption and ATP production. Knockdown of Nodal expression with specific small hairpin RNA reduced Glut-1, HK-II, and PDK-1 expressions and PDH phosphorylation. Nodal knockdown also reduced glucose uptake and lactate generation, which in turn increased mitochondrial membrane potential (Ψ), O2 utilization, and ATP synthesis. The ectopic expression of Nodal in low-expressing Nodal glioma cells resulted in the opposite results compared with those of Nodal knockdown glioma cells. Treatment of cells with recombinant Nodal increased HIF-1 expression, and this effect was regulated at the transcriptional level. Blockage of the Nodal receptor by a pharmacological inhibitor or Nodal knockdown in U87MG cells decreased HIF-1α expression. Furthermore, HIF-1α knockdown in U87MG cells decreased Glut-1, HK-II, and PDK-1 expressions and PDH phosphorylation, which were similar to results in Nodal knockdown cells. CONCLUSION Taken together, these results suggest that Nodal affects energy metabolism through HIF-1α.
Collapse
Affiliation(s)
- Jing-Huei Lai
- Corresponding Authors: Horng-Mo Lee, PhD, Department of Medical Laboratory Sciences and Biotechnology, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei 110, Taiwan. Hsin-I Ma, Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, No.325, Sec.2, Chenggong Rd., Neihu District, Taipei 114, Taiwan, ROC
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Much of the focus on the transforming growth factor-β (TGFβ) superfamily in cancer has revolved around the TGFβ ligands themselves. However, it is now becoming apparent that deregulated signalling by many of the other superfamily members also has crucial roles in both the development of tumours and metastasis. Furthermore, these signalling pathways are emerging as plausible therapeutic targets. Their roles in tumorigenesis frequently reflect their function in embryonic development or in adult tissue homeostasis, and their influence extends beyond the tumours themselves, to the tumour microenvironment and more widely to complications of cancer such as cachexia and bone loss.
Collapse
Affiliation(s)
- Lalage M Wakefield
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, Maryland 20892-4255, USA.
| | | |
Collapse
|