251
|
Steger CM, Bonaros N, Rieker RJ, Bonatti J, Schachner T. Gene therapy with antisense oligonucleotides silencing c-myc reduces neointima formation and vessel wall thickness in a mouse model of vein graft disease. Exp Mol Pathol 2018; 105:1-9. [PMID: 29775572 DOI: 10.1016/j.yexmp.2018.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Accepted: 05/13/2018] [Indexed: 10/16/2022]
Abstract
Gene therapy for avoiding intimal hyperplasia of vein grafts after coronary artery bypass grafting is still discussed controversially. A promising application of gene therapy in vein grafts is the use of antisense oligonucleotides to block the expression of genes encoding cell cycle regulatory proteins in vascular smooth muscle cells. C-myc, either directly or by regulating the expression of other proteins, controls cell proliferation, apoptosis and cell survival, tissue remodeling, angiogenesis, cell metabolism, production of inflammatory and anti-inflammatory cytokines, and also participates in cell transformation. Forty C57BL/6J mice underwent interposition of the inferior vena cava from isogenic donor mice into the common carotid artery using a previously described cuff technique. Twenty mice received periadventitial administration of antisense oligonucleotides directed against c-myc (treatment group), the other twenty mice received no treatment (control group). All vein grafts were harvested two weeks after surgery, dehydrated, wax embedded, cut into slides of 2 μm thickness, stained and histologically and immunohistochemically examined under light microscope. In our study, we could show the promising effects of antisense oligonucleotide treatment in a mouse model of vein graft disease including the significant reduction of neointimal, media and total vessel wall thickness with a significantly lower percentage of SMA positive cells, elastic fibres and acid mucopolysaccharides in the neointima and media, a decreased vascularization, and a lower expression of PDGFR ß, MMP-9 and VEGF-A positive cells throughout the whole vein graft wall.
Collapse
Affiliation(s)
- Christina Maria Steger
- Department of Pathology, Academic Teaching Hospital Feldkirch, Carinagasse 47, 6800 Feldkirch, Austria.
| | - Nikolaos Bonaros
- Department of Cardiac Surgery, Innsbruck Medical University, Anichstrasse 35, 6020 Innsbruck, Austria
| | | | - Johannes Bonatti
- Heart and Vascular Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Thomas Schachner
- Department of Cardiac Surgery, Innsbruck Medical University, Anichstrasse 35, 6020 Innsbruck, Austria
| |
Collapse
|
252
|
Garcia DA, Baek C, Estrada MV, Tysl T, Bennett EJ, Yang J, Chang JT. USP11 Enhances TGFβ-Induced Epithelial-Mesenchymal Plasticity and Human Breast Cancer Metastasis. Mol Cancer Res 2018; 16:1172-1184. [PMID: 29724812 DOI: 10.1158/1541-7786.mcr-17-0723] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 02/28/2018] [Accepted: 04/18/2018] [Indexed: 12/26/2022]
Abstract
Epithelial-mesenchymal transition (EMT) is a conserved cellular plasticity program that is reactivated in carcinoma cells and drives metastasis. Although EMT is well studied its regulatory mechanisms remain unclear. Therefore, to identify novel regulators of EMT, a data mining approach was taken using published microarray data and a group of deubiquitinases (DUB) were found to be upregulated in cells that have undergone EMT. Here, it is demonstrated that one DUB, ubiquitin-specific peptidase 11 (USP11), enhances TGFβ-induced EMT and self-renewal in immortalized human mammary epithelial cells. Furthermore, modulating USP11 expression in human breast cancer cells altered the migratory capacity in vitro and metastasis in vivo Moreover, elevated USP11 expression in human breast cancer patient clinical specimens correlated with decreased survival. Mechanistically, modulating USP11 expression altered the stability of TGFβ receptor type II (TGFBR2) and TGFβ downstream signaling in human breast cancer cells. Together, these data suggest that deubiquitination of TGFBR2 by USP11 effectively spares TGFBR2 from proteasomal degradation to promote EMT and metastasis.Implications: USP11 regulates TGFβ-induced epithelial-mesenchymal plasticity and human breast cancer metastasis and may be a potential therapeutic target for breast cancer. Mol Cancer Res; 16(7); 1172-84. ©2018 AACR.
Collapse
Affiliation(s)
- Daniel A Garcia
- Department of Medicine, University of California San Diego, La Jolla, California.,Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, California
| | - Christina Baek
- Department of Medicine, University of California San Diego, La Jolla, California
| | - M Valeria Estrada
- Biorepository and Tissue Technology Shared Resources, Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Tiffani Tysl
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Eric J Bennett
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, La Jolla, California
| | - Jing Yang
- Department of Pharmacology, University of California San Diego, La Jolla, California.,Department of Pediatrics, University of California San Diego, La Jolla, California.,Moores Cancer Center, University of California San Diego, La Jolla, California
| | - John T Chang
- Department of Medicine, University of California San Diego, La Jolla, California.
| |
Collapse
|
253
|
Oba T, Ito KI. Combination of two anti-tubulin agents, eribulin and paclitaxel, enhances anti-tumor effects on triple-negative breast cancer through mesenchymal-epithelial transition. Oncotarget 2018; 9:22986-23002. [PMID: 29796167 PMCID: PMC5955406 DOI: 10.18632/oncotarget.25184] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 04/05/2018] [Indexed: 12/16/2022] Open
Abstract
Improved prognosis for triple-negative breast cancer (TNBC) has currently plateaued and the development of novel therapeutic strategies is required. Therefore, we aimed to explore the anti-tumor effect of eribulin and paclitaxel combination therapy for TNBC. The effect of eribulin and paclitaxel in combination was tested, with both concurrent and sequential administration, using four TNBC cell lines (MDA-MB-231, Hs578T, MDA-MB-157, and Mx-1) in vitro and in an MDA-MB-231 BALB/c-nu/nu mouse xenograft model. The expression of epithelial-mesenchymal phenotypic markers was analyzed by western blotting and immunohistochemical analyses. Simultaneous administration of eribulin and paclitaxel resulted in a synergistic anti-tumor effect with MDA-MB-231 and Hs578T cells, but not MDA-MB-157 and Mx-1 cells, in vitro. Moreover, pre-treatment with one drug significantly enhanced sensitivity to the subsequently administrated second drug in MDA-MB-231 and Hs578T cells. Eribulin increased E-cadherin expression and decreased the expression of mesenchymal markers in MDA-MB-231 and Hs578T cells. In contrast, paclitaxel increased the expression of mesenchymal markers. When epithelial-mesenchymal transition was induced by TGF-β1, eribulin sensitivity was enhanced. In contrast, a TGF-β receptor kinase inhibitor decreased eribulin sensitivity. In MDA-MB-231 tumor-bearing mice, concurrent administration of low doses of eribulin and paclitaxel significantly inhibited tumor growth compared to that with either monotherapy. Moreover, single administration of eribulin before the initiation of paclitaxel treatment decreased vimentin expression and reduced the average tumor volume in a mouse xenograft model. Eribulin and paclitaxel show synergistic anti-tumor effect by altering the epithelial-mesenchymal phenotype. This combination therapy could represent a novel therapeutic strategy for TNBC.
Collapse
Affiliation(s)
- Takaaki Oba
- Division of Breast, Endocrine and Respiratory Surgery, Department of Surgery II, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| | - Ken-Ichi Ito
- Division of Breast, Endocrine and Respiratory Surgery, Department of Surgery II, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| |
Collapse
|
254
|
Frizzled-8 integrates Wnt-11 and transforming growth factor-β signaling in prostate cancer. Nat Commun 2018; 9:1747. [PMID: 29717114 PMCID: PMC5931552 DOI: 10.1038/s41467-018-04042-w] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 03/29/2018] [Indexed: 01/14/2023] Open
Abstract
Wnt-11 promotes cancer cell migration and invasion independently of β-catenin but the receptors involved remain unknown. Here, we provide evidence that FZD8 is a major Wnt-11 receptor in prostate cancer that integrates Wnt-11 and TGF-β signals to promote EMT. FZD8 mRNA is upregulated in multiple prostate cancer datasets and in metastatic cancer cell lines in vitro and in vivo. Analysis of patient samples reveals increased levels of FZD8 in cancer, correlating with Wnt-11. FZD8 co-localizes and co-immunoprecipitates with Wnt-11 and potentiates Wnt-11 activation of ATF2-dependent transcription. FZD8 silencing reduces prostate cancer cell migration, invasion, three-dimensional (3D) organotypic cell growth, expression of EMT-related genes, and TGF-β/Smad-dependent signaling. Mechanistically, FZD8 forms a TGF-β-regulated complex with TGF-β receptors that is mediated by the extracellular domains of FZD8 and TGFBR1. Targeting FZD8 may therefore inhibit aberrant activation of both Wnt and TGF-β signals in prostate cancer. Wnt11 has been shown to play a role in invasion and metastasis of prostate cancer. Here the authors show that in prostate cancer cells Wnt11 signals through the Fzd8 receptor and report an interaction between Fzd8 and TGF-β receptors regulating the transcription of a subset of TGF-beta genes.
Collapse
|
255
|
Goyette MA, Duhamel S, Aubert L, Pelletier A, Savage P, Thibault MP, Johnson RM, Carmeliet P, Basik M, Gaboury L, Muller WJ, Park M, Roux PP, Gratton JP, Côté JF. The Receptor Tyrosine Kinase AXL Is Required at Multiple Steps of the Metastatic Cascade during HER2-Positive Breast Cancer Progression. Cell Rep 2018; 23:1476-1490. [PMID: 29719259 DOI: 10.1016/j.celrep.2018.04.019] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 02/21/2018] [Accepted: 04/03/2018] [Indexed: 12/14/2022] Open
Abstract
AXL is activated by its ligand GAS6 and is expressed in triple-negative breast cancer cells. In the current study, we report AXL expression in HER2-positive (HER2+) breast cancers where it correlates with poor patient survival. Using murine models of HER2+ breast cancer, Axl, but not its ligand Gas6, was found to be essential for metastasis. We determined that AXL is required for intravasation, extravasation, and growth at the metastatic site. We found that AXL is expressed in HER2+ cancers displaying epithelial-to-mesenchymal transition (EMT) signatures where it contributes to sustain EMT. Interfering with AXL in a patient-derived xenograft (PDX) impaired transforming growth factor β (TGF-β)-induced cell invasion. Last, pharmacological inhibition of AXL specifically decreased the metastatic burden of mice developing HER2+ breast cancer. Our data identify AXL as a potential anti-metastatic co-therapeutic target for the treatment of HER2+ breast cancers.
Collapse
Affiliation(s)
- Marie-Anne Goyette
- Montreal Clinical Research Institute (IRCM), Montréal, QC H2W 1R7, Canada; Molecular Biology Programs, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Stéphanie Duhamel
- Montreal Clinical Research Institute (IRCM), Montréal, QC H2W 1R7, Canada
| | - Léo Aubert
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Ariane Pelletier
- Montreal Clinical Research Institute (IRCM), Montréal, QC H2W 1R7, Canada
| | - Paul Savage
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montréal, QC H3A 1A1, Canada
| | | | - Radia Marie Johnson
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montréal, QC H3A 1A1, Canada
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, VIB Vesalius Research Center, VIB, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, University of Leuven, Leuven 3000, Belgium
| | - Mark Basik
- Department of Oncology and Surgery, Segal Cancer Center, Lady Davis Institute for Medical Research, Sir Mortimer B. Davis Jewish General Hospital, McGill University, Montréal, QC H3T 1E2, Canada
| | - Louis Gaboury
- Department of Pathology and Cell Biology, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - William J Muller
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montréal, QC H3A 1A1, Canada
| | - Morag Park
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montréal, QC H3A 1A1, Canada
| | - Philippe P Roux
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC H3C 3J7, Canada; Department of Pathology and Cell Biology, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Jean-Philippe Gratton
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Jean-François Côté
- Montreal Clinical Research Institute (IRCM), Montréal, QC H2W 1R7, Canada; Molecular Biology Programs, Université de Montréal, Montréal, QC H3T 1J4, Canada; Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC H3C 3J7, Canada; Department of Anatomy and Cell Biology, McGill University, Montréal, QC H3A 0C7, Canada.
| |
Collapse
|
256
|
Cai J, Xia L, Li J, Ni S, Song H, Wu X. Tumor-Associated Macrophages Derived TGF-β‒Induced Epithelial to Mesenchymal Transition in Colorectal Cancer Cells through Smad2,3-4/Snail Signaling Pathway. Cancer Res Treat 2018; 51:252-266. [PMID: 29690747 PMCID: PMC6333993 DOI: 10.4143/crt.2017.613] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 04/24/2018] [Indexed: 12/22/2022] Open
Abstract
Purpose We investigated the role of tumor-associated macrophages (TAMs) on the epithelial to mesenchymal transition (EMT) of colorectal cancer cells and determined the potential mechanism involved in the metastatic process. Materials and Methods In this study, flow cytometry was used to detect the expression of target proteins. We used transwell assay to evaluate the migration of cancer cells under specific conditions. Using real-time polymerase chain reaction, we examined the expressions of cytokines and EMT-related markers in mRNA level. Animal assay was performed for analysis in vivo and hematoxylin and eosin was used to visualize the effect of TAMs on tumor metastasis. We also used immunohistochemistry and Western blotting to detect the expression of target proteins. Results Here, we observed enrichment of TAMs in colorectal tumor tissues, resulting in high metastasis in clinical therapy. Moreover, those TAMs could facilitate the EMT progression of colorectal cancer cells, which is induced by the transforming growth factor-β (TGF-β) derived from TAMs, leading to the invasion and migration of cancer cells. Conclusion Our results demonstrated that TAMs contributed the EMT progression through a TGF-β/Smad2,3-4/Snail signaling pathway, and disrupting this pathway with TGF-β receptor inhibitor could suppress metastasis, readjusting our focus to the connection of TAMs and cancer metastasis.
Collapse
Affiliation(s)
- Jianhui Cai
- Department of Colorectal and Anal Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Limin Xia
- Department of Colorectal and Anal Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jinlei Li
- Department of Colorectal and Anal Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shichang Ni
- Department of Colorectal and Anal Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huayu Song
- Department of Colorectal and Anal Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiangbin Wu
- Department of Colorectal and Anal Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
257
|
Liu ZY, Cao J, Zhang JT, Xu GL, Li XP, Wang FT, Ansari KH, Mohamed H, Fan YZ. Ring finger protein 125, as a potential highly aggressive and unfavorable prognostic biomarker, promotes the invasion and metastasis of human gallbladder cancers via activating the TGF- β1-SMAD3-ID1 signaling pathway. Oncotarget 2018; 8:49897-49914. [PMID: 28611292 PMCID: PMC5564816 DOI: 10.18632/oncotarget.18180] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 04/19/2017] [Indexed: 01/06/2023] Open
Abstract
Human gallbladder cancer (GBC) is a lethal aggressive malignant neoplasm. Identification of potential molecular biomarkers and development of targeted therapeutics for GBC patients is very necessary. In this study, we firstly investigated the correlation between ring finger protein 125 (RNF125) expression and the metastasis and prognosis of GBC, and the underlying molecular mechanism. RNF125 expression in a cohort of GBC tissues was examined; its correlation with clinicopathological and prognostic factors of GBC patients was analyzed. Moreover, the metastasis-related difference expressed genes in highly and lowly aggressive GBC cell lines were identified; and the influence of RNF125 knockdown on the metastatic phenotypes and characteristic EMT markers in highly aggressive GBC NOZ cells was detected. Furthermore, the underlying molecular mechanism of RNF125 effect was explored. The results showed that RNF125 was highly expressed in GBC tissues and related with aggressive characteristics such as Nevin stage (P = 0.041) etc. and unfavorable prognosis of GBC patients (P = 0.023, log-rank test). And, RNF125 was proved to a positive metastasis-related gene in vitro. RNF125 knockdown inhibited the invasion and migration, enhanced the adhesion, upregulated E-cadherin and β-catenin expression, and downregulated vimentin and N-cadherin expression (all P < 0.001) of NOZ cells in vitro. RNF125 promoting effect on GBC tumor progression was identified to relate with the activation of TGF-β1-SMAD3-ID1 signaling pathway. These findings firstly confirm that high RNF125 expression is related with aggressive characteristics and unfavorable prognosis of GBC patients; RNF125 promotes the invasion and metastasis of human GBCs via activating the TGF-β1-SMAD3-ID1 signaling pathway.
Collapse
Affiliation(s)
- Zhong-Yan Liu
- Department of Surgery, Tongji Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200065, P.R. China
| | - Jin Cao
- Department of Surgery, Tongji Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200065, P.R. China
| | - Jing-Tao Zhang
- Department of Surgery, Tongji Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200065, P.R. China
| | - Guo-Li Xu
- Department of Surgery, Tongji Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200065, P.R. China
| | - Xin-Ping Li
- Department of Surgery, Tongji Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200065, P.R. China
| | - Fang-Tao Wang
- Department of Surgery, Tongji Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200065, P.R. China
| | - Kamar Hasan Ansari
- Department of Surgery, Tongji Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200065, P.R. China
| | - Hassan Mohamed
- Department of Surgery, Tongji Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200065, P.R. China
| | - Yue-Zu Fan
- Department of Surgery, Tongji Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200065, P.R. China
| |
Collapse
|
258
|
Eser PÖ, Jänne PA. TGFβ pathway inhibition in the treatment of non-small cell lung cancer. Pharmacol Ther 2018; 184:112-130. [DOI: 10.1016/j.pharmthera.2017.11.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
259
|
Kimbrough-Allah MN, Millena AC, Khan SA. Differential role of PTEN in transforming growth factor β (TGF-β) effects on proliferation and migration in prostate cancer cells. Prostate 2018; 78:377-389. [PMID: 29341212 PMCID: PMC5820153 DOI: 10.1002/pros.23482] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 12/21/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Transforming growth factor-β (TGF-β) acts as a tumor suppressor in normal epithelial cells but as a tumor promoter in advanced prostate cancer cells. PI3-kinase pathway mediates TGF-β effects on prostate cancer cell migration and invasion. PTEN inhibits PI3-kinase pathway and is frequently mutated in prostate cancers. We investigated possible role(s) of PTEN in TGF-β effects on proliferation and migration in prostate cancer cells. METHODS Expression of PTEN mRNA and proteins were determined using RT-PCR and Western blotting in RWPE1 and DU145 cells. We also studied the role of PTEN in TGF-β effects on cell proliferation and migration in DU145 cells after transient silencing of endogenous PTEN. Conversely, we determined the role of PTEN in cell proliferation and migration after over-expression of PTEN in PC3 cells which lack endogenous PTEN. RESULTS TGF-β1 and TGF-β3 had no effect on PTEN mRNA levels but both isoforms increased PTEN protein levels in DU145 and RWPE1 cells indicating that PTEN may mediate TGF-β effects on cell proliferation. Knockdown of PTEN in DU145 cells resulted in significant increase in cell proliferation which was not affected by TGF-β isoforms. PTEN overexpression in PC3 cells inhibited cell proliferation. Knockdown of endogenous PTEN enhanced cell migration in DU145 cells, whereas PTEN overexpression reduced migration in PC3 cells and reduced phosphorylation of AKT in response to TGF-β. CONCLUSION We conclude that PTEN plays a role in inhibitory effects of TGF-β on cell proliferation whereas its absence may enhance TGF-β effects on activation of PI3-kinase pathway and cell migration.
Collapse
Affiliation(s)
| | - Ana C Millena
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, Georgia
| | - Shafiq A Khan
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, Georgia
| |
Collapse
|
260
|
Tumor-fibroblast interactions stimulate tumor vascularization by enhancing cytokine-driven production of MMP9 by tumor cells. Oncotarget 2018; 8:35592-35608. [PMID: 28423685 PMCID: PMC5482601 DOI: 10.18632/oncotarget.16022] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 02/24/2017] [Indexed: 12/31/2022] Open
Abstract
Advance-stage breast carcinomas include significant amounts of fibroblasts and infiltrating immune cells which have been implicated in tumor growth, recurrence, and response to therapy. The present study investigated the contribution of fibroblasts to tumor growth using direct tumor-fibroblast co-cultures and tumor xenograft models. Our findings revealed that fibroblasts enhance breast carcinoma growth by promoting the tumor vasculature via the MMP9-dependent mechanism. In tumor-fibroblast co-cultures, fibroblasts increased expression of TGF-β, TNF, and IL-1β cytokines in tumor cells. These cytokines cooperatively induced expression of matrix metalloproteinase MMP9 in tumor cells. Knockdown of MMP9 by shRNA significantly reduced tumor vascularization induced by fibroblasts. Mechanistically, our findings argue that expression of MMP9 in tumor cellsis regulated by crosstalk of TGF-β with TNF and/or IL-1β cytokines. The mechanism of this cooperative response did not involve cross-activation of the canonical signaling pathways as TGF-β did not activate RELA/p65 signaling, while TNF did not affect SMAD signaling. Instead, TGF-β and TNF cytokines co-stimulated MAP kinases and expression of JUN and JUNB, AP1 transcription factor subunits, which together with RELA/p65 were essential for the regulation of MMP9. Depletion of JUN and JUNB or RELA in tumor cells blocked the cooperative induction of MMP9 by the cytokines. Thus, our studies uncovered a previously unappreciated role of tumor-fibroblast interactions in the stimulation of tumor angiogenesis, and an essential role of the MAPK-AP1 axis in the cooperative up-regulation of the angiogenic driver MMP9 by cytokine crosstalk.
Collapse
|
261
|
Erin N, Ogan N, Yerlikaya A. Secretomes reveal several novel proteins as well as TGF-β1 as the top upstream regulator of metastatic process in breast cancer. Breast Cancer Res Treat 2018; 170:235-250. [PMID: 29557524 DOI: 10.1007/s10549-018-4752-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 03/13/2018] [Indexed: 12/11/2022]
Abstract
PURPOSE Metastatic breast cancer is resistant to many conventional treatments and novel therapeutic targets are needed. We previously isolated subsets of 4T1 murine breast cancer cells which metastasized to liver (4TLM), brain (4TBM), and heart (4THM). Among these cells, 4TLM is the most aggressive one, demonstrating mesenchymal phenotype. Here we compared secreted proteins from 4TLM, 4TBM, and 4THM cells and compared with that of hardly metastatic 67NR cells to detect differentially secreted factors involved in organ-specific metastasis. METHOD AND RESULTS Label-free LC-MS/MS proteomic technique was used to detect the differentially secreted proteins. Eighty-five of over 500 secreted proteins were significantly altered in metastatic breast cancer cells. Differential expression of several proteins such as fibulin-4, Bone Morphogenetic Protein 1, TGF-β1 MMP-3, MMP-9, and Thymic Stromal Lymphopoietin were further verified using ELISA or Western blotting. Many of these identified proteins were also present in human metastatic breast carcinomas. Annexin A1 and A5, laminin beta 1, Neutral alpha-glucosidase AB were commonly found at least in three out of six studies examined here. Ingenuity Pathway Analysis showed that proteins differentially secreted from metastatic cells are involved primarily in carcinogenesis and TGF-β1 is the top upstream regulator in all metastatic cells. CONCLUSIONS Cells metastasized to different organs displayed significant differences in several of secreted proteins. Proteins differentially altered were fibronectin, insulin-like growth factor-binding protein 7, and Procollagen-lysine, 2-oxoglutarate 5-dioxygenase 1. On the other hand, many exosomal proteins were also common to all metastatic cells, demonstrating involvement of key universal factors in distant metastatic process.
Collapse
Affiliation(s)
- Nuray Erin
- Department of Medical Pharmacology, School of Medicine, Akdeniz University, B-blok kat 1, SBAUM/Immunoloji Lab, Antalya, Turkey.
| | - Nur Ogan
- Department of Medical Pharmacology, School of Medicine, Akdeniz University, B-blok kat 1, SBAUM/Immunoloji Lab, Antalya, Turkey
| | - Azmi Yerlikaya
- Department of Medical Biology, School of Medicine, Dumlupınar University, Kütahya, Turkey
| |
Collapse
|
262
|
Sharma S, Zhuang R, Long M, Pavlovic M, Kang Y, Ilyas A, Asghar W. Circulating tumor cell isolation, culture, and downstream molecular analysis. Biotechnol Adv 2018; 36:1063-1078. [PMID: 29559380 DOI: 10.1016/j.biotechadv.2018.03.007] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 03/07/2018] [Accepted: 03/12/2018] [Indexed: 12/12/2022]
Abstract
Circulating tumor cells (CTCs) are a major contributor of cancer metastases and hold a promising prognostic significance in cancer detection. Performing functional and molecular characterization of CTCs provides an in-depth knowledge about this lethal disease. Researchers are making efforts to design devices and develop assays for enumeration of CTCs with a high capture and detection efficiency from whole blood of cancer patients. The existing and on-going research on CTC isolation methods has revealed cell characteristics which are helpful in cancer monitoring and designing of targeted cancer treatments. In this review paper, a brief summary of existing CTC isolation methods is presented. We also discuss methods of detaching CTC from functionalized surfaces (functional assays/devices) and their further use for ex-vivo culturing that aid in studies regarding molecular properties that encourage metastatic seeding. In the clinical applications section, we discuss a number of cases that CTCs can play a key role for monitoring metastases, drug treatment response, and heterogeneity profiling regarding biomarkers and gene expression studies that bring treatment design further towards personalized medicine.
Collapse
Affiliation(s)
- Sandhya Sharma
- Department of Computer & Electrical Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL 33431, USA; Asghar-Lab, Micro and Nanotechnology in Medicine, College of Engineering and Computer Science, Boca Raton, FL 33431, USA
| | - Rachel Zhuang
- Asghar-Lab, Micro and Nanotechnology in Medicine, College of Engineering and Computer Science, Boca Raton, FL 33431, USA
| | - Marisa Long
- Asghar-Lab, Micro and Nanotechnology in Medicine, College of Engineering and Computer Science, Boca Raton, FL 33431, USA
| | - Mirjana Pavlovic
- Department of Computer & Electrical Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Yunqing Kang
- Department of Ocean & Mechanical Engineering, College of Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL 33431, USA; Department of Biomedical Science, College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Azhar Ilyas
- Department of Electrical & Computer Engineering, New York Institute of Technology, Old Westbury, NY 11568, USA
| | - Waseem Asghar
- Department of Computer & Electrical Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL 33431, USA; Asghar-Lab, Micro and Nanotechnology in Medicine, College of Engineering and Computer Science, Boca Raton, FL 33431, USA; Department of Biological Sciences, Florida Atlantic University, Boca Raton, FL 33431, USA.
| |
Collapse
|
263
|
Marin-Acevedo JA, Dholaria B, Soyano AE, Knutson KL, Chumsri S, Lou Y. Next generation of immune checkpoint therapy in cancer: new developments and challenges. J Hematol Oncol 2018; 11:39. [PMID: 29544515 PMCID: PMC5856308 DOI: 10.1186/s13045-018-0582-8] [Citation(s) in RCA: 553] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 03/01/2018] [Indexed: 02/07/2023] Open
Abstract
Immune checkpoints consist of inhibitory and stimulatory pathways that maintain self-tolerance and assist with immune response. In cancer, immune checkpoint pathways are often activated to inhibit the nascent anti-tumor immune response. Immune checkpoint therapies act by blocking or stimulating these pathways and enhance the body's immunological activity against tumors. Cytotoxic T lymphocyte-associated molecule-4 (CTLA-4), programmed cell death receptor-1 (PD-1), and programmed cell death ligand-1(PD-L1) are the most widely studied and recognized inhibitory checkpoint pathways. Drugs blocking these pathways are currently utilized for a wide variety of malignancies and have demonstrated durable clinical activities in a subset of cancer patients. This approach is rapidly extending beyond CTLA-4 and PD-1/PD-L1. New inhibitory pathways are under investigation, and drugs blocking LAG-3, TIM-3, TIGIT, VISTA, or B7/H3 are being investigated. Furthermore, agonists of stimulatory checkpoint pathways such as OX40, ICOS, GITR, 4-1BB, CD40, or molecules targeting tumor microenvironment components like IDO or TLR are under investigation. In this article, we have provided a comprehensive review of immune checkpoint pathways involved in cancer immunotherapy, and discuss their mechanisms and the therapeutic interventions currently under investigation in phase I/II clinical trials. We also reviewed the limitations, toxicities, and challenges and outline the possible future research directions.
Collapse
Affiliation(s)
| | - Bhagirathbhai Dholaria
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, USA
- Present Address: Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
| | - Aixa E Soyano
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, USA
| | | | - Saranya Chumsri
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, USA
| | - Yanyan Lou
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
264
|
Okada Y, Wang T, Kasai K, Suzuki K, Takikawa Y. Regulation of transforming growth factor is involved in the efficacy of combined 5-fluorouracil and interferon alpha-2b therapy of advanced hepatocellular carcinoma. Cell Death Discov 2018; 4:42. [PMID: 29560281 PMCID: PMC5849890 DOI: 10.1038/s41420-018-0040-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Revised: 01/22/2018] [Accepted: 02/06/2018] [Indexed: 12/22/2022] Open
Abstract
Transforming growth factor-beta (TGF-β) is critical in cancer cell invasion and metastasis. The effects of a treatment that targets TGF-β using the combination of interferon alpha (IFNα)-2b and 5-fluorouracil (5-FU) are unknown. Here, we show that the serum levels of TGF-β1 prior to the therapy correlate with increased maximum tumor diameter, which is significantly (p < 0.01) decreased after the combination therapy. 5-FU increased both the expression and secretion levels of TGF-β1 in hepatoma cells, but not in normal hepatocytes. The combination of 5-FU and IFNα-2b synergistically affected cell death. However, a TGF-β1 specific inhibitor did not affect the anti-tumor activity of 5-FU. 5-FU inhibited the phosphorylation of SMAD2 and reduced the total protein levels of SMAD2, SMAD4, and pINK4b. Conversely, 5-FU stimulated the phosphorylation of extracellular signal-regulated kinase (ERK)1/2. Accordingly, the protein levels of E-cadherin and claudin-1 were reduced in 5-FU-treated cells. The combination of 5-FU and IFNα-2b, and the inhibition of ERK1/2 by a specific inhibitor neutralized the effects of 5-FU on TGF-β-related signaling molecules and restored their protein levels to those observed in the control. Interestingly, the phosphorylated protein levels of SMAD2 and the total protein levels of E-cadherin and p15INK4b were increased in 5-FU-stimulated HuH-7 cells, but not in Hep G2 cells. Our data suggest that the higher efficacy of the 5-FU and IFNα-2b combination therapy was associated with the regulation of TGF-β expression, secretion, and the signals mediated by it.
Collapse
Affiliation(s)
- Youhei Okada
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, Morioka, Iwate Japan
| | - Ting Wang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, Morioka, Iwate Japan
| | - Kazuhiro Kasai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, Morioka, Iwate Japan
| | - Kazuyuki Suzuki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, Morioka, Iwate Japan
| | - Yasuhiro Takikawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, Morioka, Iwate Japan
| |
Collapse
|
265
|
Shen M, Tsai Y, Zhu R, Keng PC, Chen Y, Chen Y, Lee SO. RETRACTED: FASN-TGF-β1-PD-L1 axis contributes to the development of resistance to NK cell cytotoxicity of cisplatin-resistant lung cancer cells. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:313-322. [DOI: 10.1016/j.bbalip.2017.12.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 12/22/2017] [Accepted: 12/31/2017] [Indexed: 12/11/2022]
|
266
|
Pindiprolu SKSS, Krishnamurthy PT, Chintamaneni PK. Pharmacological targets of breast cancer stem cells: a review. Naunyn Schmiedebergs Arch Pharmacol 2018; 391:463-479. [PMID: 29476201 DOI: 10.1007/s00210-018-1479-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 02/13/2018] [Indexed: 02/07/2023]
Abstract
Breast cancers contain small population of tumor-initiating cells called breast cancer stem cells (BCSCs), which are spared even after chemotherapy. Recently, BCSCs are implicated to be a cause of metastasis, tumor relapse, and therapy resistance in breast cancer. BCSCs have unique molecular mechanisms, which can be targeted to eliminate them. These include surface biomarkers, proteins involved in self-renewal pathways, drug efflux transporters, apoptotic/antiapoptotic proteins, autophagy, metabolism, and microenvironment regulation. The complex molecular mechanisms behind the survival of BCSCs and pharmacological targets for elimination of BCSCs are described in this review.
Collapse
Affiliation(s)
- Sai Kiran S S Pindiprolu
- Department of Pharmacology, JSS College of Pharmacy (Jagadguru Sri Shivarathreeshwara University), Rocklands, Udhagamandalam, Tamil Nadu, 643001, India
| | - Praveen T Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy (Jagadguru Sri Shivarathreeshwara University), Rocklands, Udhagamandalam, Tamil Nadu, 643001, India.
| | - Pavan Kumar Chintamaneni
- Department of Pharmacology, JSS College of Pharmacy (Jagadguru Sri Shivarathreeshwara University), Rocklands, Udhagamandalam, Tamil Nadu, 643001, India
| |
Collapse
|
267
|
Principe DR, DeCant B, Staudacher J, Vitello D, Mangan RJ, Wayne EA, Mascariñas E, Diaz AM, Bauer J, McKinney RD, Khazaie K, Pasche B, Dawson DW, Munshi HG, Grippo PJ, Jung B. Loss of TGFβ signaling promotes colon cancer progression and tumor-associated inflammation. Oncotarget 2018; 8:3826-3839. [PMID: 27270652 PMCID: PMC5354798 DOI: 10.18632/oncotarget.9830] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 05/14/2016] [Indexed: 01/05/2023] Open
Abstract
TGFβ has both tumor suppressive and tumor promoting effects in colon cancer. Also, TGFβ can affect the extent and composition of inflammatory cells present in tumors, contextually promoting and inhibiting inflammation. While colon tumors display intratumoral inflammation, the contributions of TGFβ to this process are poorly understood. In human patients, we found that epithelial loss of TGFβ signaling was associated with increased inflammatory burden; yet overexpression of TGFβ was also associated with increased inflammation. These findings were recapitulated in mutant APC models of murine tumorigenesis, where epithelial truncation of TGFBR2 led to lethal inflammatory disease and invasive colon cancer, mediated by IL8 and TGFβ1. Interestingly, mutant APC mice with global suppression of TGFβ signals displayed an intermediate phenotype, presenting with an overall increase in IL8-mediated inflammation and accelerated tumor formation, yet with a longer latency to the onset of disease observed in mice with epithelial TGFBR-deficiency. These results suggest that the loss of TGFβ signaling, particularly in colon epithelial cells, elicits a strong inflammatory response and promotes tumor progression. This implies that treating colon cancer patients with TGFβ inhibitors may result in a worse outcome by enhancing inflammatory responses.
Collapse
Affiliation(s)
- Daniel R Principe
- University of Illinois College of Medicine, Urbana-Champaign, IL, USA
| | - Brian DeCant
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Jonas Staudacher
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Dominic Vitello
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Riley J Mangan
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Elizabeth A Wayne
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Emman Mascariñas
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Andrew M Diaz
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Jessica Bauer
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Ronald D McKinney
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Khashayarsha Khazaie
- Department of Immunology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Boris Pasche
- Comprehensive Cancer Center of Wake Forest University, Winston-Salem, NC, USA
| | - David W Dawson
- Department of Pathology and Laboratory Medicine, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Hidayatullah G Munshi
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.,Department of Medicine, Northwestern University, Chicago, IL, USA
| | - Paul J Grippo
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Barbara Jung
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
268
|
MiRNAs at the Crossroads between Innate Immunity and Cancer: Focus on Macrophages. Cells 2018; 7:cells7020012. [PMID: 29419779 PMCID: PMC5850100 DOI: 10.3390/cells7020012] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 02/01/2018] [Accepted: 02/06/2018] [Indexed: 12/12/2022] Open
Abstract
Innate immune cells form an integrative component of the tumor microenvironment (TME), which can control or prevent tumor initiation and progression, due to the simultaneous processing of both anti- and pro-growth signals. This decision-making process is a consequence of gene expression changes, which are in part dependent on post-transcriptional regulatory mechanisms. In this context, microRNAs have been shown to regulate both recruitment and activation of specific tumor-associated immune cells in the TME. This review aims to describe the most important microRNAs that target cancer-related innate immune pathways. The role of exosomal microRNAs in tumor progression and microRNA-based therapeutic strategies are also discussed.
Collapse
|
269
|
Wang X, Li F, Xie L, Crane J, Zhen G, Mishina Y, Deng R, Gao B, Chen H, Liu S, Yang P, Gao M, Tu M, Wang Y, Wan M, Fan C, Cao X. Inhibition of overactive TGF-β attenuates progression of heterotopic ossification in mice. Nat Commun 2018; 9:551. [PMID: 29416028 PMCID: PMC5803194 DOI: 10.1038/s41467-018-02988-5] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 01/11/2018] [Indexed: 12/24/2022] Open
Abstract
Acquired heterotopic ossification (HO) is a painful and debilitating disease characterized by extraskeletal bone formation after injury. The exact pathogenesis of HO remains unknown. Here we show that TGF-β initiates and promotes HO in mice. We find that calcified cartilage and newly formed bone resorb osteoclasts after onset of HO, which leads to high levels of active TGF-β that recruit mesenchymal stromal/progenitor cells (MSPCs) in the HO microenvironment. Transgenic expression of active TGF-β in tendon induces spontaneous HO, whereas systemic injection of a TGF-β neutralizing antibody attenuates ectopic bone formation in traumatic and BMP-induced mouse HO models, and in a fibrodysplasia ossificans progressive mouse model. Moreover, inducible knockout of the TGF-β type II receptor in MSPCs inhibits HO progression in HO mouse models. Our study points toward elevated levels of active TGF-β as inducers and promoters of ectopic bone formation, and suggest that TGF-β might be a therapeutic target in HO.
Collapse
MESH Headings
- Achilles Tendon/drug effects
- Achilles Tendon/injuries
- Adult
- Animals
- Antibodies, Neutralizing/pharmacology
- Becaplermin/metabolism
- Bone Remodeling
- Brain Injuries, Traumatic
- Cartilage
- Case-Control Studies
- Disease Models, Animal
- Elbow Joint/surgery
- Female
- Fracture Fixation, Internal
- Fractures, Bone
- Humans
- Male
- Mesenchymal Stem Cells/metabolism
- Mice
- Mice, Knockout
- Mice, Transgenic
- Middle Aged
- Muscle, Skeletal/pathology
- Myositis Ossificans/metabolism
- Ossification, Heterotopic/metabolism
- Osteoclasts
- Osteogenesis/drug effects
- Receptor, Transforming Growth Factor-beta Type II/genetics
- Spinal Cord Injuries
- Tendon Injuries
- Tendons
- Transforming Growth Factor beta/antagonists & inhibitors
- Transforming Growth Factor beta/metabolism
- Transforming Growth Factor beta1/metabolism
- Young Adult
- Elbow Injuries
Collapse
Affiliation(s)
- Xiao Wang
- Department of Orthopedic Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Fengfeng Li
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, 200030, Shanghai, China
| | - Liang Xie
- Department of Orthopedic Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Janet Crane
- Department of Orthopedic Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Gehua Zhen
- Department of Orthopedic Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Yuji Mishina
- School of Dentistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Ruoxian Deng
- Department of Orthopedic Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Bo Gao
- Department of Orthopedic Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Hao Chen
- Department of Orthopedic Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Shen Liu
- Department of Orthopedic Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, 200030, Shanghai, China
| | - Ping Yang
- Department of Orthopedic Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Manman Gao
- Department of Orthopedic Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Manli Tu
- Department of Orthopedic Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Yiguo Wang
- Department of Orthopedic Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Mei Wan
- Department of Orthopedic Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Cunyi Fan
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, 200030, Shanghai, China
| | - Xu Cao
- Department of Orthopedic Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA.
| |
Collapse
|
270
|
Vivekanandhan S, Mukhopadhyay D. Genetic status of KRAS influences Transforming Growth Factor-beta (TGF-β) signaling: An insight into Neuropilin-1 (NRP1) mediated tumorigenesis. Semin Cancer Biol 2018; 54:72-79. [PMID: 29409705 DOI: 10.1016/j.semcancer.2018.01.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 01/25/2018] [Indexed: 02/07/2023]
Abstract
Oncogenic RAS and deregulated transforming growth factor-beta (TGF)-β signaling have been implicated in several cancers. So far, attempts to target either one of them therapeutically have been futile as both of them are involved in multiple fundamental cellular processes and the normal forms are expressed by almost all cells. Hence, their inhibition would disrupt several physiological processes. Besides, their downregulation stimulates the tumor cells to develop adaptive mechanisms and would most likely be ineffective as therapeutic targets. Furthermore, growing literature suggests that both of these signaling pathways converge to enhance tumor development. Therefore, a lot of interest has been generated to explore the areas where these pathways interface that might identify new molecules that could potentially serve as novel therapeutic targets. In this review, we focus on such convergent signaling and cross-interaction that is mediated by neuropilin-1 (NRP1), a receptor that can interact with multiple growth factors including TGF-β for promoting tumorigenesis process.
Collapse
Affiliation(s)
- Sneha Vivekanandhan
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL, United States
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL, United States.
| |
Collapse
|
271
|
Wang B, Liu T, Wu JC, Luo SZ, Chen R, Lu LG, Xu MY. STAT3 aggravates TGF-β1-induced hepatic epithelial-to-mesenchymal transition and migration. Biomed Pharmacother 2018; 98:214-221. [DOI: 10.1016/j.biopha.2017.12.035] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 12/10/2017] [Accepted: 12/13/2017] [Indexed: 12/29/2022] Open
|
272
|
Xu X, Zheng L, Yuan Q, Zhen G, Crane JL, Zhou X, Cao X. Transforming growth factor-β in stem cells and tissue homeostasis. Bone Res 2018; 6:2. [PMID: 29423331 PMCID: PMC5802812 DOI: 10.1038/s41413-017-0005-4] [Citation(s) in RCA: 268] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 11/12/2017] [Accepted: 11/15/2017] [Indexed: 02/05/2023] Open
Abstract
TGF-β 1-3 are unique multi-functional growth factors that are only expressed in mammals, and mainly secreted and stored as a latent complex in the extracellular matrix (ECM). The biological functions of TGF-β in adults can only be delivered after ligand activation, mostly in response to environmental perturbations. Although involved in multiple biological and pathological processes of the human body, the exact roles of TGF-β in maintaining stem cells and tissue homeostasis have not been well-documented until recent advances, which delineate their functions in a given context. Our recent findings, along with data reported by others, have clearly shown that temporal and spatial activation of TGF-β is involved in the recruitment of stem/progenitor cell participation in tissue regeneration/remodeling process, whereas sustained abnormalities in TGF-β ligand activation, regardless of genetic or environmental origin, will inevitably disrupt the normal physiology and lead to pathobiology of major diseases. Modulation of TGF-β signaling with different approaches has proven effective pre-clinically in the treatment of multiple pathologies such as sclerosis/fibrosis, tumor metastasis, osteoarthritis, and immune disorders. Thus, further elucidation of the mechanisms by which TGF-β is activated in different tissues/organs and how targeted cells respond in a context-dependent way can likely be translated with clinical benefits in the management of a broad range of diseases with the involvement of TGF-β.
Collapse
Affiliation(s)
- Xin Xu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liwei Zheng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Gehua Zhen
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Janet L. Crane
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD USA
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD USA
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xu Cao
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD USA
| |
Collapse
|
273
|
Ramachandran A, Vizán P, Das D, Chakravarty P, Vogt J, Rogers KW, Müller P, Hinck AP, Sapkota GP, Hill CS. TGF-β uses a novel mode of receptor activation to phosphorylate SMAD1/5 and induce epithelial-to-mesenchymal transition. eLife 2018; 7:e31756. [PMID: 29376829 PMCID: PMC5832415 DOI: 10.7554/elife.31756] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 01/26/2018] [Indexed: 12/15/2022] Open
Abstract
The best characterized signaling pathway downstream of transforming growth factor β (TGF-β) is through SMAD2 and SMAD3. However, TGF-β also induces phosphorylation of SMAD1 and SMAD5, but the mechanism of this phosphorylation and its functional relevance is not known. Here, we show that TGF-β-induced SMAD1/5 phosphorylation requires members of two classes of type I receptor, TGFBR1 and ACVR1, and establish a new paradigm for receptor activation where TGFBR1 phosphorylates and activates ACVR1, which phosphorylates SMAD1/5. We demonstrate the biological significance of this pathway by showing that approximately a quarter of the TGF-β-induced transcriptome depends on SMAD1/5 signaling, with major early transcriptional targets being the ID genes. Finally, we show that TGF-β-induced epithelial-to-mesenchymal transition requires signaling via both the SMAD3 and SMAD1/5 pathways, with SMAD1/5 signaling being essential to induce ID1. Therefore, combinatorial signaling via both SMAD pathways is essential for the full TGF-β-induced transcriptional program and physiological responses.
Collapse
Affiliation(s)
| | - Pedro Vizán
- Developmental Signalling LaboratoryThe Francis Crick InstituteLondonUnited Kingdom
| | - Debipriya Das
- Developmental Signalling LaboratoryThe Francis Crick InstituteLondonUnited Kingdom
| | - Probir Chakravarty
- Bioinformatics and Biostatistics FacilityThe Francis Crick InstituteLondonUnited Kingdom
| | - Janis Vogt
- Medical Research Council Protein Phosphorylation and Ubiquitylation UnitUniversity of DundeeDundeeUnited Kingdom
| | | | - Patrick Müller
- Friedrich Miescher Laboratory of the Max Planck SocietyTübingenGermany
| | - Andrew P Hinck
- Department of Structural BiologyUniversity of Pittsburgh School of MedicinePittsburghUnited States
| | - Gopal P Sapkota
- Medical Research Council Protein Phosphorylation and Ubiquitylation UnitUniversity of DundeeDundeeUnited Kingdom
| | - Caroline S Hill
- Developmental Signalling LaboratoryThe Francis Crick InstituteLondonUnited Kingdom
| |
Collapse
|
274
|
Lu Z, Chen Z, Li Y, Wang J, Zhang Z, Che Y, Huang J, Sun S, Mao S, Lei Y, Gao Y, He J. TGF-β-induced NKILA inhibits ESCC cell migration and invasion through NF-κB/MMP14 signaling. J Mol Med (Berl) 2018; 96:301-313. [PMID: 29379981 PMCID: PMC5859688 DOI: 10.1007/s00109-018-1621-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 11/27/2017] [Accepted: 01/15/2018] [Indexed: 12/18/2022]
Abstract
The transforming growth factor β (TGF-β) signaling pathway plays anti- and pro-tumoral roles in the vast majority of cancers, and long noncoding RNAs have been reported to play key roles in the highly contextual response process. However, the roles of long noncoding RNAs (lncRNAs) in TGF-β signaling in esophageal squamous cell carcinoma (ESCC) remain unknown. In this study, we performed RNA-seq to compare lncRNAs expression levels between TGF-β1-treated and untreated ESCC cells and observed that NF-kappaB-interacting lncRNA (NKILA) was remarkably upregulated by the classical TGF-β signaling pathway. RNA profiling of 39 pairs ESCC tumor and adjacent nontumor samples using RT-qPCR demonstrated that NKILA is significantly downregulated in ESCC tumor tissues, and NKILA expression levels were significantly decreased in advanced tumor tissues (III and IV) compared to early stages (I and II) (p < 0.01). Gain- and loss-of-function assays showed that NKILA inhibited ESCC cell metastasis in vitro and in vivo, and mechanism studies showed that NKILA repressed MMP14 expression by inhibiting IκBα phosphorylation and NF-κB activation. Collectively, these findings suggest that the TGF-β-induced lncRNA NKILA has potential as an antimetastasis therapy. KEY MESSAGES Long noncoding RNA NKILA could be remarkably upregulated by classical TGF-β signal pathway in ESCC. NKILA was significantly downregulated in esophageal squamous cell carcinoma and negatively correlated with TNM stage. NKILA inhibits ESCC cell metastasis via repressing MMP14 expression by suppressing the phosphorylation of IκBα and NF-κB activation.
Collapse
Affiliation(s)
- Zhiliang Lu
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, 100021, People's Republic of China
| | - Zhaoli Chen
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, 100021, People's Republic of China
| | - Yuan Li
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, 100021, People's Republic of China
| | - Jingnan Wang
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, 100021, People's Republic of China
| | - Zhirong Zhang
- Thoracic Surgery Department, Beijing Chao-Yang Hospital, Capital University of Medical Science, Beijing, 100020, China
| | - Yun Che
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, 100021, People's Republic of China
| | - Jianbing Huang
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, 100021, People's Republic of China
| | - Shouguo Sun
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, 100021, People's Republic of China
| | - Shuangshuang Mao
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, 100021, People's Republic of China
| | - Yuanyuan Lei
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, 100021, People's Republic of China
| | - Yibo Gao
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, 100021, People's Republic of China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, 100021, People's Republic of China.
| |
Collapse
|
275
|
Yingling JM, McMillen WT, Yan L, Huang H, Sawyer JS, Graff J, Clawson DK, Britt KS, Anderson BD, Beight DW, Desaiah D, Lahn MM, Benhadji KA, Lallena MJ, Holmgaard RB, Xu X, Zhang F, Manro JR, Iversen PW, Iyer CV, Brekken RA, Kalos MD, Driscoll KE. Preclinical assessment of galunisertib (LY2157299 monohydrate), a first-in-class transforming growth factor-β receptor type I inhibitor. Oncotarget 2018; 9:6659-6677. [PMID: 29467918 PMCID: PMC5805504 DOI: 10.18632/oncotarget.23795] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 12/21/2017] [Indexed: 01/29/2023] Open
Abstract
Transforming growth factor-β (TGFβ) is an important driver of tumor growth via intrinsic and extrinsic mechanisms, and is therefore an attractive target for developing cancer therapeutics. Using preclinical models, we characterized the anti-tumor activity of a small molecule inhibitor of TGFβ receptor I (TGFβRI), galunisertib (LY2157299 monohydrate). Galunisertib demonstrated potent and selective inhibition of TGFβRI with corresponding inhibition of downstream signaling via inhibition of SMAD phosphorylation (pSMAD). Galunisertib also inhibited TGFβ-induced pSMAD in vivo, which enabled a pharmacokinetic/pharmacodynamic profile in Calu6 and EMT6-LM2 tumors. Galunisertib demonstrated anti-tumor activity including inhibition of tumor cell migration and mesenchymal phenotype, reversal of TGFβ-mediated immune-suppression, and tumor growth delay. A concentration-effect relationship was established with a dosing schedule to achieve the optimal level of target modulation. Finally, a rat model demonstrated a correlation between galunisertib-dependent inhibition of pSMAD in tumor tissues and in PBMCs, supporting the use of PBMCs for assessing pharmacodynamic effects. Galunisertib has been tested in several clinical studies with evidence of anti-tumor activity observed in subsets of patients. Here, we demonstrate that galunisertib inhibits a number of TGFβ-dependent functions leading to anti-tumor activity. The enhanced understanding of galunisertib provides rationale for further informed clinical development of TGFβ pathway inhibitors.
Collapse
Affiliation(s)
| | - William T. McMillen
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN and New York, NY, USA
| | - Lei Yan
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN and New York, NY, USA
| | - Huocong Huang
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - J. Scott Sawyer
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN and New York, NY, USA
| | - Jeremy Graff
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN and New York, NY, USA
| | - David K. Clawson
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN and New York, NY, USA
| | - Karen S. Britt
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN and New York, NY, USA
| | - Bryan D. Anderson
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN and New York, NY, USA
| | - Douglas W. Beight
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN and New York, NY, USA
| | - Durisala Desaiah
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN and New York, NY, USA
| | - Michael M. Lahn
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN and New York, NY, USA
| | - Karim A. Benhadji
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN and New York, NY, USA
| | - Maria J. Lallena
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN and New York, NY, USA
| | - Rikke B. Holmgaard
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN and New York, NY, USA
| | - Xiaohong Xu
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN and New York, NY, USA
| | - Faming Zhang
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN and New York, NY, USA
| | - Jason R. Manro
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN and New York, NY, USA
| | - Philip W. Iversen
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN and New York, NY, USA
| | - Chandrasekar V. Iyer
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN and New York, NY, USA
| | - Rolf A. Brekken
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Michael D. Kalos
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN and New York, NY, USA
| | - Kyla E. Driscoll
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN and New York, NY, USA
| |
Collapse
|
276
|
Grelet S, Geslain R, Howe PH. EMT does not work regular shifts. Cell Cycle 2018; 17:141-142. [PMID: 29334820 DOI: 10.1080/15384101.2017.1414684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Affiliation(s)
- Simon Grelet
- a Department of Biochemistry & Molecular Biology , Medical University of South Carolina , Charleston , South Carolina , USA.,b Hollings Cancer Center , Medical University of South Carolina , Charleston , South Carolina , USA
| | - Renaud Geslain
- c Laboratory of tRNA Biology, Department of Biology , College of Charleston , Charleston , SC , USA
| | - Philip H Howe
- a Department of Biochemistry & Molecular Biology , Medical University of South Carolina , Charleston , South Carolina , USA.,b Hollings Cancer Center , Medical University of South Carolina , Charleston , South Carolina , USA
| |
Collapse
|
277
|
Nayak KB, Sajitha IS, Kumar TRS, Chakraborty S. Ecotropic viral integration site 1 promotes metastasis independent of epithelial mesenchymal transition in colon cancer cells. Cell Death Dis 2018; 9:18. [PMID: 29339729 PMCID: PMC5833819 DOI: 10.1038/s41419-017-0036-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 10/08/2017] [Accepted: 10/09/2017] [Indexed: 12/17/2022]
Abstract
The most indecipherable component of solid cancer is the development of metastasis which accounts for more than 90% of cancer-related mortalities. A developmental program termed epithelial-mesenchymal transition (EMT) has also been shown to play a critical role in promoting metastasis in epithelium-derived solid tumors. By analyzing publicly available microarray datasets, we observed that ecotropic viral integration site 1 (EVI1) correlates negatively with SLUG, a master regulator of EMT. This correlation was found to be relevant as we demonstrated that EVI1 binds to SLUG promoter element directly through the distal set of zinc fingers and downregulates its expression. Many studies have shown that the primary role of SLUG during EMT and EMT-like processes is the regulation of cell motility in most of the cancer cells. Knockdown of EVI1 in metastatic colon cancer cell and subsequent passage through matrigel not only increased the invading capacity but also induced an EMT-like morphological feature of the cells, such as spindle-shaped appearance and led to a significant reduction in the expression of the epithelial marker, E-CADHERIN and increase in the expression of the mesenchymal marker, N-CADHERIN. The cells, when injected into immunocompromised mice, failed to show any metastatic foci in distant organs however the ones with EVI1, metastasized in the intraperitoneal layer and also showed multiple micro metastatic foci in the lungs and spleen. These findings suggest that in colon cancer EVI1 is dispensable for epithelial-mesenchymal transition, however, is required for metastasis.
Collapse
Affiliation(s)
- Kasturi Bala Nayak
- Department of Gene Function and Regulation, Institute of Life Sciences Nalco Square, Bhubaneswar, Odisha, India
| | - I S Sajitha
- Department of Veterinary Pathology, College of Veterinary & Animal Sciences, Wayanad, Kerala, India
| | - T R Santhosh Kumar
- Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Soumen Chakraborty
- Department of Gene Function and Regulation, Institute of Life Sciences Nalco Square, Bhubaneswar, Odisha, India.
| |
Collapse
|
278
|
Proteinase-activated receptor 2 promotes TGF-β-dependent cell motility in pancreatic cancer cells by sustaining expression of the TGF-β type I receptor ALK5. Oncotarget 2018; 7:41095-41109. [PMID: 27248167 PMCID: PMC5173045 DOI: 10.18632/oncotarget.9600] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 05/14/2016] [Indexed: 12/14/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by high expression of transforming growth factor (TGF)-β and the G protein-coupled receptor proteinase-activated receptor 2 (PAR2), the latter of which functions as a cell-surface sensor for serine proteinases asscociated with the tumour microenvironment. Since TGF-β and PAR2 affect tumourigenesis by regulating migration, invasion and metastasis, we hypothesized that there is signalling crosstalk between them. Depleting PDAC and non-PDAC cells of PAR2 by RNA interference strongly decreased TGF-β1-induced activation of Smad2/3 and p38 mitogen-activated protein kinase, Smad dependent transcriptional activity, expression of invasion associated genes, and cell migration/invasion in vitro. Likewise, the plasminogen activator-inhibitor 1 gene in primary cultures of aortic smooth muscle cells from PAR2-/- mice displayed a greatly attenuated sensitivity to TGF-β1 stimulation. PAR2 depletion in PDAC cells resulted in reduced protein and mRNA levels of the TGF-β type I receptor activin receptor-like kinase 5 (ALK5). Forced expression of wild-type ALK5 or a kinase-active ALK5 mutant, but not a kinase-active but Smad-binding defective ALK5 mutant, was able to rescue TGF-β1-induced Smad3 activation, Smad dependent transcription, and cell migration in PAR2-depleted cells. Together, our data show that PAR2 is crucial for TGF-β1-induced cell motility by its ability to sustain expression of ALK5. Therapeutically targeting PAR2 may thus be a promising approach in preventing TGF-β-dependent driven metastatic dissemination in PDAC and possibly other stroma-rich tumour types.
Collapse
|
279
|
Loss of TGF-β signaling in osteoblasts increases basic-FGF and promotes prostate cancer bone metastasis. Cancer Lett 2018; 418:109-118. [PMID: 29337106 DOI: 10.1016/j.canlet.2018.01.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 01/02/2018] [Accepted: 01/08/2018] [Indexed: 01/02/2023]
Abstract
TGF-β plays a central role in prostate cancer (PCa) bone metastasis, and it is crucial to understand the bone cell-specific role of TGF-β signaling in this process. Thus, we used knockout (KO) mouse models having deletion of the Tgfbr2 gene specifically in osteoblasts (Tgfbr2Col1CreERT KO) or in osteoclasts (Tgfbr2LysMCre KO). We found that PCa-induced bone lesion development was promoted in the Tgfbr2Col1CreERT KO mice, but was inhibited in the Tgfbr2LysMCre KO mice, relative to their respective control Tgfbr2FloxE2 littermates. Since metastatic PCa cells attach to osteoblasts when colonized in the bone microenvironment, we focused on the mechanistic studies using the Tgfbr2Col1CreERT KO mouse model. We found that bFGF was upregulated in osteoblasts from PC3-injected tibiae of Tgfbr2Col1CreERT KO mice and correlated with increased tumor cell proliferation, angiogenesis, amounts of cancer-associated fibroblasts and osteoclasts. In vitro studies showed that osteoblastogenesis was inhibited, osteoclastogenesis was stimulated, but PC3 viability was not affected, by bFGF treatments. Lastly, the increased PC3-induced bone lesions in Tgfbr2Col1CreERT KO mice were significantly attenuated by blocking bFGF using neutralizing antibody, suggesting bFGF is a promising target inhibiting bone metastasis.
Collapse
|
280
|
Bae GY, Hong SK, Park JR, Kwon OS, Kim KT, Koo J, Oh E, Cha HJ. Chronic TGFβ stimulation promotes the metastatic potential of lung cancer cells by Snail protein stabilization through integrin β3-Akt-GSK3β signaling. Oncotarget 2018; 7:25366-76. [PMID: 27015122 PMCID: PMC5041910 DOI: 10.18632/oncotarget.8295] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/07/2016] [Indexed: 01/30/2023] Open
Abstract
Chronic exposure to TGFβ, a frequent occurrence for tumor cells in the tumor microenvironment, confers more aggressive phenotypes on cancer cells by promoting their invasion and migration while at the same time increasing their resistance to the growth-inhibitory effect of TGFβ. In this study, a transdifferentiated (TD) A549 cell model, established by chronically exposing A549 cells to TGFβ, showed highly invasive phenotypes in conjunction with attenuation of Smad-dependent signaling. We show that Snail protein, the mRNA expression of which strongly correlates with a poor prognosis in lung cancer patients, was highly stable in TD cells after TGFβ stimulation. The increased protein stability of Snail in TD cells correlated with elevated inhibitory phosphorylation of GSK3β, resulting from the high Akt activity. Notably, integrin β3, whose expression was markedly increased upon sustained exposure to TGFβ, was responsible for the high Akt activity as well as the increased Snail protein stability in TD cells. Consistently, clinical database analysis on lung cancer patients revealed a negative correlation between overall survival and integrin β3 mRNA levels. Therefore, we suggest that the integrin β3-Akt-GSK3β signaling axis plays an important role in non-canonical TGFβ signaling, determining the invasive properties of tumor cells chronically exposed to TGFβ.
Collapse
Affiliation(s)
- Gab-Yong Bae
- College of Natural Sciences, Department of Life Sciences, Sogang University, Seoul, Republic of Korea
| | - Soon-Ki Hong
- College of Natural Sciences, Department of Life Sciences, Sogang University, Seoul, Republic of Korea
| | - Jeong-Rak Park
- College of Natural Sciences, Department of Life Sciences, Sogang University, Seoul, Republic of Korea
| | - Ok-Seon Kwon
- College of Natural Sciences, Department of Life Sciences, Sogang University, Seoul, Republic of Korea
| | - Keun-Tae Kim
- College of Natural Sciences, Department of Life Sciences, Sogang University, Seoul, Republic of Korea
| | - JaeHyung Koo
- Department of Brain and Cognitive Sciences, DGIST, Daegu, Republic of Korea
| | - Ensel Oh
- Laboratory of Cancer Genomics and Molecular Pathology, Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Republic of Korea
| | - Hyuk-Jin Cha
- College of Natural Sciences, Department of Life Sciences, Sogang University, Seoul, Republic of Korea
| |
Collapse
|
281
|
Mali AV, Joshi AA, Hegde MV, Kadam SS. Enterolactone modulates the ERK/NF-κB/Snail signaling pathway in triple-negative breast cancer cell line MDA-MB-231 to revert the TGF-β-induced epithelial-mesenchymal transition. Cancer Biol Med 2018; 15:137-156. [PMID: 29951338 PMCID: PMC5994556 DOI: 10.20892/j.issn.2095-3941.2018.0012] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Objective Triple-negative breast cancer (TNBC) is highly metastatic, and there is an urgent unmet need to develop novel therapeutic strategies leading to the new drug discoveries against metastasis. The transforming growth factor-β (TGF-β) is known to promote the invasive and migratory potential of breast cancer cells through induction of epithelial-mesenchymal transition (EMT) via the ERK/NF-κB/Snail signaling pathway, leading to breast cancer metastasis. Targeting this pathway to revert the EMT would be an attractive, novel therapeutic strategy to halt breast cancer metastasis. Methods Effects of enterolactone (EL) on the cell cycle and apoptosis were investigated using flow cytometry and a cleaved caspase-3 enzyme-linked immunosorbent assay (ELISA), respectively. Effects of TGF-β induction and EL treatment on the functional malignancy of MDA-MB-231 breast cancer cells were investigated using migration and chemo-invasion assays. The effects of EL on EMT markers and the ERK/NF-κB/Snail signaling pathway after TGF-β induction were studied using confocal microscopy, quantitative reverse transcription polymerase chain reaction (qRT-PCR), Western blot, and flow cytometry. Results Herein, we report that EL exhibits a significant antimetastatic effect on MDA-MB-231 cells by almost reverting the TGF-β-induced EMT in vitro. EL downregulates the mesenchymal markers N-cadherin and vimentin, and upregulates the epithelial markers E-cadherin and occludin. It represses actin stress fiber formation via inhibition of mitogen-activated protein kinase p-38 (MAPK-p38) and cluster of differentiation 44 (CD44). EL also suppresses ERK-1/2, NF-κB, and Snail at the mRNA and protein levels. Conclusions Briefly, EL was found to inhibit TGF-β-induced EMT by blocking the ERK/NF-κB/Snail signaling pathway, which is a promising target for breast cancer metastasis therapy.
Collapse
Affiliation(s)
- Aniket V Mali
- Center for Innovation in Nutrition Health and Disease (CINHD), Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth Deemed to be University (BVDU), Dhankawadi, Pune, Maharashtra 411043, India.,Pharmaceutical Sciences, Poona College of Pharmacy, Bharati Vidyapeeth Deemed to be University (BVDU), Pune, Maharashtra 411038, India
| | - Asavari A Joshi
- Center for Innovation in Nutrition Health and Disease (CINHD), Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth Deemed to be University (BVDU), Dhankawadi, Pune, Maharashtra 411043, India
| | - Mahabaleshwar V Hegde
- Center for Innovation in Nutrition Health and Disease (CINHD), Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth Deemed to be University (BVDU), Dhankawadi, Pune, Maharashtra 411043, India
| | - Shivajirao S Kadam
- Pharmaceutical Sciences, Poona College of Pharmacy, Bharati Vidyapeeth Deemed to be University (BVDU), Pune, Maharashtra 411038, India
| |
Collapse
|
282
|
Ungefroren H, Witte D, Fiedler C, Gädeken T, Kaufmann R, Lehnert H, Gieseler F, Rauch BH. The Role of PAR2 in TGF-β1-Induced ERK Activation and Cell Motility. Int J Mol Sci 2017; 18:ijms18122776. [PMID: 29261154 PMCID: PMC5751374 DOI: 10.3390/ijms18122776] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/08/2017] [Accepted: 12/15/2017] [Indexed: 12/12/2022] Open
Abstract
Background: Recently, the expression of proteinase-activated receptor 2 (PAR2) has been shown to be essential for activin receptor-like kinase 5 (ALK5)/SMAD-mediated signaling and cell migration by transforming growth factor (TGF)-β1. However, it is not known whether activation of non-SMAD TGF-β signaling (e.g., RAS–RAF–MEK–extracellular signal-regulated kinase (ERK) signaling) is required for cell migration and whether it is also dependent on PAR2. Methods: RNA interference was used to deplete cells of PAR2, followed by xCELLigence technology to measure cell migration, phospho-immunoblotting to assess ERK1/2 activation, and co-immunoprecipitation to detect a PAR2–ALK5 physical interaction. Results: Inhibition of ERK signaling with the MEK inhibitor U0126 blunted the ability of TGF-β1 to induce migration in pancreatic cancer Panc1 cells. ERK activation in response to PAR2 agonistic peptide (PAR2–AP) was strong and rapid, while it was moderate and delayed in response to TGF-β1. Basal and TGF-β1-dependent ERK, but not SMAD activation, was blocked by U0126 in Panc1 and other cell types indicating that ERK activation is downstream or independent of SMAD signaling. Moreover, cellular depletion of PAR2 in HaCaT cells strongly inhibited TGF-β1-induced ERK activation, while the biased PAR2 agonist GB88 at 10 and 100 µM potentiated TGF-β1-dependent ERK activation and cell migration. Finally, we provide evidence for a physical interaction between PAR2 and ALK5. Our data show that both PAR2–AP- and TGF-β1-induced cell migration depend on ERK activation, that PAR2 expression is crucial for TGF-β1-induced ERK activation, and that the functional cooperation of PAR2 and TGF-β1 involves a physical interaction between PAR2 and ALK5.
Collapse
Affiliation(s)
- Hendrik Ungefroren
- First Department of Medicine, UKSH, Campus Lübeck, 23538 Lübeck, Germany.
- Department of General and Thoracic Surgery, UKSH, Campus Kiel, 24105 Kiel, Germany.
| | - David Witte
- First Department of Medicine, UKSH, Campus Lübeck, 23538 Lübeck, Germany.
| | - Christian Fiedler
- First Department of Medicine, UKSH, Campus Lübeck, 23538 Lübeck, Germany.
| | - Thomas Gädeken
- First Department of Medicine, UKSH, Campus Lübeck, 23538 Lübeck, Germany.
| | - Roland Kaufmann
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, 07747 Jena, Germany.
| | - Hendrik Lehnert
- First Department of Medicine, UKSH, Campus Lübeck, 23538 Lübeck, Germany.
| | - Frank Gieseler
- First Department of Medicine, UKSH, Campus Lübeck, 23538 Lübeck, Germany.
| | - Bernhard H Rauch
- Institute of Pharmacology, Department of General Pharmacology, University Medicine Greifswald, 17487 Greifswald, Germany.
| |
Collapse
|
283
|
Nafamostat mesilate negatively regulates the metastasis of triple-negative breast cancer cells. Arch Pharm Res 2017; 41:229-242. [PMID: 29196918 DOI: 10.1007/s12272-017-0996-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 11/26/2017] [Indexed: 12/11/2022]
Abstract
Triple-negative breast cancer (TNBC) lacking of oestrogen receptor, progesterone receptor, and epidermal growth factor receptor type 2 is a highly malignant disease which results in a poor prognosis and rare treatment options. Despite the use of conventional chemotherapy for TNBC tumours, resistance and short duration responses limit the treatment efficacy. Therefore, a need exists to develop a new chemotherapy for TNBC. The aim of this study was to examine the anti-cancer effects of nafamostat mesilate (NM), a previously known serine protease inhibitor and highly safe drug on breast cancer cells. Here, we showed that NM significantly inhibits proliferation, migration, and invasion in MDA-MB231 cells, induces G2/M phase cell-cycle arrest, and inhibits the expression of cyclin-dependent kinase 1 (CDK1). Exposure of MDA-MB231 cells to NM also resulted in decreased transcription factor activities accompanied by the regulated phosphorylation of signalling molecules and a decrease in metalloproteinases, the principal modulators of the extracellular environment during cancer progression. Especially, inhibition of TGFβ-stimulated Smad2 phosphorylation and subsequent metastasis-related gene expression, and downregulation of ERK activity may be pivotal mechanisms underlying inhibitory effects of NM on NM inhibits lung metastasis of breast cancer cells and growth of colonized tumours in mice. Taken together, our data revealed that NM inhibits cell growth and metastasis of TNBC cells and indicated that NM is a multi-targeted drug that could be an adjunct therapy for TNBC treatment.
Collapse
|
284
|
Short SP, Kondo J, Smalley-Freed WG, Takeda H, Dohn MR, Powell AE, Carnahan RH, Washington MK, Tripathi M, Payne DM, Jenkins NA, Copeland NG, Coffey RJ, Reynolds AB. p120-Catenin is an obligate haploinsufficient tumor suppressor in intestinal neoplasia. J Clin Invest 2017; 127:4462-4476. [PMID: 29130932 PMCID: PMC5707165 DOI: 10.1172/jci77217] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 10/03/2017] [Indexed: 11/17/2022] Open
Abstract
p120-Catenin (p120) functions as a tumor suppressor in intestinal cancer, but the mechanism is unclear. Here, using conditional p120 knockout in Apc-sensitized mouse models of intestinal cancer, we have identified p120 as an "obligatory" haploinsufficient tumor suppressor. Whereas monoallelic loss of p120 was associated with a significant increase in tumor multiplicity, loss of both alleles was never observed in tumors from these mice. Moreover, forced ablation of the second allele did not further enhance tumorigenesis, but instead induced synthetic lethality in combination with Apc loss of heterozygosity. In tumor-derived organoid cultures, elimination of both p120 alleles resulted in caspase-3-dependent apoptosis that was blocked by inhibition of Rho kinase (ROCK). With ROCK inhibition, however, p120-ablated organoids exhibited a branching phenotype and a substantial increase in cell proliferation. Access to data from Sleeping Beauty mutagenesis screens afforded an opportunity to directly assess the tumorigenic impact of p120 haploinsufficiency relative to other candidate drivers. Remarkably, p120 ranked third among the 919 drivers identified. Cofactors α-catenin and epithelial cadherin (E-cadherin) were also among the highest scoring candidates, indicating a mechanism at the level of the intact complex that may play an important role at very early stages of of intestinal tumorigenesis while simultaneously restricting outright loss via synthetic lethality.
Collapse
Affiliation(s)
| | - Jumpei Kondo
- Department of Medicine, Vanderbilt University Medical School, Nashville, Tennessee, USA
| | | | - Haruna Takeda
- Division of Genetics and Genomics, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
- Department of Oncologic Pathology, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Michael R. Dohn
- Department of Cancer Biology, and
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
| | - Anne E. Powell
- Department of Medicine, Vanderbilt University Medical School, Nashville, Tennessee, USA
| | | | - Mary K. Washington
- Department of Pathology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - D. Michael Payne
- CU Systems Biology Center, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Nancy A. Jenkins
- Division of Genetics and Genomics, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
- Cancer Research Program, The Methodist Hospital Research Institute, Houston, Texas, USA
| | - Neal G. Copeland
- Division of Genetics and Genomics, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
- Cancer Research Program, The Methodist Hospital Research Institute, Houston, Texas, USA
| | - Robert J. Coffey
- Department of Medicine, Vanderbilt University Medical School, Nashville, Tennessee, USA
- Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
- Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | | |
Collapse
|
285
|
Qiao X, Rao P, Zhang Y, Liu L, Pang M, Wang H, Hu M, Tian X, Zhang J, Zhao Y, Wang XM, Wang C, Yu H, Guo F, Cao Q, Wang Y, Wang YM, Zhang GY, Lee VW, Alexander SI, Zheng G, Harris DCH. Redirecting TGF- β Signaling through the β-Catenin/Foxo Complex Prevents Kidney Fibrosis. J Am Soc Nephrol 2017; 29:557-570. [PMID: 29180394 DOI: 10.1681/asn.2016121362] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Accepted: 10/25/2017] [Indexed: 01/09/2023] Open
Abstract
TGF-β is a key profibrotic factor, but targeting TGF-β to prevent fibrosis also abolishes its protective anti-inflammatory effects. Here, we investigated the hypothesis that we can redirect TGF-β signaling by preventing downstream profibrotic interaction of β-catenin with T cell factor (TCF), thereby enhancing the interaction of β-catenin with Foxo, a transcription factor that controls differentiation of TGF-β induced regulatory T cells (iTregs), and thus, enhance anti-inflammatory effects of TGF-β In iTregs derived from EL4 T cells treated with recombinant human TGF-β1 (rhTGF-β1) in vitro, inhibition of β-catenin/TCF transcription with ICG-001 increased Foxp3 expression, interaction of β-catenin and Foxo1, binding of Foxo1 to the Foxp3 promoter, and Foxo transcriptional activity. Moreover, the level of β-catenin expression positively correlated with the level of Foxo1 binding to the Foxp3 promoter and Foxo transcriptional activity. T cell fate mapping in Foxp3gfp Ly5.1/5.2 mice revealed that coadministration of rhTGF-β1 and ICG-001 further enhanced the expansion of iTregs and natural Tregs observed with rhTGF-β1 treatment alone. Coadministration of rhTGF-β1 with ICG-001 also increased the number of Tregs and reduced inflammation and fibrosis in the kidney fibrosis models of unilateral ureteric obstruction and ischemia-reperfusion injury. Notably, ICG-001 prevented the fibrosis in distant organs (lung and liver) caused by rhTGF-β1. Together, our results show that diversion of β-catenin from TCF- to Foxo-mediated transcription inhibits the β-catenin/TCF-mediated profibrotic effects of TGF-β while enhancing the β-catenin/Foxo-mediated anti-inflammatory effects. Targeting β-catenin/Foxo may be a novel therapeutic strategy in the treatment of fibrotic diseases that lead to organ failure.
Collapse
Affiliation(s)
- Xi Qiao
- Centre for Transplant and Renal Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia.,Department of Nephrology, Shanxi Kidney Disease Institute and
| | - Padmashree Rao
- Centre for Transplant and Renal Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Yun Zhang
- Centre for Transplant and Renal Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia.,Experimental Centre of Science and Research and
| | - Lixin Liu
- Centre for Transplant and Renal Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia.,Experimental Centre of Science and Research and
| | - Min Pang
- Centre for Transplant and Renal Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia.,Department of Respiratory Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| | - Hailong Wang
- Centre for Transplant and Renal Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia.,Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| | - Min Hu
- Centre for Transplant and Renal Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Xinrui Tian
- Centre for Transplant and Renal Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia.,Department of Respiratory Medicine, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| | - Jianlin Zhang
- Centre for Transplant and Renal Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia.,Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| | - Ye Zhao
- Centre for Transplant and Renal Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | | | - Chengshi Wang
- Centre for Transplant and Renal Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Hong Yu
- Cell Imaging Facility, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia; and
| | - Fei Guo
- Centre for Transplant and Renal Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Qi Cao
- Centre for Transplant and Renal Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Yiping Wang
- Centre for Transplant and Renal Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Yuan Min Wang
- Centre for Kidney Research, Kids Research Institute, The Children's Hospital at Westmead, New South Wales, Australia
| | - Geoff Yu Zhang
- Centre for Kidney Research, Kids Research Institute, The Children's Hospital at Westmead, New South Wales, Australia
| | - Vincent W Lee
- Centre for Transplant and Renal Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Stephen I Alexander
- Centre for Kidney Research, Kids Research Institute, The Children's Hospital at Westmead, New South Wales, Australia
| | - Guoping Zheng
- Centre for Transplant and Renal Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia;
| | - David C H Harris
- Centre for Transplant and Renal Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
286
|
Qiu L, Wang T, Xu X, Wu Y, Tang Q, Chen K. Long Non-Coding RNAs in Hepatitis B Virus-Related Hepatocellular Carcinoma: Regulation, Functions, and Underlying Mechanisms. Int J Mol Sci 2017; 18:ijms18122505. [PMID: 29168767 PMCID: PMC5751108 DOI: 10.3390/ijms18122505] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 11/13/2017] [Accepted: 11/20/2017] [Indexed: 12/19/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common cancer and the third leading cause of cancer death in the world. Hepatitis B virus (HBV) and its X gene-encoded protein (HBx) play important roles in the progression of HCC. Although long non-coding RNAs (lncRNAs) cannot encode proteins, growing evidence indicates that they play essential roles in HCC progression, and contribute to cell proliferation, invasion and metastasis, autophagy, and apoptosis by targeting a large number of pivotal protein-coding genes, miRNAs, and signaling pathways. In this review, we briefly outline recent findings of differentially expressed lncRNAs in HBV-related HCC, with particular focus on several key lncRNAs, and discuss their regulation by HBV/HBx, their functions, and their underlying molecular mechanisms in the progression of HCC.
Collapse
Affiliation(s)
- Lipeng Qiu
- Institute of Life Sciences, Jiangsu University, Zhenjiang 212013, China.
| | - Tao Wang
- Institute of Life Sciences, Jiangsu University, Zhenjiang 212013, China.
| | - Xiuquan Xu
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China.
| | - Yihang Wu
- Department of Pharmacy, College of Life Sciences, China Jiliang University, Hangzhou 310018, China.
| | - Qi Tang
- Institute of Life Sciences, Jiangsu University, Zhenjiang 212013, China.
| | - Keping Chen
- Institute of Life Sciences, Jiangsu University, Zhenjiang 212013, China.
| |
Collapse
|
287
|
Rose M, Meurer SK, Kloten V, Weiskirchen R, Denecke B, Antonopoulos W, Deckert M, Knüchel R, Dahl E. ITIH5 induces a shift in TGF-β superfamily signaling involving Endoglin and reduces risk for breast cancer metastasis and tumor death. Mol Carcinog 2017; 57:167-181. [PMID: 28940371 DOI: 10.1002/mc.22742] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 09/07/2017] [Accepted: 09/18/2017] [Indexed: 12/26/2022]
Abstract
ITIH5 has been proposed being a novel tumor suppressor in various tumor entities including breast cancer. Recently, ITIH5 was furthermore identified as metastasis suppressor gene in pancreatic carcinoma. In this study we aimed to specify the impact of ITIH5 on metastasis in breast cancer. Therefore, DNA methylation of ITIH5 promoter regions was assessed in breast cancer metastases using the TCGA portal and methylation-specific PCR (MSP). We reveal that the ITIH5 upstream promoter region is particularly responsible for ITIH5 gene inactivation predicting shorter survival of patients. Notably, methylation of this upstream ITIH5 promoter region was associated with disease progression, for example, abundantly found in distant metastases. In vitro, stably ITIH5-overexpressing MDA-MB-231 breast cancer clones were used to analyze cell invasion and to identify novel ITIH5-downstream targets. Indeed, ITIH5 re-expression suppresses invasive growth of MDA-MB-231 breast cancer cells while modulating expression of genes involved in metastasis including Endoglin (ENG), an accessory TGF-β receptor, which was furthermore co-expressed with ITIH5 in primary breast tumors. By performing in vitro stimulation of TGF-β signaling using TGF-β1 and BMP-2 we show that ITIH5 triggered a TGF-β superfamily signaling switch contributing to downregulation of targets like Id1, known to endorse metastasis. Moreover, ITIH5 predicts longer overall survival (OS) only in those breast tumors that feature high ENG expression or inversely regulated ID1 suggesting a clinical and functional impact of an ITIH5-ENG axis for breast cancer progression. Hence, we provide evidence that ITIH5 may represent a novel modulator of TGF-β superfamily signaling involved in suppressing breast cancer metastasis.
Collapse
Affiliation(s)
- Michael Rose
- Institute of Pathology, Medical Faculty of the RWTH Aachen University, Aachen, Germany
| | - Steffen K Meurer
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, Medical Faculty of the RWTH Aachen University, Aachen, Germany
| | - Vera Kloten
- Institute of Pathology, Medical Faculty of the RWTH Aachen University, Aachen, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, Medical Faculty of the RWTH Aachen University, Aachen, Germany
| | - Bernd Denecke
- IZKF Aachen, Medical Faculty of the RWTH Aachen University, Aachen, Germany
| | - Wiebke Antonopoulos
- Institute of Pathology, Medical Faculty of the RWTH Aachen University, Aachen, Germany
| | - Martina Deckert
- Department of Neuropathology, University of Cologne, Cologne, Germany
| | - Ruth Knüchel
- Institute of Pathology, Medical Faculty of the RWTH Aachen University, Aachen, Germany
| | - Edgar Dahl
- Institute of Pathology, Medical Faculty of the RWTH Aachen University, Aachen, Germany
| |
Collapse
|
288
|
Abstract
Correct organization of the vascular tree requires the balanced activities of several signaling pathways that regulate tubulogenesis and vascular branching, elongation, and pruning. When this balance is lost, the vessels can be malformed and fragile, and they can lose arteriovenous differentiation. In this review, we concentrate on the transforming growth factor (TGF)-β/bone morphogenetic protein (BMP) pathway, which is one of the most important and complex signaling systems in vascular development. Inactivation of these pathways can lead to altered vascular organization in the embryo. In addition, many vascular malformations are related to deregulation of TGF-β/BMP signaling. Here, we focus on two of the most studied vascular malformations that are induced by deregulation of TGF-β/BMP signaling: hereditary hemorrhagic telangiectasia (HHT) and cerebral cavernous malformation (CCM). The first of these is related to loss-of-function mutation of the TGF-β/BMP receptor complex and the second to increased signaling sensitivity to TGF-β/BMP. In this review, we discuss the potential therapeutic targets against these vascular malformations identified so far, as well as their basis in general mechanisms of vascular development and stability.
Collapse
Affiliation(s)
- Sara I Cunha
- From the Department of Immunology, Genetics, and Pathology, Uppsala University, Sweden (S.I.C., P.U.M., E.D.); FIRC Institute of Molecular Oncology, Milan, Italy (E.D., M.G.L.); and Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy (M.G.L.)
| | - Peetra U Magnusson
- From the Department of Immunology, Genetics, and Pathology, Uppsala University, Sweden (S.I.C., P.U.M., E.D.); FIRC Institute of Molecular Oncology, Milan, Italy (E.D., M.G.L.); and Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy (M.G.L.)
| | - Elisabetta Dejana
- From the Department of Immunology, Genetics, and Pathology, Uppsala University, Sweden (S.I.C., P.U.M., E.D.); FIRC Institute of Molecular Oncology, Milan, Italy (E.D., M.G.L.); and Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy (M.G.L.).
| | - Maria Grazia Lampugnani
- From the Department of Immunology, Genetics, and Pathology, Uppsala University, Sweden (S.I.C., P.U.M., E.D.); FIRC Institute of Molecular Oncology, Milan, Italy (E.D., M.G.L.); and Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy (M.G.L.)
| |
Collapse
|
289
|
Jun F, Hong J, Liu Q, Guo Y, Liao Y, Huang J, Wen S, Shen L. Epithelial membrane protein 3 regulates TGF-β signaling activation in CD44-high glioblastoma. Oncotarget 2017; 8:14343-14358. [PMID: 27527869 PMCID: PMC5362410 DOI: 10.18632/oncotarget.11102] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 07/19/2016] [Indexed: 12/18/2022] Open
Abstract
Although epithelial membrane protein 3 (EMP3) has been implicated as a candidate tumor suppressor gene for low grade glioma, its biological function in glioblastoma multiforme (GBM) still remains poorly understood. Herein, we showed that EMP3 was highly expressed in CD44-high primary GBMs. Depletion of EMP3 expression suppressed cell proliferation, impaired in vitro tumorigenic potential and induced apoptosis in CD44-high GBM cell lines. We also identified TGF-β/Smad2/3 signaling pathway as a potential target of EMP3. EMP3 interacts with TGF-βreceptor type 2 (TGFBR2) upon TGF-βstimulation in GBM cells. Consequently, the EMP3-TGFBR2 interaction regulates TGF-β/Smad2/3 signaling activation and positively impacts on TGF-βstimulated gene expression and cell proliferation in vitro and in vivo. Highly correlated protein expression of EMP3 and TGF-β/Smad2/3 signaling pathway components was also observed in GBM specimens, confirming the clinical relevancy of activated EMP3/TGF-β/Smad2/3 signaling in GBM. In conclusion, our findings revealed that EMP3 might be a potential target for CD44-high GBMs and highlight the essential functions of EMP3 in TGF-β/Smad2/3 signaling activation and tumor progression.
Collapse
Affiliation(s)
- Fu Jun
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, P. R China
| | - Jidong Hong
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, P. R China
| | - Qin Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, P. R China
| | - Yong Guo
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, P. R China
| | - Yiwei Liao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, P. R China
| | - Jianghai Huang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, P. R China
| | - Sailan Wen
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, P. R China
| | - Liangfang Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, P. R China
| |
Collapse
|
290
|
De S, Das S, Mukherjee S, Das S, Sengupta Bandyopadhyay S. Establishment of twist-1 and TGFBR2 as direct targets of microRNA-20a in mesenchymal to epithelial transition of breast cancer cell-line MDA-MB-231. Exp Cell Res 2017; 361:85-92. [PMID: 28987542 DOI: 10.1016/j.yexcr.2017.10.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 10/03/2017] [Indexed: 12/16/2022]
Abstract
Messenchymal to epithelial transition (MET) is a significant physiological phenomenon involved in embryogenesis and cancer. This study aims at investigating the mechanism of microRNA-20a (miR-20a) mediated regulation of mesenchymal to epithelial transition and identification of its direct target genes in breast cancer cell-line, MDA-MB-231. Reduced migratory and invasive property, altered cellular morphology along with reduced capability for attachment to basement membrane was acquired by over-expression of miR-20a in invasive MDA-MB-231 cell-line initially expressing low level of this micro-RNA, indicating direct correlation between abundance of miR-20a and metastatic property. The switch from mesenchymal to epithelial cells mediated by miR-20a involved post-transcriptional down-regulation of twist1, which in turn controls downstream epithelial markers like E-cadherin, claudin and mesenchymal markers like N-cadherin, fibronectin, the crucial players of mesenchymal to epithelial transition (MET). Furthermore, another key component, TGF-β and one of its receptors (TGFBR2) were found to be down-regulated by miR-20a. Additionally, reporter assay established that post-transcriptional down-regulation of TGFBR2 occurred through direct binding of miR-20a to its 3'UTR, thus abrogating the TGF-β signaling pathway resulting in inhibition of MET. Delineating the underlying molecular mechanism of miR-20a-mediated MET and defining the target genes will help us to introduce a miRNA-mediated effective therapeutic strategy against breast cancer.
Collapse
Affiliation(s)
- Soumasree De
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92A.P.C. Road, Kolkata, WB 700009, India
| | - Sayantani Das
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92A.P.C. Road, Kolkata, WB 700009, India
| | - Srimoyee Mukherjee
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92A.P.C. Road, Kolkata, WB 700009, India
| | - Sainy Das
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92A.P.C. Road, Kolkata, WB 700009, India
| | - Sumita Sengupta Bandyopadhyay
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92A.P.C. Road, Kolkata, WB 700009, India.
| |
Collapse
|
291
|
Garcia-Areas R, Libreros S, Simoes M, Castro-Silva C, Gazaniga N, Amat S, Jaczewska J, Keating P, Schilling K, Brito M, Wojcikiewicz EP, Iragavarpu-Charyulu V. Suppression of tumor-derived Semaphorin 7A and genetic ablation of host-derived Semaphorin 7A impairs tumor progression in a murine model of advanced breast carcinoma. Int J Oncol 2017; 51:1395-1404. [PMID: 29048670 PMCID: PMC5642386 DOI: 10.3892/ijo.2017.4144] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 07/31/2017] [Indexed: 12/12/2022] Open
Abstract
Solid tumors can generate a plethora of neurogenesis-related molecules that enhance their growth and metastasis. Among them, we have identified axonal guidance molecule Semaphorin 7A (SEMA7A) in breast cancer. The goal of this study was to determine the therapeutic effect of suppressing SEMA7A levels in the 4T1 murine model of advanced breast carcinoma. We used anti-SEMA7A short hairpin RNA (shRNA) to gene silence SEMA7A in 4T1 mammary tumor cells. When implanted into the mammary fat pads of syngeneic mice, SEMA7A shRNA-expressing 4T1 tumors exhibited decreased growth rates, deferred metastasis and reduced mortality. In vitro, SEMA7A shRNA-expressing 4T1 cells had weakened proliferative, migratory and invasive abilities, and decreased levels of mesenchymal factors. Atomic force microscopy studies showed that SEMA7A shRNA-expressing 4T1 cells had an increase in cell stiffness that corresponded with their decreased malignant potential. Genetic ablation of host-derived SEMA7A further enhanced the antitumor effects of SEMA7A shRNA gene silencing in 4T1 cells. Our preclinical findings demonstrate a critical role for SEMA7A in mediating mammary tumor progression.
Collapse
Affiliation(s)
- R Garcia-Areas
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - S Libreros
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - M Simoes
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - C Castro-Silva
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - N Gazaniga
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - S Amat
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - J Jaczewska
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - P Keating
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - K Schilling
- Lynn Women's Health & Wellness Institute, Boca Raton Regional Hospital, Boca Raton, FL 33431, USA
| | - M Brito
- Department of Pathology, Boca Raton Regional Hospital, Boca Raton, FL 33431, USA
| | - E P Wojcikiewicz
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - V Iragavarpu-Charyulu
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| |
Collapse
|
292
|
The effect of CT26 tumor-derived TGF-β on the balance of tumor growth and immunity. Immunol Lett 2017; 191:47-54. [PMID: 28855127 DOI: 10.1016/j.imlet.2017.08.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Accepted: 08/23/2017] [Indexed: 01/10/2023]
Abstract
INTRODUCTION TGF-β is an important target for many cancer therapies under development. In addition to suppressing anti-tumor immunity, it has pleiotropic direct pro- and anti- tumor effects. The actions of increased endogenous TGF-β production remain unclear, and may affect the outcomes of anti-TGF-β cancer therapy. We hypothesize that tumor-derived TGF-β (td-TGF-β) plays an important role in maintaining tumor remission by controlling tumor proliferation in vivo, and that decreasing td-TGF-β in the tumor microenvironment will result in tumor progression. The aim of this study was to examine the effect of TGF-β in the tumor microenvironment on the balance between its anti-proliferative and immunosuppressive effects. METHODS A murine BALB/c spontaneous colon adenocarcinoma cell line (CT26) was genetically engineered to produce increased active TGF-β (CT26-TGF-β), a dominant-negative soluble TGF-β receptor (CT26-TGF-β-R), or the empty neomycin cassette as control (CT26-neo). In vitro proliferation rates were measured. For in vivo studies, the three cell lines were injected into syngeneic BALB/c mice, and tumor growth was measured over time. Immunodeficient BALB/c nude mice were used to investigate the role of T and B cells. RESULTS In vitro, CT26-TGF-β-R and CT26-TGF-β cells showed increased and suppressed proliferation, respectively, compared to control (CT26-neo), confirming TGF-β has direct anti-tumor effects. In vivo, we found that CT26-TGF-β-R cells displayed slower growth compared to control, likely secondary to reduced suppression of anti-tumor immunity, as this effect was ablated in immunodeficient BALB/c nude mice. However, CT26-TGF-β cells (excess TGF-β) exhibited rapid early growth compared to control, but later failed to progress. The same pattern was shown in immunodeficient BALB/c nude mice, suggesting the effect on tumor growth is direct, with minimal immune system involvement. There was minimal effect on systemic antitumor immunity as determined by peripheral antigen-specific splenocyte type 1 cytokine production and tumor growth rate of CT26-neo on the contralateral flank of the same mice. CONCLUSION Although TGF-β has opposing effects on tumor growth, this study showed that excessive td-TGF-β in the tumor microenvironment renders the tumor non-proliferative. Depleting excess td-TGF-β may release this endogenous tumor suppressive mechanism, thus triggering the progression of the tumor. Therefore, our findings support cautions against using anti-TGF-β strategies in treating cancer, as this may tip the balance of anti-immunity vs. anti-tumor effects of TGF-β, leading to tumor progression instead of remission.
Collapse
|
293
|
Abstract
Transforming growth factor βs (TGF-βs) are closely related ligands that have pleiotropic activity on most cell types of the body. They act through common heterotetrameric TGF-β type II and type I transmembrane dual specificity kinase receptor complexes, and the outcome of signaling is context-dependent. In normal tissue, they serve a role in maintaining homeostasis. In many diseased states, particularly fibrosis and cancer, TGF-β ligands are overexpressed and the outcome of signaling is diverted toward disease progression. There has therefore been a concerted effort to develop drugs that block TGF-β signaling for therapeutic benefit. This review will cover the basics of TGF-β signaling and its biological activities relevant to oncology, present a summary of pharmacological TGF-β blockade strategies, and give an update on preclinical and clinical trials for TGF-β blockade in a variety of solid tumor types.
Collapse
Affiliation(s)
- Rosemary J Akhurst
- Department of Anatomy and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California 94158-9001
| |
Collapse
|
294
|
Gallardo-Pérez JC, Adán-Ladrón de Guevara A, Marín-Hernández A, Moreno-Sánchez R, Rodríguez-Enríquez S. HPI/AMF inhibition halts the development of the aggressive phenotype of breast cancer stem cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017. [DOI: 10.1016/j.bbamcr.2017.06.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
295
|
Crow J, Atay S, Banskota S, Artale B, Schmitt S, Godwin AK. Exosomes as mediators of platinum resistance in ovarian cancer. Oncotarget 2017; 8:11917-11936. [PMID: 28060758 PMCID: PMC5355315 DOI: 10.18632/oncotarget.14440] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 12/16/2016] [Indexed: 12/21/2022] Open
Abstract
Exosomes have been implicated in the cell-cell transfer of oncogenic proteins and genetic material. We speculated this may be one mechanism by which an intrinsically platinum-resistant population of epithelial ovarian cancer (EOC) cells imparts its influence on surrounding tumor cells. To explore this possibility we utilized a platinum-sensitive cell line, A2780 and exosomes derived from its resistant subclones, and an unselected, platinum-resistant EOC line, OVCAR10. A2780 cells demonstrate a ~2-fold increase in viability upon treatment with carboplatin when pre-exposed to exosomes from platinum-resistant cells as compared to controls. This coincided with increased epithelial to mesenchymal transition (EMT). DNA sequencing of EOC cell lines revealed previously unreported somatic mutations in the Mothers Against Decapentaplegic Homolog 4 (SMAD4) within platinum-resistant cells. A2780 cells engineered to exogenously express these SMAD4 mutations demonstrate up-regulation of EMT markers following carboplatin treatment, are more resistant to carboplatin, and release exosomes which impart a ~1.7-fold increase in resistance in naive A2780 recipient cells as compared to controls. These studies provide the first evidence that acquired SMAD4 mutations enhance the chemo-resistance profile of EOC and present a novel mechanism in which exchange of tumor-derived exosomes perpetuates an EMT phenotype, leading to the development of subpopulations of platinum-refractory cells.
Collapse
Affiliation(s)
- Jennifer Crow
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Safinur Atay
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Samagya Banskota
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA.,Department of Biomedical Engineering, Duke University, Durham, North Carolina, NC, USA
| | - Brittany Artale
- Bioengineering Graduate Program, University of Kansas, Lawrence, KS, USA
| | - Sarah Schmitt
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA.,University of Kansas Cancer Center, Kansas City, KS, USA
| |
Collapse
|
296
|
Mishra DK, Srivastava P, Sharma A, Prasad R, Bhuyan SK, Malage R, Kumar P, Yadava PK. Translationally controlled tumor protein (TCTP) is required for TGF-β1 induced epithelial to mesenchymal transition and influences cytoskeletal reorganization. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1865:67-75. [PMID: 28958626 DOI: 10.1016/j.bbamcr.2017.09.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 09/20/2017] [Accepted: 09/22/2017] [Indexed: 12/27/2022]
Abstract
Epithelial-mesenchymal transition (EMT) is a programed course of developmental changes resulting in the acquisition of invasiveness and mobility in cells. In cancer, this course is used by epithelial cells to attain movability. Translationally controlled tumor protein (TCTP) has been extensively characterized following the observation on tumor reversion ensuing its depletion. However, the role of TCTP in cancer progression is still elusive. Here, we demonstrate for the first time that TCTP is a target of transforming growth factor-β1 (TGF-β1), a key regulator of EMT in A549 cells. We here present changes in expression patterns of intermediate filament markers (vimentin and cytokeratin 18a) of EMT following TCTP knockdown or over expression. The TCTP over-expression in cancer cells is associated with mesenchymal characters, while downregulation promotes the epithelial markers in the cells. Interaction of TCTP with β-catenin seems to stabilize β-catenin, preparative to its nuclear localization highlighting a role for β-catenin signaling in EMT. Moreover, the induction of urokinase plasminogen activator (uPA) following ectopic expression of TCTP leads to destabilization of ECM. The cells knocked down for TCTP show diminished invasiveness and migration under TGF-β1 treatment. The present results for the first time demonstrate that TGF-β1 dependent TCTP expression is required for EMT in cells.
Collapse
Affiliation(s)
- Deepak Kumar Mishra
- Applied Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Pratibha Srivastava
- Applied Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Amod Sharma
- Applied Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Ramraj Prasad
- Applied Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Soubhagya Kumar Bhuyan
- Applied Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Rahuldev Malage
- Applied Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Pramod Kumar
- Applied Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Pramod Kumar Yadava
- Applied Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
297
|
Limoge M, Safina A, Truskinovsky AM, Aljahdali I, Zonneville J, Gruevski A, Arteaga CL, Bakin AV. Tumor p38MAPK signaling enhances breast carcinoma vascularization and growth by promoting expression and deposition of pro-tumorigenic factors. Oncotarget 2017; 8:61969-61981. [PMID: 28977919 PMCID: PMC5617479 DOI: 10.18632/oncotarget.18755] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 05/19/2017] [Indexed: 01/10/2023] Open
Abstract
The breast carcinoma microenvironment strikingly influences cancer progression and response to therapy. Various cell types in the carcinoma microenvironment show significant activity of p38 mitogen-activated protein kinase (MAPK), although the role of p38MAPK in breast cancer progression is still poorly understood. The present study examined the contribution of tumor p38MAPK to breast carcinoma microenvironment and metastatic capacity. Inactivation of p38MAPK signaling in metastatic breast carcinoma cells was achieved by forced expression of the kinase-inactive mutant of p38/MAPK14 (a dominant-negative p38, dn-p38). Disruption of tumor p38MAPK signaling reduced growth and metastases of breast carcinoma xenografts. Importantly, dn-p38 markedly decreased tumor blood-vessel density and lumen sizes. Mechanistic studies revealed that p38 controls expression of pro-angiogenic extracellular factors such as matrix protein Fibronectin and cytokines VEGFA, IL8, and HBEGF. Tumor-associated fibroblasts enhanced tumor growth and vasculature as well as increased expression of the pro-angiogenic factors. These effects were blunted by dn-p38. Metadata analysis showed elevated expression of p38 target genes in breast cancers and this was an unfavorable marker of disease recurrence and poor-outcome. Thus, our study demonstrates that tumor p38MAPK signaling promotes breast carcinoma growth, invasive and metastatic capacities. Importantly, p38 enhances carcinoma vascularization by facilitating expression and deposition of pro-angiogenic factors. These results argue that p38MAPK is a valuable target for anticancer therapy affecting tumor vasculature. Anti-p38 drugs may provide new therapeutic strategies against breast cancer, including metastatic disease.
Collapse
Affiliation(s)
- Michelle Limoge
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Alfiya Safina
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | | | - Ieman Aljahdali
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Justin Zonneville
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Aleksandar Gruevski
- State University of New York at Buffalo, Department of Biological Sciences, Buffalo, New York, USA
| | - Carlos L. Arteaga
- Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Andrei V. Bakin
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, New York, USA
| |
Collapse
|
298
|
Dai X, Fang M, Li S, Yan Y, Zhong Y, Du B. miR-21 is involved in transforming growth factor β1-induced chemoresistance and invasion by targeting PTEN in breast cancer. Oncol Lett 2017; 14:6929-6936. [PMID: 29151919 DOI: 10.3892/ol.2017.7007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 07/07/2017] [Indexed: 12/12/2022] Open
Abstract
Transforming growth factor β1 (TGF-β1) has been associated with poor outcomes in patients with breast cancer. However, the functions and underlying molecular mechanisms of TGF-β1 in breast cancer remain unknown. Therefore, the present study aimed to identify the effects of components of the TGF-β/microRNA (miR-)21/phosphatase and tensin homolog (PTEN) signaling axis in breast cancer. TGF-β1 was identified to upregulate the expression of miR-21, and miR-21 was demonstrated to be significantly upregulated in breast cancer tissues compared with benign proliferative breast disease. In addition, the expression of miR-21 was significantly associated with increased TGF-β1 and clinical characteristics in patients, including tumor grade and lymph node metastasis (all P<0.05). Furthermore, in the breast cancer MCF-7 cell line, TGF-β1 was revealed to induce the expression of miR-21 in a dose- and time-dependent manner. The results of the present study additionally demonstrated that increased miR-21, in response to TGF-β1 signaling, was associated with tumor invasion and chemoresistance in vitro. In addition, suppression of PTEN was mediated by TGF-β1-induced expression of miR-21 in breast cancer cells and using a miR-21 inhibitor revitalized the expression of PTEN. The results of the present study explored the functions of TGF-β1-stimulated expression of miR-21 to suppress the PTEN axis, which promotes breast cancer progression and chemoresistance.
Collapse
Affiliation(s)
- Xiaomeng Dai
- Department of Pathology, Medical School of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Mao Fang
- Department of Pathology, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, Guangdong 510436, P.R. China
| | - Shuang Li
- Department of Pathology, Medical School of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Yongrong Yan
- Department of Pathology, Medical School of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Ying Zhong
- Department of Pathology, Medical School of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Bin Du
- Department of Pathology, Medical School of Jinan University, Guangzhou, Guangdong 510632, P.R. China.,Division of Clinic Pathology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| |
Collapse
|
299
|
Hong D, Messier TL, Tye CE, Dobson JR, Fritz AJ, Sikora KR, Browne G, Stein JL, Lian JB, Stein GS. Runx1 stabilizes the mammary epithelial cell phenotype and prevents epithelial to mesenchymal transition. Oncotarget 2017; 8:17610-17627. [PMID: 28407681 PMCID: PMC5392273 DOI: 10.18632/oncotarget.15381] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 01/16/2017] [Indexed: 11/28/2022] Open
Abstract
Runx1 is a well characterized transcription factor essential for hematopoietic differentiation and Runx1 mutations are the cause of leukemias. Runx1 is highly expressed in normal epithelium of most glands and recently has been associated with solid tumors. Notably, the function of Runx1 in the mammary gland and how it is involved in initiation and progression of breast cancer is still unclear. Here we demonstrate the consequences of Runx1 loss in normal mammary epithelial and breast cancer cells. We first observed that Runx1 is decreased in tumorigenic and metastatic breast cancer cells. We also observed loss of Runx1 expression upon induction of epithelial-mesenchymal transition (EMT) in MCF10A (normal-like) cells. Furthermore depletion of Runx1 in MCF10A cells resulted in striking changes in cell shape, leading to mesenchymal cell morphology. The epithelial phenotype could be restored in breast cancer cells by re-expressing Runx1. Analyses of breast tumors and patient data revealed that low Runx1 expression is associated with poor prognosis and decreased survival. We addressed mechanisms for the function of Runx1 in maintaining the epithelial phenotype and find Runx1 directly regulates E-cadherin; and serves as a downstream transcription factor mediating TGFβ signaling. We also observed through global gene expression profiling of growth factor depleted cells that induction of EMT and loss of Runx1 is associated with activation of TGFβ and WNT pathways. Thus these findings have identified a novel function for Runx1 in sustaining normal epithelial morphology and preventing EMT and suggest Runx1 levels could be a prognostic indicator of tumor progression.
Collapse
Affiliation(s)
- Deli Hong
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont College of Medicine, Burlington, VT, USA.,Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Terri L Messier
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont College of Medicine, Burlington, VT, USA
| | - Coralee E Tye
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont College of Medicine, Burlington, VT, USA
| | - Jason R Dobson
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Andrew J Fritz
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont College of Medicine, Burlington, VT, USA
| | - Kenneth R Sikora
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont College of Medicine, Burlington, VT, USA
| | - Gillian Browne
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont College of Medicine, Burlington, VT, USA
| | - Janet L Stein
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont College of Medicine, Burlington, VT, USA
| | - Jane B Lian
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont College of Medicine, Burlington, VT, USA
| | - Gary S Stein
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont College of Medicine, Burlington, VT, USA
| |
Collapse
|
300
|
Li W, Zhang X, Lu X, You L, Song Y, Luo Z, Zhang J, Nie J, Zheng W, Xu D, Wang Y, Dong Y, Yu S, Hong J, Shi J, Hao H, Luo F, Hua L, Wang P, Qian X, Yuan F, Wei L, Cui M, Zhang T, Liao Q, Dai M, Liu Z, Chen G, Meckel K, Adhikari S, Jia G, Bissonnette MB, Zhang X, Zhao Y, Zhang W, He C, Liu J. 5-Hydroxymethylcytosine signatures in circulating cell-free DNA as diagnostic biomarkers for human cancers. Cell Res 2017; 27:1243-1257. [PMID: 28925386 PMCID: PMC5630683 DOI: 10.1038/cr.2017.121] [Citation(s) in RCA: 238] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 08/06/2017] [Accepted: 08/09/2017] [Indexed: 12/16/2022] Open
Abstract
DNA modifications such as 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC) are epigenetic marks known to affect global gene expression in mammals. Given their prevalence in the human genome, close correlation with gene expression and high chemical stability, these DNA epigenetic marks could serve as ideal biomarkers for cancer diagnosis. Taking advantage of a highly sensitive and selective chemical labeling technology, we report here the genome-wide profiling of 5hmC in circulating cell-free DNA (cfDNA) and in genomic DNA (gDNA) of paired tumor and adjacent tissues collected from a cohort of 260 patients recently diagnosed with colorectal, gastric, pancreatic, liver or thyroid cancer and normal tissues from 90 healthy individuals. 5hmC was mainly distributed in transcriptionally active regions coincident with open chromatin and permissive histone modifications. Robust cancer-associated 5hmC signatures were identified in cfDNA that were characteristic for specific cancer types. 5hmC-based biomarkers of circulating cfDNA were highly predictive of colorectal and gastric cancers and were superior to conventional biomarkers and comparable to 5hmC biomarkers from tissue biopsies. Thus, this new strategy could lead to the development of effective, minimally invasive methods for diagnosis and prognosis of cancer from the analyses of blood samples.
Collapse
Affiliation(s)
- Wenshuai Li
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xu Zhang
- Section of Hematology/Oncology, Department of Medicine, University of Illinois, Chicago, IL 60612, USA
| | - Xingyu Lu
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637, USA.,Shanghai Epican Genetech, Co. Ltd., Zhangjiang Hi-Tech Park, Shanghai 201203, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yanqun Song
- Shanghai Epican Genetech, Co. Ltd., Zhangjiang Hi-Tech Park, Shanghai 201203, China
| | - Zhongguang Luo
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jun Zhang
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ji Nie
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Wanwei Zheng
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Diannan Xu
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yaping Wang
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yuanqiang Dong
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Shulin Yu
- Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai 200032, China
| | - Jun Hong
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jianping Shi
- Department of Digestive Diseases, Pudong Hospital, Fudan University, Shanghai 201399, China
| | - Hankun Hao
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Fen Luo
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Luchun Hua
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Peng Wang
- Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai 200032, China
| | - Xiaoping Qian
- Department of Oncology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing 210008, China
| | - Fang Yuan
- Beijing National Laboratory for Molecular Sciences, College of Chemistry, Peking University, Beijing 100871, China.,Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing 100871, China
| | - Lianhuan Wei
- Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing 100871, China
| | - Ming Cui
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Quan Liao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Menghua Dai
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Ziwen Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Ge Chen
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Katherine Meckel
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Sarbani Adhikari
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Guifang Jia
- Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing 100871, China.,Department of Chemical Biology, Structure and Function Biomolecules Center, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Marc B Bissonnette
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Xinxiang Zhang
- Beijing National Laboratory for Molecular Sciences, College of Chemistry, Peking University, Beijing 100871, China.,Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing 100871, China
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Wei Zhang
- Department of Preventive Medicine and The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Chuan He
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637, USA.,Beijing National Laboratory for Molecular Sciences, College of Chemistry, Peking University, Beijing 100871, China
| | - Jie Liu
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China.,Department of Immunology, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, China
| |
Collapse
|