251
|
Liu S, de Boeck M, van Dam H, ten Dijke P. Regulation of the TGF-β pathway by deubiquitinases in cancer. Int J Biochem Cell Biol 2016; 76:135-45. [DOI: 10.1016/j.biocel.2016.05.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 05/02/2016] [Accepted: 05/03/2016] [Indexed: 11/26/2022]
|
252
|
Moustakas A, Heldin CH. Mechanisms of TGFβ-Induced Epithelial-Mesenchymal Transition. J Clin Med 2016; 5:jcm5070063. [PMID: 27367735 PMCID: PMC4961994 DOI: 10.3390/jcm5070063] [Citation(s) in RCA: 200] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 06/22/2016] [Accepted: 06/22/2016] [Indexed: 02/07/2023] Open
Abstract
Transitory phenotypic changes such as the epithelial–mesenchymal transition (EMT) help embryonic cells to generate migratory descendants that populate new sites and establish the distinct tissues in the developing embryo. The mesenchymal descendants of diverse epithelia also participate in the wound healing response of adult tissues, and facilitate the progression of cancer. EMT can be induced by several extracellular cues in the microenvironment of a given epithelial tissue. One such cue, transforming growth factor β (TGFβ), prominently induces EMT via a group of specific transcription factors. The potency of TGFβ is partly based on its ability to perform two parallel molecular functions, i.e. to induce the expression of growth factors, cytokines and chemokines, which sequentially and in a complementary manner help to establish and maintain the EMT, and to mediate signaling crosstalk with other developmental signaling pathways, thus promoting changes in cell differentiation. The molecules that are activated by TGFβ signaling or act as cooperating partners of this pathway are impossible to exhaust within a single coherent and contemporary report. Here, we present selected examples to illustrate the key principles of the circuits that control EMT under the influence of TGFβ.
Collapse
Affiliation(s)
- Aristidis Moustakas
- Ludwig Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, SE 751 24 Uppsala, Sweden.
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Box 582, SE 751 23 Uppsala, Sweden.
| | - Carl-Henrik Heldin
- Ludwig Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, SE 751 24 Uppsala, Sweden.
| |
Collapse
|
253
|
Hautala LC, Koistinen R, Koistinen H. Repressed PKCδ activation in glycodelin-expressing cells mediates resistance to phorbol ester and TGFβ. Cell Signal 2016; 28:1463-9. [PMID: 27373413 DOI: 10.1016/j.cellsig.2016.06.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 06/17/2016] [Accepted: 06/27/2016] [Indexed: 11/30/2022]
Abstract
Glycodelin is a glycoprotein mainly expressed in well-differentiated epithelial cells in reproductive tissues. In normal secretory endometrium, the expression of glycodelin is abundant and regulated by progesterone. In hormone-related cancers glycodelin expression is associated with well-differentiated tumors. We have previously found that glycodelin drives epithelial differentiation of HEC-1B endometrial adenocarcinoma cells, resulting in reduced tumor growth in a preclinical mouse model. Here we show that glycodelin-transfected HEC-1B cells have repressed protein kinase C delta (PKCδ) activation, likely due to downregulation of PDK1, and are resistant to phenotypic change and enhanced migration induced by phorbol 12-myristate 13-acetate (PMA). In control cells, which do not express glycodelin, the effects of PMA were abolished by using PKCδ and PDK1 inhibitors, and knockdown of PKCδ, MEK1 and 2, or ERK1 and 2 by siRNAs. Similarly, transforming growth factor β (TGFβ)-induced phenotypic change was only seen in control cells, not in glycodelin-producing cells, and it was mediated by PKCδ. Taken together, these results strongly suggest that PKCδ, via MAPK pathway, is involved in the glycodelin-driven cell differentiation rendering the cells resistant to stimulation by PMA and TGFβ.
Collapse
Affiliation(s)
- Laura C Hautala
- Department of Clinical Chemistry, Medicum, University of Helsinki and Helsinki University Central Hospital, Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Riitta Koistinen
- Department of Clinical Chemistry, Medicum, University of Helsinki and Helsinki University Central Hospital, Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Hannu Koistinen
- Department of Clinical Chemistry, Medicum, University of Helsinki and Helsinki University Central Hospital, Haartmaninkatu 8, 00290 Helsinki, Finland.
| |
Collapse
|
254
|
Mebarki S, Désert R, Sulpice L, Sicard M, Desille M, Canal F, Schneider HDP, Bergeat D, Turlin B, Bellaud P, Lavergne E, Guével RL, Corlu A, Perret C, Coulouarn C, Clément B, Musso O. De novo HAPLN1 expression hallmarks Wnt-induced stem cell and fibrogenic networks leading to aggressive human hepatocellular carcinomas. Oncotarget 2016; 7:39026-39043. [PMID: 27191501 PMCID: PMC5129911 DOI: 10.18632/oncotarget.9346] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 04/22/2016] [Indexed: 12/27/2022] Open
Abstract
About 20% hepatocellular carcinomas (HCCs) display wild-type β-catenin, enhanced Wnt signaling, hepatocyte dedifferentiation and bad outcome, suggesting a specific impact of Wnt signals on HCC stem/progenitor cells. To study Wnt-specific molecular pathways, cell fates and clinical outcome, we fine-tuned Wnt/β-catenin signaling in liver progenitor cells, using the prototypical Wnt ligand Wnt3a. Cell biology assays and transcriptomic profiling were performed in HepaRG hepatic progenitors exposed to Wnt3a after β-catenin knockdown or Wnt inhibition with FZD8_CRD. Gene expression network, molecular pathology and survival analyses were performed on HCCs and matching non-tumor livers from 70 patients by real-time PCR and tissue micro-array-based immunohistochemistry. Wnt3a reprogrammed liver progenitors to replicating fibrogenic myofibroblast-like cells displaying stem and invasive features. Invasion was inhibited by 30 nM FZD7 and FZD8 CRDs. Translation of these data to human HCCs revealed two tight gene networks associating cell surface Wnt signaling, stem/progenitor markers and mesenchymal commitment. Both networks were linked by Hyaluronan And Proteoglycan Link Protein 1 (HAPLN1), that appeared de novo in aggressive HCCs expressing cytoplasmic β-catenin and stem cell markers. HAPLN1 was independently associated with bad overall and disease-free outcome. In vitro, HAPLN1 was expressed de novo in EPCAM¯/NCAM+ mesoderm-committed progenitors, upon spontaneous epithelial-mesenchymal transition and de-differentiation of hepatocyte-like cells to liver progenitors. In these cells, HAPLN1 knockdown downregulated key markers of mesenchymal cells, such as Snail, LGR5, collagen IV and α-SMA. In conclusion, HAPLN1 reflects a signaling network leading to stemness, mesenchymal commitment and HCC progression.
Collapse
Affiliation(s)
- Sihem Mebarki
- Inserm, UMR991, Liver Metabolisms and Cancer, Rennes, France
- Université de Rennes 1, Rennes, France
| | - Romain Désert
- Inserm, UMR991, Liver Metabolisms and Cancer, Rennes, France
- Université de Rennes 1, Rennes, France
| | - Laurent Sulpice
- Inserm, UMR991, Liver Metabolisms and Cancer, Rennes, France
- Université de Rennes 1, Rennes, France
- CHU de Rennes, Department of Gastrointestinal and Hepatobiliary Surgery, Rennes, France
| | - Marie Sicard
- Inserm, UMR991, Liver Metabolisms and Cancer, Rennes, France
- Université de Rennes 1, Rennes, France
| | - Mireille Desille
- Inserm, UMR991, Liver Metabolisms and Cancer, Rennes, France
- Université de Rennes 1, Rennes, France
- CHU de Rennes, Centre de Ressources Biologiques Santé, Rennes, France
| | - Frédéric Canal
- Inserm, Institut Cochin, Paris, France
- Cnrs, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | | | - Damien Bergeat
- Inserm, UMR991, Liver Metabolisms and Cancer, Rennes, France
- Université de Rennes 1, Rennes, France
- CHU de Rennes, Department of Gastrointestinal and Hepatobiliary Surgery, Rennes, France
| | - Bruno Turlin
- Inserm, UMR991, Liver Metabolisms and Cancer, Rennes, France
- Université de Rennes 1, Rennes, France
- CHU de Rennes, Centre de Ressources Biologiques Santé, Rennes, France
| | - Pascale Bellaud
- Université de Rennes 1, UMS 18 Biosit, Biogenouest, Rennes, France
| | - Elise Lavergne
- Inserm, UMR991, Liver Metabolisms and Cancer, Rennes, France
- Université de Rennes 1, Rennes, France
| | - Rémy Le Guével
- Université de Rennes 1, UMS 18 Biosit, Biogenouest, ImPACcellCore Facility, Rennes, France
| | - Anne Corlu
- Inserm, UMR991, Liver Metabolisms and Cancer, Rennes, France
- Université de Rennes 1, Rennes, France
- Université de Rennes 1, UMS 18 Biosit, Biogenouest, ImPACcellCore Facility, Rennes, France
| | - Christine Perret
- Inserm, Institut Cochin, Paris, France
- Cnrs, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Cédric Coulouarn
- Inserm, UMR991, Liver Metabolisms and Cancer, Rennes, France
- Université de Rennes 1, Rennes, France
| | - Bruno Clément
- Inserm, UMR991, Liver Metabolisms and Cancer, Rennes, France
- Université de Rennes 1, Rennes, France
| | - Orlando Musso
- Inserm, UMR991, Liver Metabolisms and Cancer, Rennes, France
- Université de Rennes 1, Rennes, France
| |
Collapse
|
255
|
Sheller S, Papaconstantinou J, Urrabaz-Garza R, Richardson L, Saade G, Salomon C, Menon R. Amnion-Epithelial-Cell-Derived Exosomes Demonstrate Physiologic State of Cell under Oxidative Stress. PLoS One 2016; 11:e0157614. [PMID: 27333275 PMCID: PMC4917104 DOI: 10.1371/journal.pone.0157614] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 06/01/2016] [Indexed: 02/07/2023] Open
Abstract
At term, the signals of fetal maturity and feto-placental tissue aging prompt uterine readiness for delivery by transitioning quiescent myometrium to an active stage. It is still unclear how the signals reach the distant myometrium. Exosomes are a specific type of extracellular vesicle (EVs) that transport molecular signals between cells, and are released from a wide range of cells, including the maternal and fetal cells. In this study, we hypothesize that i) exosomes act as carriers of signals in utero-placental compartments and ii) exosomes reflect the physiologic status of the origin cells. The primary aims of this study were to determine exosomal contents in exosomes derived from primary amnion epithelial cells (AEC). We also determined the effect of oxidative stress on AEC derived exosomal cargo contents. AEC were isolated from amniotic membrane obtained from normal, term, not in labor placentae at delivery, and culture under standard conditions. Oxidative stress was induced using cigarette smoke extract for 48 hours. AEC-conditioned media were collected and exosomes isolated by differential centrifugations. Both growth conditions (normal and oxidative stress induced) produced cup shaped exosomes of around 50 nm, expressed exosomes enriched markers, such as CD9, CD63, CD81 and HSC70, embryonic stem cell marker Nanog, and contained similar amounts of cell free AEC DNA. Using confocal microscopy, the colocalization of histone (H) 3, heat shock protein (HSP) 70 and activated form of pro-senescence and term parturition associated marker p38 mitogen activated protein kinase (MAPK) (P-p38 MAPK) co-localized with exosome enrich marker CD9. HSP70 and P-p38 MAPK were significantly higher in exosomes from AEC grown under oxidative stress conditions than standard conditions (p<0.05). Finally, mass spectrometry and bioinformatics analysis identified 221 different proteins involved in immunomodulatory response and cell-to-cell communication. This study determined AEC exosome characteristics and their cargo reflected the physiologic status of the cell of origin and suggests that AEC-derived exosomal p38 MAPK plays a major role in determining the fate of pregnancy. Understanding the propagation of fetal signals and their mechanisms in normal term pregnancies can provide insights into pathologic activation of such signals associated with spontaneous preterm parturitions.
Collapse
Affiliation(s)
- Samantha Sheller
- Division of Maternal-Fetal Medicine & Perinatal Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - John Papaconstantinou
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Rheanna Urrabaz-Garza
- Division of Maternal-Fetal Medicine & Perinatal Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Lauren Richardson
- Division of Maternal-Fetal Medicine & Perinatal Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - George Saade
- Division of Maternal-Fetal Medicine & Perinatal Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Carlos Salomon
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, UQ Centre for Clinical Research, Faculty of Health Sciences, University of Queensland, Herston, Queensland, Australia
| | - Ramkumar Menon
- Division of Maternal-Fetal Medicine & Perinatal Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| |
Collapse
|
256
|
Potential role of targeted therapies in the treatment of triple-negative breast cancer. Anticancer Drugs 2016; 27:147-55. [PMID: 26682525 DOI: 10.1097/cad.0000000000000328] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Breast cancer is the most common cancer type that affects women and is the major cause of morbidity and mortality. Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype and accounts for 10-20% of all breast cancer cases. TNBC is commonly characterized by the absence of estrogen, progesterone, and the Her2/neu receptor and is usually diagnosed by immunohistochemistry. Mutations in the BRCA1 gene, as well as overexpression of oncogenic kinases, such as human epidermal growth factor receptor 2, vascular endothelial growth factor-A, insulin-like growth factor-1 (IGF-1)/IGF-1 receptor, and transforming growth factor-β1, have been found to be correlated with a higher risk of metastasis and poor overall survival in TNBC patients. The current review briefly discusses the various treatment options including chemotherapeutics and targeted therapies that are available currently for the therapy of TNBC patients and highlights their comparative benefits and disadvantages for clinical application.
Collapse
|
257
|
Tan J, Wang Y, Zhang N, Zhu X. Induction of epithelial to mesenchymal transition (EMT) and inhibition on adipogenesis: Two different sides of the same coin? Feasible roles and mechanisms of transforming growth factor β1 (TGF-β1) in age-related thymic involution. Cell Biol Int 2016; 40:842-6. [PMID: 27189906 DOI: 10.1002/cbin.10625] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 05/08/2016] [Accepted: 05/14/2016] [Indexed: 12/16/2022]
Abstract
Age-related thymic involution is characterized by a loss of thymic epithelial cells (TECs) and a concomitant increase in adipocytes, but the mechanisms involved in thymic adipogenesis are still not clear. Transforming growth factor β1 (TGF-β1) is a pleiotropic cytokine that has been reported to be up-regulated with age in thymic stromal cells in both human and mouse. However, the exact role of TGF-β1 in age-related thymic involution remains to be further elucidated. On the basis of previous findings, we propose a novel hypothesis that TGF-β1 functions a dual role in age-related thymic involution. On one hand, up-regulation of TGF-β1 promotes epithelial to mesenchymal transition (EMT) process in TECs via activating forkhead box protein C2 (FoxC2). On the other hand, TGF-β1 inhibits the transdifferentiation of EMT-derived mesenchymal cells to adipocytes in the thymus. If confirmed, our hypothesis will not only provide further evidence supporting that the transdifferentiation of TECs into pre-adipocytes represents a source of thymic adiposity during age-related thymic involution, but also uncover a unique role of TGF-β1 in the transdifferentiation of TECs into pre-adipocytes. Collectively, the inhibition of TGF-β1 may serve as a strategy to hinder age-related thymic involution or even to restore thymic function in the elderly.
Collapse
Affiliation(s)
- Jianxin Tan
- Research Center, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Yajun Wang
- Research Center, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Nannan Zhang
- Research Center, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China.,Department of Nerve Function, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Xike Zhu
- Research Center, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| |
Collapse
|
258
|
MicroRNA in pancreatic cancer. J Hum Genet 2016; 62:33-40. [DOI: 10.1038/jhg.2016.59] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 04/26/2016] [Accepted: 04/27/2016] [Indexed: 02/07/2023]
|
259
|
Prithviraj P, Anaka M, McKeown SJ, Permezel M, Walkiewicz M, Cebon J, Behren A, Jayachandran A. Pregnancy associated plasma protein-A links pregnancy and melanoma progression by promoting cellular migration and invasion. Oncotarget 2016; 6:15953-65. [PMID: 25940796 PMCID: PMC4599249 DOI: 10.18632/oncotarget.3643] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 03/23/2015] [Indexed: 11/25/2022] Open
Abstract
Melanoma is the most common cancer diagnosed in pregnant women and an aggressive course with poorer outcomes is commonly described during pregnancy or shortly after childbirth. The underlying mechanisms for this are not understood. Here, we report that melanoma migration, invasiveness and progression are promoted by pregnancy-associated plasma protein-A (PAPPA), a pregnancy-associated metalloproteinase produced by the placenta that increases the bioavailability of IGF1 by cleaving it from a circulating complex formed with IGFBP4. We show that PAPPA is widely expressed by metastatic melanoma tumors and is elevated in melanoma cells exhibiting mesenchymal, invasive and label-retaining phenotypes. Notably, inhibition of PAPPA significantly reduced invasion and migration of melanoma cells in vitro and in vivo within the embryonic chicken neural tube. PAPPA-enriched pregnancy serum treatment enhanced melanoma motility in vitro. Furthermore, we report that IGF1 can induce the phenotypic and functional effects of epithelial-to-mesenchymal transition (EMT) in melanoma cells. In this study, we establish a clear relationship between a pregnancy-associated protein PAPPA, melanoma and functional effects mediated through IGF1 that provides a plausible mechanism for accelerated melanoma progression during pregnancy. This opens the possibility of targeting the PAPPA/IGF1 axis therapeutically.
Collapse
Affiliation(s)
- Prashanth Prithviraj
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Cancer Immunobiology Laboratory, Heidelberg, VIC, Australia.,Olivia Newton-John Cancer Research Institute, Olivia Newton-John Cancer and Wellness Centre, Heidelberg, VIC, Australia.,Department of Medicine, University of Melbourne, VIC, Australia
| | - Matthew Anaka
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Cancer Immunobiology Laboratory, Heidelberg, VIC, Australia
| | - Sonja J McKeown
- Department of Anatomy and Neuroscience, University of Melbourne, VIC, Australia
| | | | - Marzena Walkiewicz
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Cancer Immunobiology Laboratory, Heidelberg, VIC, Australia.,Olivia Newton-John Cancer Research Institute, Olivia Newton-John Cancer and Wellness Centre, Heidelberg, VIC, Australia
| | - Jonathan Cebon
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Cancer Immunobiology Laboratory, Heidelberg, VIC, Australia.,Olivia Newton-John Cancer Research Institute, Olivia Newton-John Cancer and Wellness Centre, Heidelberg, VIC, Australia.,Department of Medicine, University of Melbourne, VIC, Australia.,School of Cancer Medicine, La Trobe University, VIC, Australia
| | - Andreas Behren
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Cancer Immunobiology Laboratory, Heidelberg, VIC, Australia.,Olivia Newton-John Cancer Research Institute, Olivia Newton-John Cancer and Wellness Centre, Heidelberg, VIC, Australia.,Department of Medicine, University of Melbourne, VIC, Australia.,School of Cancer Medicine, La Trobe University, VIC, Australia
| | - Aparna Jayachandran
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Cancer Immunobiology Laboratory, Heidelberg, VIC, Australia.,Olivia Newton-John Cancer Research Institute, Olivia Newton-John Cancer and Wellness Centre, Heidelberg, VIC, Australia.,Department of Medicine, University of Melbourne, VIC, Australia.,School of Cancer Medicine, La Trobe University, VIC, Australia
| |
Collapse
|
260
|
Man XY, Chen XB, Li W, Landeck L, Dou TT, Chen JQ, Zhou J, Cai SQ, Zheng M. Analysis of epithelial-mesenchymal transition markers in psoriatic epidermal keratinocytes. Open Biol 2016; 5:rsob.150032. [PMID: 26269426 PMCID: PMC4554915 DOI: 10.1098/rsob.150032] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Psoriasis is similar to endpoints of epithelial–mesenchymal transition (EMT), a process of epithelial cells transformed into fibroblast-like cells. The molecular epithelial and mesenchymal markers were analysed in psoriatic keratinocytes. No obvious alteration of epithelial markers E-cadherin (E-cad), keratin 10 (K10), K14 and K16 was detected in psoriatic keratinocytes. However, significantly increased expression of Vim, FN, plasminogen activator inhibitor 1 (PAI-1) and Slug was seen. IL-17A and IL-13 at 50 ng ml−1 strongly decreased expression of K10, Vim and FN. TGF-β1 at 50 ng ml−1 promoted the production of N-cad, Vim, FN and PAI-1. Slug was decreased by dexamethasone (Dex), but E-cad was upregulated by Dex. Silencing of ERK partially increased E-cad and K16, but remarkably inhibited K14, FN, Vim, β-catenin, Slug and α5 integrin. Moreover, inhibition of Rho and GSK3 by their inhibitors Y27632 and SB216763, respectively, strongly raised E-cad, β-catenin and Slug. Dex decreased Y27632-mediated increase of β-catenin. Dex at 2.0 µM inhibited SB216763-regulated E-cad, β-catenin and slug. In conclusion, EMT in psoriatic keratinocytes may be defined as an intermediate phenotype of type 2 EMT. ERK, Rho and GSK3 play active roles in the process of EMT in psoriatic keratinocytes.
Collapse
Affiliation(s)
- Xiao-Yong Man
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, People's Republic of China
| | - Xi-Bei Chen
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, People's Republic of China
| | - Wei Li
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, People's Republic of China
| | - Lilla Landeck
- Department of Dermatology, Ernst von Bergmann General Hospital, Teaching Hospital of Charité-University, Potsdam, Germany
| | - Ting-Ting Dou
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, People's Republic of China
| | - Jia-Qi Chen
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, People's Republic of China
| | - Jiong Zhou
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, People's Republic of China
| | - Sui-Qing Cai
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, People's Republic of China
| | - Min Zheng
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, People's Republic of China
| |
Collapse
|
261
|
Boehme SA, Franz-Bacon K, DiTirro DN, Ly TW, Bacon KB. MAP3K19 Is a Novel Regulator of TGF-β Signaling That Impacts Bleomycin-Induced Lung Injury and Pulmonary Fibrosis. PLoS One 2016; 11:e0154874. [PMID: 27144281 PMCID: PMC4856290 DOI: 10.1371/journal.pone.0154874] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 04/20/2016] [Indexed: 12/15/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive, debilitating disease for which two medications, pirfenidone and nintedanib, have only recently been approved for treatment. The cytokine TGF-β has been shown to be a central mediator in the disease process. We investigated the role of a novel kinase, MAP3K19, upregulated in IPF tissue, in TGF-β-induced signal transduction and in bleomycin-induced pulmonary fibrosis. MAP3K19 has a very limited tissue expression, restricted primarily to the lungs and trachea. In pulmonary tissue, expression was predominantly localized to alveolar and interstitial macrophages, bronchial epithelial cells and type II pneumocytes of the epithelium. MAP3K19 was also found to be overexpressed in bronchoalveolar lavage macrophages from IPF patients compared to normal patients. Treatment of A549 or THP-1 cells with either MAP3K19 siRNA or a highly potent and specific inhibitor reduced phospho-Smad2 & 3 nuclear translocation following TGF-β stimulation. TGF-β-induced gene transcription was also strongly inhibited by both the MAP3K19 inhibitor and nintedanib, whereas pirfenidone had a much less pronounced effect. In combination, the MAP3K19 inhibitor appeared to act synergistically with either pirfenidone or nintedanib, at the level of target gene transcription or protein production. Finally, in an animal model of IPF, inhibition of MAP3K19 strongly attenuated bleomycin-induced pulmonary fibrosis when administered either prophylactically ortherapeutically. In summary, these results strongly suggest that inhibition of MAP3K19 may have a beneficial therapeutic effect in the treatment of IPF and represents a novel strategy to target this disease.
Collapse
Affiliation(s)
- Stefen A. Boehme
- AxikinPharmaceuticals, Inc., San Diego, California, United States of America
| | - Karin Franz-Bacon
- DNA Consulting, Inc., San Diego, California, United States of America
| | - Danielle N. DiTirro
- AxikinPharmaceuticals, Inc., San Diego, California, United States of America
| | - Tai Wei Ly
- AxikinPharmaceuticals, Inc., San Diego, California, United States of America
| | - Kevin B. Bacon
- AxikinPharmaceuticals, Inc., San Diego, California, United States of America
| |
Collapse
|
262
|
Lua I, Li Y, Pappoe LS, Asahina K. Myofibroblastic Conversion and Regeneration of Mesothelial Cells in Peritoneal and Liver Fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 185:3258-73. [PMID: 26598235 DOI: 10.1016/j.ajpath.2015.08.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 07/05/2015] [Accepted: 08/04/2015] [Indexed: 01/28/2023]
Abstract
Mesothelial cells (MCs) form a single epithelial layer and line the surface of body cavities and internal organs. Patients who undergo peritoneal dialysis often develop peritoneal fibrosis that is characterized by the accumulation of myofibroblasts in connective tissue. Although MCs are believed to be the source of myofibroblasts, their contribution has remained obscure. We determined the contribution of peritoneal MCs to myofibroblasts in chlorhexidine gluconate (CG)-induced fibrosis compared with that of phenotypic changes of liver MCs. CG injections resulted in disappearance of MCs from the body wall and the accumulation of myofibroblasts in the connective tissue. Conditional linage tracing with Wilms tumor 1 (Wt1)-CreERT2 and Rosa26 reporter mice found that 17% of myofibroblasts were derived from MCs in peritoneal fibrosis. Conditional deletion of transforming growth factor-β type II receptor in Wt1(+) MCs substantially reduced peritoneal fibrosis. The CG treatment also induced myofibroblastic conversion of MCs in the liver. Lineage tracing with Mesp1-Cre mice revealed that Mesp1(+) mesoderm gave rise to liver MCs but not peritoneal MCs. During recovery from peritoneal fibrosis, peritoneal MCs, but not liver MCs, contribute to the regeneration of the peritoneal mesothelium, indicating an inherent difference between parietal and visceral MCs. In conclusion, MCs partially contribute to myofibroblasts in peritoneal and liver fibrosis, and protection of the MC layer leads to reduced development of fibrous tissue.
Collapse
Affiliation(s)
- Ingrid Lua
- Department of Pathology, Southern California Research Center for Alcoholic Liver and Pancreatic Diseases (ALPD) and Cirrhosis, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Yuchang Li
- Department of Pathology, Southern California Research Center for Alcoholic Liver and Pancreatic Diseases (ALPD) and Cirrhosis, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Lamioko S Pappoe
- Division of Nephrology, Los Angeles County+University of Southern California Medical Center, Los Angeles, California
| | - Kinji Asahina
- Department of Pathology, Southern California Research Center for Alcoholic Liver and Pancreatic Diseases (ALPD) and Cirrhosis, Keck School of Medicine, University of Southern California, Los Angeles, California.
| |
Collapse
|
263
|
Choudhary P, Gutteridge A, Impey E, Storer RI, Owen RM, Whiting PJ, Bictash M, Benn CL. Targeting the cAMP and Transforming Growth Factor-β Pathway Increases Proliferation to Promote Re-Epithelialization of Human Stem Cell-Derived Retinal Pigment Epithelium. Stem Cells Transl Med 2016; 5:925-37. [PMID: 27112176 DOI: 10.5966/sctm.2015-0247] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 02/01/2016] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED Retinal pigment epithelium (RPE) cell integrity is critical to the maintenance of retinal function. Many retinopathies such as age-related macular degeneration (AMD) are caused by the degeneration or malfunction of the RPE cell layer. Replacement of diseased RPE with healthy, stem cell-derived RPE is a potential therapeutic strategy for treating AMD. Human embryonic stem cells (hESCs) differentiated into RPE progeny have the potential to provide an unlimited supply of cells for transplantation, but challenges around scalability and efficiency of the differentiation process still remain. Using hESC-derived RPE as a cellular model, we sought to understand mechanisms that could be modulated to increase RPE yield after differentiation. We show that RPE epithelialization is a density-dependent process, and cells seeded at low density fail to epithelialize. We demonstrate that activation of the cAMP pathway increases proliferation of dissociated RPE in culture, in part through inhibition of transforming growth factor-β (TGF-β) signaling. This results in enhanced uptake of epithelial identity, even in cultures seeded at low density. In line with these findings, targeted manipulation of the TGF-β pathway with small molecules produces an increase in efficiency of RPE re-epithelialization. Taken together, these data highlight mechanisms that promote epithelial fate acquisition in stem cell-derived RPE. Modulation of these pathways has the potential to favorably impact scalability and clinical translation of hESC-derived RPE as a cell therapy. SIGNIFICANCE Stem cell-derived retinal pigment epithelium (RPE) is currently being evaluated as a cell-replacement therapy for macular degeneration. This work shows that the process of generating RPE in vitro is regulated by the cAMP and transforming growth factor-β signaling pathway. Modulation of these pathways by small molecules, as identified by phenotypic screening, leads to an increased efficiency of generating RPE cells with a higher yield. This can have a potential impact on manufacturing transplantation-ready cells at large scale and is advantageous for clinical studies using this approach in the future.
Collapse
Affiliation(s)
- Parul Choudhary
- Pfizer Neuroscience and Pain Research Unit, Pfizer Ltd., Great Abington, Cambridge, United Kingdom
| | - Alex Gutteridge
- Pfizer Neuroscience and Pain Research Unit, Pfizer Ltd., Great Abington, Cambridge, United Kingdom
| | - Emma Impey
- Pfizer Neuroscience and Pain Research Unit, Pfizer Ltd., Great Abington, Cambridge, United Kingdom
| | - R Ian Storer
- Pfizer Worldwide Medicinal Chemistry, Pfizer Ltd., Great Abington, Cambridge, United Kingdom
| | - Robert M Owen
- Pfizer Worldwide Medicinal Chemistry, Pfizer Ltd., Great Abington, Cambridge, United Kingdom
| | - Paul J Whiting
- Pfizer Neuroscience and Pain Research Unit, Pfizer Ltd., Great Abington, Cambridge, United Kingdom
| | - Magda Bictash
- Pfizer Neuroscience and Pain Research Unit, Pfizer Ltd., Great Abington, Cambridge, United Kingdom
| | - Caroline L Benn
- Pfizer Neuroscience and Pain Research Unit, Pfizer Ltd., Great Abington, Cambridge, United Kingdom
| |
Collapse
|
264
|
Hu ZY, Liu YP, Xie LY, Wang XY, Yang F, Chen SY, Li ZG. AKAP-9 promotes colorectal cancer development by regulating Cdc42 interacting protein 4. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1172-81. [PMID: 27039663 DOI: 10.1016/j.bbadis.2016.03.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 02/29/2016] [Accepted: 03/25/2016] [Indexed: 02/07/2023]
Abstract
Our previous studies have shown that PRKA kinase anchor protein 9 (AKAP-9) is involved in colorectal cancer (CRC) cell proliferation and migration in vitro. However, whether or not AKAP-9 is important for CRC development or metastasis in vivo remains unknown. In the present study, we found that AKAP-9 expression was significantly higher in human colorectal cancer tissues than the paired normal tissues. In fact, AKAP-9 level correlated with the CRC infiltrating depth and metastasis. Moreover, the higher AKAP-9 expression was associated with the lower survival rate in patients. In cultured CRC cells, knockdown of AKAP-9 inhibited cell proliferation, invasion, and migration. AKAP-9 deficiency also attenuated CRC tumor growth and metastasis in vivo. Mechanistically, AKAP-9 interacted with cdc42 interacting protein 4 (CIP4) and regulated its expression. CIP4 levels were interrelated to the AKAP-9 level in CRC cells. Functionally, AKAP-9 was essential for TGF-β1-induced epithelial-mesenchymal transition of CRC cells, and CIP4 played a critical role in mediating the function of AKAP-9. Importantly, CIP4 expression was significantly up-regulated in human CRC tissues. Taken together, our results demonstrated that AKAP-9 facilitates CRC development and metastasis via regulating CIP4-mediated epithelial-mesenchymal transition of CRC cells.
Collapse
Affiliation(s)
- Zhi-Yan Hu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Molecular Tumour Pathology, Guangzhou 510515, China
| | - Yan-Ping Liu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Molecular Tumour Pathology, Guangzhou 510515, China
| | - Lin-Ying Xie
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Molecular Tumour Pathology, Guangzhou 510515, China
| | - Xiao-Yan Wang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Molecular Tumour Pathology, Guangzhou 510515, China
| | - Fang Yang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Molecular Tumour Pathology, Guangzhou 510515, China
| | - Shi-You Chen
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA, United States.
| | - Zu-Guo Li
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Molecular Tumour Pathology, Guangzhou 510515, China.
| |
Collapse
|
265
|
AlHossiny M, Luo L, Frazier WR, Steiner N, Gusev Y, Kallakury B, Glasgow E, Creswell K, Madhavan S, Kumar R, Upadhyay G. Ly6E/K Signaling to TGFβ Promotes Breast Cancer Progression, Immune Escape, and Drug Resistance. Cancer Res 2016; 76:3376-86. [PMID: 27197181 DOI: 10.1158/0008-5472.can-15-2654] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 03/29/2016] [Indexed: 12/11/2022]
Abstract
Stem cell antigen Sca-1 is implicated in murine cancer stem cell biology and breast cancer models, but the role of its human homologs Ly6K and Ly6E in breast cancer are not established. Here we report increased expression of Ly6K/E in human breast cancer specimens correlates with poor overall survival, with an additional specific role for Ly6E in poor therapeutic outcomes. Increased expression of Ly6K/E also correlated with increased expression of the immune checkpoint molecules PDL1 and CTLA4, increased tumor-infiltrating T regulatory cells, and decreased natural killer (NK) cell activation. Mechanistically, Ly6K/E was required for TGFβ signaling and proliferation in breast cancer cells, where they contributed to phosphorylation of Smad1/5 and Smad2/3. Furthermore, Ly6K/E promoted cytokine-induced PDL1 expression and activation and binding of NK cells to cancer cells. Finally, we found that Ly6K/E promoted drug resistance and facilitated immune escape in this setting. Overall, our results establish a pivotal role for a Ly6K/E signaling axis involving TGFβ in breast cancer pathophysiology and drug response, and highlight this signaling axis as a compelling realm for therapeutic invention. Cancer Res; 76(11); 3376-86. ©2016 AACR.
Collapse
Affiliation(s)
- Midrar AlHossiny
- Department of Oncology, Georgetown University Medical Center, Washington, DC
| | - Linlin Luo
- Innovation Center for Biomedical Informatics (ICBI), Georgetown University Medical Center, Washington, DC
| | - William R Frazier
- Department of Oncology, Georgetown University Medical Center, Washington, DC
| | - Noriko Steiner
- Department of Oncology, Georgetown University Medical Center, Washington, DC
| | - Yuriy Gusev
- Department of Oncology, Georgetown University Medical Center, Washington, DC. Innovation Center for Biomedical Informatics (ICBI), Georgetown University Medical Center, Washington, DC
| | - Bhaskar Kallakury
- Department of Pathology, Georgetown University Medical Center, Washington, DC
| | - Eric Glasgow
- Department of Oncology, Georgetown University Medical Center, Washington, DC
| | - Karen Creswell
- Department of Oncology, Georgetown University Medical Center, Washington, DC
| | - Subha Madhavan
- Department of Oncology, Georgetown University Medical Center, Washington, DC. Innovation Center for Biomedical Informatics (ICBI), Georgetown University Medical Center, Washington, DC
| | - Rakesh Kumar
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC
| | - Geeta Upadhyay
- Department of Oncology, Georgetown University Medical Center, Washington, DC. Innovation Center for Biomedical Informatics (ICBI), Georgetown University Medical Center, Washington, DC.
| |
Collapse
|
266
|
Abstract
Epithelial-to-mesenchymal transition (EMT) is a complex physiological and pathological process in which epithelial cells acquire mesenchymal characteristics. EMT occurs during embryogenesis and organ development, wound healing and organ regeneration, tumor migration and invasion. In recent years, cholangiocytes have been shown to undergo EMT in different cholangiopathies including hepatolithiasis. Transforming growth factor-β/Smads signaling is considered the master regulator. The purpose of this article is to introduce the concept and type of EMT, summarize recent advances that support or refute the concept that cholangiocytes are capable of phenotype transition of hepaticlithiasis and discuss the probable mechanism.
Collapse
|
267
|
Wang H, Wang Y, Du Q, Lu P, Fan H, Lu J, Hu R. Inflammasome-independent NLRP3 is required for epithelial-mesenchymal transition in colon cancer cells. Exp Cell Res 2016; 342:184-92. [PMID: 26968633 DOI: 10.1016/j.yexcr.2016.03.009] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/07/2016] [Accepted: 03/07/2016] [Indexed: 12/22/2022]
Abstract
Inflammasome NLRP3 plays a crucial role in the process of colitis and colitis--associated colon cancer. Even though much is known regarding the NLRP3 inflammasome that regulates pro-inflammatory cytokine release in innate immune cells, the role of NLRP3 in non-immune cells is still unclear. In this study, we showed that NLRP3 was highly expressed in mesenchymal-like colon cancer cells (SW620), and was upregulated by tumor necrosis factors-α (TNF-α) and transforming growth factor-β1 (TGF-β1) respectively, during EMT in colon cancer epithelial cells HCT116 and HT29. Knockdown of NLRP3 retained epithelial spindle-like morphology of HCT116 and HT29 cells and reversed the mesenchymal characteristic of SW620 cells, indicated by the decreased expression of vimentin and MMP9 and increased expression of E-cadherin. In addition, knockdown of NLRP3 in colorectal carcinoma cells displayed diminished cell migration and invasion. Interestingly, during the EMT process induced by TNF-α or TGF-β1, the cleaved caspase-1 and ASC speck were not detected, indicating that NLRP3 functions in an inflammasome-independent way. Further studies demonstrated that NLRP3 protein expression was regulated by NF-κB signaling in TNF-α or TGF-β1-induced EMT, as verified by the NF-κB inhibitor Bay 11-7082. Moreover, NLRP3 knockdown reduced the expression of Snail1, indicating that NLRP3 may promote EMT through regulating Snail1. In summary, our results showed that the NLRP3 expression, not the inflammasome activation, was required for EMT in colorectal cancer cells.
Collapse
Affiliation(s)
- Hong Wang
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Yajing Wang
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Qianming Du
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Ping Lu
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Huimin Fan
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Jinrong Lu
- Department of Organic Chemistry, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Rong Hu
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, Jiangsu 210009, China.
| |
Collapse
|
268
|
Antognelli C, Gambelunghe A, Muzi G, Talesa VN. Glyoxalase I drives epithelial-to-mesenchymal transition via argpyrimidine-modified Hsp70, miR-21 and SMAD signalling in human bronchial cells BEAS-2B chronically exposed to crystalline silica Min-U-Sil 5: Transformation into a neoplastic-like phenotype. Free Radic Biol Med 2016; 92:110-125. [PMID: 26784015 DOI: 10.1016/j.freeradbiomed.2016.01.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 01/12/2016] [Accepted: 01/13/2016] [Indexed: 12/15/2022]
Abstract
Glyoxalase I (Glo1) is the main scavenging enzyme of methylglyoxal (MG), a potent precursor of advanced glycation end products (AGEs). AGEs are known to control multiple biological processes, including epithelial to mesenchymal transition (EMT), a multistep phenomenon associated with cell transformation, playing a major role in a variety of diseases, including cancer. Crystalline silica is a well-known occupational health hazard, responsible for a great number of human pulmonary diseases, such as silicosis. There is still much debate concerning the carcinogenic role of crystalline silica, mainly due to the lack of a causal demonstration between silica exposure and carcinogenesis. It has been suggested that EMT might play a role in crystalline silica-induced lung neoplastic transformation. The aim of this study was to investigate whether, and by means of which mechanism, the antiglycation defence Glo1 is involved in Min-U-Sil 5 (MS5) crystalline silica-induced EMT in BEAS-2B human bronchial epithelial cells chronically exposed, and whether this is associated with the beginning of a neoplastic-like transformation process. By using gene silencing/overexpression and scavenging/inhibitory agents, we demonstrated that MS5 induced hydrogen peroxide-mediated c-Jun-dependent Glo1 up-regulation which resulted in a decrease in the Argpyrimidine-modified Hsp70 protein level which triggered EMT in a novel mechanism involving miR-21 and SMAD signalling. The observed EMT was associated with a neoplastic-like phenotype. The results obtained provide a causal in vitro demonstration of the MS5 pro-carcinogenic transforming role and more importantly they provide new insights into the mechanisms involved in this process, thus opening new paths in research concerning the in vivo study of the carcinogenic potential of crystalline silica.
Collapse
Affiliation(s)
- Cinzia Antognelli
- Department of Experimental Medicine, School of Medicine and Surgery, University of Perugia, Piazzale L. Severi 1, 06129 Perugia, Italy.
| | - Angela Gambelunghe
- Department of Medicine, School of Medicine and Surgery, University of Perugia, Piazzale L. Severi 1, 06129 Perugia, Italy.
| | - Giacomo Muzi
- Department of Medicine, School of Medicine and Surgery, University of Perugia, Piazzale L. Severi 1, 06129 Perugia, Italy.
| | - Vincenzo Nicola Talesa
- Department of Experimental Medicine, School of Medicine and Surgery, University of Perugia, Piazzale L. Severi 1, 06129 Perugia, Italy.
| |
Collapse
|
269
|
Zhang C, Lu Y, Li Q, Mao J, Hou Z, Yu X, Fan S, Li J, Gao T, Yan B, Wang B, Song B, Li L. Salinomycin suppresses TGF-β1-induced epithelial-to-mesenchymal transition in MCF-7 human breast cancer cells. Chem Biol Interact 2016; 248:74-81. [PMID: 26896736 DOI: 10.1016/j.cbi.2016.02.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Revised: 11/28/2015] [Accepted: 02/07/2016] [Indexed: 12/11/2022]
Abstract
Epithelial-to-mesenchymal transition (EMT) is the major cause of breast cancer to initiate invasion and metastasis. Salinomycin (Sal) has been found as an effective chemical compound to kill breast cancer stem cells. However, the effect of Sal on invasion and metastasis of breast cancer is unclear. In the present study, we showed that Sal reversed transforming growth factor-β1 (TGF-β1) induced invasion and metastasis accompanied with down-regulation of MMP-2 by experiments on human breast cancer cell line MCF-7. Sal was able to inhibit TGF-β1-induced EMT phenotypic transition and the activation of key signaling molecules involved in Smad (p-Smad2/3,Snail1) and non-Smad (β-catenin, p-p38 MAPK) signals which cooperatively regulate the induction of EMT. Importantly, in a series of breast cancer specimens, we found strong correlation among E-cadherin expression, β-catenin expression, and the lymph node metastatic potential of breast cancer. Our research suggests that Sal is promised to be a chemotherapeutic drug by suppressing the metastasis of breast cancer.
Collapse
Affiliation(s)
- Chunying Zhang
- Department of Pathology, Dalian Medical University, Dalian 116044, PR China
| | - Ying Lu
- Department of Pathology, Dalian Medical University, Dalian 116044, PR China
| | - Qing Li
- Department of Pathology, Dalian Medical University, Dalian 116044, PR China
| | - Jun Mao
- The Key Laboratory of Tumor Stem Cell Research of Liaoning Province, Dalian Medical University, Dalian 116044, PR China
| | - Zhenhuan Hou
- Department of Pathology, Dalian Medical University, Dalian 116044, PR China
| | - Xiaotang Yu
- Department of Pathology, Dalian Medical University, Dalian 116044, PR China
| | - Shujun Fan
- Department of Pathology, Dalian Medical University, Dalian 116044, PR China
| | - Jiazhi Li
- Department of Pathology, Dalian Medical University, Dalian 116044, PR China
| | - Tong Gao
- Department of Pathology, Dalian Medical University, Dalian 116044, PR China
| | - Bing Yan
- Department of Pathology, Dalian Medical University, Dalian 116044, PR China
| | - Bo Wang
- Department of Pathology, Dalian Medical University, Dalian 116044, PR China
| | - Bo Song
- Department of Pathology, Dalian Medical University, Dalian 116044, PR China
| | - Lianhong Li
- Department of Pathology, Dalian Medical University, Dalian 116044, PR China; The Key Laboratory of Tumor Stem Cell Research of Liaoning Province, Dalian Medical University, Dalian 116044, PR China.
| |
Collapse
|
270
|
Wang H, Chen Y, Han J, Meng Q, Xi Q, Wu G, Zhang B. DCAF4L2 promotes colorectal cancer invasion and metastasis via mediating degradation of NFκb negative regulator PPM1B. Am J Transl Res 2016; 8:405-418. [PMID: 27158335 PMCID: PMC4846892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 01/03/2016] [Indexed: 06/05/2023]
Abstract
DCAF4L2 is a member of WD-repeat proteins, which commonly serve as mediators of protein-protein interplay. In this study, we reported that elevated DCAF4L2 expression in human colorectal cancer (CRC) significantly correlated with a more advanced clinical stage as in lymphatic and distant metastasis. More importantly, elevated DCAF4L2 expression is an independent prognosis factor for survival. Genetic perturbations demonstrated that DCAF4L2 overexpression in CRC cells promoted cell migration and invasion, whereas knockdown of which had opposing effects. Moreover we discovered that DCAF4L2 overexpression could promote epithelial-mesenchymal-transition (EMT) through activating NFκB signal pathway. Mass spectrometry analysis showed that DCAF4L2 could form an E3 ligase complex with Cul4A and DDB1 thus mediated degradation of PPM1B, which has been reported to negatively regulate NFκB signaling. We identified PPM1B as a substrate of Cul4A-DDB1-DCAF4L2 E3 ligase complex, as knockdown of PPM1B abrogated shDCAF4L2 mediated inhibition of cell invasion in CRC cells. For further verification, DCAF4L2 expression inversely correlated with PPM1B expression in a cohort of 87 CRC patients. These findings may provide insight into the understanding of DCAF4L2 as a novel critical factor and a candidate target for CRC treatment.
Collapse
Affiliation(s)
- Haiyu Wang
- Department of General Surgery, Zhongshan Hospital, Fudan University Shanghai, China
| | - Yusheng Chen
- Department of General Surgery, Zhongshan Hospital, Fudan University Shanghai, China
| | - Jun Han
- Department of General Surgery, Zhongshan Hospital, Fudan University Shanghai, China
| | - Qingyang Meng
- Department of General Surgery, Zhongshan Hospital, Fudan University Shanghai, China
| | - Qiulei Xi
- Department of General Surgery, Zhongshan Hospital, Fudan University Shanghai, China
| | - Guohao Wu
- Department of General Surgery, Zhongshan Hospital, Fudan University Shanghai, China
| | - Bo Zhang
- Department of General Surgery, Zhongshan Hospital, Fudan University Shanghai, China
| |
Collapse
|
271
|
Ding H, Xu Y, Gao D, Wang L. Glioma-associated oncogene homolog 1 promotes epithelial-mesenchymal transition in human renal tubular epithelial cell. Am J Transl Res 2016; 8:662-669. [PMID: 27158358 PMCID: PMC4846915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 12/15/2015] [Indexed: 06/05/2023]
Abstract
Sonic hedgehog (Shh) signaling critically regulates embryogenesis and tissue homeostasis. Here, we investigated the role of Shh signaling in mediating epithelial-mesenchymal transition (EMT) in human renal tubular epithelial cells HKC-8. Our RT-PCR assays demonstrated that TGF-β1 induced time-dependent changes in the mRNA transcript levels of Shh, with a steady rise from one hour post TGF-β1 treatment and a peak at four hours post TGF-β1 treatment. Furthermore, TGF-β1 induced a time-dependent increase in the mRNA transcript levels of Gli1. Pre-treatment with 2 or 5 µM cyclopamine significantly attenuated TGF-β1-induced rise in the mRNA transcript levels of Gli1, but failed to attenuate TGF-β1-induced rise in Shh mRNA transcript levels. Additionally, immunoblotting assays and immunofluorescence staining demonstrated that inhibition of Shh signaling by cyclopamine significantly attenuated TGF-β1-induced increase in the mRNA transcript levels of α-SMA, collagen I, and fibronectin. Gli1 overexpression induced Snail1 expression. Moreover, Gli(-/-) mice that had undergone unilateral ureteral obstruction for seven days showed significant reduction in the mRNA transcript levels of Snail1 compared to the wildtype controls. In conclusion, the current study provides novel insight into the regulation of EMT by the Shh/Gli1 signaling pathway, suggesting a critical role of Shh/Gli1 signaling in EMT of human renal tubular epithelial cells.
Collapse
Affiliation(s)
- Hong Ding
- Division of Nephrology, The Forth Affiliated Hospital, China Medical University Shenyang, Liaoning 110032, China
| | - Yanyan Xu
- Division of Nephrology, The Forth Affiliated Hospital, China Medical University Shenyang, Liaoning 110032, China
| | - Di Gao
- Division of Nephrology, The Forth Affiliated Hospital, China Medical University Shenyang, Liaoning 110032, China
| | - Lei Wang
- Division of Nephrology, The Forth Affiliated Hospital, China Medical University Shenyang, Liaoning 110032, China
| |
Collapse
|
272
|
Zhou S, Tang X, Tang F. Krüppel-like factor 17, a novel tumor suppressor: its low expression is involved in cancer metastasis. Tumour Biol 2016; 37:1505-1513. [PMID: 26662959 PMCID: PMC4842221 DOI: 10.1007/s13277-015-4588-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 12/03/2015] [Indexed: 12/18/2022] Open
Abstract
Krüppel-like factor (KLF) family is highly conserved zinc finger transcription factors that regulate cell proliferation, differentiation, apoptosis, and migration. KLF17 is a member of the KLF family. Recent studies have demonstrated that KLF17 low expression and inactivation are caused by microRNA, gene mutation, and loss of heterozygosity in human tumors, which participates in tumor progression. KLF17 low expression increases cancer metastatic viability; its mechanism is that low KLF17 mediates epithelial-mesenchymal transition (EMT) through regulating EMT-related genes expression; the reduced-KLF17 also increases cancer metastasis though upregulating inhibitor of DNA binding 1 (ID1). Additionally, mutant p53 proteins are capable of developing a complex with KLF17, which mediate the depletion of KLF17 inhibiting EMT gene transcription and increases cancer metastasis. KLF17 downregulation also mediates the activation of TGF-β pathway.
Collapse
Affiliation(s)
- Shan Zhou
- Medical Research Center and Clinical Laboratory, Zhuhai People’s Hospital and Zhuhai Hospital of Jinan University, 79 Kangning Road, Zhuhai, 519000 Guangdong China
| | - Xiaowei Tang
- Metallurgical Science and Engineering, Central South University, 21# Lushan South Road, Changsha, 410083 China
| | - Faqing Tang
- Medical Research Center and Clinical Laboratory, Zhuhai People’s Hospital and Zhuhai Hospital of Jinan University, 79 Kangning Road, Zhuhai, 519000 Guangdong China
| |
Collapse
|
273
|
Runa F, Adamian Y, Kelber JA. Ascending the PEAK1 toward targeting TGFβ during cancer progression: Recent advances and future perspectives. CANCER CELL & MICROENVIRONMENT 2016; 3:e1162. [PMID: 29392163 PMCID: PMC5790177 DOI: 10.14800/ccm.1162] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cancer is the second leading cause of death in the United States. Mortality in patients with solid, epithelial-derived tumors strongly correlates with disease stage and the systemic metastatic load. In such cancers, notable morphological and molecular changes have been attributed to cells as they pass through a continuum of epithelial-mesenchymal transition (EMT) states and many of these changes are essential for metastasis. While cancer metastasis is a complex cascade that is regulated by cell-autonomous and microenvironmental influences, it is well-accepted that understanding and controlling metastatic disease is a viable method for increasing patient survival. In the past 5 years, the novel non-receptor tyrosine kinase PEAK1 has surfaced as a central regulator of tumor progression and metastasis in the context of solid, epithelial cancers. Here, we review this literature with a special focus on our recent work demonstrating that PEAK1 mediates non-canonical pro-tumorigenic TGFβ signaling and is an intracellular control point between tumor cells and their extracellular microenvironment. We conclude with a brief discussion of potential applications derived from our current understanding of PEAK1 biology.
Collapse
Affiliation(s)
- Farhana Runa
- Department of Biology, California State University, Northridge, CA, USA
| | - Yvess Adamian
- Department of Biology, California State University, Northridge, CA, USA
| | | |
Collapse
|
274
|
Analysis of Hippo and TGFβ signaling in polarizing epithelial cells and mouse embryos. Differentiation 2016; 91:109-18. [PMID: 26803209 DOI: 10.1016/j.diff.2016.01.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 01/07/2016] [Indexed: 11/20/2022]
Abstract
The Hippo signaling pathway is involved in numerous biological events ranging from early development to organogenesis and when disrupted, impacts various human diseases including cancer. The Hippo pathway also interacts with and controls the activity of other signaling pathways such as the TGFβ/Smad pathway, in which Hippo pathway activity influences the subcellular localization of Smad transcription factors. Here, we describe techniques for examining crosstalk between Hippo and TGFβ signaling in polarizing mammary epithelial cells. In addition, we provide detailed methods for analyzing the subcellular localization of the Hippo pathway effectors, Taz and Yap using both in vitro cultured epithelial cells and in vivo in pregastrulation mouse embryos.
Collapse
|
275
|
Yoshida T, Song L, Bai Y, Kinose F, Li J, Ohaegbulam KC, Muñoz-Antonia T, Qu X, Eschrich S, Uramoto H, Tanaka F, Nasarre P, Gemmill RM, Roche J, Drabkin HA, Haura EB. ZEB1 Mediates Acquired Resistance to the Epidermal Growth Factor Receptor-Tyrosine Kinase Inhibitors in Non-Small Cell Lung Cancer. PLoS One 2016; 11:e0147344. [PMID: 26789630 PMCID: PMC4720447 DOI: 10.1371/journal.pone.0147344] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 01/01/2016] [Indexed: 01/01/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) is one mechanism of acquired resistance to inhibitors of the epidermal growth factor receptor-tyrosine kinases (EGFR-TKIs) in non-small cell lung cancer (NSCLC). The precise mechanisms of EMT-related acquired resistance to EGFR-TKIs in NSCLC remain unclear. We generated erlotinib-resistant HCC4006 cells (HCC4006ER) by chronic exposure of EGFR-mutant HCC4006 cells to increasing concentrations of erlotinib. HCC4006ER cells acquired an EMT phenotype and activation of the TGF-β/SMAD pathway, while lacking both T790M secondary EGFR mutation and MET gene amplification. We employed gene expression microarrays in HCC4006 and HCC4006ER cells to better understand the mechanism of acquired EGFR-TKI resistance with EMT. At the mRNA level, ZEB1 (TCF8), a known regulator of EMT, was >20-fold higher in HCC4006ER cells than in HCC4006 cells, and increased ZEB1 protein level was also detected. Furthermore, numerous ZEB1 responsive genes, such as CDH1 (E-cadherin), ST14, and vimentin, were coordinately regulated along with increased ZEB1 in HCC4006ER cells. We also identified ZEB1 overexpression and an EMT phenotype in several NSCLC cells and human NSCLC samples with acquired EGFR-TKI resistance. Short-interfering RNA against ZEB1 reversed the EMT phenotype and, importantly, restored erlotinib sensitivity in HCC4006ER cells. The level of micro-RNA-200c, which can negatively regulate ZEB1, was significantly reduced in HCC4006ER cells. Our results suggest that increased ZEB1 can drive EMT-related acquired resistance to EGFR-TKIs in NSCLC. Attempts should be made to explore targeting ZEB1 to resensitize TKI-resistant tumors.
Collapse
Affiliation(s)
- Takeshi Yoshida
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Lanxi Song
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Yun Bai
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Fumi Kinose
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Jiannong Li
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Kim C. Ohaegbulam
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Teresita Muñoz-Antonia
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Xiaotao Qu
- Department of Biomedical Informatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Steven Eschrich
- Department of Biomedical Informatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Hidetaka Uramoto
- Second Department of Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
| | - Fumihiro Tanaka
- Second Department of Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
| | - Patrick Nasarre
- Division of Hematology-Oncology, Department of Medicine and the Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Robert M. Gemmill
- Division of Hematology-Oncology, Department of Medicine and the Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Joëlle Roche
- Division of Hematology-Oncology, Department of Medicine and the Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Harry A. Drabkin
- Division of Hematology-Oncology, Department of Medicine and the Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Eric B. Haura
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
- * E-mail:
| |
Collapse
|
276
|
Zhao L, Mao Y, Zhou J, Zhao Y, Cao Y, Chen X. Multifunctional DDX3: dual roles in various cancer development and its related signaling pathways. Am J Cancer Res 2016; 6:387-402. [PMID: 27186411 PMCID: PMC4859668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 01/12/2016] [Indexed: 06/05/2023] Open
Abstract
DEAD-box RNA helicase 3 (DDX3) is a highly conserved family member of DEAD-box protein, which is a cluster of ATP-dependent and the largest family of RNA helicase. DEAD-box family is characterized by the regulation of ATPase and helicase activities, the modulation of RNA metabolism, and the actors of RNA binding proteins or molecular chaperones to interact with other proteins or RNA. For DDX3, it exerts its multifaceted roles in viral manipulation, stress response, hypoxia, radiation response and apoptosis, and is closely related to cancer development and progression. DDX3 has dual roles in different cancer types and can act as either an oncogene or tumor suppressor gene during cancer progression. In the present review, we mainly provide an overview of current knowledge on dual roles of DDX3 in various types of cancer, including breast cancer, lung cancer, colorectal cancer, hepatocellular carcinoma, oral squamous cell carcinoma, Ewing sarcoma, glioblastoma multiforme and gallbladder carcinoma, and illustrate the regulatory mechanisms for leading these two controversial biological effects. Furthermore, we summarize the essential signaling pathways that DDX3 participated, especially the Wnt/β-catenin signaling and EMT related signaling (TGF-β, Notch, Hedgehog pathways), which are crucial to DDX3 mediated cancer metastasis process. Thoroughly exploring the dual roles of DDX3 in cancer development and the essential signaling pathways it involved, it will help us open new perspectives to develop novel promising targets to elevate therapeutic effects and facilitate the "Personalized medicine" or "Precision medicine" to come into clinic.
Collapse
Affiliation(s)
- Luqing Zhao
- Department of Pathology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, China
- Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South UniversityChangsha 410013, Hunan, China
- Department of Dermatology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, China
| | - Yitao Mao
- Department of Radiology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, China
| | - Jianhua Zhou
- Department of Pathology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, China
- Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South UniversityChangsha 410013, Hunan, China
| | - Yuelong Zhao
- School of Computer Science and Engineering, South China University of TechnologyGuangzhou 510640, Guangdong, China
| | - Ya Cao
- Cancer Research Institute, School of Basic Medical Science, Xiangya School of Medicine, Central South UniversityChangsha 410013, Hunan, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, China
| |
Collapse
|
277
|
Revisiting Epithelial-to-Mesenchymal Transition in Liver Fibrosis: Clues for a Better Understanding of the "Reactive" Biliary Epithelial Phenotype. Stem Cells Int 2016; 2016:2953727. [PMID: 26880950 PMCID: PMC4736590 DOI: 10.1155/2016/2953727] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 10/20/2015] [Indexed: 12/27/2022] Open
Abstract
Whether liver epithelial cells contribute to the development of hepatic scarring by undergoing epithelial-to-mesenchymal transition (EMT) is a controversial issue. Herein, we revisit the concept of EMT in cholangiopathies, a group of severe hepatic disorders primarily targeting the bile duct epithelial cell (cholangiocyte), leading to progressive portal fibrosis, the main determinant of liver disease progression. Unfortunately, therapies able to halt this process are currently lacking. In cholangiopathies, fibrogenesis is part of ductular reaction, a reparative complex involving epithelial, mesenchymal, and inflammatory cells. Ductular reactive cells (DRC) are cholangiocytes derived from the activation of the hepatic progenitor cell compartment. These cells are arranged into irregular strings and express a “reactive” phenotype, which enables them to extensively crosstalk with the other components of ductular reaction. We will first discuss EMT in liver morphogenesis and then highlight how some of these developmental programs are partly reactivated in DRC. Evidence for “bona fide” EMT changes in cholangiocytes is lacking, but expression of some mesenchymal markers represents a fundamental repair mechanism in response to chronic biliary damage with potential harmful fibrogenetic effects. Understanding microenvironmental cues and signaling perturbations promoting these changes in DRC may help to identify potential targets for new antifibrotic therapies in cholangiopathies.
Collapse
|
278
|
Cives M, Rizzo F, Simone V, Bisceglia F, Stucci S, Seeber A, Spizzo G, Montrone T, Resta L, Silvestris F. Reviewing the Osteotropism in Neuroendocrine Tumors: The Role of Epithelial-Mesenchymal Transition. Neuroendocrinology 2016; 103:321-34. [PMID: 26227818 DOI: 10.1159/000438902] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 07/15/2015] [Indexed: 11/19/2022]
Abstract
BACKGROUND Neuroendocrine tumors (NETs) metastasize to the bone. However, the incidence, clinical features, management and pathogenesis of bone involvement in NET patients have been poorly investigated. METHODS We reviewed all published reports of histologically confirmed bone metastatic NETs and explored clinical, radiological, prognostic and therapeutic characteristics in a population of 152 patients. We then evaluated immunohistochemical expression of a panel of eight epithelial-mesenchymal transition (EMT)-related factors including SNAIL, TGF-β1, CTGF, IL-11, PTHrP, EpCAM, CXCR4 and RANK in an independent cohort of 44 archival primary NETs. Biomarker expression was correlated with clinicopathological variables, including skeletal involvement, and tested for survival prediction. RESULTS We found that 55% of NET patients with bone metastases were male, with a median age of 55 years at diagnosis. Metastases were restricted to the skeleton in 34% of the NET population, and axial and osteoblastic lesions were prevalent. NETs differently expressed proteins involved in EMT activation. High CXCR4 (p < 0.0001) and low TGF-β1 levels (p = 0.0015) were significantly associated with increased risk of skeletal metastases, suggesting that EMT is implicated in NET osteotropism. By applying an algorithm measuring distinct immunohistochemical predictors of osteotropism on primary tumors, we were able to identify NET patients with bone metastases with a sensitivity and specificity of 91 and 100%, respectively (p < 0.0001). Patients whose primary tumors expressed CTGF (p = 0.0007) as well as the truncated form of EpCAM (p = 0.06) showed shorter survival. CONCLUSION Although underestimated, bone metastases are a prominent feature of NETs, and the tumor expression of EMT markers at diagnosis may predict concurrent or subsequent skeleton colonization.
Collapse
Affiliation(s)
- Mauro Cives
- Department of Biomedical Sciences and Clinical Oncology, University of Bari, Bari, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
279
|
Iskender B, Izgi K, Hizar E, Jauch J, Arslanhan A, Yuksek EH, Canatan H. Inhibition of epithelial-mesenchymal transition in bladder cancer cells via modulation of mTOR signalling. Tumour Biol 2015; 37:8281-91. [PMID: 26718217 DOI: 10.1007/s13277-015-4695-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 12/16/2015] [Indexed: 11/30/2022] Open
Abstract
Mounting evidence suggests that signalling cross-talk plays a significant role in the regulation of epithelial-mesenchymal transition (EMT) in cancer cells. However, the complex network regulating the EMT in different cancer types has not been fully described yet which affects the development of novel therapeutic strategies. In the present study, we investigated the signalling pathways involved in EMT of bladder cancer cells and demonstrated the effects of two novel agents in the regulation of EMT. Myrtucommulone-A (MC-A) and thymoquinone (TQ) have been shown to possess anti-cancer properties. However, their targets in the regulation of cancer cell behavior are not well defined. Here, we defined the effects of two putative anti-cancer agents on bladder cancer cell migration and their possible intracellular targets in the regulation of EMT. Our results suggest that MC-A or TQ treatment affected N-cadherin, Snail, Slug, and β-catenin expressions and effectively attenuated mTOR activity. The downstream components in mTOR signalling were also affected. MC-A treatment resulted in the concomitant inhibition of extracellular matrix-regulated protein kinases 1 and 2 (ERK 1/2), p38 mitogen-activated protein kinase (MAPK) and Src activity. On the other hand, TQ treatment increased Src activity while exerting no effect on ERK 1/2 or p38 MAPK activity. Given the stronger inhibition of EMT-related markers in MC-A-treated samples, we concluded that this effect might be due to collective inhibition of multiple signalling pathways which result in a decrease in their cross-talk in bladder cancer cells. Overall, the data in this study proposes novel action mechanisms for MC-A or TQ in bladder cancer cells and highlights the potential use of these active compounds in the regulation of EMT.
Collapse
Affiliation(s)
- Banu Iskender
- Department of Medical Biology, Faculty of Medicine, Erciyes University, 38039, Melikgazi, Kayseri, Turkey. .,Betul-Ziya Eren Genome and Stem Cell Centre, Erciyes University, 38039, Melikgazi, Kayseri, Turkey.
| | - Kenan Izgi
- Department of Medical Biochemistry, Faculty of Medicine, Erciyes University, 38039, Melikgazi, Kayseri, Turkey.,Betul-Ziya Eren Genome and Stem Cell Centre, Erciyes University, 38039, Melikgazi, Kayseri, Turkey
| | - Esra Hizar
- Department of Medical Biology, Faculty of Medicine, Erciyes University, 38039, Melikgazi, Kayseri, Turkey.,Betul-Ziya Eren Genome and Stem Cell Centre, Erciyes University, 38039, Melikgazi, Kayseri, Turkey
| | - Johann Jauch
- Universität des Saarlandes, Organische Chemie II, Geb. C4.2, 66123, Saarbrücken, Germany
| | - Aslihan Arslanhan
- Department of Medical Biochemistry, Faculty of Medicine, Erciyes University, 38039, Melikgazi, Kayseri, Turkey.,Betul-Ziya Eren Genome and Stem Cell Centre, Erciyes University, 38039, Melikgazi, Kayseri, Turkey
| | - Esra Hilal Yuksek
- Department of Medical Biochemistry, Faculty of Medicine, Erciyes University, 38039, Melikgazi, Kayseri, Turkey.,Betul-Ziya Eren Genome and Stem Cell Centre, Erciyes University, 38039, Melikgazi, Kayseri, Turkey
| | - Halit Canatan
- Department of Medical Biology, Faculty of Medicine, Erciyes University, 38039, Melikgazi, Kayseri, Turkey.,Betul-Ziya Eren Genome and Stem Cell Centre, Erciyes University, 38039, Melikgazi, Kayseri, Turkey
| |
Collapse
|
280
|
Muthusamy BP, Budi EH, Katsuno Y, Lee MK, Smith SM, Mirza AM, Akhurst RJ, Derynck R. ShcA Protects against Epithelial-Mesenchymal Transition through Compartmentalized Inhibition of TGF-β-Induced Smad Activation. PLoS Biol 2015; 13:e1002325. [PMID: 26680585 PMCID: PMC4682977 DOI: 10.1371/journal.pbio.1002325] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 11/10/2015] [Indexed: 12/15/2022] Open
Abstract
Epithelial–mesenchymal transition (EMT) is a normal cell differentiation event during development and contributes pathologically to carcinoma and fibrosis progression. EMT often associates with increased transforming growth factor-β (TGF-β) signaling, and TGF-β drives EMT, in part through Smad-mediated reprogramming of gene expression. TGF-β also activates the Erk MAPK pathway through recruitment and Tyr phosphorylation of the adaptor protein ShcA by the activated TGF-β type I receptor. We found that ShcA protects the epithelial integrity of nontransformed cells against EMT by repressing TGF-β-induced, Smad-mediated gene expression. p52ShcA competed with Smad3 for TGF-β receptor binding, and down-regulation of ShcA expression enhanced autocrine TGF-β/Smad signaling and target gene expression, whereas increased p52ShcA expression resulted in decreased Smad3 binding to the TGF-β receptor, decreased Smad3 activation, and increased Erk MAPK and Akt signaling. Furthermore, p52ShcA sequestered TGF-β receptor complexes to caveolin-associated membrane compartments, and reducing ShcA expression enhanced the receptor localization in clathrin-associated membrane compartments that enable Smad activation. Consequently, silencing ShcA expression induced EMT, with increased cell migration, invasion, and dissemination, and increased stem cell generation and mammosphere formation, dependent upon autocrine TGF-β signaling. These findings position ShcA as a determinant of the epithelial phenotype by repressing TGF-β-induced Smad activation through differential partitioning of receptor complexes at the cell surface. The adaptor protein ShcA protects epithelial cells from transitioning toward a mesenchymal phenotype by controlling partitioning of the TGF-β receptor and repressing downstream Smad2/3 activation. TGF-β family proteins control cell differentiation and various cell functions. Increased TGF-β signaling, acting through heteromeric receptor complexes, contributes to carcinoma progression and fibrosis. TGF-β drives epithelial–mesenchymal transdifferentiation (EMT), which enables cell migration and invasion. Upon TGF-β binding, “type I” receptors activate, through phosphorylation, Smad2 and Smad3 that control target gene transcription. In EMT, Smad complexes activate the expression of EMT “master” transcription factors and cooperate with these to repress the epithelial phenotype and activate mesenchymal gene expression. TGF-β receptors also activate Erk MAPK signaling, involving association of the adaptor protein ShcA and Tyr phosphorylation of ShcA by type I receptors. We now show that the predominant ShcA isoform, p52ShcA, competes with Smad2/3 for binding to type I TGF-β receptors, thus repressing Smad2/3 activation in response to TGF-β and localizing the receptors to caveolar compartments. Consequently, decreased ShcA expression enhanced TGF-β receptor localization in clathrin compartments and autocrine Smad2/3 signaling, repressed the epithelial phenotype, and promoted EMT. The changes following decreased ShcA expression resulted in increased cell migration and invasion, as well as increased stem cell generation, dependent upon autocrine TGF-β signaling. These findings position ShcA as a determinant of the epithelial phenotype by repressing TGF-β-induced Smad activation through differential partitioning of receptor complexes at the cell surface.
Collapse
Affiliation(s)
- Baby Periyanayaki Muthusamy
- Departments of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, United States of America
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States of America
| | - Erine H. Budi
- Departments of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, United States of America
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States of America
| | - Yoko Katsuno
- Departments of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, United States of America
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States of America
| | - Matthew K. Lee
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, California, United States of America
| | - Susan M. Smith
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, California, United States of America
| | - Amer M. Mirza
- XOMA Corp., Berkeley, California, United States of America
| | - Rosemary J. Akhurst
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States of America
- Department of Anatomy, University of California, San Francisco, San Francisco, California, United States of America
- Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California, United States of America
| | - Rik Derynck
- Departments of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, United States of America
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States of America
- Department of Anatomy, University of California, San Francisco, San Francisco, California, United States of America
- Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
281
|
Du F, Li S, Wang T, Zhang HY, Li DT, Du ZX, Wang HQ, Wang YQ. BAG3 regulates ECM accumulation in renal proximal tubular cells induced by TGF-β1. Am J Transl Res 2015; 7:2805-2814. [PMID: 26885277 PMCID: PMC4731677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 12/12/2015] [Indexed: 06/05/2023]
Abstract
Previously we have demonstrated that Bcl-2-associated athanogene 3 (BAG3) is increased in renal fibrosis using a rat unilateral ureteral obstruction model. The current study investigated the role of BAG3 in renal fibrosis using transforming growth factor (TGF)-β1-treated human proximal tubular epithelial (HK-2) cells. An upregulation of BAG3 in vitro models was observed, which correlated with the increased synthesis of extracellular matrix (ECM) proteins and expression of tissue-type plasminogen activator inhibitor (PAI)-1. Blockade of BAG3 induction by shorting hairpin RNA suppressed the expression of ECM proteins but had no effect on PAI-1 expression induced by TGF-β1. Forced overexpression of BAG3 selectively increased collagens. TGF-β1-induced BAG3 expression in HK-2 cells was attenuated by ERK1/2 and JNK MAPK inhibitors. In addition, forced BAG3 overexpression blocked attenuation of collagens expression by ERK1/2 and JNK inhibitors. These data suggest that ERK1/2 and JNK signaling events are involved in modulating the expression of BAG3, which would ultimately contribute to renal fibrosis by enhancing the synthesis and deposition of ECM proteins.
Collapse
Affiliation(s)
- Feng Du
- Department of Nephrology, Shengjing Hospital, China Medical UniversityShenyang 110004, China
| | - Si Li
- Department of Endocrinology & Metabolism, The 1 Affiliated Hospital, China Medical UniversityShenyang 110001, China
| | - Tian Wang
- Department of Endocrinology & Metabolism, The 1 Affiliated Hospital, China Medical UniversityShenyang 110001, China
| | - Hai-Yan Zhang
- Department of Geriatrics, The 1 Affiliated Hospital, China Medical UniversityShenyang 110001, China
| | - De-Tian Li
- Department of Nephrology, Shengjing Hospital, China Medical UniversityShenyang 110004, China
| | - Zhen-Xian Du
- Department of Endocrinology & Metabolism, The 1 Affiliated Hospital, China Medical UniversityShenyang 110001, China
| | - Hua-Qin Wang
- Department of Biochemistry & Molecular Biology, China Medical UniversityShenyang 110001, China
| | - Yan-Qiu Wang
- Department of Nephrology, Shengjing Hospital, China Medical UniversityShenyang 110004, China
| |
Collapse
|
282
|
Liang D, Wang Y, Zhu Z, Yang G, An G, Li X, Niu P, Chen L, Tian L. BMP-7 attenuated silica-induced pulmonary fibrosis through modulation of the balance between TGF-β/Smad and BMP-7/Smad signaling pathway. Chem Biol Interact 2015; 243:72-81. [PMID: 26585589 DOI: 10.1016/j.cbi.2015.11.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Revised: 10/13/2015] [Accepted: 11/09/2015] [Indexed: 10/22/2022]
Abstract
OBJECTIVE To investigate the anti-fibrotic effects and possible mechanisms of bone morphogenetic protein-7 (BMP-7) on silica induced fibrosis in RLE-6TN cells, and compare the preventive treatment of experimental silicosis with BMP-7 with therapeutic treatment of silicosis in vitro models. METHODS RLE-6TN cells were incubated with the supernatant of RAW264.7, treated by 50 μg/mL silica in either presence or absence of BMP-7 in different phases. Morphological changes and the cellular wound-healing assays were used to evaluate the process of EMT. By using Western Blotting, the epithelial marker E-cadherin (E-cad), and the mesenchymal markers Vimentin (Vim), Snail, and fibronectin (FN) were detected as well as the Smad signaling pathway proteins, including phosphorylated Smad1/5(P-Smad1/5), phosphorylated Smad2/3(P-Smad2/3), and non-phosphorylated Smad1, Smad8, and Smad2. The progress of fibrosis was assessed by the content of hydroxyproline (Hyp) and collagen I and III protein levels. In addition, MTT assay was used to explore the toxic effects of silica as well as BMP-7. RESULTS The EMT model of RLE-6TN cells was established successfully, the cells had a fibroblast-like morphology with increasing migration activity. The expressions of Vim, Snail, FN, collagen I and collagen III were up-regulated with the increase of silica concentration. BMP-7 could attenuate the decrease of P-Smad1/5 and the increase of P-Smad2/3, collagen I, collagen III, and FN via Smad signaling pathway. BMP-7 inhibited the mesenchymal-like responses in RLE-6TN cells, including cell migration, expression of fibrosis markers, and secretion of Hyp. Furthermore, the anti-fibrotic effects in the prevention group were more effective than treatment group. CONCLUSION The restoration of BMP signaling with BMP-7 is associated with inhibiting silica-induced fibrosis through the mechanisms of activated BMP-7/Smad and suppressed TGF-β/Smad pathways. Preventive treatment of pulmonary fibrosis progression with BMP-7 may expect to be the optimized strategy than therapeutic therapy of fibrosis.
Collapse
Affiliation(s)
- Di Liang
- School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Yan Wang
- School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Zhonghui Zhu
- School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Gengxia Yang
- Oncology Minimally Invasive Interventional Center, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Guoliang An
- School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Xiaoli Li
- School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Piye Niu
- School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Li Chen
- School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Lin Tian
- School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
283
|
Su H, Jin X, Shen L, Fang Y, Fei Z, Zhang X, Xie C, Chen X. Inhibition of cyclin D1 enhances sensitivity to radiotherapy and reverses epithelial to mesenchymal transition for esophageal cancer cells. Tumour Biol 2015; 37:5355-63. [PMID: 26561473 DOI: 10.1007/s13277-015-4393-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 11/05/2015] [Indexed: 12/13/2022] Open
Abstract
Acquired radioresistance during radiotherapy has significantly affected the treatment efficacy in esophageal cancer. Many of radioresistant cancer cells demonstrated epithelial-mesenchymal transition (EMT).We found in previous study that a radioresistant cell line (KYSE-150R) possessed EMT characteristic with cyclin D1 overexpression. Cyclin D1 has been demonstrated to affect the radiation sensitivity in cancer cells. To elucidate the molecular functions of cyclin D1 on EMT phenotypes and esophageal cancer radiosensitivity, we treated the radioresistant esophageal cancer cells (KYSE-150R) and parental cells (KYSE-150) with cyclin D1 small interfering RNA (siRNA). The cell proliferation rate of KYSE-150R and the radiation survival fraction were significantly decreased in cyclin D1 siRNA treatment group. Knocking down cyclin D1 resulted in G0/G1 arrest in KYSE-150R cells. The average number of irradiation-induced γ-H2AX foci increased in the cells treated with cyclin D1 siRNA, indicating impaired DNA double-strand break (DSB) repair in KYSE-150R cells. Cyclin D1 also reversed EMT phenotypes with significantly increased expression of E-cadherin in KYSE-150R cells. However, cyclin D1 siRNA have no radiosensitizing effects on KYSE-150 cells, with no obvious change in EMT marker expression .Our work showed that EMT phenotypes can be reduced and the radiosensitivity of esophageal cancer cells can be enhanced by inhibiting cyclin D1.
Collapse
Affiliation(s)
- Huafang Su
- Department of Radiotherapy and Chemotherapy, The First Affiliated Hospital of Wenzhou Medical University, No. 2 Fuxue Lane, Wenzhou, 325000, China
| | - Xiance Jin
- Department of Radiotherapy and Chemotherapy, The First Affiliated Hospital of Wenzhou Medical University, No. 2 Fuxue Lane, Wenzhou, 325000, China
| | - Lanxiao Shen
- Department of Radiotherapy and Chemotherapy, The First Affiliated Hospital of Wenzhou Medical University, No. 2 Fuxue Lane, Wenzhou, 325000, China
| | - Ya Fang
- Department of Radiotherapy and Chemotherapy, The First Affiliated Hospital of Wenzhou Medical University, No. 2 Fuxue Lane, Wenzhou, 325000, China
| | - Zhenghua Fei
- Department of Radiotherapy and Chemotherapy, The First Affiliated Hospital of Wenzhou Medical University, No. 2 Fuxue Lane, Wenzhou, 325000, China
| | - Xuebang Zhang
- Department of Radiotherapy and Chemotherapy, The First Affiliated Hospital of Wenzhou Medical University, No. 2 Fuxue Lane, Wenzhou, 325000, China
| | - Congying Xie
- Department of Radiotherapy and Chemotherapy, The First Affiliated Hospital of Wenzhou Medical University, No. 2 Fuxue Lane, Wenzhou, 325000, China.
| | - Xiaolei Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
284
|
Chen C, Li R, Ross RS, Manso AM. Integrins and integrin-related proteins in cardiac fibrosis. J Mol Cell Cardiol 2015; 93:162-74. [PMID: 26562414 DOI: 10.1016/j.yjmcc.2015.11.010] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 11/07/2015] [Accepted: 11/07/2015] [Indexed: 12/21/2022]
Abstract
Cardiac fibrosis is one of the major components of the healing mechanism following any injury of the heart and as such may contribute to both systolic and diastolic dysfunction in a range of pathophysiologic conditions. Canonically, it can occur as part of the remodeling process that occurs following myocardial infarction or that follows as a response to pressure overload. Integrins are cell surface receptors which act in both cellular adhesion and signaling. Most importantly, in the context of the continuously contracting myocardium, they are recognized as mechanotransducers. They have been implicated in the development of fibrosis in several organs, including the heart. This review will focus on the involvement of integrins and integrin-related proteins, in cardiac fibrosis, outlining the roles of these proteins in the fibrotic responses in specific cardiac pathologies, discuss some of the common end effectors (angiotensin II, transforming growth factor beta 1 and mechanical stress) through which integrins function and finally discuss how manipulation of this set of proteins may lead to new treatments which could prove useful to alter the deleterious effects of cardiac fibrosis.
Collapse
Affiliation(s)
- Chao Chen
- Department of Medicine, Cardiology, UCSD School of Medicine, La Jolla, CA 92093-0613, USA; Veterans Administration San Diego Healthcare System, San Diego, CA 92161, USA.
| | - Ruixia Li
- Department of Medicine, Cardiology, UCSD School of Medicine, La Jolla, CA 92093-0613, USA; Veterans Administration San Diego Healthcare System, San Diego, CA 92161, USA.
| | - Robert S Ross
- Department of Medicine, Cardiology, UCSD School of Medicine, La Jolla, CA 92093-0613, USA; Veterans Administration San Diego Healthcare System, San Diego, CA 92161, USA.
| | - Ana Maria Manso
- Department of Medicine, Cardiology, UCSD School of Medicine, La Jolla, CA 92093-0613, USA; Veterans Administration San Diego Healthcare System, San Diego, CA 92161, USA.
| |
Collapse
|
285
|
FU JIANJIANG, KE XIAOQIN, TAN SONGLIN, LIU TING, WANG SHAN, MA JUNCHAO, LU HONG. The natural compound codonolactone attenuates TGF-β1-mediated epithelial-to-mesenchymal transition and motility of breast cancer cells. Oncol Rep 2015; 35:117-26. [DOI: 10.3892/or.2015.4394] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 08/05/2015] [Indexed: 11/05/2022] Open
|
286
|
Early downregulation of acute phase proteins after doxorubicin exposition in patients with breast cancer. Tumour Biol 2015; 37:3775-83. [DOI: 10.1007/s13277-015-4203-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 10/05/2015] [Indexed: 10/22/2022] Open
|
287
|
Liu RM, Desai LP. Reciprocal regulation of TGF-β and reactive oxygen species: A perverse cycle for fibrosis. Redox Biol 2015; 6:565-577. [PMID: 26496488 PMCID: PMC4625010 DOI: 10.1016/j.redox.2015.09.009] [Citation(s) in RCA: 480] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 09/17/2015] [Accepted: 09/20/2015] [Indexed: 12/21/2022] Open
Abstract
Transforming growth factor beta (TGF-β) is the most potent pro-fibrogenic cytokine and its expression is increased in almost all of fibrotic diseases. Although signaling through Smad pathway is believed to play a central role in TGF-β's fibrogenesis, emerging evidence indicates that reactive oxygen species (ROS) modulate TGF-β's signaling through different pathways including Smad pathway. TGF-β1 increases ROS production and suppresses antioxidant enzymes, leading to a redox imbalance. ROS, in turn, induce/activate TGF-β1 and mediate many of TGF-β's fibrogenic effects, forming a vicious cycle (see graphic flow chart on the right). Here, we review the current knowledge on the feed-forward mechanisms between TGF-β1 and ROS in the development of fibrosis. Therapeutics targeting TGF-β-induced and ROS-dependent cellular signaling represents a novel approach in the treatment of fibrotic disorders. TGF-β1 is the most potent ubiquitous profibrogenic cytokine. TGF- β 1 induces redox imbalance by ↑ ROS production and ↓ anti-oxidant defense system Redox imbalance, in turn, activates latent TGF-β1 and induces TGF-β1 expression. Redox imbalance also mediates many of TGF-β1’s profibrogenic effects
Collapse
Affiliation(s)
- Rui-Ming Liu
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmi ngham, Birmingham, AL, USA.
| | - Leena P Desai
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmi ngham, Birmingham, AL, USA
| |
Collapse
|
288
|
Euphol from Euphorbia tirucalli Negatively Modulates TGF-β Responsiveness via TGF-β Receptor Segregation inside Membrane Rafts. PLoS One 2015; 10:e0140249. [PMID: 26448474 PMCID: PMC4598150 DOI: 10.1371/journal.pone.0140249] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Accepted: 09/23/2015] [Indexed: 01/07/2023] Open
Abstract
Transforming growth factor-β (TGF-β) responsiveness in cultured cells can be modulated by TGF-β partitioning between lipid raft/caveolae- and clathrin-mediated endocytosis pathways. Lipid rafts are plasma membrane microdomains with an important role in cell survival signaling, and cholesterol is necessary for the lipid rafts’ structure and function. Euphol is a euphane-type triterpene alcohol that is structurally similar to cholesterol and has a wide range of pharmacological properties, including anti-inflammatory and anti-cancer effects. In the present study, euphol suppressed TGF-β signaling by inducing TGF-β receptor movement into lipid-raft microdomains and degrading TGF-β receptors.
Collapse
|
289
|
André PA, Prêle CM, Vierkotten S, Carnesecchi S, Donati Y, Chambers RC, Pache JC, Crestani B, Barazzone-Argiroffo C, Königshoff M, Laurent GJ, Irminger-Finger I. BARD1 mediates TGF-β signaling in pulmonary fibrosis. Respir Res 2015; 16:118. [PMID: 26415510 PMCID: PMC4587901 DOI: 10.1186/s12931-015-0278-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 09/17/2015] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a rapid progressive fibro-proliferative disorder with poor prognosis similar to lung cancer. The pathogenesis of IPF is uncertain, but loss of epithelial cells and fibroblast proliferation are thought to be central processes. Previous reports have shown that BARD1 expression is upregulated in response to hypoxia and associated with TGF-β signaling, both recognized factors driving lung fibrosis. Differentially spliced BARD1 isoforms, in particular BARD1β, are oncogenic drivers of proliferation in cancers of various origins. We therefore hypothesized that BARD1 and/or its isoforms might play a role in lung fibrosis. METHODS We investigated BARD1 expression as a function of TGF-β in cultured cells, in mice with experimentally induced lung fibrosis, and in lung biopsies from pulmonary fibrosis patients. RESULTS FL BARD1 and BARD1β were upregulated in response to TGF-β in epithelial cells and fibroblasts in vitro and in vivo. Protein and mRNA expression studies showed very low expression in healthy lung tissues, but upregulated expression of full length (FL) BARD1 and BARD1β in fibrotic tissues. CONCLUSION Our data suggest that FL BARD1 and BARD1β might be mediators of pleiotropic effects of TGF-β. In particular BARD1β might be a driver of proliferation and of pulmonary fibrosis pathogenesis and progression and represent a target for treatment.
Collapse
Affiliation(s)
- Pierre-Alain André
- Molecular Gynecology and Obstetrics Laboratory, Department of Gynecology and Obstetrics, Geneva University Hospitals, Geneva, Switzerland. .,Department of Genetic and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland.
| | - Cecilia M Prêle
- Institute for Respiratory Health, University of Western Australia, Nedlands, WA, Australia. .,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia, Harry Perkins Institute of Medical Research, Nedlands, WA, Australia.
| | - Sarah Vierkotten
- Comprehensive Pneumology Center Ludwig Maximilians University, University Hospital Grosshadern and Helmholtz Zentrum München, Munich, Germany.
| | - Stéphanie Carnesecchi
- Department of Pediatrics and Pathology/Immunology, University of Geneva, Geneva, Switzerland.
| | - Yves Donati
- Department of Pediatrics and Pathology/Immunology, University of Geneva, Geneva, Switzerland.
| | - Rachel C Chambers
- Centre for Inflammation and Tissue Repair, University College London, London, UK.
| | - Jean-Claude Pache
- Department of Clinical Pathology, Geneva University Hospitals, Geneva, Switzerland.
| | - Bruno Crestani
- INSERM, Unité 1152, University of Paris Diderot and Hopital Bichat, Paris, France.
| | | | - Melanie Königshoff
- Comprehensive Pneumology Center Ludwig Maximilians University, University Hospital Grosshadern and Helmholtz Zentrum München, Munich, Germany.
| | - Geoffrey J Laurent
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia, Harry Perkins Institute of Medical Research, Nedlands, WA, Australia.
| | - Irmgard Irminger-Finger
- Molecular Gynecology and Obstetrics Laboratory, Department of Gynecology and Obstetrics, Geneva University Hospitals, Geneva, Switzerland. .,Department of Genetic and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland. .,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia, Harry Perkins Institute of Medical Research, Nedlands, WA, Australia.
| |
Collapse
|
290
|
Jung SY, Yi JY, Kim MH, Song KH, Kang SM, Ahn J, Hwang SG, Nam KY, Song JY. IM-412 inhibits the invasion of human breast carcinoma cells by blocking FGFR-mediated signaling. Oncol Rep 2015; 34:2731-7. [PMID: 26351897 DOI: 10.3892/or.2015.4249] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 07/29/2015] [Indexed: 11/05/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive cancer with a poor prognosis due to its epithelial‑to-mesenchymal transition (EMT) phenotype. Cancer patients often experience several detrimental effects of cancer treatment, such as chemoresistance, radioresistance and the maintenance of cancer stem cells due to EMT. Thus, EMT signaling is considered to be a valuable therapeutic target for cancer treatment, and its inhibition is being attempted as a new treatment option for TNBC patients. Previously, we showed that 3-(2-chlorobenzyl)-1,7-dimethyl-1H-imidazo[2,1-f]purine‑2,4(3H,8H)-dione (IM-412) inhibits transforming growth factor-β (TGF-β)-induced differentiation of human lung fibroblasts through both Smad-dependent and -independent pathways. In the present study, we examined the inhibitory effect of IM-412 on EMT pathways and invasiveness in TNBC cells since the TGF-β signaling pathway is a typical signaling pathway that functions in EMT. IM-412 not only potently suppressed the migration and invasion of MDA-MB-231 cells, but also lowered the expression of mesenchymal markers and EMT-activating transcription factors in these cells. IM-412 inhibited the activation of several signaling proteins, including Smad2/Smad3, p38MAPK, Akt and JNK, and it also attenuated the phosphorylation of FGFR1 and FGFR3. Collectively, our findings suggest that the synthetic compound IM-412 suppressed the EMT process in MDA-MB-231 cells and thereby effectively inhibited the migration and invasion of these cancer cells. Thus, IM-412 could serve as a novel therapeutic agent for malignant cancers.
Collapse
Affiliation(s)
- Seung-Youn Jung
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Jae Youn Yi
- Division of Radiation Effect, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Mi-Hyoung Kim
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Kyung-Hee Song
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Seong-Mook Kang
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Jiyeon Ahn
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Sang-Gu Hwang
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Ky-Youb Nam
- Center for Development and Commercialization of Anti-Cancer Therapeutics, Asan Medical Center, Seoul, Republic of Korea
| | - Jie-Young Song
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| |
Collapse
|
291
|
TGF-β Negatively Regulates CXCL1 Chemokine Expression in Mammary Fibroblasts through Enhancement of Smad2/3 and Suppression of HGF/c-Met Signaling Mechanisms. PLoS One 2015; 10:e0135063. [PMID: 26252654 PMCID: PMC4529193 DOI: 10.1371/journal.pone.0135063] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 07/17/2015] [Indexed: 01/23/2023] Open
Abstract
Fibroblasts are major cellular components of the breast cancer stroma, and influence the growth, survival and invasion of epithelial cells. Compared to normal tissue fibroblasts, carcinoma associated fibroblasts (CAFs) show increased expression of numerous soluble factors including growth factors and cytokines. However, the mechanisms regulating expression of these factors remain poorly understood. Recent studies have shown that breast CAFs overexpress the chemokine CXCL1, a key regulator of tumor invasion and chemo-resistance. Increased expression of CXCL1 in CAFs correlated with poor patient prognosis, and was associated with decreased expression of TGF-β signaling components. The goal of these studies was to understand the role of TGF-β in regulating CXCL1 expression in CAFs, using cell culture and biochemical approaches. We found that TGF-β treatment decreased CXCL1 expression in CAFs, through Smad2/3 dependent mechanisms. Chromatin immunoprecipitation and site-directed mutagenesis assays revealed two new binding sites in the CXCL1 promoter important for Smad2/3 modulation of CXCL1 expression. Smad2/3 proteins also negatively regulated expression of Hepatocyte Growth Factor (HGF), which was found to positively regulate CXCL1 expression in CAFs through c-Met receptor dependent mechanisms. HGF/c-Met signaling in CAFs was required for activity of NF-κB, a transcriptional activator of CXCL1 expression. These studies indicate that TGF-β negatively regulates CXCL1 expression in CAFs through Smad2/3 binding to the promoter, and through suppression of HGF/c-Met autocrine signaling. These studies reveal novel insight into how TGF-β and HGF, key tumor promoting factors modulate CXCL1 chemokine expression in CAFs.
Collapse
|
292
|
Cohen-Solal KA, Boregowda RK, Lasfar A. RUNX2 and the PI3K/AKT axis reciprocal activation as a driving force for tumor progression. Mol Cancer 2015. [PMID: 26204939 PMCID: PMC4513933 DOI: 10.1186/s12943-015-0404-3] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
From the first reported role of the transcription factor RUNX2 in osteoblast and chondrocyte differentiation and migration to its involvement in promigratory/proinvasive behavior of breast, prostate, and thyroid cancer cells, osteosarcoma, or melanoma cells, RUNX2 currently emerges as a key player in metastasis. In this review, we address the interaction of RUNX2 with the PI3K/AKT signaling pathway, one of the critical axes controlling cancer growth and metastasis. AKT, either by directly phosphorylating/activating RUNX2 or phosphorylating/inactivating regulators of RUNX2 stability or activity, contributes to RUNX2 transcriptional activity. Reciprocally, the activation of the PI3K/AKT pathway by RUNX2 regulation of its different components has been described in non-transformed and transformed cells. This mutual activation in the context of cancer cells exhibiting constitutive AKT activation and high levels of RUNX2 might constitute a major driving force in tumor progression and aggressiveness.
Collapse
Affiliation(s)
- Karine A Cohen-Solal
- Rutgers Cancer Institute of New Jersey, Department of Medicine, Division of Medical Oncology - Rutgers, the State University of New Jersey, Robert Wood Johnson Medical School, 195 Little Albany Street, New Brunswick, New Jersey, 08903, USA.
| | - Rajeev K Boregowda
- Rutgers Cancer Institute of New Jersey, Department of Medicine, Division of Medical Oncology - Rutgers, the State University of New Jersey, Robert Wood Johnson Medical School, 195 Little Albany Street, New Brunswick, New Jersey, 08903, USA
| | - Ahmed Lasfar
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, 08854, USA.,Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, 08903, USA
| |
Collapse
|
293
|
Hu ZY, Xie WB, Yang F, Xiao LW, Wang XY, Chen SY, Li ZG. NDRG1 attenuates epithelial-mesenchymal transition of nasopharyngeal cancer cells via blocking Smad2 signaling. Biochim Biophys Acta Mol Basis Dis 2015; 1852:1876-86. [PMID: 26071641 DOI: 10.1016/j.bbadis.2015.06.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Revised: 05/26/2015] [Accepted: 06/08/2015] [Indexed: 12/12/2022]
Abstract
N-myc downstream-regulated gene 1 (NDRG1) has been implicated in tumorigenesis and metastasis in different cancers. However, its role in nasopharyngeal carcinoma remains unknown. We found that NDRG1 expression level was high in nasopharyngeal cancer 5-8F cells but low in 5-8F-LN cells with lymphatic metastasis potential. Knockdown of NDRG1 by shRNA promoted 5-8F cell proliferation, migration, and invasion in vitro and its tumorigenesis in vivo. Moreover, NDRG1 deficiency induced an epithelial-mesenchymal transition (EMT) of 5-8F cells as shown by an attenuation of E-cadherin and an induction of N-cadherin and vimentin expression. NDRG1 knockdown also enhanced Smad2 expression and phosphorylation. Smad2 signaling was attenuated in 5-8F cells but was significantly activated in 5-8F-LN cells. Knockdown of Smad2 restored E-cadherin but attenuated N-cadherin expression in NDRG1-deficient 5-8F cells, suggesting a reduction of EMT. Consistently, blockade of Smad2 in 5-8F-LN cells increased E-cadherin while diminishing N-cadherin and vimentin expression. These data indicate that Smad2 mediates the NDRG1 deficiency-induced EMT of 5-8F cells. In tumors derived from NDRG1-deficient 5-8F cells, E-cadherin expression was inhibited while vimentin and Smad2 were increased in a large number of cancer cells. Most importantly, NDRG1 expression was attenuated in human nasopharyngeal carcinoma tissues, resulted in a lower survival rate in patients. The NDRG1 was further decreased in the detached nasopharyngeal cancer cells, which was associated with a further reduced survival rate in patients with lymphatic metastasis. Taken together, these results demonstrated that NDRG1 prevents nasopharyngeal tumorigenesis and metastasis via inhibiting Smad2-mediated EMT of nasopharyngeal cells.
Collapse
Affiliation(s)
- Zhi-Yan Hu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Wei-Bing Xie
- Department of Forensic Science, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Fang Yang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Li-Wei Xiao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiao-Yan Wang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shi-You Chen
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA, United States.
| | - Zu-Guo Li
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
294
|
Ghahhari NM, Babashah S. Interplay between microRNAs and WNT/β-catenin signalling pathway regulates epithelial-mesenchymal transition in cancer. Eur J Cancer 2015; 51:1638-49. [PMID: 26025765 DOI: 10.1016/j.ejca.2015.04.021] [Citation(s) in RCA: 193] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 04/29/2015] [Accepted: 04/30/2015] [Indexed: 12/16/2022]
Abstract
The WNT/β-catenin signalling implies its significance in maintaining an epithelial cell phenotype, proper cell-cell junctions, and tissue homeostasis. Dysregulation of the members of this pathway involves in the development of cancer and an epithelial-mesenchymal transition (EMT) required for metastasis. Loss of E-cadherin is the major contributor to an EMT process and is largely influenced by the WNT/β-catenin signalling. An E-cadherin/β-catenin complex maintains epithelial integrity and disturbance of this complex and WNT/β-catenin pathway will ultimately lead to the nuclear translocation of β-catenin and transcription of EMT-promoting genes. WNT/β-catenin signalling is controlled by microRNAs (miRNAs), several of which are either up- or downregulated during EMT. The strong association between the expression of the WNT signalling components with miRNAs in the initiation and achievement of an EMT phenotype is suggestive of introducing these miRNAs as therapeutic targets against metastatic tumours. Therefore, this review aims to describe these putative miRNAs in altering the WNT/β-catenin signalling in EMT, and whether targeting them is a useful therapeutic option for human invasive tumours.
Collapse
Affiliation(s)
| | - Sadegh Babashah
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
295
|
Li L, Cao F, Liu B, Luo X, Ma X, Hu Z. TGF-β induces fascin expression in gastric cancer via phosphorylation of smad3 linker area. Am J Cancer Res 2015; 5:1890-1896. [PMID: 26269751 PMCID: PMC4529611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 05/10/2015] [Indexed: 06/04/2023] Open
Abstract
BACKGROUND Fascin is an actin-bundling protein critical for tumor invasion. TGF-β could induce fascin expression in gastric cancer cells. In this study, we attempted to explore the role of p-smad3L in the expression of fascin induced by TGF-β in gastric cancer cells. METHODS Pseudopodia were evaluated by immunofluorescence. Fascin expression was detected by RT-PCR and western blot. Smad3 siRNA was used to repress the endogenous smad3. The phosphorylations of smad3 linker region at sites s204, s208 and s213 were detected by western blot. The fascin promoter reporter activity was measured by dual luciferase assay. RESULTS TGF-β could increase the formation of pseudopodia and the expression of fascin in gastric cancer cells. Smad3 depletion abrogated the expression of fascin induced by TGF-β. The phosphorylation of smad3 linker region at serine 204, 208 and 213 was enhanced in gastric cancer cells after TGF-β treatment. The fascin promoter reporter activity was significantly enhanced with TGF-β treatment in both wild-type Smad3 group and Smad3EPSM group (P<0.05). Furthermore, the fascin promoter reporter activity in the wild-type Smad3 transfectant cells was significantly higher than that in Smad3EPSM cells (P<0.05). CONCLUSIONS fascin expression induced by TGF-β depends on smad3, at least in part, depends on smad3 linker phosphorylation.
Collapse
Affiliation(s)
- Liling Li
- Department of Pathology, Xiangya Hospital, Central South UniversityChangsha, China
- Department of Pathology, Xiangya Medical School, Central South UniversityChangsha, China
| | - Fang Cao
- Department of Pathology, Xiangya Hospital, Central South UniversityChangsha, China
- Department of Pathology, Xiangya Medical School, Central South UniversityChangsha, China
| | - Baoan Liu
- Department of Pathology, Xiangya Hospital, Central South UniversityChangsha, China
- Department of Pathology, Xiangya Medical School, Central South UniversityChangsha, China
| | - Xiaojuan Luo
- Department of Pathology, Xiangya Hospital, Central South UniversityChangsha, China
- Department of Pathology, Xiangya Medical School, Central South UniversityChangsha, China
| | - Xin Ma
- Department of Pathology, Xiangya Hospital, Central South UniversityChangsha, China
- Department of Pathology, Xiangya Medical School, Central South UniversityChangsha, China
| | - Zhongliang Hu
- Department of Pathology, Xiangya Hospital, Central South UniversityChangsha, China
- Department of Pathology, Xiangya Medical School, Central South UniversityChangsha, China
| |
Collapse
|
296
|
Lin X, Xu W, Shao M, Fan Q, Wen G, Li C, Jing L, Sun X. Shenling Baizhu San supresses colitis associated colorectal cancer through inhibition of epithelial-mesenchymal transition and myeloid-derived suppressor infiltration. Altern Ther Health Med 2015; 15:126. [PMID: 25897964 PMCID: PMC4428101 DOI: 10.1186/s12906-015-0649-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 04/15/2015] [Indexed: 12/11/2022]
Abstract
Background Shenling Baizhu San (SBS) is a well-known and classical Chinese medicine formula. It has been used for treatment of gastrointestinal disorders for about nine hundred years. Recent reports showed that it was effective in curing colitis and ameliorating the major manifestations of postoperational colorectal cancer (CRC). This study was to evaluate the effects of SBS on azoxymethane (AOM) and dextran sodium sulfate (DSS) induced colitis associated CRC (caCRC) and to analyze the underlying mechanism of SBS in preventing CRC. Methods The colon tissue of mice in different group was determined by immunohistochemistry and western blot. TGF-β1 in serum was measured by ELISA. Myeloid-derived suppressor cells (MDSCs) were identified by flow cytometry and immunohistochemistry. Results The formed neoplasms phenotypically resembled human caCRC with upregulated β-catenin, p53 and proliferating cell nuclear antigen (PCNA). SBS treatment reduced the death rate of mice and decreased the incidence and multiplicity of colonic neoplasms. SBS decreased the number of MDSCs and the level of transforming growth factor β1 (TGF-β1). SBS alleviated epithelial mesenchymal transition (EMT) through downregulating N-cadherin (N-cad), Vimentin, Fibronectin, Snail, and upregulating E-cadherin (E-cad). It reduced the activation of Wnt5a and EMT induced by TGF-β1. Conclusions SBS reduced the death rate through decreasing the incidence and multiplicity of colonic tumors. SBS lowered MDSCs infiltration and inhibited TGF-β1 induced EMT to exert its anti-caCRC effects. Electronic supplementary material The online version of this article (doi:10.1186/s12906-015-0649-9) contains supplementary material, which is available to authorized users.
Collapse
|
297
|
Correlation between Tgf-Β1 and Fsp-1 Expression in Chronic Viral Hepatitis - an Immunohistochemical Study. CURRENT HEALTH SCIENCES JOURNAL 2015; 41:179-185. [PMID: 30364789 PMCID: PMC6201209 DOI: 10.12865/chsj.41.02.14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 04/01/2015] [Indexed: 12/15/2022]
Abstract
Infection with hepatitis C virus (HCV) is the most important stimulus for chronic hepatitis and subsequent progression to cirrhosis and hepatocellular carcinoma. Fibrosis that follows inflammation represents the main complication. One of the mechanisms that could be associated with development of liver fibrosis is epithelial-mesenchymal transition (EMT). Transforming Growth Factor β1 (TGF-β1) is an important mediator of fibrosis and also able to trigger phenotypic changes in EMT. Fibroblast-specific protein 1 (FSP-1), a marker of fibroblasts in organs undergoing tissue remodeling, is used to identify cells that derive from EMT. In this study, we assessed the expression of TGF-β1 and FSP-1 in liver biopsies obtained from HCV-infected patients using immunohistochemistry and correlated them in order to evaluate the relation between fibrosis and EMT in liver disease progression. Staining of liver sections revealed increased amount of type III collagen and clusters of inflammatory cells invading portal spaces. The number of TGF-β1-positive cells was directly proportional to the incidence of liver injury. In cases of mild fibrosis, FSP-1 positive cells were observed in cells lining sinusoids. As fibrosis progressed, increased number of FSP-1 positive fibroblasts, isolated cholangiocytes and hepatocytes was observed. Even EMT via the activation of TGF-β signaling pathway is recognized as a pathogenic mechanism of HCV-induced liver disease, FSP-1 alone couldn’t be used as a valuable marker for cells that undergo EMT.
Collapse
|
298
|
Kong Y, Liang Y, Wang J. Foci of Entotic Nuclei in Different Grades of Noninherited Renal Cell Cancers. IUBMB Life 2015; 67:139-44. [PMID: 25855323 DOI: 10.1002/iub.1354] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 01/15/2015] [Indexed: 01/09/2023]
Abstract
We report here an intriguing pattern in nuclear appearance of renal clear cell cancer. In low grade clear cell cancer, detailed examination showed that in many cells, two or more nuclei were within the confines of a single cell membrane. This likely resulted from a cell being contained within its neighboring cell. Consequently, this resulted in appearance of multicellularity. This appearance of the nuclei were not associated with mitotic figures, suggesting that these did not result from nuclear fission. Additionally, the cells containing this nuclei did not show any evidence of cytokinesis including equatorial tapering, suggesting that the process may have resulted from cytokinesis failure. In some sections of higher grade clear cell cancer, these appearance were higher, though we did not observe any frank syncytium formation. On careful observation, there were isolated events of fusion of nuclei within a single cell in different grades of renal cell cancers. There occurrence was more frequent in higher grades of clear cell renal cancer and metastatic clear cell carcinoma. These features were also demonstrable in multiple fields of lower grades of clear cell carcinoma. This phenomenon of entosis may contribute to aneuploidy and tumor progression to dysplastic stages and genomic instability in renal cancers. Future studies are aimed at delineating the cell-cell boundaries and the mechanism contributing to this observation, either from peripheral cell engulfing or failure of cytosolic division for cell separation.
Collapse
Affiliation(s)
- Yuke Kong
- Department of Nephrology, Lanzhou University Second Hospital, Lanzhou, China
| | | | | |
Collapse
|
299
|
Abstract
The mechanism by which reactive oxygen species (ROS) are produced by tumour cells remained incompletely understood until the discovery over the last 15 years of the family of NADPH oxidases (NOXs 1–5 and dual oxidases DUOX1/2) which are structural homologues of gp91phox, the major membrane-bound component of the respiratory burst oxidase of leucocytes. Knowledge of the roles of the NOX isoforms in cancer is rapidly expanding. Recent evidence suggests that both NOX1 and DUOX2 species produce ROS in the gastrointestinal tract as a result of chronic inflammatory stress; cytokine induction (by interferon-γ, tumour necrosis factor α, and interleukins IL-4 and IL-13) of NOX1 and DUOX2 may contribute to the development of colorectal and pancreatic carcinomas in patients with inflammatory bowel disease and chronic pancreatitis, respectively. NOX4 expression is increased in pre-malignant fibrotic states which may lead to carcinomas of the lung and liver. NOX5 is highly expressed in malignant melanomas, prostate cancer and Barrett's oesophagus-associated adenocarcinomas, and in the last it is related to chronic gastro-oesophageal reflux and inflammation. Over-expression of functional NOX proteins in many tissues helps to explain tissue injury and DNA damage from ROS that accompany pre-malignant conditions, as well as elucidating the potential mechanisms of NOX-related damage that contribute to both the initiation and the progression of a wide range of solid and haematopoietic malignancies.
Collapse
|
300
|
Thien A, Prentzell MT, Holzwarth B, Kläsener K, Kuper I, Boehlke C, Sonntag AG, Ruf S, Maerz L, Nitschke R, Grellscheid SN, Reth M, Walz G, Baumeister R, Neumann-Haefelin E, Thedieck K. TSC1 activates TGF-β-Smad2/3 signaling in growth arrest and epithelial-to-mesenchymal transition. Dev Cell 2015; 32:617-30. [PMID: 25727005 DOI: 10.1016/j.devcel.2015.01.026] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 12/19/2014] [Accepted: 01/22/2015] [Indexed: 11/27/2022]
Abstract
The tuberous sclerosis proteins TSC1 and TSC2 are key integrators of growth factor signaling. They suppress cell growth and proliferation by acting in a heteromeric complex to inhibit the mammalian target of rapamycin complex 1 (mTORC1). In this study, we identify TSC1 as a component of the transforming growth factor β (TGF-β)-Smad2/3 pathway. Here, TSC1 functions independently of TSC2. TSC1 interacts with the TGF-β receptor complex and Smad2/3 and is required for their association with one another. TSC1 regulates TGF-β-induced Smad2/3 phosphorylation and target gene expression and controls TGF-β-induced growth arrest and epithelial-to-mesenchymal transition (EMT). Hyperactive Akt specifically activates TSC1-dependent cytostatic Smad signaling to induce growth arrest. Thus, TSC1 couples Akt activity to TGF-β-Smad2/3 signaling. This has implications for cancer treatments targeting phosphoinositide 3-kinases and Akt because they may impair tumor-suppressive cytostatic TGF-β signaling by inhibiting Akt- and TSC1-dependent Smad activation.
Collapse
Affiliation(s)
- Antje Thien
- Bioinformatics and Molecular Genetics (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Renal Division, University Hospital Freiburg, 79106 Freiburg, Germany
| | - Mirja Tamara Prentzell
- Bioinformatics and Molecular Genetics (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, the Netherlands
| | - Birgit Holzwarth
- Bioinformatics and Molecular Genetics (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
| | - Kathrin Kläsener
- Molecular Immunology (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Molecular Immunology, Max-Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
| | - Ineke Kuper
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, the Netherlands; Department for Neuroscience, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
| | | | - Annika G Sonntag
- Bioinformatics and Molecular Genetics (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
| | - Stefanie Ruf
- Bioinformatics and Molecular Genetics (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, the Netherlands; BIOSS Centre for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Research Training Group (RTG) 1104, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
| | - Lars Maerz
- Bioinformatics and Molecular Genetics (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
| | - Roland Nitschke
- BIOSS Centre for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Center for Biological Systems Analysis (ZBSA), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
| | | | - Michael Reth
- Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Molecular Immunology (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Molecular Immunology, Max-Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
| | - Gerd Walz
- Renal Division, University Hospital Freiburg, 79106 Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Center for Biological Systems Analysis (ZBSA), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
| | - Ralf Baumeister
- Bioinformatics and Molecular Genetics (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Research Training Group (RTG) 1104, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Center for Biological Systems Analysis (ZBSA), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; ZBMZ Centre for Biochemistry and Molecular Cell Research (Faculty of Medicine), Albert-Ludwigs-University Freiburg, 79106 Freiburg, Germany
| | | | - Kathrin Thedieck
- Bioinformatics and Molecular Genetics (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, the Netherlands; BIOSS Centre for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Department for Neuroscience, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany.
| |
Collapse
|