1
|
Goel HL, Karner ER, Kumar A, Mukhopadhyay D, Goel S, Mercurio AM. YAP/TAZ-mediated regulation of laminin 332 is enabled by β4 integrin repression of ZEB1 to promote ferroptosis resistance. J Biol Chem 2024; 300:107202. [PMID: 38508310 PMCID: PMC11017052 DOI: 10.1016/j.jbc.2024.107202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/10/2024] [Accepted: 03/13/2024] [Indexed: 03/22/2024] Open
Abstract
We are interested in the contribution of integrins and the extracellular matrix to epithelial differentiation in carcinomas. This study was motivated by our finding that the Hippo effectors YAP and TAZ can sustain the expression of laminin 332 (LM332), the predominant ECM ligand for the integrin β4, in breast carcinoma cells with epithelial differentiation. More specifically, we observed that YAP and TAZ regulate the transcription of the LAMC2 subunit of LM332. Given that the β4-LM332 axis is associated with epithelial differentiation and YAP/TAZ have been implicated in carcinoma de-differentiation, we sought to resolve this paradox. Here, we observed that the β4 integrin sustains the expression of miR-200s that target the transcription factor ZEB1 and that ZEB1 has a pivotal role in determining the nature of YAP/TAZ-mediated transcription. In the presence of β4, ZEB1 expression is repressed enabling YAP/TAZ/TEAD-mediated transcription of LAMC2. The absence of β4, however, induces ZEB1, and ZEB1 binds to the LAMC2 promoter to inhibit LAMC2 transcription. YAP/TAZ-mediated regulation of LAMC2 has important functional consequences because we provide evidence that LM332 enables carcinoma cells to resist ferroptosis in concert with the β4 integrin.
Collapse
Affiliation(s)
- Hira Lal Goel
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Emmet R Karner
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Ayush Kumar
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Dimpi Mukhopadhyay
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Shivam Goel
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Arthur M Mercurio
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA.
| |
Collapse
|
2
|
García de Herreros A. Dual role of Snail1 as transcriptional repressor and activator. Biochim Biophys Acta Rev Cancer 2024; 1879:189037. [PMID: 38043804 DOI: 10.1016/j.bbcan.2023.189037] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/27/2023] [Accepted: 11/27/2023] [Indexed: 12/05/2023]
Abstract
Snail1 transcriptional factor plays a key role in the control of epithelial to mesenchymal transition, a process that remodels tumor cells increasing their invasion and chemo-resistance as well as reprograms their metabolism and provides stemness properties. During this transition, Snail1 acts as a transcriptional repressor and, as growing evidences have demonstrated, also as a direct activator of mesenchymal genes. In this review, I describe the different proteins that interact with Snail1 and are responsible for these two different functions on gene expression; I focus on the transcriptional factors that associate to Snail1 in their target promoters, both activated and repressed. I also present working models for Snail1 action both as repressor and activator and raise some issues that still need to be investigated.
Collapse
Affiliation(s)
- Antonio García de Herreros
- Programa de Recerca en Càncer, Hospital del Mar Research Institute (IMIM), Unidad Asociada al CSIC, Barcelona, Spain; Departament de Medicina i Ciències de la Vida, Universitat Pompeu Fabra, Barcelona, Spain.
| |
Collapse
|
3
|
Valakos D, Klagkou E, Kokkalis A, Polyzos A, Kyrilis FL, Banos A, Vatsellas G, Pliatska M, Ford E, Stravopodis DJ, Thanos D. Combinatorial targeting of a specific EMT/MET network by macroH2A variants safeguards mesenchymal identity. PLoS One 2023; 18:e0288005. [PMID: 37432970 DOI: 10.1371/journal.pone.0288005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 06/16/2023] [Indexed: 07/13/2023] Open
Abstract
Generation of induced pluripotent stem cells from specialized cell types provides an excellent model to study how cells maintain their stability, and how they can change identity, especially in the context of disease. Previous studies have shown that chromatin safeguards cell identity by acting as a barrier to reprogramming. We investigated mechanisms by which the histone macroH2A variants inhibit reprogramming and discovered that they work as gate keepers of the mesenchymal cell state by blocking epithelial transition, a step required for reprogramming of mouse fibroblasts. More specifically, we found that individual macroH2A variants regulate the expression of defined sets of genes, whose overall function is to stabilize the mesenchymal gene expression program, thus resisting reprogramming. We identified a novel gene network (MSCN, mesenchymal network) composed of 63 macroH2A-regulated genes related to extracellular matrix, cell membrane, signaling and the transcriptional regulators Id2 and Snai2, all of which function as guardians of the mesenchymal phenotype. ChIP-seq and KD experiments revealed a macroH2A variant-specific combinatorial targeting of the genes reconstructing the MSCN, thus generating robustness in gene expression programs to resist cellular reprogramming.
Collapse
Affiliation(s)
- Dimitrios Valakos
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
- Section of Biochemistry and Molecular Biology, Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), Zografou, Athens, Greece
| | - Eleftheria Klagkou
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
- Section of Biochemistry and Molecular Biology, Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), Zografou, Athens, Greece
| | - Antonis Kokkalis
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | | | - Fotis L Kyrilis
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Aggelos Banos
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | | | - Maria Pliatska
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Ethan Ford
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Dimitrios J Stravopodis
- Section of Cell Biology and Biophysics, Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), Zografou, Athens, Greece
| | - Dimitris Thanos
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
| |
Collapse
|
4
|
Xiao Y, Liang D, Li Z, Feng Z, Yuan Z, Zhang F, Wang Y, Zhou Y, Shi M, Liu L, Xiao Y, Guo B. BMP-7 Upregulates Id2 Through the MAPK Signaling Pathway to Improve Diabetic Tubulointerstitial Fibrosis and the Intervention of Oxymatrine. Front Pharmacol 2022; 13:900346. [PMID: 35721136 PMCID: PMC9201778 DOI: 10.3389/fphar.2022.900346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 05/13/2022] [Indexed: 12/05/2022] Open
Abstract
Diabetic kidney disease is one of the most serious microvascular complications of diabetes. It progresses irreversibly to end-stage renal disease if left untreated. Bone morphogenetic protein (BMP)-7 is a negative regulator of organ fibrosis and may also play an essential role in tubulointerstitial fibrosis. This study aimed to investigate the precise role and potential molecular mechanisms of BMP-7 in the progression of diabetic nephropathy. In this study, BMP-7 was overexpressed in vivo after the replication of the diabetic rat model using streptozotocin. The results showed that BMP-7 inhibited the phosphorylation of related mitogen-activated protein kinase (MAPK) pathways while upregulating the inhibitor of differentiation (Id2) expression and effectively ameliorated pathological renal injury. Further in vitro validation showed that the inhibition of the phosphorylation of MAPKs at a high glucose concentration in renal tubular epithelial cells was followed by the upregulation of Id2 protein expression, suggesting that BMP-7 could improve diabetic nephropathy by upregulating Id2 protein levels through the BMP-7–MAPK signaling pathway. Previous laboratory studies found that oxymatrine improved renal fibrotic lesions. However, the exact mechanism is unclear. The present study showed that oxymatrine treatment in a diabetic rat model upregulated BMP-7 protein expression and inhibited MAPK pathway protein phosphorylation levels. These results suggested that oxymatrine improved the epithelial-to-mesenchymal transition process in the early stage of diabetic kidney disease by regulating the BMP-7–MAPK pathway and ameliorated renal tubulointerstitial fibrosis.
Collapse
Affiliation(s)
- Yawen Xiao
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guizhou, China.,Department of Pathophysiology, Guizhou Medical University, Guizhou, China
| | - Dan Liang
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guizhou, China.,Department of Pathophysiology, Guizhou Medical University, Guizhou, China
| | - Zhiyang Li
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guizhou, China.,Department of Pathophysiology, Guizhou Medical University, Guizhou, China
| | - Zhaowei Feng
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guizhou, China.,Department of Pathophysiology, Guizhou Medical University, Guizhou, China
| | - Zhiping Yuan
- School Hospital, Guizhou Medical University, Guiyang, China
| | - Fan Zhang
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guizhou, China.,Department of Pathophysiology, Guizhou Medical University, Guizhou, China
| | - Yuanyuan Wang
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guizhou, China.,Department of Pathophysiology, Guizhou Medical University, Guizhou, China
| | - Yuxia Zhou
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guizhou, China.,Department of Pathophysiology, Guizhou Medical University, Guizhou, China
| | - Mingjun Shi
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guizhou, China.,Department of Pathophysiology, Guizhou Medical University, Guizhou, China
| | - Lingling Liu
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guizhou, China.,Department of Pathophysiology, Guizhou Medical University, Guizhou, China
| | - Ying Xiao
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guizhou, China.,Department of Pathophysiology, Guizhou Medical University, Guizhou, China
| | - Bing Guo
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guizhou, China.,Department of Pathophysiology, Guizhou Medical University, Guizhou, China
| |
Collapse
|
5
|
Yang H, Xu Z, Peng Y, Wang J, Xiang Y. Integrin β4 as a Potential Diagnostic and Therapeutic Tumor Marker. Biomolecules 2021; 11:biom11081197. [PMID: 34439865 PMCID: PMC8394641 DOI: 10.3390/biom11081197] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/09/2021] [Accepted: 08/11/2021] [Indexed: 11/05/2022] Open
Abstract
Integrin β4 (ITGβ4) is a class of transmembrane adhesion molecules composed of hemidesmosomes (HDs). Its unique long intracellular domain provides intricate signal transduction functions. These signal transduction effects are especially prominent in tumors. Many recent studies have shown that integrin β4 is differentially expressed in various tumors, and it plays a vital role in tumor invasion, proliferation, epithelial–mesenchymal transition, and angiogenesis. Therefore, we categorize the research related to integrin β4, starting from its structure and function in tumor tissues, and provide a basic description. Based on its structure and function, we believe that integrin β4 can be used as a tumor marker. In clinical practice, it is described as a diagnostic marker for the targeted treatment of cancer and will be helpful in the clinical diagnosis and treatment of tumors.
Collapse
Affiliation(s)
- Haoyu Yang
- School of Basic Medical Science, Central South University, Changsha 410013, China; (H.Y.); (Z.X.); (Y.P.)
| | - Zixuan Xu
- School of Basic Medical Science, Central South University, Changsha 410013, China; (H.Y.); (Z.X.); (Y.P.)
| | - Yuqian Peng
- School of Basic Medical Science, Central South University, Changsha 410013, China; (H.Y.); (Z.X.); (Y.P.)
| | - Jiali Wang
- Xiang Ya School of Medicine, Central South University, Changsha 410013, China;
| | - Yang Xiang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410013, China
- Correspondence: ; Tel.:+86-139-7312-8943
| |
Collapse
|
6
|
Dolmatov IY. Molecular Aspects of Regeneration Mechanisms in Holothurians. Genes (Basel) 2021; 12:250. [PMID: 33578707 PMCID: PMC7916379 DOI: 10.3390/genes12020250] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 02/07/2023] Open
Abstract
Holothurians, or sea cucumbers, belong to the phylum Echinodermata. They show good regenerative abilities. The present review provides an analysis of available data on the molecular aspects of regeneration mechanisms in holothurians. The genes and signaling pathways activated during the asexual reproduction and the formation of the anterior and posterior parts of the body, as well as the molecular mechanisms that provide regeneration of the nervous and digestive systems, are considered here. Damage causes a strong stress response, the signs of which are recorded even at late regeneration stages. In holothurian tissues, the concentrations of reactive oxygen species and antioxidant enzymes increase. Furthermore, the cellular and humoral components of the immune system are activated. Extracellular matrix remodeling and Wnt signaling play a major role in the regeneration in holothurians. All available morphological and molecular data show that the dedifferentiation of specialized cells in the remnant of the organ and the epithelial morphogenesis constitute the basis of regeneration in holothurians. However, depending on the type of damage, the mechanisms of regeneration may differ significantly in the spatial organization of regeneration process, the involvement of different cell types, and the depth of reprogramming of their genome (dedifferentiation or transdifferentiation).
Collapse
Affiliation(s)
- Igor Yu Dolmatov
- A.V. Zhirmunsky National Scientifc Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Palchevsky 17, 690041 Vladivostok, Russia
| |
Collapse
|
7
|
The Eupentacta fraudatrix transcriptome provides insights into regulation of cell transdifferentiation. Sci Rep 2020; 10:1522. [PMID: 32001787 PMCID: PMC6992634 DOI: 10.1038/s41598-020-58470-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 01/15/2020] [Indexed: 12/11/2022] Open
Abstract
The holothurian Eupentacta fraudatrix is a unique organism for studying regeneration mechanisms. Moreover, E. fraudatrix can quickly restore parts of its body and entire organ systems, yet at the moment, there is no data on the participation of stem cells in the process. To the contrary, it has been repeatedly confirmed that this process is only due to the transformation of terminally differentiated cells. In this study, we examine changes in gene expression during gut regeneration of the holothurian E. fraudatrix. Transcriptomes of intestinal anlage of the three stages of regeneration, as well as the normal gut, were sequenced with an Illumina sequencer (San Diego, CA, USA). We identified 14,617 sea urchin protein homologs, of which 308 were transcription factors. After analysing the dynamics of gene expression during regeneration and the map of biological processes in which they participate, we identified 11 factors: Ef-EGR1, Ef-ELF, Ef-GATA3, Ef-ID2, Ef-KLF1/2/4, Ef-MSC, Ef-PCGF2, Ef-PRDM9, Ef-SNAI2, Ef-TBX20, and Ef-TCF24. With the exception of TCF24, they are all involved in the regeneration, development, epithelial-mesenchymal transition, and immune response in other animals. We suggest that these transcription factors may also be involved in the transdifferentiation of coelomic epithelial cells into enterocytes in holothurians.
Collapse
|
8
|
Ke J, Wu R, Chen Y, Abba ML. Inhibitor of DNA binding proteins: implications in human cancer progression and metastasis. Am J Transl Res 2018; 10:3887-3910. [PMID: 30662638 PMCID: PMC6325517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 11/29/2018] [Indexed: 06/09/2023]
Abstract
Inhibitor of DNA binding (ID) proteins are a class of helix-loop-helix (HLH) transcription regulatory factors that act as dominant-negative antagonists of other basic HLH proteins through the formation of non-functional heterodimers. These proteins have been shown to play critical roles in a wide range of tumor-associated processes, including cell differentiation, cell cycle progression, migration and invasion, epithelial-mesenchymal transition, angiogenesis, stemness, chemoresistance, tumorigenesis, and metastasis. The aberrant expression of ID proteins has not only been detected in many types of human cancers, but is also associated with advanced tumor stages and poor clinical outcome. In this review, we provide an overview of the key biological functions of ID proteins including affiliated signaling pathways. We also describe the regulation of ID proteins in cancer progression and metastasis, and elaborate on expression profiles in cancer and the implications for prognosis. Lastly, we outline strategies for the therapeutic targeting of ID proteins as a promising and effective approach for anticancer therapy.
Collapse
Affiliation(s)
- Jing Ke
- Department of Liver Disease, The Fourth Affiliated Hospital of Anhui Medical UniversityHefei 230022, China
- Center for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, University of HeidelbergMannheim 68167, Germany
| | - Ruolin Wu
- Department of Hepatopancreatobiliary Surgery and Organ Transplantation Center, Department of General Surgery, First Affiliated Hospital of Anhui Medical University218 Jixi Avenue, Hefei 230022, Anhui, China
- Center for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, University of HeidelbergMannheim 68167, Germany
| | - Yong Chen
- Department of Medical Oncology, Subei People’s HospitalYangzhou, Jiangsu 225000, China
| | - Mohammed L Abba
- Department of Hematology and Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of HeidelbergMannheim, Germany
| |
Collapse
|
9
|
Liu Z, Yang J, Ge C, Zhao F, Li H, Yao M, Li J, Tian H. Inhibitor of binding/differentiation 2 (Id2) is regulated by CCAAT/enhancer-binding protein-α (C/EBPα) and promotes the proliferation of hepatocellular carcinoma. Am J Cancer Res 2018; 8:2254-2266. [PMID: 30555742 PMCID: PMC6291656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/02/2018] [Indexed: 06/09/2023] Open
Abstract
Inhibitor of DNA binding/differentiation (Id2) is an important regulator involved in the initiation and progression of cancer. However, the function and mechanism of the regulation of Id2 in hepatocellular carcinoma (HCC) was unclear. In the present study, we found that the overexpression of Id2 increased HCC cell proliferation in vitro and in vivo. Knockdown of Id2 inhibited HCC cell proliferation in vitro and in vivo. Furthermore, knockdown of Id2 enhanced sorafenib-induced apoptosis in HCC. Conversely, overexpression of Id2 weakened sorafenib-induced apoptosis in HCC. In addition, the transcription factor CCAAT/enhancer-binding protein alpha (C/EBPα) bound to the Id2 promoter and decreased its expression in HCC cells. Therefore, all results suggest that Id2 promotes the proliferation of HCC cells by inhibiting cell apoptosis. Id2 may serve as a potential target in HCC therapy.
Collapse
Affiliation(s)
- Zheng Liu
- Shanghai Medical College, Fudan UniversityShanghai 200032, PR China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of MedicineShanghai 200032, PR China
| | - Jing Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of MedicineShanghai 200032, PR China
| | - Chao Ge
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of MedicineShanghai 200032, PR China
| | - Fangyu Zhao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of MedicineShanghai 200032, PR China
| | - Hong Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of MedicineShanghai 200032, PR China
| | - Ming Yao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of MedicineShanghai 200032, PR China
| | - Jinjun Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of MedicineShanghai 200032, PR China
| | - Hua Tian
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of MedicineShanghai 200032, PR China
| |
Collapse
|
10
|
Wen XF, Chen M, Wu Y, Chen MN, Glogowska A, Klonisch T, Zhang GJ. Inhibitor of DNA Binding 2 Inhibits Epithelial-Mesenchymal Transition via Up-Regulation of Notch3 in Breast Cancer. Transl Oncol 2018; 11:1259-1270. [PMID: 30119050 PMCID: PMC6097462 DOI: 10.1016/j.tranon.2018.07.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/23/2018] [Indexed: 02/05/2023] Open
Abstract
Breast cancer is the second leading cause of cancer death in women worldwide. Incurable metastatic breast disease presents a major clinical challenge and is the main cause of breast cancer-related death. The epithelial-mesenchymal transition (EMT) is a critical early promoter of metastasis. In the present study, we identified a novel role for the inhibitor of DNA binding 2 (Id2), a member of the basic helix-loop-helix protein family, during the EMT of breast cancer. Expression of Id2 was positively correlated with Notch3 in breast cancer cells. Low expression of Id2 and Notch3 was associated with worse distant metastasis-free survival in breast cancer patients. The present study revealed that Id2 activated Notch3 expression by blocking E2A binding to an E-box motif in the Notch3 promoter. The Id2-mediated up-regulation of Notch3 expression at both the mRNA and protein levels resulted in an attenuated EMT, which was associated with reduced motility and matrix invasion of ER-positive and -negative human breast cancer cells and the emergence of E-cadherin expression and reduction in the mesenchymal marker vimentin in triple-negative breast cancer cells. In summary, our findings identified Id2 as a suppressor of the EMT and positive transcriptional regulator of Notch3 in breast cancer. Id2 and Notch3 may serve as novel prognostic markers in a subpopulation of ER-positive breast cancer patients.
Collapse
Affiliation(s)
- Xiao-Fen Wen
- Department of Breast Medical Oncology, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, China; ChangJiang Scholar's Laboratory, Shantou University Medical College, 22 Xinling Road, Shantou, China
| | - Min Chen
- ChangJiang Scholar's Laboratory, Shantou University Medical College, 22 Xinling Road, Shantou, China; Xiang'an Hospital, Xiamen University, 2000 East Xiang'an Rd, Xiamen, Fujian, China
| | - Yang Wu
- ChangJiang Scholar's Laboratory, Shantou University Medical College, 22 Xinling Road, Shantou, China
| | - Min-Na Chen
- Department of Breast Medical Oncology, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, China; ChangJiang Scholar's Laboratory, Shantou University Medical College, 22 Xinling Road, Shantou, China
| | - Aleksandra Glogowska
- Dept. of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Thomas Klonisch
- Dept. of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.
| | - Guo-Jun Zhang
- ChangJiang Scholar's Laboratory, Shantou University Medical College, 22 Xinling Road, Shantou, China; Xiang'an Hospital, Xiamen University, 2000 East Xiang'an Rd, Xiamen, Fujian, China.
| |
Collapse
|
11
|
Elaimy AL, Guru S, Chang C, Ou J, Amante JJ, Zhu LJ, Goel HL, Mercurio AM. VEGF-neuropilin-2 signaling promotes stem-like traits in breast cancer cells by TAZ-mediated repression of the Rac GAP β2-chimaerin. Sci Signal 2018; 11:11/528/eaao6897. [PMID: 29717062 DOI: 10.1126/scisignal.aao6897] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The role of vascular endothelial growth factor (VEGF) signaling in cancer is not only well known in the context of angiogenesis but also important in the functional regulation of tumor cells. Autocrine VEGF signaling mediated by its co-receptors called neuropilins (NRPs) appears to be essential for sustaining the proliferation and survival of cancer stem cells (CSCs), which are implicated in mediating tumor growth, progression, and drug resistance. Therefore, understanding the mechanisms involved in VEGF-mediated support of CSCs is critical to successfully treating cancer patients. The expression of the Hippo effector TAZ is associated with breast CSCs and confers stem cell-like properties. We found that VEGF-NRP2 signaling contributed to the activation of TAZ in various breast cancer cells, which mediated a positive feedback loop that promoted mammosphere formation. VEGF-NRP2 signaling activated the GTPase Rac1, which inhibited the Hippo kinase LATS, thus leading to TAZ activity. In a complex with the transcription factor TEAD, TAZ then bound and repressed the promoter of the gene encoding the Rac GTPase-activating protein (Rac GAP) β2-chimaerin. By activating GTP hydrolysis, Rac GAPs effectively turn off Rac signaling; hence, the TAZ-mediated repression of β2-chimaerin resulted in sustained Rac1 activity in CSCs. Depletion of β2-chimaerin in non-CSCs increased Rac1 activity, TAZ abundance, and mammosphere formation. Analysis of a breast cancer patient database revealed an inverse correlation between β2-chimaerin and TAZ expression in tumors. Our findings highlight an unexpected role for β2-chimaerin in a feed-forward loop of TAZ activation and the acquisition of CSC properties.
Collapse
Affiliation(s)
- Ameer L Elaimy
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605, USA.,Medical Scientist Training Program, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01605, USA
| | - Santosh Guru
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605, USA
| | - Cheng Chang
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Jianhong Ou
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605, USA
| | - John J Amante
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605, USA
| | - Lihua Julie Zhu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605, USA.,Department of Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.,Department of Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Hira Lal Goel
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605, USA
| | - Arthur M Mercurio
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605, USA.
| |
Collapse
|
12
|
Integrin-β4 identifies cancer stem cell-enriched populations of partially mesenchymal carcinoma cells. Proc Natl Acad Sci U S A 2017; 114:E2337-E2346. [PMID: 28270621 DOI: 10.1073/pnas.1618298114] [Citation(s) in RCA: 233] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Neoplastic cells within individual carcinomas often exhibit considerable phenotypic heterogeneity in their epithelial versus mesenchymal-like cell states. Because carcinoma cells with mesenchymal features are often more resistant to therapy and may serve as a source of relapse, we sought to determine whether such cells could be further stratified into functionally distinct subtypes. Indeed, we find that a basal epithelial marker, integrin-β4 (ITGB4), can be used to enable stratification of mesenchymal-like triple-negative breast cancer (TNBC) cells that differ from one another in their relative tumorigenic abilities. Notably, we demonstrate that ITGB4+ cancer stem cell (CSC)-enriched mesenchymal cells reside in an intermediate epithelial/mesenchymal phenotypic state. Among patients with TNBC who received chemotherapy, elevated ITGB4 expression was associated with a worse 5-year probability of relapse-free survival. Mechanistically, we find that the ZEB1 (zinc finger E-box binding homeobox 1) transcription factor activity in highly mesenchymal SUM159 TNBC cells can repress expression of the epithelial transcription factor TAp63α (tumor protein 63 isoform 1), a protein that promotes ITGB4 expression. In addition, we demonstrate that ZEB1 and ITGB4 are important in modulating the histopathological phenotypes of tumors derived from mesenchymal TNBC cells. Hence, mesenchymal carcinoma cell populations are internally heterogeneous, and ITGB4 is a mechanistically driven prognostic biomarker that can be used to identify the more aggressive subtypes of mesenchymal carcinoma cells in TNBC. The ability to rapidly isolate and mechanistically interrogate the CSC-enriched, partially mesenchymal carcinoma cells should further enable identification of novel therapeutic opportunities to improve the prognosis for high-risk patients with TNBC.
Collapse
|
13
|
Kamata YU, Sumida T, Kobayashi Y, Ishikawa A, Kumamaru W, Mori Y. Introduction of ID2 Enhances Invasiveness in ID2-null Oral Squamous Cell Carcinoma Cells via the SNAIL Axis. Cancer Genomics Proteomics 2017; 13:493-497. [PMID: 27807072 DOI: 10.21873/cgp.20012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 07/21/2016] [Indexed: 11/10/2022] Open
Abstract
AIM Inhibitor of DNA-binding (ID) proteins are negative regulators of basic helix-loop-helix transcription factors that generally stimulate cell proliferation and inhibit differentiation. However, the role of ID2 in cancer progression remains ambiguous. Here, we investigated the function of ID2 in ID2-null oral squamous cell carcinoma (OSCC) cells. MATERIALS AND METHODS We introduced an ID2 cDNA construct into ID2-null OSCC cells and compared them with empty-vector-transfected cells in terms of cell proliferation, invasion, and activity and expression of matrix metalloproteinase (MMP). RESULTS ID2 introduction resulted in enhanced malignant phenotypes. The ID2-expressing cells showed increased N-cadherin, vimentin, and E-cadherin expression and epithelial-mesenchymal transition. In addition, cell invasion drastically increased with increased expression and activity of MMP2. Immunoprecipitation revealed a direct interaction between ID2 and zinc finger transcription factor, snail family transcriptional repressor 1 (SNAIL1). CONCLUSION ID2 expression triggered a malignant phenotype, especially of invasive properties, through the ID2-SNAIL axis. Thus, ID2 represents a potential therapeutic target for OSCC.
Collapse
Affiliation(s)
- Y U Kamata
- Section of Oral & Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Tomoki Sumida
- Section of Oral & Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yosuke Kobayashi
- Section of Oral & Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Akiko Ishikawa
- Department of Oral and Maxillofacial Surgery, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Wataru Kumamaru
- Section of Oral & Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yoshihide Mori
- Section of Oral & Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| |
Collapse
|
14
|
Sumida T, Ishikawa A, Nakano H, Yamada T, Mori Y, Desprez PY. Targeting ID2 expression triggers a more differentiated phenotype and reduces aggressiveness in human salivary gland cancer cells. Genes Cells 2016; 21:915-20. [PMID: 27364596 DOI: 10.1111/gtc.12389] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 05/21/2016] [Indexed: 11/28/2022]
Abstract
Inhibitors of DNA-binding (ID) proteins are negative regulators of basic helix-loop-helix transcription factors and generally stimulate cell proliferation and inhibit differentiation. We previously determined that ID1 was highly expressed in aggressive salivary gland cancer (SGC) cells in culture. Here, we show that ID2 is also expressed in aggressive SGC cells. ID2 knockdown triggers important changes in cell behavior, that is, it significantly reduces the expression of N-cadherin, vimentin and Snail, induces E-cadherin expression and leads to a more differentiated phenotype exemplified by changes in cell shape. Moreover, ID2 knockdown almost completely suppresses invasion and the expression of matrix metalloproteinase 9. In conclusion, ID2 expression maintains an aggressive phenotype in SGC cells, and ID2 repression triggers a reduction in cell aggressiveness. ID2 therefore represents a potential therapeutic target during SGC progression. ID proteins are negative regulators of basic helix-loop-helix transcription factors and generally stimulate cell proliferation and inhibit differentiation. ID2 knockdown triggers important changes in cell behavior, that is, it significantly reduces the expression of N-cadherin, vimentin and Snail, induces E-cadherin expression and leads to a more differentiated phenotype exemplified by changes in cell shape. ID2 therefore represents a potential therapeutic target during SGC progression.
Collapse
Affiliation(s)
- Tomoki Sumida
- Section of Oral & Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 8128582, Japan
| | - Akiko Ishikawa
- Department of Oral & Maxillofacial Surgery, Ehime University Graduate School of Medicine, 454, Shitsukawa, Toon, 7910295, Japan
| | - Hiroyuki Nakano
- Section of Oral & Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 8128582, Japan
| | - Tomohiro Yamada
- Section of Oral & Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 8128582, Japan
| | - Yoshihide Mori
- Section of Oral & Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 8128582, Japan
| | - Pierre-Yves Desprez
- California Pacific Medical Center, Cancer Research Institute, 475 Brannan Street, Suite 220, San Francisco, California, 94107, USA
| |
Collapse
|
15
|
A 3D in vitro model to explore the inter-conversion between epithelial and mesenchymal states during EMT and its reversion. Sci Rep 2016; 6:27072. [PMID: 27255191 PMCID: PMC4891772 DOI: 10.1038/srep27072] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 05/15/2016] [Indexed: 12/16/2022] Open
Abstract
Epithelial-to-mesenchymal transitions (EMT) are strongly implicated in cancer dissemination. Intermediate states, arising from inter-conversion between epithelial (E) and mesenchymal (M) states, are characterized by phenotypic heterogeneity combining E and M features and increased plasticity. Hybrid EMT states are highly relevant in metastatic contexts, but have been largely neglected, partially due to the lack of physiologically-relevant 3D platforms to study them. Here we propose a new in vitro model, combining mammary E cells with a bioengineered 3D matrix, to explore phenotypic and functional properties of cells in transition between E and M states. Optimized alginate-based 3D matrices provided adequate 3D microenvironments, where normal epithelial morphogenesis was recapitulated, with formation of acini-like structures, similar to those found in native mammary tissue. TGFβ1-driven EMT in 3D could be successfully promoted, generating M-like cells. TGFβ1 removal resulted in phenotypic switching to an intermediate state (RE cells), a hybrid cell population expressing both E and M markers at gene/protein levels. RE cells exhibited increased proliferative/clonogenic activity, as compared to M cells, being able to form large colonies containing cells with front-back polarity, suggesting a more aggressive phenotype. Our 3D model provides a powerful tool to investigate the role of the microenvironment on metastable EMT stages.
Collapse
|
16
|
Pioli PD, Whiteside SK, Weis JJ, Weis JH. Snai2 and Snai3 transcriptionally regulate cellular fitness and functionality of T cell lineages through distinct gene programs. Immunobiology 2016; 221:618-33. [PMID: 26831822 DOI: 10.1016/j.imbio.2016.01.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 01/13/2016] [Accepted: 01/14/2016] [Indexed: 01/31/2023]
Abstract
T lymphocytes are essential contributors to the adaptive immune system and consist of multiple lineages that serve various effector and regulatory roles. As such, precise control of gene expression is essential to the proper development and function of these cells. Previously, we identified Snai2 and Snai3 as being essential regulators of immune tolerance partly due to the impaired function of CD4(+) regulatory T cells in Snai2/3 conditional double knockout mice. Here we extend those previous findings using a bone marrow transplantation model to provide an environmentally unbiased view of the molecular changes imparted onto various T lymphocyte populations once Snai2 and Snai3 are deleted. The data presented here demonstrate that Snai2 and Snai3 transcriptionally regulate the cellular fitness and functionality of not only CD4(+) regulatory T cells but effector CD8(α+) and CD4(+) conventional T cells as well. This is achieved through the modulation of gene sets unique to each cell type and includes transcriptional targets relevant to the survival and function of each T cell lineage. As such, Snai2 and Snai3 are essential regulators of T cell immunobiology.
Collapse
Affiliation(s)
- Peter D Pioli
- Division of Cell Biology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84132, United States.
| | - Sarah K Whiteside
- Division of Cell Biology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84132, United States
| | - Janis J Weis
- Division of Cell Biology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84132, United States
| | - John H Weis
- Division of Cell Biology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84132, United States
| |
Collapse
|
17
|
Zhou JP, Gao ZL, Zhou ML, He MY, Xu XH, Tao DT, Yang CC, Liu LK. Snail interacts with Id2 in the regulation of TNF-α-induced cancer cell invasion and migration in OSCC. Am J Cancer Res 2015; 5:1680-1691. [PMID: 26175937 PMCID: PMC4497435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 04/10/2015] [Indexed: 06/04/2023] Open
Abstract
The inflammatory tumor microenvironment has been identified to play a pivotal role in tumor development and metastasis. Tumor necrosis factor-α (TNF-α) is one of the key cytokines that regulate the inflammatory processes in tumor promotion. In the current study, we treated three oral squamous cell carcinoma (OSCC) cell lines with TNF-α to study its role in inflammation-induced tumor progression. Here we show that TNF-α induces stabilization of the transcriptional repressor Snail and activates NF-κB pathway in the three OSCC cell lines. These activities resulted in the increased motility and invasiveness of three OSCC cell lines. In addition, upon dealing with TNF-α for the indicated time, three OSCC cell lines underwent epithelial-to-mesenchymal transition (EMT), in which they presented a fibroblast-like phenotype and had a decreased expression of epithelial marker (E-cadherin) and an increased expression of mesenchymal marker (vimentin). We further demonstrated that TNF-α can up-regulate the expression of Id2 while inducing an EMT in oral cancer cells. Finally, we showed that Id2 interacted with Snail which may constrain Snail-dependent suppression of E-cadherin. In conclusion, our study indicates that TNF-α induces Snail stabilization is dependent on the activation of NF-κB pathway and results in increasing cell invasion and migration in OSCC cells. Id2 may contribute to regulate the function of Snail during TNF-α-mediated EMT in OSCC. These findings have significant implications for inflammation-induced tumor promotion in OSCC.
Collapse
Affiliation(s)
- Jing-Ping Zhou
- Department of Oral Medicine, School of Stomatology, Wannan Medical CollegeWuhu, People’s Republic of China
| | - Zhen-Lin Gao
- Department of Oncology IV, First Hospital of ShijiazhuangShijiazhuang, People’s Republic of China
| | - Mei-Ling Zhou
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical UniversityNanjing, People’s Republic of China
- Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical UniversityNanjing, People’s Republic of China
| | - Meng-Ying He
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical UniversityNanjing, People’s Republic of China
- Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical UniversityNanjing, People’s Republic of China
| | - Xiao-Hui Xu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical UniversityNanjing, People’s Republic of China
- Department of The First Outpatient, Affiliated Hospital of Stomatology, Nanjing Medical UniversityNanjing, People’s Republic of China
| | - De-Tao Tao
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Wannan Medical CollegeWuhu, People’s Republic of China
| | - Cong-Chong Yang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical UniversityNanjing, People’s Republic of China
- Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical UniversityNanjing, People’s Republic of China
| | - Lai-Kui Liu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical UniversityNanjing, People’s Republic of China
- Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical UniversityNanjing, People’s Republic of China
| |
Collapse
|
18
|
Cheng X, Yang Y, Fan Z, Yu L, Bai H, Zhou B, Wu X, Xu H, Fang M, Shen A, Chen Q, Xu Y. MKL1 potentiates lung cancer cell migration and invasion by epigenetically activating MMP9 transcription. Oncogene 2015; 34:5570-81. [PMID: 25746000 DOI: 10.1038/onc.2015.14] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 12/17/2014] [Accepted: 01/07/2015] [Indexed: 12/24/2022]
Abstract
Malignant tumors are exemplified by excessive proliferation and aggressive migration/invasion contributing to increased mortality of cancer patients. Matrix metalloproteinase 9 (MMP9) expression is positively correlated with lung cancer malignancy. The mechanism underlying an elevated MMP9 expression is not clearly defined. We demonstrate here that the transcriptional modulator megakaryocytic leukemia 1 (MKL1) was activated by hypoxia and transforming growth factor (TGF-β), two prominent pro-malignancy factors, in cultured lung cancer cells. MKL1 levels were also increased in more invasive types of lung cancer in humans. Depletion of MKL1 in lung cancer cells attenuated migration and invasion both in vitro and in vivo. Overexpression of MKL1 potentiated the induction of MMP9 transcription by hypoxia and TGF-β, whereas MKL1 silencing diminished MMP9 expression. Of interest, MKL1 knockdown eliminated histone H3K4 methylation surrounding the MMP9 promoter. Further analyses revealed that MKL1 recruited ASH2, a component of the H3K4 methyltransferase complex, to activate MMP9 transcription. Depletion of ASH2 ameliorated cancer cell migration and invasion in an MMP9-dependent manner. Together our data indicate that MKL1 potentiates lung cancer cell migration and invasion by epigenetically activating MMP9 transcription.
Collapse
Affiliation(s)
- X Cheng
- Key Laboratory of Cardiovascular Disease and Department of Pathophysiology, Nanjing Medical University, Nanjing, China.,Jiangsu Institute of Nuclear Medicine, Wuxi, China
| | - Y Yang
- Key Laboratory of Cardiovascular Disease and Department of Pathophysiology, Nanjing Medical University, Nanjing, China.,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Z Fan
- Key Laboratory of Cardiovascular Disease and Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - L Yu
- Key Laboratory of Cardiovascular Disease and Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - H Bai
- Key Laboratory of Cardiovascular Disease and Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - B Zhou
- Key Laboratory of Cardiovascular Disease and Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - X Wu
- Key Laboratory of Cardiovascular Disease and Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - H Xu
- Key Laboratory of Cardiovascular Disease and Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - M Fang
- Key Laboratory of Cardiovascular Disease and Department of Pathophysiology, Nanjing Medical University, Nanjing, China.,Department of Nursing, Jiangsu Jiankang Vocational University, Nanjing, China
| | - A Shen
- Department of Key Laboratory of Inflammation and Molecular Targets, Medical College, Nantong University, Nantong, China
| | - Q Chen
- Key Laboratory of Cardiovascular Disease and Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Y Xu
- Key Laboratory of Cardiovascular Disease and Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
19
|
Cichon MA, Nelson CM, Radisky DC. Regulation of epithelial-mesenchymal transition in breast cancer cells by cell contact and adhesion. Cancer Inform 2015; 14:1-13. [PMID: 25698877 PMCID: PMC4325704 DOI: 10.4137/cin.s18965] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 12/29/2014] [Accepted: 01/04/2015] [Indexed: 02/06/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a physiological program that is activated during cancer cell invasion and metastasis. We show here that EMT-related processes are linked to a broad and conserved program of transcriptional alterations that are influenced by cell contact and adhesion. Using cultured human breast cancer and mouse mammary epithelial cells, we find that reduced cell density, conditions under which cell contact is reduced, leads to reduced expression of genes associated with mammary epithelial cell differentiation and increased expression of genes associated with breast cancer. We further find that treatment of cells with matrix metalloproteinase-3 (MMP-3), an inducer of EMT, interrupts a defined subset of cell contact-regulated genes, including genes encoding a variety of RNA splicing proteins known to regulate the expression of Rac1b, an activated splice isoform of Rac1 known to be a key mediator of MMP-3-induced EMT in breast, lung, and pancreas. These results provide new insights into how MMPs act in cancer progression and how loss of cell-cell interactions is a key step in the earliest stages of cancer development.
Collapse
Affiliation(s)
- Magdalena A Cichon
- Department of Cancer Biology, Mayo Clinic Cancer Center, Jacksonville, FL USA
| | - Celeste M Nelson
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic Cancer Center, Jacksonville, FL USA
| |
Collapse
|
20
|
Niu G, Ye T, Qin L, Bourbon PM, Chang C, Zhao S, Li Y, Zhou L, Cui P, Rabinovitz I, Mercurio AM, Zhao D, Zeng H. Orphan nuclear receptor TR3/Nur77 improves wound healing by upregulating the expression of integrin β4. FASEB J 2014; 29:131-40. [PMID: 25326539 DOI: 10.1096/fj.14-257550] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Tissue repair/wound healing, in which angiogenesis plays an important role, is a critical step in many diseases including chronic wound, myocardial infarction, stroke, cancer, and inflammation. Recently, we were the first to report that orphan nuclear receptor TR3/Nur77 is a critical mediator of angiogenesis and its associated microvessel permeability. Tumor growth and angiogenesis induced by VEGF-A, histamine, and serotonin are almost completely inhibited in Nur77 knockout mice. However, it is not known whether TR3/Nur77 plays any roles in wound healing. In these studies, skin wound-healing assay was performed in 3 types of genetically modified mice having various Nur77 activities. We found that ectopic induction of Nur77 in endothelial cells of mice is sufficient to improve skin wound healing. Although skin wound healing in Nur77 knockout mice is comparable to the wild-type control mice, the process is significantly delayed in the EC-Nur77-DN mice, in which a dominant negative Nur77 mutant is inducibly and specifically expressed in mouse endothelial cells. By a loss-of-function assay, we elucidate a novel feed-forward signaling pathway, integrin β4 → PI3K → Akt → FAK, by which TR3 mediates HUVEC migration. Furthermore, TR3/Nur77 regulates the expression of integrin β4 by targeting its promoter activity. In conclusion, expression of TR3/Nur77 improves wound healing by targeting integrin β4. TR3/Nur77 is a potential candidate for proangiogenic therapy. The results further suggest that TR3/Nur77 is required for pathologic angiogenesis but not for developmental/physiologic angiogenesis and that Nur77 and its family members play a redundant role in normal skin wound healing.
Collapse
Affiliation(s)
- Gengming Niu
- Center for Vascular Biology Research and Division of Molecular and Vascular Biology, Department of Medicine and Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Taiyang Ye
- Center for Vascular Biology Research and Division of Molecular and Vascular Biology, Department of Medicine and Department of Obstetrics & Gynecology, Renji Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, People's Republic of China
| | | | | | - Cheng Chang
- Department of Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Shengqiang Zhao
- Center for Vascular Biology Research and Division of Molecular and Vascular Biology, Department of Medicine and Department of Gastroenterology, Provincial Hospital Affiliated to Shandong University, Ji-nan, People's Republic of China
| | - Yan Li
- Center for Vascular Biology Research and Division of Molecular and Vascular Biology, Department of Medicine and Department of Gastroenterology, Provincial Hospital Affiliated to Shandong University, Ji-nan, People's Republic of China
| | - Lei Zhou
- Center for Vascular Biology Research and Division of Molecular and Vascular Biology, Department of Medicine and Departments of Hepatobiliary Surgery and General Surgery, the First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Pengfei Cui
- Center for Vascular Biology Research and Division of Molecular and Vascular Biology, Department of Medicine and Department of General Surgery, Pancreatic Disease Institute, Union Hospital, Huazhong University of Science & Technology (HUST), Wuhan, People's Republic of China
| | - Issac Rabinovitz
- Center for Vascular Biology Research and Division of Molecular and Vascular Biology, Department of Medicine and Department of Pathology
| | - Arthur M Mercurio
- Department of Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Dezheng Zhao
- Center for Vascular Biology Research and Division of Molecular and Vascular Biology, Department of Medicine and Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Huiyan Zeng
- Center for Vascular Biology Research and Division of Molecular and Vascular Biology, Department of Medicine and
| |
Collapse
|
21
|
Dentelli P, Traversa M, Rosso A, Togliatto G, Olgasi C, Marchiò C, Provero P, Lembo A, Bon G, Annaratone L, Sapino A, Falcioni R, Brizzi MF. miR-221/222 control luminal breast cancer tumor progression by regulating different targets. Cell Cycle 2014; 13:1811-26. [PMID: 24736554 DOI: 10.4161/cc.28758] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
α6β4 integrin is an adhesion molecule for laminin receptors involved in tumor progression. We present a link between β4 integrin expression and miR-221/222 in the most prevalent human mammary tumor: luminal invasive carcinomas (Lum-ICs). Using human primary tumors that display different β4 integrin expression and grade, we show that miR-221/222 expression inversely correlates with tumor proliferating index, Ki67. Interestingly, most high-grade tumors express β4 integrin and low miR-221/222 levels. We ectopically transfected miR-221/222 into a human-derived mammary tumor cell line that recapitulates the luminal subtype to investigate whether miR-221/222 regulates β4 expression. We demonstrate that miR-221/222 overexpression results in β4 expression downregulation, breast cancer cell proliferation, and invasion inhibition. The role of miR-221/222 in driving β4 integrin expression is also confirmed via mutating the miR-221/222 seed sequence for β4 integrin 3'UTR. Furthermore, we show that these 2 miRNAs are also key breast cancer cell proliferation and invasion regulators, via the post-transcriptional regulation of signal transducer and activator of transcription 5A (STAT5A) and of a disintegrin and metalloprotease-17 (ADAM-17). We further confirm these data by silencing ADAM-17, using a dominant-negative or an activated STAT5A form. miR-221/222-driven β4 integrin, STAT5A, and ADAM-17 did not occur in MCF-10A cells, denoted "normal" breast epithelial cells, indicating that the mechanism is cancer cell-specific. These results provide the first evidence of a post-transcriptional mechanism that regulates β4 integrin, STAT5A, and ADAM-17 expression, thus controlling breast cancer cell proliferation and invasion. Pre-miR-221/222 use in the aggressive luminal subtype may be a powerful therapeutic anti-cancer strategy.
Collapse
Affiliation(s)
| | - Matteo Traversa
- Department of Medical Sciences; University of Torino; Torino, Italy
| | - Arturo Rosso
- Department of Medical Sciences; University of Torino; Torino, Italy
| | | | - Cristina Olgasi
- Department of Medical Sciences; University of Torino; Torino, Italy
| | - Caterina Marchiò
- Department of Medical Sciences; University of Torino; Torino, Italy
| | - Paolo Provero
- Department of Molecular Biotechnology and Health Sciences; University of Torino; Torino, Italy; Center for Translational Genomics and Bioinformatics; San Raffaele Scientific Institute; Milan, Italy
| | - Antonio Lembo
- Department of Molecular Biotechnology and Health Sciences; University of Torino; Torino, Italy; Center for Translational Genomics and Bioinformatics; San Raffaele Scientific Institute; Milan, Italy
| | - Giulia Bon
- Department of Experimental Oncology; Regina Elena National Cancer Institute; Rome, Italy
| | - Laura Annaratone
- Department of Medical Sciences; University of Torino; Torino, Italy
| | - Anna Sapino
- Department of Medical Sciences; University of Torino; Torino, Italy
| | - Rita Falcioni
- Department of Experimental Oncology; Regina Elena National Cancer Institute; Rome, Italy
| | | |
Collapse
|
22
|
de Jong J, Akhtar W, Badhai J, Rust AG, Rad R, Hilkens J, Berns A, van Lohuizen M, Wessels LFA, de Ridder J. Chromatin landscapes of retroviral and transposon integration profiles. PLoS Genet 2014; 10:e1004250. [PMID: 24721906 PMCID: PMC3983033 DOI: 10.1371/journal.pgen.1004250] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 02/04/2014] [Indexed: 12/16/2022] Open
Abstract
The ability of retroviruses and transposons to insert their genetic material into host DNA makes them widely used tools in molecular biology, cancer research and gene therapy. However, these systems have biases that may strongly affect research outcomes. To address this issue, we generated very large datasets consisting of to unselected integrations in the mouse genome for the Sleeping Beauty (SB) and piggyBac (PB) transposons, and the Mouse Mammary Tumor Virus (MMTV). We analyzed (epi)genomic features to generate bias maps at both local and genome-wide scales. MMTV showed a remarkably uniform distribution of integrations across the genome. More distinct preferences were observed for the two transposons, with PB showing remarkable resemblance to bias profiles of the Murine Leukemia Virus. Furthermore, we present a model where target site selection is directed at multiple scales. At a large scale, target site selection is similar across systems, and defined by domain-oriented features, namely expression of proximal genes, proximity to CpG islands and to genic features, chromatin compaction and replication timing. Notable differences between the systems are mainly observed at smaller scales, and are directed by a diverse range of features. To study the effect of these biases on integration sites occupied under selective pressure, we turned to insertional mutagenesis (IM) screens. In IM screens, putative cancer genes are identified by finding frequently targeted genomic regions, or Common Integration Sites (CISs). Within three recently completed IM screens, we identified 7%–33% putative false positive CISs, which are likely not the result of the oncogenic selection process. Moreover, results indicate that PB, compared to SB, is more suited to tag oncogenes. Retroviruses and transposons are widely used in cancer research and gene therapy. However, these systems show integration biases that may strongly affect results. To address this issue, we generated very large datasets consisting of to unselected integrations for the Sleeping Beauty and piggyBac transposons, and the Mouse Mammary Tumor Virus (MMTV). We analyzed (epi)genomic features to generate bias maps at local and genome-wide scales. MMTV showed a remarkably uniform distribution of integrations across the genome, and a striking similarity was observed between piggyBac and the Murine Leukemia Virus. Moreover, we find that target site selection is directed at multiple scales. At larger scales, it is similar across systems, and directed by a set of domain-oriented features, including chromatin compaction, replication timing, and CpG islands. Notable differences between systems are defined at smaller scales by a diverse range of epigenetic features. As a practical application of our findings, we determined that three recent insertional mutagenesis screens - commonly used for cancer gene discovery - contained 7%–33% putative false positive integration hotspots.
Collapse
Affiliation(s)
- Johann de Jong
- Computational Cancer Biology Group, Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Netherlands Consortium for Systems Biology, Amsterdam, The Netherlands
| | - Waseem Akhtar
- Netherlands Consortium for Systems Biology, Amsterdam, The Netherlands
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jitendra Badhai
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Alistair G. Rust
- Wellcome Trust Sanger Institute, Genome Campus, Hinxton-Cambridge, United Kingdom
| | - Roland Rad
- Department of Medicine II; Klinikum Rechts der Isar; Technische Universität München, German Cancer Research Center (DKFZ), Heidelberg, & German Cancer Consortium (DKTK), Heidelberg, Germany
| | - John Hilkens
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Anton Berns
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Skoltech Center for Stem Cell Research, Skolkovo Institute for Science and Technology, Skolkovo, Odintsovsky, Moscow, Russia
| | - Maarten van Lohuizen
- Netherlands Consortium for Systems Biology, Amsterdam, The Netherlands
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Lodewyk F. A. Wessels
- Computational Cancer Biology Group, Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Netherlands Consortium for Systems Biology, Amsterdam, The Netherlands
- Delft Bioinformatics Lab, Faculty of EEMCS, TU Delft, Delft, The Netherlands
- * E-mail: (LFAW); (JdR)
| | - Jeroen de Ridder
- Delft Bioinformatics Lab, Faculty of EEMCS, TU Delft, Delft, The Netherlands
- * E-mail: (LFAW); (JdR)
| |
Collapse
|
23
|
Moustakas A, Heldin P. TGFβ and matrix-regulated epithelial to mesenchymal transition. Biochim Biophys Acta Gen Subj 2014; 1840:2621-34. [PMID: 24561266 DOI: 10.1016/j.bbagen.2014.02.004] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Accepted: 02/05/2014] [Indexed: 12/14/2022]
Abstract
BACKGROUND The progression of cancer through stages that guide a benign hyperplastic epithelial tissue towards a fully malignant and metastatic carcinoma, is driven by genetic and microenvironmental factors that remodel the tissue architecture. The concept of epithelial-mesenchymal transition (EMT) has evolved to emphasize the importance of plastic changes in tissue architecture, and the cross-communication of tumor cells with various cells in the stroma and with specific molecules in the extracellular matrix (ECM). SCOPE OF THE REVIEW Among the multitude of ECM-embedded cytokines and the regulatory potential of ECM molecules, this article focuses on the cytokine transforming growth factor β (TGFβ) and the glycosaminoglycan hyaluronan, and their roles in cancer biology and EMT. For brevity, we concentrate our effort on breast cancer. MAJOR CONCLUSIONS Both normal and abnormal TGFβ signaling can be detected in carcinoma and stromal cells, and TGFβ-induced EMT requires the expression of hyaluronan synthase 2 (HAS2). Correspondingly, hyaluronan is a major constituent of tumor ECM and aberrant levels of both hyaluronan and TGFβ are thought to promote a wounding reaction to the local tissue homeostasis. The link between EMT and metastasis also involves the mesenchymal-epithelial transition (MET). ECM components, signaling networks, regulatory non-coding RNAs and epigenetic mechanisms form the network of regulation during EMT-MET. GENERAL SIGNIFICANCE Understanding the mechanism that controls epithelial plasticity in the mammary gland promises the development of valuable biomarkers for the prognosis of breast cancer progression and even provides new ideas for a more integrative therapeutic approach against disease. This article is part of a Special Issue entitled Matrix-mediated cell behaviour and properties.
Collapse
Affiliation(s)
- Aristidis Moustakas
- Ludwig Institute for 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.
| | - Paraskevi Heldin
- Ludwig Institute for 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.
| |
Collapse
|
24
|
Lasorella A, Benezra R, Iavarone A. The ID proteins: master regulators of cancer stem cells and tumour aggressiveness. Nat Rev Cancer 2014; 14:77-91. [PMID: 24442143 DOI: 10.1038/nrc3638] [Citation(s) in RCA: 281] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Inhibitor of DNA binding (ID) proteins are transcriptional regulators that control the timing of cell fate determination and differentiation in stem and progenitor cells during normal development and adult life. ID genes are frequently deregulated in many types of human neoplasms, and they endow cancer cells with biological features that are hijacked from normal stem cells. The ability of ID proteins to function as central 'hubs' for the coordination of multiple cancer hallmarks has established these transcriptional regulators as therapeutic targets and biomarkers in specific types of human tumours.
Collapse
Affiliation(s)
- Anna Lasorella
- Institute for Cancer Genetics, Department of Pathology and Pediatrics, Columbia University Medical Center, 1130 St. Nicholas Avenue, New York, 10032 New York, USA
| | - Robert Benezra
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, Box 241, New York, 10065 New York, USA
| | - Antonio Iavarone
- Institute for Cancer Genetics, Department of Pathology and Neurology, Columbia University Medical Center, 1130 St. Nicholas Avenue, New York, 10032 New York, USA
| |
Collapse
|