1
|
Zhu R, Shirley CM, Chu SH, Li L, Nguyen BH, Seo J, Wu M, Seale T, Duffield AS, Staudt LM, Levis M, Hu Y, Small D. Inhibition of NOTCH4 sensitizes FLT3/ITD acute myeloid leukemia cells to FLT3 tyrosine kinase inhibition. Leukemia 2024; 38:1581-1591. [PMID: 38811818 DOI: 10.1038/s41375-024-02292-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 05/31/2024]
Abstract
Internal tandem duplication mutations of FLT3 (FLT3/ITD) confer poor prognosis in AML. FLT3 tyrosine kinase inhibitors (TKIs) alone have limited and transient clinical efficacy thus calling for new targets for more effective combination therapy. In a loss-of-function RNAi screen, we identified NOTCH4 as one such potential target whose inhibition proved cytotoxic to AML cells, and also sensitized them to FLT3 inhibition. Further investigation found increased NOTCH4 expression in FLT3/ITD AML cell lines and primary patient samples. Inhibition of NOTCH4 by shRNA knockdown, CRISPR-Cas9-based knockout or γ-secretase inhibitors synergized with FLT3 TKIs to kill FLT3/ITD AML cells in vitro. NOTCH4 inhibition sensitized TKI-resistant FLT3/ITD cells to FLT3 TKI inhibition. The combination reduced phospho-ERK and phospho-AKT, indicating inhibition of MAPK and PI3K/AKT signaling pathways. It also led to changes in expression of genes involved in regulating cell cycling, DNA repair and transcription. A patient-derived xenograft model showed that the combination reduced both the level of leukemic involvement of primary human FLT3/ITD AML cells and their ability to engraft secondary recipients. In summary, these results demonstrate that NOTCH4 inhibition synergizes with FLT3 TKIs to eliminate FLT3/ITD AML cells, providing a new therapeutic target for AML with FLT3/ITD mutations.
Collapse
MESH Headings
- Humans
- fms-Like Tyrosine Kinase 3/genetics
- fms-Like Tyrosine Kinase 3/antagonists & inhibitors
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
- Animals
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Mice
- Receptor, Notch4/genetics
- Xenograft Model Antitumor Assays
- Mutation
- Cell Line, Tumor
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Ruiqi Zhu
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Courtney M Shirley
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - S Haihua Chu
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Beam Therapeutics, Cambridge, MA, USA
| | - Li Li
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bao H Nguyen
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jaesung Seo
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Min Wu
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tessa Seale
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Amy S Duffield
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Louis M Staudt
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mark Levis
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yu Hu
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Donald Small
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
2
|
Pinot M, Le Borgne R. Spatio-Temporal Regulation of Notch Activation in Asymmetrically Dividing Sensory Organ Precursor Cells in Drosophila melanogaster Epithelium. Cells 2024; 13:1133. [PMID: 38994985 PMCID: PMC11240559 DOI: 10.3390/cells13131133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/13/2024] Open
Abstract
The Notch communication pathway, discovered in Drosophila over 100 years ago, regulates a wide range of intra-lineage decisions in metazoans. The division of the Drosophila mechanosensory organ precursor is the archetype of asymmetric cell division in which differential Notch activation takes place at cytokinesis. Here, we review the molecular mechanisms by which epithelial cell polarity, cell cycle and intracellular trafficking participate in controlling the directionality, subcellular localization and temporality of mechanosensitive Notch receptor activation in cytokinesis.
Collapse
Affiliation(s)
| | - Roland Le Borgne
- Univ Rennes, Centre National de la Recherche Scientifique UMR 6290, IGDR (Institut de Génétique et Développement de Rennes), F-35000 Rennes, France
| |
Collapse
|
3
|
Ren Y, Mao X, Xu H, Dang Q, Weng S, Zhang Y, Chen S, Liu S, Ba Y, Zhou Z, Han X, Liu Z, Zhang G. Ferroptosis and EMT: key targets for combating cancer progression and therapy resistance. Cell Mol Life Sci 2023; 80:263. [PMID: 37598126 PMCID: PMC10439860 DOI: 10.1007/s00018-023-04907-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/21/2023]
Abstract
Iron-dependent lipid peroxidation causes ferroptosis, a form of regulated cell death. Crucial steps in the formation of ferroptosis include the accumulation of ferrous ions (Fe2+) and lipid peroxidation, of which are controlled by glutathione peroxidase 4 (GPX4). Its crucial role in stopping the spread of cancer has been shown by numerous studies undertaken in the last ten years. Epithelial-mesenchymal transition (EMT) is the process by which epithelial cells acquire mesenchymal characteristics. EMT is connected to carcinogenesis, invasiveness, metastasis, and therapeutic resistance in cancer. It is controlled by a range of internal and external signals and changes the phenotype from epithelial to mesenchymal like. Studies have shown that mesenchymal cancer cells tend to be more ferroptotic than their epithelial counterparts. Drug-resistant cancer cells are more easily killed by inducers of ferroptosis when they undergo EMT. Therefore, understanding the interaction between ferroptosis and EMT will help identify novel cancer treatment targets. In-depth discussion is given to the regulation of ferroptosis, the potential application of EMT in the treatment of cancer, and the relationships between ferroptosis, EMT, and signaling pathways associated with tumors. Invasion, metastasis, and inflammation in cancer all include ferroptosis and EMT. The goal of this review is to provide suggestions for future research and practical guidance for applying ferroptosis and EMT in clinical practice.
Collapse
Affiliation(s)
- Yuqing Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xiangrong Mao
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Qin Dang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Shuang Chen
- Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shutong Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yuhao Ba
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Zhaokai Zhou
- Department of Pediatric Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Guojun Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
4
|
Cleymaet R, Vermassen T, Coopman R, Vermeersch H, De Keukeleire S, Rottey S. The Therapeutic Landscape of Salivary Gland Malignancies-Where Are We Now? Int J Mol Sci 2022; 23:ijms232314891. [PMID: 36499216 PMCID: PMC9740091 DOI: 10.3390/ijms232314891] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 11/23/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022] Open
Abstract
Salivary gland malignancies (SGMs) account for less than 5% of new diagnoses in head and neck tumors. If feasible, surgery is the preferred treatment modality. Nevertheless, some malignancies have a tendency of recurrence, with possible distant metastasis. Alternative treatment strategies, such as primary radiation or chemotherapeutics, often present low response rates. As a result, there is an unmet need for novel therapeutic approaches. Nowadays, target-based therapies (e.g., small inhibitors and immunotherapy) are used by the medical oncologist for possible treatment of advanced SGMs. Based on recent published trials, some novel treatments may provide additional disease control for some patients. However, sample sizes are small, the general findings are unsatisfactory, and a lot of uncertainties remain to be elucidated. Nevertheless, research shows that patients do not benefit from blind administration of systemic treatments and therefore a more personalized approach is highly needed. The aim of this review paper is to summarize the most recent advances in the biological understanding and molecular pathways of salivary gland cancers, the association of these pathways with the current treatments used and their implications for more personalized targeted-based therapies.
Collapse
Affiliation(s)
- Robbert Cleymaet
- Department of Oromaxillofacial and Plastic Surgery, Ghent University Hospital, 9000 Ghent, Belgium
| | - Tijl Vermassen
- Department Medical Oncology, University Hospital Ghent, 9000 Ghent, Belgium
- Department Basic and Applied Medical Sciences, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
- Correspondence: ; Tel.: +32-9-332-26-92
| | - Renaat Coopman
- Department of Oromaxillofacial and Plastic Surgery, Ghent University Hospital, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Hubert Vermeersch
- Department of Oromaxillofacial and Plastic Surgery, Ghent University Hospital, 9000 Ghent, Belgium
| | - Stijn De Keukeleire
- Department Internal Medicine, University Hospital Brussels, 1090 Brussels, Belgium
| | - Sylvie Rottey
- Department Medical Oncology, University Hospital Ghent, 9000 Ghent, Belgium
- Department Basic and Applied Medical Sciences, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
- Drug Research Unit Ghent, University Hospital Ghent, 9000 Ghent, Belgium
| |
Collapse
|
5
|
Lee SN, Yoon JH. The Role of Proprotein Convertases in Upper Airway Remodeling. Mol Cells 2022; 45:353-361. [PMID: 35611689 PMCID: PMC9200660 DOI: 10.14348/molcells.2022.0019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/22/2022] [Accepted: 02/27/2022] [Indexed: 11/27/2022] Open
Abstract
Chronic rhinosinusitis (CRS) is a multifactorial, heterogeneous disease characterized by persistent inflammation of the sinonasal mucosa and tissue remodeling, which can include basal/progenitor cell hyperplasia, goblet cell hyperplasia, squamous cell metaplasia, loss or dysfunction of ciliated cells, and increased matrix deposition. Repeated injuries can stimulate airway epithelial cells to produce inflammatory mediators that activate epithelial cells, immune cells, or the epithelial-mesenchymal trophic unit. This persistent inflammation can consequently induce aberrant tissue remodeling. However, the molecular mechanisms driving disease within the different molecular CRS subtypes remain inadequately characterized. Numerous secreted and cell surface proteins relevant to airway inflammation and remodeling are initially synthesized as inactive precursor proteins, including growth/differentiation factors and their associated receptors, enzymes, adhesion molecules, neuropeptides, and peptide hormones. Therefore, these precursor proteins require post-translational cleavage by proprotein convertases (PCs) to become fully functional. In this review, we summarize the roles of PCs in CRS-associated tissue remodeling and discuss the therapeutic potential of targeting PCs for CRS treatment.
Collapse
Affiliation(s)
- Sang-Nam Lee
- The Airway Mucus Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Joo-Heon Yoon
- The Airway Mucus Institute, Yonsei University College of Medicine, Seoul 03722, Korea
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
6
|
Tan H, Li M, Han L, Zhao Y, Zhang X. Gypensapogenin I Suppresses Cell Proliferation in Triple-Negative Breast Cancer Via Triggering the Closure of AKT/GSK3β/β-Catenin and Notch-1 Signaling Pathways. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:5438-5449. [PMID: 35465659 DOI: 10.1021/acs.jafc.2c02512] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Jiaogulan (Gynostemma pentaphyllum) tea is a functional food that is commercially available worldwide. Gypensapogenin I (Gyp I), which is a natural damarane-type saponin, was obtained from the hydrolysates of total gypenosides. The present research was performed to investigate the potential antiproliferation effect of Gyp I in MDA-MB-231 cells and the underlying mechanisms. Here, we found that Gyp I attenuated survival, inhibited proliferation, and induced apoptosis in MDA-MB-231 cells. Target prediction by binding molecule docking and western blot assays confirmed the mechanism by which Gyp I inhibited the proliferation of breast cancer cells via the AKT/GSK3β/β-catenin signaling pathway. We also showed that Gyp I exhibited superior in vivo efficacy that was dose dependent. Tumor tissue transcriptome analysis indicated that Gyp I could decrease the expression levels of NOTCH1 and HES1, which was in contrast to the effect on MAML and NUMBL, indicating that our compound hindered the activation of the Notch-1 signaling pathway. In summary, we report for the first time that Gyp I shows excellent anti-breast cancer activity in vivo and in vitro and that its pathway of action is related to the AKT/GSK3β/β-catenin and Notch-1 signaling pathways. Therefore, Jiaogulan tea can not only be used as a health food but also possesses the possibility to treat triple-negative breast cancer (TNBC).
Collapse
Affiliation(s)
- Hongyan Tan
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Minjie Li
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Linlin Han
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yuqing Zhao
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiaoshu Zhang
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
7
|
GIT1 protects against breast cancer growth through negative regulation of Notch. Nat Commun 2022; 13:1537. [PMID: 35318302 PMCID: PMC8940956 DOI: 10.1038/s41467-022-28631-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 01/18/2022] [Indexed: 12/20/2022] Open
Abstract
Hyperactive Notch signalling is frequently observed in breast cancer and correlates with poor prognosis. However, relatively few mutations in the core Notch signalling pathway have been identified in breast cancer, suggesting that as yet unknown mechanisms increase Notch activity. Here we show that increased expression levels of GIT1 correlate with high relapse-free survival in oestrogen receptor-negative (ER(-)) breast cancer patients and that GIT1 mediates negative regulation of Notch. GIT1 knockdown in ER(-) breast tumour cells increased signalling downstream of Notch and activity of aldehyde dehydrogenase, a predictor of poor clinical outcome. GIT1 interacts with the Notch intracellular domain (ICD) and influences signalling by inhibiting the cytoplasm-to-nucleus transport of the Notch ICD. In xenograft experiments, overexpression of GIT1 in ER(-) cells prevented or reduced Notch-driven tumour formation. These results identify GIT1 as a modulator of Notch signalling and a guardian against breast cancer growth. Notch signalling is reported to be hyperactivated in oestrogen receptor-negative (ER-) breast cancer. Here the authors show that G protein-coupled receptor kinase-interacting protein 1 (GIT1) negatively regulates Notch signalling and tumour growth in ER- breast cancer by blocking Notch ICD nuclear translocation.
Collapse
|
8
|
Liu S, Hsu EC, Shen M, Aslan M, Stoyanova T. Metastasis Model to Test the Role of Notch Signaling in Prostate Cancer. Methods Mol Biol 2022; 2472:221-233. [PMID: 35674904 DOI: 10.1007/978-1-0716-2201-8_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Distant metastasis is the main cause of death in prostate cancer patients. Notch signaling plays an important role in driving prostate cancer aggressiveness and metastasis. In this chapter, we describe a protocol to measure prostate cancer metastatic colonization, incidences of metastasis, accurately quantify the burden of metastasis, and test the role of NOTCH1 receptor on prostate cancer metastatic colonization and homing to distant sites. The metastasis model presented here is established by intracardiac injection of control human prostate cancer cells and NOTCH1 downregulated cells. The cells are engineered to express both red fluorescent protein (RFP) and luciferase. In this model, whole body bioluminescence imaging, high-resolution, and quantitative fluorescence imaging are utilized for quantitative assessment of metastatic colonization and metastasis burden. Further, histopathology analyses of diverse metastatic organs are performed. This model is a powerful and versatile tool to investigate the mechanisms underlying the function of NOTCH receptors in metastatic colonization in prostate cancer.
Collapse
Affiliation(s)
- Shiqin Liu
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA, USA
| | - En-Chi Hsu
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA, USA
| | - Michelle Shen
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA, USA
| | - Merve Aslan
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA, USA
| | - Tanya Stoyanova
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA, USA.
| |
Collapse
|
9
|
Jin L, Ma X, Lei X, Tong J, Wang R. Cyclophosphamide inhibits Pax5 methylation to regulate the growth of retinoblastoma via the Notch1 pathway. Hum Exp Toxicol 2021; 40:S497-S508. [PMID: 34658283 DOI: 10.1177/09603271211051601] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Retinoblastoma (Rb) is the most common intraocular malignant tumor in infants. Here, we investigated the function and mechanism of cyclophosphamide (CTX) in the development of Rb. Real-time quantitative polymerase chain reaction (RT-qPCR) results showed that paired box protein 5 (Pax5) expression was down-regulated in Rb tissues and cell lines. Methylation-specific PCR (MSP) results showed that the methylation level of Pax5 was up-regulated in Rb. After treatment with CTX, the Pax5 expression in Rb cell lines was increased significantly. The methylation of Pax5 and the expression of DNA methyltransferases (DNMTs) were down-regulated in the CTX group. Cyclophosphamide inhibited cell proliferation, migration, and invasion, promoted cell apoptosis via the Notch1 pathway. DNA methyltransferase inhibitor SGI-1027 had synergistic effects with CTX. Paired box protein 5 siRNA was transfected into Y79 cells treated with CTX. The expression of DNMTs, Pax5, the Notch1 pathway and apoptosis marker protein was detected by Western blotting, and changes in cell behavior were detected, respectively. Results showed that knockdown of Pax5 reversed the effects of CTX. Moreover, the Notch1 activator Valproic acid (VPA) abolished the inhibitory effects of CTX on Rb development. Moreover, CTX inhibited tumor growth in nude mice. These findings demonstrated that CTX up-regulated Pax5 expression by down-regulating DNMTs expression, and then inhibited the Notch1 signaling pathway activation and Rb growth.
Collapse
Affiliation(s)
- Lan Jin
- Department of Ophthalmology, Xi'an Hospital of Traditional Chinese Medicine, Xi'an, China
| | - Xiaojie Ma
- Department of Ophthalmology, Xi'an Hospital of Traditional Chinese Medicine, Xi'an, China
| | - Xiaoqin Lei
- Department of Ophthalmology, Xi'an No. 4 Hospital, Xi'an, China
| | - Jing'an Tong
- Department of Ophthalmology, 107652The First Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Runsheng Wang
- Department of Ophthalmology, Xi'an No. 4 Hospital, Xi'an, China
| |
Collapse
|
10
|
Guan Y, Yang YJ, Nagarajan P, Ge Y. Transcriptional and signalling regulation of skin epithelial stem cells in homeostasis, wounds and cancer. Exp Dermatol 2020; 30:529-545. [PMID: 33249665 DOI: 10.1111/exd.14247] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 10/10/2020] [Accepted: 11/13/2020] [Indexed: 02/06/2023]
Abstract
The epidermis and skin appendages are maintained by their resident epithelial stem cells, which undergo long-term self-renewal and multilineage differentiation. Upon injury, stem cells are activated to mediate re-epithelialization and restore tissue function. During this process, they often mount lineage plasticity and expand their fates in response to damage signals. Stem cell function is tightly controlled by transcription machineries and signalling transductions, many of which derail in degenerative, inflammatory and malignant dermatologic diseases. Here, by describing both well-characterized and newly emerged pathways, we discuss the transcriptional and signalling mechanisms governing skin epithelial homeostasis, wound repair and squamous cancer. Throughout, we highlight common themes underscoring epithelial stem cell plasticity and tissue-level crosstalk in the context of skin physiology and pathology.
Collapse
Affiliation(s)
- Yinglu Guan
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Youn Joo Yang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Priyadharsini Nagarajan
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yejing Ge
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
11
|
Dong Y, Li J, Liu R, Zhao Z, Wang S, Cui K. Musashi1 expression is negatively correlated with numb expression in brain metastases. Medicine (Baltimore) 2020; 99:e22000. [PMID: 33120728 PMCID: PMC7581019 DOI: 10.1097/md.0000000000022000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 07/25/2020] [Accepted: 07/31/2020] [Indexed: 11/25/2022] Open
Abstract
The expression of tumor stem cell markers musashi1 (msi1) and numb in brain metastases were detected to explore their roles in the development of brain metastases.A total of 51 cases of brain metastasis, 29 cases of primary tumor and 15 cases of normal brain tissue were selected. Immunohistochemistry and reverse transcription polymerase chain reaction (RT-PCR) were used to detect msi1 and numb expression at the protein and mRNA levels. Correlation between msi1 and numb in brain metastases were evaluated.Immunohistochemistry and RT-PCR showed that no significant difference in the expression of msi1 and numb between brain metastases and primary tumors was observed (P > .05); the expression of msi1 and numb in brain metastases was significantly higher than that in normal brain tissues (P < .05); and the expression of msi1 and numb in primary tumors was significantly higher than that in normal brain tissues (P < .05). In general, the expression of msi1 gene was negatively correlated with the expression of numb at mRNA level by Pearson correlation analysis (r = -0.345, P < .05). Additionally, the expression of msi1 and numb in brain metastases was not related to gender, age, and tissue origin (P > .05).Msi1 is highly expressed in brain metastases and primary tumors, while numb is lowly expressed in brain metastases and primary tumors; msi1 and numb are negatively correlated in brain metastases, suggesting that msi1 and numb may have regulatory mechanisms in the development of brain metastases.
Collapse
|
12
|
Xu W, Xu YN, Zhang X, Xu Y, Jian X, Chen JM, Chen GF, Zhang H, Liu P, Mu YP. Hepatic stem cell Numb gene is a potential target of Huang Qi Decoction against cholestatic liver fibrosis. Sci Rep 2020; 10:17486. [PMID: 33060633 PMCID: PMC7566460 DOI: 10.1038/s41598-020-74324-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 09/30/2020] [Indexed: 12/13/2022] Open
Abstract
Numb is a negative regulator of Notch signal pathway. Previous study has demonstrated that Notch signal pathway activation is required for hepatic progenitor cell (HPC) differentiating into cholangiocytes in cholestatic liver fibrosis (CLF), and Huang Qi Decoction (HQD) could prevent CLF through inhibition of the Notch signal pathway. However, the role of Numb in HQD against CLF is yet unclear. Thus, CLF rats transplanted into rat bone marrow-derived mesenchymal stem cells with knocked down Numb gene (BMSCNumb-KD) were treated with HQD. Simultaneously, Numb gene knockdown was also performed in WB-F344 cell line and then treated with refined HQD in vitro. In vivo study revealed that liver fibrosis was inhibited by HQD plus BMSCNumb-KD treatment, while Hyp content in liver tissue, the gene and protein expression of α-SMA, gene expression of Col I, TNF-α, and TGF-β1 were increased compared to that in HQD group. Furthermore, Notch signal pathway was inhibited by HQD plus BMSCNumb-KD, while the protein expression of Numb was decreased and RBP-Jκ and Hes1 was increased compared to that in HQD group. In vitro, HQD reduced the differentiation of WB-F344 cells into cholangiocyte phenotype, while this effect was attenuated after Numb-knockdown. This study highlights that the absence of hepatic stem cell Numb gene decreases effect of HQD against CLF, which give rise the conclusion that Numb might be a potential target for HQD against CLF.
Collapse
Affiliation(s)
- Wen Xu
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM); Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, Shanghai University of TCM, 528, Zhangheng Road, Pudong district, Shanghai, 201203, People's Republic of China.,Shanghai Key Laboratory of TCM, Shanghai, People's Republic of China
| | - Yan-Nan Xu
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM); Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, Shanghai University of TCM, 528, Zhangheng Road, Pudong district, Shanghai, 201203, People's Republic of China.,Shanghai Key Laboratory of TCM, Shanghai, People's Republic of China
| | - Xu Zhang
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM); Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, Shanghai University of TCM, 528, Zhangheng Road, Pudong district, Shanghai, 201203, People's Republic of China.,Shanghai Key Laboratory of TCM, Shanghai, People's Republic of China
| | - Ying Xu
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM); Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, Shanghai University of TCM, 528, Zhangheng Road, Pudong district, Shanghai, 201203, People's Republic of China.,Shanghai Key Laboratory of TCM, Shanghai, People's Republic of China
| | - Xun Jian
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM); Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, Shanghai University of TCM, 528, Zhangheng Road, Pudong district, Shanghai, 201203, People's Republic of China.,Shanghai Key Laboratory of TCM, Shanghai, People's Republic of China
| | - Jia-Mei Chen
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM); Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, Shanghai University of TCM, 528, Zhangheng Road, Pudong district, Shanghai, 201203, People's Republic of China.,Shanghai Key Laboratory of TCM, Shanghai, People's Republic of China
| | - Gao-Feng Chen
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM); Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, Shanghai University of TCM, 528, Zhangheng Road, Pudong district, Shanghai, 201203, People's Republic of China.,Shanghai Key Laboratory of TCM, Shanghai, People's Republic of China
| | - Hua Zhang
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM); Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, Shanghai University of TCM, 528, Zhangheng Road, Pudong district, Shanghai, 201203, People's Republic of China.,Shanghai Key Laboratory of TCM, Shanghai, People's Republic of China
| | - Ping Liu
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM); Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, Shanghai University of TCM, 528, Zhangheng Road, Pudong district, Shanghai, 201203, People's Republic of China. .,Shanghai Key Laboratory of TCM, Shanghai, People's Republic of China. .,E-Institute of Shanghai Municipal Education Commission, Shanghai University of TCM, Shanghai, People's Republic of China.
| | - Yong-Ping Mu
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM); Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, Shanghai University of TCM, 528, Zhangheng Road, Pudong district, Shanghai, 201203, People's Republic of China. .,Shanghai Key Laboratory of TCM, Shanghai, People's Republic of China.
| |
Collapse
|
13
|
Li L, Gao F, Fan L, Su C, Liang X, Gong C. Disorders of Sex Development in Individuals Harbouring MAMLD1 Variants: WES and Interactome Evidence of Oligogenic Inheritance. Front Endocrinol (Lausanne) 2020; 11:582516. [PMID: 33424767 PMCID: PMC7786837 DOI: 10.3389/fendo.2020.582516] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022] Open
Abstract
Mastermind-like domain-containing 1 (MAMLD1) has been shown to play an important role in the process of sexual development and is associated with 46,XY disorders of sex development (DSDs). However, the causative role of MAMLD1 variations in DSDs remains disputable. In this study, we have described a clinical series on children from unrelated families with 46,XY DSD harbouring MAMLD1 variants. Whole exome sequencing (WES) was performed for each patient. WES data were filtered using common tools and disease customisation algorithms, including comparison against lists of known and candidate MAMLD1-related and DSD-related genes. Lastly, we investigated the hypothesis that MAMLD1-related DSD may follow an oligogenic mode of inheritance. Forty-three potentially deleterious/candidate variants of 18 genes (RET, CDH23, MYO7A, NOTCH2, MAML1, MAML2, CYP1A1, WNT9B, GLI2, GLI3, MAML3, WNT9A, FRAS1, PIK3R3, FREM2, PTPN11, EVC, and FLNA) were identified, which may have contributed to the patient phenotypes. MYO7A was the most commonly identified gene. Specific gene combinations were also identified. In the interactome analysis, MAMLD1 exhibited direct connection with MAML1/2/3 and NOTCH1/2. Through NOTCH1/2, the following eight genes were shown to be associated with MAMLD1:WNT9A/9B, GLI2/3, RET, FLNA, PTPN11, and EYA1. Our findings provide further evidence that individuals with MAMLD1-related 46,XY DSD could carry two or more variants of known DSD-related genes, and the phenotypic outcome of affected individuals might be determined by multiple genes.
Collapse
|
14
|
Xie J, Lin LS, Huang XY, Gan RH, Ding LC, Su BH, Zhao Y, Lu YG, Zheng DL. The NOTCH1-HEY1 pathway regulates self-renewal and epithelial-mesenchymal transition of salivary adenoid cystic carcinoma cells. Int J Biol Sci 2020; 16:598-610. [PMID: 32025208 PMCID: PMC6990919 DOI: 10.7150/ijbs.36407] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 11/12/2019] [Indexed: 12/13/2022] Open
Abstract
Our previous study demonstrated a close relationship between the NOTCH signaling pathway and salivary adenoid cystic carcinoma (SACC). Its receptor gene, NOTCH1, and its downstream gene, HES1, contribute to the proliferation, invasion and metastasis of SACC. Accumulating evidence supports HEY1 as another effector of the signaling pathway. The purpose of this study was to explore the effects of the NOTCH1-HEY1 pathway on the proliferation, invasion and metastasis of SACC cells. Our results verified that HEY1 is a specific molecular target of the NOTCH signaling pathway in SACC cells and that its expression in carcinoma is much higher than that in paracarcinoma tissues. The expression of NOTCH1 and HEY1 are positively correlated in the salivary adenoid cystic carcinoma tissues. NOTCH1 is significantly related to the activation of HEY1 in SACC, and that HEY1 reciprocally regulates NOTCH1 expression in SACC. HEY1 promotes cell proliferation and spheroid formation and inhibits cell apoptosis in vitro. In addition, HEY1 enhances the tumorigenicity of SACC in vivo. Furthermore, HEY1 increases cell invasion and metastasis by driving the expression of epithelial-mesenchymal transition (EMT)-related genes and MMPs. The results of this study indicate that the NOTCH1-HEY1 pathway is specifically upregulated in SACC and promotes cell proliferation, self-renewal, invasion, metastasis and the expression of EMT-related genes and MMPs. Our findings suggest that a NOTCH1-HEY1 pathway inhibitor might therefore have potential therapeutic applications in treating SACC patients by inhibiting cancer cell growth and metastasis.
Collapse
Affiliation(s)
- Jing Xie
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, 246 Yang Qiao Middle Road, Fuzhou 350000, China.,Key laboratory of Stomatology of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, 88 Jiaotong Rd, Fuzhou 350004, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, 1 Xue Yuan Road, University Town, Fuzhou 350122, China
| | - Li-Song Lin
- Department of Oral and Maxillofacial Surgery, Affiliated First Hospital of Fujian Medical University, 20 Cha Zhong Road, Fuzhou 350005, China
| | - Xiao-Yu Huang
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, 246 Yang Qiao Middle Road, Fuzhou 350000, China.,Key laboratory of Stomatology of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, 88 Jiaotong Rd, Fuzhou 350004, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, 1 Xue Yuan Road, University Town, Fuzhou 350122, China
| | - Rui-Huan Gan
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, 246 Yang Qiao Middle Road, Fuzhou 350000, China.,Key laboratory of Stomatology of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, 88 Jiaotong Rd, Fuzhou 350004, China
| | - Lin-Can Ding
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, 246 Yang Qiao Middle Road, Fuzhou 350000, China
| | - Bo-Hua Su
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, 246 Yang Qiao Middle Road, Fuzhou 350000, China
| | - Yong Zhao
- Key laboratory of Stomatology of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, 88 Jiaotong Rd, Fuzhou 350004, China.,Department of pathology, School and Hospital of Stomatology, Fujian Medical University, 246 Yang Qiao Middle Road, Fuzhou 350000, China
| | - You-Guang Lu
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, 246 Yang Qiao Middle Road, Fuzhou 350000, China.,Key laboratory of Stomatology of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, 88 Jiaotong Rd, Fuzhou 350004, China
| | - Da-Li Zheng
- Key laboratory of Stomatology of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, 88 Jiaotong Rd, Fuzhou 350004, China
| |
Collapse
|
15
|
Cheung PF, Neff F, Neander C, Bazarna A, Savvatakis K, Liffers ST, Althoff K, Lee CL, Moding EJ, Kirsch DG, Saur D, Bazhin AV, Trajkovic-Arsic M, Heikenwalder MF, Siveke JT. Notch-Induced Myeloid Reprogramming in Spontaneous Pancreatic Ductal Adenocarcinoma by Dual Genetic Targeting. Cancer Res 2018; 78:4997-5010. [PMID: 29844119 DOI: 10.1158/0008-5472.can-18-0052] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 04/20/2018] [Accepted: 05/22/2018] [Indexed: 11/16/2022]
Abstract
Despite advances in our understanding of the genetics of pancreatic ductal adenocarcinoma (PDAC), the efficacy of therapeutic regimens targeting aberrant signaling pathways remains highly limited. Therapeutic strategies are greatly hampered by the extensive desmoplasia that comprises heterogeneous cell populations. Notch signaling is a contentious pathway exerting opposite roles in tumorigenesis depending on cellular context. Advanced model systems are needed to gain more insights into complex signaling in the multilayered tumor microenvironment. In this study, we employed a dual recombinase-based in vivo strategy to modulate Notch signaling specifically in myeloid cells to dissect the tumorigenic role of Notch in PDAC stroma. Pancreas-specific KrasG12D activation and loss of Tp53 was induced using a Pdx1-Flp transgene, whereas Notch signaling was genetically targeted using a myeloid-targeting Lyz2-Cre strain for either activation of Notch2-IC or deletion of Rbpj. Myeloid-specific Notch activation significantly decreased tumor infiltration by protumorigenic M2 macrophages in spontaneous endogenous PDAC, which translated into significant survival benefit. Further characterization revealed upregulated antigen presentation and cytotoxic T effector phenotype upon Notch-induced M2 reduction. This approach is the first proof of concept for genetic targeting and reprogramming of myeloid cells in a complex disease model of PDAC and provides evidence for a regulatory role of Notch signaling in intratumoral immune phenotypes.Significance: This study provides insight into the role of myeloid-dependent NOTCH signaling in PDAC and accentuates the need to dissect differential roles of signaling pathways in different cellular components within the tumor microenvironment. Cancer Res; 78(17); 4997-5010. ©2018 AACR.
Collapse
Affiliation(s)
- Phyllis F Cheung
- Division of Solid Tumor Translational Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany.,German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Florian Neff
- Division of Solid Tumor Translational Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany.,German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Christian Neander
- Division of Solid Tumor Translational Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany.,German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Anna Bazarna
- Division of Solid Tumor Translational Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany.,German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Konstantinos Savvatakis
- Division of Solid Tumor Translational Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany.,German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Sven-Thorsten Liffers
- Division of Solid Tumor Translational Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany.,German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Kristina Althoff
- Division of Solid Tumor Translational Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany.,German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Chang-Lung Lee
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| | - Everett J Moding
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| | - David G Kirsch
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| | - Dieter Saur
- German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany.,Medical Department, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Alexandr V Bazhin
- Department of General, Visceral and Transplant Surgery, Ludwig-Maximilians University, Munich, Germany.,German Caner Consortium (DKTK), Partner Site Munich, Germany
| | - Marija Trajkovic-Arsic
- Division of Solid Tumor Translational Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany.,German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany
| | | | - Jens T Siveke
- Division of Solid Tumor Translational Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany. .,German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany.,Medical Department, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| |
Collapse
|
16
|
Manic Fringe deficiency imposes Jagged1 addiction to intestinal tumor cells. Nat Commun 2018; 9:2992. [PMID: 30065304 PMCID: PMC6068201 DOI: 10.1038/s41467-018-05385-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 06/29/2018] [Indexed: 12/26/2022] Open
Abstract
Delta ligands regulate Notch signaling in normal intestinal stem cells, while Jagged1 activates Notch in intestinal adenomas carrying active β-catenin. We used the ApcMin/+ mouse model, tumor spheroid cultures, and patient-derived orthoxenografts to address this divergent ligand-dependent Notch function and its implication in disease. We found that intestinal-specific Jag1 deletion or antibody targeting Jag1 prevents tumor initiation in mice. Addiction to Jag1 is concomitant with the absence of Manic Fringe (MFNG) in adenoma cells, and its ectopic expression reverts Jag1 dependence. In 239 human colorectal cancer patient samples, MFNG imposes a negative correlation between Jag1 and Notch, being high Jag1 in the absence of MFNG predictive of poor prognosis. Jag1 antibody treatment reduces patient-derived tumor orthoxenograft growth without affecting normal intestinal mucosa. Our data provide an explanation to Jag1 dependence in cancer, and reveal that Jag1-Notch1 interference provides therapeutic benefit in a subset of colorectal cancer and FAP syndrome patients.
Collapse
|
17
|
Aung KL, El-Khoueiry AB, Gelmon K, Tran B, Bajaj G, He B, Chen T, Zhu L, Poojary S, Basak S, Qi Z, Spreafico A, Fischer BS, Desai J. A multi-arm phase I dose escalating study of an oral NOTCH inhibitor BMS-986115 in patients with advanced solid tumours. Invest New Drugs 2018; 36:1026-1036. [PMID: 29637471 DOI: 10.1007/s10637-018-0597-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 03/26/2018] [Indexed: 10/17/2022]
Abstract
Background Inhibiting Notch is a promising anti-cancer strategy as it plays a critical role in cancer stem cells maintenance and tumour angiogenesis. BMS-986115 is an orally active, selective inhibitor of gamma-secretase mediated Notch signalling. Method Two dose escalation schedules (Arm-A continuous daily schedule and Arm-B intermittent 2 times weekly schedule) of BMS-986115 were evaluated in advanced solid tumour patients. The primary objective was to establish the safety, tolerability and Maximum Tolerated Dose (MTD) of BMS-986115. Results Thirty six patients (24 in Arm A and 12 in Arm B) were treated. The most frequent treatment related adverse advents were diarrhoea (72%), hypophosphataemia (64%), and nausea (61%). The MTD was 1.5 mg daily in Arm A but not established in Arm B. Four patients in Arm A and 2 in Arm B experienced dose limiting toxicities (grade 3 nausea, diarrhoea, pruritus/urticaria and ileus). BMS-986115 showed dose related increase in exposure within the dose range tested. Target inhibition of Notch pathway related genes was observed. Three patients in Arm A and 2 in Arm B achieved stable disease for more than 6 months. Conclusion The daily oral dosing of BMS-986115 is safe and tolerable with biological activity demonstrated by continuous target engagement and Notch signalling inhibition.
Collapse
Affiliation(s)
- Kyaw L Aung
- Drug Development Program, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, M5G 2M9, Canada
| | - Anthony B El-Khoueiry
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, 900333, USA
| | - Karen Gelmon
- British Columbia Cancer Agency, Vancouver, British Columbia, V5Z 4E6, Canada
| | - Ben Tran
- Peter MacCallum Cancer Centre and Royal Melbourne Hospital, University of Melbourne, 305 Grattan St, Melbourne, VIC, 3000, Australia
| | - Gaurav Bajaj
- Clinical Pharmacology & Pharmacometrics, Bristol-Myers Squibb, Lawrenceville, NJ, 08648, USA
| | - Bing He
- Clinical Pharmacology & Pharmacometrics, Bristol-Myers Squibb, Lawrenceville, NJ, 08648, USA
| | - Tian Chen
- Global Biometric Sciences, Bristol-Myers Squibb, Princeton, NJ, 08540, USA
| | - Lili Zhu
- Global Biometric Sciences, Bristol-Myers Squibb, Princeton, NJ, 08540, USA
| | - Sharath Poojary
- Biocon BMS R&D Centre, Syngene International Ltd, Bangalore, 560 099, India
| | - Shashwati Basak
- Biocon BMS R&D Centre, Syngene International Ltd, Bangalore, 560 099, India
| | - Zhenhao Qi
- Clinical Genomics and Genetics, Translational Medicine, Bristol-Myers Squibb, Princeton, NJ, 08540, USA
| | - Anna Spreafico
- Drug Development Program, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, M5G 2M9, Canada
| | - Bruce S Fischer
- Oncology Early Clinical Development, Bristol-Myers Squibb, Lawrenceville, NJ, 08648, USA
| | - Jayesh Desai
- Peter MacCallum Cancer Centre and Royal Melbourne Hospital, University of Melbourne, 305 Grattan St, Melbourne, VIC, 3000, Australia.
| |
Collapse
|
18
|
Skarmoutsou E, Bevelacqua V, D' Amico F, Russo A, Spandidos DA, Scalisi A, Malaponte G, Guarneri C. FOXP3 expression is modulated by TGF‑β1/NOTCH1 pathway in human melanoma. Int J Mol Med 2018; 42:392-404. [PMID: 29620159 PMCID: PMC5979787 DOI: 10.3892/ijmm.2018.3618] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 04/04/2018] [Indexed: 12/18/2022] Open
Abstract
Forkhead box protein 3 (FOXP3) transcription factor is expressed by immune cells and several human cancers and is associated with tumor aggressiveness and unfavorable clinical outcomes. NOTCH and transforming growth factor-β (TGF-β) protumorigenic effects are mediated by FOXP3 expression in several cancer models; however, their interaction and role in melanoma is unknown. We investigated TGF-β-induced FOXP3 gene expression during NOTCH1 signaling inactivation. Primary (WM35) and metastatic melanoma (A375 and A2058) cell lines and normal melanocytes (NHEM) were used. FOXP3 subcellular distribution was evaluated by immuno cytochemical analysis. Gene expression levels were assessed by reverse transcription-quantitative polymerase chain reaction. Protein levels were assessed by western blot analysis. The γ-secretase inhibitor (GSI) was used for NOTCH1 inhibition and recombinant human (rh)TGF-β was used for melanoma cell stimulation. Cell proliferation and viability were respectively assessed by MTT and Trypan blue dye assays. FOXP3 mRNA and protein levels were progressively higher in WM35, A375 and A2058 cell lines compared to NHEM and their levels were further increased after stimulation with rh-TGF-β. TGF-β-mediated FOXP3 expression was mediated by NOTCH1 signaling. Inhibition of NOTCH1 with concomitant rh-TGF-β stimulation determined the reduction in gene expression and protein level of FOXP3. Finally, melanoma cell line proliferation and viability were reduced by NOTCH1 inhibition. The results show that nn increase in FOXP3 expression in metastatic melanoma cell lines is a potential marker of tumor aggressiveness and metastasis. NOTCH1 is a central mediator of TGF-β-mediated FOXP3 expression and NOTCH1 inhibition produces a significant reduction of melanoma cell proliferation and viability.
Collapse
Affiliation(s)
- Eva Skarmoutsou
- Department of Biomedical and Biotechnological Science, University of Catania, 95124 Catania, Italy
| | - Valentina Bevelacqua
- Department of Biomedical and Biotechnological Science, University of Catania, 95124 Catania, Italy
| | - Fabio D' Amico
- Department of Biomedical and Biotechnological Science, University of Catania, 95124 Catania, Italy
| | - Angela Russo
- Department of Biomedical and Biotechnological Science, University of Catania, 95124 Catania, Italy
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 71003 Heraklion, Crete, Greece
| | - Aurora Scalisi
- Unit of Oncologic Diseases, ASP‑Catania, 95100 Catania, Italy
| | - Grazia Malaponte
- Research Unit of the Catania Section of the Italian League Against Cancer, 95122 Catania, Italy
| | - Claudio Guarneri
- Department of Clinical and Experimental Medicine, Section of Dermatology, University of Messina, 98122 Messina, Italy
| |
Collapse
|
19
|
Salazar JL, Yamamoto S. Integration of Drosophila and Human Genetics to Understand Notch Signaling Related Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1066:141-185. [PMID: 30030826 PMCID: PMC6233323 DOI: 10.1007/978-3-319-89512-3_8] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Notch signaling research dates back to more than one hundred years, beginning with the identification of the Notch mutant in the fruit fly Drosophila melanogaster. Since then, research on Notch and related genes in flies has laid the foundation of what we now know as the Notch signaling pathway. In the 1990s, basic biological and biochemical studies of Notch signaling components in mammalian systems, as well as identification of rare mutations in Notch signaling pathway genes in human patients with rare Mendelian diseases or cancer, increased the significance of this pathway in human biology and medicine. In the 21st century, Drosophila and other genetic model organisms continue to play a leading role in understanding basic Notch biology. Furthermore, these model organisms can be used in a translational manner to study underlying mechanisms of Notch-related human diseases and to investigate the function of novel disease associated genes and variants. In this chapter, we first briefly review the major contributions of Drosophila to Notch signaling research, discussing the similarities and differences between the fly and human pathways. Next, we introduce several biological contexts in Drosophila in which Notch signaling has been extensively characterized. Finally, we discuss a number of genetic diseases caused by mutations in genes in the Notch signaling pathway in humans and we expand on how Drosophila can be used to study rare genetic variants associated with these and novel disorders. By combining modern genomics and state-of-the art technologies, Drosophila research is continuing to reveal exciting biology that sheds light onto mechanisms of disease.
Collapse
Affiliation(s)
- Jose L Salazar
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, TX, USA
| | - Shinya Yamamoto
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, TX, USA.
- Program in Developmental Biology, BCM, Houston, TX, USA.
- Department of Neuroscience, BCM, Houston, TX, USA.
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.
| |
Collapse
|
20
|
The Notch Interactome: Complexity in Signaling Circuitry. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1066:125-140. [PMID: 30030825 DOI: 10.1007/978-3-319-89512-3_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The Notch pathway controls a very broad spectrum of cell fates in metazoans during development, influencing proliferation, differentiation and cell death. Given its central role in normal development and homeostasis, misregulation of Notch signals can lead to various disorders including cancer. How the Notch pathway mediates such pleiotropic and differential effects is of fundamental importance. It is becoming increasingly clear through a number of large-scale genetic and proteomic studies that Notch interacts with a staggeringly large number of other genes and pathways in a context-dependent, complex, and highly regulated network, which determines the ultimate biological outcome. How best to interpret and analyze the continuously increasing wealth of data on Notch interactors remains a challenge. Here we review the current state of genetic and proteomic data related to the Notch interactome.
Collapse
|
21
|
Gan RH, Wei H, Xie J, Zheng DP, Luo EL, Huang XY, Xie J, Zhao Y, Ding LC, Su BH, Lin LS, Zheng DL, Lu YG. Notch1 regulates tongue cancer cells proliferation, apoptosis and invasion. Cell Cycle 2017; 17:216-224. [PMID: 29117785 DOI: 10.1080/15384101.2017.1395534] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVES Notch1 regulates tumor biology in a complex, context-dependent manner. The roles of Notch1 in tongue cancer are still controversial. The aim of this study is to investigate the roles of Notch1 in tongue cancer. MATERIALS AND METHODS The expression of Notch1 was tested between tongue cancer and normal samples by using immunohistochemistry. Tongue cancer cells were transfected with siRNA or plasmid, respectively. Cell proliferation, apoptosis, migration and invasion ability were tested in appropriate ways. The subcutaneous tumor model was established to observe the tumor growth. RESULTS Notch1 was upregulated in tongue carcinoma tissues and the expression of Notch1 was related with tumor stage and differentiation. Overexpression of Notch1 could increase tongue cancer cells proliferation, invasion and migration. But inhibited the expression of Notch1 could decrease cells proliferation, invasion and migration and promote cell apoptosis in vitro and in vivo. CONCLUSION Our results prove that the oncogenic role of Notch1 in tongue cancer and provide the direction of targeted therapy of tongue cancer.
Collapse
Affiliation(s)
- Rui-Huan Gan
- a Department of Preventive Dentistry , Affiliated Stomatological Hospital, Fujian Medical University , 246 Yang Qiao Middle Road, Fuzhou 350000 , China.,b Key laboratory of stomatology , Fujian Province University , 88 Jiao Tong Road, Fuzhou 350004 , China
| | - Hua Wei
- c The School of Basic Medical Science , Fujian Medical University , 1 Xue Yuan Road, Shang Jie Town, Min Hou County, Fuzhou 350000 , China
| | - Jing Xie
- a Department of Preventive Dentistry , Affiliated Stomatological Hospital, Fujian Medical University , 246 Yang Qiao Middle Road, Fuzhou 350000 , China.,b Key laboratory of stomatology , Fujian Province University , 88 Jiao Tong Road, Fuzhou 350004 , China
| | - Dan-Ping Zheng
- a Department of Preventive Dentistry , Affiliated Stomatological Hospital, Fujian Medical University , 246 Yang Qiao Middle Road, Fuzhou 350000 , China.,b Key laboratory of stomatology , Fujian Province University , 88 Jiao Tong Road, Fuzhou 350004 , China
| | - Er-Ling Luo
- a Department of Preventive Dentistry , Affiliated Stomatological Hospital, Fujian Medical University , 246 Yang Qiao Middle Road, Fuzhou 350000 , China.,b Key laboratory of stomatology , Fujian Province University , 88 Jiao Tong Road, Fuzhou 350004 , China
| | - Xiao-Yu Huang
- a Department of Preventive Dentistry , Affiliated Stomatological Hospital, Fujian Medical University , 246 Yang Qiao Middle Road, Fuzhou 350000 , China.,b Key laboratory of stomatology , Fujian Province University , 88 Jiao Tong Road, Fuzhou 350004 , China
| | - Jian Xie
- a Department of Preventive Dentistry , Affiliated Stomatological Hospital, Fujian Medical University , 246 Yang Qiao Middle Road, Fuzhou 350000 , China.,b Key laboratory of stomatology , Fujian Province University , 88 Jiao Tong Road, Fuzhou 350004 , China
| | - Yong Zhao
- b Key laboratory of stomatology , Fujian Province University , 88 Jiao Tong Road, Fuzhou 350004 , China.,d Department of Pathology , Affiliated Stomatological Hospital, Fujian Medical University , 246 Yang Qiao Middle Road, Fuzhou 350000 , China
| | - Lin-Can Ding
- a Department of Preventive Dentistry , Affiliated Stomatological Hospital, Fujian Medical University , 246 Yang Qiao Middle Road, Fuzhou 350000 , China.,b Key laboratory of stomatology , Fujian Province University , 88 Jiao Tong Road, Fuzhou 350004 , China
| | - Bo-Hua Su
- a Department of Preventive Dentistry , Affiliated Stomatological Hospital, Fujian Medical University , 246 Yang Qiao Middle Road, Fuzhou 350000 , China.,b Key laboratory of stomatology , Fujian Province University , 88 Jiao Tong Road, Fuzhou 350004 , China
| | - Li-Song Lin
- e Department of Oral and Maxillofacial Surgery , Affiliated First Hospital of Fujian Medical University , 20 Cha Zhong Road, Fuzhou 350000 , China
| | - Da-Li Zheng
- f Department of Molecular Medicine , University of South Florida , 19202 Bruce B Downs Blvd, Tampa , FL 33613 , United States
| | - You-Guang Lu
- a Department of Preventive Dentistry , Affiliated Stomatological Hospital, Fujian Medical University , 246 Yang Qiao Middle Road, Fuzhou 350000 , China.,b Key laboratory of stomatology , Fujian Province University , 88 Jiao Tong Road, Fuzhou 350004 , China
| |
Collapse
|
22
|
Thiel VN, Giaimo BD, Schwarz P, Soller K, Vas V, Bartkuhn M, Blätte TJ, Döhner K, Bullinger L, Borggrefe T, Geiger H, Oswald F. Heterodimerization of AML1/ETO with CBFβ is required for leukemogenesis but not for myeloproliferation. Leukemia 2017; 31:2491-2502. [PMID: 28360416 PMCID: PMC5668496 DOI: 10.1038/leu.2017.105] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 02/18/2017] [Accepted: 03/13/2017] [Indexed: 02/07/2023]
Abstract
The AML1/Runx1 transcription factor and its heterodimerization partner CBFβ are essential regulators of myeloid differentiation. The chromosomal translocation t(8;21), fusing the DNA binding domain of AML1 to the corepressor eight-twenty-one (ETO), is frequently associated with acute myeloid leukemia and generates the AML1/ETO (AE) fusion protein. AE represses target genes usually activated by AML1 and also affects the endogenous repressive function of ETO at Notch target genes. In order to analyze the contribution of CBFβ in AE-mediated leukemogenesis and deregulation of Notch target genes, we introduced two point mutations in a leukemia-initiating version of AE in mice, called AE9a, that disrupt the AML1/CBFβ interaction (AE9aNT). We report that the AE9a/CBFβ interaction is not required for the AE9a-mediated aberrant expression of AML1 target genes, while upregulation/derepression of Notch target genes does require the interaction with CBFβ. Using retroviral transduction to express AE9a in murine adult bone marrow-derived hematopoietic progenitors, we observed that both AE9a and AE9aNT lead to increased myeloproliferation in vivo. However, both development of leukemia and long-term replating capacity are only observed with AE9a but not with AE9aNT. Thus, deregulation of both AML1 and Notch target genes is required for the development of AE9a-driven leukemia.
Collapse
Affiliation(s)
- V N Thiel
- University Medical Center Ulm, Center for Internal Medicine, Department of Internal Medicine I, University of Ulm, Ulm, Germany
| | - B D Giaimo
- Institute of Biochemistry, University of Giessen, Giessen, Germany
| | - P Schwarz
- University Medical Center Ulm, Center for Internal Medicine, Department of Internal Medicine I, University of Ulm, Ulm, Germany
| | - K Soller
- Institute of Molecular Medicine, University of Ulm, Ulm, Germany
| | - V Vas
- Institute of Molecular Medicine, University of Ulm, Ulm, Germany
| | - M Bartkuhn
- Institute for Genetics, University of Giessen, Giessen, Germany
| | - T J Blätte
- University Medical Center Ulm, Center for Internal Medicine, Department of Internal Medicine III, University of Ulm, Ulm, Germany
| | - K Döhner
- University Medical Center Ulm, Center for Internal Medicine, Department of Internal Medicine III, University of Ulm, Ulm, Germany
| | - L Bullinger
- University Medical Center Ulm, Center for Internal Medicine, Department of Internal Medicine III, University of Ulm, Ulm, Germany
| | - T Borggrefe
- Institute of Biochemistry, University of Giessen, Giessen, Germany
| | - H Geiger
- Institute of Molecular Medicine, University of Ulm, Ulm, Germany
- Division of Experimental Hematology and Cancer Biology, CCHMC, Cincinnati, OH, USA
| | - F Oswald
- University Medical Center Ulm, Center for Internal Medicine, Department of Internal Medicine I, University of Ulm, Ulm, Germany
| |
Collapse
|
23
|
Abstract
MicroRNAs have broad roles in tumorigenesis and cell differentiation through regulation of target genes. Notch signaling also controls cell differentiation and tumorigenesis. However, the mechanisms through which Notch mediates microRNA expression are still unclear. In this study, we aimed to identify microRNAs regulated by Notch signaling. Our analysis found that microRNA-449a (miR-449a) was indirectly regulated by Notch signaling. Although miR-449a-deficient mice did not show any Notch-dependent defects in immune cell development, treatment of miR-449a-deficient mice with azoxymethane (AOM) or dextran sodium sulfate (DSS) increased the numbers and sizes of colon tumors. These effects were associated with an increase in intestinal epithelial cell proliferation following AOM/DSS treatment. In patients with colon cancer, miR-449a expression was inversely correlated with disease-free survival and histological scores and was positively correlated with the expression of MLH1 for which loss-of function mutations have been shown to be involved in colon cancer. Colon tissues of miR-449a-deficient mice showed reduced Mlh1 expression compared with those of wild-type mice. Thus, these data suggested that miR-449a acted as a key regulator of colon tumorigenesis by controlling the proliferation of intestinal epithelial cells. Additionally, activation of miR-449a may represent an effective therapeutic strategy and prognostic marker in colon cancer.
Collapse
|
24
|
Tabaja N, Yuan Z, Oswald F, Kovall RA. Structure-function analysis of RBP-J-interacting and tubulin-associated (RITA) reveals regions critical for repression of Notch target genes. J Biol Chem 2017; 292:10549-10563. [PMID: 28487372 DOI: 10.1074/jbc.m117.791707] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 05/04/2017] [Indexed: 11/06/2022] Open
Abstract
The Notch pathway is a cell-to-cell signaling mechanism that is essential for tissue development and maintenance, and aberrant Notch signaling has been implicated in various cancers, congenital defects, and cardiovascular diseases. Notch signaling activates the expression of target genes, which are regulated by the transcription factor CSL (CBF1/RBP-J, Su(H), Lag-1). CSL interacts with both transcriptional corepressor and coactivator proteins, functioning as both a repressor and activator, respectively. Although Notch activation complexes are relatively well understood at the structural level, less is known about how CSL interacts with corepressors. Recently, a new RBP-J (mammalian CSL ortholog)-interacting protein termed RITA has been identified and shown to export RBP-J out of the nucleus, thereby leading to the down-regulation of Notch target gene expression. However, the molecular details of RBP-J/RITA interactions are unclear. Here, using a combination of biochemical/cellular, structural, and biophysical techniques, we demonstrate that endogenous RBP-J and RITA proteins interact in cells, map the binding regions necessary for RBP-J·RITA complex formation, and determine the X-ray structure of the RBP-J·RITA complex bound to DNA. To validate the structure and glean more insights into function, we tested structure-based RBP-J and RITA mutants with biochemical/cellular assays and isothermal titration calorimetry. Whereas our structural and biophysical studies demonstrate that RITA binds RBP-J similarly to the RAM (RBP-J-associated molecule) domain of Notch, our biochemical and cellular assays suggest that RITA interacts with additional regions in RBP-J. Taken together, these results provide molecular insights into the mechanism of RITA-mediated regulation of Notch signaling, contributing to our understanding of how CSL functions as a transcriptional repressor of Notch target genes.
Collapse
Affiliation(s)
- Nassif Tabaja
- From the Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267 and
| | - Zhenyu Yuan
- From the Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267 and
| | - Franz Oswald
- the Department of Internal Medicine I, Center for Internal Medicine, University Medical Center Ulm, 89081 Ulm, Germany
| | - Rhett A Kovall
- From the Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267 and
| |
Collapse
|
25
|
Abstract
The Notch signalling cascade is an evolutionarily conserved pathway that has a crucial role in regulating development and homeostasis in various tissues. The cellular processes and events that it controls are diverse, and continued investigation over recent decades has revealed how the role of Notch signalling is multifaceted and highly context dependent. Consistent with the far-reaching impact that Notch has on development and homeostasis, aberrant activity of the pathway is also linked to the initiation and progression of several malignancies, and Notch can in fact be either oncogenic or tumour suppressive depending on the tissue and cellular context. The Notch pathway therefore represents an important target for therapeutic agents designed to treat many types of cancer. In this Review, we focus on the latest developments relating specifically to the tumour-suppressor activity of Notch signalling and discuss the potential mechanisms by which Notch can inhibit carcinogenesis in various tissues. Potential therapeutic strategies aimed at restoring or augmenting Notch-mediated tumour suppression will also be highlighted.
Collapse
Affiliation(s)
- Craig S Nowell
- CMU, Department for Pathology and Immunology, University of Geneva, Rue Michel Servet, 1211 Geneva 4, Switzerland
| | - Freddy Radtke
- Ecole Polytechnique Fédérale de Lausanne, School of Life Sciences, Swiss Institute for Experimental Cancer Research, Lausanne, Vaud 1015, Switzerland
| |
Collapse
|
26
|
Sonic Hedgehog promotes proliferation of Notch-dependent monociliated choroid plexus tumour cells. Nat Cell Biol 2016; 18:418-30. [PMID: 26999738 PMCID: PMC4814324 DOI: 10.1038/ncb3327] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Accepted: 02/09/2016] [Indexed: 12/17/2022]
Abstract
Aberrant Notch signaling has been linked to many cancers including choroid plexus (CP) tumours, a group of rare and predominantly pediatric brain neoplasms. We developed animal models of CP tumours by inducing sustained expression of Notch1 that recapitulate properties of human CP tumours with aberrant NOTCH signaling. Whole transcriptome and functional analyses showed that tumour cell proliferation is associated with Sonic Hedgehog (Shh) in the tumour microenvironment. Unlike CP epithelial cells, which have multiple primary cilia, tumour cells possess a solitary primary cilium as a result of Notch-mediated suppression of multiciliate diffferentiation. A Shh-driven signaling cascade in the primary cilium occurs in tumour cells but not in epithelial cells. Lineage studies show that CP tumours arise from mono-ciliated progenitors in the roof plate characterized by elevated Notch signaling. Abnormal SHH signaling and distinct ciliogenesis are detected in human CP tumours, suggesting SHH pathway and cilia differentiation as potential therapeutic avenues.
Collapse
|
27
|
A study of the role of Notch1 and JAG1 gene methylation in development of breast cancer. Med Oncol 2016; 33:35. [PMID: 26971121 DOI: 10.1007/s12032-016-0750-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 02/26/2016] [Indexed: 10/22/2022]
Abstract
This study is to explore the roles of gene methylation of Notch1 and JAG1 in development of invasive ductal carcinoma of breast. Quantitative analysis the DNA methylation levels of Notch1 and JAG1 gene by the MassARRAY method in invasive ductal carcinoma of breast (IDC; n = 89), atypical ductal hyperplasia of breast (ADH; n = 11), and ordinary ductal hyperplasia of breast (UDH; n = 20). The expressions of JAG1 and Notch1 protein in four breast tissues were detected by immunohistochemistry SP method. (1) Positive expression rates of Notch1 protein in IDC and DCIS were 88.7 % (79/89) and 70.0 % (14/20), respectively, which were significantly higher than the levels in ADH (36.0 %, 4/11) and UDH (25.0 %, 5/20; P < 0.05). Notch1 protein expression was significant positively correlated with lymph node metastasis, pathological grades, and TNM stages of IDC. (2) Positive expression rates of JAG1 protein in IDC and DCIS were 89.9 % (80/89) and 75.0 % (15/20), respectively, which were significantly higher than those of ADH (45.0 %, 5/11) and UDH (30.0 %, 6/20; P < 0.05). JAG1 protein expression was significant positive correlation with lymph node metastasis, pathological grades and TNM stages of IDC. There is an overall hypomethylation alteration of Notch1 and JAG gene in IDC, with corresponding over-expression of Notch1 and JAG1 protein. This inverse correlation shows that the alteration of protein expression results from hypomethylation oncogene Notch1 and JAG1, and this change may play an important role in occurrence and progression of breast cancer.
Collapse
|
28
|
Ding N, Che L, Li XL, Liu Y, Jiang LJ, Fan B, Tao JY, Chen X, Ji JF. Oncogenic potential of IDH1R132C mutant in cholangiocarcinoma development in mice. World J Gastroenterol 2016; 22:2071-2080. [PMID: 26877611 PMCID: PMC4726679 DOI: 10.3748/wjg.v22.i6.2071] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 08/04/2015] [Accepted: 09/02/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate whether IDH1R132C mutant in combination with loss of p53 and activated Notch signaling promotes intrahepatic cholangiocarcinoma (ICC) development.
METHODS: We applied hydrodynamic injection and sleeping beauty mediated somatic integration to induce loss of p53 (via shP53), activation of Notch [via intracellular domain of Notch1 (NICD)] and/or overexpression of IDH1R132C mutant together with the sleeping beauty transposase into the mouse liver. Specifically, we co-expressed shP53 and NICD (shP53/NICD, n = 4), shP53 and IDH1R132C (shP53/IDH1R132C, n = 3), NICD and IDH1R132C (NICD/IDH1R132C, n = 4), as well as NICD, shP53 and IDH1R132C (NICD/shP53/IDH1R132C, n = 9) in mice. Mice were monitored for liver tumor development and euthanized at various time points. Liver histology was analyzed by hematoxylin and eosin staining. Molecular features of NICD/shP53/IDH1R132C ICC tumor cells were characterized by Myc tag, Flag tag, Ki-67, p-Erk and p-AKT immunohistochemical staining. Desmoplastic reaction in tumor tissues was studied by Picro-Sirius red staining.
RESULTS: We found that co-expression of shP53/NICD, shP53/IDH1R132C or NICD/IDH1R132C did not lead to liver tumor formation. In striking contrast, co-expression of NICD/shP53/IDH1R132C resulted in ICC development in mice (P < 0.01). The tumors could be identified as early as 12 wk post hydrodynamic injection. Tumors rapidly progressed, and by 18 wk post hydrodynamic injection, multiple cystic lesions could be identified on the liver surface. NICD/shP53/IDH1R132C liver tumors shared multiple histological features of human ICCs, including hyperplasia of irregular glands. Importantly, all tumor cells were positive for the biliary epithelial cell marker cytokeratin 19. Extensive collagen fibers could be visualized in tumor tissues using Sirus red staining, duplicating the desmoplastic reaction observed in human ICC. Tumors were highly proliferative and expressed ectopically injected genes. Together these studies supported that NICD/shP53/IDH1R132C liver tumors were indeed ICCs. Finally, no p-AKT or p-ERK positive staining was observed, suggesting that NICD/shP53/IDH1R132C driven ICC development was independent of AKT/mTOR and Ras/MAPK signaling cascades.
CONCLUSION: We have generated a simple, non-germline murine ICC model with activated Notch, loss of p53 and IDH1R132C mutant. The study supported the oncogenic potential of IDH1R132C.
Collapse
|
29
|
|
30
|
Gil-García B, Baladrón V. The complex role of NOTCH receptors and their ligands in the development of hepatoblastoma, cholangiocarcinoma and hepatocellular carcinoma. Biol Cell 2015; 108:29-40. [DOI: 10.1111/boc.201500029] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 11/24/2015] [Indexed: 12/12/2022]
Affiliation(s)
- Borja Gil-García
- Laboratory of Biochemistry and Molecular Biology; Department of Inorganic and Organic Chemistry and Biochemistry; Medical School/CRIB/Biomedicine Unit; University of Castilla-La Mancha (UCLM)/CSIC; 02008, Albacete Spain
| | - Victoriano Baladrón
- Laboratory of Biochemistry and Molecular Biology; Department of Inorganic and Organic Chemistry and Biochemistry; Medical School/CRIB/Biomedicine Unit; University of Castilla-La Mancha (UCLM)/CSIC; 02008, Albacete Spain
| |
Collapse
|
31
|
Pang S, Shen J, Liu Y, Chen F, Zheng Z, James AW, Hsu CY, Zhang H, Lee KS, Wang C, Li C, Chen X, Jia H, Zhang X, Soo C, Ting K. Proliferation and osteogenic differentiation of mesenchymal stem cells induced by a short isoform of NELL-1. Stem Cells 2015; 33:904-15. [PMID: 25376942 DOI: 10.1002/stem.1884] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 09/19/2014] [Accepted: 09/27/2014] [Indexed: 01/11/2023]
Abstract
Neural epidermal growth factor-like (NEL)-like protein 1 (NELL-1) has been identified as an osteoinductive differentiation factor that promotes mesenchymal stem cell (MSC) osteogenic differentiation. In addition to full-length NELL-1, there are several NELL-1-related transcripts reported. We used rapid amplification of cDNA ends to recover potential cDNA of NELL-1 isoforms. A NELL-1 isoform with the N-terminal 240 amino acid (aa) residues truncated was identified. While full-length NELL-1 that contains 810 aa residues (NELL-1810 ) plays an important role in embryologic skeletal development, the N-terminal-truncated NELL-1 isoform (NELL-1570 ) was expressed postnatally. Similar to NELL-1810 , NELL-1570 induced MSC osteogenic differentiation. In addition, NELL-1570 significantly stimulated MSC proliferation in multiple MSC-like populations such as murine C3H10T1/2 MSC cell line, mouse primary MSCs, and perivascular stem cells, which is a type of stem cells proposed as the perivascular origin of MSCs. In contrast, NELL-1810 demonstrated only limited stimulation of MSC proliferation. Similar to NELL-1810 , NELL-1570 was found to be secreted from host cells. Both NELL-1570 expression lentiviral vector and column-purified recombinant protein NELL-1570 demonstrated almost identical effects in MSC proliferation and osteogenic differentiation, suggesting that NELL-1570 may function as a pro-osteogenic growth factor. In vivo, NELL-1570 induced significant calvarial defect regeneration accompanied by increased cell proliferation. Thus, NELL-1570 has the potential to be used for cell-based or hormone-based therapy of bone regeneration.
Collapse
Affiliation(s)
- Shen Pang
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Li LC, Wang DL, Wu YZ, Nian WQ, Wu ZJ, Li Y, Ma HW, Shao JH. Gastric tumor-initiating CD44 + cells and epithelial-mesenchymal transition are inhibited by γ-secretase inhibitor DAPT. Oncol Lett 2015; 10:3293-3299. [PMID: 26722328 DOI: 10.3892/ol.2015.3727] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 08/17/2015] [Indexed: 12/15/2022] Open
Abstract
It has been proposed that the Notch signaling pathway may serve a pivotal role in cellular differentiation, proliferation and apoptosis. However, the function of Notch signaling in gastric cancer stem cells (GCSCs) is largely unknown. The present study aimed to delineate the role of the Notch1 pathway in GCSCs and during epithelial-mesenchymal transition (EMT). Flow cytometry was used to isolate CD44+ cells from the human gastric cancer cell line, MKN45. CD44+ cells displayed the characteristics of CSCs and exhibited higher Notch1 expression compared with CD44- cells. To investigate the role of the Notch1 pathway in GCSCs, CD44+ cells were treated with the γ-secretase inhibitor DAPT. DAPT treatment inhibited the expression of the Notch1 downstream target Hes1 and EMT markers, suppressed the properties of CSCs and impaired the invasion and proliferation capabilities of CD44+ cells. In addition, intraperitoneal treatment with DAPT effectively inhibited the growth of CD44+ cell xenograft tumors. The present study indicated that CD44+ GCSCs possess the characteristics of CSCs and that the Notch1 pathway serves a critical role in the maintenance of CSCs and EMT.
Collapse
Affiliation(s)
- Lu-Chun Li
- Department of Oncology, Chongqing Cancer Institute, Chongqing 400030, P.R. China
| | - Dong-Lin Wang
- Department of Oncology, Chongqing Cancer Institute, Chongqing 400030, P.R. China
| | - Yong-Zhong Wu
- Department of Radiotherapy, Chongqing Cancer Institute, Chongqing 400030, P.R. China
| | - Wei-Qi Nian
- Department of Galactophore, Chongqing Cancer Institute, Chongqing 400030, P.R. China
| | - Zhi-Juan Wu
- Department of Oncology, Chongqing Cancer Institute, Chongqing 400030, P.R. China
| | - Yan Li
- Department of Oncology, Chongqing Cancer Institute, Chongqing 400030, P.R. China
| | - Hui-Wen Ma
- Department of Oncology, Chongqing Cancer Institute, Chongqing 400030, P.R. China
| | - Jiang-He Shao
- Department of Oncology, Chongqing Cancer Institute, Chongqing 400030, P.R. China
| |
Collapse
|
33
|
Stephenson NL, Avis JM. Mutational analysis of the Notch2 negative regulatory region identifies key structural elements for mechanical stability. FEBS Open Bio 2015; 5:625-33. [PMID: 26288744 PMCID: PMC4537882 DOI: 10.1016/j.fob.2015.07.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 07/27/2015] [Accepted: 07/27/2015] [Indexed: 11/23/2022] Open
Abstract
Mutations within heterodimerization (HD) domain of Notch2 reduce mechanical stability. Structural changes are observed within the LNRA:B linker region/LNRB and αC helix. The LNRC:HD domain interaction is also reduced in stability. Changes in mechanical stability versus chemical stability are highlighted.
The Notch signalling pathway is fundamental to cell differentiation in developing and self-renewing tissues. Notch is activated upon ligand-induced conformational change of the Notch negative regulatory region (NRR), unmasking a key proteolytic site (S2) and facilitating downstream events. The favoured model requires endocytosis of a tightly bound ligand to transmit force to the NRR region, sufficient to cause a structural change that exposes the S2 site. We have previously shown, using atomic force microscopy and molecular dynamics simulations, that application of force to the N-terminus of the Notch2 NRR facilitates metalloprotease cleavage at an early stage in the unfolding process. Here, mutations are made within the heterodimerization (HD) domain of the NRR that are known to cause constitutive activation of Notch1 whilst having no effect on the chemical stability of Notch2. Comparison of the mechanical stability and simulated forced unfolding of recombinant Notch2 NRR proteins demonstrates a reduced stability following mutation and identifies two critical structural elements of the NRR in its response to force – the linker region between Lin12-Notch repeats LNRA and LNRB and the α3 helix within the HD domain – both of which mask the S2 cleavage site prior to Notch activation. In two mutated proteins, the LNRC:HD domain interaction is also reduced in stability. The observed changes to mechanical stability following these HD domain mutations highlight key regions of the Notch2 NRR that are important for mechanical, but not chemical, stability. This research could also help determine the fundamental differences in the NRRs of Notch1 and Notch2.
Collapse
Affiliation(s)
- Natalie L Stephenson
- Faculty of Life Sciences, Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| | - Johanna M Avis
- Faculty of Life Sciences, Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| |
Collapse
|
34
|
Ho DM, Pallavi SK, Artavanis-Tsakonas S. The Notch-mediated hyperplasia circuitry in Drosophila reveals a Src-JNK signaling axis. eLife 2015. [PMID: 26222204 PMCID: PMC4517436 DOI: 10.7554/elife.05996] [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] [Indexed: 12/11/2022] Open
Abstract
Notch signaling controls a wide range of cell fate decisions during development and disease via synergistic interactions with other signaling pathways. Here, through a genome-wide genetic screen in Drosophila, we uncover a highly complex Notch-dependent genetic circuitry that profoundly affects proliferation and consequently hyperplasia. We report a novel synergistic relationship between Notch and either of the non-receptor tyrosine kinases Src42A and Src64B to promote hyperplasia and tissue disorganization, which results in cell cycle perturbation, JAK/STAT signal activation, and differential regulation of Notch targets. Significantly, the JNK pathway is responsible for the majority of the phenotypes and transcriptional changes downstream of Notch-Src synergy. We previously reported that Notch-Mef2 also activates JNK, indicating that there are commonalities within the Notch-dependent proliferation circuitry; however, the current data indicate that Notch-Src accesses JNK in a significantly different fashion than Notch-Mef2. DOI:http://dx.doi.org/10.7554/eLife.05996.001 The cells within animals are organized into tissues and organs that perform particular roles. To develop and maintain these structures, the ability of individual cells to divide and grow is strictly controlled by the activities of many proteins, including one called Notch. This protein is found in all multicellular organisms and allows cells to communicate with each other. Mutations in the gene that encodes Notch can cause cells to divide excessively and lead to cancer and other diseases. Notch regulates the growth and division of cells by interacting with many other proteins. For example, Mef2 works with Notch to activate a communication system called the JNK pathway. This pathway is involved in controlling cell division, cell death, and cell movement. However, it is thought that Notch may also interact with other proteins that have not yet been identified. Now, Ho et al. have conducted a genome-wide screen in fruit flies to find proteins that interact with Notch. The experiments used flies that develop abnormally large eyes because they have an over-active Notch protein. Ho et al. identified hundreds of fruit fly genes that could increase or decrease the size of the flies' eyes in the presence of Notch activity. Many of these genes are known to be involved in development, cell division, or in controlling the activity of other genes. Ho et al. found that two of these genes encode similar proteins called Src42A and Src64B, which are similar to the Src proteins that are involved in many types of human cancers. The experiments show that both proteins interact with Notch to promote uncontrolled cell division and lead to tissues in the flies becoming more disorganized. The JNK pathway is also activated by Notch working with Src42A or Src64B, but in a different manner to how it is activated by Mef2 and Notch, and with different consequences for cells. This study provides new insights into how genes work together in order to influence cell division and other events in development. Also, it suggests that Notch activity may regulate the growth of cancers linked with defects in the Src proteins. DOI:http://dx.doi.org/10.7554/eLife.05996.002
Collapse
Affiliation(s)
- Diana M Ho
- Department of Cell Biology, Harvard Medical School, Boston, United States
| | - S K Pallavi
- Department of Cell Biology, Harvard Medical School, Boston, United States
| | | |
Collapse
|
35
|
Sharma A, Gadkari RA, Ramakanth SV, Padmanabhan K, Madhumathi DS, Devi L, Appaji L, Aster JC, Rangarajan A, Dighe RR. A novel Monoclonal Antibody against Notch1 Targets Leukemia-associated Mutant Notch1 and Depletes Therapy Resistant Cancer Stem Cells in Solid Tumors. Sci Rep 2015; 5:11012. [PMID: 26046801 PMCID: PMC4457015 DOI: 10.1038/srep11012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 05/07/2015] [Indexed: 01/08/2023] Open
Abstract
Higher Notch signaling is known to be associated with hematological and solid cancers. We developed a potential immunotherapeutic monoclonal antibody (MAb) specific for the Negative Regulatory Region of Notch1 (NRR). The MAb604.107 exhibited higher affinity for the "Gain-of-function" mutants of Notch1 NRR associated with T Acute lymphoblastic Leukemia (T-ALL). Modeling of the mutant NRR with 12 amino-acid insertion demonstrated "opening" resulting in exposure of the S2-cleavage site leading to activated Notch1 signaling. The MAb, at low concentrations (1-2 μg/ml), inhibited elevated ligand-independent Notch1 signaling of NRR mutants, augmented effect of Thapsigargin, an inhibitor of mutant Notch1, but had no effect on the wild-type Notch1. The antibody decreased proliferation of the primary T-ALL cells and depleted leukemia initiating CD34/CD44 high population. At relatively high concentrations, (10-20 μg/ml), the MAb affected Notch1 signaling in the breast and colon cancer cell lines. The Notch-high cells sorted from solid-tumor cell lines exhibited characteristics of cancer stem cells, which were inhibited by the MAb. The antibody also increased the sensitivity to Doxorubucinirubicin. Further, the MAb impeded the growth of xenografts from breast and colon cancer cells potentiated regression of the tumors along with Doxorubucin. Thus, this antibody is potential immunotherapeutic tool for different cancers.
Collapse
Affiliation(s)
- Ankur Sharma
- Department of Molecular Reproduction Development and Genetics, Indian Institute of Science Bangalore, Karnataka, India
| | - Rupali A Gadkari
- Molecular Biophysics Unit, Indian Institute of Science Bangalore, Karnataka, India
| | - Satthenapalli V Ramakanth
- Department of Molecular Reproduction Development and Genetics, Indian Institute of Science Bangalore, Karnataka, India
| | - Krishnanand Padmanabhan
- Department of Molecular Reproduction Development and Genetics, Indian Institute of Science Bangalore, Karnataka, India
| | - Davanam S Madhumathi
- Department of Pathology, Kidwai Memorial Institute of Oncology, Bangalore, Karnataka, India
| | - Lakshmi Devi
- Department of Pathology, Kidwai Memorial Institute of Oncology, Bangalore, Karnataka, India
| | - Lingappa Appaji
- Department of Pediatric Oncology, Kidwai Memorial Institute of Oncology, Bangalore, Karnataka, India
| | - Jon C Aster
- Department of Pathology, Brigham &Women's Hospital, Harvard Medical School, Boston, USA
| | - Annapoorni Rangarajan
- Department of Molecular Reproduction Development and Genetics, Indian Institute of Science Bangalore, Karnataka, India
| | - Rajan R Dighe
- Department of Molecular Reproduction Development and Genetics, Indian Institute of Science Bangalore, Karnataka, India
| |
Collapse
|
36
|
Mutvei AP, Fredlund E, Lendahl U. Frequency and distribution of Notch mutations in tumor cell lines. BMC Cancer 2015; 15:311. [PMID: 25907971 PMCID: PMC4430925 DOI: 10.1186/s12885-015-1278-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 03/26/2015] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Deregulated Notch signaling is linked to a variety of tumors and it is therefore important to learn more about the frequency and distribution of Notch mutations in a tumor context. METHODS In this report, we use data from the recently developed Cancer Cell Line Encyclopedia to assess the frequency and distribution of Notch mutations in a large panel of cancer cell lines in silico. RESULTS Our results show that the mutation frequency of Notch receptor and ligand genes is at par with that for established oncogenes and higher than for a set of house-keeping genes. Mutations were found across all four Notch receptor genes, but with notable differences between protein domains, mutations were for example more prevalent in the regions encoding the LNR and PEST domains in the Notch intracellular domain. Furthermore, an in silico estimation of functional impact showed that deleterious mutations cluster to the ligand-binding and the intracellular domains of NOTCH1. For most cell line groups, the mutation frequency of Notch genes is higher than in associated primary tumors. CONCLUSIONS Our results shed new light on the spectrum of Notch mutations after in vitro culturing of tumor cells. The higher mutation frequency in tumor cell lines indicates that Notch mutations are associated with a growth advantage in vitro, and thus may be considered to be driver mutations in a tumor cell line context.
Collapse
Affiliation(s)
| | - Erik Fredlund
- Department of Oncology and Pathology, Science for Life Laboratory, Karolinska Institute, SE-171 77, Stockholm, Sweden.
| | - Urban Lendahl
- Department of Cell and Molecular Biology, SE-171 77, Stockholm, Sweden.
| |
Collapse
|
37
|
Profiling invasiveness in head and neck cancer: recent contributions of genomic and transcriptomic approaches. Cancers (Basel) 2015; 7:585-97. [PMID: 25836654 PMCID: PMC4491672 DOI: 10.3390/cancers7020585] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Revised: 03/20/2015] [Accepted: 03/24/2015] [Indexed: 01/17/2023] Open
Abstract
High-throughput molecular profiling approaches have emerged as precious research tools in the field of head and neck translational oncology. Such approaches have identified and/or confirmed the role of several genes or pathways in the acquisition/maintenance of an invasive phenotype and the execution of cellular programs related to cell invasion. Recently published new-generation sequencing studies in head and neck squamous cell carcinoma (HNSCC) have unveiled prominent roles in carcinogenesis and cell invasion of mutations involving NOTCH1 and PI3K-patwhay components. Gene-expression profiling studies combined with systems biology approaches have allowed identifying and gaining further mechanistic understanding into pathways commonly enriched in invasive HNSCC. These pathways include antigen-presenting and leucocyte adhesion molecules, as well as genes involved in cell-extracellular matrix interactions. Here we review the major insights into invasiveness in head and neck cancer provided by high-throughput molecular profiling approaches.
Collapse
|
38
|
Liu Z, Brunskill E, Boyle S, Chen S, Turkoz M, Guo Y, Grant R, Kopan R. Second-generation Notch1 activity-trap mouse line (N1IP::CreHI) provides a more comprehensive map of cells experiencing Notch1 activity. Development 2015; 142:1193-202. [PMID: 25725069 DOI: 10.1242/dev.119529] [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] [Indexed: 11/20/2022]
Abstract
We have previously described the creation and analysis of a Notch1 activity-trap mouse line, Notch1 intramembrane proteolysis-Cre6MT or N1IP::Cre(LO), that marked cells experiencing relatively high levels of Notch1 activation. Here, we report and characterize a second line with improved sensitivity (N1IP::Cre(HI)) to mark cells experiencing lower levels of Notch1 activation. This improvement was achieved by increasing transcript stability and by restoring the native carboxy terminus of Cre, resulting in a five- to tenfold increase in Cre activity. The magnitude of this effect probably impacts Cre activity in strains with carboxy-terminal Ert2 fusion. These two trap lines and the related line N1IP::Cre(ERT2) form a complementary mapping tool kit to identify changes in Notch1 activation patterns in vivo as the consequence of genetic or pharmaceutical intervention, and illustrate the variation in Notch1 signal strength from one tissue to the next and across developmental time.
Collapse
Affiliation(s)
- Zhenyi Liu
- SAGE Labs, St Louis, MO 63146, USA Department of Developmental Biology, Washington University, St Louis, MO 63110, USA
| | - Eric Brunskill
- Division of Developmental Biology, Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Scott Boyle
- Department of Developmental Biology, Washington University, St Louis, MO 63110, USA
| | - Shuang Chen
- Department of Developmental Biology, Washington University, St Louis, MO 63110, USA Division of Developmental Biology, Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Mustafa Turkoz
- Department of Developmental Biology, Washington University, St Louis, MO 63110, USA Division of Developmental Biology, Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Yuxuan Guo
- Department of Developmental Biology, Washington University, St Louis, MO 63110, USA Carnegie Institution for Science, Department of Embryology, Baltimore, MD 21218, USA
| | - Rachel Grant
- Department of Developmental Biology, Washington University, St Louis, MO 63110, USA
| | - Raphael Kopan
- Department of Developmental Biology, Washington University, St Louis, MO 63110, USA Division of Developmental Biology, Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| |
Collapse
|
39
|
Shao S, Zhao X, Zhang X, Luo M, Zuo X, Huang S, Wang Y, Gu S, Zhao X. Notch1 signaling regulates the epithelial-mesenchymal transition and invasion of breast cancer in a Slug-dependent manner. Mol Cancer 2015; 14:28. [PMID: 25645291 PMCID: PMC4322803 DOI: 10.1186/s12943-015-0295-3] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 01/13/2015] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND The epithelial-mesenchymal transition (EMT) is crucial for the invasion and metastasis of breast cancer. However, how Notch signaling regulates the EMT process and invasion in breast cancer remains largely unknown. METHODS The impact of Notch1 silencing by specific shRNAs on the EMT and invasion of human breast cancer MCF-7 and MDA-MB-231 cells as well as xenografts was tested by western blot, real-time polymerase chain reaction (RT-PCR), immunofluorescence, transwell, and immunohistochemistry assays. The effect of Slug silencing or upregulation on the EMT and invasion of breast cancer cells was analyzed, and the effect of Notch1 signaling on Slug expression was determined by the luciferase reporter assay. RESULTS The Notch1 intracellular domain (N1ICD) and Jagged1 were expressed in breast cancer cells. Notch1 silencing reversed the spontaneous EMT process and inhibited the migration and invasion of breast cancer cells and the growth of xenograft breast cancers. The expression of N1ICD was upregulated significantly by Jagged1-mediated Notch signaling activation. Moreover, Jagged1-mediated Notch signaling promoted the EMT process, migration, and invasion of breast cancer cells, which were abrogated by Notch silencing. Furthermore, the N1ICD positively regulated the Slug expression by inducing Slug promoter activation. Importantly, the knockdown of Slug weakened the invasion ability of breast cancer cells and reversed the Jagged1-induced EMT process with significantly decreased expression of vimentin and increased expression of E-cadherin. In addition, Slug overexpression restored the Notch1 knockdown-suppressed EMT process. CONCLUSIONS Our novel data indicate that Notch signaling positively regulates the EMT, invasion, and growth of breast cancer cells by inducing Slug expression. The Notch1-Slug signaling axis may represent a potential therapeutic target for breast cancer therapy.
Collapse
Affiliation(s)
- Shan Shao
- The Department of Oncology, the First Hospital Affiliated to the School of Medicine, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi Province, China.
| | - Xiaoai Zhao
- The Department of Oncology, the First Hospital Affiliated to the School of Medicine, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi Province, China.
| | - Xiaojin Zhang
- The Department of Oncology, the First Hospital Affiliated to the School of Medicine, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi Province, China.
| | - Minna Luo
- The Department of Oncology, the First Hospital Affiliated to the School of Medicine, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi Province, China.
| | - Xiaoxiao Zuo
- The Department of Oncology, the First Hospital Affiliated to the School of Medicine, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi Province, China.
| | - Shangke Huang
- The Department of Oncology, the First Hospital Affiliated to the School of Medicine, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi Province, China.
| | - Ying Wang
- The Department of Oncology, the First Hospital Affiliated to the School of Medicine, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi Province, China.
| | - Shanzhi Gu
- The Department of Forensic Medicine, Medical School, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, 710061, Shaanxi Province, China.
| | - Xinhan Zhao
- The Department of Oncology, the First Hospital Affiliated to the School of Medicine, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi Province, China.
| |
Collapse
|
40
|
Kaushik G, Venugopal A, Ramamoorthy P, Standing D, Subramaniam D, Umar S, Jensen RA, Anant S, Mammen JMV. Honokiol inhibits melanoma stem cells by targeting notch signaling. Mol Carcinog 2014; 54:1710-21. [PMID: 25491779 DOI: 10.1002/mc.22242] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Revised: 09/04/2014] [Accepted: 09/26/2014] [Indexed: 12/20/2022]
Abstract
Melanoma is an aggressive disease with limited therapeutic options. Here, we determined the effects of honokiol (HNK), a biphenolic natural compound on melanoma cells and stemness. HNK significantly inhibited melanoma cell proliferation, viability, clonogenicity and induced autophagy. In addition, HNK significantly inhibited melanosphere formation in a dose dependent manner. Western blot analyses also demonstrated reduction in stem cell markers CD271, CD166, Jarid1b, and ABCB5. We next examined the effect of HNK on Notch signaling, a pathway involved in stem cell self-renewal. Four different Notch receptors exist in cells, which when cleaved by a series of enzymatic reactions catalyzed by Tumor Necrosis Factor-α-Converting Enzyme (TACE) and γ-secretase protein complex, results in the release of the Notch intracellular domain (NICD), which then translocates to the nucleus and induces target gene expression. Western blot analyses demonstrated that in HNK treated cells there is a significant reduction in the expression of cleaved Notch-2. In addition, there was a reduction in the expression of downstream target proteins, Hes-1 and cyclin D1. Moreover, HNK treatment suppressed the expression of TACE and γ-secretase complex proteins in melanoma cells. To confirm that suppression of Notch-2 activation is critical for HNK activity, we overexpressed NICD1, NICD2, and performed HNK treatment. NICD2, but not NICD1, partially restored the expression of Hes-1 and cyclin D1, and increased melanosphere formation. Taken together, these data suggest that HNK is a potent inhibitor of melanoma cells, in part, through the targeting of melanoma stem cells by suppressing Notch-2 signaling.
Collapse
Affiliation(s)
- Gaurav Kaushik
- Departments of Surgery, The University of Kansas Medical Center, Kansas City, Kansas
| | - Anand Venugopal
- Departments of Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, Kansas
| | - Prabhu Ramamoorthy
- Departments of Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, Kansas.,University of Kansas Cancer Center, Kansas City, Kansas
| | - David Standing
- Departments of Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, Kansas
| | - Dharmalingam Subramaniam
- Departments of Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, Kansas.,University of Kansas Cancer Center, Kansas City, Kansas
| | - Shahid Umar
- Departments of Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, Kansas.,University of Kansas Cancer Center, Kansas City, Kansas
| | - Roy A Jensen
- Departments of Pathology, The University of Kansas Medical Center, Kansas City, Kansas.,University of Kansas Cancer Center, Kansas City, Kansas
| | - Shrikant Anant
- Departments of Surgery, The University of Kansas Medical Center, Kansas City, Kansas.,Departments of Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, Kansas.,University of Kansas Cancer Center, Kansas City, Kansas
| | - Joshua M V Mammen
- Departments of Surgery, The University of Kansas Medical Center, Kansas City, Kansas.,University of Kansas Cancer Center, Kansas City, Kansas
| |
Collapse
|
41
|
Li N, Fassl A, Chick J, Inuzuka H, Li X, Mansour MR, Liu L, Wang H, King B, Shaik S, Gutierrez A, Ordureau A, Otto T, Kreslavsky T, Baitsch L, Bury L, Meyer CA, Ke N, Mulry KA, Kluk MJ, Roy M, Kim S, Zhang X, Geng Y, Zagozdzon A, Jenkinson S, Gale RE, Linch DC, Zhao JJ, Mullighan CG, Harper JW, Aster JC, Aifantis I, von Boehmer H, Gygi SP, Wei W, Look AT, Sicinski P. Cyclin C is a haploinsufficient tumour suppressor. Nat Cell Biol 2014; 16:1080-91. [PMID: 25344755 PMCID: PMC4235773 DOI: 10.1038/ncb3046] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 08/29/2014] [Indexed: 12/12/2022]
Abstract
Cyclin C was cloned as a growth-promoting G1 cyclin, and was also shown to regulate gene transcription. Here we report that in vivo cyclin C acts as a haploinsufficient tumour suppressor, by controlling Notch1 oncogene levels. Cyclin C activates an 'orphan' CDK19 kinase, as well as CDK8 and CDK3. These cyclin-C-CDK complexes phosphorylate the Notch1 intracellular domain (ICN1) and promote ICN1 degradation. Genetic ablation of cyclin C blocks ICN1 phosphorylation in vivo, thereby elevating ICN1 levels in cyclin-C-knockout mice. Cyclin C ablation or heterozygosity collaborates with other oncogenic lesions and accelerates development of T-cell acute lymphoblastic leukaemia (T-ALL). Furthermore, the cyclin C encoding gene CCNC is heterozygously deleted in a significant fraction of human T-ALLs, and these tumours express reduced cyclin C levels. We also describe point mutations in human T-ALL that render cyclin-C-CDK unable to phosphorylate ICN1. Hence, tumour cells may develop different strategies to evade inhibition by cyclin C.
Collapse
Affiliation(s)
- Na Li
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Genetics, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Anne Fassl
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Genetics, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Joel Chick
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Hiroyuki Inuzuka
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Xiaoyu Li
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, and Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Marc R. Mansour
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, and Division of Hematology/ Oncology, Children's Hospital, Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02215, USA
- Department of Haematology, University College London Cancer Institute, London WC1E 6BT, UK
| | - Lijun Liu
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Genetics, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Haizhen Wang
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Genetics, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Bryan King
- Howard Hughes Medical Institute and Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
| | - Shavali Shaik
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Alejandro Gutierrez
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, and Division of Hematology/ Oncology, Children's Hospital, Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Alban Ordureau
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Tobias Otto
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Genetics, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Taras Kreslavsky
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, and Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Lukas Baitsch
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Leah Bury
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Genetics, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Clifford A. Meyer
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute and Harvard School of Public Health, Boston, MA 02115, USA
| | - Nan Ke
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Genetics, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Kristin A. Mulry
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Genetics, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Michael J. Kluk
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | - Moni Roy
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | - Sunkyu Kim
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, USA
| | - Xiaowu Zhang
- Cell Signaling Technology, Inc., Danvers MA 01923, USA
| | - Yan Geng
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Genetics, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Agnieszka Zagozdzon
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Genetics, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Sarah Jenkinson
- Department of Haematology, University College London Cancer Institute, London WC1E 6BT, UK
| | - Rosemary E. Gale
- Department of Haematology, University College London Cancer Institute, London WC1E 6BT, UK
| | - David C. Linch
- Department of Haematology, University College London Cancer Institute, London WC1E 6BT, UK
| | - Jean J. Zhao
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Charles G. Mullighan
- Department of Pathology, St. Jude Research Hospital, Memphis, Tennessee 38105, USA
| | - J. Wade Harper
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Jon C. Aster
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | - Iannis Aifantis
- Howard Hughes Medical Institute and Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
| | - Harald von Boehmer
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, and Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Steven P. Gygi
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - A. Thomas Look
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, and Division of Hematology/ Oncology, Children's Hospital, Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Piotr Sicinski
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Genetics, Harvard Medical School, Boston, Massachusetts 02215, USA
| |
Collapse
|
42
|
Evans AG, Calvi LM. Notch signaling in the malignant bone marrow microenvironment: implications for a niche-based model of oncogenesis. Ann N Y Acad Sci 2014; 1335:63-77. [PMID: 25351294 DOI: 10.1111/nyas.12562] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Fueled by the growing interest in stem cell biology and the promise of regenerative medicine, study of the hematopoietic stem cell (HSC) microenvironment has provided critical insights into normal and malignant hematopoiesis. Notch receptor signaling in this microenvironment is a critical regulator of HSC fate and differentiation. Notch signaling also has the potential to modulate the growth of various malignant cell types, as evidenced by the growing list of hematologic cancers and other malignancies associated with either mutations in Notch genes or alterations in Notch signaling. In both health and disease, activation of Notch signaling predominantly exerts influence through stromal cell interactions with the tumor or stem cell microenvironments. Definitive evidence from transgenic mouse models has shown that alterations in stromal cell signaling from the bone marrow niche can induce malignant outgrowth of preleukemic clones and leukemia. Understanding how Notch receptor signals in the bone marrow microenvironment govern stem cell behavior will advance our understanding of cancer pathogenesis in hematologic malignancies and may have implications for treating metastatic solid tumors involving bone. These microenvironmental interactions are potential therapeutic targets for treating and preventing a variety of diseases, including bone marrow failure disorders, myelodysplastic syndromes, leukemia, and lymphoma.
Collapse
Affiliation(s)
- Andrew G Evans
- Hematopathology Unit, Department of Pathology, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | | |
Collapse
|
43
|
Durinck K, Wallaert A, Van de Walle I, Van Loocke W, Volders PJ, Vanhauwaert S, Geerdens E, Benoit Y, Van Roy N, Poppe B, Soulier J, Cools J, Mestdagh P, Vandesompele J, Rondou P, Van Vlierberghe P, Taghon T, Speleman F. The Notch driven long non-coding RNA repertoire in T-cell acute lymphoblastic leukemia. Haematologica 2014; 99:1808-16. [PMID: 25344525 DOI: 10.3324/haematol.2014.115683] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Genetic studies in T-cell acute lymphoblastic leukemia have uncovered a remarkable complexity of oncogenic and loss-of-function mutations. Amongst this plethora of genetic changes, NOTCH1 activating mutations stand out as the most frequently occurring genetic defect, identified in more than 50% of T-cell acute lymphoblastic leukemias, supporting a role as an essential driver for this gene in T-cell acute lymphoblastic leukemia oncogenesis. In this study, we aimed to establish a comprehensive compendium of the long non-coding RNA transcriptome under control of Notch signaling. For this purpose, we measured the transcriptional response of all protein coding genes and long non-coding RNAs upon pharmacological Notch inhibition in the human T-cell acute lymphoblastic leukemia cell line CUTLL1 using RNA-sequencing. Similar Notch dependent profiles were established for normal human CD34(+) thymic T-cell progenitors exposed to Notch signaling activity in vivo. In addition, we generated long non-coding RNA expression profiles (array data) from ex vivo isolated Notch active CD34(+) and Notch inactive CD4(+)CD8(+) thymocytes and from a primary cohort of 15 T-cell acute lymphoblastic leukemia patients with known NOTCH1 mutation status. Integration of these expression datasets with publicly available Notch1 ChIP-sequencing data resulted in the identification of long non-coding RNAs directly regulated by Notch activity in normal and malignant T cells. Given the central role of Notch in T-cell acute lymphoblastic leukemia oncogenesis, these data pave the way for the development of novel therapeutic strategies that target hyperactive Notch signaling in human T-cell acute lymphoblastic leukemia.
Collapse
Affiliation(s)
- Kaat Durinck
- Center for Medical Genetics, Ghent University, Belgium;
| | | | - Inge Van de Walle
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, Ghent, Belgium
| | | | | | | | - Ellen Geerdens
- Laboratory for the Molecular Biology of Leukemia, Center for Human Genetics, KU Leuven and Center for the Biology of Disease, VIB, Leuven, Belgium
| | - Yves Benoit
- Center for Medical Genetics, Ghent University, Belgium
| | | | - Bruce Poppe
- Center for Medical Genetics, Ghent University, Belgium
| | - Jean Soulier
- Genome Rearrangements and Cancer Laboratory, U944 INSERM, University Paris Diderot and Hematology Laboratory, Saint-Louis Hospital, Paris, France
| | - Jan Cools
- Laboratory for the Molecular Biology of Leukemia, Center for Human Genetics, KU Leuven and Center for the Biology of Disease, VIB, Leuven, Belgium
| | | | | | - Pieter Rondou
- Center for Medical Genetics, Ghent University, Belgium
| | | | - Tom Taghon
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, Ghent, Belgium
| | | |
Collapse
|
44
|
Maemura K, Natsugoe S, Takao S. Molecular mechanism of cholangiocarcinoma carcinogenesis. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2014; 21:754-760. [DOI: 10.1002/jhbp.126] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Kosei Maemura
- Department of Digestive Surgery, Breast and Thyroid Surgery; Kagoshima University Graduate School of Medical and Dental Sciences; Kagoshima Japan
| | - Shoji Natsugoe
- Department of Digestive Surgery, Breast and Thyroid Surgery; Kagoshima University Graduate School of Medical and Dental Sciences; Kagoshima Japan
| | - Sonshin Takao
- Center for Biomedical Science and Swine Research; Kagoshima University; 8-35-1 Sakuragaoka Kagoshima 890-8520 Japan
| |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW Recent advances in sequencing technology have led to a deeper and more comprehensive understanding of the molecular biology of pancreatic ductal adenocarcinoma. This timely review seeks to summarize these recent advances which will provide a foundation for future studies in the field. RECENT FINDINGS Stereotypical genetic alterations have been identified and confirmed. However, additional alterations have highlighted the importance and complexity of a number of intracellular signaling pathways that present unique opportunities for therapeutic targeting. SUMMARY A genetic signature of pancreatic ductal adenocarcinoma has been identified. This recent and important work is currently in the process of being applied in many clinical applications from early diagnostics to customized therapeutic regimens for this disease. A fundamental understanding of these findings will thus be of utmost importance for future research in the field and in the clinical care of patients with this lethal disease.
Collapse
|
46
|
Arasada RR, Amann JM, Rahman MA, Huppert SS, Carbone DP. EGFR blockade enriches for lung cancer stem-like cells through Notch3-dependent signaling. Cancer Res 2014; 74:5572-84. [PMID: 25125655 DOI: 10.1158/0008-5472.can-13-3724] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mutations in the epidermal growth factor receptor (EGFR) are the most common actionable genetic abnormalities yet discovered in lung cancer. However, targeting these mutations with kinase inhibitors is not curative in advanced disease and has yet to demonstrate an impact on potentially curable, early-stage disease, with some data suggesting adverse outcomes. Here, we report that treatment of EGFR-mutated lung cancer cell lines with erlotinib, while showing robust cell death, enriches the ALDH(+) stem-like cells through EGFR-dependent activation of Notch3. In addition, we demonstrate that erlotinib treatment increases the clonogenicity of lung cancer cells in a sphere-forming assay, suggesting increased stem-like cell potential. We demonstrate that inhibition of EGFR kinase activity leads to activation of Notch transcriptional targets in a γ secretase inhibitor-sensitive manner and causes Notch activation, leading to an increase in ALDH high(+) cells. We also find a kinase-dependent physical association between the Notch3 and EGFR receptors and tyrosine phosphorylation of Notch3. This could explain the worsened survival observed in some studies of erlotinib treatment at early-stage disease, and suggests that specific dual targeting might overcome this adverse effect.
Collapse
Affiliation(s)
- Rajeswara Rao Arasada
- Department of Internal Medicine, The Ohio State University Medical Center, Columbus, Ohio
| | - Joseph M Amann
- Department of Internal Medicine, The Ohio State University Medical Center, Columbus, Ohio
| | - Mohammad A Rahman
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Center for Critical Care Medicine, The Ohio State University Medical Center, Columbus, Ohio
| | - Stacey S Huppert
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio.
| | - David P Carbone
- Department of Internal Medicine, The Ohio State University Medical Center, Columbus, Ohio.
| |
Collapse
|
47
|
Zhang Q, Zhang Y, Wu L, Yang Y, Li X, Gao L, Hou X, Wu Y, Hou G, Li Z, Lin X. dBrms1 Acts as a Positive Regulator of Notch Signaling in Drosophila Wing. J Genet Genomics 2014; 41:317-25. [DOI: 10.1016/j.jgg.2014.04.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 04/01/2014] [Accepted: 04/10/2014] [Indexed: 01/25/2023]
|
48
|
De Salvo M, Raimondi L, Vella S, Adesso L, Ciarapica R, Verginelli F, Pannuti A, Citti A, Boldrini R, Milano GM, Cacchione A, Ferrari A, Collini P, Rosolen A, Bisogno G, Alaggio R, Inserra A, Locatelli M, Stifani S, Screpanti I, Miele L, Locatelli F, Rota R. Hyper-activation of Notch3 amplifies the proliferative potential of rhabdomyosarcoma cells. PLoS One 2014; 9:e96238. [PMID: 24797362 PMCID: PMC4010457 DOI: 10.1371/journal.pone.0096238] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 04/04/2014] [Indexed: 11/18/2022] Open
Abstract
Rhabdomyosarcoma (RMS) is a pediatric myogenic-derived soft tissue sarcoma that includes two major histopathological subtypes: embryonal and alveolar. The majority of alveolar RMS expresses PAX3-FOXO1 fusion oncoprotein, associated with the worst prognosis. RMS cells show myogenic markers expression but are unable to terminally differentiate. The Notch signaling pathway is a master player during myogenesis, with Notch1 activation sustaining myoblast expansion and Notch3 activation inhibiting myoblast fusion and differentiation. Accordingly, Notch1 signaling is up-regulated and activated in embryonal RMS samples and supports the proliferation of tumor cells. However, it is unable to control their differentiation properties. We previously reported that Notch3 is activated in RMS cell lines, of both alveolar and embryonal subtype, and acts by inhibiting differentiation. Moreover, Notch3 depletion reduces PAX3-FOXO1 alveolar RMS tumor growth in vivo. However, whether Notch3 activation also sustains the proliferation of RMS cells remained unclear. To address this question, we forced the expression of the activated form of Notch3, Notch3IC, in the RH30 and RH41 PAX3-FOXO1-positive alveolar and in the RD embryonal RMS cell lines and studied the proliferation of these cells. We show that, in all three cell lines tested, Notch3IC over-expression stimulates in vitro cell proliferation and prevents the effects of pharmacological Notch inhibition. Furthermore, Notch3IC further increases RH30 cell growth in vivo. Interestingly, knockdown of Notch canonical ligands JAG1 or DLL1 in RMS cell lines decreases Notch3 activity and reduces cell proliferation. Finally, the expression of Notch3IC and its target gene HES1 correlates with that of the proliferative marker Ki67 in a small cohort of primary PAX-FOXO1 alveolar RMS samples. These results strongly suggest that high levels of Notch3 activation increase the proliferative potential of RMS cells.
Collapse
MESH Headings
- Cell Line, Tumor
- Cell Proliferation
- Gene Expression Regulation, Neoplastic
- Humans
- Ki-67 Antigen/genetics
- Ki-67 Antigen/metabolism
- Oncogene Proteins, Fusion/biosynthesis
- Oncogene Proteins, Fusion/genetics
- Paired Box Transcription Factors/biosynthesis
- Paired Box Transcription Factors/genetics
- Receptor, Notch1/genetics
- Receptor, Notch1/metabolism
- Receptor, Notch3
- Receptors, Notch/genetics
- Receptors, Notch/metabolism
- Rhabdomyosarcoma, Alveolar/genetics
- Rhabdomyosarcoma, Alveolar/metabolism
- Rhabdomyosarcoma, Alveolar/pathology
- Rhabdomyosarcoma, Embryonal/genetics
- Rhabdomyosarcoma, Embryonal/metabolism
- Rhabdomyosarcoma, Embryonal/pathology
- Signal Transduction
Collapse
Affiliation(s)
- Maria De Salvo
- Department of Oncohematology, Ospedale Pediatrico Bambino Gesù, IRCCS, Roma, Italy
| | - Lavinia Raimondi
- Department of Oncohematology, Ospedale Pediatrico Bambino Gesù, IRCCS, Roma, Italy
| | - Serena Vella
- Department of Oncohematology, Ospedale Pediatrico Bambino Gesù, IRCCS, Roma, Italy
| | - Laura Adesso
- Department of Oncohematology, Ospedale Pediatrico Bambino Gesù, IRCCS, Roma, Italy
| | - Roberta Ciarapica
- Department of Oncohematology, Ospedale Pediatrico Bambino Gesù, IRCCS, Roma, Italy
| | - Federica Verginelli
- Department of Oncohematology, Ospedale Pediatrico Bambino Gesù, IRCCS, Roma, Italy
| | - Antonio Pannuti
- Stanley Scott Cancer Center, Louisiana State University Health Sciences Center and Louisiana Cancer Research Consortium, New Orleans, Louisiana, United States of America
| | - Arianna Citti
- Department of Pathology, Ospedale Pediatrico Bambino Gesù, IRCCS, Roma, Italy
| | - Renata Boldrini
- Department of Pathology, Ospedale Pediatrico Bambino Gesù, IRCCS, Roma, Italy
| | - Giuseppe M. Milano
- Department of Oncohematology, Ospedale Pediatrico Bambino Gesù, IRCCS, Roma, Italy
| | - Antonella Cacchione
- Department of Oncohematology, Ospedale Pediatrico Bambino Gesù, IRCCS, Roma, Italy
| | - Andrea Ferrari
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Paola Collini
- Anatomic Pathology Unit 2, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Angelo Rosolen
- Department of Pediatrics, Oncohematology Unit, University of Padova, Padova, Italy
| | - Gianni Bisogno
- Department of Pediatrics, Oncohematology Unit, University of Padova, Padova, Italy
| | - Rita Alaggio
- Department of Pathology, University of Padova, Padova, Italy
| | - Alessandro Inserra
- Department of Surgery, Ospedale Pediatrico Bambino Gesù, IRCCS, Roma, Italy
| | - Mattia Locatelli
- Department of Scientific Directorate, Ospedale Pediatrico Bambino Gesù, IRCCS, Roma, Italy
| | - Stefano Stifani
- Centre for Neuronal Survival, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | | | - Lucio Miele
- Stanley Scott Cancer Center, Louisiana State University Health Sciences Center and Louisiana Cancer Research Consortium, New Orleans, Louisiana, United States of America
| | - Franco Locatelli
- Department of Oncohematology, Ospedale Pediatrico Bambino Gesù, IRCCS, Roma, Italy
- Dipartimento di Scienze Pediatriche, Università di Pavia, Pavia, Italy
| | - Rossella Rota
- Department of Oncohematology, Ospedale Pediatrico Bambino Gesù, IRCCS, Roma, Italy
- * E-mail:
| |
Collapse
|
49
|
Sarode B, Nowell CS, Ihm J, Kostic C, Arsenijevic Y, Moulin AP, Schorderet DF, Beermann F, Radtke F. Notch signaling in the pigmented epithelium of the anterior eye segment promotes ciliary body development at the expense of iris formation. Pigment Cell Melanoma Res 2014; 27:580-9. [DOI: 10.1111/pcmr.12236] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 03/12/2014] [Indexed: 11/27/2022]
Affiliation(s)
- Bhushan Sarode
- School of Life Science; Ecole Polytechnique Fédérale de Lausanne (EPFL); Lausanne Switzerland
- Swiss Institute for Experimental Cancer Research (ISREC); Lausanne Switzerland
| | - Craig S. Nowell
- School of Life Science; Ecole Polytechnique Fédérale de Lausanne (EPFL); Lausanne Switzerland
- Swiss Institute for Experimental Cancer Research (ISREC); Lausanne Switzerland
| | - JongEun Ihm
- School of Life Science; Ecole Polytechnique Fédérale de Lausanne (EPFL); Lausanne Switzerland
| | - Corinne Kostic
- Unit of Gene Therapy and Stem Cell Biology; Jules-Gonin Eye Hospital; University of Lausanne; Lausanne Switzerland
- Eye Pathology Laboratory; Jules-Gonin Eye Hospital; University of Lausanne; Lausanne Switzerland
| | - Yvan Arsenijevic
- Unit of Gene Therapy and Stem Cell Biology; Jules-Gonin Eye Hospital; University of Lausanne; Lausanne Switzerland
- Eye Pathology Laboratory; Jules-Gonin Eye Hospital; University of Lausanne; Lausanne Switzerland
| | - Alexandre P. Moulin
- Eye Pathology Laboratory; Jules-Gonin Eye Hospital; University of Lausanne; Lausanne Switzerland
| | - Daniel F. Schorderet
- School of Life Science; Ecole Polytechnique Fédérale de Lausanne (EPFL); Lausanne Switzerland
- Eye Pathology Laboratory; Jules-Gonin Eye Hospital; University of Lausanne; Lausanne Switzerland
- IRO - Institute for Research in Ophthalmology; Sion Switzerland
| | - Friedrich Beermann
- School of Life Science; Ecole Polytechnique Fédérale de Lausanne (EPFL); Lausanne Switzerland
- Swiss Institute for Experimental Cancer Research (ISREC); Lausanne Switzerland
| | - Freddy Radtke
- School of Life Science; Ecole Polytechnique Fédérale de Lausanne (EPFL); Lausanne Switzerland
- Swiss Institute for Experimental Cancer Research (ISREC); Lausanne Switzerland
| |
Collapse
|
50
|
Palagani V, Bozko P, El Khatib M, Belahmer H, Giese N, Sipos B, Malek NP, Plentz RR. Combined inhibition of Notch and JAK/STAT is superior to monotherapies and impairs pancreatic cancer progression. Carcinogenesis 2014; 35:859-866. [DOI: 10.1093/carcin/bgt394] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|