1
|
Gao W, Gu K, Ma L, Yang F, Deng L, Zhang Y, Miao MZ, Li W, Li G, Qian H, Zhang Z, Wang G, Yu H, Liu X. Interstitial Fluid Shear Stress Induces the Synthetic Phenotype Switching of VSMCs to Release Pro-calcified Extracellular Vesicles via EGFR-MAPK-KLF5 Pathway. Int J Biol Sci 2024; 20:2727-2747. [PMID: 38725857 PMCID: PMC11077359 DOI: 10.7150/ijbs.90725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 04/20/2024] [Indexed: 05/12/2024] Open
Abstract
Phenotypic switching (from contractile to synthetic) of vascular smooth muscle cells (VSMCs) is essential in the progression of atherosclerosis. The damaged endothelium in the atherosclerotic artery exposes VSMCs to increased interstitial fluid shear stress (IFSS). However, the precise mechanisms by which increased IFSS influences VSMCs phenotypic switching are unrevealed. Here, we employed advanced numerical simulations to calculate IFSS values accurately based on parameters acquired from patient samples. We then carefully investigated the phenotypic switching and extracellular vesicles (EVs) secretion of VSMCs under various IFSS conditions. By employing a comprehensive set of approaches, we found that VSMCs exhibited synthetic phenotype upon atherosclerotic IFSS. This synthetic phenotype is the upstream regulator for the enhanced secretion of pro-calcified EVs. Mechanistically, as a mechanotransducer, the epidermal growth factor receptor (EGFR) initiates the flow-based mechanical cues to MAPK signaling pathway, facilitating the nuclear accumulation of the transcription factor krüppel-like factor 5 (KLF5). Furthermore, pharmacological inhibiting either EGFR or MAPK signaling pathway blocks the nuclear accumulation of KLF5 and finally results in the maintenance of contractile VSMCs even under increased IFSS stimulation. Collectively, targeting this signaling pathway holds potential as a novel therapeutic strategy to inhibit VSMCs phenotypic switching and mitigate the progression of atherosclerosis.
Collapse
Affiliation(s)
- Wenbo Gao
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Kaiyun Gu
- Department of Cardiac Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Lunjie Ma
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Fan Yang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Li Deng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Yaojia Zhang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Michael Z. Miao
- Division of Oral & Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, NC, 27599, USA
| | - Wenjun Li
- Division of Oral & Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, NC, 27599, USA
| | - Gang Li
- Department of Genome Sciences, University of Washington, William H. Foege Hall, 3720 15th Ave NE, Seattle 98195, USA
| | - Hong Qian
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Zhen Zhang
- Department of Cardiology, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
- JinFeng Laboratory, Chongqing 401329, China
| | - Hongchi Yu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| |
Collapse
|
2
|
Su A, Yao K, Zhang H, Wang Y, Zhang H, Tang J. DANCR Induces Cisplatin Resistance of Triple-Negative Breast Cancer by KLF5/p27 Signaling. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:248-258. [PMID: 36509121 DOI: 10.1016/j.ajpath.2022.11.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/14/2022]
Abstract
An increasing body of evidence suggests that long noncoding RNAs play critical roles in human cancer. Breast cancer is a heterogeneous disease and the potential involvement of long noncoding RNAs in breast cancer remains poorly understood. Herein, the study identified a long noncoding RNA, DANCR, which promotes cisplatin chemoresistance in triple-negative breast cancer (TNBC) cells. Mechanistically, binding of DANCR to Krüppel-like factor 5 (KLF5) induced acetylation of KLF5 at lysine 369 (K369), and DANCR knockdown resulted in down-regulation of KLF5 protein levels. Furthermore, DANCR/KLF5 signaling pathway induced hypersensitivity to cisplatin in chemoresistant patients by inhibiting p27 transcription. In summary, this study reinforced the potential presence of a growth regulatory network in TNBC cells, and documented a DANCR/KLF5/p27 signaling pathway mediating cisplatin chemoresistance in TNBC.
Collapse
Affiliation(s)
- Anchen Su
- The First Hospital of Lanzhou University, Gansu International Scientific and Technological Cooperation Base of Reproductive Medicine Transformation Application, Lanzhou, China
| | - Kun Yao
- Gansu Provincial Hospital, Lanzhou, China
| | - Hanru Zhang
- Gynecology and Obstetrics, Gansu Provincial Maternity and Child Care Hospital, Lanzhou, China
| | - Yiqing Wang
- The First Hospital of Lanzhou University, Gansu International Scientific and Technological Cooperation Base of Reproductive Medicine Transformation Application, Lanzhou, China
| | - Haibo Zhang
- Oncology Center, Department of Radiation Oncology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China.
| | - Jianming Tang
- The First Hospital of Lanzhou University, Gansu International Scientific and Technological Cooperation Base of Reproductive Medicine Transformation Application, Lanzhou, China.
| |
Collapse
|
3
|
Houschyar KS, Borrelli MR, Rein S, Tapking C, Popp D, Puladi B, Ooms M, Schulz T, Maan ZN, Branski LK, Siemers F, Philipp-Dormston WG, Yazdi AS, Duscher D. Wnt ligand expression in malignant melanoma: new insights. EUROPEAN JOURNAL OF PLASTIC SURGERY 2022. [DOI: 10.1007/s00238-022-01941-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
4
|
Sachs M, Wetzel S, Reichelt J, Sachs W, Schebsdat L, Zielinski S, Seipold L, Heintz L, Müller SA, Kretz O, Lindenmeyer M, Wiech T, Huber TB, Lüllmann-Rauch R, Lichtenthaler SF, Saftig P, Meyer-Schwesinger C. ADAM10-Mediated Ectodomain Shedding Is an Essential Driver of Podocyte Damage. J Am Soc Nephrol 2021; 32:1389-1408. [PMID: 33785583 PMCID: PMC8259650 DOI: 10.1681/asn.2020081213] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 02/08/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Podocytes embrace the glomerular capillaries with foot processes, which are interconnected by a specialized adherens junction to ultimately form the filtration barrier. Altered adhesion and loss are common features of podocyte injury, which could be mediated by shedding of cell-adhesion molecules through the regulated activity of cell surface-expressed proteases. A Disintegrin and Metalloproteinase 10 (ADAM10) is such a protease known to mediate ectodomain shedding of adhesion molecules, among others. Here we evaluate the involvement of ADAM10 in the process of antibody-induced podocyte injury. METHODS Membrane proteomics, immunoblotting, high-resolution microscopy, and immunogold electron microscopy were used to analyze human and murine podocyte ADAM10 expression in health and kidney injury. The functionality of ADAM10 ectodomain shedding for podocyte development and injury was analyzed, in vitro and in vivo, in the anti-podocyte nephritis (APN) model in podocyte-specific, ADAM10-deficient mice. RESULTS ADAM10 is selectively localized at foot processes of murine podocytes and its expression is dispensable for podocyte development. Podocyte ADAM10 expression is induced in the setting of antibody-mediated injury in humans and mice. Podocyte ADAM10 deficiency attenuates the clinical course of APN and preserves the morphologic integrity of podocytes, despite subepithelial immune-deposit formation. Functionally, ADAM10-related ectodomain shedding results in cleavage of the cell-adhesion proteins N- and P-cadherin, thus decreasing their injury-related surface levels. This favors podocyte loss and the activation of downstream signaling events through the Wnt signaling pathway in an ADAM10-dependent manner. CONCLUSIONS ADAM10-mediated ectodomain shedding of injury-related cadherins drives podocyte injury.
Collapse
Affiliation(s)
- Marlies Sachs
- Institute of Cellular and Integrative Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sebastian Wetzel
- Institute of Biochemistry, Christian-Albrechts University Kiel, Kiel, Germany
| | - Julia Reichelt
- Institute of Cellular and Integrative Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Wiebke Sachs
- Institute of Cellular and Integrative Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lisa Schebsdat
- Institute of Cellular and Integrative Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stephanie Zielinski
- Institute of Cellular and Integrative Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lisa Seipold
- Institute of Biochemistry, Christian-Albrechts University Kiel, Kiel, Germany
| | - Lukas Heintz
- Institute of Cellular and Integrative Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stephan A. Müller
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - Oliver Kretz
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maja Lindenmeyer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Wiech
- Nephropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias B. Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Stefan F. Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Paul Saftig
- Institute of Biochemistry, Christian-Albrechts University Kiel, Kiel, Germany
| | - Catherine Meyer-Schwesinger
- Institute of Cellular and Integrative Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
5
|
Li Y, Kong R, Chen H, Zhao Z, Li L, Li J, Hu J, Zhang G, Pan S, Wang Y, Wang G, Chen H, Sun B. Overexpression of KLF5 is associated with poor survival and G1/S progression in pancreatic cancer. Aging (Albany NY) 2020; 11:5035-5057. [PMID: 31327760 PMCID: PMC6682527 DOI: 10.18632/aging.102096] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 07/11/2019] [Indexed: 01/05/2023]
Abstract
Despite improvements in surgical procedures and comprehensive therapies, pancreatic cancer remains one of the most aggressive and deadly human malignancies. It is therefore necessary to determine which cellular mediators associate with prognosis in pancreatic cancer so as to improve the treatment of this disease. In the present study, mRNA array and immunohistochemical analyses showed that KLF5 is highly expressed in tissue samples from three short-surviving patients with pancreatic cancer. Survival analysis using data from The Cancer Genome Atlas showed that patients highly expressing KLF5 exhibited shorter overall and tumor-free survival times. Mechanistically, KLF5 promoted expression of E2F1, cyclin D1 and Rad51, while inhibiting expression of p16 in pancreatic cancer cells. Finally, flow cytometric analyses verified that KLF5 promotes G1/S progression of the cell cycle in pancreatic cancer cells. Collectively, these findings demonstrate that KLF5 is an important prognostic biomarker in pancreatic cancer patients, and they shed light on the molecular mechanism by which KLF5 stimulates cell cycle progression in pancreatic cancer.
Collapse
Affiliation(s)
- Yilong Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin 150001, China
| | - Rui Kong
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin 150001, China
| | - Hongze Chen
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin 150001, China
| | - Zhongjie Zhao
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin 150001, China
| | - Le Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin 150001, China
| | - Jiating Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin 150001, China
| | - Jisheng Hu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin 150001, China
| | - Guangquan Zhang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin 150001, China
| | - Shangha Pan
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin 150001, China
| | - Yongwei Wang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin 150001, China
| | - Gang Wang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin 150001, China
| | - Hua Chen
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin 150001, China
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin 150001, China
| |
Collapse
|
6
|
Okada H, Yamada M, Kamimoto K, Kok CYY, Kaneko K, Ema M, Miyajima A, Itoh T. The transcription factor Klf5 is essential for intrahepatic biliary epithelial tissue remodeling after cholestatic liver injury. J Biol Chem 2018. [PMID: 29523685 DOI: 10.1074/jbc.ra118.002372] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Under various conditions of liver injury, the intrahepatic biliary epithelium undergoes dynamic tissue expansion and remodeling, a process known as ductular reaction. Mouse models defective in inducing such a tissue-remodeling process are more susceptible to liver injury, suggesting a crucial role of this process in liver regeneration. However, the molecular mechanisms regulating the biliary epithelial cell (BEC) dynamics in the ductular reaction remain largely unclear. Here, we demonstrate that the transcription factor Krüppel-like factor 5 (Klf5) is highly enriched in mouse liver BECs and plays a key role in regulating the ductular reaction, specifically under cholestatic injury conditions. Although mice lacking Klf5 in the entire liver epithelium, including both hepatocytes and BECs (Klf5-LKO (liver epithelial-specific knockout) mice), did not exhibit any apparent phenotype in the hepatobiliary system under normal conditions, they exhibited significant defects in biliary epithelial tissue remodeling upon 3,5-diethoxycarbonyl-1,4-dihydrocollidine-induced cholangitis, concomitantly with exacerbated cholestasis and reduced survival rate. In contrast, mice lacking Klf5 solely in hepatocytes did not exhibit any such phenotypes, confirming Klf5's specific role in BECs. RNA-sequencing analyses of BECs isolated from the Klf5-LKO mouse livers revealed that the Klf5 deficiency primarily affected expression of cell cycle-related genes. Moreover, immunostaining analysis with the proliferation marker Ki67 disclosed that the Klf5-LKO mice had significantly reduced BEC proliferation levels upon injury. These results indicate that Klf5 plays a critical role in the ductular reaction and biliary epithelial tissue expansion and remodeling by inducing BEC proliferation and thereby contributing to liver regeneration.
Collapse
Affiliation(s)
- Hajime Okada
- From the Laboratory of Cell Growth and Differentiation, Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032 and
| | - Minami Yamada
- From the Laboratory of Cell Growth and Differentiation, Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032 and
| | - Kenji Kamimoto
- From the Laboratory of Cell Growth and Differentiation, Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032 and
| | - Cindy Yuet-Yin Kok
- From the Laboratory of Cell Growth and Differentiation, Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032 and
| | - Kota Kaneko
- From the Laboratory of Cell Growth and Differentiation, Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032 and
| | - Masatsugu Ema
- the Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Seta, Tsukinowa-cho, Otsu, Shiga 520-2192, Japan
| | - Atsushi Miyajima
- From the Laboratory of Cell Growth and Differentiation, Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032 and
| | - Tohru Itoh
- From the Laboratory of Cell Growth and Differentiation, Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032 and
| |
Collapse
|
7
|
Li Z, Dong J, Zou T, Du C, Li S, Chen C, Liu R, Wang K. Dexamethasone induces docetaxel and cisplatin resistance partially through up-regulating Krüppel-like factor 5 in triple-negative breast cancer. Oncotarget 2017; 8:11555-11565. [PMID: 28030791 PMCID: PMC5355285 DOI: 10.18632/oncotarget.14135] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 11/22/2016] [Indexed: 11/25/2022] Open
Abstract
PURPOSE Dexamethasone (Dex), a glucocorticoid (GC), is used as a pretreatment drug in cancer patients undergoing chemotherapy. Dex functions by binding to the glucocorticoid receptor (GR) to prevent allergic reactions and severe chemotherapeutic side effects such as nausea and vomiting. However, the mechanisms by which Dex causes chemoresistance remain unknown. METHODS We used docetaxel and cisplatin to treat triple-negative breast cancer (TNBC) cells with or without Dex and assessed cell proliferation using a sulforhodamine B colorimetric (SRB) assay. Additionally, Western blotting was employed to measure Krüppel-like factor 5 (KLF5), GR and several apoptosis-related proteins. To determine how the GR regulates KLF5, we used qRT-PCR, luciferase reporter assays and ChIP assays. Finally, we detected the involvement of Dex in TNBC chemotherapeutic resistance using HCC1806 xenograft model in vivo. RESULTS In this study, we demonstrated that Dex induces docetaxel and cisplatin resistance in TNBC cells in vitro and in vivo. Dex up-regulates pro-survival transcription factor KLF5 expression at both mRNA and protein levels dependent on GR. Importantly, Dex failed to promote cancer cell survival and tumor growth when KLF5 induction was blocked. CONCLUSIONS We conclude that KLF5 is a Dex-induced gene that contributes to Dex-mediated drug chemoresistance, providing a potential novel target for TNBC treatment.
Collapse
Affiliation(s)
- Zhen Li
- Department of Gastrointestinal and Hernia Surgery, Institute of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China
- Kunming Digestive Disease Treatment Engineering Technology Center, Kunming, Yunnan 650032, China
| | - Jian Dong
- Department of Oncology, Yunnan Tumor Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650118, China
| | - Tianning Zou
- Department of Breast Surgery, Yunnan Tumor Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650118, China
| | - Chengzhi Du
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China
| | - Siyuan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China
| | - Ceshi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China
| | - Rong Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China
| | - Kunhua Wang
- Department of Gastrointestinal and Hernia Surgery, Institute of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
- Kunming Digestive Disease Treatment Engineering Technology Center, Kunming, Yunnan 650032, China
| |
Collapse
|
8
|
Early Transcriptional Changes Induced by Wnt/ β-Catenin Signaling in Hippocampal Neurons. Neural Plast 2016; 2016:4672841. [PMID: 28116168 PMCID: PMC5223035 DOI: 10.1155/2016/4672841] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/20/2016] [Accepted: 11/27/2016] [Indexed: 01/04/2023] Open
Abstract
Wnt/β-catenin signaling modulates brain development and function and its deregulation underlies pathological changes occurring in neurodegenerative and neurodevelopmental disorders. Since one of the main effects of Wnt/β-catenin signaling is the modulation of target genes, in the present work we examined global transcriptional changes induced by short-term Wnt3a treatment (4 h) in primary cultures of rat hippocampal neurons. RNAseq experiments allowed the identification of 170 differentially expressed genes, including known Wnt/β-catenin target genes such as Notum, Axin2, and Lef1, as well as novel potential candidates Fam84a, Stk32a, and Itga9. Main biological processes enriched with differentially expressed genes included neural precursor (GO:0061364, p-adjusted = 2.5 × 10−7), forebrain development (GO:0030900, p-adjusted = 7.3 × 10−7), and stem cell differentiation (GO:0048863 p-adjusted = 7.3 × 10−7). Likewise, following activation of the signaling cascade, the expression of a significant number of genes with transcription factor activity (GO:0043565, p-adjusted = 4.1 × 10−6) was induced. We also studied molecular networks enriched upon Wnt3a activation and detected three highly significant expression modules involved in glycerolipid metabolic process (GO:0046486, p-adjusted = 4.5 × 10−19), learning or memory (GO:0007611, p-adjusted = 4.0 × 10−5), and neurotransmitter secretion (GO:0007269, p-adjusted = 5.3 × 10−12). Our results indicate that Wnt/β-catenin mediated transcription controls multiple biological processes related to neuronal structure and activity that are affected in synaptic dysfunction disorders.
Collapse
|
9
|
Lenfert E, Maenz C, Heinlein C, Jannasch K, Schumacher U, Pantel K, Tolstonog GV, Deppert W, Wegwitz F. Mutant p53 promotes epithelial-mesenchymal plasticity and enhances metastasis in mammary carcinomas of WAP-T mice. Int J Cancer 2014; 136:E521-33. [PMID: 25195563 DOI: 10.1002/ijc.29186] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 07/31/2014] [Accepted: 08/21/2014] [Indexed: 12/20/2022]
Abstract
To study the postulated mutant p53 (mutp53) "gain of function" effects in mammary tumor development, progression and metastasis, we crossed SV40 transgenic WAP-T mice with mutant p53 transgenic WAP-mutp53 mice. Compared to tumors in monotransgenic WAP-T mice, tumors in bitransgenic WAP-T x WAP-mutp53 mice showed higher tumor grading, enhanced vascularization, and significantly increased metastasis. Bitransgenic tumors revealed a gene signature associated with the oncogenic epithelial-mesenchymal transition pathway (EMT gene signature). In cultures of WAP-T tumor-derived G-2 cancer cells, which are comprised of subpopulations displaying "mesenchymal" and "epithelial" phenotypes, this EMT gene signature was associated with the "mesenchymal" compartment. Furthermore, ectopic expression of mutp53 in G-2 cells sufficed to induce a strong EMT phenotype. In contrast to these in vitro effects, monotransgenic and bitransgenic tumors were phenotypically similar suggesting that in vivo the tumor cell phenotype might be under control of the tumor microenvironment. In support, orthotopic transplantation of G-2 cells as well as of G-2 cells expressing ectopic mutp53 into syngeneic mice resulted in tumors with a predominantly epithelial phenotype, closely similar to that of endogenous primary tumors. We conclude that induction of an EMT gene signature by mutp53 in bitransgenic tumors primarily promotes tumor cell plasticity, that is, the probability of tumor cells to undergo EMT processes under appropriate stimuli, thereby possibly increasing their potential to disseminate and metastasize.
Collapse
Affiliation(s)
- Eva Lenfert
- Department for Tumor Biology, University Medical Center Hamburg-Eppendorf (UKE), D-20246, Hamburg, Germany; Department of Tumor Virology, Heinrich-Pette-Institute, Leibniz Institute for Experimental Virology, D-20251, Hamburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Nakaya T, Ogawa S, Manabe I, Tanaka M, Sanada M, Sato T, Taketo MM, Nakao K, Clevers H, Fukayama M, Kuroda M, Nagai R. KLF5 regulates the integrity and oncogenicity of intestinal stem cells. Cancer Res 2014; 74:2882-91. [PMID: 24626089 DOI: 10.1158/0008-5472.can-13-2574] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The intestinal epithelium maintains homeostasis by a self-renewal process involving resident stem cells, including Lgr5(+) crypt-base columnar cells, but core mechanisms and their contributions to intestinal cancer are not fully defined. In this study, we examined a hypothesized role for KLF5, a zinc-finger transcription factor that is critical to maintain the integrity of embryonic and induced pluripotent stem cells, in intestinal stem-cell integrity and cancer in the mouse. Klf5 was indispensable for the integrity and oncogenic transformation of intestinal stem cells. In mice, inducible deletion of Klf5 in Lgr5(+) stem cells suppressed their proliferation and survival in a manner associated with nuclear localization of β-catenin (Catnb), generating abnormal apoptotic cells in intestinal crypts. Moreover, production of lethal adenomas and carcinomas by specific expression of an oncogenic mutant of β-catenin in Lgr5(+) stem cells was suppressed completely by Klf5 deletion in the same cells. Given that activation of the Wnt/β-catenin pathway is the most frequently altered pathway in human colorectal cancer, our results argue that KLF5 acts as a fundamental core regulator of intestinal oncogenesis at the stem-cell level, and they suggest KLF5 targeting as a rational strategy to eradicate stem-like cells in colorectal cancer.
Collapse
Affiliation(s)
- Takeo Nakaya
- Authors' Affiliations: Department of Molecular Pathology and Translational Research Unit, Tokyo Medical University; Cancer Genomics Project; Departments of Pathology, and Cardiovascular Medicine; Animal Resources, Graduate School of Medicine, University of Tokyo; Honeybee Science Research Center, Tamagawa University; Department of Gastroenterology, School of Medicine, Keio University, Tokyo; Department of Pharmacology, Graduate School of Medicine, Kyoto University, Kyoto; Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Kobe; Jichi Medical University, Shimotsuke, Tochigi, Japan; and Hubrecht Institute and KNAW, Utrecht, the NetherlandsAuthors' Affiliations: Department of Molecular Pathology and Translational Research Unit, Tokyo Medical University; Cancer Genomics Project; Departments of Pathology, and Cardiovascular Medicine; Animal Resources, Graduate School of Medicine, University of Tokyo; Honeybee Science Research Center, Tamagawa University; Department of Gastroenterology, School of Medicine, Keio University, Tokyo; Department of Pharmacology, Graduate School of Medicine, Kyoto University, Kyoto; Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Kobe; Jichi Medical University, Shimotsuke, Tochigi, Japan; and Hubrecht Institute and KNAW, Utrecht, the NetherlandsAuthors' Affiliations: Department of Molecular Pathology and Translational Research Unit, Tokyo Medical University; Cancer Genomics Project; Departments of Pathology, and Cardiovascular Medicine; Animal Resources, Graduate School of Medicine, University of Tokyo; Honeybee Science Research Center, Tamagawa University; Department of Gastroenterology, School of Medicine, Keio University, Tokyo; Department of Pharmacology, Graduate School of Medicine, Kyoto University, Kyoto; Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Kobe; Jichi Medical University, Shimotsuke
| | - Seishi Ogawa
- Authors' Affiliations: Department of Molecular Pathology and Translational Research Unit, Tokyo Medical University; Cancer Genomics Project; Departments of Pathology, and Cardiovascular Medicine; Animal Resources, Graduate School of Medicine, University of Tokyo; Honeybee Science Research Center, Tamagawa University; Department of Gastroenterology, School of Medicine, Keio University, Tokyo; Department of Pharmacology, Graduate School of Medicine, Kyoto University, Kyoto; Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Kobe; Jichi Medical University, Shimotsuke, Tochigi, Japan; and Hubrecht Institute and KNAW, Utrecht, the Netherlands
| | - Ichiro Manabe
- Authors' Affiliations: Department of Molecular Pathology and Translational Research Unit, Tokyo Medical University; Cancer Genomics Project; Departments of Pathology, and Cardiovascular Medicine; Animal Resources, Graduate School of Medicine, University of Tokyo; Honeybee Science Research Center, Tamagawa University; Department of Gastroenterology, School of Medicine, Keio University, Tokyo; Department of Pharmacology, Graduate School of Medicine, Kyoto University, Kyoto; Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Kobe; Jichi Medical University, Shimotsuke, Tochigi, Japan; and Hubrecht Institute and KNAW, Utrecht, the Netherlands
| | - Masami Tanaka
- Authors' Affiliations: Department of Molecular Pathology and Translational Research Unit, Tokyo Medical University; Cancer Genomics Project; Departments of Pathology, and Cardiovascular Medicine; Animal Resources, Graduate School of Medicine, University of Tokyo; Honeybee Science Research Center, Tamagawa University; Department of Gastroenterology, School of Medicine, Keio University, Tokyo; Department of Pharmacology, Graduate School of Medicine, Kyoto University, Kyoto; Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Kobe; Jichi Medical University, Shimotsuke, Tochigi, Japan; and Hubrecht Institute and KNAW, Utrecht, the NetherlandsAuthors' Affiliations: Department of Molecular Pathology and Translational Research Unit, Tokyo Medical University; Cancer Genomics Project; Departments of Pathology, and Cardiovascular Medicine; Animal Resources, Graduate School of Medicine, University of Tokyo; Honeybee Science Research Center, Tamagawa University; Department of Gastroenterology, School of Medicine, Keio University, Tokyo; Department of Pharmacology, Graduate School of Medicine, Kyoto University, Kyoto; Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Kobe; Jichi Medical University, Shimotsuke, Tochigi, Japan; and Hubrecht Institute and KNAW, Utrecht, the Netherlands
| | - Masashi Sanada
- Authors' Affiliations: Department of Molecular Pathology and Translational Research Unit, Tokyo Medical University; Cancer Genomics Project; Departments of Pathology, and Cardiovascular Medicine; Animal Resources, Graduate School of Medicine, University of Tokyo; Honeybee Science Research Center, Tamagawa University; Department of Gastroenterology, School of Medicine, Keio University, Tokyo; Department of Pharmacology, Graduate School of Medicine, Kyoto University, Kyoto; Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Kobe; Jichi Medical University, Shimotsuke, Tochigi, Japan; and Hubrecht Institute and KNAW, Utrecht, the Netherlands
| | - Toshiro Sato
- Authors' Affiliations: Department of Molecular Pathology and Translational Research Unit, Tokyo Medical University; Cancer Genomics Project; Departments of Pathology, and Cardiovascular Medicine; Animal Resources, Graduate School of Medicine, University of Tokyo; Honeybee Science Research Center, Tamagawa University; Department of Gastroenterology, School of Medicine, Keio University, Tokyo; Department of Pharmacology, Graduate School of Medicine, Kyoto University, Kyoto; Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Kobe; Jichi Medical University, Shimotsuke, Tochigi, Japan; and Hubrecht Institute and KNAW, Utrecht, the Netherlands
| | - Makoto M Taketo
- Authors' Affiliations: Department of Molecular Pathology and Translational Research Unit, Tokyo Medical University; Cancer Genomics Project; Departments of Pathology, and Cardiovascular Medicine; Animal Resources, Graduate School of Medicine, University of Tokyo; Honeybee Science Research Center, Tamagawa University; Department of Gastroenterology, School of Medicine, Keio University, Tokyo; Department of Pharmacology, Graduate School of Medicine, Kyoto University, Kyoto; Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Kobe; Jichi Medical University, Shimotsuke, Tochigi, Japan; and Hubrecht Institute and KNAW, Utrecht, the Netherlands
| | - Kazuki Nakao
- Authors' Affiliations: Department of Molecular Pathology and Translational Research Unit, Tokyo Medical University; Cancer Genomics Project; Departments of Pathology, and Cardiovascular Medicine; Animal Resources, Graduate School of Medicine, University of Tokyo; Honeybee Science Research Center, Tamagawa University; Department of Gastroenterology, School of Medicine, Keio University, Tokyo; Department of Pharmacology, Graduate School of Medicine, Kyoto University, Kyoto; Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Kobe; Jichi Medical University, Shimotsuke, Tochigi, Japan; and Hubrecht Institute and KNAW, Utrecht, the NetherlandsAuthors' Affiliations: Department of Molecular Pathology and Translational Research Unit, Tokyo Medical University; Cancer Genomics Project; Departments of Pathology, and Cardiovascular Medicine; Animal Resources, Graduate School of Medicine, University of Tokyo; Honeybee Science Research Center, Tamagawa University; Department of Gastroenterology, School of Medicine, Keio University, Tokyo; Department of Pharmacology, Graduate School of Medicine, Kyoto University, Kyoto; Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Kobe; Jichi Medical University, Shimotsuke, Tochigi, Japan; and Hubrecht Institute and KNAW, Utrecht, the Netherlands
| | - Hans Clevers
- Authors' Affiliations: Department of Molecular Pathology and Translational Research Unit, Tokyo Medical University; Cancer Genomics Project; Departments of Pathology, and Cardiovascular Medicine; Animal Resources, Graduate School of Medicine, University of Tokyo; Honeybee Science Research Center, Tamagawa University; Department of Gastroenterology, School of Medicine, Keio University, Tokyo; Department of Pharmacology, Graduate School of Medicine, Kyoto University, Kyoto; Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Kobe; Jichi Medical University, Shimotsuke, Tochigi, Japan; and Hubrecht Institute and KNAW, Utrecht, the Netherlands
| | - Masashi Fukayama
- Authors' Affiliations: Department of Molecular Pathology and Translational Research Unit, Tokyo Medical University; Cancer Genomics Project; Departments of Pathology, and Cardiovascular Medicine; Animal Resources, Graduate School of Medicine, University of Tokyo; Honeybee Science Research Center, Tamagawa University; Department of Gastroenterology, School of Medicine, Keio University, Tokyo; Department of Pharmacology, Graduate School of Medicine, Kyoto University, Kyoto; Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Kobe; Jichi Medical University, Shimotsuke, Tochigi, Japan; and Hubrecht Institute and KNAW, Utrecht, the Netherlands
| | - Masahiko Kuroda
- Authors' Affiliations: Department of Molecular Pathology and Translational Research Unit, Tokyo Medical University; Cancer Genomics Project; Departments of Pathology, and Cardiovascular Medicine; Animal Resources, Graduate School of Medicine, University of Tokyo; Honeybee Science Research Center, Tamagawa University; Department of Gastroenterology, School of Medicine, Keio University, Tokyo; Department of Pharmacology, Graduate School of Medicine, Kyoto University, Kyoto; Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Kobe; Jichi Medical University, Shimotsuke, Tochigi, Japan; and Hubrecht Institute and KNAW, Utrecht, the Netherlands
| | - Ryozo Nagai
- Authors' Affiliations: Department of Molecular Pathology and Translational Research Unit, Tokyo Medical University; Cancer Genomics Project; Departments of Pathology, and Cardiovascular Medicine; Animal Resources, Graduate School of Medicine, University of Tokyo; Honeybee Science Research Center, Tamagawa University; Department of Gastroenterology, School of Medicine, Keio University, Tokyo; Department of Pharmacology, Graduate School of Medicine, Kyoto University, Kyoto; Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Kobe; Jichi Medical University, Shimotsuke, Tochigi, Japan; and Hubrecht Institute and KNAW, Utrecht, the NetherlandsAuthors' Affiliations: Department of Molecular Pathology and Translational Research Unit, Tokyo Medical University; Cancer Genomics Project; Departments of Pathology, and Cardiovascular Medicine; Animal Resources, Graduate School of Medicine, University of Tokyo; Honeybee Science Research Center, Tamagawa University; Department of Gastroenterology, School of Medicine, Keio University, Tokyo; Department of Pharmacology, Graduate School of Medicine, Kyoto University, Kyoto; Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Kobe; Jichi Medical University, Shimotsuke, Tochigi, Japan; and Hubrecht Institute and KNAW, Utrecht, the Netherlands
| |
Collapse
|
11
|
Baljinnyam B, Klauzinska M, Saffo S, Callahan R, Rubin JS. Recombinant R-spondin2 and Wnt3a up- and down-regulate novel target genes in C57MG mouse mammary epithelial cells. PLoS One 2012; 7:e29455. [PMID: 22238613 PMCID: PMC3251591 DOI: 10.1371/journal.pone.0029455] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 11/29/2011] [Indexed: 01/05/2023] Open
Abstract
R-spondins (Rspos) comprise a family of four secreted proteins that have important roles in cell proliferation, cell fate determination and organogenesis. Rspos typically exert their effects by potentiating the Wnt/β-catenin signaling pathway. To systematically investigate the impact of Rspo/Wnt on gene expression, we performed a microarray analysis using C57MG mouse mammary epithelial cells treated with recombinant Rspo2 and/or Wnt3a. We observed the up- and down-regulation of several previously unidentified target genes, including ones that encode proteins involved in immune responses, effectors of other growth factor signaling pathways and transcription factors. Dozens of these changes were validated by quantitative real time RT-PCR. Time course experiments showed that Rspo2 typically had little or no effect on Wnt-dependent gene expression at 3 or 6 h, but enhanced expression at 24 h, consistent with biochemical data indicating that Rspo2 acts primarily to sustain rather than acutely increase Wnt pathway activation. Up-regulation of gene expression was inhibited by pre-treatment with Dickkopf1, a Wnt/β-catenin pathway antagonist, and by siRNA knockdown of β-catenin expression. While Dickkopf1 blocked Rspo2/Wnt3a-dependent down-regulation, a number of down-regulated genes were not affected by β-catenin knockdown, suggesting that in these instances down-regulation was mediated by a β-catenin-independent mechanism.
Collapse
Affiliation(s)
- Bolormaa Baljinnyam
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Malgorzata Klauzinska
- Oncogenetics Section, Mammary Biology and Tumorigenesis Laboratory, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Saad Saffo
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Robert Callahan
- Oncogenetics Section, Mammary Biology and Tumorigenesis Laboratory, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Jeffrey S. Rubin
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
12
|
Yeo HC, Beh TT, Quek JJL, Koh G, Chan KKK, Lee DY. Integrated transcriptome and binding sites analysis implicates E2F in the regulation of self-renewal in human pluripotent stem cells. PLoS One 2011; 6:e27231. [PMID: 22076139 PMCID: PMC3208628 DOI: 10.1371/journal.pone.0027231] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Accepted: 10/12/2011] [Indexed: 11/18/2022] Open
Abstract
Rapid cellular growth and multiplication, limited replicative senescence, calibrated sensitivity to apoptosis, and a capacity to differentiate into almost any cell type are major properties that underline the self-renewal capabilities of human pluripotent stem cells (hPSCs). We developed an integrated bioinformatics pipeline to understand the gene regulation and functions involved in maintaining such self-renewal properties of hPSCs compared to matched fibroblasts. An initial genome-wide screening of transcription factor activity using in silico binding-site and gene expression microarray data newly identified E2F as one of major candidate factors, revealing their significant regulation of the transcriptome. This is underscored by an elevated level of its transcription factor activity and expression in all tested pluripotent stem cell lines. Subsequent analysis of functional gene groups demonstrated the importance of the TFs to self-renewal in the pluripotency-coupled context; E2F directly targets the global signaling (e.g. self-renewal associated WNT and FGF pathways) and metabolic network (e.g. energy generation pathways, molecular transports and fatty acid metabolism) to promote its canonical functions that are driving the self-renewal of hPSCs. In addition, we proposed a core self-renewal module of regulatory interplay between E2F and, WNT and FGF pathways in these cells. Thus, we conclude that E2F plays a significant role in influencing the self-renewal capabilities of hPSCs.
Collapse
Affiliation(s)
- Hock Chuan Yeo
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Thian Thian Beh
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jovina Jia Ling Quek
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore, Singapore
| | - Geoffrey Koh
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Ken Kwok Keung Chan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- * E-mail: (KKKC); (DYL)
| | - Dong-Yup Lee
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore, Singapore
- * E-mail: (KKKC); (DYL)
| |
Collapse
|
13
|
Walker F, Zhang HH, Odorizzi A, Burgess AW. LGR5 is a negative regulator of tumourigenicity, antagonizes Wnt signalling and regulates cell adhesion in colorectal cancer cell lines. PLoS One 2011; 6:e22733. [PMID: 21829496 PMCID: PMC3145754 DOI: 10.1371/journal.pone.0022733] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Accepted: 07/04/2011] [Indexed: 02/07/2023] Open
Abstract
Background LGR5 (Leucine-rich repeat-containing G-protein coupled receptor 5) is the most established marker for intestinal stem cells. Mouse models show that LGR5+ cells are the cells of origin of intestinal cancer, and LGR5 expression is elevated in human colorectal cancers, however very little is known about LGR5 function or its contribution to the stem cell phenotype and to colorectal cancer. Principal Findings We have modulated the expression of LGR5 by RNAi (inhibitory RNAs) or overexpression in colorectal cancer cell lines. Paradoxically, ablation of LGR5 induces increased invasion and anchorage-independent growth, and enhances tumourigenicity in xenografts experiments. Conversely, overexpression of LGR5 augments cell adhesion, reduces clonogenicity and attenuates tumourigenicity. Expression profiling revealed enhanced wnt signalling and upregulation of EMT genes upon knockdown of LGR5, with opposite changes in LGR5 overexpressing cells. These findings suggest that LGR5 is important in restricting stem cells to their niche, and that loss of LGR5 concomitant with activated wnt signalling may contribute to the invasive phenotype of colorectal carcinomas.
Collapse
MESH Headings
- Animals
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Blotting, Western
- Cell Adhesion/physiology
- Cell Movement
- Cell Proliferation
- Colony-Forming Units Assay
- Colorectal Neoplasms/genetics
- Colorectal Neoplasms/metabolism
- Colorectal Neoplasms/pathology
- Fluorescent Antibody Technique
- Gene Expression Profiling
- Humans
- Mice
- Mice, Nude
- Oligonucleotide Array Sequence Analysis
- RNA, Messenger/genetics
- RNA, Small Interfering/genetics
- Receptors, G-Protein-Coupled/antagonists & inhibitors
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Tumor Cells, Cultured
- Wnt Proteins/genetics
- Wnt Proteins/metabolism
- Wound Healing
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Francesca Walker
- Epithelial Biochemistry Laboratory, Ludwig Institute for Cancer Research, Parkville, Victoria, Australia.
| | | | | | | |
Collapse
|
14
|
Meyer SE, Hasenstein JR, Baktula A, Velu CS, Xu Y, Wan H, Whitsett JA, Gilks CB, Grimes HL. Kruppel-like factor 5 is not required for K-RasG12D lung tumorigenesis, but represses ABCG2 expression and is associated with better disease-specific survival. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:1503-13. [PMID: 20639455 DOI: 10.2353/ajpath.2010.090651] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
K-RAS mutations are found in approximately 30% of lung cancers. The transcription factor Krüppel-like Factor 5 (KLF5) has been shown to mediate cellular transformation signaling events downstream of oncogenic RAS in other cancers, but a role for KLF5 in lung tumorigenesis has not been defined. We show here that knockdown of KLF5 expression significantly decreased anchorage-independent growth, but did not affect proliferation of human lung adenocarcinoma cells. Moreover, Klf5 is not required for lung tumor formation in an inducible oncogenic K-Ras(G12D) mouse model of lung tumorigenesis, and non-small cell lung cancer patients expressing high levels of KLF5 (21/258) have a significantly better disease-specific survival than those with intermediate to no KLF5 expression. Further, KLF5 knockdown in K-RAS-mutant human lung cancer cells resulted in a fivefold increase in ATP-binding cassette, subfamily G (WHITE), member 2 (ABCG2), an anthracycline drug transporter, which lead to significantly increased resistance to doxorubicin treatment, a chemotherapeutic agent clinically used to treat lung cancer. In summary, while KLF5 is not required for oncogenic mutant K-Ras-induced lung tumorigenesis, KLF5 regulation of ABCG2 expression may be important for chemotherapeutic resistance and patient survival.
Collapse
Affiliation(s)
- Sara E Meyer
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Regulation of the Rho family small GTPase Wrch-1/RhoU by C-terminal tyrosine phosphorylation requires Src. Mol Cell Biol 2010; 30:4324-38. [PMID: 20547754 DOI: 10.1128/mcb.01646-09] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Wrch-1 is an atypical Rho family small GTPase with roles in migration, epithelial cell morphogenesis, osteoclastogenesis, and oncogenic transformation. Here, we observed rapid relocalization of Wrch-1 from the plasma membrane upon serum stimulation. Studies revealed a requirement for serum-stimulated tyrosine phosphorylation of Wrch-1 at residue Y254 within its C-terminal membrane targeting domain, mediated by the nonreceptor tyrosine kinase Src. Genetic or pharmacological loss of Src kinase activity blocked both phosphorylation and relocalization of Wrch-1. Functionally, Y254 was required for proper Wrch-1 modulation of cystogenesis in three-dimensional culture, and the phospho-deficient mutant, Y254F, was enhanced in Wrch-1-mediated anchorage-independent growth. Mechanistically, C-terminal tyrosine phosphorylation and subsequent relocalization of Wrch-1 downregulated its ability to interact with and activate its effectors by decreasing active Wrch-1-GTP, perhaps by altering proximity to a GEF or GAP. Phospho-deficient Wrch-1(Y254F) remained at the plasma membrane and GTP bound and continued to recruit and activate its effector PAK, even upon serum stimulation. In contrast, a phospho-mimetic mutant, Y254E, was constitutively endosomally localized and GDP bound and failed to recruit PAK unless mutated to be constitutively active/GAP insensitive. C-terminal tyrosine phosphorylation thus represents a new paradigm in posttranslational control of small GTPase localization, activation, and biological function.
Collapse
|
16
|
Foss B, Tronstad KJ, Bruserud Ø. Connexin-based signaling in acute myelogenous leukemia (AML). BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2009; 1798:1-8. [PMID: 19883623 DOI: 10.1016/j.bbamem.2009.10.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2009] [Revised: 09/15/2009] [Accepted: 10/26/2009] [Indexed: 10/20/2022]
Abstract
Normal and malignant hematopoiesis are regulated by intercellular communication in the hematopoietic microenvironments, and both soluble mediators as well as direct cell-cell contact play important functional roles. Gap junctions are complex membrane structures that transfer molecules between neighboring cells and thereby alter intracellular signaling and metabolism. The gap junction building blocks, the connexins, are also involved in gap junction-independent intercellular communication by forming hemichannels that transfer substances between the intra- and extracellular spaces. Connexins are furthermore involved in cell regulation as single molecules by modulating intracellular pathways and possibly gene transcription. The role of connexins in leukemogenesis and leukemic cell functions are not well characterized. In this review, we describe the known effects of gap junctions and connexins in acute myelogenous leukemia and the diverse potential of connexins in acute myelogenous leukemia chemosensitivity, intracellular signaling and cell death regulation.
Collapse
Affiliation(s)
- Brynjar Foss
- Department of Health Studies, University of Stavanger, Stavanger, Norway.
| | | | | |
Collapse
|
17
|
Wang Y, Hanifi-Moghaddam P, Hanekamp EE, Kloosterboer HJ, Franken P, Veldscholte J, van Doorn HC, Ewing PC, Kim JJ, Grootegoed JA, Burger CW, Fodde R, Blok LJ. Progesterone inhibition of Wnt/beta-catenin signaling in normal endometrium and endometrial cancer. Clin Cancer Res 2009; 15:5784-93. [PMID: 19737954 DOI: 10.1158/1078-0432.ccr-09-0814] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Wnt signaling regulates the fine balance between stemness and differentiation. Here, the role of Wnt signaling to maintain the balance between estrogen-induced proliferation and progesterone-induced differentiation during the menstrual cycle, as well as during the induction of hyperplasia and carcinogenesis of the endometrium, was investigated. EXPERIMENTAL DESIGN Endometrial gene expression profiles from estradiol (E(2)) and E(2) + medroxyprogesterone acetate-treated postmenopausal patients were combined with profiles obtained during the menstrual cycle (PubMed; GEO DataSets). Ishikawa cells were transfected with progesterone receptors and Wnt inhibitors dickkopf homologue 1 (DKK1) and forkhead box O1 (FOXO1), measuring Wnt activation. Expression of DKK1 and FOXO1 was inhibited by use of sequence-specific short hairpins. Furthermore, patient samples (hormone-treated endometria, hyperplasia, and endometrial cancer) were stained for Wnt activation using nuclear beta-catenin and CD44. RESULTS In vivo, targets and components of the Wnt signaling pathway (among them DKK1 and FOXO1) are regulated by E(2) and progesterone. In Wnt-activated Ishikawa cells, progesterone inhibits Wnt signaling by induction of DKK1 and FOXO1. Furthermore, using siRNA-mediated knockdown of both DKK1 and FOXO1, progesterone inhibition of Wnt signaling was partly circumvented. Subsequently, immunohistochemical analysis of the Wnt target gene CD44 showed that progesterone acted as an inhibitor of Wnt signaling in hyperplasia and in well-differentiated endometrial cancer. CONCLUSION Progesterone induction of DKK1 and FOXO1 results in inhibition of Wnt signaling in the human endometrium. This Wnt inhibitory effect of progesterone is likely to play a rate-limiting role in the maintenance of endometrial homeostasis and, on its loss, in tumor onset and progression toward malignancy.
Collapse
Affiliation(s)
- Yongyi Wang
- Department of Obstetrics and Gynecology, Josephine Nefkens Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Dong JT, Chen C. Essential role of KLF5 transcription factor in cell proliferation and differentiation and its implications for human diseases. Cell Mol Life Sci 2009; 66:2691-706. [PMID: 19448973 PMCID: PMC11115749 DOI: 10.1007/s00018-009-0045-z] [Citation(s) in RCA: 211] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2009] [Revised: 04/22/2009] [Accepted: 04/24/2009] [Indexed: 02/08/2023]
Abstract
KLF5 (Kruppel-like factor 5) is a basic transcription factor binding to GC boxes at a number of gene promoters and regulating their transcription. KLF5 is expressed during development and, in adults, with higher levels in proliferating epithelial cells. The expression and activity of KLF5 are regulated by multiple signaling pathways, including Ras/MAPK, PKC, and TGFbeta, and various posttranslational modifications, including phosphorylation, acetylation, ubiquitination, and sumoylation. Consistently, KLF5 mediates the signaling functions in cell proliferation, cell cycle, apoptosis, migration, differentiation, and stemness by regulating gene expression in response to environment stimuli. The expression of KLF5 is frequently abnormal in human cancers and in cardiovascular disease-associated vascular smooth muscle cells (VSMCs). Due to its significant functions in cell proliferation, survival, and differentiation, KLF5 could be a potential diagnostic biomarker and therapeutic target for cancer and cardiovascular diseases.
Collapse
Affiliation(s)
- Jin-Tang Dong
- Department of Hematology and Medical Oncology, Department of Urology and Winship Cancer Institute, Emory University School of Medicine, 1365-C Clifton Road, Atlanta, GA 30322, USA.
| | | |
Collapse
|
19
|
Railo A, Pajunen A, Itäranta P, Naillat F, Vuoristo J, Kilpeläinen P, Vainio S. Genomic response to Wnt signalling is highly context-dependent--evidence from DNA microarray and chromatin immunoprecipitation screens of Wnt/TCF targets. Exp Cell Res 2009; 315:2690-704. [PMID: 19563800 DOI: 10.1016/j.yexcr.2009.06.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2007] [Revised: 05/15/2009] [Accepted: 06/22/2009] [Indexed: 11/27/2022]
Abstract
Wnt proteins are important regulators of embryonic development, and dysregulated Wnt signalling is involved in the oncogenesis of several human cancers. Our knowledge of the downstream target genes is limited, however. We used a chromatin immunoprecipitation-based assay to isolate and characterize the actual gene segments through which Wnt-activatable transcription factors, TCFs, regulate transcription and an Affymetrix microarray analysis to study the global transcriptional response to the Wnt3a ligand. The anti-beta-catenin immunoprecipitation of DNA-protein complexes from mouse NIH3T3 fibroblasts expressing a fusion protein of beta-catenin and TCF7 resulted in the identification of 92 genes as putative TCF targets. GeneChip assays of gene expression performed on NIH3T3 cells and the rat pheochromocytoma cell line PC12 revealed 355 genes in NIH3T3 and 129 genes in the PC12 cells with marked changes in expression after Wnt3a stimulus. Only 2 Wnt-regulated genes were shared by both cell lines. Surprisingly, Disabled-2 was the only gene identified by the chromatin immunoprecipitation approach that displayed a marked change in expression in the GeneChip assay. Taken together, our approaches give an insight into the complex context-dependent nature of Wnt pathway transcriptional responses and identify Disabled-2 as a potential new direct target for Wnt signalling.
Collapse
Affiliation(s)
- Antti Railo
- Oulu Centre for Cell Matrix Research, Biocenter Oulu, Laboratory of Developmental Biology and Department of Medical Biochemistry and Molecular Biology, FIN-90014, University of Oulu, P. O. Box 5000, Finland
| | | | | | | | | | | | | |
Collapse
|
20
|
Liu R, Zheng HQ, Zhou Z, Dong JT, Chen C. KLF5 promotes breast cell survival partially through fibroblast growth factor-binding protein 1-pERK-mediated dual specificity MKP-1 protein phosphorylation and stabilization. J Biol Chem 2009; 284:16791-16798. [PMID: 19411256 DOI: 10.1074/jbc.m808919200] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Krüpple-like transcription factor 5 (KLF5) is a zinc-finger transcription factor promoting cell survival and tumorigenesis in multiple cancers. A high expression level of KLF5 has been shown to be associated with shorter breast cancer patient survival. However, the role of KLF5 and mechanism of KLF5 actions in breast cancer remain unclear. In this study, we found that KLF5 knockdown by small interfering RNA in two breast cell lines, MCF10A and BT20, induces apoptosis. Interestingly, a pro-survival phosphatase, dual specificity mitogen-activated protein kinase phosphatase 1 (MKP-1), is down-regulated by KLF5 ablation. Consistently, KLF5 overexpression increases the MKP-1 protein expression in Hs578T and MCF7. We further found that MKP-1 is essential and sufficient for KLF5 to promote breast cell survival. However, MKP-1 is not a KLF5 direct transcription target because the MKP-1 mRNA level is not regulated by KLF5. By cycloheximide chase assays, we found that KLF5 decreases MKP-1 protein degradation via activating the ERK signaling. Inhibition of pERK by the pharmacological inhibitor U0126 specifically blocks KLF5-induced MKP-1 phosphorylation and stabilization. Additionally, constitutive activation of ERK by constitutively activated MEK1 rescues the KLF5 depletion-induced MKP-1 down-regulation. Consistently, the phosphorylation-deficient MKP-1 mutant cannot be stabilized by KLF5. Finally, the activation of ERK by KLF5 is very likely through the KLF5 direct target gene FGF-BP in breast cells. These findings suggest that KLF5 is a pro-survival factor that promotes breast cell survival partially through pERK-mediated MKP-1 phosphorylation and stabilization. The KLF5-FGF-BP-pERK-MKP-1 signaling axis may provide new therapeutic targets for invasive breast cancer.
Collapse
Affiliation(s)
- Rong Liu
- From the Center for Cell Biology and Cancer Research, Albany Medical College, Albany, New York 12208
| | - Han-Qiu Zheng
- From the Center for Cell Biology and Cancer Research, Albany Medical College, Albany, New York 12208
| | - Zhongmei Zhou
- From the Center for Cell Biology and Cancer Research, Albany Medical College, Albany, New York 12208
| | - Jin-Tang Dong
- Winship Cancer Institute and Department of Hematology and Oncology, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Ceshi Chen
- From the Center for Cell Biology and Cancer Research, Albany Medical College, Albany, New York 12208.
| |
Collapse
|
21
|
Koçer SS, Djurić PM, Bugallo MF, Simon SR, Matic M. Transcriptional profiling of putative human epithelial stem cells. BMC Genomics 2008; 9:359. [PMID: 18667080 PMCID: PMC2536675 DOI: 10.1186/1471-2164-9-359] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2007] [Accepted: 07/30/2008] [Indexed: 12/30/2022] Open
Abstract
Background Human interfollicular epidermis is sustained by the proliferation of stem cells and their progeny, transient amplifying cells. Molecular characterization of these two cell populations is essential for better understanding of self renewal, differentiation and mechanisms of skin pathogenesis. The purpose of this study was to obtain gene expression profiles of alpha 6+/MHCI+, transient amplifying cells and alpha 6+/MHCI-, putative stem cells, and to compare them with existing data bases of gene expression profiles of hair follicle stem cells. The expression of Major Histocompatibility Complex (MHC) class I, previously shown to be absent in stem cells in several tissues, and alpha 6 integrin were used to isolate MHCI positive basal cells, and MHCI low/negative basal cells. Results Transcriptional profiles of the two cell populations were determined and comparisons made with published data for hair follicle stem cell gene expression profiles. We demonstrate that presumptive interfollicular stem cells, alpha 6+/MHCI- cells, are enriched in messenger RNAs encoding surface receptors, cell adhesion molecules, extracellular matrix proteins, transcripts encoding members of IFN-alpha family proteins and components of IFN signaling, but contain lower levels of transcripts encoding proteins which take part in energy metabolism, cell cycle, ribosome biosynthesis, splicing, protein translation, degradation, DNA replication, repair, and chromosome remodeling. Furthermore, our data indicate that the cell signaling pathways Notch1 and NF-κB are downregulated/inhibited in MHC negative basal cells. Conclusion This study demonstrates that alpha 6+/MHCI- cells have additional characteristics attributed to stem cells. Moreover, the transcription profile of alpha 6+/MHCI- cells shows similarities to transcription profiles of mouse hair follicle bulge cells known to be enriched for stem cells. Collectively, our data suggests that alpha 6+/MHCI- cells may be enriched for stem cells. This study is the first comprehensive gene expression profile of putative human epithelial stem cells and their progeny that were isolated directly from neonatal foreskin tissue. Our study is important for understanding self renewal and differentiation of epidermal stem cells, and for elucidating signaling pathways involved in those processes. The generated data base may serve those working with other human epithelial tissue progenitors.
Collapse
Affiliation(s)
- Salih S Koçer
- Department of Biochemistry and Cell Biology, State University of New York at Stony Brook, Stony Brook, NY, USA.
| | | | | | | | | |
Collapse
|
22
|
Königshoff M, Balsara N, Pfaff EM, Kramer M, Chrobak I, Seeger W, Eickelberg O. Functional Wnt signaling is increased in idiopathic pulmonary fibrosis. PLoS One 2008; 3:e2142. [PMID: 18478089 PMCID: PMC2374879 DOI: 10.1371/journal.pone.0002142] [Citation(s) in RCA: 382] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2008] [Accepted: 04/02/2008] [Indexed: 12/17/2022] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a fatal lung disease, characterized by distorted lung architecture and loss of respiratory function. Alveolar epithelial cell injury and hyperplasia, enhanced extracellular matrix deposition, and (myo)fibroblast activation are features of IPF. Wnt/β-catenin signaling has been shown to determine epithelial cell fate during development. As aberrant reactivation of developmental signaling pathways has been suggested to contribute to IPF pathogenesis, we hypothesized that Wnt/β-catenin signaling is activated in epithelial cells in IPF. Thus, we quantified and localized the expression and activity of the Wnt/β-catenin pathway in IPF. Methodology/Principal Findings The expression of Wnt1, 3a, 7b, and 10b, the Wnt receptors Fzd1-4, Lrp5-6, as well as the intracellular signal transducers Gsk-3β, β-catenin, Tcf1, 3, 4, and Lef1 was analyzed in IPF and transplant donor lungs by quantitative real-time (q)RT-PCR. Wnt1, 7b and 10b, Fzd2 and 3, β-catenin, and Lef1 expression was significantly increased in IPF. Immunohistochemical analysis localized Wnt1, Wnt3a, β-catenin, and Gsk-3β expression largely to alveolar and bronchial epithelium. This was confirmed by qRT-PCR of primary alveolar epithelial type II (ATII) cells, demonstrating a significant increase of Wnt signaling in ATII cells derived from IPF patients. In addition, Western blot analysis of phospho-Gsk-3β, phospho-Lrp6, and β-catenin, and qRT-PCR of the Wnt target genes cyclin D1, Mmp 7, or Fibronectin 1 demonstrated increased functional Wnt/β-catenin signaling in IPF compared with controls. Functional in vitro studies further revealed that Wnt ligands induced lung epithelial cell proliferation and (myo)fibroblast activation and collagen synthesis. Conclusions/Significance Our study demonstrates that the Wnt/β-catenin pathway is expressed and operative in adult lung epithelium. Increased Wnt/β-catenin signaling may be involved in epithelial cell injury and hyperplasia, as well as impaired epithelial-mesenchymal cross-talk in IPF. Thus, modification of Wnt signaling may represent a therapeutic option in IPF.
Collapse
Affiliation(s)
- Melanie Königshoff
- Department of Medicine, University of Giessen Lung Center, University of Giessen, Giessen, Germany
| | - Nisha Balsara
- Department of Medicine, University of Giessen Lung Center, University of Giessen, Giessen, Germany
| | - Eva-Maria Pfaff
- Department of Medicine, University of Giessen Lung Center, University of Giessen, Giessen, Germany
| | - Monika Kramer
- Department of Medicine, University of Giessen Lung Center, University of Giessen, Giessen, Germany
| | - Izabella Chrobak
- Department of Medicine, University of Giessen Lung Center, University of Giessen, Giessen, Germany
| | - Werner Seeger
- Department of Medicine, University of Giessen Lung Center, University of Giessen, Giessen, Germany
| | - Oliver Eickelberg
- Department of Medicine, University of Giessen Lung Center, University of Giessen, Giessen, Germany
- * E-mail:
| |
Collapse
|
23
|
Vlad A, Röhrs S, Klein-Hitpass L, Müller O. The first five years of the Wnt targetome. Cell Signal 2008; 20:795-802. [DOI: 10.1016/j.cellsig.2007.10.031] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2007] [Accepted: 10/30/2007] [Indexed: 02/04/2023]
|
24
|
Kumaran R, Kingsbury A, Coulter I, Lashley T, Williams D, de Silva R, Mann D, Revesz T, Lees A, Bandopadhyay R. DJ-1 (PARK7) is associated with 3R and 4R tau neuronal and glial inclusions in neurodegenerative disorders. Neurobiol Dis 2007; 28:122-32. [PMID: 17719794 DOI: 10.1016/j.nbd.2007.07.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2007] [Revised: 06/25/2007] [Accepted: 07/01/2007] [Indexed: 12/13/2022] Open
Abstract
Mutations in the DJ-1 gene are associated with autosomal recessive Parkinson's disease (PD), but its role in disease pathogenesis is unknown. This study examines DJ-1 immunoreactivity (DJ-1 IR) in a variety of neurodegenerative disorders, Alzheimer's disease (AD), frontotemporal lobar degeneration (FTLD) with Pick bodies, FTLD with MAPT mutations, progressive supranuclear palsy (PSP) and corticobasal degeneration (CBD), in which hyperphosphorylated tau inclusions are the major pathological signature. DJ-1 IR was seen in a subset of neurofibrillary tangles (NFTs), neuropil threads (NTs), and neurites in extracellular plaques in AD; tau inclusions in AD contained both 3R and 4R tau. A subset of Pick bodies in FTLD showed DJ-1 IR. In PSP, DJ-1 IR was present in a few NFTs, NTs and glial cell inclusions. In CBD, DJ-1 IR was seen only in astrocytic plaques. In cases of FTLD with MAPT mutations that were 4R tau positive (i.e. N279K and exon 10+16 mutations), DJ-1 IR was present mostly in oligodendroglial coiled bodies. However, in MAPT R406W mutation cases, DJ-1 IR was associated mainly with NFTs and NTs and these were both 3R and 4R tau positive. No DJ-1 IR was present in FTLD with ubiquitin inclusions (FTLD-U). In AD and FTLD with Pick bodies, DJ-1 protein was enriched in the sarkosyl-insoluble fractions of frozen brain tissue containing insoluble hyperphosphorylated tau, thus strengthening the association of DJ-1 with tau pathology. Additionally using two-dimensional gel electrophoresis, we demonstrated accumulation of acidic pI isoforms of DJ-1 in AD brain, which may compromise its normal function. Our observations confirm previous findings that DJ-1 is present in a subpopulation of glial and neuronal tau inclusions in tau diseases and associated with both 3R and 4R tau isoforms.
Collapse
Affiliation(s)
- Ravindran Kumaran
- Reta Lila Weston Institute of Neurological Studies, Institute of Neurology, 1, Wakefield Street, WC1N 1PJ, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Chen C, Benjamin MS, Sun X, Otto KB, Guo P, Dong XY, Bao Y, Zhou Z, Cheng X, Simons JW, Dong JT. KLF5 promotes cell proliferation and tumorigenesis through gene regulation and the TSU-Pr1 human bladder cancer cell line. Int J Cancer 2007; 118:1346-55. [PMID: 16184550 DOI: 10.1002/ijc.21533] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
KLF5 is a transcription factor that plays important roles in multiple physical and pathological processes, including cell growth, cell cycle regulation, and angiogenesis. To better characterize KLF5 function in bladder carcinogenesis, we established stable TSU-Pr1 cell clones expressing different levels of KLF5. These clones were then characterized for cell growth, cell cycle progression, tumorigenesis, and alteration in gene expression. Overexpression of KLF5 promoted tumorigenesis of the TSU-Pr1 cancer cells in mice. Consistently, KLF5 increased G1 to S phase transition, which was accompanied by the upregulation of cyclin D1, phosphorylation of MAPK and Akt, and reduced protein levels for CDK inhibitors p27 and p15. Microarray analysis combined with expression verification in different cell systems identified a number of additional genes that are potentially regulated by KLF5, including HBP17, ITGA6, and RAIG1. These findings suggest that the KLF5 transcription factor plays an oncogenic role in the TSU-Pr1 bladder cancer cell line through the regulation of a subset of genes.
Collapse
MESH Headings
- Animals
- Blotting, Northern
- Blotting, Western
- Cell Line, Tumor
- Cell Proliferation
- Clone Cells
- Cyclin D1/metabolism
- Gene Expression Regulation, Neoplastic
- Humans
- Kruppel-Like Transcription Factors/genetics
- Kruppel-Like Transcription Factors/metabolism
- Kruppel-Like Transcription Factors/physiology
- Male
- Mice
- Mice, SCID
- Mitogen-Activated Protein Kinases/metabolism
- Neoplasm Transplantation
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/pathology
- Oligonucleotide Array Sequence Analysis
- Proto-Oncogene Proteins c-akt/metabolism
- RNA, Small Interfering/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- S Phase/genetics
- S Phase/physiology
- Transplantation, Heterologous
- Tumor Burden
- Urinary Bladder Neoplasms/genetics
- Urinary Bladder Neoplasms/metabolism
- Urinary Bladder Neoplasms/pathology
Collapse
Affiliation(s)
- Ceshi Chen
- Winship Cancer Institute and Department of Oncology and Hematology, Emory University School of Medicine, Atlanta, GA, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Labbé E, Lock L, Letamendia A, Gorska AE, Gryfe R, Gallinger S, Moses HL, Attisano L. Transcriptional Cooperation between the Transforming Growth Factor-β and Wnt Pathways in Mammary and Intestinal Tumorigenesis. Cancer Res 2007; 67:75-84. [PMID: 17210685 DOI: 10.1158/0008-5472.can-06-2559] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Transforming growth factor-beta (TGF-beta) and Wnt ligands function in numerous developmental processes, and alterations of both signaling pathways are associated with common pathologic conditions, including cancer. To obtain insight into the extent of interdependence of the two signaling cascades in regulating biological responses, we used an oligonucleotide microarray approach to identify Wnt and TGF-beta target genes using normal murine mammary gland epithelial cells as a model. Combination treatment of TGF-beta and Wnt revealed a novel transcriptional program that could not have been predicted from single ligand treatments and included a cohort of genes that were cooperatively induced by both pathways. These included both novel and known components or modulators of TGF-beta and Wnt pathways, suggesting that mutual feedback is a feature of the coordinated activities of the ligands. The majority of the cooperative targets display increased expression in tumors derived from either Min (many intestinal neoplasia) or mouse mammary tumor virus (MMTV)-Wnt1 mice, two models of Wnt-induced tumors, with nine of these genes (Ankrd1, Ccnd1, Ctgf, Gpc1, Hs6st2, IL11, Inhba, Mmp14, and Robo1) showing increases in both. Reduction of TGF-beta signaling by expression of a dominant-negative TGF-beta type II receptor in bigenic MMTV-Wnt1/DNIIR mice increased mammary tumor latency and was correlated with a decrease in expression of Gpc1, Inhba, and Robo1, three of the TGF-beta/Wnt cooperative targets. Our results indicate that the TGF-beta and Wnt/beta-catenin pathways are firmly intertwined and generate a unique gene expression pattern that can contribute to tumor progression.
Collapse
MESH Headings
- Adenoma/genetics
- Adenoma/metabolism
- Adenoma/pathology
- Animals
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Intestinal Neoplasms/genetics
- Intestinal Neoplasms/metabolism
- Intestinal Neoplasms/pathology
- L Cells
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Signal Transduction
- Transcription, Genetic
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/metabolism
- Wnt Proteins/genetics
- Wnt Proteins/metabolism
- Wnt3 Protein
Collapse
Affiliation(s)
- Etienne Labbé
- Departments of Medical Biophysics and Biochemistry, University of Toronto, 160 College Street, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Uddin RK, Singh SM. cis-Regulatory sequences of the genes involved in apoptosis, cell growth, and proliferation may provide a target for some of the effects of acute ethanol exposure. Brain Res 2006; 1088:31-44. [PMID: 16631145 DOI: 10.1016/j.brainres.2006.02.125] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2005] [Revised: 01/31/2006] [Accepted: 02/26/2006] [Indexed: 01/22/2023]
Abstract
The physiological effects of alcohol are known to include drunkenness, toxicity, and addiction leading to alcohol-related health and societal problems. Some of these effects are mediated by regulation of expression of many genes involved in alcohol response pathways. Analysis of the regulatory elements and biological interaction of the genes that show coexpression in response to alcohol may give an insight into how they are regulated. Fifty-two ethanol-responsive (ER) genes displaying differential expression in mouse brain in response to acute ethanol exposure were subjected to bioinformatics analysis to identify known or putative transcription factor binding sites and cis-regulatory modules in the promoter regions that may be involved in their responsiveness to alcohol. Functional interactions of these genes were also examined to assess their cumulative contribution to metabolomic pathways. Clustering and promoter sequence analysis of the ER genes revealed the DNA binding site for nuclear transcription factor Y (NFY) as the most significant. NFY also take part in the proposed biological association network of a number of ER genes, where these genes interact with themselves and other cellular components, and may generate a major cumulative effect on apoptosis, cell survival, and proliferation in response to alcohol. NFY has the potential to play a critical role in mediating the expression of a set of ER genes whose interactions contribute to apoptosis, cell survival, and proliferation, which in turn may affect alcohol-related behaviors.
Collapse
Affiliation(s)
- Raihan K Uddin
- Department of Biology and Division of Medical Genetics, The University of Western Ontario, London, Ontario, Canada N6A 5B7.
| | | |
Collapse
|
28
|
Chen C, Sun X, Guo P, Dong XY, Sethi P, Cheng X, Zhou J, Ling J, Simons JW, Lingrel JB, Dong JT. Human Kruppel-like factor 5 is a target of the E3 ubiquitin ligase WWP1 for proteolysis in epithelial cells. J Biol Chem 2005; 280:41553-61. [PMID: 16223724 DOI: 10.1074/jbc.m506183200] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The transcription factor KLF5 plays an important role in human carcinogenesis. In epithelial cells, the KLF5 protein is tightly regulated by the ubiquitin-proteasome pathway. To better understand the mechanisms for the regulation of KLF5 protein, we identified and characterized an E3 ubiquitin ligase for KLF5, i.e. WWP1. We found that WWP1 formed a protein complex with KLF5 in vivo and in vitro. Furthermore, WWP1 mediated the ubiquitination and degradation of KLF5, and the catalytic cysteine residue of WWP1 is essential for its function. A PY motif in a transactivation domain of KLF5 is necessary for its interaction with WWP1. Finally, WWP1 was amplified and overexpressed in some cancer cell lines from the prostate and breast, which negatively regulated the function of KLF5 in gene regulation. These findings not only established WWP1 as an E3 ubiquitin ligase for KLF5, they also further implicated the KLF5 pathway in human carcinogenesis.
Collapse
Affiliation(s)
- Ceshi Chen
- Winship Cancer Institute and Department of Hematology and Oncology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Ghiselli G, Liu CG. Global gene expression profiling of cells overexpressing SMC3. Mol Cancer 2005; 4:34. [PMID: 16156898 PMCID: PMC1242249 DOI: 10.1186/1476-4598-4-34] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2004] [Accepted: 09/12/2005] [Indexed: 12/24/2022] Open
Abstract
Background The Structural Maintenance of Chromosome 3 protein (SMC3) plays an essential role during the sister chromatid separation, is involved in DNA repair and recombination and participates in microtubule-mediated intracellular transport. SMC3 is frequently elevated in human colon carcinoma and overexpression of the protein transforms murine NIH3T3 fibroblasts. In order to gain insight into the mechanism of SMC3-mediated tumorigenesis a gene expression profiling was performed on human 293 cells line stably overexpressing SMC3. Results Biotinylated complementary RNA (cRNA) was used for hybridization of a cDNAmicroarray chip harboring 18,861 65-mer oligos derived from the published dEST sequences. After filtering, the hybridization data were normalized and statistically analyzed. Sixty-five genes for which a putative function could be assigned displayed at least two-fold change in their expression level. Eighteen of the affected genes is either a transcriptional factor or is involved in DNA and chromatin related mechanisms whereas most of those involved in signal transduction are members or modulators of the ras-rho/GTPase and cAMP signaling pathways. In particular the expression of RhoB and CRE-BPa, two mediators of cellular transformation, was significantly enhanced. This association was confirmed by analyzing the RhoB and CRE-BPa transcript levels in cells transiently transfected with an SMC3 expression vector. Consistent with the idea that the activation of ras-rho/GTPase and cAMP pathways is relevant in the context of the cellular changes following SMC3 overexpression, gene transactivation through the related serum (SRE) and cAMP (CRE) cis-acting response elements was significantly increased. Conclusion We have documented a selective effect of the ectopic expression of SMC3 on a set of genes and transcriptional signaling pathways that are relevant for tumorigenesis. The results lead to postulate that RhoB and CRE-BPa two known oncogenic mediators whose expression is significantly increased following SMC3 overexpression play a significant role in mediating SMC3 tumorigenesis.
Collapse
Affiliation(s)
- Giancarlo Ghiselli
- Department of Pathology and Cell Biology, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA 19107, USA
- Kimmel Cancer Center, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA 19107, USA
| | - Chang-Gong Liu
- Kimmel Cancer Center, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA 19107, USA
- Department of Microbiology and Immunology, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA 19107, USA
| |
Collapse
|
30
|
Taneyhill LA, Bronner-Fraser M. Dynamic alterations in gene expression after Wnt-mediated induction of avian neural crest. Mol Biol Cell 2005; 16:5283-93. [PMID: 16135532 PMCID: PMC1266426 DOI: 10.1091/mbc.e05-03-0210] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The Wnt signaling pathway is important in the formation of neural crest cells in many vertebrates, but the downstream targets of neural crest induction by Wnt are largely unknown. Here, we examined quantitative changes in gene expression regulated by Wnt-mediated neural crest induction using quantitative PCR (QPCR). Induction was recapitulated in vitro by adding soluble Wnt to intermediate neural plate tissue cultured in collagen, and induced versus control tissue were assayed using gene-specific primers at times corresponding to premigratory (18 and 24 h) or early (36 h) stages of crest migration. The results show that Wnt signaling up-regulates in a distinct temporal pattern the expression of several genes normally expressed in the dorsal neural tube (slug, Pax3, Msx1, FoxD3, cadherin 6B) at "premigratory" stages. While slug is maintained in early migrating crest cells, Pax3, FoxD3, Msx1 and cadherin 6B all are down-regulated by the start of migration. These results differ from the temporal profile of these genes in response to the addition of recombinant BMP4, where gene expression seems to be maintained. Interestingly, expression of rhoB is unchanged or even decreased in response to Wnt-mediated induction at all times examined, though it is up-regulated by BMP signals. The temporal QPCR profiles in our culture paradigm approximate in vivo expression patterns of these genes before neural crest migration, and are consistent with Wnt being an initial neural crest inducer with additional signals like BMP and other factors maintaining expression of these genes in vivo. Our results are the first to quantitatively describe changes in gene expression in response to a Wnt or BMP signal during transformation of a neural tube cell into a migratory neural crest cell.
Collapse
Affiliation(s)
- Lisa A Taneyhill
- Division of Biology, California Institute of Technology, Pasadena, CA 91125, USA
| | | |
Collapse
|
31
|
Chen C, Sun X, Ran Q, Wilkinson KD, Murphy TJ, Simons JW, Dong JT. Ubiquitin-proteasome degradation of KLF5 transcription factor in cancer and untransformed epithelial cells. Oncogene 2005; 24:3319-27. [PMID: 15735697 DOI: 10.1038/sj.onc.1208497] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Ubiquitin-mediated proteolysis plays a central role in controlling intracellular levels of essential regulatory molecules such as p53, cyclins, myc, BRCA1, HIF-1alpha, etc. The Kruppel-like factor 5 (KLF5) transcription factor regulates biological processes involved in carcinogenesis, angiogenesis, and smooth muscle cell differentiation. In carcinogenesis, KLF5's role has been indicated by frequent genetic deletion as well as functional studies. Here we show that KLF5 is an unstable protein with a short half-life. Destruction of KLF5 was prevented by each of the proteasome-specific inhibitors tested but not by an inhibitor for trypsin-like proteases and cysteine proteases or by a lysosome inhibitor in epithelial cells. Furthermore, KLF5 underwent ubiquitination, and deletion of a 56-amino-acid sequence adjacent to a known transactivation domain of KLF5 significantly reduced its ubiquitination and degradation. Interestingly, cancer cells appeared to be more active in KLF5 degradation than untransformed epithelial cells, yet their proteasome activity was not higher. These results suggest that KLF5 protein is degraded at least in part through ubiquitination-proteasome pathway, which may have become hyperactive for KLF5 in cancer cells.
Collapse
Affiliation(s)
- Ceshi Chen
- Department of Oncology and Hematology, Winship Cancer Institute, Emory University School of Medicine, 1365-C Clifton Road, Atlanta, GA, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
The complex cross-regulation between Wnt signaling, cell-cell adhesion, and cell-matrix adhesion has revealed a number of regulatory components important in development and cancer progression. In the following, we would like to highlight and summarize some of the steps where pathways converge or diverge in regulating Wnt activity, matrix-induced pathways, and cell adhesion. We would like to focus on the involvement of heparan sulfate proteoglycan-rich proteins (HSPGs), integrin-mediated outside-in signaling, and cadherin-mediated cell-cell adhesion on Wnt pathways and the transcriptional regulation of extracellular matrix components and cell adhesion molecules by Wnt signaling.
Collapse
Affiliation(s)
- Alexandra Schambony
- Universität Karlsruhe, Zoologisches Institut II, Kaiserstrasse 12, D-76128 Karlsruhe, Germany.
| | | | | |
Collapse
|