1
|
Lyu J, Okada H, Sunagozaka H, Kawaguchi K, Shimakami T, Nio K, Murai K, Shirasaki T, Yoshida M, Arai K, Yamashita T, Tanaka T, Harada K, Takamura T, Kaneko S, Yamashita T, Honda M. Potential utility of l-carnitine for preventing liver tumors derived from metabolic dysfunction-associated steatohepatitis. Hepatol Commun 2024; 8:e0425. [PMID: 38619434 PMCID: PMC11019826 DOI: 10.1097/hc9.0000000000000425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 01/26/2024] [Indexed: 04/16/2024] Open
Abstract
BACKGROUND Recent reports have unveiled the potential utility of l-carnitine to alleviate metabolic dysfunction-associated steatohepatitis (MASH) by enhancing mitochondrial metabolic function. However, its efficacy at preventing the development of HCC has not been assessed fully. METHODS l-carnitine (2 g/d) was administered to 11 patients with MASH for 10 weeks, and blood liver function tests were performed. Five patients received a serial liver biopsy, and liver histology and hepatic gene expression were evaluated using this tissue. An atherogenic plus high-fat diet MASH mouse model received long-term l-carnitine administration, and liver histology and liver tumor development were evaluated. RESULTS Ten-week l-carnitine administration significantly improved serum alanine transaminase and aspartate transaminase levels along with a histological improvement in the NAFLD activity score, while steatosis and fibrosis were not improved. Gene expression profiling revealed a significant improvement in the inflammation and profibrotic gene signature as well as the recovery of lipid metabolism. Long-term l-carnitine administration to atherogenic plus high-fat diet MASH mice substantially improved liver histology (inflammation, steatosis, and fibrosis) and significantly reduced the incidence of liver tumors. l-carnitine directly reduced the expression of the MASH-associated and stress-induced transcriptional factor early growth response 1. Early growth response 1 activated the promoter activity of neural precursor cell expressed, developmentally downregulated protein 9 (NEDD9), an oncogenic protein. Thus, l-carnitine reduced the activation of the NEDD9, focal adhesion kinase 1, and AKT oncogenic signaling pathway. CONCLUSIONS Short-term l-carnitine administration ameliorated MASH through its anti-inflammatory effects. Long-term l-carnitine administration potentially improved the steatosis and fibrosis of MASH and may eventually reduce the risk of HCC.
Collapse
Affiliation(s)
- Junyan Lyu
- Department of Clinical Laboratory Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Hikari Okada
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Hajime Sunagozaka
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Kazunori Kawaguchi
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Tetsuro Shimakami
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Kouki Nio
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Kazuhisa Murai
- Department of Clinical Laboratory Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Takayoshi Shirasaki
- Department of Clinical Laboratory Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Mika Yoshida
- Department of Clinical Laboratory Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Kuniaki Arai
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Tatsuya Yamashita
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Takuji Tanaka
- Research Center of Diagnostic Pathology, Gifu Municipal Hospital, Gifu, Japan
| | - Kenichi Harada
- Department of Human Pathology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Toshinari Takamura
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Shuichi Kaneko
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Taro Yamashita
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Masao Honda
- Department of Clinical Laboratory Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| |
Collapse
|
2
|
NEDD9 sustains hexokinase expression to promote glycolysis. Oncogenesis 2022; 11:15. [PMID: 35410460 PMCID: PMC9001639 DOI: 10.1038/s41389-022-00391-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 03/18/2022] [Accepted: 03/23/2022] [Indexed: 11/08/2022] Open
Abstract
AbstractElevated rates of glycolysis in cancer cells support tumor growth, in a process that typically depends on oncogene-induced increases in the expression and/or activity of enzymes in the glycolytic pathway. The NEDD9 scaffolding protein is upregulated in many advanced tumors, with increased NEDD9 promoting the activity of SRC and other effectors that promote invasion and metastasis. We here define a new role for NEDD9 in support of glycolysis. NEDD9 knockdown significantly impaired glycolysis in multiple lung cancer cell lines This was accompanied by post-transcriptional downregulation of steady-state levels of hexokinases (HK1 and HK2), which catalyze early steps in the glycolytic cascade, key rate limiting enzyme phosphofructokinase (PFK1), and downstream glyceraldehyde phosphate dehydrogenase (GAPDH). In mice, protein levels of HK1, HK2, PFK1, and GAPDH were depressed in Krastm4Tyj/J/Trp53tm1Brn/J (KP) non-small cell lung tumors with null versus wild type Nedd9. Reciprocally, depletion of HK1 or HK2 elevated NEDD9 expression, as did the treatment of cells with 2-deoxyglucose (2DG), an inhibitor of glycolysis; whereas overexpression of hexokinases promoted NEDD9 dephosphorylation, associated with reduced NEDD9 activity. Together, these data for the first time suggest a negative feedback circuit involving NEDD9 and glycolytic enzymes that may contribute to NEDD9 action in promoting the aggressive growth of advanced tumors.
Collapse
|
3
|
Haouari S, Vourc’h P, Jeanne M, Marouillat S, Veyrat-Durebex C, Lanznaster D, Laumonnier F, Corcia P, Blasco H, Andres CR. The Roles of NEDD4 Subfamily of HECT E3 Ubiquitin Ligases in Neurodevelopment and Neurodegeneration. Int J Mol Sci 2022; 23:ijms23073882. [PMID: 35409239 PMCID: PMC8999422 DOI: 10.3390/ijms23073882] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 02/06/2023] Open
Abstract
The ubiquitin pathway regulates the function of many proteins and controls cellular protein homeostasis. In recent years, it has attracted great interest in neurodevelopmental and neurodegenerative diseases. Here, we have presented the first review on the roles of the 9 proteins of the HECT E3 ligase NEDD4 subfamily in the development and function of neurons in the central nervous system (CNS). We discussed their regulation and their direct or indirect involvement in neurodevelopmental diseases, such as intellectual disability, and neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease or Amyotrophic Lateral Sclerosis. Further studies on the roles of these proteins, their regulation and their targets in neurons will certainly contribute to a better understanding of neuronal function and dysfunction, and will also provide interesting information for the development of therapeutics targeting them.
Collapse
Affiliation(s)
- Shanez Haouari
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
| | - Patrick Vourc’h
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
- CHRU de Tours, Service de Biochimie et Biologie Moléculaire, 37044 Tours, France
- Correspondence: ; Tel.: +33-(0)2-34-37-89-10; Fax: +33-(0)2-47-36-61-85
| | - Médéric Jeanne
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
- CHRU de Tours, Service de Génétique, 37044 Tours, France
| | - Sylviane Marouillat
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
| | - Charlotte Veyrat-Durebex
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
- CHRU de Tours, Service de Biochimie et Biologie Moléculaire, 37044 Tours, France
| | - Débora Lanznaster
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
| | - Frédéric Laumonnier
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
| | - Philippe Corcia
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
- CHRU de Tours, Service de Neurologie, 37044 Tours, France
| | - Hélène Blasco
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
- CHRU de Tours, Service de Biochimie et Biologie Moléculaire, 37044 Tours, France
| | - Christian R. Andres
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
- CHRU de Tours, Service de Biochimie et Biologie Moléculaire, 37044 Tours, France
| |
Collapse
|
4
|
Li B, Sun G, Yu H, Meng J, Wei F. Exosomal circTAOK1 contributes to diabetic kidney disease progression through regulating SMAD3 expression by sponging miR-520h. Int Urol Nephrol 2022; 54:2343-2354. [PMID: 35142978 DOI: 10.1007/s11255-022-03139-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 01/30/2022] [Indexed: 12/21/2022]
Abstract
BACKGROUND Diabetic nephropathy (DN) is a frequent diabetes complication with complex pathogenesis. Circular RNA (circRNA) circTAOK1 (also named circ_0003928) has been reported to be upregulated in high glucose (HG)-treated human umbilical vein endothelial cells. Also, exosomal circRNAs can exert significant roles in the pathology of various diseases. This study is designed to explore the role and mechanism of exosomal circTAOK1 on the glomerular mesangial cell (GMC) injury in DN. METHODS Exosomes were detected by a transmission electron microscope. The protein levels of CD9, CD63, proliferating cell nuclear antigen (PCNA), cyclinD1, α-SMA, fibronectin, E-cadherin, N-cadherin, and SMAD family member 3 (SMAD3) were examined by western blot assay. circTAOK1, microRNA-520h (miR-520h), and SMAD3 levels were detected by real-time quantitative polymerase chain reaction (RT-qPCR). Cell proliferation and cell cycle progression were detected by cell counting kit-8 (CCK-8), 5-ethynyl-2'-deoxyuridine (EdU), and flow cytometry assays. The binding relationship between miR-520h and circTAOK1 or SMAD3 was predicted by Starbase and then verified by a dual-luciferase reporter and RNA immunoprecipitation (RIP), RNA pull-down assays. RESULTS CircTAOK1 expression was upregulated in the exosomes isolated from HG-treated glomerular epithelial cells (GEC). Moreover, GEC-circTAOK1-Exo could promote proliferation, fibrosis, and epithelial-mesenchymal transition (EMT) of glomerular mesangial cells (GMCs). Mechanically, circTAOK1 could regulate SMAD3 expression by sponging miR-520h, GEO-si-circTAOK1 Exo-induced miR-520h and repressed SMAD3 expression in GMC. CONCLUSION GEC-circTAOK1-Exo could boost proliferation, fibrosis, and EMT of GMC through targeting the miR-520h/SMAD3 axis, providing new insights into the pathogenesis of DN.
Collapse
Affiliation(s)
- Bo Li
- Department of Blood Purification, The Second Hospital of Tianjin Medical University, No. 23 Pingjiang Road, Hexi District, Tianjin, China
| | - Guijiang Sun
- Department of Blood Purification, The Second Hospital of Tianjin Medical University, No. 23 Pingjiang Road, Hexi District, Tianjin, China
| | - Haibo Yu
- Department of Blood Purification, The Second Hospital of Tianjin Medical University, No. 23 Pingjiang Road, Hexi District, Tianjin, China
| | - Jia Meng
- Department of Blood Purification, The Second Hospital of Tianjin Medical University, No. 23 Pingjiang Road, Hexi District, Tianjin, China
| | - Fang Wei
- Department of Blood Purification, The Second Hospital of Tianjin Medical University, No. 23 Pingjiang Road, Hexi District, Tianjin, China.
| |
Collapse
|
5
|
Deneka AY, Kopp MC, Nikonova AS, Gaponova AV, Kiseleva AA, Hensley HH, Flieder DB, Serebriiskii IG, Golemis EA. Nedd9 Restrains Autophagy to Limit Growth of Early Stage Non-Small Cell Lung Cancer. Cancer Res 2021; 81:3717-3726. [PMID: 34006524 DOI: 10.1158/0008-5472.can-20-3626] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 03/16/2021] [Accepted: 04/26/2021] [Indexed: 01/22/2023]
Abstract
Non-small cell lung cancer (NSCLC) is the most common cancer worldwide. With overall 5-year survival estimated at <17%, it is critical to identify factors that regulate NSCLC disease prognosis. NSCLC is commonly driven by mutations in KRAS and TP53, with activation of additional kinases such as SRC promoting tumor invasion. In this study, we investigated the role of NEDD9, a SRC activator and scaffolding protein, in NSCLC tumorigenesis. In an inducible model of NSCLC dependent on Kras mutation and Trp53 loss (KP mice), deletion of Nedd9 (KPN mice) led to the emergence of larger tumors characterized by accelerated rates of tumor growth and elevated proliferation. Orthotopic injection of KP and KPN tumors into the lungs of Nedd9-wild-type and -null mice indicated the effect of Nedd9 loss was cell-autonomous. Tumors in KPN mice displayed reduced activation of SRC and AKT, indicating that activation of these pathways did not mediate enhanced growth of KPN tumors. NSCLC tumor growth has been shown to require active autophagy, a process dependent on activation of the kinases LKB1 and AMPK. KPN tumors contained high levels of active LKB1 and AMPK and increased autophagy compared with KP tumors. Treatment with the autophagy inhibitor chloroquine completely eliminated the growth advantage of KPN tumors. These data for the first time identify NEDD9 as a negative regulator of LKB1/AMPK-dependent autophagy during early NSCLC tumor growth. SIGNIFICANCE: This study demonstrates a novel role for the scaffolding protein NEDD9 in regulating LKB1-AMPK signaling in early stage non-small cell lung cancer, suppressing autophagy and tumor growth.
Collapse
Affiliation(s)
- Alexander Y Deneka
- Program in Molecular Therapeutics Fox Chase Cancer Center, Philadelphia, PA.,Kazan Federal University, Kazan, Russian Federation, Kazan, Tatarstan, Russia
| | - Meghan C Kopp
- Program in Molecular Therapeutics Fox Chase Cancer Center, Philadelphia, PA.,Cancer Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Anna S Nikonova
- Program in Molecular Therapeutics Fox Chase Cancer Center, Philadelphia, PA
| | - Anna V Gaponova
- Program in Molecular Therapeutics Fox Chase Cancer Center, Philadelphia, PA
| | - Anna A Kiseleva
- Program in Molecular Therapeutics Fox Chase Cancer Center, Philadelphia, PA
| | - Harvey H Hensley
- Program in Molecular Therapeutics Fox Chase Cancer Center, Philadelphia, PA
| | - Douglas B Flieder
- Program in Molecular Therapeutics Fox Chase Cancer Center, Philadelphia, PA.,Department of Pathology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | | | - Erica A Golemis
- Program in Molecular Therapeutics Fox Chase Cancer Center, Philadelphia, PA.
| |
Collapse
|
6
|
Radix Scutellariae Ameliorates Stress-Induced Depressive-Like Behaviors via Protecting Neurons through the TGF β3-Smad2/3-Nedd9 Signaling Pathway. Neural Plast 2020; 2020:8886715. [PMID: 33273910 PMCID: PMC7683137 DOI: 10.1155/2020/8886715] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/29/2020] [Accepted: 10/31/2020] [Indexed: 12/18/2022] Open
Abstract
Chronic stress can impair hippocampal neurogenesis, increase neuronal apoptosis, and cause depressive-like behaviors. Our previous studies found that Radix Scutellariae (RS) can rescue the stress-induced neuronal injury, but the mechanism is not clear. Here, we continued to investigate the underlying antidepressant mechanisms of the RS extract. A 7-week chronic unpredictable mild stress (CUMS) procedure was used to establish a murine depression model. 0.75 g/kg or 1.5 g/kg RS was administered daily to the mice during the last 4 weeks. Depressive-like behaviors were evaluated by the sucrose preference test (SPT), forced swimming test (FST), open field test (OFT), and tail suspension test (TST). The neuroprotective effect of RS was evaluated with the expression of hippocampal neuron-related markers and apoptosis-associated proteins by Nissl staining, immunohistochemistry, and western blot. Transforming growth factor-β3 (TGFβ3) pathway-related proteins were detected by western blot. Results showed that RS could ameliorate depressive-like behaviors, increase the expression of the antiapoptotic protein B-cell lymphoma 2 (BCL-2), reduce the expression of the proapoptotic protein BCL-2-associated X (BAX), and increase the number of doublecortin- (DCX-), microtubule-associated protein 2- (MAP2-), and neuronal nucleus- (NeuN-) positive cells in the hippocampus. Moreover, RS could reverse the CUMS-induced decrease of TGFβ3 protein, promote the phosphorylation of SMAD2/3, and increase the expression of downstream NEDD9 protein. These results suggest that RS could exert antidepressant effects via protecting neurons. And the molecular mechanism might be related to the regulation of the TGFβ3-SMAD2/3-NEDD9 pathway.
Collapse
|
7
|
Lee KH. Involvement of Wnt signaling in primary cilia assembly and disassembly. FEBS J 2020; 287:5027-5038. [PMID: 33015954 DOI: 10.1111/febs.15579] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/21/2020] [Accepted: 09/29/2020] [Indexed: 11/28/2022]
Abstract
The primary cilium is a nonmotile microtubule-based structure, which functions as an antenna-like cellular sensing organelle. The primary cilium is assembled from the basal body, a mother centriole-based structure, during interphase or a quiescent cell stage, and rapidly disassembles before entering mitosis in a dynamic cycle. Defects in this ciliogenesis dynamics are associated with human diseases such as ciliopathy and cancer, but the molecular mechanisms of the ciliogenesis dynamics are still largely unknown. To date, various cellular signaling pathways associated with primary cilia have been proposed, but the main signaling pathways regulating primary cilia assembly/disassembly remain enigmatic. This review describes recent findings in Wnt-induced primary cilia assembly/disassembly and potential future directions for the study of the cellular signaling related to the primary ciliogenesis dynamics.
Collapse
Affiliation(s)
- Kyung Ho Lee
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Korea
| |
Collapse
|
8
|
Zhang C, Wang T, Wu H, Zhang L, Li K, Wang F, Chen Y, Jin J, Hua D. HEF1 regulates differentiation through the Wnt5a/β-catenin signaling pathway in human gastric cancer. Biochem Biophys Res Commun 2019; 509:201-208. [DOI: 10.1016/j.bbrc.2018.12.104] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 12/14/2018] [Indexed: 02/06/2023]
|
9
|
Akhtar N, Syed DN, Lall RK, Mirza B, Mukhtar H. Targeting epithelial to mesenchymal transition in prostate cancer by a novel compound, plectranthoic acid, isolated fromFicus microcarpa. Mol Carcinog 2018; 57:653-663. [DOI: 10.1002/mc.22790] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 01/30/2018] [Accepted: 02/12/2018] [Indexed: 01/04/2023]
Affiliation(s)
- Nosheen Akhtar
- Department of Dermatology; School of Medicine and Public Health; University of Wisconsin; Madison Wisconsin
- Department of Molecular Medicine; National University of Medical Sciences; Rawalpindi Pakistan
| | - Deeba N. Syed
- Department of Dermatology; School of Medicine and Public Health; University of Wisconsin; Madison Wisconsin
| | - Rahul K. Lall
- Department of Dermatology; School of Medicine and Public Health; University of Wisconsin; Madison Wisconsin
| | - Bushra Mirza
- Department of Biochemistry; Faculty of Biological Sciences; Quaid-i-Azam University; Islamabad Pakistan
| | - Hasan Mukhtar
- Department of Dermatology; School of Medicine and Public Health; University of Wisconsin; Madison Wisconsin
| |
Collapse
|
10
|
Halakou F, Kilic ES, Cukuroglu E, Keskin O, Gursoy A. Enriching Traditional Protein-protein Interaction Networks with Alternative Conformations of Proteins. Sci Rep 2017; 7:7180. [PMID: 28775330 PMCID: PMC5543104 DOI: 10.1038/s41598-017-07351-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 06/27/2017] [Indexed: 12/19/2022] Open
Abstract
Traditional Protein-Protein Interaction (PPI) networks, which use a node and edge representation, lack some valuable information about the mechanistic details of biological processes. Mapping protein structures to these PPI networks not only provides structural details of each interaction but also helps us to find the mutual exclusive interactions. Yet it is not a comprehensive representation as it neglects the conformational changes of proteins which may lead to different interactions, functions, and downstream signalling. In this study, we proposed a new representation for structural PPI networks inspecting the alternative conformations of proteins. We performed a large-scale study by creating breast cancer metastasis network and equipped it with different conformers of proteins. Our results showed that although 88% of proteins in our network has at least two structures in Protein Data Bank (PDB), only 22% of them have alternative conformations and the remaining proteins have different regions saved in PDB. However, using even this small set of alternative conformations we observed a considerable increase in our protein docking predictions. Our protein-protein interaction predictions increased from 54% to 76% using the alternative conformations. We also showed the benefits of investigating structural data and alternative conformations of proteins through three case studies.
Collapse
Affiliation(s)
- Farideh Halakou
- Department of Computer Engineering, Koc University, Istanbul, 34450, Turkey
| | - Emel Sen Kilic
- Department of Chemical and Biological Engineering, Koc University, Istanbul, 34450, Turkey.,Microbiology, Immunology and Cell Biology Department, West Virginia University, Morgantown, 26505, WV, USA
| | - Engin Cukuroglu
- Computational Sciences and Engineering, Graduate School of Sciences and Engineering, Koc University, Istanbul, 34450, Turkey
| | - Ozlem Keskin
- Department of Chemical and Biological Engineering, Koc University, Istanbul, 34450, Turkey
| | - Attila Gursoy
- Department of Computer Engineering, Koc University, Istanbul, 34450, Turkey.
| |
Collapse
|
11
|
Kumar S, Sharma G, Chakraborty C, Sharma AR, Kim J. Regulatory functional territory of PLK-1 and their substrates beyond mitosis. Oncotarget 2017; 8:37942-37962. [PMID: 28415805 PMCID: PMC5514964 DOI: 10.18632/oncotarget.16290] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 03/03/2017] [Indexed: 12/04/2022] Open
Abstract
Polo-like kinase 1 (PLK-1) is a well-known (Ser/Thr) mitotic protein kinase and is considered as a proto-oncogene. As hyper-activation of PLK-1 is broadly associated with poor prognosis and cancer progression, it is one of the most extensively studied mitotic kinases. During mitosis, PLK-1 regulates various cell cycle events, such as spindle pole maturation, chromosome segregation and cytokinesis. However, studies have demonstrated that the role of PLK-1 is not only restricted to mitosis, but PLK-1 can also regulate other vital events beyond mitosis, including transcription, translation, ciliogenesis, checkpoint adaptation and recovery, apoptosis, chromosomes dynamics etc. Recent reviews have tried to define the regulatory role of PLK-1 during mitosis progression and tumorigenesis, but its' functional role beyond mitosis is still largely unexplored. PLK-1 can regulate the activity of many proteins that work outside of its conventional territory. The dysregulation of these proteins can cause diseases such as Alzheimer's disease, tumorigenesis etc. and may also lead to drug resistance. Thus, in this review, we discussed the versatile role of PLK-1 and tried to collect data to validate its' functional role in cell cycle regulation apart from mitosis.
Collapse
Affiliation(s)
- Shiv Kumar
- Department of Biochemistry, Institute of Cell Differentiation and Aging, Hallym University, College of Medicine, Chucheonsi, Gangwondo, Republic of Korea
| | - Garima Sharma
- Institute For Skeletal Aging & Orthopedic Surgery, Hallym University, College of Medicine, Chucheonsi, Gangwondo, Republic of Korea
| | - Chiranjib Chakraborty
- Department of Bio-informatics, School of Computer and Information Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Ashish Ranjan Sharma
- Institute For Skeletal Aging & Orthopedic Surgery, Hallym University, College of Medicine, Chucheonsi, Gangwondo, Republic of Korea
| | - Jaebong Kim
- Department of Biochemistry, Institute of Cell Differentiation and Aging, Hallym University, College of Medicine, Chucheonsi, Gangwondo, Republic of Korea
| |
Collapse
|
12
|
Bradbury PM, Turner K, Mitchell C, Griffin KR, Middlemiss S, Lau L, Dagg R, Taran E, Cooper-White J, Fabry B, O’Neill GM. The focal adhesion targeting (FAT) domain of p130 Crk associated substrate (p130Cas) confers mechanosensing function. J Cell Sci 2017; 130:1263-1273. [DOI: 10.1242/jcs.192930] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 02/02/2017] [Indexed: 11/20/2022] Open
Abstract
The Cas family of focal adhesion proteins contain a highly conserved C-terminal focal adhesion targeting (FAT) domain. To determine the role of the FAT domain we compared wildtype exogenous NEDD9 with a hybrid construct in which the NEDD9 FAT domain is exchanged for the p130Cas FAT domain. Fluorescence recovery after photobleaching (FRAP) revealed significantly slowed exchange of the fusion protein at focal adhesions and significantly slower 2D migration. No differences were detected in cell stiffness measured with Atomic Force Microscopy (AFM) and cell adhesion forces measured with a magnetic tweezer device. Thus the slowed migration was not due to changes in cell stiffness or adhesion strength. Analysis of cell migration on surfaces of increasing rigidity revealed a striking reduction of cell motility in cells expressing the p130Cas FAT domain. The p130Cas FAT domain induced rigidity-dependent tyrosine phosphorylation of the NEDD9 substrate domain. This in turn reduced post-translational cleavage of NEDD9 which we show inhibits NEDD9-induced migration. Collectively, our data therefore suggest that the p130Cas FAT domain uniquely confers mechanosensing function.
Collapse
Affiliation(s)
- Peta M. Bradbury
- Children’s Cancer Research Unit, Kids Research Institute, The Children’s Hospital at Westmead, Westmead, 2145, New South Wales, Australia
- Discipline of Paediatrics and Child Health, The University of Sydney, Sydney, 2000, New South Wales, Australia
| | - Kylie Turner
- Children’s Cancer Research Unit, Kids Research Institute, The Children’s Hospital at Westmead, Westmead, 2145, New South Wales, Australia
| | - Camilla Mitchell
- Children’s Cancer Research Unit, Kids Research Institute, The Children’s Hospital at Westmead, Westmead, 2145, New South Wales, Australia
| | - Kaitlyn R. Griffin
- Children’s Cancer Research Unit, Kids Research Institute, The Children’s Hospital at Westmead, Westmead, 2145, New South Wales, Australia
| | - Shiloh Middlemiss
- Children’s Cancer Research Unit, Kids Research Institute, The Children’s Hospital at Westmead, Westmead, 2145, New South Wales, Australia
| | - Loretta Lau
- Children’s Cancer Research Unit, Kids Research Institute, The Children’s Hospital at Westmead, Westmead, 2145, New South Wales, Australia
| | - Rebecca Dagg
- Children’s Cancer Research Unit, Kids Research Institute, The Children’s Hospital at Westmead, Westmead, 2145, New South Wales, Australia
| | - Elena Taran
- Australian National Fabrication Facility- Queensland node, Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St. Lucia, Queensland, Australia
| | - Justin Cooper-White
- Tissue Engineering and Microfluidics Laboratory, Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St. Lucia, Queensland, Australia
| | - Ben Fabry
- Department of Physics, University of Erlangen-Nuremberg, Germany
| | - Geraldine M. O’Neill
- Children’s Cancer Research Unit, Kids Research Institute, The Children’s Hospital at Westmead, Westmead, 2145, New South Wales, Australia
- Discipline of Paediatrics and Child Health, The University of Sydney, Sydney, 2000, New South Wales, Australia
| |
Collapse
|
13
|
Omata Y, Nakamura S, Koyama T, Yasui T, Hirose J, Izawa N, Matsumoto T, Imai Y, Seo S, Kurokawa M, Tsutsumi S, Kadono Y, Morimoto C, Aburatani H, Miyamoto T, Tanaka S. Identification of Nedd9 as a TGF-β-Smad2/3 Target Gene Involved in RANKL-Induced Osteoclastogenesis by Comprehensive Analysis. PLoS One 2016; 11:e0157992. [PMID: 27336669 PMCID: PMC4918979 DOI: 10.1371/journal.pone.0157992] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 06/08/2016] [Indexed: 12/18/2022] Open
Abstract
TGF-ß is a multifunctional cytokine that is involved in cell proliferation, differentiation and function. We previously reported an essential role of the TGF-ß -Smad2/3 pathways in RANKL-induced osteoclastogenesis. Using chromatin immunoprecipitation followed by sequencing, we comprehensively identified Smad2/3 target genes in bone marrow macrophages. These genes were enriched in the gene population upregulated by TGF-ß and downregulated by RANKL. Recent studies have revealed that histone modifications, such as trimethylation of histone H3 lysine 4 (H3K4me3) and lysine 27 (H3K27me3), critically regulate key developmental steps. We identified Nedd9 as a Smad2/3 target gene whose histone modification pattern was converted from H3K4me3(+)/H3K4me27(+) to H3K4me3(+)/H3K4me27(-) by TGF-ß. Nedd9 expression was increased by TGF-ß and suppressed by RANKL. Overexpression of Nedd9 partially rescued an inhibitory effect of a TGF-ß inhibitor, while gene silencing of Nedd9 suppressed RANKL-induced osteoclastogenesis. RANKL-induced osteoclastogenesis were reduced and stimulatory effects of TGF-ß on RANKL-induced osteoclastogenesis were partially abrogated in cells from Nedd9-deficient mice although knockout mice did not show abnormal skeletal phenotypes. These results suggest that Nedd9 is a Smad2/3 target gene implicated in RANKL-induced osteoclastogenesis.
Collapse
Affiliation(s)
- Yasunori Omata
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Shinya Nakamura
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Takuma Koyama
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Tetsuro Yasui
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Jun Hirose
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Naohiro Izawa
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Takumi Matsumoto
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yuuki Imai
- Division of Integrative Pathophysiology, Proteo-Science Center, Graduate School of Medicine, Ehime University, Ehime 791–0295, Japan
| | - Sachiko Seo
- Department of Hematology and Oncology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Mineo Kurokawa
- Department of Hematology and Oncology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Shuichi Tsutsumi
- Genome Science Division, Research Center for Advanced Science and Technology (RCAST), The University of Tokyo, Tokyo, Japan
| | - Yuho Kadono
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Chikao Morimoto
- Department of Therapy Development and Innovation for Immune Disorders and Cancers, Juntendo University, Tokyo, Japan
| | - Hiroyuki Aburatani
- Genome Science Division, Research Center for Advanced Science and Technology (RCAST), The University of Tokyo, Tokyo, Japan
| | - Takeshi Miyamoto
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Department of Orthopedic Surgery, Keio University, Tokyo, Japan
| | - Sakae Tanaka
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- * E-mail:
| |
Collapse
|
14
|
Abstract
Itch or itchy E3 ubiquitin ligase was initially discovered by genetic studies on the mouse coat color changes, and its deletion results in an itchy phenotype with constant skin scratching and multi-organ inflammation. It is a member of the homologous to E6-associated protein C-terminus (HECT)-type family of E3 ligases, with the protein-interacting WW-domains for the recruitment of substrate and the HECT domain for the transfer of ubiquitin to the substrate. Since its discovery, numerous studies have demonstrated that Itch is involved in the control of many aspects of immune responses including T-cell activation and tolerance and T-helper cell differentiation. Itch is also implicated in other biological contexts such as tumorigenesis, development, and stress responses. Many signaling pathways are regulated by Itch-promoted ubiquitylation of diverse target proteins. Itch is also involved in human diseases. Here, we discuss the major progress in understanding the biological significance of Itch-promoted protein ubiquitylation in the immune and other systems and in Itch-mediated regulation of signal transduction.
Collapse
Affiliation(s)
- Daisuke Aki
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China.,Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Wen Zhang
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Yun-Cai Liu
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China.,Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| |
Collapse
|
15
|
Latasa MJ, Jiménez-Lara AM, Cosgaya JM. Retinoic acid regulates Schwann cell migration via NEDD9 induction by transcriptional and post-translational mechanisms. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1510-8. [PMID: 27085739 DOI: 10.1016/j.bbamcr.2016.04.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 03/30/2016] [Accepted: 04/11/2016] [Indexed: 12/14/2022]
Abstract
Schwann cell migration is essential during the regenerative response to nerve injury, however, the factors that regulate this phenomenon are not yet clear. Here we describe that retinoic acid (RA), whose production and signaling activity are greatly enhanced during nerve regeneration, increases Schwann cell migration. This is accompanied by the up-regulation of NEDD9, a member of the CAS family of scaffold proteins previously implicated in migratory and invasive behavior in gliomas, melanomas and the neural crest cells from which Schwann cells derive. This RA-induced NEDD9 accumulation is due to augmented mRNA levels, as well as an increase of NEDD9 protein stability. Although all NEDD9 phospho-isoforms present in Schwann cells are induced by the retinoid, the hormone also changes its phosphorylation status, thus altering the ratio between the different isoforms. Silencing NEDD9 in Schwann cells had no effect on basal migratory ability, but completely abrogated RA-induced enhanced migration. Collectively, our results indicate that RA could be a major regulator of Schwann cell migration after nerve injury, thus offering a new insight into peripheral nerve repair.
Collapse
Affiliation(s)
- Maria-Jesus Latasa
- Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Spain; Department of Endocrine and Nervous System Physiopathology, Arturo Duperier, 4, 28029 Madrid, Spain
| | - Ana María Jiménez-Lara
- Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Spain; Department of Endocrine and Nervous System Physiopathology, Arturo Duperier, 4, 28029 Madrid, Spain
| | - Jose Miguel Cosgaya
- Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Spain; Department of Endocrine and Nervous System Physiopathology, Arturo Duperier, 4, 28029 Madrid, Spain.
| |
Collapse
|
16
|
ZHANG SISEN, WU LIHUA. Roles of neural precursor cell expressed, developmentally downregulated 9 in tumor-associated cellular processes (Review). Mol Med Rep 2015; 12:6415-21. [DOI: 10.3892/mmr.2015.4240] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 06/15/2015] [Indexed: 11/05/2022] Open
|
17
|
Ibrahim R, Lemoine A, Bertoglio J, Raingeaud J. Human enhancer of filamentation 1-induced colorectal cancer cell migration: Role of serine phosphorylation and interaction with the breast cancer anti-estrogen resistance 3 protein. Int J Biochem Cell Biol 2015; 64:45-57. [DOI: 10.1016/j.biocel.2015.03.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 03/11/2015] [Accepted: 03/18/2015] [Indexed: 02/02/2023]
|
18
|
Shagisultanova E, Gaponova AV, Gabbasov R, Nicolas E, Golemis EA. Preclinical and clinical studies of the NEDD9 scaffold protein in cancer and other diseases. Gene 2015; 567:1-11. [PMID: 25967390 DOI: 10.1016/j.gene.2015.04.086] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 04/20/2015] [Accepted: 04/21/2015] [Indexed: 12/17/2022]
Abstract
Cancer progression requires a significant reprogramming of cellular signaling to support the essential tumor-specific processes that include hyperproliferation, invasion (for solid tumors) and survival of metastatic colonies. NEDD9 (also known as CasL and HEF1) encodes a multi-domain scaffolding protein that assembles signaling complexes regulating multiple cellular processes relevant to cancer. These include responsiveness to signals emanating from the T and B cell receptors, integrins, chemokine receptors, and receptor tyrosine kinases, as well as cytoplasmic oncogenes such as BCR-ABL and FAK- and SRC-family kinases. Downstream, NEDD9 regulation of partners including CRKL, WAVE, PI3K/AKT, ERK, E-cadherin, Aurora-A (AURKA), HDAC6, and others allow NEDD9 to influence functions as pleiotropic as migration, invasion, survival, ciliary resorption, and mitosis. In this review, we summarize a growing body of preclinical and clinical data that indicate that while NEDD9 is itself non-oncogenic, changes in expression of NEDD9 (most commonly elevation of expression) are common features of tumors, and directly impact tumor aggressiveness, metastasis, and response to at least some targeted agents inhibiting NEDD9-interacting proteins. These data strongly support the relevance of further development of NEDD9 as a biomarker for therapeutic resistance. Finally, we briefly discuss emerging evidence supporting involvement of NEDD9 in additional pathological conditions, including stroke and polycystic kidney disease.
Collapse
Affiliation(s)
- Elena Shagisultanova
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Anna V Gaponova
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Rashid Gabbasov
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; Department of Genetics, Kazan Federal University (Volga Region), Kazan, Tatarstan, Russia
| | - Emmanuelle Nicolas
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Erica A Golemis
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.
| |
Collapse
|
19
|
Beck TN, Chikwem AJ, Solanki NR, Golemis EA. Bioinformatic approaches to augment study of epithelial-to-mesenchymal transition in lung cancer. Physiol Genomics 2014; 46:699-724. [PMID: 25096367 PMCID: PMC4187119 DOI: 10.1152/physiolgenomics.00062.2014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 08/04/2014] [Indexed: 12/22/2022] Open
Abstract
Bioinformatic approaches are intended to provide systems level insight into the complex biological processes that underlie serious diseases such as cancer. In this review we describe current bioinformatic resources, and illustrate how they have been used to study a clinically important example: epithelial-to-mesenchymal transition (EMT) in lung cancer. Lung cancer is the leading cause of cancer-related deaths and is often diagnosed at advanced stages, leading to limited therapeutic success. While EMT is essential during development and wound healing, pathological reactivation of this program by cancer cells contributes to metastasis and drug resistance, both major causes of death from lung cancer. Challenges of studying EMT include its transient nature, its molecular and phenotypic heterogeneity, and the complicated networks of rewired signaling cascades. Given the biology of lung cancer and the role of EMT, it is critical to better align the two in order to advance the impact of precision oncology. This task relies heavily on the application of bioinformatic resources. Besides summarizing recent work in this area, we use four EMT-associated genes, TGF-β (TGFB1), NEDD9/HEF1, β-catenin (CTNNB1) and E-cadherin (CDH1), as exemplars to demonstrate the current capacities and limitations of probing bioinformatic resources to inform hypothesis-driven studies with therapeutic goals.
Collapse
Affiliation(s)
- Tim N Beck
- Developmental Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania; Program in Molecular and Cell Biology and Genetics, Drexel University College of Medicine, Philadelphia, Pennsylvania; and
| | - Adaeze J Chikwem
- Developmental Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania; Temple University School of Medicine, Philadelphia, Pennsylvania; and
| | - Nehal R Solanki
- Immune Cell Development and Host Defense Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania; Program in Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Erica A Golemis
- Developmental Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania; Temple University School of Medicine, Philadelphia, Pennsylvania; and Program in Molecular and Cell Biology and Genetics, Drexel University College of Medicine, Philadelphia, Pennsylvania; and
| |
Collapse
|
20
|
Bradbury P, Bach CT, Paul A, O'Neill GM. Src kinase determines the dynamic exchange of the docking protein NEDD9 (neural precursor cell expressed developmentally down-regulated gene 9) at focal adhesions. J Biol Chem 2014; 289:24792-800. [PMID: 25059660 DOI: 10.1074/jbc.m113.544106] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Dynamic exchange of molecules between the cytoplasm and integrin-based focal adhesions provides a rapid response system for modulating cell adhesion. Increased residency time of molecules that regulate adhesion turnover contributes to adhesion stability, ultimately determining migration speed across two-dimensional surfaces. In the present study we test the role of Src kinase in regulating dynamic exchange of the focal adhesion protein NEDD9/HEF1/Cas-L. Using either chemical inhibition or fibroblasts genetically null for Src together with fluorescence recovery after photobleaching (FRAP), we find that Src significantly reduces NEDD9 exchange at focal adhesions. Analysis of NEDD9 mutant constructs with the two major Src-interacting domains disabled revealed the greatest effects were due to the NEDD9 SH2 binding domain. This correlated with a significant change in two-dimensional migratory speed. Given the emerging role of NEDD9 as a regulator of focal adhesion stability, the time of NEDD9 association at the focal adhesions is key in modulating rates of migration and invasion. Our study suggests that Src kinase activity determines NEDD9 exchange at focal adhesions and may similarly modulate other focal adhesion-targeted Src substrates to regulate cell migration.
Collapse
Affiliation(s)
- Peta Bradbury
- From the Children's Cancer Research Unit, Kids Research Institute at The Children's Hospital at Westmead, Westmead 2145, New South Wales and Discipline of Paediatrics and Child Health, University of Sydney, New South Wales 2006, Australia
| | - Cuc T Bach
- From the Children's Cancer Research Unit, Kids Research Institute at The Children's Hospital at Westmead, Westmead 2145, New South Wales and
| | - Andre Paul
- From the Children's Cancer Research Unit, Kids Research Institute at The Children's Hospital at Westmead, Westmead 2145, New South Wales and
| | - Geraldine M O'Neill
- From the Children's Cancer Research Unit, Kids Research Institute at The Children's Hospital at Westmead, Westmead 2145, New South Wales and Discipline of Paediatrics and Child Health, University of Sydney, New South Wales 2006, Australia
| |
Collapse
|
21
|
Adaptors for disorders of the brain? The cancer signaling proteins NEDD9, CASS4, and PTK2B in Alzheimer's disease. Oncoscience 2014; 1:486-503. [PMID: 25594051 PMCID: PMC4278314 DOI: 10.18632/oncoscience.64] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 07/23/2014] [Indexed: 12/19/2022] Open
Abstract
No treatment strategies effectively limit the progression of Alzheimer's disease (AD), a common and debilitating neurodegenerative disorder. The absence of viable treatment options reflects the fact that the pathophysiology and genotypic causes of the disease are not well understood. The advent of genome-wide association studies (GWAS) has made it possible to broadly investigate genotypic alterations driving phenotypic occurrences. Recent studies have associated single nucleotide polymorphisms (SNPs) in two paralogous scaffolding proteins, NEDD9 and CASS4, and the kinase PTK2B, with susceptibility to late-onset AD (LOAD). Intriguingly, NEDD9, CASS4, and PTK2B have been much studied as interacting partners regulating oncogenesis and metastasis, and all three are known to be active in the brain during development and in cancer. However, to date, the majority of studies of these proteins have emphasized their roles in the directly cancer relevant processes of migration and survival signaling. We here discuss evidence for roles of NEDD9, CASS4 and PTK2B in additional processes, including hypoxia, vascular changes, inflammation, microtubule stabilization and calcium signaling, as potentially relevant to the pathogenesis of LOAD. Reciprocally, these functions can better inform our understanding of the action of NEDD9, CASS4 and PTK2B in cancer.
Collapse
|
22
|
TGF-β signaling in stem cells and tumorigenesis. Mol Oncol 2013. [DOI: 10.1017/cbo9781139046947.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
23
|
Rink L, Ochs MF, Zhou Y, von Mehren M, Godwin AK. ZNF-mediated resistance to imatinib mesylate in gastrointestinal stromal tumor. PLoS One 2013; 8:e54477. [PMID: 23372733 PMCID: PMC3556080 DOI: 10.1371/journal.pone.0054477] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 12/12/2012] [Indexed: 12/14/2022] Open
Abstract
Although imatinib mesylate (IM) has transformed the treatment of gastrointestinal stromal tumors (GIST), many patients experience primary/secondary drug resistance. In a previous study, we identified a gene signature, consisting mainly of Kruppel-associated box (KRAB) domain containing zinc finger (ZNF) transcriptional repressors that predict short-term response to IM. To determine if these genes have functional significance, a siRNA library targeting these genes was constructed and applied to GIST cells in vitro. These screens identified seventeen “IM sensitizing genes” in GIST cells (sensitization index (SI) <0.85 ratio of drug/vehicle) with a false discovery rate (FDR) <15%, including twelve ZNF genes, the majority of which are located within the HSA19p12–13.1 locus. These genes were shown to be highly specific to IM and another tyrosine kinase inhibitor (TKI), sunitinib, in GIST cells. In order to determine mechanistically how these ZNFs might be modulating response to IM, RNAi approaches were used to individually silence genes within the predictive signature in GIST cells and expression profiling was performed. Knockdown of the 14 IM-sensitizing genes (10 ZNFs) universally led to downregulation of six genes, including TGFb3, periostin, and NEDD9. These studies implicate a role of KRAB-ZNFs in modulating response to TKIs in GIST.
Collapse
Affiliation(s)
- Lori Rink
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America.
| | | | | | | | | |
Collapse
|
24
|
Lee KH, Johmura Y, Yu LR, Park JE, Gao Y, Bang JK, Zhou M, Veenstra TD, Yeon Kim B, Lee KS. Identification of a novel Wnt5a-CK1ɛ-Dvl2-Plk1-mediated primary cilia disassembly pathway. EMBO J 2012; 31:3104-17. [PMID: 22609948 PMCID: PMC3400010 DOI: 10.1038/emboj.2012.144] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2011] [Accepted: 04/18/2012] [Indexed: 01/17/2023] Open
Abstract
Non-motile primary cilium is an antenna-like structure whose defect is associated with a wide range of pathologies, including developmental disorders and cancer. Although mechanisms regulating cilia assembly have been extensively studied, how cilia disassembly is regulated remains poorly understood. Here, we report unexpected roles of Dishevelled 2 (Dvl2) and interphase polo-like kinase 1 (Plk1) in primary cilia disassembly. We demonstrated that Dvl2 is phosphorylated at S143 and T224 in a manner that requires both non-canonical Wnt5a ligand and casein kinase 1 epsilon (CK1ɛ), and that this event is critical to interact with Plk1 in early stages of the cell cycle. The resulting Dvl2-Plk1 complex mediated Wnt5a-CK1ɛ-Dvl2-dependent primary cilia disassembly by stabilizing the HEF1 scaffold and activating its associated Aurora-A (AurA), a kinase crucially required for primary cilia disassembly. Thus, via the formation of the Dvl2-Plk1 complex, Plk1 plays an unanticipated role in primary cilia disassembly by linking Wnt5a-induced biochemical steps to HEF1/AurA-dependent cilia disassembly. This study may provide new insights into the mechanism underlying ciliary disassembly processes and various cilia-related disorders.
Collapse
Affiliation(s)
- Kyung Ho Lee
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Yoshikazu Johmura
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Li-Rong Yu
- Division of Systems Biology, Center for Proteomics, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA
| | - Jung-Eun Park
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Yuan Gao
- Division of Systems Biology, Center for Proteomics, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA
| | - Jeong K Bang
- Division of Magnetic Resonance, Korea Basic Science Institute, Chung-Buk, Republic of Korea
| | - Ming Zhou
- Laboratory of Proteomics and Analytical Technologies, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Timothy D Veenstra
- Laboratory of Proteomics and Analytical Technologies, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Bo Yeon Kim
- Chemical Biology Research Center and World Class Institute, Korea Research Institute of Bioscience and Biotechnology, Chung-Buk, Republic of Korea
| | - Kyung S Lee
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
25
|
Yu Y, Fay NC, Smoligovets AA, Wu HJ, Groves JT. Myosin IIA modulates T cell receptor transport and CasL phosphorylation during early immunological synapse formation. PLoS One 2012; 7:e30704. [PMID: 22347397 PMCID: PMC3275606 DOI: 10.1371/journal.pone.0030704] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 12/28/2011] [Indexed: 01/10/2023] Open
Abstract
Activation of T cell receptor (TCR) by antigens occurs in concert with an elaborate multi-scale spatial reorganization of proteins at the immunological synapse, the junction between a T cell and an antigen-presenting cell (APC). The directed movement of molecules, which intrinsically requires physical forces, is known to modulate biochemical signaling. It remains unclear, however, if mechanical forces exert any direct influence on the signaling cascades. We use T cells from AND transgenic mice expressing TCRs specific to the moth cytochrome c 88–103 peptide, and replace the APC with a synthetic supported lipid membrane. Through a series of high spatiotemporal molecular tracking studies in live T cells, we demonstrate that the molecular motor, non-muscle myosin IIA, transiently drives TCR transport during the first one to two minutes of immunological synapse formation. Myosin inhibition reduces calcium influx and colocalization of active ZAP-70 (zeta-chain associated protein kinase 70) with TCR, revealing an influence on signaling activity. More tellingly, its inhibition also significantly reduces phosphorylation of the mechanosensing protein CasL (Crk-associated substrate the lymphocyte type), raising the possibility of a direct mechanical mechanism of signal modulation involving CasL.
Collapse
Affiliation(s)
- Yan Yu
- Howard Hughes Medical Institute, Department of Chemistry, University of California, Berkeley, California, United States of America
- Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Nicole C. Fay
- Howard Hughes Medical Institute, Department of Chemistry, University of California, Berkeley, California, United States of America
- Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
- Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Alexander A. Smoligovets
- Howard Hughes Medical Institute, Department of Chemistry, University of California, Berkeley, California, United States of America
- Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
- Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Hung-Jen Wu
- Howard Hughes Medical Institute, Department of Chemistry, University of California, Berkeley, California, United States of America
- Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Jay T. Groves
- Howard Hughes Medical Institute, Department of Chemistry, University of California, Berkeley, California, United States of America
- Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- * E-mail:
| |
Collapse
|
26
|
Zhao M, Vuori K. The docking protein p130Cas regulates cell sensitivity to proteasome inhibition. BMC Biol 2011; 9:73. [PMID: 22034875 PMCID: PMC3215977 DOI: 10.1186/1741-7007-9-73] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Accepted: 10/28/2011] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND The focal adhesion protein p130Cas (Cas) activates multiple intracellular signaling pathways upon integrin or growth factor receptor ligation. Full-length Cas frequently promotes cell survival and migration, while its C-terminal fragment (Cas-CT) produced upon intracellular proteolysis is known to induce apoptosis in some circumstances. Here, we have studied the putative role of Cas in regulating cell survival and death pathways upon proteasome inhibition. RESULTS We found that Cas-/- mouse embryonic fibroblasts (MEFs), as well as empty vector-transfected Cas-/- MEFs (Cas-/- (EV)) are significantly resistant to cell death induced by proteasome inhibitors, such as MG132 and Bortezomib. As expected, wild-type MEFs (WT) and Cas-/- MEFs reconstituted with full-length Cas (Cas-FL) were sensitive to MG132- and Bortezomib-induced apoptosis that involved activation of a caspase-cascade, including Caspase-8. Cas-CT generation was not required for MG132-induced cell death, since expression of cleavage-resistant Cas mutants effectively increased sensitivity of Cas-/- MEFs to MG132. At the present time, the domains in Cas and the downstream pathways that are required for mediating cell death induced by proteasome inhibitors remain unknown. Interestingly, however, MG132 or Bortezomib treatment resulted in activation of autophagy in cells that lacked Cas, but not in cells that expressed Cas. Furthermore, autophagy was found to play a protective role in Cas-deficient cells, as inhibition of autophagy either by chemical or genetic means enhanced MG132-induced apoptosis in Cas-/- (EV) cells, but not in Cas-FL cells. Lack of Cas also contributed to resistance to the DNA-damaging agent Doxorubicin, which coincided with Doxorubicin-induced autophagy in Cas-/- (EV) cells. Thus, Cas may have a regulatory role in cell death signaling in response to multiple different stimuli. The mechanisms by which Cas inhibits induction of autophagy and affects cell death pathways are currently being investigated. CONCLUSION Our study demonstrates that Cas is required for apoptosis that is induced by proteasome inhibition, and potentially by other death stimuli. We additionally show that Cas may promote such apoptosis, at least partially, by inhibiting autophagy. This is the first demonstration of Cas being involved in the regulation of autophagy, adding to the previous findings by others linking focal adhesion components to the process of autophagy.
Collapse
Affiliation(s)
- Ming Zhao
- Cancer Center, Sanford-Burnham Medical Research Institute, 10901 N, Torrey Pines Road, La Jolla, CA 92037, USA
| | | |
Collapse
|
27
|
Bradbury P, Mahmassani M, Zhong J, Turner K, Paul A, Verrills NM, O'Neill GM. PP2A phosphatase suppresses function of the mesenchymal invasion regulator NEDD9. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2011; 1823:290-7. [PMID: 22061964 DOI: 10.1016/j.bbamcr.2011.10.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Revised: 10/18/2011] [Accepted: 10/18/2011] [Indexed: 12/26/2022]
Abstract
The mesenchymal mode of cancer cell invasion characterized by active adhesion turnover and a polarized actin cytoskeleton, is critically regulated by the adaptor protein NEDD9/HEF1/Cas-L. While it is known that NEDD9 is subject to extensive phosphorylation modification, the molecules that determine NEDD9 phosphorylation to stimulate adhesion turnover and mesenchymal cell morphologies are currently unknown. Earlier studies have suggested that the serine/threonine phosphatase PP2A regulates interconversion between a low molecular mass NEDD9 phosphoform and higher molecular mass phosphoforms. However, previous studies have used chemical inhibitors to block PP2A activity. In the present study we therefore aimed to specifically inhibit PP2A activity via siRNA and dominant negative approaches to investigate the effect of PP2A on interconversion between 115 kDa and 105 kDa NEDD9 and determine the functional consequence of PP2A activity for NEDD9 function. Strikingly, we find that while the phosphatase inhibitor Calyculin A indeed abrogates detachment-induced dephosphorylation of the 115 kDa NEDD9 phosphoform, PP2A depletion does not inhibit 115 kDa to 105 kDa interconversion. Our data suggest instead that PP2A targets discrete NEDD9 phosphorylation modifications separate to the events that mediate interconversion between the two forms. Functionally, PP2A depletion increases NEDD9 mediated cell spreading and mutation of S369 in the serine-rich region of NEDD9 to aspartate mimics this effect. Importantly, mutation of S369 to alanine abrogates the ability of dominant negative PP2A to increase NEDD9-mediated cell spreading. Collectively, our data reveal that the tumour suppressor PP2A may act via S369 to regulated NEDD9-mediated cell spreading.
Collapse
Affiliation(s)
- Peta Bradbury
- Children's Cancer Research Unit, Kids Research Institute, The Children's Hospital at Westmead, Westmead, NSW, 2145 Australia
| | | | | | | | | | | | | |
Collapse
|
28
|
Estradiol stabilizes the 105-kDa phospho-form of the adhesion docking protein NEDD9 and suppresses NEDD9-dependent cell spreading in breast cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2011; 1813:340-5. [DOI: 10.1016/j.bbamcr.2010.11.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Revised: 11/24/2010] [Accepted: 11/29/2010] [Indexed: 11/21/2022]
|
29
|
Singh MK, Izumchenko E, Klein-Szanto AJ, Egleston BL, Wolfson M, Golemis EA. Enhanced genetic instability and dasatinib sensitivity in mammary tumor cells lacking NEDD9. Cancer Res 2010; 70:8907-16. [PMID: 20940402 DOI: 10.1158/0008-5472.can-10-0353] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Elevated expression of the NEDD9/HEF1/Cas-L scaffolding protein promotes tumor cell invasion and metastasis in multiple cancer cell types. Conversely, generation of mammary tumors in the mouse mammary tumor virus (MMTV)-polyoma virus middle T (PyVT) genetic model is delayed by a Nedd9(-/-) genotype. These activities arise from the role of NEDD9 in assembling complexes and supporting activity of cancer signaling proteins, including FAK, Src, Shc, and AKT, and would support evaluation of NEDD9 expression as an unambiguous biomarker for tumor aggressiveness. However, we here show that despite the initial delay in tumor growth, cells derived from MMTV-PyVT;Nedd9(-/-) tumors are characteristically hyperaggressive versus MMTV-PyVT;Nedd9(+/+) cells in anchorage-independent growth, in growth on three-dimensional matrix produced by tumor-associated fibroblasts, and in formation of tumors after mammary orthotopic reinjection and of lung metastases after tail vein injection. This reversal suggests the specific selection of MMTV-PyVT;Nedd9(-/-) cells for growth in an in vivo microenvironment. Indeed, MMTV-PyVT;Nedd9(-/-) cells have increased cell cycle, centrosomal, and mitotic defects, phenotypes compatible with the increased selection of these cells for aggressive growth. Intriguingly, in spite of their aggressive phenotype, MMTV-PyVT;Nedd9(-/-) cells persistently have low levels of Src activation and are hypersensitive to the Src kinase inhibitor dasatinib. These studies identify NEDD9 as a complex modulator of different aspects of mammary tumor growth.
Collapse
Affiliation(s)
- Mahendra K Singh
- Departments of Developmental Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111, USA
| | | | | | | | | | | |
Collapse
|
30
|
Moore FE, Osmundson EC, Koblinski J, Pugacheva E, Golemis EA, Ray D, Kiyokawa H. The WW-HECT protein Smurf2 interacts with the Docking Protein NEDD9/HEF1 for Aurora A activation. Cell Div 2010; 5:22. [PMID: 20825672 PMCID: PMC2941750 DOI: 10.1186/1747-1028-5-22] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2010] [Accepted: 09/08/2010] [Indexed: 12/12/2022] Open
Abstract
The multi-functional adaptor protein NEDD9/HEF1/Cas-L regulates cell motility, invasion and cell cycle progression, and plays key roles in cancer progression and metastasis. NEDD9 is localized to the centrosome and is required for activation of Aurora A kinase in mitosis. Here we demonstrate that the HECT-WW protein Smurf2 physically associates with NEDD9 and is required for the stability of NEDD9 protein. Smurf2 depletion results in a marked decrease in NEDD9 protein levels, by facilitating polyubiquitination and proteasomal degradation of NEDD9. Conversely, forced overexpression of Smurf2 results in upregulation of endogenous NEDD9 protein, confirming the role for Smurf2 in NEDD9 stability. Cells with Smurf2 depletion fail to activate Aurora A at the G2/M boundary, leading to a marked delay in mitotic entry. These observations suggest that the stable complex of Smurf2 and NEDD9 is required for timely entry into mitosis via Aurora A activation.
Collapse
Affiliation(s)
- Finola E Moore
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Evan C Osmundson
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jennifer Koblinski
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Elena Pugacheva
- Department of Biochemistry, Mary Babb Randolph Cancer Center, West Virginia University; Morgantown, WV, USA
| | - Erica A Golemis
- Program in Molecular and Translational Medicine, Fox Chase Cancer Center, Philadelphia, PA USA
| | - Dipankar Ray
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Hiroaki Kiyokawa
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
31
|
Tikhmyanova N, Tulin AV, Roegiers F, Golemis EA. Dcas supports cell polarization and cell-cell adhesion complexes in development. PLoS One 2010; 5:e12369. [PMID: 20808771 PMCID: PMC2927436 DOI: 10.1371/journal.pone.0012369] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2010] [Accepted: 07/29/2010] [Indexed: 01/17/2023] Open
Abstract
Mammalian Cas proteins regulate cell migration, division and survival, and are often deregulated in cancer. However, the presence of four paralogous Cas family members in mammals (BCAR1/p130Cas, EFS/Sin1, NEDD9/HEF1/Cas-L, and CASS4/HEPL) has limited their analysis in development. We deleted the single Drosophila Cas gene, Dcas, to probe the developmental function of Dcas. Loss of Dcas had limited effect on embryonal development. However, we found that Dcas is an important modulator of the severity of the developmental phenotypes of mutations affecting integrins (If and mew) and their downstream effectors Fak56D or Src42A. Strikingly, embryonic lethal Fak56D-Dcas double mutant embryos had extensive cell polarity defects, including mislocalization and reduced expression of E-cadherin. Further genetic analysis established that loss of Dcas modified the embryonal lethal phenotypes of embryos with mutations in E-cadherin (Shg) or its signaling partners p120- and beta-catenin (Arm). These results support an important role for Cas proteins in cell-cell adhesion signaling in development.
Collapse
Affiliation(s)
- Nadezhda Tikhmyanova
- Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
- Department of Biochemistry, Drexel University Medical School, Philadelphia, Pennsylvania, United States of America
| | - Alexei V. Tulin
- Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Fabrice Roegiers
- Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Erica A. Golemis
- Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
32
|
Tikhmyanova N, Little JL, Golemis EA. CAS proteins in normal and pathological cell growth control. Cell Mol Life Sci 2010; 67:1025-48. [PMID: 19937461 PMCID: PMC2836406 DOI: 10.1007/s00018-009-0213-1] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Revised: 11/03/2009] [Accepted: 11/09/2009] [Indexed: 12/20/2022]
Abstract
Proteins of the CAS (Crk-associated substrate) family (BCAR1/p130Cas, NEDD9/HEF1/Cas-L, EFS/SIN and CASS4/HEPL) are integral players in normal and pathological cell biology. CAS proteins act as scaffolds to regulate protein complexes controlling migration and chemotaxis, apoptosis, cell cycle, and differentiation, and have more recently been linked to a role in progenitor cell function. Reflecting these complex functions, over-expression of CAS proteins has now been strongly linked to poor prognosis and increased metastasis in cancer, as well as resistance to first-line chemotherapeutics in multiple tumor types including breast and lung cancers, glioblastoma, and melanoma. Further, CAS proteins have also been linked to additional pathological conditions including inflammatory disorders, Alzheimer's and Parkinson's disease, as well as developmental defects. This review will explore the roles of the CAS proteins in normal and pathological states in the context of the many mechanistic insights into CAS protein function that have emerged in the past decade.
Collapse
Affiliation(s)
- Nadezhda Tikhmyanova
- Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA 19111 USA
- Department of Biochemistry, Drexel University Medical School, Philadelphia, PA 19102 USA
| | - Joy L. Little
- Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA 19111 USA
| | - Erica A. Golemis
- Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA 19111 USA
| |
Collapse
|
33
|
Barouki R, Coumoul X. Cell migration and metastasis markers as targets of environmental pollutants and the Aryl hydrocarbon receptor. Cell Adh Migr 2010; 4:72-6. [PMID: 20009531 DOI: 10.4161/cam.4.1.10313] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
During the last few years, several studies have pointed to a surprising link between environmental pollutants cellular signaling and important cell functions such as plasticity, adhesion and migration. This unexpected link could be related to endogenous functions of pollutants receptors that may be disrupted by environmental factors, which is supported by observations in invertebrate species. It could also reveal novel toxic end-points and mechanisms of those pollutants, such as teratogenesis and cancer metastasis that are highly relevant from a public health point of view. In the present short article, we will review our recent observations on the aryl hydrocarbon receptor and its new molecular and cellular targets. We identified HEF1/NEDD9/CAS-L, a multifunctional protein involved in integrin-based signaling as a transcriptional target of the receptor, and showed that its induction was critical for cell plasticity mediated by environmental pollutants. We will put our studies in perspective with other observations made by several groups.
Collapse
|
34
|
Abstract
Cilia are microtubule-based structures that protrude from the cell surface and function as sensors for mechanical and chemical environmental cues that regulate cellular differentiation or division. In metazoans, ciliary signaling is important during organismal development and in the homeostasis controls of adult tissues, with receptors for the Hedgehog, platelet derived growth factor (PDGF), Wnt, and other signaling cascades arrayed and active along the ciliary membrane. In normal cells, cilia are dynamically regulated during cell cycle progression: present in G0 and G1 cells, and usually in S/G2 cells, but almost invariably resorbed before mitotic entry, to reappear post-cytokinesis. This periodic resorption and reassembly of cilia, specified by the intrinsic cell cycle the intrinsic cell cycle machinery, influences the susceptibility of cells to the influence of extrinsic signals with cilia-associated receptors. Pathogenic conditions of mammals associated with loss of or defects in ciliary integrity include a number of developmental disorders, cystic syndromes in adults, and some cancers. With the continuing expansion of the list of human diseases associated with ciliary abnormalities, the identification of the cellular mechanisms regulating ciliary growth and disassembly has become a topic of intense research interest. Although these mechanisms are far from being understood, a number of recent studies have begun to identify key regulatory factors that may begin to offer insight into disease pathogenesis and treatment. In this chapter we will discuss the current state of knowledge regarding cell cycle control of ciliary dynamics, and provide general methods that can be applied to investigate cell cycle-dependent ciliary growth and disassembly.
Collapse
|
35
|
Izumchenko E, Singh MK, Plotnikova OV, Tikhmyanova N, Little JL, Serebriiskii IG, Seo S, Kurokawa M, Egleston BL, Klein-Szanto A, Pugacheva EN, Hardy RR, Wolfson M, Connolly DC, Golemis EA. NEDD9 promotes oncogenic signaling in mammary tumor development. Cancer Res 2009; 69:7198-206. [PMID: 19738060 DOI: 10.1158/0008-5472.can-09-0795] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In the past 3 years, altered expression of the HEF1/CAS-L/NEDD9 scaffolding protein has emerged as contributing to cancer metastasis in multiple cancer types. However, whereas some studies have identified elevated NEDD9 expression as prometastatic, other work has suggested a negative role in tumor progression. We here show that the Nedd9-null genetic background significantly limits mammary tumor initiation in the MMTV-polyoma virus middle T genetic model. Action of NEDD9 is tumor cell intrinsic, with immune cell infiltration, stroma, and angiogenesis unaffected. The majority of the late-appearing mammary tumors of MMTV-polyoma virus middle T;Nedd9(-/-) mice are characterized by depressed activation of proteins including AKT, Src, FAK, and extracellular signal-regulated kinase, emphasizing an important role of NEDD9 as a scaffolding protein for these prooncogenic proteins. Analysis of cells derived from primary Nedd9(+/+) and Nedd9(-/-) tumors showed persistently reduced FAK activation, attachment, and migration, consistent with a role for NEDD9 activation of FAK in promoting tumor aggressiveness. This study provides the first in vivo evidence of a role for NEDD9 in breast cancer progression and suggests that NEDD9 expression may provide a biomarker for tumor aggressiveness.
Collapse
Affiliation(s)
- Eugene Izumchenko
- Program in Molecular and Translational Medicine, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Vogel T, Ahrens S, Büttner N, Krieglstein K. Transforming growth factor beta promotes neuronal cell fate of mouse cortical and hippocampal progenitors in vitro and in vivo: identification of Nedd9 as an essential signaling component. ACTA ACUST UNITED AC 2009; 20:661-71. [PMID: 19587023 PMCID: PMC2820705 DOI: 10.1093/cercor/bhp134] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Transforming Growth Factor β (Tgfβ) and associated signaling effectors are expressed in the forebrain, but little is known about the role of this multifunctional cytokine during forebrain development. Using hippocampal and cortical primary cell cultures of developing mouse brains, this study identified Tgfβ-regulated genes not only associated with cell cycle exit of progenitors but also with adoption of neuronal cell fate. Accordingly, we observed not only an antimitotic effect of Tgfβ on progenitors but also an increased expression of neuronal markers in Tgfβ treated cultures. This effect was dependent upon Smad4. Furthermore, in vivo loss-of-function analyses using Tgfβ2−/−/Tgfβ3−/− double mutant mice showed the opposite effect of increased cell proliferation and fewer neurons in the cerebral cortex and hippocampus. Gata2, Runx1, and Nedd9 were candidate genes regulated by Tgfβ and known to be involved in developmental processes of neuronal progenitors. Using siRNA-mediated knockdown, we identified Nedd9 as an essential signaling component for the Tgfβ-dependent increase in neuronal cell fate. Expression of this scaffolding protein, which is mainly described as a signaling molecule of the β1-integrin pathway, was not only induced after Tgfβ treatment but was also associated with morphological changes of the Nestin-positive progenitor pool observed upon exposure to Tgfβ.
Collapse
Affiliation(s)
- Tanja Vogel
- Department of Neuroanatomy, Centre of Anatomy, Georg-August-University, 37075 Goettingen, Germany
| | | | | | | |
Collapse
|
37
|
Hivert V, Pierre J, Raingeaud J. Phosphorylation of human enhancer of filamentation (HEF1) on serine 369 induces its proteasomal degradation. Biochem Pharmacol 2009; 78:1017-25. [PMID: 19539609 DOI: 10.1016/j.bcp.2009.06.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2009] [Revised: 05/29/2009] [Accepted: 06/05/2009] [Indexed: 11/29/2022]
Abstract
Human enhancer of filamentation 1 (HEF1) is a multi-domain docking protein of the p130 Cas family. HEF1 is present at focal adhesions and is involved in integrin signalling mediating cytoskeleton reorganization associated with cell migration, adhesion or apoptosis. HEF1 functions are regulated in part by phosphorylation on tyrosine residues. HEF1 is also phosphorylated on serines/threonines leading to two isoforms refered to as p105 and p115. In most cases, the serine/threonine kinase(s) responsible for HEF1 phosphorylation have not been identified. In the present study, we have investigated HEF1 ser/thr phosphorylation. In the HCT-116 cell line transiently overexpressing Flag-HEF1 we showed that Hesperadin, a synthetic indolinone displaying antiproliferative effect and described as an inhibitor of various kinases including Aurora-B, prevented HEF1 phosphorylation induced by the ser/thr phosphatase PP2A inhibitor: okadaic acid (OA). In addition we showed that conversion of endogenous HEF1 p105 to p115 in HaCaT cells was prevented upon treatment with Hesperadin, resulting in accumulation of p105HEF1. We also identified serine 369 as the target site of phosphorylation by this Hesperadin-inhibited kinase in HCT-116. Finally, we provide evidence that phosphorylation on serine 369 but not phosphorylation on serine 296, triggers HEF1 degradation by the proteasomal machinery. These data suggest that conversion of p105 to p115 results from a ser-369-dependent phosphorylation mediated by an Hesperadin-sensitive kinase and regulates the half-life of HEF1.
Collapse
Affiliation(s)
- Virginie Hivert
- INSERM U749, Université Paris-sud 11, Faculté de Pharmacie, 5 rue JB Clement, 92296 Chatenay-Malabry, France
| | | | | |
Collapse
|
38
|
Aquino JB, Marmigère F, Lallemend F, Lundgren TK, Villar MJ, Wegner M, Ernfors P. Differential expression and dynamic changes of murine NEDD9 in progenitor cells of diverse tissues. Gene Expr Patterns 2008; 8:217-26. [DOI: 10.1016/j.gep.2008.01.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2007] [Revised: 01/11/2008] [Accepted: 01/11/2008] [Indexed: 11/30/2022]
|
39
|
Kim W, Seok Kang Y, Soo Kim J, Shin NY, Hanks SK, Song WK. The integrin-coupled signaling adaptor p130Cas suppresses Smad3 function in transforming growth factor-beta signaling. Mol Biol Cell 2008; 19:2135-46. [PMID: 18321991 DOI: 10.1091/mbc.e07-10-0991] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Reciprocal cooperative signaling by integrins and growth factor receptors at G1 phase during cell cycle progression is well documented. By contrast, little is known about the cross-talk between integrin and transforming growth factor (TGF)-beta signaling. Here, we show that integrin signaling counteracts the inhibitory effects of TGF-beta on cell growth and that this effect is mediated by p130Cas (Crk-associated substrate, 130 kDa). Adhesion to fibronectin or laminin reduces TGF-beta-induced Smad3 phosphorylation and thus inhibits TGF-beta-mediated growth arrest; loss of p130Cas abrogates these effects. Loss and gain of function studies demonstrated that, once tyrosine-phosphorylated via integrin signaling, p130Cas binds to Smad3 and reduces phosphorylation of Smad3. That in turn leads to inhibition of p15 and p21 expression and facilitation of cell cycle progression. Thus, p130Cas-mediated control of TGF-beta/Smad signaling may provide an additional clue to the mechanism underlying resistance to TGF-beta-induced growth inhibition.
Collapse
Affiliation(s)
- Wook Kim
- Cell Dynamics Research Center and Bioimaging Research Center, Gwangju Institute of Science and Technology, Gwangju 500-712, Korea
| | | | | | | | | | | |
Collapse
|
40
|
Abstract
Transforming growth factor-beta (TGF-beta) represents a large family of growth and differentiation factors that mobilize complex signaling networks to regulate cellular differentiation, proliferation, motility, adhesion, and apoptosis. TGF-beta signaling is tightly regulated by multiple complex mechanisms, and its deregulation plays a key role in the progression of many forms of cancer. Upon ligand binding, TGF-beta signals are transduced by Smad proteins, which in turn are tightly dependent on modulation by adaptor proteins such as embryonic liver fodrin, Smad anchor for receptor activation, filamin, and crkl. A further layer of regulation is imposed by ubiquitin-mediated targeting and proteasomal degradation of specific components of the TGF-beta signaling pathway. This review focuses on the ubiquitinators that regulate TGF-beta signaling and the association of these ubiquitin ligases with various forms of cancer. Delineating the role of ubiquitinators in the TGF-beta signaling pathway could yield powerful novel therapeutic targets for designing new cancer treatments.
Collapse
Affiliation(s)
- Eric Glasgow
- Laboratory of Cancer Genetics, Digestive Diseases, and GI Developmental Biology, Department of Surgery, Medicine and Lombardi Cancer Center, Georgetown University Medical Center, Washington, DC 20007, USA.
| | | |
Collapse
|
41
|
Singh MK, Cowell L, Seo S, O’Neill GM, Golemis EA. Molecular basis for HEF1/NEDD9/Cas-L action as a multifunctional co-ordinator of invasion, apoptosis and cell cycle. Cell Biochem Biophys 2007; 48:54-72. [PMID: 17703068 PMCID: PMC1976382 DOI: 10.1007/s12013-007-0036-3] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2007] [Revised: 04/11/2007] [Accepted: 11/30/1999] [Indexed: 10/23/2022]
Abstract
Upregulation of the scaffolding protein HEF1, also known as NEDD9 and Cas-L, has recently been identified as a pro-metastatic stimulus in a number of different solid tumors, and has also been strongly associated with pathogenesis of BCR-Abl-dependent tumors. As the evidence mounts for HEF1/NEDD9/Cas-L as a key player in metastatic cancer, it is timely to review the molecular regulation of HEF1/NEDD9/Cas-L. Most of the mortality associated with cancer arises from uncontrolled metastases, thus a better understanding of the properties of proteins specifically associated with promotion of this process may yield insights that improve cancer diagnosis and treatment. In this review, we summarize the extensive literature regarding HEF1/NEDD9/Cas-L expression and function in signaling relevant to cell attachment, migration, invasion, cell cycle, apoptosis, and oncogenic signal transduction. The complex function of HEF1/NEDD9/Cas-L revealed by this analysis leads us to propose a model in which alleviation of cell cycle checkpoints and acquired resistance to apoptosis is permissive for a HEF1/NEDD9/Cas-L-promoted pro-metastatic phenotype.
Collapse
Affiliation(s)
- Mahendra K. Singh
- Division of Basic Science, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Lauren Cowell
- Oncology Research Unit, The Children’s Hospital at Westmead, NSW, Australia
- Discipline of Paediatrics and Child Health, The University of Sydney, NSW, Australia
| | - Sachiko Seo
- Department of Hematology and Oncology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Geraldine M. O’Neill
- Oncology Research Unit, The Children’s Hospital at Westmead, NSW, Australia
- Discipline of Paediatrics and Child Health, The University of Sydney, NSW, Australia
| | - Erica A. Golemis
- Division of Basic Science, Fox Chase Cancer Center, Philadelphia, PA, USA
- corresponding author: Erica A. Golemis, Fox Chase Cancer Center, 333 Cottman Ave. Philadelphia, PA 19111 USA, Phone: 215-728-2860, FAX: 215-728-3616,
| |
Collapse
|
42
|
Abstract
The transforming growth factorbeta (TGFbeta) superfamily regulates a broad spectrum of biological responses throughout embryonic development and adult life, including cell proliferation and differentiation, epithelial-to-mesenchymal transition, apoptosis, and angiogenesis. TGFbeta members initiate signaling by bringing together a complex of serine/threonine kinase receptors that transmit signals through intracellular Smad proteins. Genetic alterations in numerous components of the TGFbeta signaling pathway have been associated with several human cancers. In addition, tight regulation of TGFbeta signaling is pivotal to the maintenance of homeostasis and the prevention of carcinogenesis. The ubiquitin/proteosome system is one mechanism by which cells regulate the expression and activity of effectors of the TGFbeta signaling cascade. Mounting evidence also suggests that disruption of the ubiquitin-dependent degradation of components of the TGFbeta pathway leads to the development and progression of cancer. Therefore, understanding how these two pathways intertwine will contribute to the advancement of our knowledge of cancer development.
Collapse
Affiliation(s)
- Luisa Izzi
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Liliana Attisano
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
43
|
Inamoto S, Iwata S, Inamoto T, Nomura S, Sasaki T, Urasaki Y, Hosono O, Kawasaki H, Tanaka H, Dang NH, Morimoto C. Crk-associated substrate lymphocyte type regulates transforming growth factor-beta signaling by inhibiting Smad6 and Smad7. Oncogene 2006; 26:893-904. [PMID: 16909115 DOI: 10.1038/sj.onc.1209848] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Crk-associated substrate lymphocyte type (Cas-L) is a 105 kDa docking protein with diverse functional properties, including regulation of cell division, proliferation, migration and adhesion. Cas-L is also involved in beta1 integrin- or antigen receptor-mediated signaling in B and T cells. In the present study, we demonstrate that Cas-L potentiates transforming growth factor-beta (TGF-beta) signaling pathway by interacting with Smad6 and Smad7. Immunoprecipitation experiments reveal that single domain deletion of full-length Cas-L completely abolishes its docking function with Smad6 and Smad7, suggesting that the natural structure of Cas-L is necessary for its association with Smad6 and Smad7. On the other hand, both N-terminal and C-terminal deletion mutants of Smad6 and Smad7 still retain their docking ability to Cas-L, suggesting that Smad6 and Smad7 possess several binding motifs to Cas-L. Moreover, Cas-L interaction with Mad-homology (MH)2 domain, but not with MH1 domain of Smad6 or Smad7, ameliorates TGF-beta-induced signaling pathway. Finally, depletion of Cas-L by small-interfering RNA oligo attenuates TGF-beta-induced growth inhibition of Huh-7 cells, with a concomitant reduction in phosphorylation of Smad2 and Smad3. These results strongly suggest that Cas-L is a potential regulator of TGF-beta signaling pathway.
Collapse
Affiliation(s)
- S Inamoto
- Division of Clinical Immunology, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Mao X, Kikani CK, Riojas RA, Langlais P, Wang L, Ramos FJ, Fang Q, Christ-Roberts CY, Hong JY, Kim RY, Liu F, Dong LQ. APPL1 binds to adiponectin receptors and mediates adiponectin signalling and function. Nat Cell Biol 2006; 8:516-23. [PMID: 16622416 DOI: 10.1038/ncb1404] [Citation(s) in RCA: 521] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2005] [Accepted: 03/17/2006] [Indexed: 02/06/2023]
Abstract
Adiponectin, also known as Acrp30, is an adipose tissue-derived hormone with anti-atherogenic, anti-diabetic and insulin sensitizing properties. Two seven-transmembrane domain-containing proteins, AdipoR1 and AdipoR2, have recently been identified as adiponectin receptors, yet signalling events downstream of these receptors remain poorly defined. By using the cytoplasmic domain of AdipoR1 as bait, we screened a yeast two-hybrid cDNA library derived from human fetal brain. This screening led to the identification of a phosphotyrosine binding domain and a pleckstrin homology domain-containing adaptor protein, APPL1 (adaptor protein containing pleckstrin homology domain, phosphotyrosine binding (PTB) domain and leucine zipper motif). APPL1 interacts with adiponectin receptors in mammalian cells and the interaction is stimulated by adiponectin. Overexpression of APPL1 increases, and suppression of APPL1 level reduces, adiponectin signalling and adiponectin-mediated downstream events (such as lipid oxidation, glucose uptake and the membrane translocation of glucose transport 4 (GLUT4)). Adiponectin stimulates the interaction between APPL1 and Rab5 (a small GTPase) interaction, leading to increased GLUT4 membrane translocation. APPL1 also acts as a critical regulator of the crosstalk between adiponectin signalling and insulin signalling pathways. These results demonstrate a key function for APPL1 in adiponectin signalling and provide a molecular mechanism for the insulin sensitizing function of adiponectin.
Collapse
Affiliation(s)
- Xuming Mao
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Saha T, Vardhini D, Tang Y, Katuri V, Jogunoori W, Volpe EA, Haines D, Sidawy A, Zhou X, Gallicano I, Schlegel R, Mishra B, Mishra L. RING finger-dependent ubiquitination by PRAJA is dependent on TGF-beta and potentially defines the functional status of the tumor suppressor ELF. Oncogene 2006; 25:693-705. [PMID: 16247473 DOI: 10.1038/sj.onc.1209123] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In gastrointestinal cells, biological signals for transforming growth factor-beta (TGF-beta) are transduced through transmembrane serine/threonine kinase receptors that signal to Smad proteins. Smad4, a tumor suppressor, is often mutated in human gastrointestinal cancers. The mechanism of Smad4 inactivation, however, remains uncertain and could be through E3-mediated ubiquitination of Smad4/adaptor protein complexes. Disruption of ELF (embryonic liver fodrin), a Smad4 adaptor protein, modulates TGF-beta signaling. We have found that PRAJA, a RING-H2 protein, interacts with ELF in a TGF-beta-dependent manner, with a fivefold increase of PRAJA expression and a subsequent decrease in ELF and Smad4 expression, in gastrointestinal cancer cell lines (P < 0.05). Strikingly, PRAJA manifests substantial E3-dependent ubiquitination of ELF and Smad3, but not Smad4. Delta-PRAJA, which has a deleted RING finger domain at the C terminus, abolishes ubiquitination of ELF. A stable cell line that overexpresses PRAJA exhibits low levels of ELF in comparison to a Delta-PRAJA stable cell line, where ELF expression is high compared to normal controls. The alteration of ELF and/or Smad4 expression and/or function in the TGF-beta signaling pathway may be induced by enhancement of ELF degradation, which is mediated by a high-level expression of PRAJA in gastrointestinal cancers. In hepatocytes, half-life (t(1/2)) and rate constant for degradation (k(D)) of ELF is 1.91 h and 21.72 min(-1) when coupled with ectopic expression of PRAJA in cells stimulated by TGF-beta, compared to PRAJA-transfected unstimulated cells (t(1/2) = 4.33 h and k(D) = 9.6 min(-1)). These studies reveal a mechanism for tumorigenesis whereby defects in adaptor proteins for Smads, such as ELF, can undergo degradation by PRAJA, through the ubiquitin-mediated pathway.
Collapse
Affiliation(s)
- T Saha
- Department of Surgical Sciences, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20007, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Pugacheva EN, Golemis EA. HEF1-aurora A interactions: points of dialog between the cell cycle and cell attachment signaling networks. Cell Cycle 2006; 5:384-91. [PMID: 16479169 PMCID: PMC2547350 DOI: 10.4161/cc.5.4.2439] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Regulated timing of cell division cycles, and geometrical precision in the planar orientation of cell division, are critical during organismal development and remain important for the maintenance of polarized structures in adults. Mounting evidence suggests that these processes are coordinated at the centrosome through the action of proteins that mediate both cell cycle and cell attachment. Our recent work identifying HEF1 as an activator of the Aurora A kinase suggests a novel hub for such integrated signaling. We suggest that defects in components of the machinery specifying the temporal and spatial integration of cell division may induce cancer and other diseases through pleiotropic effects on cell migration, proliferation, apoptosis, and genomic stability.
Collapse
Affiliation(s)
- Elena N. Pugacheva
- Division of Basic Science, Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA 19111
| | - Erica A. Golemis
- Division of Basic Science, Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA 19111
| |
Collapse
|
47
|
Zheng M, McKeown-Longo PJ. Cell adhesion regulates Ser/Thr phosphorylation and proteasomal degradation of HEF1. J Cell Sci 2005; 119:96-103. [PMID: 16352661 DOI: 10.1242/jcs.02712] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human enhancer of filamentation 1 (HEF1), a multifunctional docking protein of the Cas family, participates in integrin and growth factor signaling pathways that regulate global cellular processes including growth, motility and apoptosis. HEF1 consists of two isoforms, p105 and p115, the larger molecular weight form resulting from Ser/Thr phosphorylation of p105HEF1. The molecular mechanisms that regulate the interconversion of the two HEF1 species as well as the function of HEF1 Ser/Thr phosphorylation are unknown. Our study reveals that cell adhesion and detachment regulate the interconversion of the two HEF1 isoforms. Experiments using various inhibitors of cytoskeletal organization indicated that disruption of actin microfilaments but not intermediate filaments or microtubules resulted in a complete conversion of p115HEF1 to p105HEF1. The conversion of p115HEF1 to p105HEF1 was prevented by inhibition of protein phosphatase 2A (PP2A), suggesting that cytoskeletal regulation of PP2A activity controlled the dephosphorylation of p115HEF1. Degradation of endogenous HEF1 was dependent on proteasomes with the p115 species of HEF1 being preferentially targeted for turnover. Dephosphorylation of HEF1 by suspending cells or disrupting actin filaments protected HEF1 from degradation. These results suggest that the adhesion-dependent actin organization regulates proteasomal turnover of HEF1 through the activity of PP2A.
Collapse
Affiliation(s)
- Mingzhe Zheng
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, NY 12208, USA
| | | |
Collapse
|
48
|
Sun Y, Lowther W, Kato K, Bianco C, Kenney N, Strizzi L, Raafat D, Hirota M, Khan NI, Bargo S, Jones B, Salomon D, Callahan R. Notch4 intracellular domain binding to Smad3 and inhibition of the TGF-beta signaling. Oncogene 2005; 24:5365-74. [PMID: 16007227 DOI: 10.1038/sj.onc.1208528] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We present evidence that Notch4ICD attenuates TGF-beta signaling. Cells expressing the activated form of the Notch4 receptor (ICD4) were resistant to the growth-inhibitory effects of TGF-beta. Notch4ICD was found to bind to Smad2, Smad3 and Smad4 but with higher affinity to Smad3. Deletion analysis showed that binding of Smad3 to ICD4 was mediated by its MH2 domain and was not dependent on the presence of the RAM23 region in ICD4. Using two TGF-beta/Activin reporter luciferase assays, RT-PCR and Western blot analysis, we demonstrate that ICD4 and ICD4 deltaRAM23 inhibit Smad-binding element and 3TP luciferase reporter activity and PAI-1 gene expression. MCF-7 human breast cancer cells express Notch4ICD (ICD4) and are resistant to the growth-inhibitory effects of TGF-beta. Blockage of Notch4 processing to ICD4 by gamma-secretase inhibitor renders MCF-7 cells sensitive to growth inhibition by TGF-beta. The interplay between these two signaling pathways may be a significant determinant during mammary tumorigenesis.
Collapse
Affiliation(s)
- Youping Sun
- The Tumor Growth Factor Section, Mammary Biology and Tumorigenesis Laboratory, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Ray D, Terao Y, Nimbalkar D, Chu LH, Donzelli M, Tsutsui T, Zou X, Ghosh AK, Varga J, Draetta GF, Kiyokawa H. Transforming growth factor beta facilitates beta-TrCP-mediated degradation of Cdc25A in a Smad3-dependent manner. Mol Cell Biol 2005; 25:3338-47. [PMID: 15798217 PMCID: PMC1069597 DOI: 10.1128/mcb.25.8.3338-3347.2005] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Ubiquitin-dependent degradation of Cdc25A is a major mechanism for damage-induced S-phase checkpoint. Two ubiquitin ligases, the Skp1-cullin-beta-TrCP (SCFbeta-TrCP) complex and the anaphase-promoting complex (APCCdh1), are involved in Cdc25A degradation. Here we demonstrate that the transforming growth factor beta (TGF-beta)-Smad3 pathway promotes SCF(beta-TrCP)-mediated Cdc25A ubiquitination. Cells treated with TGF-beta, as well as cells transfected with Smad3 or a constitutively active type I TGF-beta receptor, exhibit increased ubiquitination and markedly shortened half-lives of Cdc25A. Furthermore, Cdc25A is stabilized in cells transfected with Smad3 small interfering RNA (siRNA) and cells from Smad3-null mice. TGF-beta-induced ubiquitination is associated with Cdc25A phosphorylation at the beta-TrCP docking site (DS82G motif) and physical association of Cdc25A with Smad3 and beta-TrCP. Cdc25A mutant proteins deficient in DS82G phosphorylation are resistant to TGF-beta-Smad3-induced degradation, whereas a Cdc25A mutant protein defective in APCCdh1 recognition undergoes efficient degradation. Smad3 siRNA inhibits beta-TrCP-Cdc25A interaction and Cdc25A degradation in response to TGF-beta. beta-TrCP2 siRNA also inhibits Smad3-induced Cdc25A degradation. In contrast, Cdh1 siRNA had no effect on Cdc25A down-regulation by Smad3. These data suggest that Smad3 plays a key role in the regulation of Cdc25A ubiquitination by SCFbeta-TrCP and that Cdc25A stabilization observed in various cancers could be associated with defects in the TGF-beta-Smad3 pathway.
Collapse
Affiliation(s)
- Dipankar Ray
- Department of Biochemistry and Molecular Genetics, University of Illinois College of Medicine, Chicago, IL 60607, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Iwata S, Souta-Kuribara A, Yamakawa A, Sasaki T, Shimizu T, Hosono O, Kawasaki H, Tanaka H, Dang NH, Watanabe T, Arima N, Morimoto C. HTLV-I Tax induces and associates with Crk-associated substrate lymphocyte type (Cas-L). Oncogene 2005; 24:1262-71. [PMID: 15592516 DOI: 10.1038/sj.onc.1208261] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Crk-associated substrate lymphocyte type (Cas-L) is a docking protein that is heavily tyrosine phosphorylated by the engagement of beta1 integrins in T cells. In the present study, we attempted to evaluate the role of Cas-L in the pathophysiology of adult T-cell leukemia (ATL). Examination of peripheral blood mononuclear cells from ATL patients as well as ATL-derived T cell lines showed an elevation of Cas-L in these cells. We showed that tyrosine phosphorylation as well as expression of Cas-L was markedly elevated through the induction of human T-lymphotropic virus type I (HTLV-I) Tax in JPX-9 cells, with these cells showing marked motile behavior on the ligands for integrins. We next performed yeast two-hybrid screening of cDNA library from an HTLV-I-transformed T cell line, which resulted in the identification of Tax as a putative binding partner for Cas-L. Co-precipitation experiments revealed that the serine-rich region of Cas-L might serve as the binding site with the highest affinity for Tax. Co-localization study showed that Tax and Cas-L partly merged in the cytoplasm. Finally, we showed that exogenous Cas-L inhibited Tax-mediated transactivation of nuclear factor kappaB (NF-kappaB), while Tax-independent activation of NF-kappaB remained intact, hence indicating that Cas-L might specifically regulate Tax-NF-kappaB pathway.
Collapse
Affiliation(s)
- Satoshi Iwata
- Division of Clinical Immunology, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|