37 results on '"Su Sien Ong"'
Search Results
2. Transcription factor ZNF148 is a negative regulator of human muscle differentiation
- Author
-
Jesse Bakke, William C. Wright, Anthony E. Zamora, Su Sien Ong, Yue-Ming Wang, Jessica D. Hoyer, Christopher T. Brewer, Paul G. Thomas, and Taosheng Chen
- Subjects
Medicine ,Science - Abstract
Abstract Muscle differentiation is a complex process in which muscle progenitor cells undergo determination and eventually cellular fusion. This process is heavily regulated by such master transcription factors as MYOD and members of the MEF2 family. Here, we show that the transcription factor ZNF148 plays a direct role in human muscle cell differentiation. Downregulation of ZNF148 drives the formation of a muscle phenotype with rapid expression of myosin heavy chain, even in proliferative conditions. This phenotype was most likely mediated by the robust and swift upregulation of MYOD and MEF2C.
- Published
- 2017
- Full Text
- View/download PDF
3. A phosphotyrosine switch regulates organic cation transporters
- Author
-
Jason A. Sprowl, Su Sien Ong, Alice A. Gibson, Shuiying Hu, Guoqing Du, Wenwei Lin, Lie Li, Shashank Bharill, Rachel A. Ness, Adrian Stecula, Steven M. Offer, Robert B. Diasio, Anne T. Nies, Matthias Schwab, Guido Cavaletti, Eberhard Schlatter, Giuliano Ciarimboli, Jan H. M. Schellens, Ehud Y. Isacoff, Andrej Sali, Taosheng Chen, Sharyn D. Baker, Alex Sparreboom, and Navjotsingh Pabla
- Subjects
Science - Abstract
Organic cation transporters are important drug transporters that influence therapeutic outcomes. Here, the authors find that these transporters are regulated by tyrosine phosphorylation and propose that tyrosine kinase inhibitors can influence drug transporter function through post-translational mechanisms.
- Published
- 2016
- Full Text
- View/download PDF
4. Cyclin-dependent kinase 4 phosphorylates and positively regulates PAX3-FOXO1 in human alveolar rhabdomyosarcoma cells.
- Author
-
Lingling Liu, Jing Wu, Su Sien Ong, and Taosheng Chen
- Subjects
Medicine ,Science - Abstract
Alveolar rhabdomyosarcoma (ARMS) is an aggressive childhood muscle sarcoma with a 5-year survival rate of less than 30%. More than 80% of ARMSs harbor a PAX3-FOXO1 fusion transcription factor. However, expression of PAX3-FOXO1 in muscle cells alone is not sufficient and requires the loss of function of Ink4a/ARF to promote malignant proliferation of muscle cells in vitro or initiate ARMS tumor formation in vivo. This prompted us to examine the signaling pathways required to activate the function of PAX3-FOXO1 and to explore the functional interaction between the Ink4a/ARF and PAX3-FOXO1 signaling pathways. Here we report that inhibition of cyclin-dependent kinase 4 (Cdk4) by fascaplysin (a small molecule selective inhibitor of Cdk4/cyclin D1 that we identified in a screen for compounds that inhibit PAX3-FOXO1) led to inhibition of the transcriptional activity of PAX3-FOXO1 in ARMS cell line Rh30. Consistent with this finding, activation of Cdk4 enhanced the activity of PAX3-FOXO1. In vitro kinase assays revealed that Cdk4 directly phosphorylated PAX3-FOXO1 at Ser(430). Whereas fascaplysin did not affect the protein level of PAX3-FOXO1, it did increase the cytoplasmic level of PAX3-FOXO1 in a portion of cells. Our findings indicate that Cdk4 phosphorylates and positively regulates PAX3-FOXO1 and suggest that inhibition of Cdk4 activity should be explored as a promising avenue for developing therapy for ARMS.
- Published
- 2013
- Full Text
- View/download PDF
5. GUItars: a GUI tool for analysis of high-throughput RNA interference screening data.
- Author
-
Asli N Goktug, Su Sien Ong, and Taosheng Chen
- Subjects
Medicine ,Science - Abstract
High-throughput RNA interference (RNAi) screening has become a widely used approach to elucidating gene functions. However, analysis and annotation of large data sets generated from these screens has been a challenge for researchers without a programming background. Over the years, numerous data analysis methods were produced for plate quality control and hit selection and implemented by a few open-access software packages. Recently, strictly standardized mean difference (SSMD) has become a widely used method for RNAi screening analysis mainly due to its better control of false negative and false positive rates and its ability to quantify RNAi effects with a statistical basis. We have developed GUItars to enable researchers without a programming background to use SSMD as both a plate quality and a hit selection metric to analyze large data sets.The software is accompanied by an intuitive graphical user interface for easy and rapid analysis workflow. SSMD analysis methods have been provided to the users along with traditionally-used z-score, normalized percent activity, and t-test methods for hit selection. GUItars is capable of analyzing large-scale data sets from screens with or without replicates. The software is designed to automatically generate and save numerous graphical outputs known to be among the most informative high-throughput data visualization tools capturing plate-wise and screen-wise performances. Graphical outputs are also written in HTML format for easy access, and a comprehensive summary of screening results is written into tab-delimited output files.With GUItars, we demonstrated robust SSMD-based analysis workflow on a 3840-gene small interfering RNA (siRNA) library and identified 200 siRNAs that increased and 150 siRNAs that decreased the assay activities with moderate to stronger effects. GUItars enables rapid analysis and illustration of data from large- or small-scale RNAi screens using SSMD and other traditional analysis methods. The software is freely available at http://sourceforge.net/projects/guitars/.
- Published
- 2012
- Full Text
- View/download PDF
6. Figure S3 from Regulation of OATP1B1 Function by Tyrosine Kinase–mediated Phosphorylation
- Author
-
Jason A. Sprowl, Shuiying Hu, Alex Sparreboom, Aviv Paz, Kevin M. Huang, Muhammad Erfan Uddin, Yan Jin, Taosheng Chen, Su Sien Ong, Jun Qu, Shichen Shen, James J. Petti, Alice A. Gibson, Kyle Z. Pasquariello, Mingqing Chen, and Elizabeth R. Hayden
- Abstract
A structural model of OATP1B1. The model was generated by Phyre2 and presented as a cartoon. Y640 and Y645 are highlighted in blue and the image was produced with PyMOL.
- Published
- 2023
- Full Text
- View/download PDF
7. Data from Regulation of OATP1B1 Function by Tyrosine Kinase–mediated Phosphorylation
- Author
-
Jason A. Sprowl, Shuiying Hu, Alex Sparreboom, Aviv Paz, Kevin M. Huang, Muhammad Erfan Uddin, Yan Jin, Taosheng Chen, Su Sien Ong, Jun Qu, Shichen Shen, James J. Petti, Alice A. Gibson, Kyle Z. Pasquariello, Mingqing Chen, and Elizabeth R. Hayden
- Abstract
Purpose:OATP1B1 (SLCO1B1) is the most abundant and pharmacologically relevant uptake transporter in the liver and a key mediator of xenobiotic clearance. However, the regulatory mechanisms that determine OATP1B1 activity remain uncertain, and as a result, unexpected drug–drug interactions involving OATP1B1 substrates continue to be reported, including several involving tyrosine kinase inhibitors (TKI).Experimental Design:OATP1B1-mediated activity in overexpressing HEK293 cells and hepatocytes was assessed in the presence of FDA-approved TKIs, while rosuvastatin pharmacokinetics in the presence of an OATP1B1 inhibiting TKI were measured in vivo. Tyrosine phosphorylation of OATP1B1 was determined by LC/MS-MS–based proteomics and transport function was measured following exposure to siRNAs targeting 779 different kinases.Results:Twenty-nine of 46 FDA-approved TKIs studied significantly inhibit OATP1B1 function. Inhibition of OATP1B1 by TKIs, such as nilotinib, is predominantly noncompetitive, can increase systemic concentrations of rosuvastatin in vivo, and is associated with reduced phosphorylation of OATP1B1 at tyrosine residue 645. Using genetic screens and functional validation studies, the Src kinase LYN was identified as a potential regulator of OATP1B1 activity that is highly sensitive to inhibition by various TKIs at clinically relevant concentrations.Conclusions:A novel kinase-dependent posttranslational mechanism of OATP1B1 activation was identified and interference with this process by TKIs can influence the elimination of a broad range of xenobiotic substrates.
- Published
- 2023
- Full Text
- View/download PDF
8. Table S2 from Regulation of OATP1B1 Function by Tyrosine Kinase–mediated Phosphorylation
- Author
-
Jason A. Sprowl, Shuiying Hu, Alex Sparreboom, Aviv Paz, Kevin M. Huang, Muhammad Erfan Uddin, Yan Jin, Taosheng Chen, Su Sien Ong, Jun Qu, Shichen Shen, James J. Petti, Alice A. Gibson, Kyle Z. Pasquariello, Mingqing Chen, and Elizabeth R. Hayden
- Abstract
Cellular viability and uptake of 8FcA by OATP1B1 following reverse transfection with 25 nM siRNA for various protein kinases. Percent uptake was determined relative to OATP1B1 uptake with control siRNA.
- Published
- 2023
- Full Text
- View/download PDF
9. Data from Targeting Histone Demethylases in MYC-Driven Neuroblastomas with Ciclopirox
- Author
-
Andrew M. Davidoff, Taosheng Chen, R. Kiplin Guy, Stephen White, Douglas R. Green, Junmin Peng, Chunxu Qu, Yong Cheng, Ashutosh Mishra, Jie J. Zheng, Zhenmei Li, Yinan Wu, Anang Shelat, Sourav Das, Brandon Young, Vincent A. Boyd, Su Sien Ong, Yingdi Wang, Ruoning Wang, Amit C. Nathwani, Pollyanna Goh, Ju Bao, Wenwei Lin, Jonathan Low, Jesse Davidson, Junfang Zhou, Rodrigo B. Interiano, Alaa M. AlTahan, Dongli Hu, Sandra Milasta, and Jun Yang
- Abstract
Histone lysine demethylases facilitate the activity of oncogenic transcription factors, including possibly MYC. Here we show that multiple histone demethylases influence the viability and poor prognosis of neuroblastoma cells, where MYC is often overexpressed. We also identified the approved small-molecule antifungal agent ciclopirox as a novel pan-histone demethylase inhibitor. Ciclopirox targeted several histone demethylases, including KDM4B implicated in MYC function. Accordingly, ciclopirox inhibited Myc signaling in parallel with mitochondrial oxidative phosphorylation, resulting in suppression of neuroblastoma cell viability and inhibition of tumor growth associated with an induction of differentiation. Our findings provide new insights into epigenetic regulation of MYC function and suggest a novel pharmacologic basis to target histone demethylases as an indirect MYC-targeting approach for cancer therapy. Cancer Res; 77(17); 4626–38. ©2017 AACR.
- Published
- 2023
- Full Text
- View/download PDF
10. Figure S1-S6, Table S1-S2 from Targeting Histone Demethylases in MYC-Driven Neuroblastomas with Ciclopirox
- Author
-
Andrew M. Davidoff, Taosheng Chen, R. Kiplin Guy, Stephen White, Douglas R. Green, Junmin Peng, Chunxu Qu, Yong Cheng, Ashutosh Mishra, Jie J. Zheng, Zhenmei Li, Yinan Wu, Anang Shelat, Sourav Das, Brandon Young, Vincent A. Boyd, Su Sien Ong, Yingdi Wang, Ruoning Wang, Amit C. Nathwani, Pollyanna Goh, Ju Bao, Wenwei Lin, Jonathan Low, Jesse Davidson, Junfang Zhou, Rodrigo B. Interiano, Alaa M. AlTahan, Dongli Hu, Sandra Milasta, and Jun Yang
- Abstract
Figure S1 shows siRNA screening in neuroblastoma cells. Figure S2 shows the dentification of ciclopirox that targets KDM4B. Figure S3 shows the CPX effect on other histone methyl marks. Figure S4 shows that CPX targets the Myc pathway. Figure S5 shows that CPX inhibits oxidative phosphorylation. Figure S6 shows the CPX effect on BE2C xenografts with KDM4B knockdown. Table S1 shows the sequences of the focused KDM siRNA library. Table S2 shows the primers for real time PCR.
- Published
- 2023
- Full Text
- View/download PDF
11. Table S2 from Multikinase Inhibitors Induce Cutaneous Toxicity through OAT6-Mediated Uptake and MAP3K7-Driven Cell Death
- Author
-
Sharyn D. Baker, Alex Sparreboom, Michael L. Maitland, Hiroto Inaba, Peter Vogel, Taosheng Chen, Su Sien Ong, Gerard P. Mascara, Guoqing Du, Shelley J. Orwick, Aksana Vasilyeva, Shuiying Hu, Alice A. Gibson, and Eric I. Zimmerman
- Abstract
Cellular uptake of sorafenib following transporter-directed siRNA screen.
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary References from Multikinase Inhibitors Induce Cutaneous Toxicity through OAT6-Mediated Uptake and MAP3K7-Driven Cell Death
- Author
-
Sharyn D. Baker, Alex Sparreboom, Michael L. Maitland, Hiroto Inaba, Peter Vogel, Taosheng Chen, Su Sien Ong, Gerard P. Mascara, Guoqing Du, Shelley J. Orwick, Aksana Vasilyeva, Shuiying Hu, Alice A. Gibson, and Eric I. Zimmerman
- Abstract
Supplementary References used.
- Published
- 2023
- Full Text
- View/download PDF
13. Figure S2 from Multikinase Inhibitors Induce Cutaneous Toxicity through OAT6-Mediated Uptake and MAP3K7-Driven Cell Death
- Author
-
Sharyn D. Baker, Alex Sparreboom, Michael L. Maitland, Hiroto Inaba, Peter Vogel, Taosheng Chen, Su Sien Ong, Gerard P. Mascara, Guoqing Du, Shelley J. Orwick, Aksana Vasilyeva, Shuiying Hu, Alice A. Gibson, and Eric I. Zimmerman
- Abstract
Identification of OAT-dependent sorafenib transport in human primary keratinocytes
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Figure Legends from Multikinase Inhibitors Induce Cutaneous Toxicity through OAT6-Mediated Uptake and MAP3K7-Driven Cell Death
- Author
-
Sharyn D. Baker, Alex Sparreboom, Michael L. Maitland, Hiroto Inaba, Peter Vogel, Taosheng Chen, Su Sien Ong, Gerard P. Mascara, Guoqing Du, Shelley J. Orwick, Aksana Vasilyeva, Shuiying Hu, Alice A. Gibson, and Eric I. Zimmerman
- Abstract
Legends for Supplementary Figures S1-S4.
- Published
- 2023
- Full Text
- View/download PDF
15. Data from Multikinase Inhibitors Induce Cutaneous Toxicity through OAT6-Mediated Uptake and MAP3K7-Driven Cell Death
- Author
-
Sharyn D. Baker, Alex Sparreboom, Michael L. Maitland, Hiroto Inaba, Peter Vogel, Taosheng Chen, Su Sien Ong, Gerard P. Mascara, Guoqing Du, Shelley J. Orwick, Aksana Vasilyeva, Shuiying Hu, Alice A. Gibson, and Eric I. Zimmerman
- Abstract
The use of multikinase inhibitors (MKI) in oncology, such as sorafenib, is associated with a cutaneous adverse event called hand–foot skin reaction (HFSR), in which sites of pressure or friction become inflamed and painful, thus significantly impacting quality of life. The pathogenesis of MKI-induced HFSR is unknown, and the only available treatment options involve dose reduction or discontinuation of therapy, which have negative effects on primary disease management. To investigate the underlying mechanisms by which sorafenib promotes keratinocyte cytotoxicity and subsequent HFSR induction, we performed a transporter-directed RNAi screen in human epidermal keratinocytes and identified SLC22A20 (OAT6) as an uptake carrier of sorafenib. Further investigations into the intracellular mechanism of sorafenib activity through in situ kinome profiling identified the mitogen-activated protein kinase MAP3K7 (TAK1) as a target of sorafenib that induces cell death. Finally, we demonstrate that sorafenib induced keratinocyte injury in vivo and that this effect could be reversed by cotreatment with the OAT6 inhibitor probenecid. Collectively, our findings reveal a novel pathway that regulates the entry of some MKIs into keratinocytes and explains the basis underlying sorafenib-induced skin toxicity, with important implications for the therapeutic management of HFSR. Cancer Res; 76(1); 117–26. ©2015 AACR.
- Published
- 2023
- Full Text
- View/download PDF
16. A kinome-wide screen identifies a CDKL5-SOX9 regulatory axis in epithelial cell death and kidney injury
- Author
-
Katalin Susztak, Taosheng Chen, John W. Christman, Yuntao Bai, Stuart Cobb, Ming Poi, Harpreet Singh, Sharyn D. Baker, Rajgopal Govindarajan, Rachel E. Cianciolo, Jiyoung Kim, Sangwoon Chung, Radhika Raj, Alex Sparreboom, Laura A. Jayne, Navjotsingh Pabla, Shreshtha Rojesh, David S. Gardner, Ralph D. Hector, Moray J. Campbell, Mei Ji He Ho Feng, Avinash K. Persaud, and Su Sien Ong
- Subjects
0301 basic medicine ,Keratinocytes ,Male ,Programmed cell death ,Science ,Green Fluorescent Proteins ,CDKL5 ,General Physics and Astronomy ,Protein Serine-Threonine Kinases ,Kidney ,General Biochemistry, Genetics and Molecular Biology ,Nephrotoxicity ,03 medical and health sciences ,Mice ,0302 clinical medicine ,RNA interference ,medicine ,Animals ,Humans ,Kinome ,RNA, Small Interfering ,lcsh:Science ,Mice, Knockout ,Neurons ,Multidisciplinary ,Cell Death ,Kinase ,business.industry ,Acute kidney injury ,Kidney metabolism ,Epithelial Cells ,SOX9 Transcription Factor ,General Chemistry ,Acute Kidney Injury ,medicine.disease ,Mice, Inbred C57BL ,030104 developmental biology ,Kidney Tubules ,030220 oncology & carcinogenesis ,Cancer research ,lcsh:Q ,Female ,RNA Interference ,business - Abstract
Renal tubular epithelial cells (RTECs) perform the essential function of maintaining the constancy of body fluid composition and volume. Toxic, inflammatory, or hypoxic-insults to RTECs can cause systemic fluid imbalance, electrolyte abnormalities and metabolic waste accumulation- manifesting as acute kidney injury (AKI), a common disorder associated with adverse long-term sequelae and high mortality. Here we report the results of a kinome-wide RNAi screen for cellular pathways involved in AKI-associated RTEC-dysfunction and cell death. Our screen and validation studies reveal an essential role of Cdkl5-kinase in RTEC cell death. In mouse models, genetic or pharmacological Cdkl5 inhibition mitigates nephrotoxic and ischemia-associated AKI. We propose that Cdkl5 is a stress-responsive kinase that promotes renal injury in part through phosphorylation-dependent suppression of pro-survival transcription regulator Sox9. These findings reveal a surprising non-neuronal function of Cdkl5, identify a pathogenic Cdkl5-Sox9 axis in epithelial cell-death, and support CDKL5 antagonism as a therapeutic approach for AKI.
- Published
- 2020
- Full Text
- View/download PDF
17. Regulation of OATP1B1 Function by Tyrosine Kinase-mediated Phosphorylation
- Author
-
Muhammad Erfan Uddin, Kyle Z. Pasquariello, Alice A. Gibson, Elizabeth R. Hayden, Shuiying Hu, Mingqing Chen, Alex Sparreboom, Yan Jin, Su Sien Ong, Kevin M. Huang, Aviv Paz, Jason A. Sprowl, James J. Petti, Shichen Shen, Taosheng Chen, and Jun Qu
- Subjects
Cancer Research ,Chemistry ,Kinase ,Liver-Specific Organic Anion Transporter 1 ,Tyrosine phosphorylation ,Pharmacology ,Protein-Tyrosine Kinases ,Article ,respiratory tract diseases ,chemistry.chemical_compound ,Mice ,HEK293 Cells ,Oncology ,Nilotinib ,LYN ,medicine ,Hepatocytes ,Phosphorylation ,Animals ,Humans ,Tyrosine ,Tyrosine kinase ,medicine.drug ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Purpose: OATP1B1 (SLCO1B1) is the most abundant and pharmacologically relevant uptake transporter in the liver and a key mediator of xenobiotic clearance. However, the regulatory mechanisms that determine OATP1B1 activity remain uncertain, and as a result, unexpected drug–drug interactions involving OATP1B1 substrates continue to be reported, including several involving tyrosine kinase inhibitors (TKI). Experimental Design: OATP1B1-mediated activity in overexpressing HEK293 cells and hepatocytes was assessed in the presence of FDA-approved TKIs, while rosuvastatin pharmacokinetics in the presence of an OATP1B1 inhibiting TKI were measured in vivo. Tyrosine phosphorylation of OATP1B1 was determined by LC/MS-MS–based proteomics and transport function was measured following exposure to siRNAs targeting 779 different kinases. Results: Twenty-nine of 46 FDA-approved TKIs studied significantly inhibit OATP1B1 function. Inhibition of OATP1B1 by TKIs, such as nilotinib, is predominantly noncompetitive, can increase systemic concentrations of rosuvastatin in vivo, and is associated with reduced phosphorylation of OATP1B1 at tyrosine residue 645. Using genetic screens and functional validation studies, the Src kinase LYN was identified as a potential regulator of OATP1B1 activity that is highly sensitive to inhibition by various TKIs at clinically relevant concentrations. Conclusions: A novel kinase-dependent posttranslational mechanism of OATP1B1 activation was identified and interference with this process by TKIs can influence the elimination of a broad range of xenobiotic substrates.
- Published
- 2021
18. Transcription factor ZNF148 is a negative regulator of human muscle differentiation
- Author
-
Jessica D. Hoyer, Christopher Trent Brewer, William C. Wright, Yue-Ming Wang, Su Sien Ong, Jesse Bakke, Taosheng Chen, Paul G. Thomas, and Anthony E. Zamora
- Subjects
0301 basic medicine ,Mef2 ,Cellular differentiation ,Science ,Biology ,MyoD ,Muscle Development ,Article ,Cell Line ,Myoblasts ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Myosin ,Humans ,MEF2C ,RNA, Small Interfering ,Muscle, Skeletal ,Transcription factor ,Cell Line, Transformed ,MyoD Protein ,Multidisciplinary ,Myogenesis ,Gene Expression Profiling ,Cell Differentiation ,Cell biology ,DNA-Binding Proteins ,030104 developmental biology ,Phenotype ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,Gene Knockdown Techniques ,Immunology ,Medicine ,Transcription Factors - Abstract
Muscle differentiation is a complex process in which muscle progenitor cells undergo determination and eventually cellular fusion. This process is heavily regulated by such master transcription factors as MYOD and members of the MEF2 family. Here, we show that the transcription factor ZNF148 plays a direct role in human muscle cell differentiation. Downregulation of ZNF148 drives the formation of a muscle phenotype with rapid expression of myosin heavy chain, even in proliferative conditions. This phenotype was most likely mediated by the robust and swift upregulation of MYOD and MEF2C.
- Published
- 2017
19. Multikinase Inhibitors Induce Cutaneous Toxicity through OAT6-Mediated Uptake and MAP3K7-Driven Cell Death
- Author
-
Sharyn D. Baker, Taosheng Chen, Shuiying Hu, Gerard P. Mascara, Guoqing Du, Su Sien Ong, Peter Vogel, Shelley Orwick, Eric I. Zimmerman, Aksana Vasilyeva, Alice A. Gibson, Michael L. Maitland, Alex Sparreboom, and Hiroto Inaba
- Subjects
Keratinocytes ,Niacinamide ,0301 basic medicine ,Sorafenib ,Cancer Research ,Programmed cell death ,Organic Anion Transporters ,Transfection ,urologic and male genital diseases ,MAP3K7 ,Skin Diseases ,Article ,Nuclear Receptor Subfamily 2, Group C, Member 2 ,Mice ,Random Allocation ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Kinome ,Protein kinase A ,Protein Kinase Inhibitors ,neoplasms ,Skin ,Cell Death ,business.industry ,Phenylurea Compounds ,Hep G2 Cells ,MAP Kinase Kinase Kinases ,digestive system diseases ,female genital diseases and pregnancy complications ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,Female ,Keratinocyte ,business ,Intracellular ,medicine.drug - Abstract
The use of multikinase inhibitors (MKI) in oncology, such as sorafenib, is associated with a cutaneous adverse event called hand–foot skin reaction (HFSR), in which sites of pressure or friction become inflamed and painful, thus significantly impacting quality of life. The pathogenesis of MKI-induced HFSR is unknown, and the only available treatment options involve dose reduction or discontinuation of therapy, which have negative effects on primary disease management. To investigate the underlying mechanisms by which sorafenib promotes keratinocyte cytotoxicity and subsequent HFSR induction, we performed a transporter-directed RNAi screen in human epidermal keratinocytes and identified SLC22A20 (OAT6) as an uptake carrier of sorafenib. Further investigations into the intracellular mechanism of sorafenib activity through in situ kinome profiling identified the mitogen-activated protein kinase MAP3K7 (TAK1) as a target of sorafenib that induces cell death. Finally, we demonstrate that sorafenib induced keratinocyte injury in vivo and that this effect could be reversed by cotreatment with the OAT6 inhibitor probenecid. Collectively, our findings reveal a novel pathway that regulates the entry of some MKIs into keratinocytes and explains the basis underlying sorafenib-induced skin toxicity, with important implications for the therapeutic management of HFSR. Cancer Res; 76(1); 117–26. ©2015 AACR.
- Published
- 2016
- Full Text
- View/download PDF
20. Serine 350 of human pregnane X receptor is crucial for its heterodimerization with retinoid X receptor alpha and transactivation of target genes in vitro and in vivo
- Author
-
Wenwei Lin, Jing Wu, Sergio C. Chai, Xiaojuan Chai, Wen Xie, Jordan A. Beard, Erin G. Schuetz, Su Zeng, Taosheng Chen, Satyanarayana R. Pondugula, Ayesha Elias, Yue-Ming Wang, and Su Sien Ong
- Subjects
Models, Molecular ,Transcriptional Activation ,Receptors, Steroid ,Transgene ,Mice, Transgenic ,Biology ,Biochemistry ,Article ,Transactivation ,Cell Line, Tumor ,Serine ,Transcriptional regulation ,Animals ,Cytochrome P-450 CYP3A ,Humans ,Phosphorylation ,Pharmacology ,Regulation of gene expression ,Pregnane X receptor ,Retinoid X Receptor alpha ,Retinoid X receptor alpha ,Pregnane X Receptor ,Membrane Proteins ,Molecular biology ,Cell biology ,Liver ,Nuclear receptor ,Mutation ,Protein Multimerization - Abstract
The human pregnane X receptor (hPXR), a member of the nuclear receptor superfamily, senses xenobiotics and controls the transcription of genes encoding drug-metabolizing enzymes and transporters. The regulation of hPXR's transcriptional activation of its target genes is important for xenobiotic detoxification and endobiotic metabolism, and hPXR dysregulation can cause various adverse drug effects. Studies have implicated the putative phosphorylation site serine 350 (Ser 350 ) in regulating hPXR transcriptional activity, but the mechanism of regulation remains elusive. Here we investigated the transactivation of hPXR target genes in vitro and in vivo by hPXR with a phosphomimetic mutation at Ser 350 (hPXR S350D ). The S350D phosphomimetic mutation reduced the endogenous expression of cytochrome P450 3A4 (an hPXR target gene) in HepG2 and LS180 cells. Biochemical assays and structural modeling revealed that Ser 350 of hPXR is crucial for formation of the hPXR–retinoid X receptor alpha (RXRα) heterodimer. The S350D mutation abrogated heterodimerization in a ligand-independent manner, impairing hPXR-mediated transactivation. Further, in a novel humanized transgenic mouse model expressing the hPXR S350D transgene, we demonstrated that the S350D mutation alone is sufficient to impair hPXR transcriptional activity in mouse liver. This transgenic mouse model provides a unique tool to investigate the regulation and function of hPXR, including its non-genomic function, in vivo . Our finding that phosphorylation regulates hPXR activity has implications for development of novel hPXR antagonists and for safety evaluation during drug development.
- Published
- 2015
- Full Text
- View/download PDF
21. Targeting Histone Demethylases in MYC-Driven Neuroblastomas with Ciclopirox
- Author
-
Dongli Hu, Jie Zheng, Sandra Milasta, Pollyanna Goh, Brandon Young, Ju Bao, Junmin Peng, Alaa Altahan, Jesse T. Davidson, Rodrigo B. Interiano, Stephen W. White, Vincent A. Boyd, Yinan Wu, R. Kiplin Guy, Ashutosh Mishra, Su Sien Ong, Junfang Zhou, Sourav Das, Douglas R. Green, Wenwei Lin, Taosheng Chen, Yingdi Wang, Yong Cheng, Jun J. Yang, Chunxu Qu, Amit C. Nathwani, Anang A. Shelat, Ruoning Wang, Jonathan Low, Andrew M. Davidoff, and Zhenmei Li
- Subjects
0301 basic medicine ,Enzymologic ,Cancer Research ,Antifungal Agents ,Transcription, Genetic ,Cellular differentiation ,Mice, SCID ,Oxidative Phosphorylation ,Epigenesis, Genetic ,Histones ,Mice ,Neuroblastoma ,Tumor Cells, Cultured ,RNA, Small Interfering ,Cancer ,Histone Demethylases ,Pediatric ,Cultured ,biology ,Cell Differentiation ,Tumor Cells ,Histone ,Oncology ,Transcription ,medicine.drug ,Pyridones ,Pediatric Cancer ,Oncology and Carcinogenesis ,SCID ,Small Interfering ,Gene Expression Regulation, Enzymologic ,Article ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,Rare Diseases ,Genetic ,medicine ,Genetics ,Animals ,Humans ,Epigenetics ,Oncology & Carcinogenesis ,Transcription factor ,Cell Proliferation ,Ciclopirox ,Cell growth ,Neurosciences ,030104 developmental biology ,Gene Expression Regulation ,biology.protein ,Cancer research ,Demethylase ,RNA ,Epigenesis - Abstract
Histone lysine demethylases facilitate the activity of oncogenic transcription factors, including possibly MYC. Here we show that multiple histone demethylases influence the viability and poor prognosis of neuroblastoma cells, where MYC is often overexpressed. We also identified the approved small-molecule antifungal agent ciclopirox as a novel pan-histone demethylase inhibitor. Ciclopirox targeted several histone demethylases, including KDM4B implicated in MYC function. Accordingly, ciclopirox inhibited Myc signaling in parallel with mitochondrial oxidative phosphorylation, resulting in suppression of neuroblastoma cell viability and inhibition of tumor growth associated with an induction of differentiation. Our findings provide new insights into epigenetic regulation of MYC function and suggest a novel pharmacologic basis to target histone demethylases as an indirect MYC-targeting approach for cancer therapy. Cancer Res; 77(17); 4626–38. ©2017 AACR.
- Published
- 2017
22. Stability of the human pregnane X receptor is regulated by E3 ligase UBR5 and serine/threonine kinase DYRK2
- Author
-
Ayesha Elias, Darren N. Saunders, Taosheng Chen, Su Sien Ong, Asli N. Goktug, and Jing Wu
- Subjects
Receptors, Steroid ,Ubiquitin-Protein Ligases ,Protein Serine-Threonine Kinases ,Biochemistry ,Article ,Ubiquitin ,Humans ,Molecular Biology ,Transcription factor ,Cells, Cultured ,Serine/threonine-specific protein kinase ,Gene knockdown ,Pregnane X receptor ,biology ,Protein Stability ,Kinase ,Pregnane X Receptor ,Ubiquitination ,Hep G2 Cells ,Cell Biology ,Protein-Tyrosine Kinases ,Ubiquitin ligase ,HEK293 Cells ,Hepatocytes ,biology.protein ,Phosphorylation - Abstract
The hPXR (human pregnane X receptor), a major chemical toxin sensor, is a ligand-induced transcription factor activated by various xenobiotics and toxins, resulting in the transcriptional up-regulation of detoxifying enzymes. To date, little is known about the upstream regulation of hPXR. Using MS analysis and a kinome-wide siRNA screen, we report that the E3 ligase UBR5 (ubiquitin protein ligase E3 component n-recognin 5) and DYRK2 (dual-specificity tyrosine-phosphorylation-regulated kinase 2) regulate hPXR stability. UBR5 knockdown resulted in accumulation of cellular hPXR and a concomitant increase in hPXR activity, whereas the rescue of UBR5 knockdown decreased the cellular hPXR level and activity. Importantly, UBR5 exerted its effect in concert with the serine/threonine kinase DYRK2, as the knockdown of DYRK2 phenocopied UBR5 knockdown. hPXR was shown to be a substrate for DYRK2, and DYRK2-dependent phosphorylation of hPXR facilitated its subsequent ubiquitination by UBR5. This is the first report of the post-translational regulation of hPXR via phosphorylation-facilitated ubiquitination by DYRK2 and UBR5. The results of the present study reveal the role of the ubiquitin–proteasomal pathway in modulating hPXR activity and indicate that pharmacological inhibitors of the ubiquitin–proteasomal pathway that regulate hPXR stability may negatively affect treatment outcome from unintended hPXR-mediated drug–drug interactions.
- Published
- 2014
- Full Text
- View/download PDF
23. Identification and characterization of phosphorylation sites within the pregnane X receptor protein
- Author
-
Taosheng Chen, Ashutosh Mishra, Junmin Peng, Anthony A. High, Su Sien Ong, Ayesha Elias, and Jing Wu
- Subjects
Protein Folding ,Receptors, Steroid ,Insecta ,Transcription, Genetic ,Mutant ,Response element ,digestive system ,Biochemistry ,Article ,Animals ,Humans ,Phosphorylation ,Pharmacology ,Pregnane X receptor ,Binding Sites ,biology ,Kinase ,Cyclin-Dependent Kinase 2 ,Cyclin-dependent kinase 2 ,Pregnane X Receptor ,Hep G2 Cells ,Subcellular localization ,digestive system diseases ,HEK293 Cells ,Phosphoprotein ,biology.protein - Abstract
Pregnane X receptor (PXR) is a xenobiotic sensor regulating the expression of genes involved in xenobiotic detoxification and elimination. Phosphorylation plays an important role in modulating PXR activity and several phosphorylation sites have been predicted and characterized in in vitro experiments. Although PXR has been shown to be a phosphoprotein in vivo, the exact residues that are phosphorylated remain elusive. Using mass spectrometry, we identified for the first time S114, T133/135, S167, and S200 residues that are phosphorylated in PXR following an in vitro kinase assay using cyclin-dependent kinase 2. We further found that the phosphorylation at S114, T133, and T135 occurred in PXR isolated from cells. We tested the phosphodeficient and phosphomimetic mutants corresponding to all the sites identified and determined that phosphorylation at S114 attenuates the transcriptional activity of PXR, consistent with the observation that the S114D mutant displayed reduced association with the PXR-targeted DNA response element. Phosphomimetic mutations at either T133 or T135 did not show a significant change in transcriptional activity however, the dual phosphomimetic mutant T133D/T135D displayed reduced transcriptional activity. Subcellular localization studies showed a varied distribution of the mutants suggesting that the regulation of PXR is much more complex than what we can observe by just overexpressing the mutants. Thus, our results provide the first direct evidence that PXR is phosphorylated at specific residues and suggest that further investigation is warranted to fully understand the regulation of PXR by phosphorylation.
- Published
- 2014
- Full Text
- View/download PDF
24. Piperine activates human pregnane X receptor to induce the expression of cytochrome P450 3A4 and multidrug resistance protein 1
- Author
-
Sergio C. Chai, Taosheng Chen, Yue-Ming Wang, Su Sien Ong, Wenwei Lin, Erin G. Schuetz, and Jing Wu
- Subjects
Male ,Models, Molecular ,Transcriptional Activation ,Receptors, Steroid ,Polyunsaturated Alkamides ,Pharmacology ,Real-Time Polymerase Chain Reaction ,Toxicology ,Binding, Competitive ,Article ,Cell Line ,Mice ,Nuclear Receptor Coactivator 2 ,chemistry.chemical_compound ,Alkaloids ,Piperidines ,Multidrug Resistance Protein 1 ,Coactivator ,Animals ,Cytochrome P-450 CYP3A ,Humans ,ATP Binding Cassette Transporter, Subfamily B, Member 1 ,Benzodioxoles ,RNA, Small Interfering ,P-glycoprotein ,Pregnane X receptor ,biology ,CYP3A4 ,Chimera ,fungi ,Hemodynamics ,Pregnane X Receptor ,Cytochrome P450 ,Mice, Inbred C57BL ,chemistry ,Piperine ,biology.protein ,Nuclear receptor coactivator 2 ,RNA ,lipids (amino acids, peptides, and proteins) ,Plasmids - Abstract
Activation of the pregnane X receptor (PXR) and subsequently its target genes, including those encoding drug transporters and metabolizing enzymes, while playing substantial roles in xenobiotics detoxification, might cause undesired drug-drug interactions. Recently, an increased awareness has been given to dietary components for potential induction of diet-drug interactions through activation of PXR. Here, we studied, whether piperine (PIP), a major component extracted from the widely-used daily spice black pepper, could induce PXR-mediated expression of cytochrome P450 3A4 (CYP3A4) and multidrug resistance protein 1 (MDR1). Our results showed that PIP activated human PXR (hPXR)-mediated CYP3A4 and MDR1 expression in human hepatocytes, intestine cells, and a mouse model; PIP activated hPXR by recruiting its coactivator SRC-1 in both cellular and cell-free systems; PIP bound to the hPXR ligand binding domain in a competitive ligand binding assay in vitro. The dichotomous effects of PIP on induction of CYP3A4 and MDR1 expression observed here and inhibition of their activity reported elsewhere challenges the potential use of PIP as a bioavailability enhancer and suggests that cautions should be taken for PIP consumption during drug treatment in patients, particularly those who favor daily pepper spice or rely on certain pepper remedies.
- Published
- 2013
- Full Text
- View/download PDF
25. A phosphotyrosine switch regulates organic cation transporters
- Author
-
Guido Cavaletti, Jason A. Sprowl, Guoqing Du, Robert B. Diasio, Navjotsingh Pabla, Taosheng Chen, Steven M. Offer, Matthias Schwab, Lie Li, Eberhard Schlatter, Rachel A. Ness, Su Sien Ong, Alice A. Gibson, Wenwei Lin, Alex Sparreboom, Adrian Stecula, Giuliano Ciarimboli, Jan H.M. Schellens, Shashank Bharill, Anne T. Nies, Ehud Y. Isacoff, Andrej Sali, Sharyn D. Baker, Shuiying Hu, Sprowl, J, Ong, S, Gibson, A, Hu, S, Du, G, Lin, W, Li, L, Bharill, S, Ness, R, Stecula, A, Offer, S, Diasio, R, Nies, A, Schwab, M, Cavaletti, G, Schlatter, E, Ciarimboli, G, Schellens, J, Isacoff, E, Sali, A, Chen, T, Baker, S, Sparreboom, A, and Pabla, N
- Subjects
Models, Molecular ,0301 basic medicine ,Organoplatinum Compounds ,General Physics and Astronomy ,Organic Anion Transporters ,Pharmacology ,chemistry.chemical_compound ,Mice ,Models ,Ganglia, Spinal ,Src family kinase ,Proto-Oncogene Proteins c-yes ,Multidisciplinary ,Organic cation transport proteins ,Kinase ,Liver-Specific Organic Anion Transporter 1 ,Organic Cation Transporter 1 ,Organic Cation Transporter 2 ,Protein-Tyrosine Kinases ,3. Good health ,Oxaliplatin ,5.1 Pharmaceuticals ,Efflux ,oxaliplatin, neurotoxicity ,Patient Safety ,Development of treatments and therapeutic interventions ,Tyrosine kinase ,YES1 ,Spinal ,Organic Cation Transport Proteins ,Science ,1.1 Normal biological development and functioning ,Antineoplastic Agents ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Underpinning research ,Animals ,Humans ,Phosphotyrosine ,Protein Kinase Inhibitors ,Molecular ,Tyrosine phosphorylation ,Transporter ,General Chemistry ,respiratory tract diseases ,030104 developmental biology ,HEK293 Cells ,chemistry ,Hela Cells ,biology.protein ,Ganglia ,HeLa Cells - Abstract
Membrane transporters are key determinants of therapeutic outcomes. They regulate systemic and cellular drug levels influencing efficacy as well as toxicities. Here we report a unique phosphorylation-dependent interaction between drug transporters and tyrosine kinase inhibitors (TKIs), which has uncovered widespread phosphotyrosine-mediated regulation of drug transporters. We initially found that organic cation transporters (OCTs), uptake carriers of metformin and oxaliplatin, were inhibited by several clinically used TKIs. Mechanistic studies showed that these TKIs inhibit the Src family kinase Yes1, which was found to be essential for OCT2 tyrosine phosphorylation and function. Yes1 inhibition in vivo diminished OCT2 activity, significantly mitigating oxaliplatin-induced acute sensory neuropathy. Along with OCT2, other SLC-family drug transporters are potentially part of an extensive ‘transporter-phosphoproteome' with unique susceptibility to TKIs. On the basis of these findings we propose that TKIs, an important and rapidly expanding class of therapeutics, can functionally modulate pharmacologically important proteins by inhibiting protein kinases essential for their post-translational regulation., Organic cation transporters are important drug transporters that influence therapeutic outcomes. Here, the authors find that these transporters are regulated by tyrosine phosphorylation and propose that tyrosine kinase inhibitors can influence drug transporter function through post-translational mechanisms.
- Published
- 2016
26. Role of CAR and PXR in xenobiotic sensing and metabolism
- Author
-
Sergio C. Chai, Su Sien Ong, Yue-Ming Wang, and Taosheng Chen
- Subjects
Receptors, Steroid ,Receptors, Cytoplasmic and Nuclear ,Computational biology ,Biology ,Ligands ,Toxicology ,Article ,Xenobiotics ,chemistry.chemical_compound ,Detoxification ,Constitutive androstane receptor ,Animals ,Humans ,Drug Interactions ,Constitutive Androstane Receptor ,Pharmacology ,Regulation of gene expression ,Pregnane X receptor ,Pregnane X Receptor ,General Medicine ,Gene Expression Regulation ,Biochemistry ,chemistry ,Post translational ,Expert opinion ,Inactivation, Metabolic ,Models, Animal ,Signal transduction ,Xenobiotic ,Protein Processing, Post-Translational ,Protein Binding ,Signal Transduction - Abstract
The xenobiotic detoxification system, which protects the human body from external chemicals, comprises drug-metabolizing enzymes and transporters whose expressions are regulated by pregnane X receptor (PXR) and the constitutive androstane receptor (CAR). The progress made in a large number of recent studies calls for a timely review to summarize and highlight these key discoveries.This review summarizes recent advances in elucidating the roles of PXR and CAR in the xenobiotic detoxification system. It also highlights the progress in understanding the regulation of PXR and CAR activity at the post-translational levels, as well as the structural basis for the regulation of these two xenobiotic sensors.Future efforts are needed to discover novel agonists and antagonists with species and isoform selectivity, to systematically understand the regulation of PXR and CAR at multiple levels (transcriptional, post-transcriptional and post-translational levels) in response to xenobiotics exposure, and to solve the structures of the full-length receptors, which will be enabled by improved protein expression and purification techniques and approaches. In addition, more efforts will be needed to validate PXR and CAR as disease-related therapeutic targets and thus expand their roles as master xenobiotic sensors.
- Published
- 2012
- Full Text
- View/download PDF
27. Regulation of Syk by Phosphorylation on Serine in the Linker Insert
- Author
-
W. Andy Tao, Marietta L. Harrison, Jianjie Hu, Jacob A. Galan, Su Sien Ong, Victoria A. Martin, Robert L. Geahlen, Haiyan Ma, and Leela L. Paris
- Subjects
Receptors, Antigen, B-Cell ,Syk ,chemical and pharmacologic phenomena ,Serine threonine protein kinase ,Peptide Mapping ,environment and public health ,Biochemistry ,Serine ,Humans ,Syk Kinase ,Protein phosphorylation ,Phosphorylation ,Molecular Biology ,Protein kinase C ,ets-Domain Protein Elk-1 ,Serine/threonine-specific protein kinase ,ZAP-70 Protein-Tyrosine Kinase ,NFATC Transcription Factors ,Chemistry ,Intracellular Signaling Peptides and Proteins ,hemic and immune systems ,U937 Cells ,Cell Biology ,Protein-Tyrosine Kinases ,enzymes and coenzymes (carbohydrates) ,Signal transduction ,Signal Transduction - Abstract
The Syk protein-tyrosine kinase is phosphorylated on multiple tyrosines after the aggregation of the B cell antigen receptor. However, metabolic labeling experiments indicate that Syk is inducibly phosphorylated to an even greater extent on serine after receptor ligation. A combination of phosphopeptide mapping and mass spectrometric analyses indicates that serine 291 is a major site of phosphorylation. Serine 291 lies within a 23-amino acid insert located within the linker B region that distinguishes Syk from SykB and Zap-70. The phosphorylation of serine-291 by protein kinase C enhances the ability of Syk to couple the antigen receptor to the activation of the transcription factors NFAT and Elk-1. Protein interaction studies indicate a role for the phosphorylated linker insert in promoting an interaction between Syk and the chaperone protein, prohibitin.
- Published
- 2010
- Full Text
- View/download PDF
28. The SH3 Domain of Lck Modulates T-Cell Receptor-Dependent Activation of Extracellular Signal-Regulated Kinase through Activation of Raf-1
- Author
-
Robert L. Geahlen, Bartek Rajwa, Su Sien Ong, Vivian T. Thieu, Marietta L. Harrison, and Manqing Li
- Subjects
MAPK/ERK pathway ,CD3 Complex ,MAP Kinase Signaling System ,Receptors, Antigen, T-Cell ,Golgi Apparatus ,chemical and pharmacologic phenomena ,SRC Family Tyrosine Kinase ,Biology ,Mitogen-activated protein kinase kinase ,Gene Expression Regulation, Enzymologic ,SH3 domain ,Substrate Specificity ,src Homology Domains ,Jurkat Cells ,Phosphoserine ,symbols.namesake ,Humans ,Phosphorylation ,Extracellular Signal-Regulated MAP Kinases ,Phosphotyrosine ,Molecular Biology ,Mitogen-Activated Protein Kinase Kinases ,ZAP-70 Protein-Tyrosine Kinase ,Phospholipase C gamma ,T-cell receptor ,hemic and immune systems ,Articles ,Cell Biology ,Golgi apparatus ,Cell biology ,Enzyme Activation ,Proto-Oncogene Proteins c-raf ,Protein Transport ,Lymphocyte Specific Protein Tyrosine Kinase p56(lck) ,symbols ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Engagement of the T-cell antigen receptor (TCR) results in the proximal activation of the Src family tyrosine kinase Lck. The activation of Lck leads to the downstream activation of the Ras/Raf/MEK/ERK signaling pathway (where ERK is extracellular signal-related kinase). Under conditions of weak, but not strong, stimulation through the TCR, a version of Lck that contains a single point mutation in the SH3 (Src homology 3) domain (W97ALck) fails to support the activation of ERK, despite initiating signaling through the TCR, as demonstrated by the robust activation of ZAP-70, PLC-gamma, and Ras. We determined that the signaling lesion in W97ALck-expressing cells lies at the level of Raf-1 activation and is dependent on the presence of tyrosines 340/341 in the Raf-1 sequence. These data demonstrate a second function for Lck in TCR-mediated signaling to ERK. Additionally, we found that a significant fraction of Lck is localized to the Golgi apparatus and that, compared with wild-type Lck, W97ALck displays aberrant Golgi membrane localization. Our results support a model where under conditions of weak stimulation through the TCR, in addition to activated Ras, Golgi apparatus-localized Lck is needed for the full activation of Raf-1.
- Published
- 2008
- Full Text
- View/download PDF
29. Mitigation of acute kidney injury by cell-cycle inhibitors that suppress both CDK4/6 and OCT2 functions
- Author
-
Eberhard Schlatter, Su Sien Ong, Aksana Vasilyeva, Lie Li, Guoqing Du, Laura J. Janke, Michael J Buege, Alice A. Gibson, Taosheng Chen, Alex Sparreboom, Navjotsingh Pabla, Jason A. Sprowl, Shuiying Hu, and Giuliano Ciarimboli
- Subjects
Organic Cation Transport Proteins ,Pyridines ,Aminopyridines ,Palbociclib ,urologic and male genital diseases ,Protective Agents ,Piperazines ,Nephrotoxicity ,Small Molecule Libraries ,Mice ,medicine ,Animals ,Humans ,Cisplatin ,Multidisciplinary ,biology ,urogenital system ,Cyclin-dependent kinase 4 ,Acute kidney injury ,Cyclin-Dependent Kinase 4 ,Organic Cation Transporter 2 ,Cell Cycle Checkpoints ,Cyclin-Dependent Kinase 6 ,Cell cycle ,Acute Kidney Injury ,Biological Sciences ,medicine.disease ,Enzyme Activation ,Disease Models, Animal ,HEK293 Cells ,Kidney Tubules ,Toxic injury ,Biochemistry ,Purines ,biology.protein ,Cancer research ,Cyclin-dependent kinase 6 ,medicine.drug ,HeLa Cells - Abstract
Acute kidney injury (AKI) is a potentially fatal syndrome characterized by a rapid decline in kidney function caused by ischemic or toxic injury to renal tubular cells. The widely used chemotherapy drug cisplatin accumulates preferentially in the renal tubular cells and is a frequent cause of drug-induced AKI. During the development of AKI the quiescent tubular cells reenter the cell cycle. Strategies that block cell-cycle progression ameliorate kidney injury, possibly by averting cell division in the presence of extensive DNA damage. However, the early signaling events that lead to cell-cycle activation during AKI are not known. In the current study, using mouse models of cisplatin nephrotoxicity, we show that the G1/S-regulating cyclin-dependent kinase 4/6 (CDK4/6) pathway is activated in parallel with renal cell-cycle entry but before the development of AKI. Targeted inhibition of CDK4/6 pathway by small-molecule inhibitors palbociclib (PD-0332991) and ribociclib (LEE011) resulted in inhibition of cell-cycle progression, amelioration of kidney injury, and improved overall survival. Of additional significance, these compounds were found to be potent inhibitors of organic cation transporter 2 (OCT2), which contributes to the cellular accumulation of cisplatin and subsequent kidney injury. The unique cell-cycle and OCT2-targeting activities of palbociclib and LEE011, combined with their potential for clinical translation, support their further exploration as therapeutic candidates for prevention of AKI.
- Published
- 2015
30. Cyclin-dependent kinase 4 phosphorylates and positively regulates PAX3-FOXO1 in human alveolar rhabdomyosarcoma cells
- Author
-
Jing Wu, Taosheng Chen, Su Sien Ong, and Lingling Liu
- Subjects
Cytoplasm ,Transcription, Genetic ,Gene Expression ,lcsh:Medicine ,Biochemistry ,Molecular Cell Biology ,Signaling in Cellular Processes ,Paired Box Transcription Factors ,Myocyte ,Phosphorylation ,lcsh:Science ,Cyclin ,Multidisciplinary ,Protein Kinase Signaling Cascade ,biology ,Forkhead Box Protein O1 ,Kinase ,food and beverages ,Forkhead Transcription Factors ,Signaling in Selected Disciplines ,musculoskeletal system ,Signaling Cascades ,Nuclear Signaling ,Mitotic Signaling ,Protein Transport ,embryonic structures ,Alveolar rhabdomyosarcoma ,Signal transduction ,hormones, hormone substitutes, and hormone antagonists ,Research Article ,Signal Transduction ,endocrine system ,Cyclin D1 ,Cell Line, Tumor ,DNA-binding proteins ,medicine ,Humans ,Biology ,PAX3 Transcription Factor ,Protein Kinase Inhibitors ,Transcription factor ,Rhabdomyosarcoma, Alveolar ,Oncogenic Signaling ,Cyclin-dependent kinase 4 ,lcsh:R ,Proteins ,Cyclin-Dependent Kinase 4 ,medicine.disease ,Enzyme Activation ,biology.protein ,Cancer research ,lcsh:Q ,Transcriptional Signaling - Abstract
Alveolar rhabdomyosarcoma (ARMS) is an aggressive childhood muscle sarcoma with a 5-year survival rate of less than 30%. More than 80% of ARMSs harbor a PAX3-FOXO1 fusion transcription factor. However, expression of PAX3-FOXO1 in muscle cells alone is not sufficient and requires the loss of function of Ink4a/ARF to promote malignant proliferation of muscle cells in vitro or initiate ARMS tumor formation in vivo. This prompted us to examine the signaling pathways required to activate the function of PAX3-FOXO1 and to explore the functional interaction between the Ink4a/ARF and PAX3-FOXO1 signaling pathways. Here we report that inhibition of cyclin-dependent kinase 4 (Cdk4) by fascaplysin (a small molecule selective inhibitor of Cdk4/cyclin D1 that we identified in a screen for compounds that inhibit PAX3-FOXO1) led to inhibition of the transcriptional activity of PAX3-FOXO1 in ARMS cell line Rh30. Consistent with this finding, activation of Cdk4 enhanced the activity of PAX3-FOXO1. In vitro kinase assays revealed that Cdk4 directly phosphorylated PAX3-FOXO1 at Ser(430). Whereas fascaplysin did not affect the protein level of PAX3-FOXO1, it did increase the cytoplasmic level of PAX3-FOXO1 in a portion of cells. Our findings indicate that Cdk4 phosphorylates and positively regulates PAX3-FOXO1 and suggest that inhibition of Cdk4 activity should be explored as a promising avenue for developing therapy for ARMS.
- Published
- 2013
31. Pregnane X Receptor in Drug Development
- Author
-
Su Sien Ong, Sergio C. Chai, Yue-Ming Wang, and Taosheng Chen
- Subjects
0303 health sciences ,03 medical and health sciences ,Pregnane X receptor ,0302 clinical medicine ,Drug development ,Chemistry ,030220 oncology & carcinogenesis ,Pharmacology ,digestive system ,digestive system diseases ,030304 developmental biology - Published
- 2011
32. Abstract 4415: Tyrosine phosphorylation is essential for OCT2 function and a target of inhibition by TKIs
- Author
-
Alice A. Gibson, Alex Sparreboom, Su Sien Ong, Guoqing Du, Navjotsingh Pabla, Taosheng Chen, Wenwei Lin, Shuiying Hu, Jason A. Sprowl, and Lie Li
- Subjects
Cancer Research ,chemistry.chemical_compound ,Oncology ,chemistry ,Tyrosine phosphorylation ,Function (biology) ,Cell biology - Abstract
Cisplatin and oxaliplatin are among the most widely used anticancer drugs in both children and adults. The clinical use of these agents is associated with dose-limiting damage to kidneys (nephrotoxicity) and peripheral nerves (neurotoxicity). Mechanistic understanding of the underlying etiology remains incomplete and preventive strategies are currently unavailable. We previously reported that cisplatin nephrotoxicity (Filipski et al, CPT 2009) and oxaliplatin neurotoxicity (Sprowl et al, PNAS 2013) are dependent on organic cation transporter-mediated uptake mechanisms and can be reduced by genetic or pharmacological inhibition of OCT2. With a goal to identify small molecule OCT2 inhibitors, that could be used to prevent these toxicities, we carried out a high-throughput chemical library screen. Multiple tyrosine kinase inhibitors (TKIs), including dasatinib and vandetanib, were identified as potent OCT2 inhibitors. Further studies showed that TKIs inhibit OCT2 through a non-competitive mechanism raising the possibility that tyrosine phosphorylation by an unknown TKI-sensitive protein kinase(s) may be essential for OCT2 function. Using in vitro cell culture models and proteomics, site-directed mutagenesis and functional assays we found that OCT2 is indeed tyrosine phosphorylated and this phosphorylation is essential for OCT2 function and sensitive to inhibition by TKIs. A subsequent RNAi kinase library screen identified the Src-family member YES1 as the OCT2-phosphorylating kinase. This was further supported by experiments where a TKI-resistant YES1 mutant was able to rescue OCT2 inhibition by dasatinib. Non-TKI inhibitors of YES1 were also found to suppress OCT2 function. Importantly, tyrosine phosphorylation of OCT2 was observed in vivo, in murine kidneys and dorsal root ganglions, major sites of OCT2 expression and toxicities of platinum-based drugs. siRNA mediated in vivo knockdown of YES1 reduced OCT2 phosphorylation and suppressed its function in the kidneys. Treatment with the YES1 targeting TKI, Dasatinib, suppressed OCT2 phosphorylation in dorsal root ganglions, reduced oxaliplatin uptake and significantly prevented oxaliplatin induced neurotoxicity in mice models. These findings have not only provided new mechanistic insights into the regulation of membrane transporters; but have also provided a rationale for the translational development of new targeted interventions using TKI pulse-exposure to mitigate the debilitating side effects associated with platinum-based chemotherapeutics. Note: This abstract was not presented at the meeting. Citation Format: Navjotsingh Pabla, Jason A. Sprowl, Su Sien Ong, Alice A. Gibson, Guoqing Du, Wenwei Lin, Shuiying Hu, Lie Li, Taosheng Chen, Alex Sparreboom. Tyrosine phosphorylation is essential for OCT2 function and a target of inhibition by TKIs. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 4415. doi:10.1158/1538-7445.AM2015-4415
- Published
- 2015
- Full Text
- View/download PDF
33. Role of the SH3 Domain in the Trafficking and Localization of Lck in T Lymphocytes
- Author
-
Andy W. Tao, Robert L. Geahlen, Marietta L. Harrison, and Su Sien Ong
- Subjects
Chemistry ,T cell ,CD28 ,chemical and pharmacologic phenomena ,hemic and immune systems ,Biochemistry ,SH3 domain ,Cell biology ,medicine.anatomical_structure ,Genetics ,medicine ,biological phenomena, cell phenomena, and immunity ,Molecular Biology ,Src family ,Tyrosine kinase ,Biotechnology - Abstract
Lck, a member of the Src family of non-receptor protein tyrosine kinase, plays a pivotal role in normal T cell development and activation. The SH3 domain of Lck is a protein-protein interacting dom...
- Published
- 2006
- Full Text
- View/download PDF
34. Stability of the human pregnane X receptor is regulated by E3 ligase UBR5 and serine/threonine kinase DYRK2.
- Author
-
Su Sien ONG, Asli N. GOKTUG, Ayesha ELIAS, Jing WU, SAUNDERS, Darren, and Taosheng CHEN
- Subjects
- *
PREGNANE X receptor , *SERINE/THREONINE kinases , *LIGASES , *TRANSCRIPTION factors , *XENOBIOTICS , *UBIQUITINATION , *PHOSPHORYLATION - Abstract
The hPXR (human pregnane X receptor), a major chemical toxin sensor, is a ligand-induced transcription factor activated by various xenobiotics and toxins, resulting in the transcriptional up-regulation of detoxifying enzymes. To date, little is known about the upstream regulation of hPXR. Using MS analysis and a kinome-wide siRNA screen, we report that the E3 ligase UBR5 (ubiquitin protein ligase E3 component n-recognin 5) and DYRK2 (dual-specificity tyrosine-phosphorylation-regulated kinase 2) regulate hPXR stability. UBR5 knockdown resulted in accumulation of cellular hPXR and a concomitant increase in hPXR activity, where as the rescue ofUBR5 knockdown decreased the cellular hPXR level and activity. Importantly, UBR5 exerted its effect in concert with the serine/threonine kinase DYRK2, as the knockdown of DYRK2 phenocopied UBR5 knockdown. hPXR was shown to be a substrate for DYRK2, and DYRK2-dependent phosphorylation of hPXR facilitated its subsequent ubiquitination by UBR5. This is the first report of the posttranslational regulation of hPXR via phosphorylation-facilitated ubiquitination by DYRK2 and UBR5. The results of the present study reveal the role of the ubiquitin-proteasomal pathway in modulating hPXR activity and indicate that pharmacological inhibitors of the ubiquitin-proteasomal pathway that regulate hPXR stability may negatively affect treatment outcome from unintended hPXR-mediated drug-drug interactions. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
35. The SH3 Domain of Lck Modulates T-Cell Receptor-Dependent Activation of Extracellular Signal-Regulated Kinase through Activation of Raf-1.
- Author
-
Manqing Li, Su Sien Ong, Rajwa, Bartek, Thieu, Vivian T., Geahlen, Robert L., and Harrison, Marietta L.
- Subjects
- *
T cell receptors , *ANTIGENS , *PROTEIN-tyrosine kinases , *CELLS , *TYROSINE , *GOLGI apparatus - Abstract
Engagement of the T-cell antigen receptor (TCR) results in the proximal activation of the Src family tyrosine kinase Lck. The activation of Lck leads to the downstream activation of the Ras/Raf/MEK/ERK signaling pathway (where ERK is extracellular signal-related kinase). Under conditions of weak, but not strong, stimulation through the TCR, a version of Lck that contains a single point mutation in the SH3 (Src homology 3) domain (W97ALck) fails to support the activation of ERK, despite initiating signaling through the TCR, as demonstrated by the robust activation of ZAP-70, PLC-γ, and Ras. We determined that the signaling lesion in W97ALck-expressing cells lies at the level of Raf-1 activation and is dependent on the presence of tyrosines 340/341 in the Raf-1 sequence. These data demonstrate a second function for Lck in TCR-mediated signaling to ERK. Additionally, we found that a significant fraction of Lck is localized to the Golgi apparatus and that, compared with wild-type Lck, W97ALck displays aberrant Golgi membrane localization. Our results support a model where under conditions of weak stimulation through the TCR, in addition to activated Ras, Golgi apparatus-localized Lck is needed for the full activation of Raf-1. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
36. Multikinase Inhibitors Induce Cutaneous Toxicity through OAT6-Mediated Uptake and MAP3K7-Driven Cell Death.
- Author
-
Zimmerman, Eric I., Gibson, Alice A., Shuiying Hu, Vasilyeva, Aksana, Orwick, Shelley J., Du, Guoqing, Mascara, Gerard P., Su Sien Ong, Taosheng Chen, Vogel, Peter, Inaba, Hiroto, Maitland, Michael L., Sparreboom, Alex, and Baker, Sharyn D.
- Subjects
- *
DERMATOTOXICOLOGY , *KERATINOCYTES , *HAND-foot syndrome , *MITOGEN-activated protein kinases , *SORAFENIB - Abstract
The use of multikinase inhibitors (MKI) in oncology, such as sorafenib, is associated with a cutaneous adverse event called hand-foot skin reaction (HFSR), in which sites of pressure or friction become inflamed and painful, thus significantly impacting quality of life. The pathogenesis of MKI-induced HFSR is unknown, and the only available treatment options involve dose reduction or discontinuation of therapy, which have negative effects on primary disease management. To investigate the underlying mechanisms by which sorafenib promotes keratinocyte cytotoxicity and subsequent HFSR induction, we performed a transporter-directed RNAi screen in human epidermal keratinocytes and identified SLC22A20 (OAT6) as an uptake carrier of sorafenib. Further investigations into the intracellular mechanism of sorafenib activity through in situ kinome profiling identified the mitogen-activated protein kinase MAP3K7 (TAK1) as a target of sorafenib that induces cell death. Finally, we demonstrate that sorafenib induced keratinocyte injury in vivo and that this effect could be reversed by cotreatment with the OAT6 inhibitor probenecid. Collectively, our findings reveal a novel pathway that regulates the entry of some MKIs into keratinocytes and explains the basis underlying sorafenib-induced skin toxicity, with important implications for the therapeutic management of HFSR. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
37. Regulation of Syk by Phosphorylation on Serine in the Linker Insert.
- Author
-
Paris, Leela L., Jianjie Hu, Galan, Jacob, Su Sien Ong, Martin, Victoria A., Ma, Haiyan, Tao, W. Andy, Harrison, Marietta L., and Geahlen, Robert L.
- Subjects
- *
AMINO acids , *CHEMICAL reactions , *CHEMICAL processes , *FLUORINATION , *OZONIZATION , *PHOSPHORYLATION - Abstract
The Syk protein-tyrosine kinase is phosphorylated on multiple tyrosines after the aggregation of the B cell antigen receptor. However, metabolic labeling experiments indicate that Syk is inducibly phosphorylated to an even greater extent on serine after receptor ligation. A combination of phosphopeptide mapping and mass spectrometric analyses indicates that serine 291 is a major site of phosphorylation. Serine 291 lies within a 23-amino acid insert located within the linker B region that distinguishes Syk from SykB and Zap-70. The phosphorylation of serine-291 by protein kinase C enhances the ability of Syk to couple the antigen receptor to the activation of the transcription factors NFAT and Elk-1. Protein interaction studies indicate a role for the phosphorylated linker insert in promoting an interaction between Syk and the chaperone protein, prohibitin. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.