1
|
Angelika P, Anniina V, Olli S, Teemu H. Development of an enzyme-coupled activity assay for Janus kinase 2 inhibitor screening. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2023:S2472-5552(23)00036-9. [PMID: 37149038 DOI: 10.1016/j.slasd.2023.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/05/2023] [Accepted: 05/02/2023] [Indexed: 05/08/2023]
Abstract
JAK2 transmits signals of several important cytokines, such as growth hormone and erythropoietin. The interest toward the therapeutic targeting of JAK2 was boosted in 2005, when the somatic JAK2 V617F mutation, responsible for the majority of myeloproliferative neoplasms (MPNs) was discovered. JAK2 inhibitors have been approved for MPN therapy and they are effective in alleviating symptoms and improving the quality of life of the patients, but they do not lead to molecular remission. This calls for the discovery of new compounds for JAK2-targeted therapeutic approaches. Here we describe the development of a fluorescence-based activity assay for the screening of versatile inhibitor types against JAK2. The assay was utilized to screen a diverse set of small molecule weight natural products and the assay performance was compared to that of differential scanning fluorimetry. We identified 37 hits and further analysis of the most potent hits revealed that most of them displayed non-ATP competitive binding modes. The hits were profiled against other JAK family members and showed distinctive selectivity profiles. The developed assay is consistent, simple and inexpensive to use, and can be utilized for inhibitor screening of diverse compound classes against all JAK family members.
Collapse
Affiliation(s)
- Pölläniemi Angelika
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Virtanen Anniina
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Silvennoinen Olli
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Haikarainen Teemu
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| |
Collapse
|
2
|
Smirnova TD, Shtykov SN, Zhelobitskaya EA. Energy transfer in liquid and solid nanoobjects: application in luminescent analysis. PHYSICAL SCIENCES REVIEWS 2019. [DOI: 10.1515/psr-2018-9981] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Abstract
Radiationless resonance electronic excitation energy transfer (ET) is a fundamental physical phenomenon in luminescence spectroscopy playing an important role in natural processes, especially in photosynthesis and biochemistry. Besides, it is widely used in photooptics, optoelectronics, and protein chemistry, coordination chemistry of transition metals and lanthanides as well as in luminescent analysis. ET involves the transfer of electronic energy from a donor (D) (molecules or particles) which is initially excited, to an acceptor (A) at the ground state to emit it later. Fluorescence or phosphorescence of the acceptor that occurs during ET is known as sensitized. There do many kinds of ET exist but in all cases along with other factors the rate and efficiency of ET in common solvents depends to a large extent on the distance between the donor and the acceptor. This dependency greatly limits the efficiency of ET and, correspondingly, does not allow the determination of analytes in highly diluted (10–9–10–15 M) solutions. To solve the problem of distance-effect, the effects of concentrating and bring close together the donor and acceptor in surfactant micelles (liquid nanosystems) or sorption on solid nanoparticles are used. Various approaches to promote the efficiency of ET for improvement determination selectivity and sensitivity using liquid and solid nanoobjects is reviewed and analyzed.
Collapse
|
3
|
Stone SR, Heinrich T, Juraja SM, Satiaputra JN, Hall CM, Anastasas M, Mills AD, Chamberlain CA, Winslow S, Priebatsch K, Cunningham PT, Hoffmann K, Milech N. β-Lactamase Tools for Establishing Cell Internalization and Cytosolic Delivery of Cell Penetrating Peptides. Biomolecules 2018; 8:biom8030051. [PMID: 29997382 PMCID: PMC6163455 DOI: 10.3390/biom8030051] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/22/2018] [Accepted: 06/29/2018] [Indexed: 12/15/2022] Open
Abstract
The ability of cell penetrating peptides (CPPs) to deliver biologically relevant cargos into cells is becoming more important as targets in the intracellular space continue to be explored. We have developed two assays based on CPP-dependent, intracellular delivery of TEM-1 β-lactamase enzyme, a functional biological molecule comparable in size to many protein therapeutics. The first assay focuses on the delivery of full-length β-lactamase to evaluate the internalization potential of a CPP sequence. The second assay uses a split-protein system where one component of β-lactamase is constitutively expressed in the cytoplasm of a stable cell line and the other component is delivered by a CPP. The delivery of a split β-lactamase component evaluates the cytosolic delivery capacity of a CPP. We demonstrate that these assays are rapid, flexible and have potential for use with any cell type and CPP sequence. Both assays are validated using canonical and novel CPPs, with limits of detection from <500 nM to 1 µM. Together, the β-lactamase assays provide compatible tools for functional characterization of CPP activity and the delivery of biological cargos into cells.
Collapse
Affiliation(s)
- Shane R Stone
- Telethon Kids Institute, University of Western Australia, Subiaco, WA 6008, Australia.
- Phylogica Pty Ltd., Subiaco, WA 6008, Australia.
| | - Tatjana Heinrich
- Telethon Kids Institute, University of Western Australia, Subiaco, WA 6008, Australia.
- Phylogica Pty Ltd., Subiaco, WA 6008, Australia.
| | - Suzy M Juraja
- Telethon Kids Institute, University of Western Australia, Subiaco, WA 6008, Australia.
- Phylogica Pty Ltd., Subiaco, WA 6008, Australia.
| | - Jiulia N Satiaputra
- Telethon Kids Institute, University of Western Australia, Subiaco, WA 6008, Australia.
- Phylogica Pty Ltd., Subiaco, WA 6008, Australia.
| | - Clinton M Hall
- Telethon Kids Institute, University of Western Australia, Subiaco, WA 6008, Australia.
- Phylogica Pty Ltd., Subiaco, WA 6008, Australia.
| | - Mark Anastasas
- Telethon Kids Institute, University of Western Australia, Subiaco, WA 6008, Australia.
- Phylogica Pty Ltd., Subiaco, WA 6008, Australia.
| | - Anna D Mills
- Telethon Kids Institute, University of Western Australia, Subiaco, WA 6008, Australia.
- Phylogica Pty Ltd., Subiaco, WA 6008, Australia.
| | - Christopher A Chamberlain
- Telethon Kids Institute, University of Western Australia, Subiaco, WA 6008, Australia.
- Phylogica Pty Ltd., Subiaco, WA 6008, Australia.
| | - Scott Winslow
- Telethon Kids Institute, University of Western Australia, Subiaco, WA 6008, Australia.
- Phylogica Pty Ltd., Subiaco, WA 6008, Australia.
| | - Kristin Priebatsch
- Telethon Kids Institute, University of Western Australia, Subiaco, WA 6008, Australia.
- Phylogica Pty Ltd., Subiaco, WA 6008, Australia.
| | - Paula T Cunningham
- Telethon Kids Institute, University of Western Australia, Subiaco, WA 6008, Australia.
- Phylogica Pty Ltd., Subiaco, WA 6008, Australia.
| | - Katrin Hoffmann
- Telethon Kids Institute, University of Western Australia, Subiaco, WA 6008, Australia.
- Phylogica Pty Ltd., Subiaco, WA 6008, Australia.
| | - Nadia Milech
- Telethon Kids Institute, University of Western Australia, Subiaco, WA 6008, Australia.
- Phylogica Pty Ltd., Subiaco, WA 6008, Australia.
| |
Collapse
|
4
|
Goryashchenko AS, Khrenova MG, Bochkova AA, Ivashina TV, Vinokurov LM, Savitsky AP. Genetically Encoded FRET-Sensor Based on Terbium Chelate and Red Fluorescent Protein for Detection of Caspase-3 Activity. Int J Mol Sci 2015; 16:16642-54. [PMID: 26204836 PMCID: PMC4519970 DOI: 10.3390/ijms160716642] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 06/30/2015] [Accepted: 07/06/2015] [Indexed: 12/01/2022] Open
Abstract
This article describes the genetically encoded caspase-3 FRET-sensor based on the terbium-binding peptide, cleavable linker with caspase-3 recognition site, and red fluorescent protein TagRFP. The engineered construction performs two induction-resonance energy transfer processes: from tryptophan of the terbium-binding peptide to Tb(3+) and from sensitized Tb(3+) to acceptor--the chromophore of TagRFP. Long-lived terbium-sensitized emission (microseconds), pulse excitation source, and time-resolved detection were utilized to eliminate directly excited TagRFP fluorescence and background cellular autofluorescence, which lasts a fraction of nanosecond, and thus to improve sensitivity of analyses. Furthermore the technique facilitates selective detection of fluorescence, induced by uncleaved acceptor emission. For the first time it was shown that fluorescence resonance energy transfer between sensitized terbium and TagRFP in the engineered construction can be studied via detection of microsecond TagRFP fluorescence intensities. The lifetime and distance distribution between donor and acceptor were calculated using molecular dynamics simulation. Using this data, quantum yield of terbium ions with binding peptide was estimated.
Collapse
Affiliation(s)
| | - Maria G Khrenova
- M. V. Lomonosov Moscow State University, Department of Chemistry, 119991 Moscow, Russia.
| | - Anna A Bochkova
- M. V. Lomonosov Moscow State University, Department of Chemistry, 119991 Moscow, Russia.
| | - Tatiana V Ivashina
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Russian Academy of Sciences, 142290 Pushchino, Russia.
| | - Leonid M Vinokurov
- Branch of Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 142290 Pushchino, Russia.
| | - Alexander P Savitsky
- A. N. Bach Institute of Biochemistry, Russian Academy of Sciences, 119071 Moscow, Russia.
- M. V. Lomonosov Moscow State University, Department of Chemistry, 119991 Moscow, Russia.
| |
Collapse
|
5
|
Vuojola J, Soukka T. Luminescent lanthanide reporters: new concepts for use in bioanalytical applications. Methods Appl Fluoresc 2014; 2:012001. [DOI: 10.1088/2050-6120/2/1/012001] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
6
|
Zherdeva VV, Savitsky AP. Using lanthanide-based resonance energy transfer for in vitro and in vivo studies of biological processes. BIOCHEMISTRY (MOSCOW) 2013; 77:1553-74. [DOI: 10.1134/s0006297912130111] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
7
|
Qian J, Mason JL, Holskin BP, Murray KA, Meyer SL, Ator MA, Angeles TS. Comparison of two homogeneous cell-based kinase assays for JAK2 V617F: SureFire pSTAT5 and GeneBLAzer fluorescence resonance energy transfer assays. Assay Drug Dev Technol 2011; 10:212-7. [PMID: 22132729 DOI: 10.1089/adt.2011.0399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling pathway plays an important role in cellular responses to cytokines and growth factors. Recent studies have identified a recurrent somatic activating mutation (JAK2 V617F) in majority of patients with myeloproliferative disorders (MPDs). Development of drugs that target JAK2 V617F is, therefore, of therapeutic relevance. To discover small molecule inhibitors for this target, robust and reliable cell-based assays are important. Here, we present a comparison of two homogeneous, 384-well plate-based cellular assays using Invitrogen's CellSensor® JAK2 V617F interferon regulatory factor-1 (irf1)-beta-lactamase (bla) human erythroleukemia line (HEL): (1) SureFire® pSTAT5 AlphaScreen® assay from PerkinElmer; and (2) GeneBLAzer® fluorescence resonance energy transfer assay from Invitrogen. HEL cells are growth factor-independent due to JAK2 V617F mutation that causes constitutive STAT5 activation. The SureFire assay measures levels of phosphorylated STAT5 downstream of JAKs, while the GeneBLAzer assay is a reporter assay that monitors bla activity further downstream of STAT5. Evaluation of a number of chemically diverse JAK2 inhibitors in the two cellular assays yielded comparable half-maximal inhibitory concentration (IC₅₀) values, boding well for the utility of these assay formats in compound profiling.
Collapse
Affiliation(s)
- Jie Qian
- Worldwide Discovery Research, Cephalon, Inc., West Chester, Pennsylvania 19380, USA
| | | | | | | | | | | | | |
Collapse
|
8
|
Machleidt T, Robers MB, Hermanson SB, Dudek JM, Bi K. TR-FRET cellular assays for interrogating posttranslational modifications of histone H3. ACTA ACUST UNITED AC 2011; 16:1236-46. [PMID: 21972037 DOI: 10.1177/1087057111422943] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Posttranslational modifications such as phosphorylation, acetylation, and methylation play important roles in regulating the structures and functions of histones, which in turn regulate gene expression and DNA repair and replication. Histone-modifying enzymes, such as deacetylases, methyltransferases and demethylases, have been pursued as therapeutic targets for various diseases. However, detection of the activities of these enzymes in high-throughput cell-based formats has remained challenging. The authors have developed high-throughput LanthaScreen cellular assays for Histone H3 site-specific modifications. These assays use cells expressing green fluorescence protein-tagged Histone H3 transiently delivered via BacMam and terbium-labeled anti-Histone H3 modification-specific antibodies. Robust time-resolved Förster resonance energy transfer signals were detected for H3 lysine-9 acetylation and dimethylation (H3K9me2), serine-10 phosphorylation, K4 di- and trimethylation, and K27 trimethylation. Consistent with previous reports, hypoxic stress increased K4 methylation levels, and methyltransferase G9a inhibitor UNC-0638 decreased K9me2 levels significantly, with little effects on other modifications. To demonstrate the utility of this assay platform in screening, the K9 acetylation assay was used to profile the Enzo Epigenetics Library. Twelve known HDAC inhibitors were identified as hits and followed up in a dose-response format. In conclusion, this assay platform enables high-throughput cell-based analysis of diverse types of posttranslational modifications of Histone H3.
Collapse
Affiliation(s)
- Thomas Machleidt
- Life Technologies Corporation, Discovery and ADMET Systems, Madison, WI 53719, USA
| | | | | | | | | |
Collapse
|
9
|
Mason JL, Holskin BP, Murray KA, Meyer SL, Wells-Knecht KJ, Ator MA, Angeles TS. Modification of CellSensor irf1-bla TF-1 and irf1-bla HEL Assays for Direct Comparison of Wild-Type JAK2 and JAK2 V617F Inhibition. Assay Drug Dev Technol 2011; 9:311-8. [PMID: 21133674 DOI: 10.1089/adt.2010.0297] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Jennifer L. Mason
- Lead Discovery and Profiling, Worldwide Discovery Research, Cephalon Inc., West Chester, Pennsylvania
| | - Beverly P. Holskin
- Lead Discovery and Profiling, Worldwide Discovery Research, Cephalon Inc., West Chester, Pennsylvania
| | - Kristen A. Murray
- Lead Discovery and Profiling, Worldwide Discovery Research, Cephalon Inc., West Chester, Pennsylvania
| | - Sheryl L. Meyer
- Lead Discovery and Profiling, Worldwide Discovery Research, Cephalon Inc., West Chester, Pennsylvania
| | - Kevin J. Wells-Knecht
- Analytical Development, Worldwide Analytical Research, Cephalon Inc., West Chester, Pennsylvania
| | - Mark A. Ator
- Lead Discovery and Profiling, Worldwide Discovery Research, Cephalon Inc., West Chester, Pennsylvania
| | - Thelma S. Angeles
- Lead Discovery and Profiling, Worldwide Discovery Research, Cephalon Inc., West Chester, Pennsylvania
| |
Collapse
|
10
|
Robers MB, Loh C, Carlson CB, Yang H, Frey EA, Hermanson SB, Bi K. Measurement of the cellular deacetylase activity of SIRT1 on p53 via LanthaScreen® technology. MOLECULAR BIOSYSTEMS 2010; 7:59-66. [PMID: 20931131 DOI: 10.1039/c0mb00026d] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Upon genomic insult, the tumor suppressor p53 is phosphorylated and acetylated at specific serine and lysine residues, increasing its stability and transactivation function. Deacetylases, including the type III histone deacetylase SIRT1, remove acetyl groups from p53 and counterbalance acetyltransferase activity during a DNA damage response. This report describes a series of high-throughput LanthaScreen® time-resolved Förster resonance energy transfer (TR-FRET) immunoassays for detection of intracellular p53 phosphorylation of Ser15 and acetylation of Lys382 upon treatment with DNA damage agents, such as etoposide. These assays were used to measure the deacetylase activity of SIRT1 and/or Type I/II Histone deacetylases (HDACs). First, BacMam-mediated overexpression of SIRT1 resulted in dose-dependent deacetylation of GFP-p53 following etoposide treatment of U-2 OS cells, confirming that GFP-p53 serves as a SIRT1 substrate in this assay format. Further, overexpression of the acetyltransferase p300 via BacMam increased the acetylation of GFP-p53 at Lys382. Next, siRNA-mediated knockdown of SIRT1 resulted in increased GFP-p53 acetylation, indicating that endogenous SIRT1 activity can also be measured in U-2 OS cells. Consistent with these results, GFP-p53 acetylation was also increased upon treatment of cells with a small-molecule inhibitor of SIRT1, EX-527. The effect of this compound was dramatically increased when used in combination with chemotherapeutic drug and/or the HDAC inhibitor Trichostatin A, confirming a proposed synergistic mechanism of p53 deacetylation by SIRT1 and Type I/II HDACs. Taken together, the cellular assays described here can be used as high-throughput alternatives to traditional immunoassays such as western blotting for identifying pharmacological modulators of specific p53-modifying enzymes.
Collapse
Affiliation(s)
- Matthew B Robers
- Discovery Assays and Services, Life Technologies Corporation, Madison, WI 53719, USA
| | | | | | | | | | | | | |
Collapse
|
11
|
Malemud CJ. Suppression of Autoimmune Arthritis by Small Molecule Inhibitors of the JAK/STAT Pathway. Pharmaceuticals (Basel) 2010; 3:1446-1455. [PMID: 27713312 PMCID: PMC4033991 DOI: 10.3390/ph3051446] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2010] [Revised: 04/20/2010] [Accepted: 05/11/2010] [Indexed: 12/23/2022] Open
Abstract
A skewed ratio of pro-inflammatory to anti-inflammatory cytokines, elevated growth factor synthesis and T- and B-lymphocyte activation are 3 hallmarks of rheumatoid arthritis (RA) pathology. Interleukin-6 (IL-6), IL-7, IL-17, IL-12/IL-23 and growth factors, granulocyte macrophage-colony stimulating factor, IL-3, and erythropoietin activate the Janus Kinase/Signal Transducers and Activators of Transcription (JAK/STAT) pathway. Evidence showed that STAT protein phosphorylation (p-STAT) by activated JAKs is permissive for p-STAT to act as transcription factors by binding to STAT-responsive gene promoter sequences. This event is critical for perpetuating RA, in part, by up-regulating pro-inflammatory cytokine gene transcription. Activation of JAK/STAT by cytokines and growth factors can induce ‘cross-talk’ with other signaling pathways by which Stress-Activated Protein/Mitogen-Activated Protein Kinase (SAP/MAPK) and Phosphatidylinositide-3-Kinase (PI3K)-mediated signaling are also activated. JAK-specific small molecule inhibitors (SMIs) were developed to test whether JAK/STAT pathway blockade would regulate autoimmune-mediated inflammation. JAK-specific SMI blockade inhibited p-STAT induced by pro-inflammatory cytokines in vitro. Systemically administered JAK-specific SMI blockade also ameliorated biomarkers of inflammation in well-validated arthritis animal models. A few JAK-specific SMIs have made their way into RA clinical trials. In fact, the JAK3-specific SMI, CP-690,500 is the first JAK/STAT SMI to be assessed for clinical efficacy in a Phase III RA trial.
Collapse
Affiliation(s)
- Charles J Malemud
- Division of Rheumatic Diseases, Departments of Medicine & Anatomy, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA.
| |
Collapse
|
12
|
Osmond RIW, Das S, Crouch MF. Development of cell-based assays for cytokine receptor signaling, using an AlphaScreen SureFire assay format. Anal Biochem 2010; 403:94-101. [PMID: 20382104 DOI: 10.1016/j.ab.2010.04.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 03/21/2010] [Accepted: 04/05/2010] [Indexed: 11/27/2022]
Abstract
The signal transducers and activators of transcription (STAT) proteins are a small family of signaling proteins that are crucial for cytokine and growth factor receptor-mediated signaling in various blood cell types. Despite their central role in immune and hematopoietic cellular regulation, there are relatively few options for monitoring receptor-mediated JAK/STAT signaling events in a cell-based format, without the need for cellular transfections or labor intensive methodology. Indeed, traditional methods such as the Western blot or ELISA remain a standard method for determining the phosphorylation status of endogenous STAT proteins. Here we present data for the rapid detection of endogenous receptor-mediated phosphorylation of multiple STAT proteins using the bead-based AlphaScreen SureFire technology. With three different cell lines (human acute monocytic leukemia THP-1 cells, human erythroleukemic TF-1 cells, and human T lymphocytic Jurkat cells), we have optimized a rapid and homogeneous methodology for monitoring endogenous, receptor-mediated signaling via STAT 1, STAT 3, or STAT 5 phosphorylation, in response to several agonists. These assays, which can be tailored for both standard research applications or high-throughput drug screening applications, afford quantitative data for receptor-mediated signaling mechanisms in an endogenous, cellular environment.
Collapse
|
13
|
Kim SH, Gunther JR, Katzenellenbogen JA. Monitoring a coordinated exchange process in a four-component biological interaction system: development of a time-resolved terbium-based one-donor/three-acceptor multicolor FRET system. J Am Chem Soc 2010; 132:4685-92. [PMID: 20230029 PMCID: PMC2860875 DOI: 10.1021/ja100248q] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Hormonal regulation of cellular function involves the binding of small molecules with receptors that then coordinate subsequent interactions with other signal transduction proteins. These dynamic, multicomponent processes are difficult to track in cells and even in reconstituted in vitro systems, and most methods can monitor only two-component interactions, often with limited capacity to follow dynamic changes. Through a judicious choice of three organic acceptor fluorophores paired with a terbium donor fluorophore, we have developed the first example of a one-donor/three-acceptor multicolor time-resolved fluorescence energy transfer (TR-FRET) system, and we have exemplified its use by monitoring a ligand-regulated protein-protein exchange process in a four-component biological system. By careful quantification of the emission from each of the three acceptors at the four channels for terbium donor emission, we demonstrate that any of these donor channels can be used to estimate the magnitude of the three FRET signals in this terbium-donor triple-acceptor system with minimal bleedthrough. Using this three-channel terbium-based, TR-FRET assay system, we show in one experiment that the addition of a fluorescein-labeled estrogen agonist displaces a SNAPFL-labeled antiestrogen from the ligand binding pocket of a terbium-labeled estrogen receptor, at the same time causing a Cy5-labeled coactivator to be recruited to the estrogen receptor. This experiment demonstrates the power of a four-color TR-FRET experiment, and it shows that the overall process of estrogen receptor ligand exchange and coactivator binding is a dynamic but precisely coordinated process.
Collapse
Affiliation(s)
- Sung Hoon Kim
- Department of Chemistry, University of Illinois, 600 S. Mathews Ave., Urbana, IL 61801
| | - Jillian R. Gunther
- Department of Chemistry, University of Illinois, 600 S. Mathews Ave., Urbana, IL 61801
| | | |
Collapse
|
14
|
Carlson CB, Mashock MJ, Bi K. BacMam-enabled LanthaScreen cellular assays for PI3K/Akt pathway compound profiling in disease-relevant cell backgrounds. ACTA ACUST UNITED AC 2010; 15:327-34. [PMID: 20145103 DOI: 10.1177/1087057109357788] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The authors recently reported the development and application of multiple LanthaScreen cellular assays to interrogate specific steps within the PI3K/Akt pathway. The importance of this signaling cascade in regulating fundamental aspects of cell growth and survival, as well as in the progression of cancer, underscores the need for portable cell-based assays for compound profiling in multiple disease-relevant cell backgrounds. To meet this need, the authors have now expanded their LanthaScreen assay platform across a variety of cell types using a gene delivery technology known as BacMam. Here, they have demonstrated the successful detection of Akt-dependent phosphorylation of PRAS40 at Thr246 in 10 different cell lines harboring mutations known to activate the PI3K/Akt pathway. In addition, they generated inhibitory profiles of 17 known pathway inhibitors in these same cells to validate the approach of using the BacMam-enabled LanthaScreen cellular assay format to rapidly profile compounds in disease-relevant cell types. Importantly, their results provide a broad illustration of how the genetic alterations that affect PI3K/Akt signaling can also influence the inhibitory profile of a given compound.
Collapse
Affiliation(s)
- Coby B Carlson
- Invitrogen Discovery Assays and Services, Cell Systems Division, Invitrogen (Part of Life Technologies), Madison, Wisconsin 53719, USA.
| | | | | |
Collapse
|
15
|
Hancock MK, Lebakken CS, Wang J, Bi K. Multi-pathway cellular analysis of compound selectivity. MOLECULAR BIOSYSTEMS 2010; 6:1834-43. [DOI: 10.1039/c003669b] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
16
|
Lieu PT, Machleidt T, Thyagarajan B, Fontes A, Frey E, Fuerstenau-Sharp M, Thompson DV, Swamilingiah GM, Derebail SS, Piper D, Chesnut JD. Generation of Site-Specific Retargeting Platform Cell Lines for Drug Discovery Using phiC31 and R4 Integrases. ACTA ACUST UNITED AC 2009; 14:1207-15. [DOI: 10.1177/1087057109348941] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
One of the challenges in developing cell lines for high-throughput screening in drug discovery is the labor- and time-intensive process required to create stable clonal cell lines that express specific reporters or drug targets. The authors report here the generation of a site-specific retargeting platform in 3 different cell lines: adherent HEK293, suspension CHO-S, and a human embryonic cell line (BGO1V). These platform cell lines were generated by using a combination of 2 site-specific integrases to develop a system that allows one to efficiently target a gene of interest to a specific locus and generates rapid production of homogeneous cell pools that stably express the gene of interest. The phiC31 integrase was used to create a platform line by placing a target site for the R4 integrase into a pseudo attP site, and then the R4 integrase was used to place a gene of interest into specific R4 target site. The authors demonstrate the successful and rapid retargeting of a G-protein-coupled receptor (cholecystokinin receptor A, CCKAR), an ion channel (the transient receptor potential cation channel, subfamily M, member 8, TRPM8), and a GFP-c-Jun(1-79) fusion protein into the specific loci in these cell lines and show that these retargeted cell lines exhibit functional and pharmacological responses consistent with those reported in the literature.
Collapse
|
17
|
Huwiler KG, Machleidt T, Chase L, Hanson B, Robers MB. Characterization of serotonin 5-hydroxytryptamine-1A receptor activation using a phospho-extracellular-signal regulated kinase 2 sensor. Anal Biochem 2009; 393:95-104. [DOI: 10.1016/j.ab.2009.06.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2009] [Accepted: 06/15/2009] [Indexed: 11/16/2022]
|
18
|
Johnson RL, Huang R, Jadhav A, Southall N, Wichterman J, MacArthur R, Xia M, Bi K, Printen J, Austin CP, Inglese J. A quantitative high-throughput screen for modulators of IL-6 signaling: a model for interrogating biological networks using chemical libraries. MOLECULAR BIOSYSTEMS 2009; 5:1039-50. [PMID: 19668870 DOI: 10.1039/b902021g] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Small molecule modulators are critical for dissecting and understanding signaling pathways at the molecular level. Interleukin 6 (IL-6) is a cytokine that signals via the JAK-STAT pathway and is implicated in cancer and inflammation. To identify modulators of this pathway, we screened a chemical collection against an IL-6 responsive cell line stably expressing a beta-lactamase reporter gene fused to a sis-inducible element (SIE-bla cells). This assay was optimized for a 1536-well microplate format and screened against 11 693 small molecules using quantitative high-throughput screening (qHTS), a method that assays a chemical library at multiple concentrations to generate titration-response profiles for each compound. The qHTS recovered 564 actives with well-fit curves that clustered into 32 distinct chemical series of 13 activators and 19 inhibitors. A retrospective analysis of the qHTS data indicated that single concentration data at 1.5 and 7.7 microM scored 35 and 71% of qHTS actives, respectively, as inactive and were therefore false negatives. Following counter screens to identify fluorescent and non-selective series, we found four activator and one inhibitor series that modulated SIE-bla cells but did not show similar activity in reporter gene assays induced by EGF and hypoxia. Small molecules within these series will make useful tool compounds to investigate IL-6 signaling mediated by JAK-STAT activation.
Collapse
Affiliation(s)
- Ronald L Johnson
- NIH Chemical Genomics Center, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Carlson CB, Robers MB, Vogel KW, Machleidt T. Development of LanthaScreen cellular assays for key components within the PI3K/AKT/mTOR pathway. ACTA ACUST UNITED AC 2009; 14:121-32. [PMID: 19196698 DOI: 10.1177/1087057108328132] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The PI3K/AKT/mTOR pathway is central to cell growth and survival, cell cycle regulation, and programmed cell death. Aberrant activation of this signaling cascade is linked to several disease states, and thus many components of the pathway are attractive targets for therapeutic intervention. However, the considerable degree of complexity, crosstalk, and feedback regulation that exists within the pathway (especially with respect to the regulation of mTOR and its complexes) underscores the need for a comprehensive set of cell-based assays to properly identify and characterize small-molecule modulators. Here, the development and application of time-resolved Förster resonance energy transfer (TR-FRET)-based assays to enable the phosphoprotein analysis of key pathway components in a cellular format are reported. The LanthaScreen cellular assay platform uses FRET between a terbium-labeled phosphorylation site-specific antibody and an expressed green fluorescent protein fusion of particular kinase substrate and provides an assay readout that is ratiometric, robust, and amenable to high-throughput screening applications. Assays specific for 5 different targets within the pathway are highlighted: Ser183 and Thr246 on the proline-rich AKT substrate 40 kDa (PRAS40), Ser457 on programmed cell death protein 4 (PDCD4), and Thr308 and Ser473 on AKT. Each assay was evaluated under various experimental conditions and individually optimized for performance. Known pathway agonists and a small panel of commercially available compounds were also used to complete the assay validation. Taken together, these data demonstrate the utility of a related set of cell-based assays to interrogate PI3K/AKT/mTOR signaling and provide a template for the development of similar assays for other targets.
Collapse
Affiliation(s)
- Coby B Carlson
- Invitrogen Discovery Sciences, Madison, Wisconsin 53719, USA.
| | | | | | | |
Collapse
|