1
|
Sutton MN, Glazer SE, Muzzioli R, Yang P, Gammon ST, Piwnica-Worms D. Dimerization of the 4Ig isoform of B7-H3 in tumor cells mediates enhanced proliferation and tumorigenic signaling. Commun Biol 2024; 7:21. [PMID: 38182652 PMCID: PMC10770396 DOI: 10.1038/s42003-023-05736-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 12/20/2023] [Indexed: 01/07/2024] Open
Abstract
B7-H3 (CD276) has two isoforms (2Ig and 4Ig), no confirmed cognate receptor, and physiological functions that remain elusive. While differentially expressed on many solid tumors correlating with poor survival, mechanisms of how B7-H3 signals in cis (tumor cell) versus in trans (immune cell co-regulator) to elicit pro-tumorigenic phenotypes remain poorly defined. Herein, we characterized a tumorigenic and signaling role for tumor cell-expressed 4Ig-B7-H3, the dominant human isoform, in gynecological cancers that could be abrogated upon CRISPR/Cas9 knockout of B7-H3; tumorigenesis was rescued upon re-expression of 4Ig-B7-H3. Size exclusion chromatography revealed dimerization states for the extracellular domains of both human 4Ig- and murine 2Ig-B7-H3. mEGFP lifetimes of expressed 4Ig-B7-H3-mEGFP fusions determined by FRET-FLIM assays confirmed close-proximity interactions of 4Ig-B7-H3 and identified two distinct homo-FRET lifetime populations, consistent with monomeric and homo-dimer interactions. In live cells, bioluminescence imaging of 4Ig-B7-H3-mediated split luciferase complementation showed dimerization of 4Ig-B7-H3. To separate basal from dimer state activities in the absence of a known receptor, C-terminus (cytosolic) chemically-induced dimerization of 4Ig-B7-H3 increased tumor cell proliferation and cell activation signaling pathways (AKT, Jak/STAT, HIF1α, NF-κβ) significantly above basal expression of 4Ig-B7-H3 alone. These results revealed a new, dimerization-dependent intrinsic tumorigenic signaling role for 4Ig-B7-H3, likely acting in cis, and provide a therapeutically-actionable target for intervention of B7-H3-dependent tumorigenesis.
Collapse
Affiliation(s)
- Margie N Sutton
- Department of Cancer Systems Imaging, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Sarah E Glazer
- Department of Cancer Systems Imaging, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Riccardo Muzzioli
- Department of Cancer Systems Imaging, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ping Yang
- Department of Cancer Systems Imaging, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Seth T Gammon
- Department of Cancer Systems Imaging, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - David Piwnica-Worms
- Department of Cancer Systems Imaging, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
2
|
Lo CH. Recent advances in cellular biosensor technology to investigate tau oligomerization. Bioeng Transl Med 2021; 6:e10231. [PMID: 34589603 PMCID: PMC8459642 DOI: 10.1002/btm2.10231] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 12/12/2022] Open
Abstract
Tau is a microtubule binding protein which plays an important role in physiological functions but it is also involved in the pathogenesis of Alzheimer's disease and related tauopathies. While insoluble and β-sheet containing tau neurofibrillary tangles have been the histopathological hallmark of these diseases, recent studies suggest that soluble tau oligomers, which are formed prior to fibrils, are the primary toxic species. Substantial efforts have been made to generate tau oligomers using purified recombinant protein strategies to study oligomer conformations as well as their toxicity. However, no specific toxic tau species has been identified to date, potentially due to the lack of cellular environment. Hence, there is a need for cell-based models for direct monitoring of tau oligomerization and aggregation. This review will summarize the recent advances in the cellular biosensor technology, with a focus on fluorescence resonance energy transfer, bimolecular fluorescence complementation, and split luciferase complementation approaches, to monitor formation of tau oligomers and aggregates in living cells. We will discuss the applications of the cellular biosensors in examining the heterogeneous tau conformational ensembles and factors affecting tau self-assembly, as well as detecting cell-to-cell propagation of tau pathology. We will also compare the advantages and limitations of each type of tau biosensors, and highlight their translational applications in biomarker development and therapeutic discovery.
Collapse
Affiliation(s)
- Chih Hung Lo
- Department of Neurology, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
3
|
Functional Imaging Using Bioluminescent Reporter Genes in Living Subjects. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00004-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
4
|
Liu Y, Jiang J, Lepik B, Zhang Y, Zinn KR, Frank SJ. Subdomain 2, Not the Transmembrane Domain, Determines the Dimerization Partner of Growth Hormone Receptor and Prolactin Receptor. Endocrinology 2017; 158:3235-3248. [PMID: 28977606 PMCID: PMC5659695 DOI: 10.1210/en.2017-00469] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 07/27/2017] [Indexed: 01/09/2023]
Abstract
Growth hormone receptor (GHR) and prolactin (PRL) receptor (PRLR) are homologous transmembrane class I cytokine receptors. In humans, GH interacts with GHR homodimers or PRLR homodimers and PRL interacts with only PRLR homodimers to promote signaling. In human breast cancer cells endogenously expressing both receptors, GHR and PRLR specifically coimmunoprecipitate. We previously devised a split luciferase complementation assay to study GHR and PRLR assemblages. In this technique, firefly luciferase is split into two fragments (N- and C-terminal fragments of the luciferase), each without enzyme activity and tethered to the tails of two receptors. The fragments restore luciferase activity when brought close to each other by the receptors. Real-time ligand-induced complementation changes reflect the arrangement of receptors and indicate that GHR/PRLR is arranged as a heteromultimer comprised of GHR-GHR homodimers and PRLR-PRLR homodimers. We now dissect determinants for GHR and PRLR homodimerization versus heteroassociation. GHR and PRLR have extracellular domains comprised of the ligand-binding N-terminal subdomain 1 and a membrane-proximal subdomain 2 (S2), which fosters receptor-receptor contact. Based on previous studies of S2 versus the transmembrane domain (TMD) in GHR dimerization, we constructed GHR(PRLRS2), GHR(PRLRS2-TMD), and GHR(PRLRTMD), replacing GHR's S2 alone, S2 plus TMD, and TMD alone with PRLR's counterpart. We tested by complementation the ability of these chimeras and GHR or PRLR to homodimerize or heteroassociate. Comparing various combinations, we found GHR(PRLRS2) and GHR(PRLRS2-TMD) behaved as PRLR, whereas GHR(PRLRTMD) behaved as GHR regarding their dimerization partners. We conclude that S2 of GHR and PRLR, rather than their TMDs, determines their dimerization partner.
Collapse
Affiliation(s)
- Ying Liu
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Jing Jiang
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Bradford Lepik
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Yue Zhang
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Kurt R. Zinn
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Stuart J. Frank
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, Alabama 35294
- Endocrinology Section, Medical Service, Veterans Affairs Medical Center, Birmingham, Alabama 35233
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294
| |
Collapse
|
5
|
Hsu WCJ, Wildburger NC, Haidacher SJ, Nenov MN, Folorunso O, Singh AK, Chesson BC, Franklin WF, Cortez I, Sadygov RG, Dineley KT, Rudra JS, Taglialatela G, Lichti CF, Denner L, Laezza F. PPARgamma agonists rescue increased phosphorylation of FGF14 at S226 in the Tg2576 mouse model of Alzheimer's disease. Exp Neurol 2017; 295:1-17. [PMID: 28522250 DOI: 10.1016/j.expneurol.2017.05.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 04/13/2017] [Accepted: 05/13/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Cognitive impairment in humans with Alzheimer's disease (AD) and in animal models of Aβ-pathology can be ameliorated by treatments with the nuclear receptor peroxisome proliferator-activated receptor-gamma (PPARγ) agonists, such as rosiglitazone (RSG). Previously, we demonstrated that in the Tg2576 animal model of AD, RSG treatment rescued cognitive deficits and reduced aberrant activity of granule neurons in the dentate gyrus (DG), an area critical for memory formation. METHODS We used a combination of mass spectrometry, confocal imaging, electrophysiology and split-luciferase assay and in vitro phosphorylation and Ingenuity Pathway Analysis. RESULTS Using an unbiased, quantitative nano-LC-MS/MS screening, we searched for potential molecular targets of the RSG-dependent rescue of DG granule neurons. We found that S226 phosphorylation of fibroblast growth factor 14 (FGF14), an accessory protein of the voltage-gated Na+ (Nav) channels required for neuronal firing, was reduced in Tg2576 mice upon treatment with RSG. Using confocal microscopy, we confirmed that the Tg2576 condition decreased PanNav channels at the AIS of the DG, and that RSG treatment of Tg2576 mice reversed the reduction in PanNav channels. Analysis from previously published data sets identified correlative changes in action potential kinetics in RSG-treated T2576 compared to untreated and wildtype controls. In vitro phosphorylation and mass spectrometry confirmed that the multifunctional kinase GSK-3β, a downstream target of insulin signaling highly implicated in AD, phosphorylated FGF14 at S226. Assembly of the FGF14:Nav1.6 channel complex and functional regulation of Nav1.6-mediated currents by FGF14 was impaired by a phosphosilent S226A mutation. Bioinformatics pathway analysis of mass spectrometry and biochemistry data revealed a highly interconnected network encompassing PPARγ, FGF14, SCN8A (Nav 1.6), and the kinases GSK-3 β, casein kinase 2β, and ERK1/2. CONCLUSIONS These results identify FGF14 as a potential PPARγ-sensitive target controlling Aβ-induced dysfunctions of neuronal activity in the DG underlying memory loss in early AD.
Collapse
Affiliation(s)
- Wei-Chun J Hsu
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Biochemistry and Molecular Biology Graduate Program, Graduate School of Biomedical Sciences, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; M.D./Ph.D. Combined Degree Program, Graduate School of Biomedical Sciences, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Norelle C Wildburger
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Neuroscience Graduate Program, Graduate School of Biomedical Sciences, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Department of Neurology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, United States
| | - Sigmund J Haidacher
- Department of Internal Medicine, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Miroslav N Nenov
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Oluwarotimi Folorunso
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Aditya K Singh
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Brent C Chesson
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Whitney F Franklin
- Neuroscience Graduate Program, Graduate School of Biomedical Sciences, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Department of Neurology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Ibdanelo Cortez
- Neuroscience Graduate Program, Graduate School of Biomedical Sciences, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Rovshan G Sadygov
- Biochemistry and Molecular Biology Graduate Program, Graduate School of Biomedical Sciences, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Sealy Center for Molecular Medicine, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Kelly T Dineley
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Department of Neurology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Center for Addiction Research, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Jay S Rudra
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Giulio Taglialatela
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Department of Neurology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Cheryl F Lichti
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Larry Denner
- Sealy Center for Molecular Medicine, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Department of Internal Medicine, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Center for Addiction Research, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Fernanda Laezza
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Center for Addiction Research, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Center for Biomedical Engineering, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States.
| |
Collapse
|
6
|
Smirnova DV, Ugarova NN. Firefly Luciferase-based Fusion Proteins and their Applications in Bioanalysis. Photochem Photobiol 2016; 93:436-447. [PMID: 27796044 DOI: 10.1111/php.12656] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 08/31/2016] [Indexed: 12/19/2022]
Abstract
Firefly luciferase is widely used in molecular biology and bioanalytical systems as a reporter molecule due to the high quantum yield of the bioluminescence, availability of stable mutant forms of the enzyme with prescribed spectral characteristics and abundance of bacterial expression systems suitable for production of recombinant proteins in limitless quantities. In this review, we described fusion proteins of luciferase with biotin-binding domain and streptavidin, with proteins A and G, antibodies, with DNA- and RNA-binding proteins, as well as fusion proteins designed for BRET systems. The firefly luciferase-based fusion proteins are represented as an effective tool for the development of different bioanalytical systems such as (1) systems in which luciferase is attached to the surface of the target and the bioluminescence signal is detected from the specific complexes formed; (2) BRET-based systems, in which the specific interaction induces changes in the bioluminescence spectrum; and (3) systems that use modified or split luciferases, in which the luciferase activity changes under the action of the analyte. All these systems have wide application in biochemical analysis of physiologically important compounds, for the detection of pathogenic bacteria and viruses, for evaluation of protein-protein interactions, assaying of metabolites involved in cell communication and cell signaling.
Collapse
Affiliation(s)
- Daria V Smirnova
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| | - Natalia N Ugarova
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
7
|
Liu Y, Zhang Y, Jiang J, Lobie PE, Paulmurugan R, Langenheim JF, Chen WY, Zinn KR, Frank SJ. GHR/PRLR Heteromultimer Is Composed of GHR Homodimers and PRLR Homodimers. Mol Endocrinol 2016; 30:504-517. [PMID: 27003442 PMCID: PMC4853563 DOI: 10.1210/me.2015-1319] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 03/17/2016] [Indexed: 01/04/2023] Open
Abstract
GH receptor (GHR) and prolactin (PRL) receptor (PRLR) are homologous transmembrane cytokine receptors. Each prehomodimerizes and ligand binding activates Janus Kinase 2 (JAK2)-signal transducer and activator of transcription (STAT) signaling pathways by inducing conformational changes within receptor homodimers. In humans, GHR is activated by GH, whereas PRLR is activated by both GH and PRL. We previously devised a split luciferase complementation assay, in which 1 receptor is fused to an N-terminal luciferase (Nluc) fragment, and the other receptor is fused to a C-terminal luciferase (Cluc) fragment. When receptors approximate, luciferase activity (complementation) results. Using this assay, we reported ligand-independent GHR-GHR complementation and GH-induced complementation changes characterized by acute augmentation above basal signal, consistent with induction of conformational changes that bring GHR cytoplasmic tails closer. We also demonstrated association between GHR and PRLR in T47D human breast cancer cells by coimmunoprecipitation, suggesting that, in addition to forming homodimers, these receptors form hetero-assemblages with functional consequences. We now extend these analyses to examine basal and ligand-induced complementation of coexpressed PRLR-Nluc and PRLR-Cluc chimeras and coexpressed GHR-Nluc and PRLR-Cluc chimeras. We find that PRLR-PRLR and GHR-PRLR form specifically interacting ligand-independent assemblages and that either GH or PRL augments PRLR-PRLR complementation, much like the GH-induced changes in GHR-GHR dimers. However, in contrast to the complementation patterns for GHR-GHR or PRLR-PRLR homomers, both GH and PRL caused decline in luciferase activity for GHR-PRLR heteromers. These and other data suggest that GHR and PRLR associate in complexes comprised of GHR-GHR/PRLR-PRLR heteromers consisting of GHR homodimers and PRLR homodimers, rather than GHR-PRLR heterodimers.
Collapse
Affiliation(s)
- Ying Liu
- Department of Medicine (Y.L., Y.Z., J.J., S.J.F.), Division of Endocrinology, Diabetes, and Metabolism; Department of Radiology (K.R.Z.); and Department of Cell, Developmental, and Integrative Biology (S.J.F.), University of Alabama at Birmingham, Birmingham, Alabama 35294; Cancer Science Institute of Singapore and Department of Pharmacology (P.E.L.), National University of Singapore, Singapore 119077; Department of Radiology (R.P.), Stanford University School of Medicine, Palo Alto, California 94304; Department of Biological Sciences (J.F.L., W.Y.C.), Clemson University, Clemson, South Carolina 29634; and Endocrinology Section (S.J.F.), Medical Service, Veterans Affairs Medical Center, Birmingham, Alabama 35233
| | - Yue Zhang
- Department of Medicine (Y.L., Y.Z., J.J., S.J.F.), Division of Endocrinology, Diabetes, and Metabolism; Department of Radiology (K.R.Z.); and Department of Cell, Developmental, and Integrative Biology (S.J.F.), University of Alabama at Birmingham, Birmingham, Alabama 35294; Cancer Science Institute of Singapore and Department of Pharmacology (P.E.L.), National University of Singapore, Singapore 119077; Department of Radiology (R.P.), Stanford University School of Medicine, Palo Alto, California 94304; Department of Biological Sciences (J.F.L., W.Y.C.), Clemson University, Clemson, South Carolina 29634; and Endocrinology Section (S.J.F.), Medical Service, Veterans Affairs Medical Center, Birmingham, Alabama 35233
| | - Jing Jiang
- Department of Medicine (Y.L., Y.Z., J.J., S.J.F.), Division of Endocrinology, Diabetes, and Metabolism; Department of Radiology (K.R.Z.); and Department of Cell, Developmental, and Integrative Biology (S.J.F.), University of Alabama at Birmingham, Birmingham, Alabama 35294; Cancer Science Institute of Singapore and Department of Pharmacology (P.E.L.), National University of Singapore, Singapore 119077; Department of Radiology (R.P.), Stanford University School of Medicine, Palo Alto, California 94304; Department of Biological Sciences (J.F.L., W.Y.C.), Clemson University, Clemson, South Carolina 29634; and Endocrinology Section (S.J.F.), Medical Service, Veterans Affairs Medical Center, Birmingham, Alabama 35233
| | - Peter E Lobie
- Department of Medicine (Y.L., Y.Z., J.J., S.J.F.), Division of Endocrinology, Diabetes, and Metabolism; Department of Radiology (K.R.Z.); and Department of Cell, Developmental, and Integrative Biology (S.J.F.), University of Alabama at Birmingham, Birmingham, Alabama 35294; Cancer Science Institute of Singapore and Department of Pharmacology (P.E.L.), National University of Singapore, Singapore 119077; Department of Radiology (R.P.), Stanford University School of Medicine, Palo Alto, California 94304; Department of Biological Sciences (J.F.L., W.Y.C.), Clemson University, Clemson, South Carolina 29634; and Endocrinology Section (S.J.F.), Medical Service, Veterans Affairs Medical Center, Birmingham, Alabama 35233
| | - Ramasamy Paulmurugan
- Department of Medicine (Y.L., Y.Z., J.J., S.J.F.), Division of Endocrinology, Diabetes, and Metabolism; Department of Radiology (K.R.Z.); and Department of Cell, Developmental, and Integrative Biology (S.J.F.), University of Alabama at Birmingham, Birmingham, Alabama 35294; Cancer Science Institute of Singapore and Department of Pharmacology (P.E.L.), National University of Singapore, Singapore 119077; Department of Radiology (R.P.), Stanford University School of Medicine, Palo Alto, California 94304; Department of Biological Sciences (J.F.L., W.Y.C.), Clemson University, Clemson, South Carolina 29634; and Endocrinology Section (S.J.F.), Medical Service, Veterans Affairs Medical Center, Birmingham, Alabama 35233
| | - John F Langenheim
- Department of Medicine (Y.L., Y.Z., J.J., S.J.F.), Division of Endocrinology, Diabetes, and Metabolism; Department of Radiology (K.R.Z.); and Department of Cell, Developmental, and Integrative Biology (S.J.F.), University of Alabama at Birmingham, Birmingham, Alabama 35294; Cancer Science Institute of Singapore and Department of Pharmacology (P.E.L.), National University of Singapore, Singapore 119077; Department of Radiology (R.P.), Stanford University School of Medicine, Palo Alto, California 94304; Department of Biological Sciences (J.F.L., W.Y.C.), Clemson University, Clemson, South Carolina 29634; and Endocrinology Section (S.J.F.), Medical Service, Veterans Affairs Medical Center, Birmingham, Alabama 35233
| | - Wen Y Chen
- Department of Medicine (Y.L., Y.Z., J.J., S.J.F.), Division of Endocrinology, Diabetes, and Metabolism; Department of Radiology (K.R.Z.); and Department of Cell, Developmental, and Integrative Biology (S.J.F.), University of Alabama at Birmingham, Birmingham, Alabama 35294; Cancer Science Institute of Singapore and Department of Pharmacology (P.E.L.), National University of Singapore, Singapore 119077; Department of Radiology (R.P.), Stanford University School of Medicine, Palo Alto, California 94304; Department of Biological Sciences (J.F.L., W.Y.C.), Clemson University, Clemson, South Carolina 29634; and Endocrinology Section (S.J.F.), Medical Service, Veterans Affairs Medical Center, Birmingham, Alabama 35233
| | - Kurt R Zinn
- Department of Medicine (Y.L., Y.Z., J.J., S.J.F.), Division of Endocrinology, Diabetes, and Metabolism; Department of Radiology (K.R.Z.); and Department of Cell, Developmental, and Integrative Biology (S.J.F.), University of Alabama at Birmingham, Birmingham, Alabama 35294; Cancer Science Institute of Singapore and Department of Pharmacology (P.E.L.), National University of Singapore, Singapore 119077; Department of Radiology (R.P.), Stanford University School of Medicine, Palo Alto, California 94304; Department of Biological Sciences (J.F.L., W.Y.C.), Clemson University, Clemson, South Carolina 29634; and Endocrinology Section (S.J.F.), Medical Service, Veterans Affairs Medical Center, Birmingham, Alabama 35233
| | - Stuart J Frank
- Department of Medicine (Y.L., Y.Z., J.J., S.J.F.), Division of Endocrinology, Diabetes, and Metabolism; Department of Radiology (K.R.Z.); and Department of Cell, Developmental, and Integrative Biology (S.J.F.), University of Alabama at Birmingham, Birmingham, Alabama 35294; Cancer Science Institute of Singapore and Department of Pharmacology (P.E.L.), National University of Singapore, Singapore 119077; Department of Radiology (R.P.), Stanford University School of Medicine, Palo Alto, California 94304; Department of Biological Sciences (J.F.L., W.Y.C.), Clemson University, Clemson, South Carolina 29634; and Endocrinology Section (S.J.F.), Medical Service, Veterans Affairs Medical Center, Birmingham, Alabama 35233
| |
Collapse
|
8
|
Liu Y, Berry PA, Zhang Y, Jiang J, Lobie PE, Paulmurugan R, Langenheim JF, Chen WY, Zinn KR, Frank SJ. Dynamic analysis of GH receptor conformational changes by split luciferase complementation. Mol Endocrinol 2014; 28:1807-1819. [PMID: 25188449 PMCID: PMC4213363 DOI: 10.1210/me.2014-1153] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 08/28/2014] [Indexed: 11/19/2022] Open
Abstract
The transmembrane GH receptor (GHR) exists at least in part as a preformed homodimer on the cell surface. Structural and biochemical studies suggest that GH binds GHR in a 1:2 stoichiometry to effect acute GHR conformational changes that trigger the activation of the receptor-associated tyrosine kinase, Janus kinase 2 (JAK2), and downstream signaling. Despite information about GHR-GHR association derived from elegant fluorescence resonance energy transfer/bioluminescence resonance energy transfer studies, an assessment of the dynamics of GH-induced GHR conformational changes has been lacking. To this end, we used a split luciferase complementation assay that allowed detection in living cells of specific ligand-independent GHR-GHR interaction. Furthermore, GH treatment acutely augmented complementation of enzyme activity between GHRs fused, respectively, to N- and C-terminal fragments of firefly luciferase. Analysis of the temporal pattern of GH-induced complementation changes, pharmacological manipulation, genetic alteration of JAK2 levels, and truncation of the GHR intracellular domain (ICD) tail suggested that GH acutely enhances proximity of the GHR homodimer partners independent of the presence of JAK2, phosphorylation of GHR-luciferase chimeras, or an intact ICD. However, subsequent reduction of complementation requires JAK2 kinase activity and the ICD tail. This conclusion is in contrast to existing models of the GHR activation process.
Collapse
Affiliation(s)
- Ying Liu
- Department of Medicine (Y.L., P.A.B., Y.Z., J.J., S.J.F.), Division of Endocrinology, Diabetes, and Metabolism, and Departments of Radiology (K.R.Z.), and Cell, Developmental, and Integrative Biology (S.J.F.), University of Alabama at Birmingham, Birmingham, Alabama 35294; Cancer Science Institute of Singapore and Department of Pharmacology (P.E.L.), National University of Singapore, Singapore 119077; Department of Radiology (R.P.), Stanford University School of Medicine, Palo Alto, California 94304; Department of Biological Sciences (J.F.L., W.Y.C.), Clemson University, Clemson, South Carolina 29634; and Endocrinology Section (S.J.F.), Medical Service, Veterans Affairs Medical Center, Birmingham, Alabama 35233
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Gan Y, Buckels A, Liu Y, Zhang Y, Paterson AJ, Jiang J, Zinn KR, Frank SJ. Human GH receptor-IGF-1 receptor interaction: implications for GH signaling. Mol Endocrinol 2014; 28:1841-1854. [PMID: 25211187 PMCID: PMC4213361 DOI: 10.1210/me.2014-1174] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 09/04/2014] [Indexed: 01/18/2023] Open
Abstract
GH signaling yields multiple anabolic and metabolic effects. GH binds the transmembrane GH receptor (GHR) to activate the intracellular GHR-associated tyrosine kinase, Janus kinase 2 (JAK2), and downstream signals, including signal transducer and activator of transcription 5 (STAT5) activation and IGF-1 gene expression. Some GH effects are partly mediated by GH-induced IGF-1 via IGF-1 receptor (IGF-1R), a tyrosine kinase receptor. We previously demonstrated in non-human cells that GH causes formation of a GHR-JAK2-IGF-1R complex and that presence of IGF-1R (even without IGF-1 binding) augments proximal GH signaling. In this study, we use human LNCaP prostate cancer cells as a model system to further study the IGF-1R's role in GH signaling. GH promoted JAK2 and GHR tyrosine phosphorylation and STAT5 activation in LNCaP cells. By coimmunoprecipitation and a new split luciferase complementation assay, we find that GH augments GHR/IGF-1R complex formation, which is inhibited by a Fab of an antagonistic anti-GHR monoclonal antibody. Short hairpin RNA-mediated IGF-1R silencing in LNCaP cells reduced GH-induced GHR, JAK2, and STAT5 phosphorylation. Similarly, a soluble IGF-1R extracellular domain fragment (sol IGF-1R) interacts with GHR in response to GH and blunts GH signaling. Sol IGF-1R also markedly inhibits GH-induced IGF-1 gene expression in both LNCaP cells and mouse primary osteoblast cells. On the basis of these and other findings, we propose a model in which IGF-1R augments GH signaling by allowing a putative IGF-1R-associated molecule that regulates GH signaling to access the activated GHR/JAK2 complex and envision sol IGF-1R as a dominant-negative inhibitor of this IGF-1R-mediated augmentation. Physiological implications of this new model are discussed.
Collapse
Affiliation(s)
- Yujun Gan
- Department of Medicine (Y.G., A.B., Y.L., Y.Z., A.J.P., J.J., S.J.F.), Division of Endocrinology, Diabetes, and Metabolism, and Departments of Radiology (K.R.Z.) and Cell, Developmental, and Integrative Biology (S.J.F.), University of Alabama at Birmingham, Birmingham, Alabama 35294; and Endocrinology Section (S.J.F.), Medical Service, Veterans Affairs Medical Center, Birmingham, Alabama 35233
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Diversity in genetic in vivo methods for protein-protein interaction studies: from the yeast two-hybrid system to the mammalian split-luciferase system. Microbiol Mol Biol Rev 2012; 76:331-82. [PMID: 22688816 DOI: 10.1128/mmbr.05021-11] [Citation(s) in RCA: 135] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The yeast two-hybrid system pioneered the field of in vivo protein-protein interaction methods and undisputedly gave rise to a palette of ingenious techniques that are constantly pushing further the limits of the original method. Sensitivity and selectivity have improved because of various technical tricks and experimental designs. Here we present an exhaustive overview of the genetic approaches available to study in vivo binary protein interactions, based on two-hybrid and protein fragment complementation assays. These methods have been engineered and employed successfully in microorganisms such as Saccharomyces cerevisiae and Escherichia coli, but also in higher eukaryotes. From single binary pairwise interactions to whole-genome interactome mapping, the self-reassembly concept has been employed widely. Innovative studies report the use of proteins such as ubiquitin, dihydrofolate reductase, and adenylate cyclase as reconstituted reporters. Protein fragment complementation assays have extended the possibilities in protein-protein interaction studies, with technologies that enable spatial and temporal analyses of protein complexes. In addition, one-hybrid and three-hybrid systems have broadened the types of interactions that can be studied and the findings that can be obtained. Applications of these technologies are discussed, together with the advantages and limitations of the available assays.
Collapse
|
11
|
Abstract
Bioluminescence imaging of live cells has recently been recognized as an important alternative to fluorescence imaging. Fluorescent probes are much brighter than bioluminescent probes (luciferase enzymes) and, therefore, provide much better spatial and temporal resolution and much better contrast for delineating cell structure. However, with bioluminescence imaging there is virtually no background or toxicity. As a result, bioluminescence can be superior to fluorescence for detecting and quantifying molecules and their interactions in living cells, particularly in long-term studies. Structurally diverse luciferases from beetle and marine species have been used for a wide variety of applications, including tracking cells in vivo, detecting protein-protein interactions, measuring levels of calcium and other signaling molecules, detecting protease activity, and reporting circadian clock gene expression. Such applications can be optimized by the use of brighter and variously colored luciferases, brighter microscope optics, and ultrasensitive, low-noise cameras. This article presents a review of how bioluminescence differs from fluorescence, its applications to cellular imaging, and available probes, optics, and detectors. It also gives practical suggestions for optimal bioluminescence imaging of single cells.
Collapse
|
12
|
James ML, Gambhir SS. A molecular imaging primer: modalities, imaging agents, and applications. Physiol Rev 2012; 92:897-965. [PMID: 22535898 DOI: 10.1152/physrev.00049.2010] [Citation(s) in RCA: 742] [Impact Index Per Article: 57.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Molecular imaging is revolutionizing the way we study the inner workings of the human body, diagnose diseases, approach drug design, and assess therapies. The field as a whole is making possible the visualization of complex biochemical processes involved in normal physiology and disease states, in real time, in living cells, tissues, and intact subjects. In this review, we focus specifically on molecular imaging of intact living subjects. We provide a basic primer for those who are new to molecular imaging, and a resource for those involved in the field. We begin by describing classical molecular imaging techniques together with their key strengths and limitations, after which we introduce some of the latest emerging imaging modalities. We provide an overview of the main classes of molecular imaging agents (i.e., small molecules, peptides, aptamers, engineered proteins, and nanoparticles) and cite examples of how molecular imaging is being applied in oncology, neuroscience, cardiology, gene therapy, cell tracking, and theranostics (therapy combined with diagnostics). A step-by-step guide to answering biological and/or clinical questions using the tools of molecular imaging is also provided. We conclude by discussing the grand challenges of the field, its future directions, and enormous potential for further impacting how we approach research and medicine.
Collapse
Affiliation(s)
- Michelle L James
- Molecular Imaging Program, Department of Radiology, Stanford University, Palo Alto, CA 94305, USA
| | | |
Collapse
|
13
|
Yivgi-Ohana N, Eifer M, Addadi Y, Neeman M, Gross A. Utilizing mitochondrial events as biomarkers for imaging apoptosis. Cell Death Dis 2011; 2:e166. [PMID: 21633389 PMCID: PMC3168990 DOI: 10.1038/cddis.2011.47] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 04/18/2011] [Accepted: 04/19/2011] [Indexed: 01/20/2023]
Abstract
Cells undergoing apoptosis show a plethora of time-dependent changes. The available tools for imaging apoptosis in live cells rely either on the detection of the activity of caspases, or on the visualization of exposure of phosphatidyl serine in the outer leaflet of the cell membrane. We report here a novel method for the detection of mitochondrial events during apoptosis, namely translocation of Bax to mitochondria and release of cytochrome c (Cyt c) using bimolecular fluorescence complementation. Expression of split yellow fluorescent protein (YFP) fragments fused to Bax and Cyt c, resulted in robust induction of YFP fluorescence at the mitochondria of apoptotic cells with very low background. In vivo expression of split YFP protein fragments in liver hepatocytes and intra-vital imaging of subcutaneous tumor showed elevated YFP fluorescence upon apoptosis induction. Thus, YFP complementation could be applied for high-throughput screening and in vivo molecular imaging of mitochondrial events during apoptosis.
Collapse
Affiliation(s)
- N Yivgi-Ohana
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - M Eifer
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Y Addadi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - M Neeman
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - A Gross
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
14
|
Pétrin D, Hébert TE. Imaging-based approaches to understanding g protein-coupled receptor signalling complexes. Methods Mol Biol 2011; 756:37-60. [PMID: 21870219 DOI: 10.1007/978-1-61779-160-4_2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
In the last 10 years, imaging assays based on resonance energy transfer (RET) and protein fragment complementation have made it possible to study interactions between components of G protein-coupled receptor (GPCR) signalling complexes in living cells under physiological conditions. Here, we consider the history of such approaches, the current tools available and how they have changed our understanding of GPCR signalling. We also discuss some theoretical and methodological issues important when combining the different types of assay.
Collapse
Affiliation(s)
- Darlaine Pétrin
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
| | | |
Collapse
|
15
|
Lajoie P, Snapp EL. Formation and toxicity of soluble polyglutamine oligomers in living cells. PLoS One 2010; 5:e15245. [PMID: 21209946 PMCID: PMC3011017 DOI: 10.1371/journal.pone.0015245] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2010] [Accepted: 11/16/2010] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Aggregation and cytotoxicity of mutant proteins containing an expanded number of polyglutamine (polyQ) repeats is a hallmark of several diseases, including Huntington's disease (HD). Within cells, mutant Huntingtin (mHtt) and other polyglutamine expansion mutant proteins exist as monomers, soluble oligomers, and insoluble inclusion bodies (IBs). Determining which of these forms constitute a toxic species has proven difficult. Recent studies support a role for IBs as a cellular coping mechanism to sequester levels of potentially toxic soluble monomeric and oligomeric species of mHtt. METHODOLOGY/PRINCIPAL FINDINGS When fused to a fluorescent reporter (GFP) and expressed in cells, the soluble monomeric and oligomeric polyglutamine species are visually indistinguishable. Here, we describe two complementary biophysical fluorescence microscopy techniques to directly detect soluble polyglutamine oligomers (using Htt exon 1 or Htt(ex1)) and monitor their fates in live cells. Photobleaching analyses revealed a significant reduction in the mobilities of mHtt(ex1) variants consistent with their incorporation into soluble microcomplexes. Similarly, when fused to split-GFP constructs, both wildtype and mHtt(ex1) formed oligomers, as evidenced by the formation of a fluorescent reporter. Only the mHtt(ex1) split-GFP oligomers assembled into IBs. Both FRAP and split-GFP approaches confirmed the ability of mHtt(ex1) to bind and incorporate wildtype Htt into soluble oligomers. We exploited the irreversible binding of split-GFP fragments to forcibly increase levels of soluble oligomeric mHtt(ex1). A corresponding increase in the rate of IBs formation and the number formed was observed. Importantly, higher levels of soluble mHtt(ex1) oligomers significantly correlated with increased mutant cytotoxicity, independent of the presence of IBs. CONCLUSIONS/SIGNIFICANCE Our study describes powerful and sensitive tools for investigating soluble oligomeric forms of expanded polyglutamine proteins, and their impact on cell viability. Moreover, these methods should be applicable for the detection of soluble oligomers of a wide variety of aggregation prone proteins.
Collapse
Affiliation(s)
- Patrick Lajoie
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Erik Lee Snapp
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| |
Collapse
|
16
|
Michelini E, Cevenini L, Mezzanotte L, Coppa A, Roda A. Cell-based assays: fuelling drug discovery. Anal Bioanal Chem 2010; 398:227-38. [PMID: 20623273 DOI: 10.1007/s00216-010-3933-z] [Citation(s) in RCA: 131] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Revised: 06/16/2010] [Accepted: 06/16/2010] [Indexed: 12/15/2022]
Abstract
It has been estimated that over a billion dollars in resources can be consumed to obtain clinical approval, and only a few new chemical entities are approved by the US Food and Drug Administration (FDA) each year. Therefore it is of utmost importance to obtain the maximum amount of information about biological activity, toxicological profile, biochemical mechanisms, and off-target interactions of drug-candidate leads in the earliest stages of drug discovery. Cell-based assays, because of their peculiar advantages of predictability, possibility of automation, multiplexing, and miniaturization, seem the most appealing tool for the high demands of the early stages of the drug-discovery process. Nevertheless, cellular screening, relying on different strategies ranging from reporter gene technology to protein fragment complementation assays, still presents a variety of challenges. This review focuses on main advantages and limitations of different cell-based approaches, and future directions and trends in this fascinating field.
Collapse
Affiliation(s)
- Elisa Michelini
- Department of Pharmaceutical Sciences, University of Bologna, Via Mentana, 7, 40126 Bologna, Italy
| | | | | | | | | |
Collapse
|
17
|
Yang RYC, Yang KS, Pike LJ, Marshall GR. Targeting the dimerization of epidermal growth factor receptors with small-molecule inhibitors. Chem Biol Drug Des 2010; 76:1-9. [PMID: 20456371 PMCID: PMC2906783 DOI: 10.1111/j.1747-0285.2010.00986.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The epidermal growth factor (EGF) receptor is a receptor tyrosine kinase involved in the control of cell proliferation, and its overexpression is strongly associated with a variety of aggressive cancers. For example, 70-80% of metaplastic (cancer cells of mixed type) breast carcinomas overexpress EGF receptors. In addition, the EGF receptor is a highly significant contributor to common brain tumors (glioblastoma multiforme), both in initiation and progression (Huang P.H., Xu A.M., White F.M. (2009) Oncogenic EGFR signaling networks in glioma. Sci Signal;2:re6.). Brain metastases, an unmet medical need, are also common in metastatic cancer associated with overexpression of EGF receptors. Formation of EGF receptor homodimers is essential for kinase activation and was the basis for exploring direct inhibition of EGF receptor activation by blocking dimerization with small molecules. While inhibitors of protein/protein interactions are often considered difficult therapeutic targets, NSC56452, initially identified by virtual screening, was shown experimentally to inhibit EGF receptor kinase activation in a dose-dependent manner. This compound blocked EGF-stimulated dimer formation as measured by chemical cross-linking and luciferase fragment complementation. The compound was further shown to inhibit the growth of HeLa cells. This first-generation lead compound represents the first drug-like, small-molecule inhibitor of EGF receptor activation that is not directed against the intracellular kinase domain.
Collapse
Affiliation(s)
- Robert Y. C. Yang
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis School of Medicine. St. Louis, MO 63110
| | - Katherine S. Yang
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis School of Medicine. St. Louis, MO 63110
| | - Linda J. Pike
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis School of Medicine. St. Louis, MO 63110
| | - Garland R. Marshall
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis School of Medicine. St. Louis, MO 63110
| |
Collapse
|
18
|
Ketteler R. The Feynman trajectories: determining the path of a protein using fixed-endpoint assays. ACTA ACUST UNITED AC 2010; 15:321-6. [PMID: 20130209 DOI: 10.1177/1087057109357116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Richard Feynman postulated in 1948 that the path of an electron can be best described by the sum or functional integral of all possible trajectories rather than by the notion of a single, unique trajectory. As a consequence, the position of an electron does not harbor any information about the paths that contributed to this position. This observation constitutes a classical endpoint observation. The endpoint assay is the desired type of experiment for high-throughput screening applications, mainly because of limitations in data acquisition and handling. Quite contrary to electrons, it is possible to extract information about the path of a protein using endpoint assays, and these types of applications are reviewed in this article.
Collapse
Affiliation(s)
- Robin Ketteler
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom.
| |
Collapse
|
19
|
Broome AM, Bhavsar N, Ramamurthy G, Newton G, Basilion JP. Expanding the utility of beta-galactosidase complementation: piece by piece. Mol Pharm 2010; 7:60-74. [PMID: 19899815 PMCID: PMC2835542 DOI: 10.1021/mp900188e] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The ability to image and quantify multiple biomarkers in disease necessitates the development of split reporter fragment platforms. We have divided the beta-galactosidase enzyme into unique, independent polypeptides that are able to reassemble and complement enzymatic activity in bacteria and in mammalian cells. We created two sets of complementing pairs that individually have no enzymatic activity. However, when brought into close geometric proximity, the complementing pairs associated resulting in detectable enzymatic activity. We then constructed a stable ligand complex composed of reporter fragment, linker, and targeting moiety. The targeting moiety, in this case a ligand, allowed cell surface receptor targeting in vitro. Further, we were able to simultaneously visualize two cell surface receptors implicated in cancer development, epidermal growth factor receptor and transferrin receptor, using complementing pairs of the ligand-reporter fragment complex.
Collapse
Affiliation(s)
- Ann-Marie Broome
- Department of Biomedical Engineering, Case Center for Imaging Research and National Foundation for Cancer Research Center for Molecular Imaging, Case Western Reserve University
- Department of Radiology, Case Center for Imaging Research and National Foundation for Cancer Research Center for Molecular Imaging, Case Western Reserve University
- Department of Dermatology, Case Western Reserve University
| | - Nihir Bhavsar
- Department of Biomedical Engineering, Case Center for Imaging Research and National Foundation for Cancer Research Center for Molecular Imaging, Case Western Reserve University
| | - Gopal Ramamurthy
- Department of Radiology, Case Center for Imaging Research and National Foundation for Cancer Research Center for Molecular Imaging, Case Western Reserve University
| | - Gail Newton
- Department of Pathology at Harvard Medical School, Brigham and Women’s Hospital, Boston, Massachusetts
| | - James P. Basilion
- Department of Biomedical Engineering, Case Center for Imaging Research and National Foundation for Cancer Research Center for Molecular Imaging, Case Western Reserve University
- Department of Radiology, Case Center for Imaging Research and National Foundation for Cancer Research Center for Molecular Imaging, Case Western Reserve University
| |
Collapse
|
20
|
Morell M, Ventura S, Avilés FX. Protein complementation assays: Approaches for the in vivo analysis of protein interactions. FEBS Lett 2009; 583:1684-91. [DOI: 10.1016/j.febslet.2009.03.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2009] [Accepted: 03/02/2009] [Indexed: 11/28/2022]
|
21
|
Roda A, Guardigli M, Michelini E, Mirasoli M. Bioluminescence in analytical chemistry and in vivo imaging. Trends Analyt Chem 2009. [DOI: 10.1016/j.trac.2008.11.015] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|