66 results on '"Wandless TJ"'
Search Results
2. Ubiquitin-Derived Fragment as a Peptide Linker for the Efficient Cleavage of a Target Protein from a Degron.
- Author
-
Utsugi Y, Nishimura K, Yamanaka S, Nishino K, Kosako H, Sawasaki T, Shigemori H, Wandless TJ, and Miyamae Y
- Subjects
- Proteins metabolism, Ubiquitination, Peptides metabolism, Ubiquitin metabolism, Degrons
- Abstract
The chemogenetic control of cellular protein stability using degron tags is a powerful experimental strategy in biomedical research. However, this technique requires permanent fusion of the degron to a target protein, which may interfere with the proper function of the protein. Here, we report a peptide fragment from the carboxyl terminus of ubiquitin as a cleavable linker that exhibits the slow but efficient cleavage of a degron tag via cellular deubiquitinating enzymes (DUBs). We designed a fusion protein consisting of a cleavable linker and a destabilizing domain (DD), which conditionally controls the expression and release of a target protein in a ligand-induced state, allowing the free unmodified protein to perform its function. Insertion of an AGIA epitope at the carboxyl terminus of the linker made space for the DUBs to access the site to assist the cleavage reaction when the amino terminus of the target protein caused steric hindrance. The developed system, termed a cleavable degron using ubiquitin-derived linkers (c-DUB), provides robust and tunable regulation of target proteins in their native forms. The c-DUB system is a useful tool for the regulation of proteins that have terminal sites that are essential for the proper localization and function. In addition, a mechanistic investigation using proximity labeling showed that DUBs associate with the refolded DD to reverse ubiquitination, suggesting a cellular surveillance system for distinguishing the refolded DD from misfolded proteins. The c-DUB method may benefit from this machinery so that DUBs subsequently cleave the neighboring linker.
- Published
- 2024
- Full Text
- View/download PDF
3. A cAMP Sensor Based on Ligand-Dependent Protein Stabilization.
- Author
-
Sidoli M, Chen LC, Lu AJ, Wandless TJ, and Talbot WS
- Subjects
- Animals, Cyclic AMP-Dependent Protein Kinases metabolism, Fluorescence Resonance Energy Transfer, Ligands, Zebrafish metabolism, Biosensing Techniques, Cyclic AMP metabolism
- Abstract
cAMP is a ubiquitous second messenger with many functions in diverse organisms. Current cAMP sensors, including Föster resonance energy transfer (FRET)-based and single-wavelength-based sensors, allow for real time visualization of this small molecule in cultured cells and in some cases in vivo. Nonetheless the observation of cAMP in living animals is still difficult, typically requiring specialized microscopes and ex vivo tissue processing. Here we used ligand-dependent protein stabilization to create a new cAMP sensor. This sensor allows specific and sensitive detection of cAMP in living zebrafish embryos, which may enable new understanding of the functions of cAMP in living vertebrates.
- Published
- 2022
- Full Text
- View/download PDF
4. Transient rest restores functionality in exhausted CAR-T cells through epigenetic remodeling.
- Author
-
Weber EW, Parker KR, Sotillo E, Lynn RC, Anbunathan H, Lattin J, Good Z, Belk JA, Daniel B, Klysz D, Malipatlolla M, Xu P, Bashti M, Heitzeneder S, Labanieh L, Vandris P, Majzner RG, Qi Y, Sandor K, Chen LC, Prabhu S, Gentles AJ, Wandless TJ, Satpathy AT, Chang HY, and Mackall CL
- Subjects
- Animals, Cell Line, Tumor, Cytotoxicity, Immunologic, Down-Regulation, Enhancer of Zeste Homolog 2 Protein metabolism, Epigenome, Female, Hepatocyte Nuclear Factor 1-alpha metabolism, High Mobility Group Proteins metabolism, Humans, Immunologic Memory, Lymphocyte Activation, Lymphoid Enhancer-Binding Factor 1 metabolism, Male, Mice, Neoplasms, Experimental therapy, Protein Domains, Protein Stability, Receptors, Chimeric Antigen chemistry, Receptors, Chimeric Antigen immunology, Signal Transduction, T-Lymphocytes metabolism, Transcription, Genetic, Xenograft Model Antitumor Assays, Dasatinib pharmacology, Epigenesis, Genetic, Immunotherapy, Adoptive, Receptors, Chimeric Antigen metabolism, T-Lymphocytes immunology
- Abstract
T cell exhaustion limits immune responses against cancer and is a major cause of resistance to chimeric antigen receptor (CAR)-T cell therapeutics. Using murine xenograft models and an in vitro model wherein tonic CAR signaling induces hallmark features of exhaustion, we tested the effect of transient cessation of receptor signaling, or rest, on the development and maintenance of exhaustion. Induction of rest through enforced down-regulation of the CAR protein using a drug-regulatable system or treatment with the multikinase inhibitor dasatinib resulted in the acquisition of a memory-like phenotype, global transcriptional and epigenetic reprogramming, and restored antitumor functionality in exhausted CAR-T cells. This work demonstrates that rest can enhance CAR-T cell efficacy by preventing or reversing exhaustion, and it challenges the notion that exhaustion is an epigenetically fixed state., (Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2021
- Full Text
- View/download PDF
5. A Method for Conditional Regulation of Protein Stability in Native or Near-Native Form.
- Author
-
Miyamae Y, Chen LC, Utsugi Y, Farrants H, and Wandless TJ
- Subjects
- Animals, Peptide Hydrolases metabolism, Protein Domains, Protein Stability, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Ubiquitin chemistry, Ubiquitin genetics, Protein Engineering methods, Ubiquitin metabolism
- Abstract
Here, we report a method to regulate cellular protein levels by introducing a ubiquitin variant between a destabilizing domain (DD) and the regulated protein. When produced in the absence of a stabilizing ligand the DD dominates and the entire fusion protein is processively degraded by the proteasome. In the presence of the stabilizing ligand the fusion protein is metabolically stable and becomes a substrate for abundant ubiquitin-specific proteases, liberating a native, or a near-native protein-of-interest. This technique is thus particularly useful for the study of proteins whose free N terminus is required for proper function. In addition, removal of the DD in the presence of stabilizing ligand leads to higher expression levels of regulated protein when cells experience transient exposure to a stabilizing ligand, such as in a living animal receiving a single dose of a pharmacological agent as the stabilizing ligand., Competing Interests: Declaration of Interests T.J.W. is the founder of and consultant to Obsidian Therapeutics, which is pursuing therapeutic applications of the destabilizing domains., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
6. A Novel Destabilizing Domain Based on a Small-Molecule Dependent Fluorophore.
- Author
-
Navarro R, Chen LC, Rakhit R, and Wandless TJ
- Subjects
- Animals, Mice, Microscopy, Fluorescence, NIH 3T3 Cells, Fluorescent Dyes chemistry
- Abstract
Tools that can directly regulate the activity of any protein-of-interest are valuable in the study of complex biological processes. Herein, we describe the development of a novel protein domain that exhibits small molecule-dependent stability and fluorescence based on the bilirubin-inducible fluorescent protein, UnaG. When genetically fused to any protein-of-interest, this fluorescent destabilizing domain (FDD) confers its instability to the entire fusion protein, facilitating the rapid degradation of the fusion. In the presence of its cognate ligand bilirubin (BR), the FDD fusion becomes stable and fluorescent. This new chemical genetic tool allows for rapid, reversible, and tunable control over the stability and fluorescence of a wide range of protein targets.
- Published
- 2016
- Full Text
- View/download PDF
7. A method to rapidly create protein aggregates in living cells.
- Author
-
Miyazaki Y, Mizumoto K, Dey G, Kudo T, Perrino J, Chen LC, Meyer T, and Wandless TJ
- Subjects
- Animals, Binding Sites genetics, Caenorhabditis elegans genetics, Caenorhabditis elegans metabolism, Cell Line, Tumor, Green Fluorescent Proteins chemistry, Green Fluorescent Proteins genetics, HEK293 Cells, Humans, Mice, Microscopy, Confocal, Microscopy, Fluorescence, NIH 3T3 Cells, Tacrolimus Binding Protein 1A chemistry, Tacrolimus Binding Protein 1A genetics, Time-Lapse Imaging methods, Green Fluorescent Proteins metabolism, Protein Aggregates, Protein Aggregation, Pathological, Tacrolimus Binding Protein 1A metabolism
- Abstract
The accumulation of protein aggregates is a common pathological hallmark of many neurodegenerative diseases. However, we do not fully understand how aggregates are formed or the complex network of chaperones, proteasomes and other regulatory factors involved in their clearance. Here, we report a chemically controllable fluorescent protein that enables us to rapidly produce small aggregates inside living cells on the order of seconds, as well as monitor the movement and coalescence of individual aggregates into larger structures. This method can be applied to diverse experimental systems, including live animals, and may prove valuable for understanding cellular responses and diseases associated with protein aggregates.
- Published
- 2016
- Full Text
- View/download PDF
8. Distinct transcriptional responses elicited by unfolded nuclear or cytoplasmic protein in mammalian cells.
- Author
-
Miyazaki Y, Chen LC, Chu BW, Swigut T, and Wandless TJ
- Subjects
- Animals, Cell Line, Cell Nucleus metabolism, Cytoplasm metabolism, Mice, Cell Physiological Phenomena, Gene Expression Regulation, Signal Transduction, Transcription, Genetic, Unfolded Protein Response
- Abstract
Eukaryotic cells possess a variety of signaling pathways that prevent accumulation of unfolded and misfolded proteins. Chief among these is the heat shock response (HSR), which is assumed to respond to unfolded proteins in the cytosol and nucleus alike. In this study, we probe this axiom further using engineered proteins called 'destabilizing domains', whose folding state we control with a small molecule. The sudden appearance of unfolded protein in mammalian cells elicits a robust transcriptional response, which is distinct from the HSR and other known pathways that respond to unfolded proteins. The cellular response to unfolded protein is strikingly different in the nucleus and the cytosol, although unfolded protein in either compartment engages the p53 network. This response provides cross-protection during subsequent proteotoxic stress, suggesting that it is a central component of protein quality control networks, and like the HSR, is likely to influence the initiation and progression of human pathologies.
- Published
- 2015
- Full Text
- View/download PDF
9. Using light to shape chemical gradients for parallel and automated analysis of chemotaxis.
- Author
-
Collins SR, Yang HW, Bonger KM, Guignet EG, Wandless TJ, and Meyer T
- Subjects
- Adenosine Triphosphate pharmacology, Cell Line, Tumor, Chemotactic Factors pharmacology, Dose-Response Relationship, Drug, Equipment Design, Fluoresceins analysis, Fluorescent Dyes analysis, GTP-Binding Protein alpha Subunits, Gi-Go physiology, Humans, Image Processing, Computer-Assisted, Leukemia, Myeloid, Acute pathology, Microscopy, Fluorescence methods, N-Formylmethionine Leucyl-Phenylalanine pharmacology, Neoplasm Proteins physiology, Neutrophils cytology, Neutrophils drug effects, Photochemistry, RNA, Small Interfering pharmacology, Time-Lapse Imaging, Chemotaxis drug effects, Ultraviolet Rays
- Abstract
Numerous molecular components have been identified that regulate the directed migration of eukaryotic cells toward sources of chemoattractant. However, how the components of this system are wired together to coordinate multiple aspects of the response, such as directionality, speed, and sensitivity to stimulus, remains poorly understood. Here we developed a method to shape chemoattractant gradients optically and analyze cellular chemotaxis responses of hundreds of living cells per well in 96-well format by measuring speed changes and directional accuracy. We then systematically characterized migration and chemotaxis phenotypes for 285 siRNA perturbations. A key finding was that the G-protein Giα subunit selectively controls the direction of migration while the receptor and Gβ subunit proportionally control both speed and direction. Furthermore, we demonstrate that neutrophils chemotax persistently in response to gradients of fMLF but only transiently in response to gradients of ATP. The method we introduce is applicable for diverse chemical cues and systematic perturbations, can be used to measure multiple cell migration and signaling parameters, and is compatible with low- and high-resolution fluorescence microscopy., (© 2015 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2015
- Full Text
- View/download PDF
10. Chemical biology strategies for posttranslational control of protein function.
- Author
-
Rakhit R, Navarro R, and Wandless TJ
- Subjects
- Allosteric Regulation, Protein Binding, Protein Engineering, Protein Multimerization, Protein Precursors metabolism, Protein Processing, Post-Translational, Protein Stability, Proteins chemistry, Proteins genetics, Small Molecule Libraries chemistry, Small Molecule Libraries metabolism, Proteins metabolism
- Abstract
A common strategy to understand a biological system is to selectively perturb it and observe its response. Although technologies now exist to manipulate cellular systems at the genetic and transcript level, the direct manipulation of functions at the protein level can offer significant advantages in precision, speed, and reversibility. Combining the specificity of genetic manipulation and the spatiotemporal resolution of light- and small molecule-based approaches now allows exquisite control over biological systems to subtly perturb a system of interest in vitro and in vivo. Conditional perturbation mechanisms may be broadly characterized by change in intracellular localization, intramolecular activation, or degradation of a protein-of-interest. Here we review recent advances in technologies for conditional regulation of protein function and suggest further areas of potential development., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
11. General method for regulating protein stability with light.
- Author
-
Bonger KM, Rakhit R, Payumo AY, Chen JK, and Wandless TJ
- Subjects
- Animals, Avena chemistry, Light, Mice, NIH 3T3 Cells, Protein Engineering, Protein Structure, Tertiary, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins genetics, Zebrafish, Avena genetics, Luminescent Proteins chemistry, Luminescent Proteins genetics, Photolysis, Phototropins chemistry, Phototropins genetics, Protein Stability
- Abstract
Post-translational regulation of protein abundance in cells is a powerful tool for studying protein function. Here, we describe a novel genetically encoded protein domain that is degraded upon exposure to nontoxic blue light. We demonstrate that fusion proteins containing this domain are rapidly degraded in cultured cells and in zebrafish upon illumination.
- Published
- 2014
- Full Text
- View/download PDF
12. The E3 ubiquitin ligase UBE3C enhances proteasome processivity by ubiquitinating partially proteolyzed substrates.
- Author
-
Chu BW, Kovary KM, Guillaume J, Chen LC, Teruel MN, and Wandless TJ
- Subjects
- Benzoquinones pharmacology, Gene Knockdown Techniques, Green Fluorescent Proteins metabolism, HSP90 Heat-Shock Proteins antagonists & inhibitors, HSP90 Heat-Shock Proteins metabolism, HeLa Cells, Humans, Lactams, Macrocyclic pharmacology, Proteasome Endopeptidase Complex genetics, Proteasome Endopeptidase Complex metabolism, Protein Biosynthesis drug effects, Protein Biosynthesis genetics, Ubiquitin-Protein Ligases genetics, Ubiquitination drug effects, Protein Folding, Proteolysis drug effects, Ubiquitin-Protein Ligases metabolism, Ubiquitination genetics
- Abstract
To maintain protein homeostasis, cells must balance protein synthesis with protein degradation. Accumulation of misfolded or partially degraded proteins can lead to the formation of pathological protein aggregates. Here we report the use of destabilizing domains, proteins whose folding state can be reversibly tuned using a high affinity ligand, as model substrates to interrogate cellular protein quality control mechanisms in mammalian cells using a forward genetic screen. Upon knockdown of UBE3C, an E3 ubiquitin ligase, a reporter protein consisting of a destabilizing domain fused to GFP is degraded more slowly and incompletely by the proteasome. Partial proteolysis is also observed when UBE3C is present but cannot ubiquitinate substrates because its active site has been mutated, it is unable to bind to the proteasome, or the substrate lacks lysine residues. UBE3C knockdown also results in less substrate polyubiquitination. Finally, knockdown renders cells more susceptible to the Hsp90 inhibitor 17-AAG, suggesting that UBE3C protects against the harmful accumulation of protein fragments arising from incompletely degraded proteasome substrates.
- Published
- 2013
- Full Text
- View/download PDF
13. Inducible control of gene expression with destabilized Cre.
- Author
-
Sando R 3rd, Baumgaertel K, Pieraut S, Torabi-Rander N, Wandless TJ, Mayford M, and Maximov A
- Subjects
- Animals, Humans, Mice, Recombination, Genetic drug effects, Trimethoprim pharmacology, Gene Expression Regulation drug effects, Integrases metabolism
- Abstract
Acute manipulation of gene and protein function in the brain is essential for understanding the mechanisms of nervous system development, plasticity and information processing. Here we describe a technique based on a destabilized Cre recombinase (DD-Cre) whose activity is controlled by the antibiotic trimethoprim (TMP). We show that DD-Cre triggers rapid TMP-dependent recombination of loxP-flanked ('floxed') alleles in mouse neurons in vivo and validate the use of this system for neurobehavioral research.
- Published
- 2013
- Full Text
- View/download PDF
14. Rapid and tunable control of protein stability in Caenorhabditis elegans using a small molecule.
- Author
-
Cho U, Zimmerman SM, Chen LC, Owen E, Kim JV, Kim SK, and Wandless TJ
- Subjects
- Animals, Mice, NIH 3T3 Cells, Tetrahydrofolate Dehydrogenase genetics, Tetrahydrofolate Dehydrogenase metabolism, Caenorhabditis elegans metabolism, Caenorhabditis elegans Proteins metabolism, Protein Stability
- Abstract
Destabilizing domains are conditionally unstable protein domains that can be fused to a protein of interest resulting in degradation of the fusion protein in the absence of stabilizing ligand. These engineered protein domains enable rapid, reversible and dose-dependent control of protein expression levels in cultured cells and in vivo. To broaden the scope of this technology, we have engineered new destabilizing domains that perform well at temperatures of 20-25°C. This raises the possibility that our technology could be adapted for use at any temperature. We further show that these new destabilizing domains can be used to regulate protein concentrations in C. elegans. These data reinforce that DD can function in virtually any organism and temperature.
- Published
- 2013
- Full Text
- View/download PDF
15. FK506 activates BMPR2, rescues endothelial dysfunction, and reverses pulmonary hypertension.
- Author
-
Spiekerkoetter E, Tian X, Cai J, Hopper RK, Sudheendra D, Li CG, El-Bizri N, Sawada H, Haghighat R, Chan R, Haghighat L, de Jesus Perez V, Wang L, Reddy S, Zhao M, Bernstein D, Solow-Cordero DE, Beachy PA, Wandless TJ, Ten Dijke P, and Rabinovitch M
- Subjects
- Animals, Apoptosis, Bone Morphogenetic Protein 4 physiology, Bone Morphogenetic Protein Receptors, Type II genetics, Cell Hypoxia, Cell Line, Tumor, Cell Proliferation, Endothelial Cells drug effects, Endothelium, Vascular pathology, Endothelium, Vascular physiopathology, High-Throughput Screening Assays, Humans, Hypertension, Pulmonary metabolism, Hypertension, Pulmonary pathology, Inhibitor of Differentiation Protein 1 genetics, Inhibitor of Differentiation Protein 1 metabolism, Male, Mice, Mice, Knockout, Microvessels pathology, Neointima drug therapy, Neointima metabolism, Neointima pathology, Pulmonary Artery pathology, Rats, Rats, Sprague-Dawley, Signal Transduction, Smad Proteins metabolism, Tacrolimus Binding Protein 1A metabolism, Bone Morphogenetic Protein Receptors, Type II metabolism, Endothelial Cells physiology, Hypertension, Pulmonary drug therapy, Tacrolimus pharmacology
- Abstract
Dysfunctional bone morphogenetic protein receptor-2 (BMPR2) signaling is implicated in the pathogenesis of pulmonary arterial hypertension (PAH). We used a transcriptional high-throughput luciferase reporter assay to screen 3,756 FDA-approved drugs and bioactive compounds for induction of BMPR2 signaling. The best response was achieved with FK506 (tacrolimus), via a dual mechanism of action as a calcineurin inhibitor that also binds FK-binding protein-12 (FKBP12), a repressor of BMP signaling. FK506 released FKBP12 from type I receptors activin receptor-like kinase 1 (ALK1), ALK2, and ALK3 and activated downstream SMAD1/5 and MAPK signaling and ID1 gene regulation in a manner superior to the calcineurin inhibitor cyclosporine and the FKBP12 ligand rapamycin. In pulmonary artery endothelial cells (ECs) from patients with idiopathic PAH, low-dose FK506 reversed dysfunctional BMPR2 signaling. In mice with conditional Bmpr2 deletion in ECs, low-dose FK506 prevented exaggerated chronic hypoxic PAH associated with induction of EC targets of BMP signaling, such as apelin. Low-dose FK506 also reversed severe PAH in rats with medial hypertrophy following monocrotaline and in rats with neointima formation following VEGF receptor blockade and chronic hypoxia. Our studies indicate that low-dose FK506 could be useful in the treatment of PAH.
- Published
- 2013
- Full Text
- View/download PDF
16. Par-4 downregulation promotes breast cancer recurrence by preventing multinucleation following targeted therapy.
- Author
-
Alvarez JV, Pan TC, Ruth J, Feng Y, Zhou A, Pant D, Grimley JS, Wandless TJ, Demichele A, and Chodosh LA
- Subjects
- Animals, Apoptosis Regulatory Proteins antagonists & inhibitors, Breast Neoplasms drug therapy, Calcium-Calmodulin-Dependent Protein Kinases physiology, Cardiac Myosins metabolism, Death-Associated Protein Kinases, Down-Regulation, Female, Humans, Mice, Myosin Light Chains metabolism, Phosphorylation, Receptor, ErbB-2 analysis, Tumor Suppressor Protein p53 physiology, Apoptosis Regulatory Proteins physiology, Breast Neoplasms etiology, Neoplasm Recurrence, Local etiology
- Abstract
Most deaths from breast cancer result from tumor recurrence, but mechanisms underlying tumor relapse are largely unknown. We now report that Par-4 is downregulated during tumor recurrence and that Par-4 downregulation is necessary and sufficient to promote recurrence. Tumor cells with low Par-4 expression survive therapy by evading a program of Par-4-dependent multinucleation and apoptosis that is otherwise engaged following treatment. Low Par-4 expression is associated with poor response to neoadjuvant chemotherapy and an increased risk of relapse in patients with breast cancer, and Par-4 is downregulated in residual tumor cells that survive neoadjuvant chemotherapy. Our findings identify Par-4-induced multinucleation as a mechanism of cell death in oncogene-addicted cells and establish Par-4 as a negative regulator of breast cancer recurrence., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
17. Visualizing cellular interactions with a generalized proximity reporter.
- Author
-
Sellmyer MA, Bronsart L, Imoto H, Contag CH, Wandless TJ, and Prescher JA
- Subjects
- Animals, Cell Line, Tumor, Female, HEK293 Cells, Humans, Mammary Neoplasms, Experimental genetics, Mammary Neoplasms, Experimental pathology, Cell Communication immunology, Genes, Reporter, Immunologic Surveillance, Mammary Neoplasms, Experimental immunology, Models, Biological
- Abstract
Interactions among neighboring cells underpin many physiological processes ranging from early development to immune responses. When these interactions do not function properly, numerous pathologies, including infection and cancer, can result. Molecular imaging technologies, especially optical imaging, are uniquely suited to illuminate complex cellular interactions within the context of living tissues in the body. However, no tools yet exist that allow the detection of microscopic events, such as two cells coming into close proximity, on a global, whole-animal scale. We report here a broadly applicable, longitudinal strategy for probing interactions among cells in living subjects. This approach relies on the generation of bioluminescent light when two distinct cell populations come into close proximity, with the intensity of the optical signal correlating with relative cellular location. We demonstrate the ability of this reporter strategy to gauge cell-cell proximity in culture models in vitro and then evaluate this approach for imaging tumor-immune cell interactions using a murine breast cancer model. In these studies, our imaging strategy enabled the facile visualization of features that are otherwise difficult to observe with conventional imaging techniques, including detection of micrometastatic lesions and potential sites of tumor immunosurveillance. This proximity reporter will facilitate probing of numerous types of cell-cell interactions and will stimulate the development of similar techniques to detect rare events and pathological processes in live animals.
- Published
- 2013
- Full Text
- View/download PDF
18. Networks of polarized actin filaments in the axon initial segment provide a mechanism for sorting axonal and dendritic proteins.
- Author
-
Watanabe K, Al-Bassam S, Miyazaki Y, Wandless TJ, Webster P, and Arnold DB
- Subjects
- Actin Cytoskeleton ultrastructure, Animals, Axons ultrastructure, Cells, Cultured, Dendrites ultrastructure, Microscopy, Electron, Scanning, Protein Transport physiology, Rats, Actin Cytoskeleton metabolism, Actins metabolism, Axons metabolism, Dendrites metabolism, Nerve Tissue Proteins metabolism
- Abstract
Trafficking of proteins specifically to the axonal or somatodendritic membrane allows neurons to establish and maintain polarized compartments with distinct morphology and function. Diverse evidence suggests that an actin-dependent vesicle filter within the axon initial segment (AIS) plays a critical role in polarized trafficking; however, no distinctive actin-based structures capable of comprising such a filter have been found within the AIS. Here, using correlative light and scanning electron microscopy, we visualized networks of actin filaments several microns wide within the AIS of cortical neurons in culture. Individual filaments within these patches are predominantly oriented with their plus ends facing toward the cell body, consistent with models of filter selectivity. Vesicles carrying dendritic proteins are much more likely to stop in regions occupied by the actin patches than in other regions, indicating that the patches likely prevent movement of dendritic proteins to the axon and thereby act as a vesicle filter., (Copyright © 2012 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
19. Differential trafficking of transport vesicles contributes to the localization of dendritic proteins.
- Author
-
Al-Bassam S, Xu M, Wandless TJ, and Arnold DB
- Subjects
- Animals, Biological Transport physiology, Cells, Cultured, Dendrites ultrastructure, Embryo, Mammalian, Models, Biological, Myosins metabolism, Rats, Rats, Sprague-Dawley, Single-Cell Analysis, Time-Lapse Imaging, Tissue Distribution, Axonal Transport physiology, Dendrites metabolism, Nerve Tissue Proteins metabolism, Transport Vesicles metabolism, Transport Vesicles physiology
- Abstract
In neurons, transmembrane proteins are targeted to dendrites in vesicles that traffic solely within the somatodendritic compartment. How these vesicles are retained within the somatodendritic domain is unknown. Here, we use a novel pulse-chase system, which allows synchronous release of exogenous transmembrane proteins from the endoplasmic reticulum to follow movements of post-Golgi transport vesicles. Surprisingly, we found that post-Golgi vesicles carrying dendritic proteins were equally likely to enter axons and dendrites. However, once such vesicles entered the axon, they very rarely moved beyond the axon initial segment but instead either halted or reversed direction in an actin and Myosin Va-dependent manner. In contrast, vesicles carrying either an axonal or a nonspecifically localized protein only rarely halted or reversed and instead generally proceeded to the distal axon. Thus, our results are consistent with the axon initial segment behaving as a vesicle filter that mediates the differential trafficking of transport vesicles., (Copyright © 2012 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
20. Destabilizing domains derived from the human estrogen receptor.
- Author
-
Miyazaki Y, Imoto H, Chen LC, and Wandless TJ
- Subjects
- Animals, Bacterial Proteins chemistry, Bacterial Proteins genetics, Bacterial Proteins metabolism, Binding Sites drug effects, Humans, Ligands, Luminescent Proteins chemistry, Luminescent Proteins genetics, Luminescent Proteins metabolism, Mice, Models, Molecular, Molecular Structure, Mutation, NIH 3T3 Cells, Protein Engineering, Receptors, Estrogen genetics, Receptors, Estrogen metabolism, Structure-Activity Relationship, Tamoxifen analogs & derivatives, Tamoxifen chemistry, Tamoxifen pharmacology, Receptors, Estrogen chemistry
- Abstract
Methods to rapidly and reversibly perturb the functions of specific proteins are desirable tools for studies of complex biological processes. We have demonstrated an experimental strategy to regulate the intracellular concentration of any protein of interest by using an engineered destabilizing protein domain and a cell-permeable small molecule. Destabilizing domains have general utility to confer instability to a wide range of proteins including integral transmembrane proteins. This study reports a destabilizing domain system based on the ligand binding domain of the estrogen receptor that can be regulated by one of two synthetic ligands, CMP8 or 4-hydroxytamoxifen., (© 2012 American Chemical Society)
- Published
- 2012
- Full Text
- View/download PDF
21. Imaging the impact of chemically inducible proteins on cellular dynamics in vivo.
- Author
-
Leong HS, Lizardo MM, Ablack A, McPherson VA, Wandless TJ, Chambers AF, and Lewis JD
- Subjects
- Animals, Birds embryology, Cell Line, Tumor, Diagnostic Imaging, Epithelial-Mesenchymal Transition drug effects, Female, Humans, Microscopy, Confocal, Microscopy, Fluorescence, Morpholines pharmacokinetics, Morpholines pharmacology, Transplantation, Heterologous, Vimentin metabolism, Breast Neoplasms metabolism, Cadherins metabolism
- Abstract
The analysis of dynamic events in the tumor microenvironment during cancer progression is limited by the complexity of current in vivo imaging models. This is coupled with an inability to rapidly modulate and visualize protein activity in real time and to understand the consequence of these perturbations in vivo. We developed an intravital imaging approach that allows the rapid induction and subsequent depletion of target protein levels within human cancer xenografts while assessing the impact on cell behavior and morphology in real time. A conditionally stabilized fluorescent E-cadherin chimera was expressed in metastatic breast cancer cells, and the impact of E-cadherin induction and depletion was visualized using real-time confocal microscopy in a xenograft avian embryo model. We demonstrate the assessment of protein localization, cell morphology and migration in cells undergoing epithelial-mesenchymal and mesenchymal-epithelial transitions in breast tumors. This technique allows for precise control over protein activity in vivo while permitting the temporal analysis of dynamic biophysical parameters.
- Published
- 2012
- Full Text
- View/download PDF
22. Intracellular context affects levels of a chemically dependent destabilizing domain.
- Author
-
Sellmyer MA, Chen LC, Egeler EL, Rakhit R, and Wandless TJ
- Subjects
- Cell Nucleus drug effects, Cell Nucleus metabolism, DNA-Binding Proteins genetics, Endoplasmic Reticulum drug effects, Endoplasmic Reticulum metabolism, HEK293 Cells, Humans, Intracellular Space drug effects, Mitochondria drug effects, Mitochondria metabolism, Protein Stability drug effects, Protein Structure, Tertiary, Protein Transport drug effects, RNA Splicing drug effects, RNA Splicing genetics, Recombinant Fusion Proteins metabolism, Regulatory Factor X Transcription Factors, Transcription Factors genetics, Tunicamycin pharmacology, Unfolded Protein Response drug effects, Intracellular Space metabolism, Proteins chemistry, Proteins metabolism
- Abstract
The ability to regulate protein levels in live cells is crucial to understanding protein function. In the interest of advancing the tool set for protein perturbation, we developed a protein destabilizing domain (DD) that can confer its instability to a fused protein of interest. This destabilization and consequent degradation can be rescued in a reversible and dose-dependent manner with the addition of a small molecule that is specific for the DD, Shield-1. Proteins encounter different local protein quality control (QC) machinery when targeted to cellular compartments such as the mitochondrial matrix or endoplasmic reticulum (ER). These varied environments could have profound effects on the levels and regulation of the cytoplasmically derived DD. Here we show that DD fusions in the cytoplasm or nucleus can be efficiently degraded in mammalian cells; however, targeting fusions to the mitochondrial matrix or ER lumen leads to accumulation even in the absence of Shield-1. Additionally, we characterize the behavior of the DD with perturbants that modulate protein production, degradation, and local protein QC machinery. Chemical induction of the unfolded protein response in the ER results in decreased levels of an ER-targeted DD indicating the sensitivity of the DD to the degradation environment. These data reinforce that DD is an effective tool for protein perturbation, show that the local QC machinery affects levels of the DD, and suggest that the DD may be a useful probe for monitoring protein quality control machinery.
- Published
- 2012
- Full Text
- View/download PDF
23. Ligand-switchable substrates for a ubiquitin-proteasome system.
- Author
-
Egeler EL, Urner LM, Rakhit R, Liu CW, and Wandless TJ
- Subjects
- Animals, Cell Line, Humans, Ligands, Mice, Protein Stability, Substrate Specificity, Tacrolimus Binding Protein 1A metabolism, Tacrolimus Binding Proteins, Ubiquitination, Proteasome Endopeptidase Complex metabolism, Protein Unfolding, Ubiquitin metabolism
- Abstract
Cellular maintenance of protein homeostasis is essential for normal cellular function. The ubiquitin-proteasome system (UPS) plays a central role in processing cellular proteins destined for degradation, but little is currently known about how misfolded cytosolic proteins are recognized by protein quality control machinery and targeted to the UPS for degradation in mammalian cells. Destabilizing domains (DDs) are small protein domains that are unstable and degraded in the absence of ligand, but whose stability is rescued by binding to a high affinity cell-permeable ligand. In the work presented here, we investigate the biophysical properties and cellular fates of a panel of FKBP12 mutants displaying a range of stabilities when expressed in mammalian cells. Our findings correlate observed cellular instability to both the propensity of the protein domain to unfold in vitro and the extent of ubiquitination of the protein in the non-permissive (ligand-free) state. We propose a model in which removal of stabilizing ligand causes the DD to unfold and be rapidly ubiquitinated by the UPS for degradation at the proteasome. The conditional nature of DD stability allows a rapid and non-perturbing switch from stable protein to unstable UPS substrate unlike other methods currently used to interrogate protein quality control, providing tunable control of degradation rates.
- Published
- 2011
- Full Text
- View/download PDF
24. Evaluation of FKBP and DHFR based destabilizing domains in Saccharomyces cerevisiae.
- Author
-
Rakhit R, Edwards SR, Iwamoto M, and Wandless TJ
- Subjects
- Animals, Flow Cytometry, Humans, Mice, NIH 3T3 Cells, Saccharomyces cerevisiae growth & development, Tacrolimus Binding Protein 1A chemistry, Tetrahydrofolate Dehydrogenase chemistry, Saccharomyces cerevisiae metabolism, Tacrolimus Binding Protein 1A metabolism, Tetrahydrofolate Dehydrogenase metabolism
- Abstract
Two orthogonal destabilizing domains have been developed based on mutants of human FKBP12 as well as bacterial DHFR and these engineered domains have been used to control protein concentration in a variety of contexts in vitro and in vivo. FKBP12 based destabilizing domains cannot be rescued in the yeast Saccharomyces cerevisiae; ecDHFR based destabilizing domains are not degraded as efficiently in S. cerevisiae as in mammalian cells or Plasmodium, but provide a starting point for the development of domains with increased signal-to-noise in S. cerevisiae., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
25. Small-molecule displacement of a cryptic degron causes conditional protein degradation.
- Author
-
Bonger KM, Chen LC, Liu CW, and Wandless TJ
- Subjects
- Amino Acid Sequence, Animals, Bacterial Proteins, Catalytic Domain, Cell Line, Cloning, Molecular, Gene Expression Regulation physiology, Luminescent Proteins, Mice, Models, Molecular, Morpholines pharmacology, Peptides metabolism, Protein Structure, Tertiary, Tacrolimus Binding Proteins genetics, Tacrolimus Binding Proteins metabolism, Protein Denaturation, Tacrolimus Binding Proteins chemistry
- Abstract
The ability to rapidly regulate the functions of specific proteins in living cells is a valuable tool for biological research. Here we describe a new technique by which the degradation of a specific protein is induced by a small molecule. A protein of interest is fused to a ligand-induced degradation (LID) domain, resulting in the expression of a stable and functional fusion protein. The LID domain is comprised of the FK506- and rapamycin-binding protein (FKBP) and a 19-amino-acid degron fused to the C terminus of FKBP. In the absence of the small molecule Shield-1, the degron is bound to the FKBP fusion protein and the protein is stable. When present, Shield-1 binds tightly to FKBP, displacing the degron and inducing rapid and processive degradation of the LID domain and any fused partner protein. Structure-function studies of the 19-residue peptide showed that a 4-amino-acid sequence within the peptide is responsible for degradation.
- Published
- 2011
- Full Text
- View/download PDF
26. Chemical control of FGF-2 release for promoting calvarial healing with adipose stem cells.
- Author
-
Kwan MD, Sellmyer MA, Quarto N, Ho AM, Wandless TJ, and Longaker MT
- Subjects
- Animals, Fibroblast Growth Factor 2 genetics, Male, Mice, Mice, Nude, Tissue Engineering, Tissue Scaffolds, Transplantation, Homologous, Adipocytes metabolism, Adipocytes transplantation, Fibroblast Growth Factor 2 biosynthesis, Fracture Healing, Skull injuries, Skull Fractures therapy, Stem Cell Transplantation, Stem Cells metabolism
- Abstract
Chemical control of protein secretion using a small molecule approach provides a powerful tool to optimize tissue engineering strategies by regulating the spatial and temporal dimensions that are exposed to a specific protein. We placed fibroblast growth factor 2 (FGF-2) under conditional control of a small molecule and demonstrated greater than 50-fold regulation of FGF-2 release as well as tunability, reversibility, and functionality in vitro. We then applied conditional control of FGF-2 secretion to a cell-based, skeletal tissue engineering construct consisting of adipose stem cells (ASCs) on a biomimetic scaffold to promote bone formation in a murine critical-sized calvarial defect model. ASCs are an easily harvested and abundant source of postnatal multipotent cells and have previously been demonstrated to regenerate bone in critical-sized defects. These results suggest that chemically controlled FGF-2 secretion can significantly increase bone formation by ASCs in vivo. This study represents a novel approach toward refining protein delivery for tissue engineering applications.
- Published
- 2011
- Full Text
- View/download PDF
27. Asparagine repeat function in a Plasmodium falciparum protein assessed via a regulatable fluorescent affinity tag.
- Author
-
Muralidharan V, Oksman A, Iwamoto M, Wandless TJ, and Goldberg DE
- Subjects
- Erythrocytes cytology, Erythrocytes drug effects, Erythrocytes parasitology, Genes, Essential genetics, Mutant Proteins chemistry, Mutant Proteins metabolism, Plasmodium falciparum drug effects, Proteasome Endopeptidase Complex metabolism, Protein Stability drug effects, Protein Structure, Tertiary, Protozoan Proteins genetics, Recombinant Proteins metabolism, Reproducibility of Results, Sequence Deletion genetics, Trimethoprim pharmacology, Ubiquitination drug effects, Asparagine metabolism, Fluorescent Dyes metabolism, Plasmodium falciparum metabolism, Protozoan Proteins chemistry, Protozoan Proteins metabolism, Repetitive Sequences, Amino Acid
- Abstract
One in four proteins in Plasmodium falciparum contains asparagine repeats. We probed the function of one such 28-residue asparagine repeat present in the P. falciparum proteasome lid subunit 6, Rpn6. To aid our efforts, we developed a regulatable, fluorescent affinity (RFA) tag that allows cellular localization, manipulation of cellular levels, and affinity isolation of a chosen protein in P. falciparum. The tag comprises a degradation domain derived from Escherichia coli dihydrofolate reductase together with GFP. The expression of RFA-tagged proteins is regulated by the simple folate analog trimethoprim (TMP). Parasite lines were generated in which full-length Rpn6 and an asparagine repeat-deletion mutant of Rpn6 were fused to the RFA tag. The knockdown of Rpn6 upon removal of TMP revealed that this protein is essential for ubiquitinated protein degradation and for parasite survival, but the asparagine repeat is dispensable for protein expression, stability, and function. The data point to a genomic mechanism for repeat perpetuation rather than a positive cellular role. The RFA tag should facilitate study of the role of essential genes in parasite biology.
- Published
- 2011
- Full Text
- View/download PDF
28. A general chemical method to regulate protein stability in the mammalian central nervous system.
- Author
-
Iwamoto M, Björklund T, Lundberg C, Kirik D, and Wandless TJ
- Subjects
- Animals, Cell Line, Escherichia coli enzymology, Folic Acid Antagonists pharmacology, Ligands, Mice, Protein Engineering, Protein Stability, Protein Structure, Tertiary, Rats, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins metabolism, Tacrolimus Binding Proteins chemistry, Tacrolimus Binding Proteins metabolism, Tetrahydrofolate Dehydrogenase genetics, Tetrahydrofolate Dehydrogenase metabolism, Trimethoprim pharmacology, Brain metabolism, Folic Acid Antagonists chemistry, Tetrahydrofolate Dehydrogenase chemistry, Trimethoprim chemistry
- Abstract
The ability to make specific perturbations to biological molecules in a cell or organism is a central experimental strategy in modern research biology. We have developed a general technique in which the stability of a specific protein is regulated by a cell-permeable small molecule. Mutants of the Escherichia coli dihydrofolate reductase (ecDHFR) were engineered to be degraded, and, when this destabilizing domain is fused to a protein of interest, its instability is conferred to the fused protein resulting in rapid degradation of the entire fusion protein. A small-molecule ligand trimethoprim (TMP) stabilizes the destabilizing domain in a rapid, reversible, and dose-dependent manner, and protein levels in the absence of TMP are barely detectable. The ability of TMP to cross the blood-brain barrier enables the tunable regulation of proteins expressed in the mammalian central nervous system., (Copyright © 2010 Elsevier Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
29. A plant-like kinase in Plasmodium falciparum regulates parasite egress from erythrocytes.
- Author
-
Dvorin JD, Martyn DC, Patel SD, Grimley JS, Collins CR, Hopp CS, Bright AT, Westenberger S, Winzeler E, Blackman MJ, Baker DA, Wandless TJ, and Duraisingh MT
- Subjects
- Calcium-Binding Proteins chemistry, Calcium-Binding Proteins genetics, Cyclic GMP-Dependent Protein Kinases antagonists & inhibitors, Cyclic GMP-Dependent Protein Kinases metabolism, Enzyme Inhibitors pharmacology, Host-Parasite Interactions, Humans, Ligands, Merozoites enzymology, Merozoites physiology, Models, Biological, Morpholines metabolism, Plasmodium falciparum cytology, Plasmodium falciparum enzymology, Plasmodium falciparum growth & development, Protein Kinases chemistry, Protein Kinases genetics, Protozoan Proteins chemistry, Protozoan Proteins genetics, Pyridines pharmacology, Pyrroles pharmacology, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins metabolism, Schizonts cytology, Schizonts enzymology, Schizonts physiology, Calcium-Binding Proteins metabolism, Erythrocytes parasitology, Plasmodium falciparum physiology, Protein Kinases metabolism, Protozoan Proteins metabolism
- Abstract
Clinical malaria is associated with the proliferation of Plasmodium parasites in human erythrocytes. The coordinated processes of parasite egress from and invasion into erythrocytes are rapid and tightly regulated. We have found that the plant-like calcium-dependent protein kinase PfCDPK5, which is expressed in invasive merozoite forms of Plasmodium falciparum, was critical for egress. Parasites deficient in PfCDPK5 arrested as mature schizonts with intact membranes, despite normal maturation of egress proteases and invasion ligands. Merozoites physically released from stalled schizonts were capable of invading new erythrocytes, separating the pathways of egress and invasion. The arrest was downstream of cyclic guanosine monophosphate-dependent protein kinase (PfPKG) function and independent of protease processing. Thus, PfCDPK5 plays an essential role during the blood stage of malaria replication.
- Published
- 2010
- Full Text
- View/download PDF
30. Dicistronic regulation of fluorescent proteins in the budding yeast Saccharomyces cerevisiae.
- Author
-
Edwards SR and Wandless TJ
- Subjects
- Base Sequence, Gene Expression, Green Fluorescent Proteins metabolism, Luminescent Proteins metabolism, Molecular Sequence Data, Promoter Regions, Genetic, Saccharomyces cerevisiae metabolism, Red Fluorescent Protein, Gene Expression Regulation, Fungal, Genetic Engineering, Green Fluorescent Proteins genetics, Luminescent Proteins genetics, Saccharomyces cerevisiae genetics
- Abstract
Fluorescent proteins are convenient tools for measuring protein expression levels in the budding yeast Saccharomyces cerevisiae. Co-expression of proteins from distinct vectors has been seen by fluorescence microscopy; however, the expression of two fluorescent proteins on the same vector would allow for monitoring of linked events. We engineered constructs to allow dicistronic expression of red and green fluorescent proteins and found that expression levels of the proteins correlated with their order in the DNA sequence, with the protein encoded by the 5'-gene more highly expressed. To increase expression levels of the second gene, we tested four regulatory elements inserted between the two genes: the IRES sequences for the YAP1 and p150 genes, and the promoters for the TEF1 gene from both S. cerevisiae and Ashbya gossypii. We generated constructs encoding the truncated ADH1 promoter driving expression of the red protein, yeast-enhanced Cherry, followed by a regulatory element driving expression of the green protein, yeast-enhanced GFP. Three of the four regulatory elements successfully enhanced expression of the second gene in our dicistronic construct. We have developed a method to express two genes simultaneously from one vector. Both genes are codon-optimized to produce high protein levels in yeast, and the protein products can be visualized by microscopy or flow cytometry. With this method of regulation, the two genes can be driven in a dicistronic manner, with one protein marking cells harbouring the vector and the other protein free to mark any event of interest., (Copyright 2009 John Wiley & Sons, Ltd.)
- Published
- 2010
- Full Text
- View/download PDF
31. A general method for conditional regulation of protein stability in living animals.
- Author
-
Sellmyer MA, Thorne SH, Banaszynski LA, Contag CH, and Wandless TJ
- Subjects
- Animals, Binding Sites genetics, Female, Gene Transfer Techniques, HCT116 Cells, Humans, Male, Mice, Morpholines metabolism, Neoplasm Transplantation, Neoplasms, Experimental genetics, Recombinant Fusion Proteins genetics, Tacrolimus Binding Proteins genetics, Transplantation, Heterologous, Neoplasms, Experimental metabolism, Protein Stability, Recombinant Fusion Proteins metabolism
- Published
- 2009
- Full Text
- View/download PDF
32. Regulating protein stability in mammalian cells using small molecules.
- Author
-
Hagan EL, Banaszynski LA, Chen LC, Maynard-Smith LA, and Wandless TJ
- Subjects
- Animals, Binding Sites genetics, Cell Line, Humans, Ligands, Mice, Mutation, NIH 3T3 Cells, Plasmids genetics, Protein Binding, Recombinant Fusion Proteins genetics, Transfection, Protein Stability, Recombinant Fusion Proteins metabolism, Tacrolimus Binding Protein 1A genetics
- Published
- 2009
- Full Text
- View/download PDF
33. The proteasome makes sense of mixed signals.
- Author
-
Wandless TJ
- Subjects
- Ubiquitination, Gene Expression Regulation physiology, Proteasome Endopeptidase Complex metabolism, Proteins metabolism
- Published
- 2009
- Full Text
- View/download PDF
34. Recent progress with FKBP-derived destabilizing domains.
- Author
-
Chu BW, Banaszynski LA, Chen LC, and Wandless TJ
- Subjects
- Amino Acid Motifs, Cell Cycle drug effects, Cyclin B metabolism, Cyclin B1, HeLa Cells, Humans, Structure-Activity Relationship, Tacrolimus Binding Proteins genetics, Tacrolimus Binding Proteins metabolism, Models, Molecular, Tacrolimus Binding Proteins chemistry
- Abstract
The FKBP-derived destabilizing domains are increasingly being used to confer small molecule-dependent stability to many different proteins. The L106P domain confers instability to yellow fluorescent protein when it is fused to the N-terminus, the C-terminus, or spliced into the middle of yellow fluorescent protein, however multiple copies of L106P do not confer greater instability. These engineered destabilizing domains are not dominant to endogenous degrons that regulate protein stability.
- Published
- 2008
- Full Text
- View/download PDF
35. Chemical control of protein stability and function in living mice.
- Author
-
Banaszynski LA, Sellmyer MA, Contag CH, Wandless TJ, and Thorne SH
- Subjects
- Animals, HCT116 Cells, Humans, Interleukin-2 metabolism, Mice, Mice, SCID, Neoplasm Transplantation, Neoplasms, Experimental immunology, Protein Structure, Tertiary, Transplantation, Heterologous, Tumor Necrosis Factor-alpha metabolism, Neoplasms, Experimental drug therapy, Tacrolimus Binding Proteins chemistry, Tacrolimus Binding Proteins physiology
- Abstract
Conditional control of protein function in vivo offers great potential for deconvoluting the roles of individual proteins in complicated systems. We recently developed a method in which a small protein domain, termed a destabilizing domain, confers instability to fusion protein partners in cultured cells. Instability is reversed when a cell-permeable small molecule binds this domain. Here we describe the use of this system to regulate protein function in living mammals. We show regulation of secreted proteins and their biological activity with conditional secretion of an immunomodulatory cytokine, resulting in tumor burden reduction in mouse models. Additionally, we use this approach to control the function of a specific protein after systemic delivery of the gene that encodes it to a tumor, suggesting uses for enhancing the specificity and efficacy of targeted gene-based therapies. This method represents a new strategy to regulate protein function in living organisms with a high level of control.
- Published
- 2008
- Full Text
- View/download PDF
36. Synthesis and analysis of stabilizing ligands for FKBP-derived destabilizing domains.
- Author
-
Grimley JS, Chen DA, Banaszynski LA, and Wandless TJ
- Subjects
- Animals, Humans, Ligands, Mice, Tacrolimus Binding Proteins chemistry
- Abstract
We recently identified mutants of the human FKBP12 protein that are unstable and rapidly degraded when expressed in mammalian cells. We call these FKBP mutants destabilizing domains (DDs), because their instability is conferred to any protein fused to the DDs. A cell-permeable ligand binds tightly to the DDs and prevents their degradation, thus providing small molecule control over intracellular protein levels. We now report the synthesis and functional characterization of a stabilizing ligand called Shield-2. The synthesis of Shield-2 is efficient, and this ligand binds to the FKBP(F36V) protein with a dissociation constant of 29 nM.
- Published
- 2008
- Full Text
- View/download PDF
37. Rapid control of protein level in the apicomplexan Toxoplasma gondii.
- Author
-
Herm-Götz A, Agop-Nersesian C, Münter S, Grimley JS, Wandless TJ, Frischknecht F, and Meissner M
- Subjects
- Animals, Gene Expression Regulation physiology, Gene Silencing, Gene Targeting methods, Genetic Engineering methods, Protozoan Proteins genetics, Toxoplasma genetics
- Abstract
Analysis of gene function in apicomplexan parasites is limited by the absence of reverse genetic tools that allow easy and rapid modulation of protein levels. The fusion of a ligand-controlled destabilization domain (ddFKBP) to a protein of interest enables rapid and reversible protein stabilization in T. gondii. This allows an efficient functional analysis of proteins that have a dual role during host cell invasion and/or intracellular growth of the parasite.
- Published
- 2007
- Full Text
- View/download PDF
38. A directed approach for engineering conditional protein stability using biologically silent small molecules.
- Author
-
Maynard-Smith LA, Chen LC, Banaszynski LA, Ooi AG, and Wandless TJ
- Subjects
- Animals, Bacterial Proteins chemistry, Biophysics methods, Humans, Kinetics, Ligands, Luminescent Proteins chemistry, Mice, Models, Molecular, Mutation, NIH 3T3 Cells, Oligonucleotide Array Sequence Analysis, Protein Binding, Tacrolimus Binding Protein 1A chemistry, Thermodynamics, Protein Engineering methods
- Abstract
The ability to regulate the function of specific proteins using cell-permeable molecules can be a powerful method for interrogating biological systems. To bring this type of "chemical genetic" control to a wide range of proteins, we recently developed an experimental system in which the stability of a small protein domain expressed in mammalian cells depends on the presence of a high affinity ligand. This ligand-dependent stability is conferred to any fused partner protein. The FK506- and rapamycin-binding protein (FKBP12) has been the subject of extensive biophysical analyses, including both kinetic and thermodynamic studies of the wild-type protein as well as dozens of mutants. The goal of this study was to determine if the thermodynamic stabilities (DeltaDeltaG(U-F)) of various amino acid substitutions within a given protein are predictive for engineering additional ligand-dependent destabilizing domains. We used FKBP12 as a model system and found that in vitro thermodynamic stability correlates weakly with intracellular degradation rates of the mutants and that the ability of a given mutation to destabilize the protein is context-dependent. We evaluated several new FKBP12 ligands for their ability to stabilize these mutants and found that a cell-permeable molecule called Shield-1 is the most effective stabilizing ligand. We then performed an unbiased microarray analysis of NIH3T3 cells treated with various concentrations of Shield-1. These studies show that Shield-1 does not elicit appreciable cellular responses.
- Published
- 2007
- Full Text
- View/download PDF
39. Rescue of degradation-prone mutants of the FK506-rapamycin binding (FRB) protein with chemical ligands.
- Author
-
Stankunas K, Bayle JH, Havranek JJ, Wandless TJ, Baker D, Crabtree GR, and Gestwicki JE
- Subjects
- Amino Acid Sequence, Animals, COS Cells, Cells, Cultured, Chlorocebus aethiops, Fibroblasts metabolism, Ligands, Mice, Models, Chemical, Molecular Sequence Data, Point Mutation, Tacrolimus Binding Proteins chemistry, Thermodynamics, Tryptophan chemistry, Mutation, Tacrolimus Binding Proteins genetics
- Abstract
We recently reported that certain mutations in the FK506-rapamycin binding (FRB) domain disrupt its stability in vitro and in vivo (Stankunas et al. Mol. Cell, 2003, 12, 1615). To determine the precise residues that cause instability, we calculated the folding free energy (Delta G) of a collection of FRB mutants by measuring their intrinsic tryptophan fluorescence during reversible chaotropic denaturation. Our results implicate the T2098L point mutation as a key determinant of instability. Further, we found that some of the mutants in this collection were destabilized by up to 6 kcal mol(-1) relative to the wild type. To investigate how these mutants behave in cells, we expressed firefly luciferase fused to FRB mutants in African green monkey kidney (COS) cell lines and mouse embryonic fibroblasts (MEFs). When unstable FRB mutants were used, we found that the protein levels and the luminescence intensities were low. However, addition of a chemical ligand for FRB, rapamycin, restored luciferase activity. Interestingly, we found a roughly linear relationship between the Delta G of the FRB mutants calculated in vitro and the relative chemical rescue in cells. Because rapamycin is capable of simultaneously binding both FRB and the chaperone, FK506-binding protein (FKBP), we next examined whether FKBP might contribute to the protection of FRB mutants. Using both in vitro experiments and a cell-based model, we found that FKBP stabilizes the mutants. These findings are consistent with recent models that suggest damage to intrinsic Delta G can be corrected by pharmacological chaperones. Further, these results provide a collection of conditionally stable fusion partners for use in controlling protein stability.
- Published
- 2007
- Full Text
- View/download PDF
40. Engineering small molecule specificity in nearly identical cellular environments.
- Author
-
Sellmyer MA, Stankunas K, Briesewitz R, Crabtree GR, and Wandless TJ
- Subjects
- Engineering, Environment, Folic Acid Antagonists chemistry, Humans, Methotrexate pharmacology, Tacrolimus Binding Protein 1A metabolism, Folic Acid Antagonists pharmacology, Methotrexate chemistry, Tacrolimus Binding Protein 1A antagonists & inhibitors, Tetrahydrofolate Dehydrogenase metabolism
- Abstract
Methotrexate (MTX), an inhibitor of dihydrofolate reductase, was tethered to an FKBP12 ligand (SLF), and the resulting bifunctional molecule (MTXSLF) potently inhibits either enzyme but not both simultaneously. MTXSLF is cytotoxic to fibroblasts derived from FKBP12-null mice but is detoxified 40-fold by FKBP12 in wild-type fibroblasts. These studies demonstrate that non-target proteins in an otherwise identical genetic background can be used to predictably regulate the biological activity of synthetic molecules.
- Published
- 2007
- Full Text
- View/download PDF
41. The rapamycin-binding domain of the protein kinase mammalian target of rapamycin is a destabilizing domain.
- Author
-
Edwards SR and Wandless TJ
- Subjects
- Amino Acid Motifs genetics, Animals, Enzyme Stability drug effects, Enzyme Stability genetics, Ligands, Mass Spectrometry, Mice, Mutation, NIH 3T3 Cells, Protein Binding drug effects, Protein Binding genetics, Protein Kinases genetics, Protein Structure, Quaternary drug effects, Saccharomyces cerevisiae genetics, Sirolimus analogs & derivatives, TOR Serine-Threonine Kinases, Tacrolimus Binding Protein 1A genetics, Two-Hybrid System Techniques, Immunosuppressive Agents pharmacology, Protein Kinases metabolism, Sirolimus pharmacology, Tacrolimus Binding Protein 1A metabolism
- Abstract
Rapamycin is an immunosuppressive drug that binds simultaneously to the 12-kDa FK506- and rapamycin-binding protein (FKBP12, or FKBP) and the FKBP-rapamycin binding (FRB) domain of the mammalian target of rapamycin (mTOR) kinase. The resulting ternary complex has been used to conditionally perturb protein function, and one such method involves perturbation of a protein of interest through its mislocalization. We synthesized two rapamycin derivatives that possess large substituents at the C-16 position within the FRB-binding interface, and these derivatives were screened against a library of FRB mutants using a three-hybrid assay in Saccharomyces cerevisiae. Several FRB mutants responded to one of the rapamycin derivatives, and twenty of these mutants were further characterized in mammalian cells. The mutants most responsive to the ligand were fused to yellow fluorescent protein, and fluorescence levels in the presence and absence of the ligand were measured to determine stability of the fusion proteins. Wild-type and mutant FRB domains were expressed at low levels in the absence of the rapamycin derivative, and expression levels rose up to 10-fold upon treatment with ligand. The synthetic rapamycin derivatives were further analyzed using quantitative mass spectrometry, and one of the compounds was found to contain contaminating rapamycin. Furthermore, uncontaminated analogs retained the ability to inhibit mTOR, although with diminished potency relative to rapamycin. The ligand-dependent stability displayed by wild-type FRB and FRB mutants as well as the inhibitory potential and purity of the rapamycin derivatives should be considered as potentially confounding experimental variables when using these systems.
- Published
- 2007
- Full Text
- View/download PDF
42. The enantioselective synthesis of phomopsin B.
- Author
-
Grimley JS, Sawayama AM, Tanaka H, Stohlmeyer MM, Woiwode TF, and Wandless TJ
- Subjects
- Chromatography, High Pressure Liquid, Magnetic Resonance Spectroscopy, Mass Spectrometry, Molecular Structure, Mycotoxins chemistry, Mycotoxins pharmacology, Stereoisomerism, Mycotoxins chemical synthesis
- Published
- 2007
- Full Text
- View/download PDF
43. PI(3,4,5)P3 and PI(4,5)P2 lipids target proteins with polybasic clusters to the plasma membrane.
- Author
-
Heo WD, Inoue T, Park WS, Kim ML, Park BO, Wandless TJ, and Meyer T
- Subjects
- ADP-Ribosylation Factors chemistry, ADP-Ribosylation Factors metabolism, Amino Acid Motifs, Amino Acid Sequence, Animals, GTP Phosphohydrolases chemistry, HeLa Cells, Humans, Hydrophobic and Hydrophilic Interactions, Kinetics, Mice, Molecular Sequence Data, NIH 3T3 Cells, Second Messenger Systems, Signal Transduction, Static Electricity, rab GTP-Binding Proteins chemistry, rab GTP-Binding Proteins metabolism, ras Proteins chemistry, ras Proteins metabolism, rho GTP-Binding Proteins metabolism, Cell Membrane metabolism, GTP Phosphohydrolases metabolism, Phosphatidylinositol 4,5-Diphosphate metabolism, Phosphatidylinositol Phosphates metabolism
- Abstract
Many signaling, cytoskeletal, and transport proteins have to be localized to the plasma membrane (PM) in order to carry out their function. We surveyed PM-targeting mechanisms by imaging the subcellular localization of 125 fluorescent protein-conjugated Ras, Rab, Arf, and Rho proteins. Out of 48 proteins that were PM-localized, 37 contained clusters of positively charged amino acids. To test whether these polybasic clusters bind negatively charged phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] lipids, we developed a chemical phosphatase activation method to deplete PM PI(4,5)P2. Unexpectedly, proteins with polybasic clusters dissociated from the PM only when both PI(4,5)P2 and phosphatidylinositol 3,4,5-trisphosphate [PI(3,4,5)P3] were depleted, arguing that both lipid second messengers jointly regulate PM targeting.
- Published
- 2006
- Full Text
- View/download PDF
44. A rapid, reversible, and tunable method to regulate protein function in living cells using synthetic small molecules.
- Author
-
Banaszynski LA, Chen LC, Maynard-Smith LA, Ooi AG, and Wandless TJ
- Subjects
- Animals, Ligands, Luminescent Proteins genetics, Mice, Mutation, NIH 3T3 Cells, Phenotype, Protein Binding, Protein Structure, Tertiary, Tacrolimus Binding Protein 1A chemistry, Tacrolimus Binding Proteins chemistry, Transfection, Gene Expression Regulation, Morpholines metabolism, Proteasome Endopeptidase Complex metabolism, Recombinant Fusion Proteins metabolism, Tacrolimus Binding Protein 1A genetics, Tacrolimus Binding Proteins genetics
- Abstract
Rapid and reversible methods for perturbing the function of specific proteins are desirable tools for probing complex biological systems. We have developed a general technique to regulate the stability of specific proteins in mammalian cells using cell-permeable, synthetic molecules. We engineered mutants of the human FKBP12 protein that are rapidly and constitutively degraded when expressed in mammalian cells, and this instability is conferred to other proteins fused to these destabilizing domains. Addition of a synthetic ligand that binds to the destabilizing domains shields them from degradation, allowing fused proteins to perform their cellular functions. Genetic fusion of the destabilizing domain to a gene of interest ensures specificity, and the attendant small-molecule control confers speed, reversibility, and dose-dependence to this method. This general strategy for regulating protein stability should enable conditional perturbation of specific proteins with unprecedented control in a variety of experimental settings.
- Published
- 2006
- Full Text
- View/download PDF
45. Rapamycin analogs with differential binding specificity permit orthogonal control of protein activity.
- Author
-
Bayle JH, Grimley JS, Stankunas K, Gestwicki JE, Wandless TJ, and Crabtree GR
- Subjects
- Glycogen Synthase Kinase 3 metabolism, Glycogen Synthase Kinase 3 beta, Models, Molecular, Molecular Structure, Mutation genetics, Protein Kinases chemistry, Protein Kinases genetics, Protein Structure, Tertiary, Sirolimus metabolism, TOR Serine-Threonine Kinases, Protein Kinases metabolism, Sirolimus analogs & derivatives, Sirolimus pharmacology
- Abstract
Controlling protein dimerization with small molecules has broad application to the study of protein function. Rapamycin has two binding surfaces: one that binds to FKBP12 and the other to the Frb domain of mTor/FRAP, directing their dimerization. Rapamycin is a potent cell growth inhibitor, but chemical modification of the surface contacting Frb alleviates this effect. Productive interactions with Frb-fused proteins can be restored by mutation of Frb to accommodate the rapamycin analog (a rapalog). We have quantitatively assessed the interaction between rapalogs functionalized at C16 and C20 and a panel of Frb mutants. Several drug-Frb mutant combinations have different and nonoverlapping specificities. These Frb-rapalog partners permit the selective control of different Frb fusion proteins without crossreaction. The orthogonal control of multiple target proteins broadens the capabilities of chemical induction of dimerization to regulate biologic processes.
- Published
- 2006
- Full Text
- View/download PDF
46. Conditional control of protein function.
- Author
-
Banaszynski LA and Wandless TJ
- Subjects
- Animals, Hormones metabolism, Hormones pharmacology, Protein Binding drug effects, Protein Engineering, Proteins genetics, RNA Splicing drug effects, Proteins metabolism
- Abstract
Deciphering the myriad ways in which proteins interact with each other to give rise to complex behaviors that define living systems is a significant challenge. Using perturbations of DNA, genetic analyses have provided many insights into the functions of proteins encoded by specific genes. However, it can be difficult to study essential genes using these approaches, and many biological processes occur on a fast timescale that precludes study using genetic methods. For these reasons and others, it is often desirable to target proteins directly rather than the genes that encode them. Over the past 20 years, several methods to regulate protein function have been developed. In this review, we discuss the genesis and use of these methods, with particular emphasis on the elements of specificity, speed, and reversibility.
- Published
- 2006
- Full Text
- View/download PDF
47. A cell-permeable, activity-based probe for protein and lipid kinases.
- Author
-
Yee MC, Fas SC, Stohlmeyer MM, Wandless TJ, and Cimprich KA
- Subjects
- Androstadienes pharmacology, Biotin chemistry, Boron Compounds chemistry, Cell Line, Enzyme Inhibitors pharmacology, Fluorescent Dyes pharmacology, Glutathione Transferase metabolism, HeLa Cells, Humans, Inhibitory Concentration 50, Models, Chemical, Phosphatidylinositol 3-Kinases chemistry, Phosphatidylinositol 3-Kinases metabolism, Phosphorylation, Protein Binding, Protein Processing, Post-Translational, Rhodamines chemistry, Time Factors, Transfection, Wortmannin, Androstadienes chemistry, Biochemistry methods, Lipids chemistry
- Abstract
Protein and lipid kinases are two important classes of biomedically relevant enzymes. The expression and activity of many kinases are known to be dysregulated in a variety of diseases, and proteomic tools that can assess the presence and activity of these enzymes are likely to be useful for their evaluation. Because many of the mechanisms by which protein kinases can become unregulated involve post-translational modifications or changes in protein localization, they can only be detected by examining protein activity, sometimes within the context of the living cell. Wortmannin is a steroid-derived fungal metabolite that covalently inhibits both protein and lipid kinases. Here we describe the synthesis of three wortmannin derivatives, biotin-wortmannin, BODIPY-wortmannin, and tetramethylrhodamine-wortmannin. We demonstrate that these reagents exhibit reactivity similarly as wortmannin and react with members of the phosphatidylinositol 3-kinase and PI3-kinase related kinase families in cellular lysates. Moreover, in some cases these reagents can differentiate between the active and inactive forms of the enzyme, indicating that they are activity-based probes. The reagents also exhibit complementary properties. The biotin-wortmannin reagent is effective in the isolation of labeled proteins; all three can be used for protein labeling, and BODIPY-wortmannin is cell-permeable and can be used to label proteins within cells.
- Published
- 2005
- Full Text
- View/download PDF
48. An inducible translocation strategy to rapidly activate and inhibit small GTPase signaling pathways.
- Author
-
Inoue T, Heo WD, Grimley JS, Wandless TJ, and Meyer T
- Subjects
- Animals, Cell Membrane drug effects, Enzyme Inhibitors, Kinetics, Mice, NIH 3T3 Cells, Recombinant Fusion Proteins metabolism, Signal Transduction drug effects, Translocation, Genetic genetics, Cell Membrane metabolism, Protein Engineering methods, Signal Transduction physiology, Sirolimus pharmacology, rho GTP-Binding Proteins antagonists & inhibitors, rho GTP-Binding Proteins metabolism
- Abstract
We made substantial advances in the implementation of a rapamycin-triggered heterodimerization strategy. Using molecular engineering of different targeting and enzymatic fusion constructs and a new rapamycin analog, Rho GTPases were directly activated or inactivated on a timescale of seconds, which was followed by pronounced cell morphological changes. As signaling processes often occur within minutes, such rapid perturbations provide a powerful tool to investigate the role, selectivity and timing of Rho GTPase-mediated signaling processes.
- Published
- 2005
- Full Text
- View/download PDF
49. Characterization of the FKBP.rapamycin.FRB ternary complex.
- Author
-
Banaszynski LA, Liu CW, and Wandless TJ
- Subjects
- Binding, Competitive, Fluorescence Polarization, Kinetics, Models, Molecular, Nuclear Magnetic Resonance, Biomolecular, Protein Binding, Protein Kinases metabolism, Protein Structure, Tertiary, Sirolimus metabolism, Sirolimus pharmacology, Surface Plasmon Resonance, TOR Serine-Threonine Kinases, Tacrolimus Binding Protein 1A chemistry, Tacrolimus Binding Protein 1A metabolism, Tacrolimus Binding Proteins metabolism, Protein Kinases chemistry, Sirolimus chemistry, Tacrolimus Binding Proteins chemistry
- Abstract
Rapamycin is an important immunosuppressant, a possible anticancer therapeutic, and a widely used research tool. Essential to its various functions is its ability to bind simultaneously to two different proteins, FKBP and mTOR. Despite its widespread use, a thorough analysis of the interactions between FKBP, rapamycin, and the rapamycin-binding domain of mTOR, FRB, is lacking. To probe the affinities involved in the formation of the FKBP.rapamycin.FRB complex, we used fluorescence polarization, surface plasmon resonance, and NMR spectroscopy. Analysis of the data shows that rapamycin binds to FRB with moderate affinity (K(d) = 26 +/- 0.8 microM). The FKBP12.rapamycin complex, however, binds to FRB 2000-fold more tightly (K(d) = 12 +/- 0.8 nM) than rapamycin alone. No interaction between FKBP and FRB was detected in the absence of rapamycin. These studies suggest that rapamycin's ability to bind to FRB, and by extension to mTOR, in the absence of FKBP is of little consequence under physiological conditions. Furthermore, protein-protein interactions at the FKBP12-FRB interface play a role in the stability of the ternary complex.
- Published
- 2005
- Full Text
- View/download PDF
50. Total synthesis of ustiloxin D and considerations on the origin of selectivity of the asymmetric allylic alkylation.
- Author
-
Sawayama AM, Tanaka H, and Wandless TJ
- Subjects
- Alkylation, Models, Molecular, Molecular Conformation, Peptides, Cyclic, Antineoplastic Agents chemical synthesis, Mycotoxins chemical synthesis, Peptides chemical synthesis
- Abstract
As part of investigations into cell cycle checkpoint inhibitors, an asymmetric synthesis of the antimitotic natural product, ustiloxin D, has been completed. A salen-Al-catalyzed aldol reaction was employed to construct a chiral oxazoline 9 (99% yield, 98% ee) that served the dual purpose of installing the necessary 1,2-amino alcohol functionality as well as providing an efficient synthon for the requisite methylamino group at C9. The chiral aryl-alkyl ether was assembled using a Pd-catalyzed asymmetric allylic alkylation that notably delivered a product with stereochemistry opposite to that predicted by precedent. The linear tetrapeptide was subsequently cyclized to produce ustiloxin D. The mechanistic origin of the allylic alkylation selectivity was further investigated, and a working hypothesis for the origin of the observed stereoselectivity has been proposed.
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.