1
|
Galindo G, Fixen GM, Heredia A, Morisaki T, Stasevich TJ. All Probes Plasmids (APPs) for multicolor and long-term tracking of single-mRNA translation dynamics. Mol Biol Cell 2025; 36:mr6. [PMID: 40366872 DOI: 10.1091/mbc.e25-02-0091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025] Open
Abstract
Live-cell single-mRNA imaging of translation is inherently challenging, demanding precise optimization of multiple imaging components. To simplify these experiments, we developed All Probes Plasmids (APPs)-a panel of plasmids encoding all the necessary probes for imaging at optimized relative expression levels. APPs incorporate widely used translation tags, fluorescent proteins, and mRNA labeling systems, streamlining both multiplexed imaging and reporter immobilization. By cotransfecting just two plasmids-a reporter and an APP-individual translation sites can be visualized in living cells with high signal-to-noise. We demonstrate how APPs facilitate high-fidelity multicolor translation imaging, long-term single-mRNA tracking, and fluorescence correlation spectroscopy to quantify ribosome kinetics. By lowering technical barriers and enhancing experimental flexibility, APPs provide a versatile platform for advancing single-mRNA translation research in living cells.
Collapse
Affiliation(s)
- Gabriel Galindo
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| | - Gretchen M Fixen
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| | - Amelia Heredia
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| | - Tatsuya Morisaki
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| | - Timothy J Stasevich
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
2
|
de Luis M, Xu S, Zinn K. Fluorescent labeling of proteins in vitro and in vivo using encoded peptide tags. J Biol Chem 2025:110229. [PMID: 40378958 DOI: 10.1016/j.jbc.2025.110229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 04/17/2025] [Accepted: 05/02/2025] [Indexed: 05/19/2025] Open
Abstract
Epitope tags are a simple and versatile way to label proteins as their sequences can easily be inserted into protein coding sequences, so that the expressed proteins will bear the tag(s). These tags can be used to identify and purify proteins in vitro using Western blots, flow cytometry, affinity chromatography, and other techniques. When labeled with a fluorescent probe, tagged proteins can be visualized in live or fixed cells or tissues using fluorescence microscopy, allowing for the study of protein dynamics. The most widely used epitope tags are those that have affinity to an antibody, which can be used in fixed-sample immunohistochemistry studies. While this will allow insight into a protein's localization, it will not provide any information on its dynamics. Other tags were developed with the intended use in live imaging. In this mini review, we discuss epitope tags that have affinity to antibodies, nanobodies, and small molecules and their use in fluorescence microscopy for fixed and live imaging.
Collapse
Affiliation(s)
- Maya de Luis
- Division of Biology and Biological Engineering, California Institute of Technology
| | - Shuwa Xu
- Division of Biology and Biological Engineering, California Institute of Technology
| | - Kai Zinn
- Division of Biology and Biological Engineering, California Institute of Technology
| |
Collapse
|
3
|
Rihtar E, Fink T, Lebar T, Lainšček D, Kolenc Ž, Polajnar LK, Jerala R. Ligand-induced assembly of antibody variable fragments for the chemical regulation of biological processes. Cell Chem Biol 2025; 32:474-485.e5. [PMID: 39952240 PMCID: PMC11935766 DOI: 10.1016/j.chembiol.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/30/2024] [Accepted: 01/26/2025] [Indexed: 02/17/2025]
Abstract
Precise control of biological processes by the application of small molecules can increase the safety and efficiency of therapies. Adverse side effects of small molecule signals and/or immunogenicity of regulatory domains hinder their biomedical utility. Here, we designed small molecule-responsive switches, based on the conditional reassembly of human antibody variable fragments, called Fv-CID switches. The principle was validated using high-affinity antibodies against nicotine and β-estradiol to construct chemically responsive transcription factors. Further, we developed an Fv-CID switch responsive to bio-inert, clinically approved compound fluorescein, which was used to control the activity of chimeric antigen receptor (CAR) T cells and bispecific T cell engagers (BiTEs) in vivo. This study provides a framework to regulate the expression of endogenous genes, combine multiple chemical signals, and regulate T cell-based immunotherapy in an animal cancer model using a clinically approved small molecule regulator that could be customized for regulating therapeutic proteins or cells.
Collapse
Affiliation(s)
- Erik Rihtar
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Tina Fink
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia; Centre for Technologies of Gene and Cell Therapy, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Tina Lebar
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Duško Lainšček
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia; Centre for Technologies of Gene and Cell Therapy, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Živa Kolenc
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Lucija Kadunc Polajnar
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Roman Jerala
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia; Centre for Technologies of Gene and Cell Therapy, Hajdrihova 19, 1000 Ljubljana, Slovenia.
| |
Collapse
|
4
|
Streit M, Budiarta M, Jungblut M, Beliu G. Fluorescent labeling strategies for molecular bioimaging. BIOPHYSICAL REPORTS 2025; 5:100200. [PMID: 39947326 PMCID: PMC11914189 DOI: 10.1016/j.bpr.2025.100200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/28/2025] [Accepted: 02/06/2025] [Indexed: 03/16/2025]
Abstract
Super-resolution microscopy (SRM) has transformed biological imaging by circumventing the diffraction limit of light and enabling the visualization of cellular structures and processes at the molecular level. Central to the capabilities of SRM is fluorescent labeling, which ensures the precise attachment of fluorophores to biomolecules and has direct impact on the accuracy and resolution of imaging. Continuous innovation and optimization in fluorescent labeling are essential for the successful application of SRM in cutting-edge biological research. In this review, we discuss recent advances in fluorescent labeling strategies for molecular bioimaging, with a special focus on protein labeling. We compare different approaches, highlight technological breakthroughs, and address challenges such as linkage error and labeling density. By evaluating both established and emerging methods, we aim to guide researchers through all aspects that should be considered before opting for any labeling technique.
Collapse
Affiliation(s)
- Marcel Streit
- Rudolf Virchow Center, Research Center for Integrative and Translational Bioimaging, University of Würzburg, 97080 Würzburg, Germany
| | - Made Budiarta
- Rudolf Virchow Center, Research Center for Integrative and Translational Bioimaging, University of Würzburg, 97080 Würzburg, Germany
| | - Marvin Jungblut
- Rudolf Virchow Center, Research Center for Integrative and Translational Bioimaging, University of Würzburg, 97080 Würzburg, Germany
| | - Gerti Beliu
- Rudolf Virchow Center, Research Center for Integrative and Translational Bioimaging, University of Würzburg, 97080 Würzburg, Germany.
| |
Collapse
|
5
|
Sescil J, Havens SM, Wang W. Principles and Design of Molecular Tools for Sensing and Perturbing Cell Surface Receptor Activity. Chem Rev 2025; 125:2665-2702. [PMID: 39999110 PMCID: PMC11934152 DOI: 10.1021/acs.chemrev.4c00582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
Cell-surface receptors are vital for controlling numerous cellular processes with their dysregulation being linked to disease states. Therefore, it is necessary to develop tools to study receptors and the signaling pathways they control. This Review broadly describes molecular approaches that enable 1) the visualization of receptors to determine their localization and distribution; 2) sensing receptor activation with permanent readouts as well as readouts in real time; and 3) perturbing receptor activity and mimicking receptor-controlled processes to learn more about these processes. Together, these tools have provided valuable insight into fundamental receptor biology and helped to characterize therapeutics that target receptors.
Collapse
Affiliation(s)
- Jennifer Sescil
- Department of Chemistry, University of Michigan, Ann Arbor,
MI, 48109
- Life Sciences Institute, University of Michigan, Ann Arbor,
MI, 48109
| | - Steven M. Havens
- Department of Chemistry, University of Michigan, Ann Arbor,
MI, 48109
- Life Sciences Institute, University of Michigan, Ann Arbor,
MI, 48109
| | - Wenjing Wang
- Department of Chemistry, University of Michigan, Ann Arbor,
MI, 48109
- Life Sciences Institute, University of Michigan, Ann Arbor,
MI, 48109
- Neuroscience Graduate Program, University of Michigan, Ann
Arbor, MI, 48109
- Program in Chemical Biology, University of Michigan, Ann
Arbor, MI, 48109
| |
Collapse
|
6
|
Jiang Q, Shao S, Li N, Zhang Z, Zhao L, Zhang H, Liu B. Live MSCs Characterizer Displays Stemness and Differentiation Using Colorful LV-cp Biosensors. ACS Sens 2025; 10:825-834. [PMID: 39907518 DOI: 10.1021/acssensors.4c02356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Mesenchymal stem cells (MSCs) have garnered significant attention in biomedical research due to their accessibility and remarkable differentiation potential. However, the lack of efficient and convenient living cell monitoring methods limits their widespread application in tissue engineering and stem cell therapy. Therefore, we present progress in the development of a novel series of fluorescent protein (FP) sensors based on turn-on fluorescent protein biosensors (Turn-on FPBs), termed the LV-cp biosensor system (novel live cell permuted fluorescent protein biosensors). Utilizing phage display technology to screen for affinity peptides specifically targeting MSCs and chondrocytes, the LV-cp were engineered by subcloning these peptides into permuted fluorescent proteins, thereby integrating the fluorescence activation mechanism with the affinity peptides and achieving highly accurate detection and identification of these two cell types using living cells as "fluorescence keys." This system provides a simplified, nontoxic method to replace traditional antibody kits, and strong fluorescence signals can be obtained through various fluorescence detection devices. In addition, the LV-cp biosensors enabled dynamic observation of MSCs differentiation into chondrocytes through changes in the cell fluorescence colors.
Collapse
Affiliation(s)
- Qingyun Jiang
- Cancer Hospital of Dalian University of Technology, Shenyang 110042, P. R. China
- Faculty of Medicine, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian 116024, P. R. China
| | - Shuai Shao
- Cancer Hospital of Dalian University of Technology, Shenyang 110042, P. R. China
- Faculty of Medicine, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian 116024, P. R. China
| | - Na Li
- Cancer Hospital of Dalian University of Technology, Shenyang 110042, P. R. China
- Faculty of Medicine, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian 116024, P. R. China
| | - Zhengyao Zhang
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin 124221, P. R. China
| | - Luming Zhao
- Cancer Hospital of Dalian University of Technology, Shenyang 110042, P. R. China
- Faculty of Medicine, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian 116024, P. R. China
| | - Hangyu Zhang
- Cancer Hospital of Dalian University of Technology, Shenyang 110042, P. R. China
- Faculty of Medicine, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian 116024, P. R. China
| | - Bo Liu
- Cancer Hospital of Dalian University of Technology, Shenyang 110042, P. R. China
- Faculty of Medicine, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian 116024, P. R. China
| |
Collapse
|
7
|
Galindo G, Maejima D, DeRoo J, Burlingham SR, Fixen G, Morisaki T, Febvre HP, Hasbrook R, Zhao N, Ghosh S, Mayton EH, Snow CD, Geiss BJ, Ohkawa Y, Sato Y, Kimura H, Stasevich TJ. AI-assisted protein design to rapidly convert antibody sequences to intrabodies targeting diverse peptides and histone modifications. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.06.636921. [PMID: 39975170 PMCID: PMC11839053 DOI: 10.1101/2025.02.06.636921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Intrabodies are engineered antibodies that function inside living cells, enabling therapeutic, diagnostic, and imaging applications. While powerful, their development has been hindered by challenges associated with their folding, solubility, and stability in the reduced intracellular environment. Here, we present an AI-driven pipeline integrating AlphaFold2, ProteinMPNN, and live-cell screening to optimize antibody framework regions while preserving epitope-binding complementarity-determining regions. Using this approach, we successfully converted 19 out of 26 antibody sequences into functional single-chain variable fragment (scFv) intrabodies, including a panel targeting diverse histone modifications for real-time imaging of chromatin dynamics and gene regulation. Notably, 18 of these 19 sequences had failed to convert using the standard approach, demonstrating the unique effectiveness of our method. As antibody sequence databases expand, our method will accelerate intrabody design, making their development easier, more cost-effective, and broadly accessible for biological research.
Collapse
Affiliation(s)
- Gabriel Galindo
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Daiki Maejima
- School of Life Science and Technology, Institute of Science Tokyo, Yokohama, Japan
| | - Jacob DeRoo
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Scott R Burlingham
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Gretchen Fixen
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Tatsuya Morisaki
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Hallie P Febvre
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Ryan Hasbrook
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Ning Zhao
- Department of Biochemistry and Molecular Genetics, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA
| | - Soham Ghosh
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, USA
| | - E Handly Mayton
- Department of Microbiology, Immunology, & Pathology, Colorado State University, Fort Collins, CO, USA
| | - Christopher D Snow
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
- Department of Chemical & Biological Engineering, Colorado State University, Fort Collins, CO, USA
| | - Brian J Geiss
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
- Department of Microbiology, Immunology, & Pathology, Colorado State University, Fort Collins, CO, USA
| | - Yasuyuki Ohkawa
- Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Yuko Sato
- Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| | - Hiroshi Kimura
- School of Life Science and Technology, Institute of Science Tokyo, Yokohama, Japan
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
- Cell Biology Center, Institute of Integrated Research, Institute of Science Tokyo, Yokohama, Japan
| | - Timothy J Stasevich
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
- Department of Chemical & Biological Engineering, Colorado State University, Fort Collins, CO, USA
- Cell Biology Center, Institute of Integrated Research, Institute of Science Tokyo, Yokohama, Japan
| |
Collapse
|
8
|
Banna HA, Berg K, Sadat T, Das N, Paudel R, D'Souza V, Koirala D. Synthetic anti-RNA antibody derivatives for RNA visualization in mammalian cells. Nucleic Acids Res 2025; 53:gkae1275. [PMID: 39739875 PMCID: PMC11879077 DOI: 10.1093/nar/gkae1275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 11/15/2024] [Accepted: 12/12/2024] [Indexed: 01/02/2025] Open
Abstract
Although antibody derivatives, such as Fabs and scFvs, have revolutionized the cellular imaging, quantification and tracking of proteins, analogous tools and strategies are unavailable for cellular RNA visualization. Here, we developed four synthetic anti-RNA scFv (sarabody) probes and their green fluorescent protein (GFP) fusions and demonstrated their potential to visualize RNA in live mammalian cells. We expressed these sarabodies and sarabody-GFP modules, purified them as soluble proteins, characterized their binding interactions with their corresponding epitopes and finally employed two of the four modules, sara1-GFP and sara1c-GFP, to visualize a target messenger RNA in live U2OS cells. Our current RNA imaging strategy is analogous to the existing MCP-MS2 system for RNA visualization, but additionally, our approach provides robust flexibility for developing target RNA-specific imaging modules, as epitope-specific probes can be selected from a library generated by diversifying the sarabody complementarity determining regions. While we continue to optimize these probes, develop new probes for various target RNAs and incorporate other fluorescence proteins like mCherry and HaloTag, our groundwork results demonstrated that these first-of-a-kind immunofluorescent probes will have tremendous potential for tracking mature RNAs and may aid in visualizing and quantifying many cellular processes as well as examining the spatiotemporal dynamics of various RNAs.
Collapse
Affiliation(s)
- Hasan Al Banna
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, MD 21250, USA
| | - Kimberley Berg
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Tasnia Sadat
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, MD 21250, USA
| | - Naba Krishna Das
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, MD 21250, USA
| | - Roshan Paudel
- Department of Computer Science, Morgan State University, Baltimore, MD 21251, USA
| | - Victoria D'Souza
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Deepak Koirala
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, MD 21250, USA
| |
Collapse
|
9
|
Cho Y, Jeong I, Kim KE, Rhee HW. Painting Cell-Cell Interactions by Horseradish Peroxidase and Endogenously Generated Hydrogen Peroxide. ACS Chem Biol 2025; 20:86-93. [PMID: 39692451 DOI: 10.1021/acschembio.4c00419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Cell-cell interactions are fundamental in biology for maintaining physiological conditions with direct contact being the most straightforward mode of interaction. Recent advancements have led to the development of various chemical tools for detecting or identifying these interactions. However, the use of exogenous cues, such as toxic reagents, bulky probes, and light irradiation, can disrupt normal cell physiology. For example, the toxicity of hydrogen peroxide (H2O2) limits the applications of peroxidases in the proximity labeling field. In this study, we aimed to address this limitation by demonstrating that membrane-localized horseradish peroxidase (HRP-TM) efficiently utilizes endogenously generated extracellular H2O2. By harnessing endogenous H2O2, we observed that HRP-TM-expressing cells can effectively label contacting cells without the need for exogenous H2O2 treatment. Furthermore, we confirmed that HRP-TM labels proximal cells in an interaction-dependent manner. These findings offer a novel approach for studying cell-cell interactions under more physiological conditions without the confounding effects of exogenous stimuli. Our study contributes to elucidating cell-cell interaction networks in various model organisms, providing valuable insights into the dynamic interplay between cells in their native network.
Collapse
Affiliation(s)
- Youngjoon Cho
- Department of Chemistry, Seoul National University, Seoul 08826, Korea
| | - Inyoung Jeong
- Department of Chemistry, Seoul National University, Seoul 08826, Korea
| | - Kwang-Eun Kim
- Department of Chemistry, Seoul National University, Seoul 08826, Korea
- Department of Convergence Medicine, Yonsei University Wonju College of Medicine, Wonju 26426, Korea
| | - Hyun-Woo Rhee
- Department of Chemistry, Seoul National University, Seoul 08826, Korea
- School of Biological Sciences, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
10
|
Lyon KR, Morisaki T, Stasevich TJ. Imaging and Quantifying Ribosomal Frameshifting Dynamics with Single-RNA Precision in Live Cells. Methods Mol Biol 2025; 2875:99-110. [PMID: 39535643 PMCID: PMC11633442 DOI: 10.1007/978-1-0716-4248-1_9] [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] [Indexed: 11/16/2024]
Abstract
Recent advances in fluorescence microscopy have now made it possible to measure the translation dynamics of individual RNA in living cells and in multiple colors. Here we describe a protocol that exploits these recent advances to simultaneously image the translation of two open reading frames encoded on a single reporter RNA yet frameshifted with respect to each other. This enables precise measurements of frameshifting dynamics and efficiency from specific frameshift stimulatory sequences, all with single-RNA precision.
Collapse
Affiliation(s)
- Kenneth R Lyon
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Tatsuya Morisaki
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Timothy J Stasevich
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
11
|
Amabile A, Phelan M, Yu Z, Silva P, Marks A, Morla-Folch J, Sohn M, Mollaoglu G, Falcomata C, Teunissen AJP, Brody JD, Dong Y, Brown BD. Bispecific antibody targeting of lipid nanoparticles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.20.629467. [PMID: 39763831 PMCID: PMC11702604 DOI: 10.1101/2024.12.20.629467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Abstract
Lipid nanoparticles (LNP) are the most clinically advanced non-viral gene delivery system. While progress has been made for enhancing delivery, cell specific targeting remains a challenge. Targeting moieties such as antibodies can be chemically-conjugated to LNPs however, this approach is complex and has challenges for scaling up. Here, we developed an approach to generate antibody-conjugated LNPs that utilizes a bispecific antibody (bsAb) as the targeting bridge. As a docking site for the bsAb, we generated LNPs with a short epitope, derived from hemagglutinin antigen (HA), embedded in the PEG component of the particle (LNPHA). We generated bsAb in which one domain binds HA and the other binds different cell surface proteins, including PD-L1, CD4, CD5, and SunTag. Non-chemical conjugation of the bsAb and LNP resulted in a major increase in the efficiency and specificity of transfecting cells expressing the cognate target. LNP/bsAb mediated a 4-fold increase in in vivo transfection of PD-L1 expressing cancer cells, and a 26-fold increase in ex vivo transfection of quiescent primary human T cells. Additionally, we created a universal bsAb recognizing HA and anti-rat IgG2, enabling LNP tethering to off-the-shelf antibodies such as CD4, CD8, CD20, CD45, and CD3. By utilizing a molecular dock and bsAb technology, these studies demonstrate a simple and effective strategy to generate antibody-conjugated LNPs, enabling precise and efficient mRNA delivery.
Collapse
Affiliation(s)
- Angelo Amabile
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY, USA
- RNA NanoCore - Lipid Nanoparticle Therapeutics Core, ISMMS, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, ISMMS, New York, New York, USA
| | - Matthew Phelan
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, ISMMS, New York, New York, USA
| | - Zhixin Yu
- RNA NanoCore - Lipid Nanoparticle Therapeutics Core, ISMMS, New York, NY, USA
| | - Pedro Silva
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY, USA
- RNA NanoCore - Lipid Nanoparticle Therapeutics Core, ISMMS, New York, NY, USA
| | - Adam Marks
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, ISMMS, New York, New York, USA
| | - Judit Morla-Folch
- Biomedical Engineering and Imaging Institute, ISMMS, New Yok, NY, USA
| | - Moah Sohn
- Marc and Jennifer Lipschultz Precision Immunology Institute, ISMMS, New York, New York, USA
| | - Gurkan Mollaoglu
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, ISMMS, New York, New York, USA
| | - Chiara Falcomata
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, ISMMS, New York, New York, USA
| | | | - Joshua D Brody
- Marc and Jennifer Lipschultz Precision Immunology Institute, ISMMS, New York, New York, USA
- Hematology and Medical Oncology, ISMMS, New York, New York, USA
| | - Yizhou Dong
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY, USA
- RNA NanoCore - Lipid Nanoparticle Therapeutics Core, ISMMS, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, ISMMS, New York, New York, USA
| | - Brian D Brown
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY, USA
- RNA NanoCore - Lipid Nanoparticle Therapeutics Core, ISMMS, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, ISMMS, New York, New York, USA
| |
Collapse
|
12
|
DeRoo J, Terry JS, Zhao N, Stasevich TJ, Snow CD, Geiss BJ. PAbFold: Linear Antibody Epitope Prediction using AlphaFold2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.19.590298. [PMID: 38659833 PMCID: PMC11042291 DOI: 10.1101/2024.04.19.590298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Defining the binding epitopes of antibodies is essential for understanding how they bind to their antigens and perform their molecular functions. However, while determining linear epitopes of monoclonal antibodies can be accomplished utilizing well-established empirical procedures, these approaches are generally labor- and time-intensive and costly. To take advantage of the recent advances in protein structure prediction algorithms available to the scientific community, we developed a calculation pipeline based on the localColabFold implementation of AlphaFold2 that can predict linear antibody epitopes by predicting the structure of the complex between antibody heavy and light chains and target peptide sequences derived from antigens. We found that this AlphaFold2 pipeline, which we call PAbFold, was able to accurately flag known epitope sequences for several well-known antibody targets (HA / Myc) when the target sequence was broken into small overlapping linear peptides and antibody complementarity determining regions (CDRs) were grafted onto several different antibody framework regions in the single-chain antibody fragment (scFv) format. To determine if this pipeline was able to identify the epitope of a novel antibody with no structural information publicly available, we determined the epitope of a novel anti-SARS-CoV-2 nucleocapsid targeted antibody using our method and then experimentally validated our computational results using peptide competition ELISA assays. These results indicate that the AlphaFold2-based PAbFold pipeline we developed is capable of accurately identifying linear antibody epitopes in a short time using just antibody and target protein sequences. This emergent capability of the method is sensitive to methodological details such as peptide length, AlphaFold2 neural network versions, and multiple-sequence alignment database. PAbFold is available at https://github.com/jbderoo/PAbFold.
Collapse
Affiliation(s)
- Jacob DeRoo
- School of Biomedical Engineering, Colorado State University, Fort Collins CO USA
| | - James S. Terry
- Department of Microbiology, Immunology, & Pathology, Colorado State University, Fort Collins CO USA
| | - Ning Zhao
- Department of Biochemistry and Molecular Genetics, University of Colorado-Anschutz Medical Campus, Aurora, CO USA
| | - Timothy J. Stasevich
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins CO USA
| | - Christopher D. Snow
- School of Biomedical Engineering, Colorado State University, Fort Collins CO USA
- Department of Chemical & Biological Engineering, Colorado State University, Fort Collins CO USA
| | - Brian J. Geiss
- School of Biomedical Engineering, Colorado State University, Fort Collins CO USA
- Department of Microbiology, Immunology, & Pathology, Colorado State University, Fort Collins CO USA
| |
Collapse
|
13
|
Kareemo DJ, Winborn CS, Olah SS, Miller CN, Kim J, Kadgien CA, Actor-Engel HS, Ramsay HJ, Ramsey AM, Aoto J, Kennedy MJ. Genetically encoded intrabody probes for labeling and manipulating AMPA-type glutamate receptors. Nat Commun 2024; 15:10374. [PMID: 39613728 PMCID: PMC11607441 DOI: 10.1038/s41467-024-54530-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 11/14/2024] [Indexed: 12/01/2024] Open
Abstract
Tools for visualizing and manipulating protein dynamics in living cells are critical for understanding cellular function. Here we leverage recently available monoclonal antibody sequences to generate a set of affinity tags for labeling and manipulating AMPA-type glutamate receptors (AMPARs), which mediate nearly all excitatory neurotransmission in the central nervous system. These antibodies can be produced from heterologous cells for exogenous labeling applications or directly expressed in living neurons as intrabodies, where they bind their epitopes in the endoplasmic reticulum and co-traffic to the cell surface for visualization with cell impermeant fluorescent dyes. We show these reagents do not perturb AMPAR trafficking, function, mobility, or synaptic recruitment during plasticity and therefore can be used as probes for monitoring endogenous receptors in living neurons. We also adapt these reagents to deplete AMPARs from the cell surface by trapping them in the endoplasmic reticulum, providing a simple approach for loss of excitatory neurotransmission. The strategies outlined here serve as a template for generating similar reagents targeting diverse proteins as more antibody sequences become available.
Collapse
Affiliation(s)
- Dean J Kareemo
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Christina S Winborn
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Samantha S Olah
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Carley N Miller
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - JungMin Kim
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Chelsie A Kadgien
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Hannah S Actor-Engel
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Harrison J Ramsay
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Austin M Ramsey
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Jason Aoto
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Matthew J Kennedy
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
14
|
Gonzalez-Magaldi M, Gullapalli A, Papoulas O, Liu C, Leung AYH, Guo L, Brilot A, Marcotte EM, Ke Z, Leahy DJ. Structure and organization of full-length Epidermal Growth Factor Receptor in extracellular vesicles by cryo-electron tomography. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.25.625301. [PMID: 39651119 PMCID: PMC11623583 DOI: 10.1101/2024.11.25.625301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
We report here transport of the Epidermal Growth Factor Receptor (EGFR), Insulin Receptor, 7-pass transmembrane receptor Smoothened, and 13-pass Sodium-iodide symporter to extracellular vesicles (EVs) for structural and functional studies. Mass spectrometry confirmed the transported proteins as the most abundant in EV membranes, and the presence of many receptor-interacting proteins demonstrates the utility of EVs for characterizing membrane protein interactomes. Cryo-electron tomography of EGFR-containing EVs reveals that EGFR forms clusters in the presence of EGF with a ∼3 nm gap between the inner membrane and cytoplasmic density. EGFR extracellular regions do not form regular arrays, suggesting that clustering is mediated by the intracellular region. Subtomogram averaging of the EGFR extracellular region (ECR) yielded a 15 Å map into which the crystal structure of the ligand-bound EGFR ECR dimer fits well. These findings refine our understanding of EGFR activation, clustering, and signaling, and they establish EVs as a versatile platform for structural and functional characterization of human membrane proteins in a native-like environment. Significance Statement Atomic or near-atomic resolution structural studies of proteins embedded in cell membranes have proven challenging. We show that transporting integral membrane proteins to cell-derived extracellular vesicles enables structural and functional studies of human membrane proteins in a native membrane environment. We have used this approach to visualize an active form of full-length Epidermal Growth Factor Receptor (EGFR) and show that it forms clusters in the membrane and projects its cytoplasmic signaling domains ∼3 nm away from the membrane surface. EGFR is essential for normal development, but abnormal EGFR activity is associated with several human cancers and is the target of many anticancer therapies. Our studies refine current models of how ligand binding to EGFR transmits signals across cell membranes.
Collapse
|
15
|
Ell CM, Safyan A, Chayengia M, Kustermann MMM, Lorenz J, Schächtle M, Pyrowolakis G. A genome-engineered tool set for Drosophila TGF-β/BMP signaling studies. Development 2024; 151:dev204222. [PMID: 39494616 DOI: 10.1242/dev.204222] [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: 07/01/2024] [Accepted: 10/15/2024] [Indexed: 11/05/2024]
Abstract
Ligands of the TGF-β/BMP superfamily are crucially involved in the regulation of growth, patterning and organogenesis and can act as long-range morphogens. Essential for understanding TGF-β/BMP signaling dynamics and regulation are tools that allow monitoring and manipulating pathway components at physiological expression levels and endogenous spatiotemporal patterns. We used genome engineering to generate a comprehensive library of endogenously epitope- or fluorescent-tagged versions of receptors, co-receptors, transcription factors and key feedback regulators of the Drosophila BMP and Activin signaling pathways. We demonstrate that the generated alleles are biologically active and can be used for assessing tissue and subcellular distribution of the corresponding proteins. Furthermore, we show that the genomic platforms can be used for in locus structure-function and cis-regulatory analyses. Finally, we present a complementary set of protein binder-based tools, which allow visualization as well as manipulation of the stability and subcellular localization of epitope-tagged proteins, providing new tools for the analysis of BMP signaling and beyond.
Collapse
Affiliation(s)
- Clara-Maria Ell
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany
- CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
- Institute for Biology I, Faculty of Biology, HMH, Habsburgerstr. 49, University of Freiburg, 79104 Freiburg, Germany
| | - Abu Safyan
- CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
- Institute for Biology I, Faculty of Biology, HMH, Habsburgerstr. 49, University of Freiburg, 79104 Freiburg, Germany
- International Max Planck Research School for Immunobiology, Epigenetics, and Metabolism, 79108 Freiburg, Germany
| | - Mrinal Chayengia
- Institute for Biology I, Faculty of Biology, HMH, Habsburgerstr. 49, University of Freiburg, 79104 Freiburg, Germany
| | - Manuela M M Kustermann
- Institute for Biology I, Faculty of Biology, HMH, Habsburgerstr. 49, University of Freiburg, 79104 Freiburg, Germany
| | - Jennifer Lorenz
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany
- Institute for Biology I, Faculty of Biology, HMH, Habsburgerstr. 49, University of Freiburg, 79104 Freiburg, Germany
| | - Melanie Schächtle
- Institute for Biology I, Faculty of Biology, HMH, Habsburgerstr. 49, University of Freiburg, 79104 Freiburg, Germany
| | - George Pyrowolakis
- CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
- Institute for Biology I, Faculty of Biology, HMH, Habsburgerstr. 49, University of Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
16
|
Kays I, Chen BE. Tracking and measuring local protein synthesis in vivo. Development 2024; 151:dev202908. [PMID: 39373391 DOI: 10.1242/dev.202908] [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: 03/27/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024]
Abstract
Detecting when and how much of a protein molecule is synthesized is important for understanding cell function, but current methods either cannot be performed in vivo or have poor temporal resolution. Here, we developed a technique to detect and quantify subcellular protein synthesis events in real time in vivo. This Protein Translation Reporting (PTR) technique uses a genetic tag that produces a stoichiometric ratio of a small peptide portion of a split fluorescent protein and the protein of interest during protein synthesis. We show that the split fluorescent protein peptide can generate fluorescence within milliseconds upon binding the larger portion of the fluorescent protein, and that the fluorescence intensity is directly proportional to the number of molecules of the protein of interest synthesized. Using PTR, we tracked and measured protein synthesis events in single cells over time in vivo. We use different color split fluorescent proteins to detect multiple genes or alleles in single cells simultaneously. We also split a photoswitchable fluorescent protein to photoconvert the reconstituted fluorescent protein to a different channel to continually reset the time of detection of synthesis events.
Collapse
Affiliation(s)
- Ibrahim Kays
- Centre for Research in Neuroscience, Research Institute of the McGill University Health Centre, Montréal, Québec, H3G 1A4, Canada
| | - Brian E Chen
- Centre for Research in Neuroscience, Research Institute of the McGill University Health Centre, Montréal, Québec, H3G 1A4, Canada
- Departments of Medicine and Neurology & Neurosurgery, McGill University, Montréal, Québec, H3G 1A4, Canada
| |
Collapse
|
17
|
Schnider ST, Vigano MA, Affolter M, Aguilar G. Functionalized Protein Binders in Developmental Biology. Annu Rev Cell Dev Biol 2024; 40:119-142. [PMID: 39038471 DOI: 10.1146/annurev-cellbio-112122-025214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Developmental biology has greatly profited from genetic and reverse genetic approaches to indirectly studying protein function. More recently, nanobodies and other protein binders derived from different synthetic scaffolds have been used to directly dissect protein function. Protein binders have been fused to functional domains, such as to lead to protein degradation, relocalization, visualization, or posttranslational modification of the target protein upon binding. The use of such functionalized protein binders has allowed the study of the proteome during development in an unprecedented manner. In the coming years, the advent of the computational design of protein binders, together with further advances in scaffold engineering and synthetic biology, will fuel the development of novel protein binder-based technologies. Studying the proteome with increased precision will contribute to a better understanding of the immense molecular complexities hidden in each step along the way to generate form and function during development.
Collapse
Affiliation(s)
| | | | | | - Gustavo Aguilar
- Current affiliation: Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA;
- Biozentrum, Universität Basel, Basel, Switzerland;
| |
Collapse
|
18
|
Bellec M, Chen R, Dhayni J, Trullo A, Avinens D, Karaki H, Mazzarda F, Lenden-Hasse H, Favard C, Lehmann R, Bertrand E, Lagha M, Dufourt J. Boosting the toolbox for live imaging of translation. RNA (NEW YORK, N.Y.) 2024; 30:1374-1394. [PMID: 39060168 PMCID: PMC11404453 DOI: 10.1261/rna.080140.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 06/30/2024] [Indexed: 07/28/2024]
Abstract
Live imaging of translation based on tag recognition by a single-chain antibody is a powerful technique to assess translation regulation in living cells. However, this approach is challenging and requires optimization in terms of expression level and detection sensitivity of the system, especially in a multicellular organism. Here, we improved existing fluorescent tools and developed new ones to image and quantify nascent translation in the living Drosophila embryo and in mammalian cells. We tested and characterized five different green fluorescent protein variants fused to the single-chain fragment variable (scFv) and uncovered photobleaching, aggregation, and intensity disparities. Using different strengths of germline and somatic drivers, we determined that the availability of the scFv is critical in order to detect translation throughout development. We introduced a new translation imaging method based on a nanobody/tag system named ALFA-array, allowing the sensitive and simultaneous detection of the translation of several distinct mRNA species. Finally, we developed a largely improved RNA imaging system based on an MCP-tdStaygold fusion.
Collapse
Affiliation(s)
- Maëlle Bellec
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, 34293 Montpellier, France
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Ruoyu Chen
- Vilcek Institute of Graduate Studies, NYU School of Medicine, New York 10016, USA
- Whitehead Institute for Biomedical Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Jana Dhayni
- Institut de Génétique Humaine, University of Montpellier, CNRS, 34396 Montpellier, France
| | - Antonello Trullo
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, 34293 Montpellier, France
| | - Damien Avinens
- Institut de Recherche en Infectiologie de Montpellier, CNRS UMR 9004, University of Montpellier, Montpellier, 34293 Cedex 5, France
| | - Hussein Karaki
- Institut de Génétique Humaine, University of Montpellier, CNRS, 34396 Montpellier, France
| | - Flavia Mazzarda
- Institut de Génétique Humaine, University of Montpellier, CNRS, 34396 Montpellier, France
| | - Helene Lenden-Hasse
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, 34293 Montpellier, France
| | - Cyril Favard
- Institut de Recherche en Infectiologie de Montpellier, CNRS UMR 9004, University of Montpellier, Montpellier, 34293 Cedex 5, France
| | - Ruth Lehmann
- Whitehead Institute for Biomedical Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Edouard Bertrand
- Institut de Génétique Humaine, University of Montpellier, CNRS, 34396 Montpellier, France
| | - Mounia Lagha
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, 34293 Montpellier, France
| | - Jeremy Dufourt
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, 34293 Montpellier, France
- Institut de Recherche en Infectiologie de Montpellier, CNRS UMR 9004, University of Montpellier, Montpellier, 34293 Cedex 5, France
| |
Collapse
|
19
|
Li XC, Zhu XY, Wang YY, Tong SL, Chen ZL, Lu ZY, Zhang JH, Song LL, Wang XH, Zhang C, Sun YH, Zhong CY, Su LH, Wang LX, Huang XY. Canagliflozin alleviates pulmonary hypertension by activating PPARγ and inhibiting its S225 phosphorylation. Acta Pharmacol Sin 2024; 45:1861-1878. [PMID: 38719955 PMCID: PMC11335861 DOI: 10.1038/s41401-024-01286-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/07/2024] [Indexed: 08/22/2024]
Abstract
Pulmonary hypertension (PH) is a progressive fatal disease with no cure. Canagliflozin (CANA), a novel medication for diabetes, has been found to have remarkable cardiovascular benefits. However, few studies have addressed the effect and pharmacological mechanism of CANA in the treatment of PH. Therefore, our study aimed to investigate the effect and pharmacological mechanism of CANA in treating PH. First, CANA suppressed increased pulmonary artery pressure, right ventricular hypertrophy, and vascular remodeling in both mouse and rat PH models. Network pharmacology, transcriptomics, and biological results suggested that CANA could ameliorate PH by suppressing excessive oxidative stress and pulmonary artery smooth muscle cell proliferation partially through the activation of PPARγ. Further studies demonstrated that CANA inhibited phosphorylation of PPARγ at Ser225 (a novel serine phosphorylation site in PPARγ), thereby promoting the nuclear translocation of PPARγ and increasing its ability to resist oxidative stress and proliferation. Taken together, our study not only highlighted the potential pharmacological effect of CANA on PH but also revealed that CANA-induced inhibition of PPARγ Ser225 phosphorylation increases its capacity to counteract oxidative stress and inhibits proliferation. These findings may stimulate further research and encourage future clinical trials exploring the therapeutic potential of CANA in PH treatment.
Collapse
Affiliation(s)
- Xiu-Chun Li
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China
| | - Xia-Yan Zhu
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China
| | - Yang-Yue Wang
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China
| | | | - Zhi-Li Chen
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China
| | - Zi-Yi Lu
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China
| | | | - Lan-Lan Song
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China
| | - Xing-Hong Wang
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China
| | - Chi Zhang
- Wenzhou Medical University, Wenzhou, 325000, China
| | - Yi-Han Sun
- Wenzhou Medical University, Wenzhou, 325000, China
| | | | - Li-Huang Su
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China
| | - Liang-Xing Wang
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China
| | - Xiao-Ying Huang
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China.
| |
Collapse
|
20
|
Yao Y, Lou X, Jin L, Sun W, Liu J, Chen Y, Cheng S, Zhao T, Ke S, Zhang L, Xu Y, He L, Li H. Optogenetic Strategies for Optimizing the Performance of Phospholipids Biosensors. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403026. [PMID: 39073033 PMCID: PMC11422808 DOI: 10.1002/advs.202403026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 07/07/2024] [Indexed: 07/30/2024]
Abstract
High-performance biosensors play a crucial role in elucidating the intricate spatiotemporal regulatory roles and dynamics of membrane phospholipids. However, enhancing the sensitivity and imaging performance remains a significant challenge. Here, optogenetic-based strategies are presented to optimize phospholipid biosensors. These strategies involves presequestering unbound biosensors in the cell nucleus and regulating their cytosolic levels with blue light to minimize background signal interference in phospholipid detection, particularly under conditions of high expression levels of biosensor. Furthermore, optically controlled phase separation and the SunTag system are employed to generate punctate probes for substrate detection, thereby amplifying biosensor signals and enhancing visualization of the detection process. These improved phospholipid biosensors hold great potential for enhancing the understanding of the spatiotemporal dynamics and regulatory roles of membrane lipids in live cells and the methodological insights in this study might be valuable for developing other high-performance biosensors.
Collapse
Affiliation(s)
- Yuanfa Yao
- Institute of PharmacologyCollege of Pharmaceutical ScienceZhejiang University of TechnologyHangzhou310014China
| | - Xiayan Lou
- Institute of PharmacologyCollege of Pharmaceutical ScienceZhejiang University of TechnologyHangzhou310014China
| | - Luhong Jin
- School of Information Science and TechnologyHangzhou Normal UniversityHangzhouZhejiang311121China
| | - Weiyun Sun
- Institute of PharmacologyCollege of Pharmaceutical ScienceZhejiang University of TechnologyHangzhou310014China
| | - Jingfang Liu
- Department of Biomedical EngineeringKey Laboratory of Biomedical Engineering of Ministry of EducationState Key Laboratory of Extreme Photonics and InstrumentationZhejiang Provincial Key Laboratory of Cardio‐Cerebral Vascular Detection Technology and Medicinal Effectiveness AppraisalZhejiang UniversityHangzhou310027China
| | - Yunyue Chen
- Department of Biomedical EngineeringKey Laboratory of Biomedical Engineering of Ministry of EducationState Key Laboratory of Extreme Photonics and InstrumentationZhejiang Provincial Key Laboratory of Cardio‐Cerebral Vascular Detection Technology and Medicinal Effectiveness AppraisalZhejiang UniversityHangzhou310027China
| | - Sunying Cheng
- Institute of PharmacologyCollege of Pharmaceutical ScienceZhejiang University of TechnologyHangzhou310014China
| | - Tengjiao Zhao
- Institute of PharmacologyCollege of Pharmaceutical ScienceZhejiang University of TechnologyHangzhou310014China
| | - Shuwei Ke
- Institute of PharmacologyCollege of Pharmaceutical ScienceZhejiang University of TechnologyHangzhou310014China
| | - Luhao Zhang
- School of Information Science and TechnologyHangzhou Normal UniversityHangzhouZhejiang311121China
| | - Yingke Xu
- Department of Biomedical EngineeringKey Laboratory of Biomedical Engineering of Ministry of EducationState Key Laboratory of Extreme Photonics and InstrumentationZhejiang Provincial Key Laboratory of Cardio‐Cerebral Vascular Detection Technology and Medicinal Effectiveness AppraisalZhejiang UniversityHangzhou310027China
- Department of EndocrinologyChildren's Hospital of Zhejiang University School of MedicineNational Clinical Research Center for Children's HealthHangzhouZhejiang310051China
| | - Lian He
- Department of PharmacologyJoint Laboratory of Guangdong‐Hong Kong Universities for Vascular Homeostasis and DiseasesSchool of MedicineSouthern University of Science and TechnologyShenzhen518055China
| | - Hanbing Li
- Institute of PharmacologyCollege of Pharmaceutical ScienceZhejiang University of TechnologyHangzhou310014China
| |
Collapse
|
21
|
Abstract
The translation of messenger RNA (mRNA) into proteins represents the culmination of gene expression. Recent technological advances have revolutionized our ability to investigate this process with unprecedented precision, enabling the study of translation at the single-molecule level in real time within live cells. In this review, we provide an overview of single-mRNA translation reporters. We focus on the core technology, as well as the rapid development of complementary probes, tags, and accessories that enable the visualization and quantification of a wide array of translation dynamics. We then highlight notable studies that have utilized these reporters in model systems to address key biological questions. The high spatiotemporal resolution of these studies is shedding light on previously unseen phenomena, uncovering the full heterogeneity and complexity of translational regulation.
Collapse
Affiliation(s)
- Tatsuya Morisaki
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado, USA;
| | - O'Neil Wiggan
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado, USA;
| | - Timothy J Stasevich
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado, USA;
- Cell Biology Center and World Research Hub Initiative, Tokyo Institute of Technology, Yokohama, Japan
| |
Collapse
|
22
|
Jones G, Akter Y, Shifflett V, Hruska M. Nanoscale analysis of functionally diverse glutamatergic synapses in the neocortex reveals input and layer-specific organization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.01.592008. [PMID: 38746319 PMCID: PMC11092571 DOI: 10.1101/2024.05.01.592008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Discovery of synaptic nanostructures suggests a molecular logic for the flexibility of synaptic function. We still have little understanding of how functionally diverse synapses in the brain organize their nanoarchitecture due to challenges associated with super-resolution imaging in complex brain tissue. Here, we characterized single-domain camelid nanobodies for the 3D quantitative multiplex imaging of synaptic nano-organization in 6 µm brain cryosections using STED nanoscopy. We focused on thalamocortical (TC) and corticocortical (CC) synapses along the apical-basal axis of layer 5 pyramidal neurons as models of functionally diverse glutamatergic synapses in the brain. Spines receiving TC input were larger than CC spines in all layers examined. However, TC synapses on apical and basal dendrites conformed to different organizational principles. TC afferents on apical dendrites frequently contacted spines with multiple aligned PSD-95/Bassoon nanomodules, which are larger. TC spines on basal dendrites contained mostly one aligned PSD-95/Bassoon nanocluster. However, PSD-95 nanoclusters were larger and scaled with spine volume. The nano-organization of CC synapses did not change across cortical layers. These results highlight striking nanoscale diversity of functionally distinct glutamatergic synapses, relying on afferent input and sub-cellular localization of individual synaptic connections.
Collapse
|
23
|
Xu J, Zhu N, Du Y, Han T, Zheng X, Li J, Zhu S. Biomimetic NIR-II fluorescent proteins created from chemogenic protein-seeking dyes for multicolor deep-tissue bioimaging. Nat Commun 2024; 15:2845. [PMID: 38565859 PMCID: PMC10987503 DOI: 10.1038/s41467-024-47063-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 03/19/2024] [Indexed: 04/04/2024] Open
Abstract
Near-infrared-I/II fluorescent proteins (NIR-I/II FPs) are crucial for in vivo imaging, yet the current NIR-I/II FPs face challenges including scarcity, the requirement for chromophore maturation, and limited emission wavelengths (typically < 800 nm). Here, we utilize synthetic protein-seeking NIR-II dyes as chromophores, which covalently bind to tag proteins (e.g., human serum albumin, HSA) through a site-specific nucleophilic substitution reaction, thereby creating proof-of-concept biomimetic NIR-II FPs. This chemogenic protein-seeking strategy can be accomplished under gentle physiological conditions without catalysis. Proteomics analysis identifies specific binding site (Cys 477 on DIII). NIR-II FPs significantly enhance chromophore brightness and photostability, while improving biocompatibility, allowing for high-performance NIR-II lymphography and angiography. This strategy is universal and applicable in creating a wide range of spectrally separated NIR-I/II FPs for real-time visualization of multiple biological events. Overall, this straightforward biomimetic approach holds the potential to transform fluorescent protein-based bioimaging and enables in-situ albumin targeting to create NIR-I/II FPs for deep-tissue imaging in live organisms.
Collapse
Affiliation(s)
- Jiajun Xu
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun, 130021, P.R. China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, P.R. China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Materials Science, Hebei University, Baoding, 071002, P.R. China
| | - Ningning Zhu
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun, 130021, P.R. China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, P.R. China
| | - Yijing Du
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun, 130021, P.R. China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, P.R. China
| | - Tianyang Han
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun, 130021, P.R. China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, P.R. China
| | - Xue Zheng
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun, 130021, P.R. China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, P.R. China
| | - Jia Li
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun, 130021, P.R. China
| | - Shoujun Zhu
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun, 130021, P.R. China.
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, P.R. China.
| |
Collapse
|
24
|
Tan QH, Otgonbaatar A, Kaur P, Ga AF, Harmston NP, Tolwinski NS. The Wnt Co-Receptor PTK7/Otk and Its Homolog Otk-2 in Neurogenesis and Patterning. Cells 2024; 13:365. [PMID: 38474329 PMCID: PMC10930971 DOI: 10.3390/cells13050365] [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: 11/20/2023] [Revised: 02/09/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Wnt signaling is a highly conserved metazoan pathway that plays a crucial role in cell fate determination and morphogenesis during development. Wnt ligands can induce disparate cellular responses. The exact mechanism behind these different outcomes is not fully understood but may be due to interactions with different receptors on the cell membrane. PTK7/Otk is a transmembrane receptor that is implicated in various developmental and physiological processes including cell polarity, cell migration, and invasion. Here, we examine two roles of Otk-1 and Otk-2 in patterning and neurogenesis. We find that Otk-1 is a positive regulator of signaling and Otk-2 functions as its inhibitor. We propose that PTK7/Otk functions in signaling, cell migration, and polarity contributing to the diversity of cellular responses seen in Wnt-mediated processes.
Collapse
Affiliation(s)
- Qian Hui Tan
- Division of Science, Yale-NUS College, Singapore 138527, Singapore (A.O.); (P.K.); (A.F.G.); (N.P.H.)
| | - Agimaa Otgonbaatar
- Division of Science, Yale-NUS College, Singapore 138527, Singapore (A.O.); (P.K.); (A.F.G.); (N.P.H.)
| | - Prameet Kaur
- Division of Science, Yale-NUS College, Singapore 138527, Singapore (A.O.); (P.K.); (A.F.G.); (N.P.H.)
| | - Angelica Faye Ga
- Division of Science, Yale-NUS College, Singapore 138527, Singapore (A.O.); (P.K.); (A.F.G.); (N.P.H.)
| | - Nathan P. Harmston
- Division of Science, Yale-NUS College, Singapore 138527, Singapore (A.O.); (P.K.); (A.F.G.); (N.P.H.)
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore 169857, Singapore
- Molecular Biosciences Division, Cardiff School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | - Nicholas S. Tolwinski
- Division of Science, Yale-NUS College, Singapore 138527, Singapore (A.O.); (P.K.); (A.F.G.); (N.P.H.)
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore 169857, Singapore
| |
Collapse
|
25
|
Lee SE, Chang S. nArgBP2 together with GKAP and SHANK3 forms a dynamic layered structure. Front Cell Neurosci 2024; 18:1354900. [PMID: 38440150 PMCID: PMC10909995 DOI: 10.3389/fncel.2024.1354900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/06/2024] [Indexed: 03/06/2024] Open
Abstract
nArgBP2, a protein whose disruption is implicated in intellectual disability, concentrates in excitatory spine-synapses. By forming a triad with GKAP and SHANK, it regulates spine structural rearrangement. We here find that GKAP and SHANK3 concentrate close to the synaptic contact, whereas nArgBP2 concentrates more centrally in the spine. The three proteins collaboratively form biomolecular condensates in living fibroblasts, exhibiting distinctive layered localizations. nArgBP2 concentrates in the inner phase, SHANK3 in the outer phase, and GKAP partially in both. Upon co-expression of GKAP and nArgBP2, they evenly distribute within condensates, with a notable peripheral localization of SHANK3 persisting when co-expressed with either GKAP or nArgBP2. Co-expression of SHANK3 and GKAP with CaMKIIα results in phase-in-phase condensates, with CaMKIIα at the central locus and SHANK3 and GKAP exhibiting peripheral localization. Additional co-expression of nArgBP2 maintains the layered organizational structure within condensates. Subsequent CaMKIIα activation disperses a majority of the condensates, with an even distribution of all proteins within the extant deformed condensates. Our findings suggest that protein segregation via phase separation may contribute to establishing layered organization in dendritic spines.
Collapse
Affiliation(s)
- Sang-Eun Lee
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Sunghoe Chang
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
26
|
Juan T, Bellec M, Cardoso B, Athéa H, Fukuda N, Albu M, Günther S, Looso M, Stainier DYR. Control of cardiac contractions using Cre-lox and degron strategies in zebrafish. Proc Natl Acad Sci U S A 2024; 121:e2309842121. [PMID: 38194447 PMCID: PMC10801847 DOI: 10.1073/pnas.2309842121] [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: 06/12/2023] [Accepted: 11/27/2023] [Indexed: 01/11/2024] Open
Abstract
Cardiac contractions and hemodynamic forces are essential for organ development and homeostasis. Control over cardiac contractions can be achieved pharmacologically or optogenetically. However, these approaches lack specificity or require direct access to the heart. Here, we compare two genetic approaches to control cardiac contractions by modulating the levels of the essential sarcomeric protein Tnnt2a in zebrafish. We first recombine a newly generated tnnt2a floxed allele using multiple lines expressing Cre under the control of cardiomyocyte-specific promoters, and show that it does not recapitulate the tnnt2a/silent heart mutant phenotype in embryos. We show that this lack of early cardiac contraction defects is due, at least in part, to the long half-life of tnnt2a mRNA, which masks the gene deletion effects until the early larval stages. We then generate an endogenous Tnnt2a-eGFP fusion line that we use together with the zGRAD system to efficiently degrade Tnnt2a in all cardiomyocytes. Using single-cell transcriptomics, we find that Tnnt2a depletion leads to cardiac phenotypes similar to those observed in tnnt2a mutants, with a loss of blood and pericardial flow-dependent cell types. Furthermore, we achieve conditional degradation of Tnnt2a-eGFP by splitting the zGRAD protein into two fragments that, when combined with the cpFRB2-FKBP system, can be reassembled upon rapamycin treatment. Thus, this Tnnt2a degradation line enables non-invasive control of cardiac contractions with high spatial and temporal specificity and will help further understand how they shape organ development and homeostasis.
Collapse
Affiliation(s)
- Thomas Juan
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim61231, Germany
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz- Kreislaufforschung), Bad Nauheim61231, Germany
- Cardio-Pulmonary Institute, Bad Nauheim61231, Germany
| | - Maëlle Bellec
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim61231, Germany
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz- Kreislaufforschung), Bad Nauheim61231, Germany
| | - Bárbara Cardoso
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim61231, Germany
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz- Kreislaufforschung), Bad Nauheim61231, Germany
| | - Héloïse Athéa
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim61231, Germany
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz- Kreislaufforschung), Bad Nauheim61231, Germany
| | - Nana Fukuda
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim61231, Germany
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz- Kreislaufforschung), Bad Nauheim61231, Germany
| | - Marga Albu
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim61231, Germany
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz- Kreislaufforschung), Bad Nauheim61231, Germany
| | - Stefan Günther
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz- Kreislaufforschung), Bad Nauheim61231, Germany
- Cardio-Pulmonary Institute, Bad Nauheim61231, Germany
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim61231, Germany
| | - Mario Looso
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz- Kreislaufforschung), Bad Nauheim61231, Germany
- Cardio-Pulmonary Institute, Bad Nauheim61231, Germany
- Bioinformatics Core Unit, Max Planck Institute for Heart and Lung Research, Bad Nauheim61231, Germany
| | - Didier Y. R. Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim61231, Germany
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz- Kreislaufforschung), Bad Nauheim61231, Germany
- Cardio-Pulmonary Institute, Bad Nauheim61231, Germany
| |
Collapse
|
27
|
Harris MT, Marr MT. The intrinsically disordered region of eIF5B stimulates IRES usage and nucleates biological granule formation. Cell Rep 2023; 42:113283. [PMID: 37862172 PMCID: PMC10680144 DOI: 10.1016/j.celrep.2023.113283] [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: 08/27/2022] [Revised: 03/22/2023] [Accepted: 09/29/2023] [Indexed: 10/22/2023] Open
Abstract
Cells activate stress response pathways to survive adverse conditions. Such responses involve the inhibition of global cap-dependent translation. This inhibition is a block that essential transcripts must escape via alternative methods of translation initiation, e.g., an internal ribosome entry site (IRES). IRESs have distinct structures and generally require a limited repertoire of translation factors. Cellular IRESs have been identified in many critical cellular stress response transcripts. We previously identified cellular IRESs in the murine insulin receptor (Insr) and insulin-like growth factor 1 receptor (Igf1r) transcripts and demonstrated their resistance to eukaryotic initiation factor 4F (eIF4F) inhibition. Here, we find that eIF5B preferentially promotes Insr, Igf1r, and hepatitis C virus IRES activity through a non-canonical mechanism that requires its highly charged and disordered N terminus. We find that the N-terminal region of eIF5B can drive cytoplasmic granule formation. This eIF5B granule is triggered by cellular stress and is sufficient to specifically promote IRES activity.
Collapse
Affiliation(s)
- Meghan T Harris
- Department of Biology and Rosenstiel Basic Medical Sciences Research Center, Brandeis University, Waltham, MA 02453, USA
| | - Michael T Marr
- Department of Biology and Rosenstiel Basic Medical Sciences Research Center, Brandeis University, Waltham, MA 02453, USA.
| |
Collapse
|
28
|
Eichenberger BT, Griesbach E, Mitchell J, Chao JA. Following the Birth, Life, and Death of mRNAs in Single Cells. Annu Rev Cell Dev Biol 2023; 39:253-275. [PMID: 37843928 DOI: 10.1146/annurev-cellbio-022723-024045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2023]
Abstract
Recent advances in single-molecule imaging of mRNAs in fixed and living cells have enabled the lives of mRNAs to be studied with unprecedented spatial and temporal detail. These approaches have moved beyond simply being able to observe specific events and have begun to allow an understanding of how regulation is coupled between steps in the mRNA life cycle. Additionally, these methodologies are now being applied in multicellular systems and animals to provide more nuanced insights into the physiological regulation of RNA metabolism.
Collapse
Affiliation(s)
- Bastian T Eichenberger
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland;
- University of Basel, Basel, Switzerland
| | - Esther Griesbach
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland;
| | - Jessica Mitchell
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland;
| | - Jeffrey A Chao
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland;
| |
Collapse
|
29
|
Zhang X, Wang L, Huang R, Wang J, Yan Q. Perfluoro-tert-butyl Group-Derived Capmatinib: Synthesis, Biological Evaluation and Its Application in 19 F Magnetic Resonance Imaging. Chembiochem 2023; 24:e202300354. [PMID: 37345408 DOI: 10.1002/cbic.202300354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/17/2023] [Accepted: 06/20/2023] [Indexed: 06/23/2023]
Abstract
Capmatinib is an FDA-approved drug to treat metastatic non-small cell lung cancer with MET-exon 14 skipping. Herein, the perfluoro-tert-butyl group, which possesses nine chemically identical fluorine atoms, was introduced on Capmatinib to afford a targeted 19 F magnetic resonance imaging (MRI) probe, perfluoro-tert-butyl group-derived Capmatinib (9F-CAP). The 19 F MRI concentration limit was found to be 25 mM in FLASH sequence. Molecular docking simulation, surface plasmon resonance (SPR) (with a Kd of 40.7 μM), half-inhibitory concentration (with a IC50 of 168 nM), Annexin V, and cytotoxicity assays jointly demonstrated that the 9F-CAP targeted cMET protein specifically. Therefore, the targeted imaging capability of 9F-CAP is of great significance for the preoperative diagnosis of specific cancers.
Collapse
Affiliation(s)
- Xinnan Zhang
- Key Laboratory for Advanced Materials and Feringa Nobel, Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai, 200237, China
| | - Luting Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Rd., Shanghai, 201203, China
| | - Ruimin Huang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Rd., Shanghai, 201203, China
| | - Jingbo Wang
- Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Rd., Shanghai, 200025, China
| | - Qifan Yan
- Key Laboratory for Advanced Materials and Feringa Nobel, Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai, 200237, China
| |
Collapse
|
30
|
Mishra PK, Sharma N, Kim H, Lee C, Rhee HW. GEN-Click: Genetically Encodable Click Reactions for Spatially Restricted Metabolite Labeling. ACS CENTRAL SCIENCE 2023; 9:1650-1657. [PMID: 37637744 PMCID: PMC10450880 DOI: 10.1021/acscentsci.3c00511] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Indexed: 08/29/2023]
Abstract
Chemical reactions for the in situ modification of biomolecules within living cells are under development. Among these reactions, bio-orthogonal reactions such as click chemistry using copper(I) and Staudinger ligation are widely used for specific biomolecule tracking in live systems. However, currently available live cell copper(I)-catalyzed azide/alkyne cycloaddition reactions are not designed in a spatially resolved manner. Therefore, we developed the "GEN-Click" system, which can target the copper(I)-catalyzed azide/alkyne cycloaddition reaction catalysts proximal to the protein of interest and can be genetically expressed in a live cell. The genetically controlled, spatially restricted, metal-catalyzed biorthogonal reaction can be used for proximity biotin labeling of various azido-bearing biomolecules (e.g., protein, phospholipid, oligosaccharides) in living cell systems. Using GEN-Click, we successfully detected local metabolite-transferring events at cell-cell contact sites.
Collapse
Affiliation(s)
| | - Nirmali Sharma
- Department
of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyunwoo Kim
- Department
of Biological Sciences, Ulsan National Institute
of Science and Technology, Ulsan 44919, Republic
of Korea
| | - Changwook Lee
- Department
of Biological Sciences, Ulsan National Institute
of Science and Technology, Ulsan 44919, Republic
of Korea
| | - Hyun-Woo Rhee
- Department
of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
- School
of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
31
|
Rudd JC, Maity S, Grunkemeyer JA, Snyder JC, Lovas S, Hansen LA. Membrane structure and internalization dynamics of human Flower isoforms hFWE3 and hFWE4 indicate a conserved endocytic role for hFWE4. J Biol Chem 2023; 299:104945. [PMID: 37348560 PMCID: PMC10366549 DOI: 10.1016/j.jbc.2023.104945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 06/24/2023] Open
Abstract
Human Flower (hFWE) isoforms hFWE1-4 are putative transmembrane (TM) proteins that reportedly mediate fitness comparisons during cell competition through extracellular display of their C-terminal tails. Isoform topology, subcellular localization, and duration of plasma membrane presentation are essential to this function. However, disagreement persists regarding the structure of orthologous fly and mouse FWEs, and experimental evidence for hFWE isoform subcellular localization or membrane structure is lacking. Here, we used AlphaFold2 and subsequent molecular dynamics-based structural predictions to construct epitope-tagged hFWE3 and hFWE4, the most abundant human isoforms, for experimental determination of their structure and internalization dynamics. We demonstrate that hFWE3 resides in the membrane of the endoplasmic reticulum (ER), while hFWE4 partially colocalizes with Rab4-, Rab5-, and Rab11-positive vesicles as well as with the plasma membrane. An array of imaging techniques revealed that hFWE4 positions both N- and C-terminal tails and a loop between second and third TM segments within the cytosol, while small (4-12aa) loops between the first and second and the third and fourth TM segments are either exposed to the extracellular space or within the lumen of cytoplasmic vesicles. Similarly, we found hFWE3 positions both N- and C-terminal tails in the cytosol, while a short loop between TM domains extends into the ER lumen. Finally, we demonstrate that hFWE4 exists only transiently at the cell surface and is rapidly internalized in an AP-2- and dynamin-1-dependent manner. Collectively, these data are consistent with a conserved role for hFWE4 in endocytic processes.
Collapse
Affiliation(s)
- Justin C Rudd
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, Nebraska, USA
| | - Sibaprasad Maity
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, Nebraska, USA
| | - James A Grunkemeyer
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, Nebraska, USA
| | - Joshua C Snyder
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA; Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Sándor Lovas
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, Nebraska, USA
| | - Laura A Hansen
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, Nebraska, USA.
| |
Collapse
|
32
|
Hertzog M, Erdel F. The Material Properties of the Cell Nucleus: A Matter of Scale. Cells 2023; 12:1958. [PMID: 37566037 PMCID: PMC10416959 DOI: 10.3390/cells12151958] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/26/2023] [Accepted: 07/26/2023] [Indexed: 08/12/2023] Open
Abstract
Chromatin regulatory processes physically take place in the environment of the cell nucleus, which is filled with the chromosomes and a plethora of smaller biomolecules. The nucleus contains macromolecular assemblies of different sizes, from nanometer-sized protein complexes to micrometer-sized biomolecular condensates, chromosome territories, and nuclear bodies. This multiscale organization impacts the transport processes within the nuclear interior, the global mechanical properties of the nucleus, and the way the nucleus senses and reacts to mechanical stimuli. Here, we discuss recent work on these aspects, including microrheology and micromanipulation experiments assessing the material properties of the nucleus and its subcomponents. We summarize how the properties of multiscale media depend on the time and length scales probed in the experiment, and we reconcile seemingly contradictory observations made on different scales. We also revisit the concept of liquid-like and solid-like material properties for complex media such as the nucleus. We propose that the nucleus can be considered a multiscale viscoelastic medium composed of three major components with distinct properties: the lamina, the chromatin network, and the nucleoplasmic fluid. This multicomponent organization enables the nucleus to serve its different functions as a reaction medium on the nanoscale and as a mechanosensor and structural scaffold on the microscale.
Collapse
Affiliation(s)
| | - Fabian Erdel
- MCD, Center for Integrative Biology (CBI), University of Toulouse, CNRS, 169 Avenue Marianne Grunberg-Manago, 31062 Toulouse, France
| |
Collapse
|
33
|
Boeynaems S, Dorone Y, Zhuang Y, Shabardina V, Huang G, Marian A, Kim G, Sanyal A, Şen NE, Griffith D, Docampo R, Lasker K, Ruiz-Trillo I, Auburger G, Holehouse AS, Kabashi E, Lin Y, Gitler AD. Poly(A)-binding protein is an ataxin-2 chaperone that regulates biomolecular condensates. Mol Cell 2023; 83:2020-2034.e6. [PMID: 37295429 PMCID: PMC10318123 DOI: 10.1016/j.molcel.2023.05.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 03/14/2023] [Accepted: 05/17/2023] [Indexed: 06/12/2023]
Abstract
Biomolecular condensation underlies the biogenesis of an expanding array of membraneless assemblies, including stress granules (SGs), which form under a variety of cellular stresses. Advances have been made in understanding the molecular grammar of a few scaffold proteins that make up these phases, but how the partitioning of hundreds of SG proteins is regulated remains largely unresolved. While investigating the rules that govern the condensation of ataxin-2, an SG protein implicated in neurodegenerative disease, we unexpectedly identified a short 14 aa sequence that acts as a condensation switch and is conserved across the eukaryote lineage. We identify poly(A)-binding proteins as unconventional RNA-dependent chaperones that control this regulatory switch. Our results uncover a hierarchy of cis and trans interactions that fine-tune ataxin-2 condensation and reveal an unexpected molecular function for ancient poly(A)-binding proteins as regulators of biomolecular condensate proteins. These findings may inspire approaches to therapeutically target aberrant phases in disease.
Collapse
Affiliation(s)
- Steven Boeynaems
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Therapeutic Innovation Center (THINC), Baylor College of Medicine, Houston, TX 77030, USA; Center for Alzheimer's and Neurodegenerative Diseases (CAND), Texas Children's Hospital, Houston, TX 77030, USA; Dan L Duncan Comprehensive Cancer Center (DLDCCC), Baylor College of Medicine, Houston, TX 77030, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA.
| | - Yanniv Dorone
- Department of Plant Biology, Carnegie Institution for Science, Stanford, CA 94305, USA; Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Yanrong Zhuang
- IDG/McGovern Institute for Brain Research, Tsinghua-Peking Joint Centre for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Victoria Shabardina
- Institut de Biologia Evolutiva (CSIC-Universitat Pompeu Fabra), Passeig Marítim de la Barceloneta 37-49, Barcelona 08003 Catalonia, Spain
| | - Guozhong Huang
- Department of Cellular Biology and Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602, USA
| | - Anca Marian
- Imagine Institute, Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1163, Paris Descartes Université, 75015 Paris, France
| | - Garam Kim
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Anushka Sanyal
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Nesli-Ece Şen
- Experimental Neurology, Goethe-University Hospital, 60590 Frankfurt, Germany
| | - Daniel Griffith
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA; Center for Biomolecular Condensates, Washington University in St Louis, St. Louis, MO 63130, USA
| | - Roberto Docampo
- Department of Cellular Biology and Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602, USA
| | - Keren Lasker
- The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Iñaki Ruiz-Trillo
- Institut de Biologia Evolutiva (CSIC-Universitat Pompeu Fabra), Passeig Marítim de la Barceloneta 37-49, Barcelona 08003 Catalonia, Spain; ICREA, Passeig Lluís Companys 23, Barcelona 08010 Catalonia, Spain
| | - Georg Auburger
- Experimental Neurology, Goethe-University Hospital, 60590 Frankfurt, Germany
| | - Alex S Holehouse
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA; Center for Biomolecular Condensates, Washington University in St Louis, St. Louis, MO 63130, USA
| | - Edor Kabashi
- Imagine Institute, Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1163, Paris Descartes Université, 75015 Paris, France
| | - Yi Lin
- IDG/McGovern Institute for Brain Research, Tsinghua-Peking Joint Centre for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Aaron D Gitler
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|
34
|
Tsirkas I, Zur T, Dovrat D, Paleiov Z, Ravkaie L, Aharoni A. Enhanced fluorescent imaging of proteins in live yeast cells using fluorescently labeled scFv. STAR Protoc 2023; 4:102299. [PMID: 37270779 DOI: 10.1016/j.xpro.2023.102299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/25/2023] [Accepted: 04/20/2023] [Indexed: 06/06/2023] Open
Abstract
Fluorescent labeling of proteins is a widespread approach for the microscopic examination of protein function, expression, and localization in the cell. Here, we present a protocol for the labeling of hemagglutinin (HA)-tagged protein of interest (POI) with the single-chain antibody (scFv) 2E2 fused to different fluorescent proteins (FPs) in Saccharomyces cerevisiae. We describe steps for expressing 2E2-FP, and HA tagging and labeling of POI. We detail in vivo fluorescent imaging of proteins at different cellular compartments and with diverse expression levels. For complete details on the use and execution of this protocol, please refer to Tsirkas et al. (2022).1.
Collapse
Affiliation(s)
- Ioannis Tsirkas
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Be'er Sheva 84105, Israel.
| | - Tomer Zur
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Be'er Sheva 84105, Israel
| | - Daniel Dovrat
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Be'er Sheva 84105, Israel
| | - Zohar Paleiov
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Be'er Sheva 84105, Israel
| | - Lior Ravkaie
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Be'er Sheva 84105, Israel
| | - Amir Aharoni
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Be'er Sheva 84105, Israel.
| |
Collapse
|
35
|
Hanson EK, Whelan RJ. Application of the Nicoya OpenSPR to Studies of Biomolecular Binding: A Review of the Literature from 2016 to 2022. SENSORS (BASEL, SWITZERLAND) 2023; 23:4831. [PMID: 37430747 DOI: 10.3390/s23104831] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 07/12/2023]
Abstract
The Nicoya OpenSPR is a benchtop surface plasmon resonance (SPR) instrument. As with other optical biosensor instruments, it is suitable for the label-free interaction analysis of a diverse set of biomolecules, including proteins, peptides, antibodies, nucleic acids, lipids, viruses, and hormones/cytokines. Supported assays include affinity/kinetics characterization, concentration analysis, yes/no assessment of binding, competition studies, and epitope mapping. OpenSPR exploits localized SPR detection in a benchtop platform and can be connected with an autosampler (XT) to perform automated analysis over an extended time period. In this review article, we provide a comprehensive survey of the 200 peer-reviewed papers published between 2016 and 2022 that use the OpenSPR platform. We highlight the range of biomolecular analytes and interactions that have been investigated using the platform, provide an overview on the most common applications for the instrument, and point out some representative research that highlights the flexibility and utility of the instrument.
Collapse
Affiliation(s)
- Eliza K Hanson
- Department of Chemistry, University of Kansas, Lawrence, KS 66045, USA
| | - Rebecca J Whelan
- Department of Chemistry, University of Kansas, Lawrence, KS 66045, USA
| |
Collapse
|
36
|
Stevens AJ, Harris AR, Gerdts J, Kim KH, Trentesaux C, Ramirez JT, McKeithan WL, Fattahi F, Klein OD, Fletcher DA, Lim WA. Programming multicellular assembly with synthetic cell adhesion molecules. Nature 2023; 614:144-152. [PMID: 36509107 PMCID: PMC9892004 DOI: 10.1038/s41586-022-05622-z] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/02/2022] [Indexed: 12/14/2022]
Abstract
Cell adhesion molecules are ubiquitous in multicellular organisms, specifying precise cell-cell interactions in processes as diverse as tissue development, immune cell trafficking and the wiring of the nervous system1-4. Here we show that a wide array of synthetic cell adhesion molecules can be generated by combining orthogonal extracellular interactions with intracellular domains from native adhesion molecules, such as cadherins and integrins. The resulting molecules yield customized cell-cell interactions with adhesion properties that are similar to native interactions. The identity of the intracellular domain of the synthetic cell adhesion molecules specifies interface morphology and mechanics, whereas diverse homotypic or heterotypic extracellular interaction domains independently specify the connectivity between cells. This toolkit of orthogonal adhesion molecules enables the rationally programmed assembly of multicellular architectures, as well as systematic remodelling of native tissues. The modularity of synthetic cell adhesion molecules provides fundamental insights into how distinct classes of cell-cell interfaces may have evolved. Overall, these tools offer powerful abilities for cell and tissue engineering and for systematically studying multicellular organization.
Collapse
Affiliation(s)
- Adam J Stevens
- UCSF Cell Design Institute, University of California, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
- Center for Cellular Construction, University of California, San Francisco, CA, USA
| | - Andrew R Harris
- Center for Cellular Construction, University of California, San Francisco, CA, USA
- Department of Bioengineering, University of California, Berkeley, CA, USA
- Department of Mechanical and Aerospace Engineering, Carleton University, Ottawa, Ontario, Canada
| | - Josiah Gerdts
- UCSF Cell Design Institute, University of California, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
- Center for Cellular Construction, University of California, San Francisco, CA, USA
- Department of Neurology, Weill Institute for Neuroscience, University of California, San Francisco, CA, USA
| | - Ki H Kim
- UCSF Cell Design Institute, University of California, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
- Center for Cellular Construction, University of California, San Francisco, CA, USA
| | - Coralie Trentesaux
- Program in Craniofacial Biology, University of California, San Francisco, CA, USA
- Department of Orofacial Sciences, University of California, San Francisco, CA, USA
| | - Jonathan T Ramirez
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
| | - Wesley L McKeithan
- UCSF Cell Design Institute, University of California, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
- Center for Cellular Construction, University of California, San Francisco, CA, USA
- Maze Therapeutics, San Francisco, CA, USA
| | - Faranak Fattahi
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
| | - Ophir D Klein
- Program in Craniofacial Biology, University of California, San Francisco, CA, USA
- Department of Orofacial Sciences, University of California, San Francisco, CA, USA
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Daniel A Fletcher
- Center for Cellular Construction, University of California, San Francisco, CA, USA
- Department of Bioengineering, University of California, Berkeley, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Wendell A Lim
- UCSF Cell Design Institute, University of California, San Francisco, CA, USA.
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.
- Center for Cellular Construction, University of California, San Francisco, CA, USA.
| |
Collapse
|
37
|
Sisila V, Indhu M, Radhakrishnan J, Ayyadurai N. Building biomaterials through genetic code expansion. Trends Biotechnol 2023; 41:165-183. [PMID: 35908989 DOI: 10.1016/j.tibtech.2022.07.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 06/30/2022] [Accepted: 07/08/2022] [Indexed: 01/24/2023]
Abstract
Genetic code expansion (GCE) enables directed incorporation of noncoded amino acids (NCAAs) and unnatural amino acids (UNAAs) into the active core that confers dedicated structure and function to engineered proteins. Many protein biomaterials are tandem repeats that intrinsically include NCAAs generated through post-translational modifications (PTMs) to execute assigned functions. Conventional genetic engineering approaches using prokaryotic systems have limited ability to biosynthesize functionally active biomaterials with NCAAs/UNAAs. Codon suppression and reassignment introduce NCAAs/UNAAs globally, allowing engineered proteins to be redesigned to mimic natural matrix-cell interactions for tissue engineering. Expanding the genetic code enables the engineering of biomaterials with catechols - growth factor mimetics that modulate cell-matrix interactions - thereby facilitating tissue-specific expression of genes and proteins. This method of protein engineering shows promise in achieving tissue-informed, tissue-compliant tunable biomaterials.
Collapse
Affiliation(s)
- Valappil Sisila
- Department of Biochemistry and Biotechnology, Council of Scientific and Industrial Research (CSIR) Central Leather Research Institute (CLRI), Chennai, Tamil Nadu 600020, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India
| | - Mohan Indhu
- Department of Biochemistry and Biotechnology, Council of Scientific and Industrial Research (CSIR) Central Leather Research Institute (CLRI), Chennai, Tamil Nadu 600020, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India
| | - Janani Radhakrishnan
- Department of Biochemistry and Biotechnology, Council of Scientific and Industrial Research (CSIR) Central Leather Research Institute (CLRI), Chennai, Tamil Nadu 600020, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India.
| | - Niraikulam Ayyadurai
- Department of Biochemistry and Biotechnology, Council of Scientific and Industrial Research (CSIR) Central Leather Research Institute (CLRI), Chennai, Tamil Nadu 600020, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India.
| |
Collapse
|
38
|
Cnpy3 2xHA mice reveal neuronal expression of Cnpy3 in the brain. J Neurosci Methods 2023; 383:109730. [PMID: 36280087 DOI: 10.1016/j.jneumeth.2022.109730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/04/2022] [Accepted: 10/17/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Identification of biallelic CNPY3 mutations in patients with epileptic encephalopathy and abnormal electroencephalography findings of Cnpy3 knock-out mice have indicated that the loss of CNPY3 function causes neurological disorders such as epilepsy. However, the basic property of CNPY3 in the brain remains unclear. NEW METHOD We generated C-terminal 2xHA-tag knock-in Cnpy3 mice by i-GONAD in vivo genome editing system to investigate the expression and function of Cnpy3 in the mouse brain. RESULTS 2xHA-tagged Cnpy3 was confirmed by immunoblot analysis using anti-HA and CNPY3 antibodies, although HA tagging caused the decreased Cnpy3 protein level. Immunohistochemical analysis of Cnpy32xHA knock-in mice showed that Cnpy3-2xHA was predominantly expressed in the neuron. In addition, Cnpy3 and Cnpy3-2xHA were both localized in the endoplasmic reticulum and synaptosome and showed age-dependent expression changes in the brain. COMPARISON WITH EXISTING METHODS Conventional Cnpy3 antibodies could not allow us to investigate the distribution of Cnpy3 expression in the brain, while HA-tagging revealed the expression of CNPY3 in neuronal cells. CONCLUSIONS Taken together, we demonstrated that Cnpy32xHA knock-in mice would be useful to further elucidate the property of Cnpy3 in brain function and neurological disorders.
Collapse
|
39
|
Tsirkas I, Zur T, Dovrat D, Cohen A, Ravkaie L, Aharoni A. Protein fluorescent labeling in live yeast cells using scFv-based probes. CELL REPORTS METHODS 2022; 2:100357. [PMID: 36590693 PMCID: PMC9795370 DOI: 10.1016/j.crmeth.2022.100357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 09/19/2022] [Accepted: 11/10/2022] [Indexed: 12/12/2022]
Abstract
The fusion of fluorescent proteins (FPs) to endogenous proteins is a widespread approach for microscopic examination of protein function, expression, and localization in the cell. However, proteins that are sensitive to FP fusion or expressed at low levels are difficult to monitor using this approach. Here, we develop a single-chain fragment variable (scFv)-FP approach to efficiently label Saccharomyces cerevisiae proteins that are tagged with repeats of hemagglutinin (HA)-tag sequences. We demonstrate the successful labeling of DNA-binding proteins and proteins localized to different cellular organelles including the nuclear membrane, peroxisome, Golgi apparatus, and mitochondria. This approach can lead to a significant increase in fluorescence intensity of the labeled protein, allows C'-terminal labeling of difficult-to-tag proteins and increased detection sensitivity of DNA-damage foci. Overall, the development of a scFv-FP labeling approach in yeast provides a general and simple tool for the function and localization analysis of the yeast proteome.
Collapse
Affiliation(s)
- Ioannis Tsirkas
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Be’er Sheva 84105, Israel
| | - Tomer Zur
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Be’er Sheva 84105, Israel
| | - Daniel Dovrat
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Be’er Sheva 84105, Israel
| | - Amit Cohen
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Be’er Sheva 84105, Israel
| | - Lior Ravkaie
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Be’er Sheva 84105, Israel
| | - Amir Aharoni
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Be’er Sheva 84105, Israel
| |
Collapse
|
40
|
DeLuca KF, Mick JE, DeLuca JG. Production and purification of recombinant monoclonal antibodies from human cells based on a primary sequence. STAR Protoc 2022; 3:101915. [PMID: 36595892 PMCID: PMC9763751 DOI: 10.1016/j.xpro.2022.101915] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/26/2022] [Accepted: 11/16/2022] [Indexed: 12/14/2022] Open
Abstract
There are challenges to using commercially available antibodies generated in animals, including concerns with reproducibility, high costs, and ethical issues. Here, we present a protocol for generating and purifying recombinant antibodies from human HEK293 suspension culture cells from a primary sequence. We describe the steps to generate antibody heavy and light chain plasmids, followed by transfection of the plasmids into cells and purification of antibodies. This protocol can produce high-yield recombinant monoclonal antibodies at a relatively low cost. For complete details on the use and execution of this protocol, please refer to DeLuca et al. (2021).1.
Collapse
Affiliation(s)
- Keith F DeLuca
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Jeanne E Mick
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Jennifer G DeLuca
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
41
|
Design and synthesis of DNA hydrogel based on EXPAR and CRISPR/Cas14a for ultrasensitive detection of creatine kinase MB. Biosens Bioelectron 2022; 218:114792. [DOI: 10.1016/j.bios.2022.114792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 10/06/2022] [Indexed: 11/20/2022]
|
42
|
Morales-Polanco F, Lee JH, Barbosa NM, Frydman J. Cotranslational Mechanisms of Protein Biogenesis and Complex Assembly in Eukaryotes. Annu Rev Biomed Data Sci 2022; 5:67-94. [PMID: 35472290 PMCID: PMC11040709 DOI: 10.1146/annurev-biodatasci-121721-095858] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The formation of protein complexes is crucial to most biological functions. The cellular mechanisms governing protein complex biogenesis are not yet well understood, but some principles of cotranslational and posttranslational assembly are beginning to emerge. In bacteria, this process is favored by operons encoding subunits of protein complexes. Eukaryotic cells do not have polycistronic mRNAs, raising the question of how they orchestrate the encounter of unassembled subunits. Here we review the constraints and mechanisms governing eukaryotic co- and posttranslational protein folding and assembly, including the influence of elongation rate on nascent chain targeting, folding, and chaperone interactions. Recent evidence shows that mRNAs encoding subunits of oligomeric assemblies can undergo localized translation and form cytoplasmic condensates that might facilitate the assembly of protein complexes. Understanding the interplay between localized mRNA translation and cotranslational proteostasis will be critical to defining protein complex assembly in vivo.
Collapse
Affiliation(s)
| | - Jae Ho Lee
- Department of Biology, Stanford University, Stanford, California, USA;
| | - Natália M Barbosa
- Department of Biology, Stanford University, Stanford, California, USA;
| | - Judith Frydman
- Department of Biology, Stanford University, Stanford, California, USA;
- Department of Genetics, Stanford University, Stanford, California, USA
| |
Collapse
|
43
|
Cialek CA, Galindo G, Morisaki T, Zhao N, Montgomery TA, Stasevich TJ. Imaging translational control by Argonaute with single-molecule resolution in live cells. Nat Commun 2022; 13:3345. [PMID: 35688806 PMCID: PMC9187665 DOI: 10.1038/s41467-022-30976-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 05/24/2022] [Indexed: 11/29/2022] Open
Abstract
A major challenge to our understanding of translational control has been deconvolving the individual impact specific regulatory factors have on the complex dynamics of mRNA translation. MicroRNAs (miRNAs), for example, guide Argonaute and associated proteins to target mRNAs, where they direct gene silencing in multiple ways that are not well understood. To better deconvolve these dynamics, we have developed technology to directly visualize and quantify the impact of human Argonaute2 (Ago2) on the translation and subcellular localization of individual reporter mRNAs in living cells. We show that our combined translation and Ago2 tethering sensor reflects endogenous miRNA-mediated gene silencing. Using the sensor, we find that Ago2 association leads to progressive silencing of translation at individual mRNA. Silencing was occasionally interrupted by brief bursts of translational activity and took 3–4 times longer than a single round of translation, consistent with a gradual increase in the inhibition of translation initiation. At later time points, Ago2-tethered mRNAs cluster and coalesce with P-bodies, where a translationally silent state is maintained. These results provide a framework for exploring miRNA-mediated gene regulation in live cells at the single-molecule level. Furthermore, our tethering-based, single-molecule reporter system will likely have wide-ranging application in studying RNA-protein interactions. Guided by miRNA, Argonaute proteins silence mRNA in multiple ways that are not well understood. Here, the authors develop live-cell biosensors to image the impact tethered regulatory factors, such as Argonaute, have on single-mRNA translation dynamics.
Collapse
Affiliation(s)
- Charlotte A Cialek
- Department of Biochemistry & Molecular Biology, Colorado State University, Fort Collins, CO, 80523, USA
| | - Gabriel Galindo
- Department of Biochemistry & Molecular Biology, Colorado State University, Fort Collins, CO, 80523, USA
| | - Tatsuya Morisaki
- Department of Biochemistry & Molecular Biology, Colorado State University, Fort Collins, CO, 80523, USA
| | - Ning Zhao
- Department of Biochemistry & Molecular Biology, Colorado State University, Fort Collins, CO, 80523, USA
| | - Taiowa A Montgomery
- Department of Biology, Colorado State University, Fort Collins, CO, 80523, USA.
| | - Timothy J Stasevich
- Department of Biochemistry & Molecular Biology, Colorado State University, Fort Collins, CO, 80523, USA. .,Cell Biology Center and World Research Hub Initiative, Tokyo Institute of Technology, Yokohama, Japan.
| |
Collapse
|
44
|
Murakawa T, Nakamura T, Kawaguchi K, Murayama F, Zhao N, Stasevich TJ, Kimura H, Fujita N. A Drosophila toolkit for HA-tagged proteins unveils a block in autophagy flux in the last instar larval fat body. Development 2022; 149:dev200243. [PMID: 35319746 DOI: 10.1242/dev.200243] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 01/27/2022] [Indexed: 01/04/2025]
Abstract
For in vivo functional analysis of a protein of interest (POI), multiple transgenic strains with a POI that harbors different tags are needed but generation of these strains is still labor-intensive work. To overcome this, we have developed a versatile Drosophila toolkit with a genetically encoded single-chain variable fragment for the HA epitope tag: 'HA Frankenbody'. This system allows various analyses of HA-tagged POI in live tissues by simply crossing an HA Frankenbody fly with an HA-tagged POI fly. Strikingly, the GFP-mCherry tandem fluorescent-tagged HA Frankenbody revealed a block in autophagic flux and an accumulation of enlarged autolysosomes in the last instar larval and prepupal fat body. Mechanistically, lysosomal activity was downregulated at this stage, and endocytosis, but not autophagy, was indispensable for the swelling of lysosomes. Furthermore, forced activation of lysosomes by fat body-targeted overexpression of Mitf, the single MiTF/TFE family gene in Drosophila, suppressed the lysosomal swelling and resulted in pupal lethality. Collectively, we propose that downregulated lysosomal function in the fat body plays a role in the metamorphosis of Drosophila.
Collapse
Affiliation(s)
- Tadayoshi Murakawa
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, 4259-S2-11 Nagatsuta-cho, Midori-ku, Yokohama 226-8503, Japan
- Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokohama 226-8503, Japan
| | - Tsuyoshi Nakamura
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, 4259-S2-11 Nagatsuta-cho, Midori-ku, Yokohama 226-8503, Japan
| | - Kohei Kawaguchi
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, 4259-S2-11 Nagatsuta-cho, Midori-ku, Yokohama 226-8503, Japan
| | - Futoshi Murayama
- Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokohama 226-8503, Japan
| | - Ning Zhao
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| | - Timothy J Stasevich
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
- World Research Hub Initiative, Tokyo Institute of Technology, Yokohama 226-8503, Japan
| | - Hiroshi Kimura
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, 4259-S2-11 Nagatsuta-cho, Midori-ku, Yokohama 226-8503, Japan
- Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokohama 226-8503, Japan
- World Research Hub Initiative, Tokyo Institute of Technology, Yokohama 226-8503, Japan
| | - Naonobu Fujita
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, 4259-S2-11 Nagatsuta-cho, Midori-ku, Yokohama 226-8503, Japan
- Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokohama 226-8503, Japan
- Precursory Research for Embryonic Science & Technology (PRESTO), Japan Science & Technology Agency (JST), 4-1-8 Honcho Kawaguchi, Saitama 332-0012, Japan
| |
Collapse
|
45
|
Uchino S, Ito Y, Sato Y, Handa T, Ohkawa Y, Tokunaga M, Kimura H. Live imaging of transcription sites using an elongating RNA polymerase II-specific probe. J Cell Biol 2022; 221:212888. [PMID: 34854870 PMCID: PMC8647360 DOI: 10.1083/jcb.202104134] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 10/12/2021] [Accepted: 11/10/2021] [Indexed: 12/15/2022] Open
Abstract
In eukaryotic nuclei, most genes are transcribed by RNA polymerase II (RNAP2), whose regulation is a key to understanding the genome and cell function. RNAP2 has a long heptapeptide repeat (Tyr1-Ser2-Pro3-Thr4-Ser5-Pro6-Ser7), and Ser2 is phosphorylated on an elongation form. To detect RNAP2 Ser2 phosphorylation (RNAP2 Ser2ph) in living cells, we developed a genetically encoded modification-specific intracellular antibody (mintbody) probe. The RNAP2 Ser2ph-mintbody exhibited numerous foci, possibly representing transcription “factories,” and foci were diminished during mitosis and in a Ser2 kinase inhibitor. An in vitro binding assay using phosphopeptides confirmed the mintbody’s specificity. RNAP2 Ser2ph-mintbody foci were colocalized with proteins associated with elongating RNAP2 compared with factors involved in the initiation. These results support the view that mintbody localization represents the sites of RNAP2 Ser2ph in living cells. RNAP2 Ser2ph-mintbody foci showed constrained diffusional motion like chromatin, but they were more mobile than DNA replication domains and p300-enriched foci, suggesting that the elongating RNAP2 complexes are separated from more confined chromatin domains.
Collapse
Affiliation(s)
- Satoshi Uchino
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Yuma Ito
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Yuko Sato
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan.,Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| | - Tetsuya Handa
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Makio Tokunaga
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Hiroshi Kimura
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan.,Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| |
Collapse
|
46
|
Xu J, Kim AR, Cheloha RW, Fischer FA, Li JSS, Feng Y, Stoneburner E, Binari R, Mohr SE, Zirin J, Ploegh HL, Perrimon N. Protein visualization and manipulation in Drosophila through the use of epitope tags recognized by nanobodies. eLife 2022; 11:74326. [PMID: 35076390 PMCID: PMC8853664 DOI: 10.7554/elife.74326] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
Expansion of the available repertoire of reagents for visualization and manipulation of proteins will help understand their function. Short epitope tags linked to proteins of interest and recognized by existing binders such as nanobodies facilitate protein studies by obviating the need to isolate new antibodies directed against them. Nanobodies have several advantages over conventional antibodies, as they can be expressed and used as tools for visualization and manipulation of proteins in vivo. Here, we characterize two short (<15aa) NanoTag epitopes, 127D01 and VHH05, and their corresponding high-affinity nanobodies. We demonstrate their use in Drosophila for in vivo protein detection and re-localization, direct and indirect immunofluorescence, immunoblotting, and immunoprecipitation. We further show that CRISPR-mediated gene targeting provides a straightforward approach to tagging endogenous proteins with the NanoTags. Single copies of the NanoTags, regardless of their location, suffice for detection. This versatile and validated toolbox of tags and nanobodies will serve as a resource for a wide array of applications, including functional studies in Drosophila and beyond.
Collapse
Affiliation(s)
- Jun Xu
- Department of Genetics, Harvard Medical School
| | - Ah-Ram Kim
- Department of Genetics, Harvard Medical School
| | | | | | | | - Yuan Feng
- Department of Genetics, Harvard Medical School
| | | | | | | | | | | | | |
Collapse
|
47
|
Gunasekara H, Munaweera R, Novotná L, Lillemeier BF, Hu YS. Chaotropic Perturbation of Noncovalent Interactions of the Hemagglutinin Tag Monoclonal Antibody Fragment Enables Superresolution Molecular Census. ACS NANO 2022; 16:129-139. [PMID: 34797055 PMCID: PMC11196025 DOI: 10.1021/acsnano.1c04237] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Antibody-antigen interactions represent one of the most exploited biomolecular interactions in experimental biology. While numerous techniques harnessed immobilized antibodies for nanoscale fluorescence imaging, few utilized their reversible binding kinetics. Here, we investigated noncovalent interactions of the monoclonal hemagglutinin (HA) epitope tag antibody, 12CA5, in the fixed cellular environment. We observed that the use of a chaotropic agent, potassium thiocyanate (KSCN), promoted the dissociation of the 12CA5 antibody fragment (Fab), which already displayed faster dissociation compared to its immunoglobulin G (IgG) counterpart. Molecular dynamic simulations revealed notable root-mean-square deviations and destabilizations in the presence of KSCN, while the hydrogen-bonding network remained primarily unaffected at the antigen-binding site. The reversible interactions enabled us to achieve a superresolution molecular census of local populations of 3xHA tagged microtubule fibers with improved molecular quantification consistency compared to single-molecule localization microscopy (SMLM) techniques utilizing standard immunofluorescence staining for sample labeling. Our technique, termed superresolution census of molecular epitope tags (SR-COMET), highlights the utilization of reversible antibody-antigen interactions for SMLM-based quantitative superresolution imaging.
Collapse
Affiliation(s)
- Hirushi Gunasekara
- Department of Chemistry, College of Liberal Arts and Sciences, University of Illinois at Chicago, Chicago, IL, 60607-7061, United States
| | - Rangika Munaweera
- Department of Chemistry, College of Liberal Arts and Sciences, University of Illinois at Chicago, Chicago, IL, 60607-7061, United States
| | - Lucie Novotná
- Nomis Center for Immunobiology and Microbial Pathogenesis & Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, La Jolla, California 92037, United States
| | - Björn F. Lillemeier
- Nomis Center for Immunobiology and Microbial Pathogenesis & Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, La Jolla, California 92037, United States
- Faculty of Biology and Centre for Integrative Biological Signalling Studies (CIBSS), Albert-Ludwigs-University of Freiburg, Freiburg 79104, Germany
| | - Ying S. Hu
- Department of Chemistry, College of Liberal Arts and Sciences, University of Illinois at Chicago, Chicago, IL, 60607-7061, United States
| |
Collapse
|
48
|
DeLuca KF, Mick JE, Ide AH, Lima WC, Sherman L, Schaller KL, Anderson SM, Zhao N, Stasevich TJ, Varma D, Nilsson J, DeLuca JG. Generation and diversification of recombinant monoclonal antibodies. eLife 2021; 10:72093. [PMID: 34970967 PMCID: PMC8763395 DOI: 10.7554/elife.72093] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 12/20/2021] [Indexed: 11/13/2022] Open
Abstract
Antibodies are indispensable tools used for a large number of applications in both foundational and translational bioscience research; however, there are drawbacks to using traditional antibodies generated in animals. These include a lack of standardization leading to problems with reproducibility, high costs of antibodies purchased from commercial sources, and ethical concerns regarding the large number of animals used to generate antibodies. To address these issues, we have developed practical methodologies and tools for generating low-cost, high-yield preparations of recombinant monoclonal antibodies and antibody fragments directed to protein epitopes from primary sequences. We describe these methods here, as well as approaches to diversify monoclonal antibodies, including customization of antibody species specificity, generation of genetically encoded small antibody fragments, and conversion of single chain antibody fragments (e.g. scFv) into full-length, bivalent antibodies. This study focuses on antibodies directed to epitopes important for mitosis and kinetochore function; however, the methods and reagents described here are applicable to antibodies and antibody fragments for use in any field.
Collapse
Affiliation(s)
- Keith F DeLuca
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, United States
| | - Jeanne E Mick
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, United States
| | - Amy Hodges Ide
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, United States
| | - Wanessa C Lima
- Geneva Antibody Facility, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Lori Sherman
- CU Cancer Center Cell Technologies Shared Resource, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Kristin L Schaller
- Department of Pediatric Hematology, Oncology and Bone Marrow Transplant, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Steven M Anderson
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Ning Zhao
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, United States
| | - Timothy J Stasevich
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, United States
| | - Dileep Varma
- Department of Cell and Developmental Biology, Northwestern University, Chicago, United States
| | - Jakob Nilsson
- The Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Germany
| | - Jennifer G DeLuca
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, United States
| |
Collapse
|
49
|
Zhang HN, Xue JB, Wang AZL, Jiang HW, Merugu SB, Li DW, Tao SC. EASINESS: E. coli Assisted Speedy affINity-maturation Evolution SyStem. Front Immunol 2021; 12:747267. [PMID: 34925322 PMCID: PMC8677947 DOI: 10.3389/fimmu.2021.747267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/10/2021] [Indexed: 11/13/2022] Open
Abstract
Antibodies are one of the most important groups of biomolecules for both clinical and basic research and have been developed as potential therapeutics. Affinity is the key feature for biological activity and clinical efficacy of an antibody, especially of therapeutic antibodies, and thus antibody affinity improvement is indispensable and still remains challenging. To address this issue, we developed the E. coli Assisted Speed affINity-maturation Evolution SyStem (EASINESS) for continuous directed evolution of Ag-Ab interactions. Two key components of EASINESS include a mutation system modified from error-prone DNA polymerase I (Pol I) that selectively mutates ColE1 plasmids in E. coli and a protein-protein interaction selection system from mDHFR split fragments. We designed a GCN4 variant which barely forms a homodimer, and during a single round of evolution, we reversed the homodimer formation activity from the GCN4 variant to verify the feasibility of EASINESS. We then selected a potential therapeutic antibody 18A4Hu and improved the affinity of the antibody (18A4Hu) to its target (ARG2) 12-fold in 7 days while requiring very limited hands-on time. Remarkably, these variants of 18A4Hu revealed a significant improved ability to inhibit melanoma pulmonary metastasis in a mouse model. These results indicate EASINESS could be as an attractive choice for antibody affinity maturation.
Collapse
Affiliation(s)
- Hai-Nan Zhang
- Shanghai Center for Systems Biomedicine, Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Jun-Biao Xue
- Shanghai Center for Systems Biomedicine, Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Aru Ze-Ling Wang
- Engineering Research Center of Cell and Therapeutic Antibody of Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - He-Wei Jiang
- Shanghai Center for Systems Biomedicine, Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Siva Bhararth Merugu
- Engineering Research Center of Cell and Therapeutic Antibody of Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Da-Wei Li
- Engineering Research Center of Cell and Therapeutic Antibody of Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Sheng-Ce Tao
- Shanghai Center for Systems Biomedicine, Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
50
|
Genetically encoded intrabodies as high-precision tools to visualize and manipulate neuronal function. Semin Cell Dev Biol 2021; 126:117-124. [PMID: 34782184 DOI: 10.1016/j.semcdb.2021.11.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 11/01/2021] [Accepted: 11/05/2021] [Indexed: 11/24/2022]
Abstract
Basic neuroscience research employs numerous forms of antibodies as key reagents in diverse applications. While the predominant use of antibodies is as immunolabeling reagents, neuroscientists are making increased use of intracellular antibodies or intrabodies. Intrabodies are recombinant antibodies genetically encoded for expression within neurons. These can be used to target various cargo (fluorescent proteins, reporters, enzymes, etc.) to specific molecules and subcellular domains to report on and manipulate neuronal function with high precision. Intrabodies have the advantages inherent in all genetically encoded recombinant antibodies but represent a distinct subclass in that their structure allows for their expression and function within cells. The high precision afforded by the ability to direct their expression to specific cell types, and the selective binding of intrabodies to targets within these allows intrabodies to offer unique advantages for neuroscience research, given the tremendous molecular, cellular and morphological complexity of brain neurons. Intrabodies expressed within neurons have been used for a variety of purposes in basic neuroscience research. Here I provide a general background to intrabodies and their development, and examples of their emerging utility as valuable basic neuroscience research tools.
Collapse
|