1
|
Hou J, Du K, Li J, Li Z, Cao S, Zhang S, Huang W, Liu H, Yang X, Sun S, Mo S, Qin T, Zhang X, Yin S, Nie X, Lu X. Research trends in the use of nanobodies for cancer therapy. J Control Release 2025; 381:113454. [PMID: 39922288 DOI: 10.1016/j.jconrel.2025.01.045] [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: 08/15/2024] [Revised: 01/15/2025] [Accepted: 01/17/2025] [Indexed: 02/10/2025]
Abstract
Although there are many challenges in using nanobodies for treating various complex tumor diseases, including rapid renal clearance and the complex blood-brain barrier environment, nanobodies have shown great potential due to their high antigen affinity, excellent tumor penetration ability, and favorable safety profile. Since the discovery of the variable domain (VHH) of camelid heavy-chain antibodies in 1993, nanobodies have been progressively applied to various cancer therapy platforms, such as antagonistic drugs and targeting agents for effector domains. In recent years, several nanobody-based drugs, including Caplacizumab, KN-035, and Ozoralizumab, have been approved for clinical use. Among them, KN-035 is used for treating advanced solid tumors, and these advancements have propelled nanobody development to new heights. Currently, nanobodies are being rapidly applied to the treatment of a wide range of diseases, from viral infections to cancer, demonstrating strong advantages in areas such as targeted protein degradation, bioimaging, nanobody-drug conjugation, bispecific T-cell engagers, and vaccine applications. Bibliometric tools, including CiteSpace, HisCite Pro, and Alluvial Generator, were employed to trace the historical development of nanobodies in cancer research. The contributions of authors, countries, and institutions in this field were analyzed, and research hotspots and emerging trends were identified through keyword analysis and influential articles. Future trends were also predicted. This study provides a unique, comprehensive, and objective perspective on the use of nanobodies in tumor research, laying a foundation for future research directions and offering valuable insights for researchers in the field.
Collapse
Affiliation(s)
- Jun Hou
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Kejiang Du
- Department of Otorhinolaryngology-Head and Neck Surgery, Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China; Liuzhou People's Hospital affiliated to Guangxi Medical University, Liuzhou 545006, China
| | - Jinling Li
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Zhenghui Li
- Department of Neurosurgery, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450000, China
| | - Shaorui Cao
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Shilin Zhang
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Wenxing Huang
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Heng Liu
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Xiaomei Yang
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Shuyang Sun
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Shanzhao Mo
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Tianyu Qin
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Xilei Zhang
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Shihua Yin
- Department of Otorhinolaryngology-Head and Neck Surgery, Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China.
| | - Xinyu Nie
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230002, China.
| | - Xiaoling Lu
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China.
| |
Collapse
|
2
|
Camlin NJ. Protein-targeting reverse genetic approaches: the future of oocyte and preimplantation embryo research. Mol Hum Reprod 2025; 31:gaaf008. [PMID: 40100642 PMCID: PMC12000532 DOI: 10.1093/molehr/gaaf008] [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: 11/22/2024] [Revised: 02/10/2025] [Indexed: 03/20/2025] Open
Abstract
Reverse genetic approaches are the standard in molecular biology to determine a protein's function. Traditionally, nucleic acid targeting via gene knockout (DNA) and knockdown (RNA) has been the method of choice to remove proteins-of-interest. However, the nature of mammalian oocyte maturation and preimplantation embryo development can make nucleic acid-targeting approaches difficult. Gene knockout allows time for compensatory mechanisms and secondary phenotypes to develop which can make interpretation of a protein's function difficult. Furthermore, genes can be essential for animal and/or oocyte survival, and therefore, gene knockout is not always a viable approach to investigate oocyte maturation and preimplantation embryo development. Conversely, RNA-targeting approaches, i.e. RNA interference (RNAi) and morpholinos, rely on protein half-life and therefore are unable to knockdown every protein-of-interest. An increasing number of reverse genetic approaches that directly target proteins have been developed to overcome the limitations of nucleic acid-based approaches, including Trim-Away and auxin-inducible degradation. These protein-targeting approaches give researchers exquisite and fast control of protein loss. This review will discuss how Trim-Away and auxin-inducible degradation can overcome many of the challenges of nucleic acid-based reverse genetic approaches. Furthermore, it highlights the unique research opportunities these approaches afford, such as targeting post-translationally modified proteins.
Collapse
Affiliation(s)
- Nicole J Camlin
- Cell and Molecular Biology, School of Biological, Environmental and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS, USA
| |
Collapse
|
3
|
Rega C, Tsitsa I, Roumeliotis TI, Krystkowiak I, Portillo M, Yu L, Vorhauser J, Pines J, Mansfeld J, Choudhary J, Davey NE. High resolution profiling of cell cycle-dependent protein and phosphorylation abundance changes in non-transformed cells. Nat Commun 2025; 16:2579. [PMID: 40089461 PMCID: PMC11910661 DOI: 10.1038/s41467-025-57537-8] [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: 06/20/2024] [Accepted: 02/24/2025] [Indexed: 03/17/2025] Open
Abstract
The cell cycle governs a precise series of molecular events, regulated by coordinated changes in protein and phosphorylation abundance, that culminates in the generation of two daughter cells. Here, we present a proteomic and phosphoproteomic analysis of the human cell cycle in hTERT-RPE-1 cells using deep quantitative mass spectrometry by isobaric labelling. By analysing non-transformed cells and improving the temporal resolution and coverage of key cell cycle regulators, we present a dataset of cell cycle-dependent protein and phosphorylation site oscillation that offers a foundational reference for investigating cell cycle regulation. These data reveal regulatory intricacies including proteins and phosphorylation sites exhibiting cell cycle-dependent oscillation, and proteins targeted for degradation during mitotic exit. Integrated with complementary resources, our data link cycle-dependent abundance dynamics to functional changes and are accessible through the Cell Cycle database (CCdb), an interactive web-based resource for the cell cycle community.
Collapse
Affiliation(s)
- Camilla Rega
- Division of Cancer Biology, The Institute of Cancer Research, London, UK
| | - Ifigenia Tsitsa
- Division of Cancer Biology, The Institute of Cancer Research, London, UK
| | | | | | - Maria Portillo
- Division of Cancer Biology, The Institute of Cancer Research, London, UK
| | - Lu Yu
- Division of Cancer Biology, The Institute of Cancer Research, London, UK
| | - Julia Vorhauser
- Division of Cancer Biology, The Institute of Cancer Research, London, UK
| | - Jonathon Pines
- Division of Cancer Biology, The Institute of Cancer Research, London, UK
| | - Jörg Mansfeld
- Division of Cancer Biology, The Institute of Cancer Research, London, UK
| | - Jyoti Choudhary
- Division of Cancer Biology, The Institute of Cancer Research, London, UK
| | - Norman E Davey
- Division of Cancer Biology, The Institute of Cancer Research, London, UK.
| |
Collapse
|
4
|
Pöschko P, Berrou CM, Pakari K, Ziegler MJ, Kern C, Koch B, Wittbrodt J, Wombacher R. Photoactivatable Plant Hormone-Based Chemical Inducers of Proximity for In Vivo Applications. ACS Chem Biol 2025; 20:332-339. [PMID: 39868662 PMCID: PMC11851429 DOI: 10.1021/acschembio.4c00592] [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: 09/06/2024] [Revised: 01/07/2025] [Accepted: 01/10/2025] [Indexed: 01/28/2025]
Abstract
Protein interactions play a crucial role in regulating cellular mechanisms, highlighting the need for effective methods to control these processes. In this regard, chemical inducers of proximity (CIPs) offer a promising approach to precisely manipulate protein-protein interactions in live cells and in vivo. In this study, we introduce pMandi, a photocaged version of the plant hormone-based CIP mandipropamid (Mandi), which allows the use of light as an external trigger to induce protein proximity in live mammalian cells. Furthermore, we present opabactin (OP) as a new plant hormone-based CIP that is effective in live mammalian cells at low nanomolar concentration and in live medaka embryos at submicromolar concentration. Its photocaged derivative, pOP, enables the induction of protein proximity upon light exposure in individual cells, enhancing spatiotemporal control to the level of single-cell resolution. Additionally, we explored the use of both photocaged CIPs to promote protein proximity in live medaka embryos.
Collapse
Affiliation(s)
- Philipp Pöschko
- Department
of Chemical Biology, Max Planck Institute
for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany
- Faculty
of Biosciences, Heidelberg University, Im Neuenheimer Feld 234, 69120 Heidelberg, Germany
| | - Caroline M. Berrou
- Department
of Chemical Biology, Max Planck Institute
for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany
| | - Kaisa Pakari
- Centre
for Organismal Studies Heidelberg (COS), Heidelberg University, Im Neuenheimer Feld 230, 69120 Heidelberg, Germany
- Heidelberg
Biosciences International Graduate School (HBIGS), Heidelberg University, Im Neuenheimer Feld 501, 69120 Heidelberg, Germany
| | - Michael J. Ziegler
- Department
of Chemical Biology, Max Planck Institute
for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany
| | - Christoph Kern
- Department
of Chemical Biology, Max Planck Institute
for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany
| | - Birgit Koch
- Department
of Chemical Biology, Max Planck Institute
for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany
| | - Joachim Wittbrodt
- Centre
for Organismal Studies Heidelberg (COS), Heidelberg University, Im Neuenheimer Feld 230, 69120 Heidelberg, Germany
| | - Richard Wombacher
- Department
of Chemical Biology, Max Planck Institute
for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany
| |
Collapse
|
5
|
Morabito RD, Tatarakis D, Swick R, Stettnisch S, Schilling TF, Horsfield JA, Martin BL. The ratio of Wnt signaling activity to Sox2 transcription factor levels predicts neuromesodermal fate potential. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.16.633481. [PMID: 39868081 PMCID: PMC11761523 DOI: 10.1101/2025.01.16.633481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Neuromesodermal progenitors (NMPs) are a vertebrate cell type that contribute descendants to both the spinal cord and the mesoderm. The undifferentiated bipotential NMP state is maintained when both Wnt signaling is active and Sox2 is present. We used transgenic reporter lines to live-image both Wnt activity and Sox2 levels in NMPs and observed a unique cellular ratio in NMPs compared to NMP-derived mesoderm or neural tissue. We used this unique signature to identify the previously unknown anatomical position of a progenitor population that gives rise to the midline tissues of the floor plate of the spinal cord and the mesodermal notochord. Thus, quantification of the active Wnt signaling to Sox2 ratio can be used to predict and identify cells with neuromesodermal potential. We also developed the auxin inducible degron 2 system for use in zebrafish to test the temporal role that Sox2 plays during midline formation. We found ectopic Sox2 in the presence of Wnt activity holds cells in the undifferentiated floor plate/notochord progenitor state, and that degradation of the ectopic Sox2 is required for cells to adopt a notochord fate.
Collapse
Affiliation(s)
- Robert D. Morabito
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11733, USA
| | - David Tatarakis
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
| | - Ryan Swick
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11733, USA
| | - Samantha Stettnisch
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11733, USA
| | - Thomas F. Schilling
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
| | - Julia A. Horsfield
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
- The Maurice Wilkins Centre for Biodiscovery, The University of Auckland, Auckland, New Zealand
- Genetics Otago Research Centre, University of Otago, Dunedin, New Zealand
| | - Benjamin L. Martin
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11733, USA
| |
Collapse
|
6
|
Heinzle C, Höfler A, Yu J, Heid P, Kremer N, Schunk R, Stengel F, Bange T, Boland A, Mayer TU. Positively charged specificity site in cyclin B1 is essential for mitotic fidelity. Nat Commun 2025; 16:853. [PMID: 39833154 PMCID: PMC11747444 DOI: 10.1038/s41467-024-55669-x] [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: 03/23/2024] [Accepted: 12/19/2024] [Indexed: 01/22/2025] Open
Abstract
Phosphorylation of substrates by cyclin-dependent kinases (CDKs) is the driving force of cell cycle progression. Several CDK-activating cyclins are involved, yet how they contribute to substrate specificity is still poorly understood. Here, we discover that a positively charged pocket in cyclin B1, which is exclusively conserved within B-type cyclins and binds phosphorylated serine- or threonine-residues, is essential for correct execution of mitosis. HeLa cells expressing pocket mutant cyclin B1 are strongly delayed in anaphase onset due to multiple defects in mitotic spindle function and timely activation of the E3 ligase APC/C. Pocket integrity is essential for APC/C phosphorylation particularly at non-consensus CDK1 sites and full in vitro ubiquitylation activity. Our results support a model in which cyclin B1's pocket facilitates sequential substrate phosphorylations involving initial priming events that assist subsequent pocket-dependent phosphorylations even at non-consensus CDK1 motifs.
Collapse
Affiliation(s)
- Christian Heinzle
- Department of Biology, University of Konstanz, Konstanz, Germany
- Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
| | - Anna Höfler
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Jun Yu
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Peter Heid
- Department of Biology, University of Konstanz, Konstanz, Germany
- Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
| | - Nora Kremer
- Institute of Medical Psychology and Biomedical Center (BMC), Faculty of Medicine, LMU, Munich, Germany
| | - Rebecca Schunk
- Department of Biology, University of Konstanz, Konstanz, Germany
- Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
| | - Florian Stengel
- Department of Biology, University of Konstanz, Konstanz, Germany
- Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
| | - Tanja Bange
- Institute of Medical Psychology and Biomedical Center (BMC), Faculty of Medicine, LMU, Munich, Germany
| | - Andreas Boland
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland.
| | - Thomas U Mayer
- Department of Biology, University of Konstanz, Konstanz, Germany.
- Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany.
| |
Collapse
|
7
|
Ko S, Mizumoto K. Comparison among bright green fluorescent proteins in C. elegans. MICROPUBLICATION BIOLOGY 2025; 2025:10.17912/micropub.biology.001447. [PMID: 39897165 PMCID: PMC11787626 DOI: 10.17912/micropub.biology.001447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 12/14/2024] [Accepted: 01/17/2025] [Indexed: 02/04/2025]
Abstract
Green fluorescent proteins (GFPs) are invaluable tools for visualizing cells and proteins across model systems. Efforts have been made to generate brighter fluorescent proteins such as eGFP, GFPnovo2, mNeonGreen, and mStayGold. Here, we generated single-copy knock-in C. elegans strains for these GFP variants and directly compared their brightness and photostability. We confirmed that mStayGold is brighter and more photostable than eGFP, GFPnovo2, and mNeonGreen, suggesting that it may hold advantages over other GFP variants in experiments where brightness and photostability are important factors.
Collapse
Affiliation(s)
- Sydney Ko
- Graduate Program in Cell and Developmental Biology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kota Mizumoto
- Department of Zoology, Life Sciences Institute, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
8
|
Rentsch D, Bergs A, Shao J, Elvers N, Ruse C, Seidenthal M, Aoki I, Gottschalk A. Tools and methods for cell ablation and cell inhibition in Caenorhabditis elegans. Genetics 2025; 229:1-48. [PMID: 39110015 PMCID: PMC11708922 DOI: 10.1093/genetics/iyae119] [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: 05/12/2024] [Accepted: 07/16/2024] [Indexed: 01/11/2025] Open
Abstract
To understand the function of cells such as neurons within an organism, it can be instrumental to inhibit cellular function, or to remove the cell (type) from the organism, and thus to observe the consequences on organismic and/or circuit function and animal behavior. A range of approaches and tools were developed and used over the past few decades that act either constitutively or acutely and reversibly, in systemic or local fashion. These approaches make use of either drugs or genetically encoded tools. Also, there are acutely acting inhibitory tools that require an exogenous trigger like light. Here, we give an overview of such methods developed and used in the nematode Caenorhabditis elegans.
Collapse
Affiliation(s)
- Dennis Rentsch
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue Strasse 15, D-60438 Frankfurt, Germany
- Institute for Biophysical Chemistry, Goethe University, Max-von-Laue Strasse 9, D-60438 Frankfurt, Germany
| | - Amelie Bergs
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue Strasse 15, D-60438 Frankfurt, Germany
- Institute for Biophysical Chemistry, Goethe University, Max-von-Laue Strasse 9, D-60438 Frankfurt, Germany
| | - Jiajie Shao
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue Strasse 15, D-60438 Frankfurt, Germany
- Institute for Biophysical Chemistry, Goethe University, Max-von-Laue Strasse 9, D-60438 Frankfurt, Germany
| | - Nora Elvers
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue Strasse 15, D-60438 Frankfurt, Germany
- Institute for Biophysical Chemistry, Goethe University, Max-von-Laue Strasse 9, D-60438 Frankfurt, Germany
| | - Christiane Ruse
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue Strasse 15, D-60438 Frankfurt, Germany
- Institute for Biophysical Chemistry, Goethe University, Max-von-Laue Strasse 9, D-60438 Frankfurt, Germany
| | - Marius Seidenthal
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue Strasse 15, D-60438 Frankfurt, Germany
- Institute for Biophysical Chemistry, Goethe University, Max-von-Laue Strasse 9, D-60438 Frankfurt, Germany
| | - Ichiro Aoki
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue Strasse 15, D-60438 Frankfurt, Germany
- Institute for Biophysical Chemistry, Goethe University, Max-von-Laue Strasse 9, D-60438 Frankfurt, Germany
| | - Alexander Gottschalk
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue Strasse 15, D-60438 Frankfurt, Germany
- Institute for Biophysical Chemistry, Goethe University, Max-von-Laue Strasse 9, D-60438 Frankfurt, Germany
| |
Collapse
|
9
|
Silvester E, Baker LA. Molecular tags for electron cryo-tomography. Emerg Top Life Sci 2024:ETLS20240006. [PMID: 39636021 DOI: 10.1042/etls20240006] [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: 10/09/2024] [Revised: 10/09/2024] [Accepted: 11/05/2024] [Indexed: 12/07/2024]
Abstract
Electron cryotomography enables the direct visualisation of biological specimens without stains or fixation, revealing complex molecular landscapes at high resolution. However, identifying specific proteins within these crowded environments is challenging. Molecular tagging offers a promising solution by attaching visually distinctive markers to proteins of interest, differentiating them from the background. This review explores available tagging strategies, including gold nanoparticles, metal-binding proteins, nucleic acid nanostructures and protein-based tags. The identification and targeting strategies for each approach are discussed, highlighting their respective advantages and limitations. Future directions for advancing these tagging techniques to expand their applicability to broader research questions are also considered.
Collapse
Affiliation(s)
- Emma Silvester
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, U.K
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, U.K
| | - Lindsay A Baker
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, U.K
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, U.K
| |
Collapse
|
10
|
Maia-Gil M, Gorjão M, Belousov R, Espina JA, Coelho J, Gouhier J, Ramos AP, Barriga EH, Erzberger A, Norden C. Nuclear deformability facilitates apical nuclear migration in the developing zebrafish retina. Curr Biol 2024; 34:5429-5443.e8. [PMID: 39481375 DOI: 10.1016/j.cub.2024.10.015] [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: 03/27/2024] [Revised: 08/23/2024] [Accepted: 10/03/2024] [Indexed: 11/02/2024]
Abstract
Nuclear positioning is a crucial aspect of cell and developmental biology. One example is the apical movement of nuclei in neuroepithelia before mitosis, which is essential for proper tissue formation. While the cytoskeletal mechanisms that drive nuclei to the apical side have been explored, the influence of nuclear properties on apical nuclear migration is less understood. Nuclear properties, such as deformability, can be linked to lamin A/C expression levels, as shown in various in vitro studies. Interestingly, many nuclei in early development, including neuroepithelial nuclei, express only low levels of lamin A/C. Therefore, we investigated whether increased lamin A expression in the densely packed zebrafish retinal neuroepithelium affects nuclear deformability and, consequently, migration phenomena. We found that overexpressing lamin A in retinal nuclei increases nuclear stiffness, which in turn indeed impairs apical nuclear migration. Interestingly, nuclei that do not overexpress lamin A but are embedded in a stiffer lamin A-overexpressing environment also exhibit impaired apical nuclear migration, indicating that these effects can be cell non-autonomous. Additionally, in the less crowded hindbrain neuroepithelium, only minor effects on apical nuclear migration are observed. Together, this suggests that the material properties of the nucleus influence nuclear movements in a tissue-dependent manner.
Collapse
Affiliation(s)
- Mariana Maia-Gil
- Gulbenkian Institute for Molecular Medicine (GIMM) (previously Instituto Gulbenkian de Ciência), Rua da Quinta Grande 6, 2780-156 Oeiras, Portugal
| | - Maria Gorjão
- Gulbenkian Institute for Molecular Medicine (GIMM) (previously Instituto Gulbenkian de Ciência), Rua da Quinta Grande 6, 2780-156 Oeiras, Portugal
| | - Roman Belousov
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Jaime A Espina
- Gulbenkian Institute for Molecular Medicine (GIMM) (previously Instituto Gulbenkian de Ciência), Rua da Quinta Grande 6, 2780-156 Oeiras, Portugal; Cluster of Excellence Physics of Life, Arnoldstrasse 18, 01307 Dresden, Germany
| | - João Coelho
- Gulbenkian Institute for Molecular Medicine (GIMM) (previously Instituto Gulbenkian de Ciência), Rua da Quinta Grande 6, 2780-156 Oeiras, Portugal
| | - Juliette Gouhier
- Gulbenkian Institute for Molecular Medicine (GIMM) (previously Instituto Gulbenkian de Ciência), Rua da Quinta Grande 6, 2780-156 Oeiras, Portugal
| | - Ana P Ramos
- Gulbenkian Institute for Molecular Medicine (GIMM) (previously Instituto Gulbenkian de Ciência), Rua da Quinta Grande 6, 2780-156 Oeiras, Portugal
| | - Elias H Barriga
- Gulbenkian Institute for Molecular Medicine (GIMM) (previously Instituto Gulbenkian de Ciência), Rua da Quinta Grande 6, 2780-156 Oeiras, Portugal; Cluster of Excellence Physics of Life, Arnoldstrasse 18, 01307 Dresden, Germany
| | - Anna Erzberger
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Caren Norden
- Gulbenkian Institute for Molecular Medicine (GIMM) (previously Instituto Gulbenkian de Ciência), Rua da Quinta Grande 6, 2780-156 Oeiras, Portugal.
| |
Collapse
|
11
|
Zhou Z, Wang YQ, Zheng XN, Zhang XH, Ji LY, Han JY, Zuo ZC, Mo WL, Zhang L. Optimizing ABA-based chemically induced proximity for enhanced intracellular transcriptional activation and modification response to ABA. SCIENCE CHINA. LIFE SCIENCES 2024; 67:2650-2663. [PMID: 39172347 DOI: 10.1007/s11427-024-2707-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024]
Abstract
Abscisic acid (ABA)-based chemically induced proximity (CIP) is primarily mediated by the interaction of the ABA receptor pyrabactin resistance 1-like 1 (PYL1) and the 2C-type protein phosphatase ABI1, which confers ABA-induced proximity to their fusion proteins, and offers precise temporal control of a wide array of biological processes. However, broad application of ABA-based CIP has been limited by ABA response intensity. In this study, we demonstrated that ABA-induced interaction between another ABA receptor pyrabactin resistance 1 (PYR1) and ABI1 exhibited higher ABA response intensity than that between PYL1 and ABI1 in HEK293T cells. We engineered PYR1-ABI1 and PYL1-ABI1 into ABA-induced transcriptional activation tools in mammalian cells by integration with CRISPR/dCas9 and found that the tool based on PYR1-ABI1 demonstrated better ABA response intensity than that based on PYL1-ABI1 for both exogenous and endogenous genes in mammalian cells. We further achieved ABA-induced RNA m6A modification installation and erasure by combining ABA-induced PYR1-ABI1 interaction with CRISPR/dCas13, successfully inhibiting tumor cell proliferation. We subsequently improved the interaction of PYR1-ABI1 through phage-assisted continuous evolution (PACE), successfully generating a PYR1 mutant (PYR1m) whose interaction with ABI1 exhibited a higher ABA response intensity than that of the wild-type. In addition, we tested the transcriptional activation tool based on PYRm-ABI1 and found that it also showed a higher ABA response intensity than that of the wild type. These results demonstrate that we have developed a novel ABA-based CIP and further improved upon it using PACE, providing a new approach for the modification of other CIP systems.
Collapse
Affiliation(s)
- Zeng Zhou
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
- Basic Forestry and Proteomics Research Center, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Yue-Qi Wang
- Jilin Province Engineering Laboratory of Plant Genetic Improvement, College of Plant Science, Jilin University, Changchun, 130062, China
| | - Xu-Nan Zheng
- Jilin Province Engineering Laboratory of Plant Genetic Improvement, College of Plant Science, Jilin University, Changchun, 130062, China
| | - Xiao-Hong Zhang
- Basic Forestry and Proteomics Research Center, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Lu-Yao Ji
- Basic Forestry and Proteomics Research Center, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Jun-You Han
- Jilin Province Engineering Laboratory of Plant Genetic Improvement, College of Plant Science, Jilin University, Changchun, 130062, China
| | - Ze-Cheng Zuo
- Jilin Province Engineering Laboratory of Plant Genetic Improvement, College of Plant Science, Jilin University, Changchun, 130062, China.
| | - Wei-Liang Mo
- Jilin Province Engineering Laboratory of Plant Genetic Improvement, College of Plant Science, Jilin University, Changchun, 130062, China.
| | - Li Zhang
- Jilin Province Engineering Laboratory of Plant Genetic Improvement, College of Plant Science, Jilin University, Changchun, 130062, China.
| |
Collapse
|
12
|
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
|
13
|
Huang L, Rojas-Pierce M. Rapid depletion of target proteins in plants by an inducible protein degradation system. THE PLANT CELL 2024; 36:3145-3161. [PMID: 38446628 PMCID: PMC11371150 DOI: 10.1093/plcell/koae072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 02/14/2024] [Accepted: 02/26/2024] [Indexed: 03/08/2024]
Abstract
Inducible protein knockdowns are excellent tools to test the function of essential proteins in short time scales and to capture the role of proteins in dynamic events. Current approaches destroy or sequester proteins by exploiting plant biological mechanisms such as the activity of photoreceptors for optogenetics or auxin-mediated ubiquitination in auxin degrons. It follows that these are not applicable for plants as light and auxin are strong signals for plant cells. We describe here an inducible protein degradation system in plants named E3-DART for E3-targeted Degradation of Plant Proteins. The E3-DART system is based on the specific and well-characterized interaction between the Salmonella-secreted protein H1 (SspH1) and its human target protein kinase N1 (PKN1). This system harnesses the E3 catalytic activity of SspH1 and the SspH1-binding activity of the homology region 1b (HR1b) domain from PKN1. Using Nicotiana benthamiana and Arabidopsis (Arabidopsis thaliana), we show that a chimeric protein containing the leucine-rich repeat and novel E3 ligase domains of SspH1 efficiently targets protein fusions of varying sizes containing HR1b for degradation. Target protein degradation was induced by transcriptional control of the chimeric E3 ligase using a glucocorticoid transactivation system, and target protein depletion was detected as early as 3 h after induction. This system could be used to study the loss of any plant protein with high-temporal resolution and may become an important tool in plant cell biology.
Collapse
Affiliation(s)
- Linzhou Huang
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC 27695, USA
| | - Marcela Rojas-Pierce
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC 27695, USA
| |
Collapse
|
14
|
Kim M, Bhargava HK, Shavey GE, Lim WA, El-Samad H, Ng AH. Degron-Based bioPROTACs for Controlling Signaling in CAR T Cells. ACS Synth Biol 2024; 13:2313-2327. [PMID: 38991546 PMCID: PMC11334183 DOI: 10.1021/acssynbio.4c00109] [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: 02/16/2024] [Revised: 06/02/2024] [Accepted: 06/03/2024] [Indexed: 07/13/2024]
Abstract
Chimeric antigen receptor (CAR) T cells have made a tremendous impact in the clinic, but potent signaling through the CAR can be detrimental to treatment safety and efficacy. The use of protein degradation to control CAR signaling can address these issues in preclinical models. Existing strategies for regulating CAR stability rely on small molecules to induce systemic degradation. In contrast to small molecule regulation, genetic circuits offer a more precise method to control CAR signaling in an autonomous cell-by-cell fashion. Here, we describe a programmable protein degradation tool that adopts the framework of bioPROTACs, heterobifunctional proteins that are composed of a target recognition domain fused to a domain that recruits the endogenous ubiquitin proteasome system. We develop novel bioPROTACs that utilize a compact four-residue degron and demonstrate degradation of cytosolic and membrane protein targets using either a nanobody or synthetic leucine zipper as a protein binder. Our bioPROTACs exhibit potent degradation of CARs and can inhibit CAR signaling in primary human T cells. We demonstrate the utility of our bioPROTACs by constructing a genetic circuit to degrade the tyrosine kinase ZAP70 in response to recognition of a specific membrane-bound antigen. This circuit can disrupt CAR T cell signaling only in the presence of a specific cell population. These results suggest that bioPROTACs are powerful tools for expanding the CAR T cell engineering toolbox.
Collapse
Affiliation(s)
- Matthew
S. Kim
- Tetrad
Graduate Program, University of California
San Francisco, San Francisco, California 94158, United States
- Cell
Design Institute, University of California
San Francisco, San Francisco, California 94158, United States
- Department
of Biochemistry and Biophysics, University
of California San Francisco, San
Francisco, California 94158, United States
| | - Hersh K. Bhargava
- Cell
Design Institute, University of California
San Francisco, San Francisco, California 94158, United States
- Department
of Biochemistry and Biophysics, University
of California San Francisco, San
Francisco, California 94158, United States
- Biophysics
Graduate Program, University of California
San Francisco, San Francisco, California 94158, United States
| | - Gavin E. Shavey
- Cell
Design Institute, University of California
San Francisco, San Francisco, California 94158, United States
| | - Wendell A. Lim
- Cell
Design Institute, University of California
San Francisco, San Francisco, California 94158, United States
- Department
of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California 94158, United States
| | - Hana El-Samad
- Cell
Design Institute, University of California
San Francisco, San Francisco, California 94158, United States
- Department
of Biochemistry and Biophysics, University
of California San Francisco, San
Francisco, California 94158, United States
- Chan-Zuckerberg
Biohub, San Francisco, California 94158, United States
- Altos
Labs Inc., Redwood City, California, 94065, United States
| | - Andrew H. Ng
- Cell
Design Institute, University of California
San Francisco, San Francisco, California 94158, United States
- Department
of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California 94158, United States
- Department
of Molecular Biology, Genentech Inc., South San Francisco, California 94080, United States
| |
Collapse
|
15
|
Chan A, Haley RM, Najar MA, Gonzalez-Martinez D, Bugaj LJ, Burslem GM, Mitchell MJ, Tsourkas A. Lipid-mediated intracellular delivery of recombinant bioPROTACs for the rapid degradation of undruggable proteins. Nat Commun 2024; 15:5808. [PMID: 38987546 PMCID: PMC11237011 DOI: 10.1038/s41467-024-50235-x] [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: 11/02/2023] [Accepted: 07/04/2024] [Indexed: 07/12/2024] Open
Abstract
Recently, targeted degradation has emerged as a powerful therapeutic modality. Relying on "event-driven" pharmacology, proteolysis targeting chimeras (PROTACs) can degrade targets and are superior to conventional inhibitors against undruggable proteins. Unfortunately, PROTAC discovery is limited by warhead scarcity and laborious optimization campaigns. To address these shortcomings, analogous protein-based heterobifunctional degraders, known as bioPROTACs, have been developed. Compared to small-molecule PROTACs, bioPROTACs have higher success rates and are subject to fewer design constraints. However, the membrane impermeability of proteins severely restricts bioPROTAC deployment as a generalized therapeutic modality. Here, we present an engineered bioPROTAC template able to complex with cationic and ionizable lipids via electrostatic interactions for cytosolic delivery. When delivered by biocompatible lipid nanoparticles, these modified bioPROTACs can rapidly degrade intracellular proteins, exhibiting near-complete elimination (up to 95% clearance) of targets within hours of treatment. Our bioPROTAC format can degrade proteins localized to various subcellular compartments including the mitochondria, nucleus, cytosol, and membrane. Moreover, substrate specificity can be easily reprogrammed, allowing modular design and targeting of clinically-relevant proteins such as Ras, Jnk, and Erk. In summary, this work introduces an inexpensive, flexible, and scalable platform for efficient intracellular degradation of proteins that may elude chemical inhibition.
Collapse
Affiliation(s)
- Alexander Chan
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Rebecca M Haley
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohd Altaf Najar
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cancer Biology and Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David Gonzalez-Martinez
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Lukasz J Bugaj
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - George M Burslem
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cancer Biology and Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael J Mitchell
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew Tsourkas
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
16
|
Sural S, Botero JQ, Hobert O, Tekle-Smith M. Protocol to synthesize the auxin analog 5-Ph-IAA for conditional protein depletion in C. elegans using the AID2 system. STAR Protoc 2024; 5:102901. [PMID: 38377002 PMCID: PMC10884774 DOI: 10.1016/j.xpro.2024.102901] [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: 12/18/2023] [Revised: 01/17/2024] [Accepted: 02/05/2024] [Indexed: 02/22/2024] Open
Abstract
The auxin-inducible degron (AID) system is a broadly used tool for spatiotemporal and reversible control of protein depletion in multiple experimental model systems. AID2 technology relies on a synthetic ligand, 5-phenyl-indole-3-acetic acid (5-Ph-IAA), for improved specificity and efficiency of protein degradation. Here, we provide a protocol for cost-effective 5-Ph-IAA synthesis utilizing the Suzuki coupling of 5-chloroindole and phenylboronic acid. We describe steps for evaluating the quality of lab-synthesized 5-Ph-IAA using a C. elegans AID2 tester strain.
Collapse
Affiliation(s)
- Surojit Sural
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, NY 10027, USA
| | | | - Oliver Hobert
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, NY 10027, USA.
| | | |
Collapse
|
17
|
Kim MS, Bhargava HK, Shavey GE, Lim WA, El-Samad H, Ng AH. Degron-based bioPROTACs for controlling signaling in CAR T cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.16.580396. [PMID: 38405763 PMCID: PMC10888892 DOI: 10.1101/2024.02.16.580396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Chimeric antigen receptor (CAR) T cells have made a tremendous impact in the clinic, but potent signaling through the CAR can be detrimental to treatment safety and efficacy. The use of protein degradation to control CAR signaling can address these issues in pre-clinical models. Existing strategies for regulating CAR stability rely on small molecules to induce systemic degradation. In contrast to small molecule regulation, genetic circuits offer a more precise method to control CAR signaling in an autonomous, cell-by-cell fashion. Here, we describe a programmable protein degradation tool that adopts the framework of bioPROTACs, heterobifunctional proteins that are composed of a target recognition domain fused to a domain that recruits the endogenous ubiquitin proteasome system. We develop novel bioPROTACs that utilize a compact four residue degron and demonstrate degradation of cytosolic and membrane protein targets using either a nanobody or synthetic leucine zipper as a protein binder. Our bioPROTACs exhibit potent degradation of CARs and can inhibit CAR signaling in primary human T cells. We demonstrate the utility of our bioPROTACs by constructing a genetic circuit to degrade the tyrosine kinase ZAP70 in response to recognition of a specific membrane-bound antigen. This circuit is able to disrupt CAR T cell signaling only in the presence of a specific cell population. These results suggest that bioPROTACs are a powerful tool for expanding the cell engineering toolbox for CAR T cells.
Collapse
Affiliation(s)
- Matthew S Kim
- Tetrad Graduate Program, University of California, San Francisco, San Francisco, CA; Cell Design Institute, University of California, San Francisco, San Francisco, CA
| | - Hersh K Bhargava
- Biophysics Graduate Program, University of California, San Francisco, San Francisco, CA; Cell Design Institute, University of California, San Francisco, San Francisco, CA; Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA
| | - Gavin E Shavey
- Current: Arsenal Biociences, Inc., South San Francisco, CA; Cell Design Institute, University of California, San Francisco, San Francisco, CA
| | - Wendell A Lim
- Cell Design Institute, University of California, San Francisco, San Francisco, CA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA
| | - Hana El-Samad
- Current: Altos Labs, Redwood City, CA; Cell Design Institute, University of California, San Francisco, San Francisco, CA; Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA; Chan-Zuckerberg Biohub, San Francisco, CA; Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA
| | - Andrew H Ng
- Current: Department of Molecular Biology, Genentech Inc., South San Francisco, CA, USA; Cell Design Institute, University of California, San Francisco, San Francisco, CA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
18
|
Kenny-Ganzert IW, Sherwood DR. The C. elegans anchor cell: A model to elucidate mechanisms underlying invasion through basement membrane. Semin Cell Dev Biol 2024; 154:23-34. [PMID: 37422376 PMCID: PMC10592375 DOI: 10.1016/j.semcdb.2023.07.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 06/30/2023] [Accepted: 07/01/2023] [Indexed: 07/10/2023]
Abstract
Cell invasion through basement membrane barriers is crucial during many developmental processes and in immune surveillance. Dysregulation of invasion also drives the pathology of numerous human diseases, such as metastasis and inflammatory disorders. Cell invasion involves dynamic interactions between the invading cell, basement membrane, and neighboring tissues. Owing to this complexity, cell invasion is challenging to study in vivo, which has hampered the understanding of mechanisms controlling invasion. Caenorhabditis elegans anchor cell invasion is a powerful in vivo model where subcellular imaging of cell-basement membrane interactions can be combined with genetic, genomic, and single-cell molecular perturbation studies. In this review, we outline insights gained by studying anchor cell invasion, which span transcriptional networks, translational regulation, secretory apparatus expansion, dynamic and adaptable protrusions that breach and clear basement membrane, and a complex, localized metabolic network that fuels invasion. Together, investigation of anchor cell invasion is building a comprehensive understanding of the mechanisms that underlie invasion, which we expect will ultimately facilitate better therapeutic strategies to control cell invasive activity in human disease.
Collapse
Affiliation(s)
| | - David R Sherwood
- Department of Biology, Duke University, Box 90338, Durham, NC 27708, USA.
| |
Collapse
|
19
|
Asana Marican HT, Shen H. Dynamics of Chromosome Aberrations and Cell Death in Zebrafish Embryos Exposed to 137Cs Total-Body Irradiation. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:2204-2213. [PMID: 38269402 DOI: 10.1021/acs.est.3c05389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
Abstract
Ionizing radiation exposure induces significant DNA damage and cell death in aquatic species. Accurate sensing and quantification play pivotal roles in environmental monitoring and surveillance. Zebrafish (Danio rerio) is a well-suited animal model for research into this aspect, especially with recent development of cytogenetic and transgenic tools. In this study, we present time-course studies of chromosome aberrations and cell death in zebrafish embryos exposed to 2 Gy 137Cs total-body irradiation. Using a cytogenetic approach, we quantified chromosome and chromatid aberrations in irradiated embryos at 6, 14, 20, and 24 h postirradiation. Metaphases with aberrations showed rapid declining kinetics, accompanied by incomplete karyotypes and irregular chromatin contents. Using an apoptosis-reporting transgenic zebrafish, we found increasing cell death along these time points, with the embryonic eyes and brain contributing the majority of the cell death volumes. We provide evidence that self-proliferating progenitor cells form the underlying linkage between the two kinetics and their positions define radiosensitive niches in zebrafish embryos. Our results provide detailed chromosome aberration and cell death dynamics in 137Cs-irradiated zebrafish embryos and unveil the appropriate timeline and tissue positions for accurate sensing and quantification of radiation-induced damages in zebrafish embryos.
Collapse
Affiliation(s)
- Halida Thanveer Asana Marican
- Singapore Nuclear Research and Safety Initiative, National University of Singapore, CREATE Tower, 1 Create Way, Singapore 138602, Singapore
| | - Hongyuan Shen
- Singapore Nuclear Research and Safety Initiative, National University of Singapore, CREATE Tower, 1 Create Way, Singapore 138602, Singapore
| |
Collapse
|
20
|
Arnold B, Riegger RJ, Okuda EK, Slišković I, Keller M, Bakisoglu C, McNicoll F, Zarnack K, Müller-McNicoll M. hGRAD: A versatile "one-fits-all" system to acutely deplete RNA binding proteins from condensates. J Cell Biol 2024; 223:e202304030. [PMID: 38108808 PMCID: PMC10726014 DOI: 10.1083/jcb.202304030] [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: 04/12/2023] [Revised: 09/18/2023] [Accepted: 11/21/2023] [Indexed: 12/19/2023] Open
Abstract
Nuclear RNA binding proteins (RBPs) are difficult to study because they often belong to large protein families and form extensive networks of auto- and crossregulation. They are highly abundant and many localize to condensates with a slow turnover, requiring long depletion times or knockouts that cannot distinguish between direct and indirect or compensatory effects. Here, we developed a system that is optimized for the rapid degradation of nuclear RBPs, called hGRAD. It comes as a "one-fits-all" plasmid, and integration into any cell line with endogenously GFP-tagged proteins allows for an inducible, rapid, and complete knockdown. We show that the nuclear RBPs SRSF3, SRSF5, SRRM2, and NONO are completely cleared from nuclear speckles and paraspeckles within 2 h. hGRAD works in various cell types, is more efficient than previous methods, and does not require the expression of exogenous ubiquitin ligases. Combining SRSF5 hGRAD degradation with Nascent-seq uncovered transient transcript changes, compensatory mechanisms, and an effect of SRSF5 on transcript stability.
Collapse
Affiliation(s)
- Benjamin Arnold
- Institute of Molecular Biosciences, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Ricarda J. Riegger
- Institute of Molecular Biosciences, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Ellen Kazumi Okuda
- Institute of Molecular Biosciences, Goethe University Frankfurt, Frankfurt am Main, Germany
- International Max Planck Research School for Cellular Biophysics, Frankfurt am Main, Germany
| | - Irena Slišković
- Institute of Molecular Biosciences, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Mario Keller
- Institute of Molecular Biosciences, Goethe University Frankfurt, Frankfurt am Main, Germany
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Cem Bakisoglu
- Institute of Molecular Biosciences, Goethe University Frankfurt, Frankfurt am Main, Germany
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - François McNicoll
- Institute of Molecular Biosciences, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Kathi Zarnack
- Institute of Molecular Biosciences, Goethe University Frankfurt, Frankfurt am Main, Germany
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt am Main, Germany
| | | |
Collapse
|
21
|
Ogawa Y, Ueda TP, Obara K, Nishimura K, Kamura T. Targeted Protein Degradation Systems: Controlling Protein Stability Using E3 Ubiquitin Ligases in Eukaryotic Species. Cells 2024; 13:175. [PMID: 38247866 PMCID: PMC10814424 DOI: 10.3390/cells13020175] [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: 12/12/2023] [Revised: 01/12/2024] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
This review explores various methods for modulating protein stability to achieve target protein degradation, which is a crucial aspect in the study of biological processes and drug design. Thirty years have passed since the introduction of heat-inducible degron cells utilizing the N-end rule, and methods for controlling protein stability using the ubiquitin-proteasome system have moved from academia to industry. This review covers protein stability control methods, from the early days to recent advancements, and discusses the evolution of techniques in this field. This review also addresses the challenges and future directions of protein stability control techniques by tracing their development from the inception of protein stability control methods to the present day.
Collapse
Affiliation(s)
| | | | | | - Kohei Nishimura
- Department of Biological Science, Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya 464-8601, Japan; (Y.O.); (T.P.U.); (K.O.)
| | - Takumi Kamura
- Department of Biological Science, Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya 464-8601, Japan; (Y.O.); (T.P.U.); (K.O.)
| |
Collapse
|
22
|
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
|
23
|
Fung HKH, Hayashi Y, Salo VT, Babenko A, Zagoriy I, Brunner A, Ellenberg J, Müller CW, Cuylen-Haering S, Mahamid J. Genetically encoded multimeric tags for subcellular protein localization in cryo-EM. Nat Methods 2023; 20:1900-1908. [PMID: 37932397 PMCID: PMC10703698 DOI: 10.1038/s41592-023-02053-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 09/19/2023] [Indexed: 11/08/2023]
Abstract
Cryo-electron tomography (cryo-ET) allows for label-free high-resolution imaging of macromolecular assemblies in their native cellular context. However, the localization of macromolecules of interest in tomographic volumes can be challenging. Here we present a ligand-inducible labeling strategy for intracellular proteins based on fluorescent, 25-nm-sized, genetically encoded multimeric particles (GEMs). The particles exhibit recognizable structural signatures, enabling their automated detection in cryo-ET data by convolutional neural networks. The coupling of GEMs to green fluorescent protein-tagged macromolecules of interest is triggered by addition of a small-molecule ligand, allowing for time-controlled labeling to minimize disturbance to native protein function. We demonstrate the applicability of GEMs for subcellular-level localization of endogenous and overexpressed proteins across different organelles in human cells using cryo-correlative fluorescence and cryo-ET imaging. We describe means for quantifying labeling specificity and efficiency, and for systematic optimization for rare and abundant protein targets, with emphasis on assessing the potential effects of labeling on protein function.
Collapse
Affiliation(s)
- Herman K H Fung
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Yuki Hayashi
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Veijo T Salo
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Anastasiia Babenko
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- University of Heidelberg, Heidelberg, Germany
| | - Ievgeniia Zagoriy
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Andreas Brunner
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Faculty of Biosciences, Collaboration for Joint PhD Degree between EMBL and Heidelberg University, Heidelberg, Germany
| | - Jan Ellenberg
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Christoph W Müller
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Sara Cuylen-Haering
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| | - Julia Mahamid
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany.
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| |
Collapse
|
24
|
Frecot DI, Froehlich T, Rothbauer U. 30 years of nanobodies - an ongoing success story of small binders in biological research. J Cell Sci 2023; 136:jcs261395. [PMID: 37937477 DOI: 10.1242/jcs.261395] [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] [Indexed: 11/09/2023] Open
Abstract
A milestone in the field of recombinant binding molecules was achieved 30 years ago with the discovery of single-domain antibodies from which antigen-binding variable domains, better known as nanobodies (Nbs), can be derived. Being only one tenth the size of conventional antibodies, Nbs feature high affinity and specificity, while being highly stable and soluble. In addition, they display accessibility to cryptic sites, low off-target accumulation and deep tissue penetration. Efficient selection methods, such as (semi-)synthetic/naïve or immunized cDNA libraries and display technologies, have facilitated the isolation of Nbs against diverse targets, and their single-gene format enables easy functionalization and high-yield production. This Review highlights recent advances in Nb applications in various areas of biological research, including structural biology, proteomics and high-resolution and in vivo imaging. In addition, we provide insights into intracellular applications of Nbs, such as live-cell imaging, biosensors and targeted protein degradation.
Collapse
Affiliation(s)
- Desiree I Frecot
- Pharmaceutical Biotechnology, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Markwiesenstrasse 55, 72770 Reutlingen, Reutlingen, Germany
| | - Theresa Froehlich
- Pharmaceutical Biotechnology, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany
| | - Ulrich Rothbauer
- Pharmaceutical Biotechnology, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany
| |
Collapse
|
25
|
Myles KM, Vo AA, Ragle JM, Ward JD. A spontaneous TIR1 loss-of-function allele in C. elegans. MICROPUBLICATION BIOLOGY 2023; 2023:10.17912/micropub.biology.000994. [PMID: 37908494 PMCID: PMC10613879 DOI: 10.17912/micropub.biology.000994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/12/2023] [Accepted: 10/11/2023] [Indexed: 11/02/2023]
Abstract
The auxin-inducible degron (AID) system is a widely-used system for conditional protein depletion. During the course of an experiment, we depleted the nuclear hormone receptor transcription factor NHR-23 to study molting, and we recovered a spontaneous suppressor allele that bypassed the L1 larval arrest caused by NHR-23 depletion. These mutants also failed to deplete a BFP::AID reporter in the strain background, suggesting a broader defect in the AID system. These animals carried an in-frame 18 base pair insertion that produced a 6 amino acid repeat in TIR1. The larval arrest in these animals could be restored by expressing a wild-type TIR1 transgene from an extrachromosomal array. Sister siblings that lost this array developed normally on auxin. Together, these experiments indicate that the TIR1 mutation was causing the loss of developmental arrest in the nhr-23::AID strain. This result highlights the importance of setting up a robust secondary screen to detect such mutants if performing forward genetic screens in conjunction with the AID system.
Collapse
Affiliation(s)
- Krista M. Myles
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, California, United States
| | - An A. Vo
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, California, United States
| | - James Matthew Ragle
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, California, United States
| | - Jordan D. Ward
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, California, United States
| |
Collapse
|
26
|
Kaji H, Mori F, Ito H. Enhanced precision of circadian rhythm by output system. J Theor Biol 2023; 574:111621. [PMID: 37717817 DOI: 10.1016/j.jtbi.2023.111621] [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: 01/11/2023] [Revised: 08/31/2023] [Accepted: 09/06/2023] [Indexed: 09/19/2023]
Abstract
Circadian rhythms are biological rhythms with a period of approximately 24 h that persist even under constant conditions without daily environmental cues. The molecular circadian clock machinery generates physiological rhythms, which can be transmitted into the downstream output system. Owing to the stochastic nature of the biochemical reactions and the extracellular environment, the oscillation period of circadian rhythms exhibited by individual organisms or cells is not constant on a daily basis with variations as high as 10%, as reflected by the coefficient of variation. Although the fluctuations in the circadian rhythm are measured through a reporter system such as bioluminescence or fluorescence, which is an example of output systems, experimentally confirming whether the fluctuations found in the reporter system are the same as those in the circadian clock is challenging. This study investigated a coupled system of a circadian clock and its output system numerically and analytically, and then compared the fluctuations in the oscillation period of the two systems. We found that the amount of fluctuations in the output system is smaller than that in the circadian clock, assuming the degradation rate of the molecules responsible for the output system is a typical value for protein degradation. The results indicate that the output system can improve the accuracy of the circadian rhythm without the need for any denoising processes.
Collapse
Affiliation(s)
- Hotaka Kaji
- Faculty of Design, Kyushu University, 4-9-1, Shiobaru, Fukuoka, 815-8540, Japan
| | - Fumito Mori
- Faculty of Design, Kyushu University, 4-9-1, Shiobaru, Fukuoka, 815-8540, Japan; Education and Research Center for Mathematical and Data Science, Kyushu University, 744, Motoka, Fukuoka, 819-0395, Japan
| | - Hiroshi Ito
- Faculty of Design, Kyushu University, 4-9-1, Shiobaru, Fukuoka, 815-8540, Japan.
| |
Collapse
|
27
|
Ogawa Y, Nishimura K, Obara K, Kamura T. Development of AlissAID system targeting GFP or mCherry fusion protein. PLoS Genet 2023; 19:e1010731. [PMID: 37315088 DOI: 10.1371/journal.pgen.1010731] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 04/04/2023] [Indexed: 06/16/2023] Open
Abstract
Conditional control of target proteins using the auxin-inducible degron (AID) system provides a powerful tool for investigating protein function in eukaryotes. Here, we established an Affinity-linker based super-sensitive auxin-inducible degron (AlissAID) system in budding yeast by using a single domain antibody (a nanobody). In this system, target proteins fused with GFP or mCherry were degraded depending on a synthetic auxin, 5-Adamantyl-IAA (5-Ad-IAA). In AlissAID system, nanomolar concentration of 5-Ad-IAA induces target degradation, thus minimizing the side effects from chemical compounds. In addition, in AlissAID system, we observed few basal degradations which was observed in other AID systems including ssAID system. Furthermore, AlissAID based conditional knockdown cell lines are easily generated by using budding yeast GFP Clone Collection. Target protein, which has antigen recognition sites exposed in cytosol or nucleus, can be degraded by the AlissAID system. From these advantages, the AlissAID system would be an ideal protein-knockdown system in budding yeast cells.
Collapse
Affiliation(s)
- Yoshitaka Ogawa
- Department of Biological Science, Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Kohei Nishimura
- Department of Biological Science, Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Keisuke Obara
- Department of Biological Science, Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Takumi Kamura
- Department of Biological Science, Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| |
Collapse
|
28
|
Phanindhar K, Mishra RK. Auxin-inducible degron system: an efficient protein degradation tool to study protein function. Biotechniques 2023; 74:186-198. [PMID: 37191015 DOI: 10.2144/btn-2022-0108] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023] Open
Abstract
Targeted protein degradation, with its rapid protein depletion kinetics, allows the measurement of acute changes in the cell. The auxin-inducible degron (AID) system, rapidly degrades AID-tagged proteins only in the presence of auxin. The AID system being inducible makes the study of essential genes and dynamic processes like cell differentiation, cell cycle and genome organization feasible. The AID degradation system has been adapted to yeast, protozoans, C. elegans, Drosophila, zebrafish, mouse and mammalian cell lines. Using the AID system, researchers have unveiled novel functions for essential proteins at developmental stages that were previously difficult to investigate due to early lethality. This comprehensive review discusses the development, advancements, applications and drawbacks of the AID system and compares it with other available protein degradation systems.
Collapse
Affiliation(s)
- Kundurthi Phanindhar
- CSIR-Centre for Cellular & Molecular Biology (CCMB), Uppal Road, Hyderabad, 500007, India
| | - Rakesh K Mishra
- CSIR-Centre for Cellular & Molecular Biology (CCMB), Uppal Road, Hyderabad, 500007, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
- Tata Institute for Genetics & Society (TIGS), Bangalore, 560065, India
| |
Collapse
|
29
|
Sun X, Zhou C, Xia S, Chen X. Small molecule-nanobody conjugate induced proximity controls intracellular processes and modulates endogenous unligandable targets. Nat Commun 2023; 14:1635. [PMID: 36964170 PMCID: PMC10039045 DOI: 10.1038/s41467-023-37237-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 03/08/2023] [Indexed: 03/26/2023] Open
Abstract
Chemically induced proximity (CIP) is a powerful tool to study cellular functions. However with current CIP inducers it is difficult to directly modulate unligandable and endogenous targets, and therapeutic translational potential is also restricted. Herein, we combine CIP and chemical nanobody engineering and create cell-permeable small molecule-nanobody conjugate inducers of proximity (SNACIPs). The SNACIP inducer cRGT carrying a cyclic cell-penetrating peptide rapidly enters live cells and dimerizes eDHFR and GFP-variants. cRGT enables minute-scale, reversible, no-wash and dose-dependent control of cellular processes including signaling cascade, cargo transport and ferroptosis. Small-molecule motifs can also be installed via post-translational modifications. Therefore, latent-type SNACIPs including cRTC are designed that are functionally assembled inside living cells. cRTC contains a nanobody against an intrinsically disordered protein TPX2, a microtubule nucleation factor overexpressed in various cancers. Cancer cell proliferation is inhibited and tumor growth is suppressed in vivo. Hence, SNACIPs are valuable proximity inducers for regulating cellular functions.
Collapse
Affiliation(s)
- Xiaofeng Sun
- Laboratory of Chemical Biology & Frontier Biotechnologies, The HIT Center for Life Sciences (HCLS), Harbin Institute of Technology (HIT), Harbin, 150001, PR China
- School of Life Science and Technology, HIT, Harbin, 150001, PR China
| | - Chengjian Zhou
- Laboratory of Chemical Biology & Frontier Biotechnologies, The HIT Center for Life Sciences (HCLS), Harbin Institute of Technology (HIT), Harbin, 150001, PR China
- School of Life Science and Technology, HIT, Harbin, 150001, PR China
| | - Simin Xia
- Laboratory of Chemical Biology & Frontier Biotechnologies, The HIT Center for Life Sciences (HCLS), Harbin Institute of Technology (HIT), Harbin, 150001, PR China
| | - Xi Chen
- Laboratory of Chemical Biology & Frontier Biotechnologies, The HIT Center for Life Sciences (HCLS), Harbin Institute of Technology (HIT), Harbin, 150001, PR China.
- School of Life Science and Technology, HIT, Harbin, 150001, PR China.
| |
Collapse
|
30
|
Nanobody-based RFP-dependent Cre recombinase for selective anterograde tracing in RFP-expressing transgenic animals. Commun Biol 2022; 5:979. [PMID: 36114373 PMCID: PMC9481622 DOI: 10.1038/s42003-022-03944-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 09/05/2022] [Indexed: 11/09/2022] Open
Abstract
AbstractTransgenic animals expressing fluorescent proteins are widely used to label specific cells and proteins. By using a split Cre recombinase fused with mCherry-binding nanobodies or designed ankyrin repeat proteins, we created Cre recombinase dependent on red fluorescent protein (RFP) (Cre-DOR). Functional binding units for monomeric RFPs are different from those for polymeric RFPs. We confirmed selective target RFP-dependent gene expression in the mouse cerebral cortex using stereotaxic injection of adeno-associated virus vectors. In estrogen receptor-beta (Esr2)-mRFP1 mice and gastrin-releasing peptide receptor (Grpr)-mRFP1 rats, we confirmed that Cre-DOR can be used for selective tracing of the neural projection from RFP-expressing specific neurons. Cellular localization of RFPs affects recombination efficiency of Cre-DOR, and light and chemical-induced nuclear translocation of an RFP-fused protein can modulate Cre-DOR efficiency. Our results provide a method for manipulating gene expression in specific cells expressing RFPs and expand the repertory of nanobody-based genetic tools.
Collapse
|
31
|
Li J, Dai C, Xie W, Zhang H, Huang X, Chronis C, Ye Y, Zhang W. A One-step strategy to target essential factors with auxin-inducible degron system in mouse embryonic stem cells. Front Cell Dev Biol 2022; 10:964119. [PMID: 36003152 PMCID: PMC9393215 DOI: 10.3389/fcell.2022.964119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
The self-renewal and pluripotency of embryonic stem cells (ESCs) are conferred by networks including transcription factors and histone modifiers. The Auxin-inducible degron (AID) system can rapidly and reversibly degrade its target proteins and is becoming a powerful tool to explore novel function of key pluripotent and histone modifier genes in ESCs. However, the low biallelic tagging efficiency and a basal degradation level of the current AID systems deem it unsuitable to target key pluripotent genes with tightly controlled expression levels. Here, we develop a one-step strategy to successfully target and repress the endogenous pluripotent genes in mouse ESCs and replace their expression with AID fused transgenes. Therefore, this work provides an efficient way for employing the AID system to uncover novel function of essential pluripotent and chromatin modifier genes in ESCs.
Collapse
Affiliation(s)
- Jingsheng Li
- Cam-Su Genomic Resource Center, Medical College of Soochow University, Suzhou, China
| | - Chunhong Dai
- Cam-Su Genomic Resource Center, Medical College of Soochow University, Suzhou, China
| | - Wenyan Xie
- Cam-Su Genomic Resource Center, Medical College of Soochow University, Suzhou, China
| | - Heyao Zhang
- Cam-Su Genomic Resource Center, Medical College of Soochow University, Suzhou, China
| | - Xin Huang
- Department of Computational Biology St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Constantinos Chronis
- Department of Biochemistry and Molecular Genetics University of Illinois at Chicago, Chicago, IL, United States
| | - Ying Ye
- Cam-Su Genomic Resource Center, Medical College of Soochow University, Suzhou, China
- *Correspondence: Ying Ye, ; Wensheng Zhang,
| | - Wensheng Zhang
- Cam-Su Genomic Resource Center, Medical College of Soochow University, Suzhou, China
- Department of Physiology School of Basic Medical Sciences Binzhou Medical University, Yantai, China
- *Correspondence: Ying Ye, ; Wensheng Zhang,
| |
Collapse
|
32
|
Gunkel P, Cordes VC. ZC3HC1 is a structural element of the nuclear basket effecting interlinkage of TPR polypeptides. Mol Biol Cell 2022; 33:ar82. [PMID: 35609216 DOI: 10.1091/mbc.e22-02-0037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The nuclear basket (NB), anchored to the nuclear pore complex (NPC), is commonly looked upon as a structure built solely of protein TPR polypeptides, the latter thus regarded as the NB's only scaffold-forming components. In the current study, we report ZC3HC1 as a second structural element of the NB. Recently described as an NB-appended protein omnipresent in vertebrates, we now show that ZC3HC1, both in vivo and in vitro, enables in a stepwise manner the recruitment of TPR subpopulations to the NB and their linkage to already NPC-anchored TPR polypeptides. We further demonstrate that the degron-mediated rapid elimination of ZC3HC1 results in the prompt detachment of the ZC3HC1-appended TPR polypeptides from the NB and their release into the nucleoplasm, underscoring the role of ZC3HC1 as a natural structural element of the NB. Finally, we show that ZC3HC1 can keep TPR polypeptides positioned and linked to each other even at sites remote from the NB, in line with ZC3HC1 functioning as a protein connecting TPR polypeptides.
Collapse
Affiliation(s)
- Philip Gunkel
- Max Planck Institute for Multidisciplinary Sciences, 37077 Göttingen, Germany
| | - Volker C Cordes
- Max Planck Institute for Multidisciplinary Sciences, 37077 Göttingen, Germany
| |
Collapse
|
33
|
Naruse C, Sugihara K, Miyazaki T, Pan X, Sugiyama F, Asano M. A degron system targeting endogenous PD-1 inhibits the growth of tumor cells in mice. NAR Cancer 2022; 4:zcac019. [PMID: 35734392 PMCID: PMC9204894 DOI: 10.1093/narcan/zcac019] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 05/26/2022] [Accepted: 06/03/2022] [Indexed: 11/12/2022] Open
Abstract
Recently, targeted protein degradation systems have been developed using the ubiquitin-proteasome system. Here, we established Programmed cell death-1 (PD-1) knockdown mice as a model system for subjecting endogenous mouse proteins to the small molecule-assisted shutoff (SMASh) degron system. SMASh degron-tagged PD-1-mCherry in Jurkat cells and CD3+ splenocytes were degraded by the NS3/4A protease inhibitors, asunaprevir (ASV) or grazoprevir (GRV). Growth of MC-38 colon adenocarcinoma cells injected in Pdcd1-mCherry-SMASh homozygous knock-in (KI) mice was repressed by ASV or GRV. Moreover, growth of MC-38 cells was suppressed in wild-type mice transplanted with KI bone marrow cells after GRV treatment. This is the first study to use a degron tag targeting an endogenous mouse protein in vivo. Our experimental system using the SMASh degron may be employed for treating diseases and characterizing the cellular functions of essential proteins.
Collapse
Affiliation(s)
- Chie Naruse
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Kazushi Sugihara
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Tatsuhiko Miyazaki
- Department of Pathology, Gifu University Hospital, 1-1 Yanagido, Gifu 501-1104, Japan
| | - Xuchi Pan
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Fumihiro Sugiyama
- Laboratory Animal Resource Center, Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Masahide Asano
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
34
|
Fagbadebo FO, Kaiser PD, Zittlau K, Bartlick N, Wagner TR, Froehlich T, Jarjour G, Nueske S, Scholz A, Traenkle B, Macek B, Rothbauer U. A Nanobody-Based Toolset to Monitor and Modify the Mitochondrial GTPase Miro1. Front Mol Biosci 2022; 9:835302. [PMID: 35359597 PMCID: PMC8960383 DOI: 10.3389/fmolb.2022.835302] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/08/2022] [Indexed: 12/24/2022] Open
Abstract
The mitochondrial outer membrane (MOM)-anchored GTPase Miro1, is a central player in mitochondrial transport and homeostasis. The dysregulation of Miro1 in amyotrophic lateral sclerosis (ALS) and Parkinson’s disease (PD) suggests that Miro1 may be a potential biomarker or drug target in neuronal disorders. However, the molecular functionality of Miro1 under (patho-) physiological conditions is poorly known. For a more comprehensive understanding of the molecular functions of Miro1, we have developed Miro1-specific nanobodies (Nbs) as novel research tools. We identified seven Nbs that bind either the N- or C-terminal GTPase domain of Miro1 and demonstrate their application as research tools for proteomic and imaging approaches. To visualize the dynamics of Miro1 in real time, we selected intracellularly functional Nbs, which we reformatted into chromobodies (Cbs) for time-lapse imaging of Miro1. By genetic fusion to an Fbox domain, these Nbs were further converted into Miro1-specific degrons and applied for targeted degradation of Miro1 in live cells. In summary, this study presents a collection of novel Nbs that serve as a toolkit for advanced biochemical and intracellular studies and modulations of Miro1, thereby contributing to the understanding of the functional role of Miro1 in disease-derived model systems.
Collapse
Affiliation(s)
| | - Philipp D. Kaiser
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Katharina Zittlau
- Quantitative Proteomics, Department of Biology, Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Natascha Bartlick
- Interfaculty Institute of Biochemistry, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Teresa R. Wagner
- Pharmaceutical Biotechnology, Eberhard Karls University Tübingen, Tübingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Theresa Froehlich
- Pharmaceutical Biotechnology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Grace Jarjour
- Pharmaceutical Biotechnology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Stefan Nueske
- Livestock Center of the Faculty of Veterinary Medicine, Ludwig Maximilians University Munich, Oberschleissheim, Germany
| | - Armin Scholz
- Livestock Center of the Faculty of Veterinary Medicine, Ludwig Maximilians University Munich, Oberschleissheim, Germany
| | - Bjoern Traenkle
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Boris Macek
- Quantitative Proteomics, Department of Biology, Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Ulrich Rothbauer
- Pharmaceutical Biotechnology, Eberhard Karls University Tübingen, Tübingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
- *Correspondence: Ulrich Rothbauer,
| |
Collapse
|
35
|
Kanemaki MT. Ligand-induced degrons for studying nuclear functions. Curr Opin Cell Biol 2022; 74:29-36. [DOI: 10.1016/j.ceb.2021.12.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 01/21/2023]
|
36
|
Yu D, Won Do H. Optogenetic Activation of Intracellular Nanobodies. Methods Mol Biol 2022; 2446:595-606. [PMID: 35157296 DOI: 10.1007/978-1-0716-2075-5_31] [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: 06/14/2023]
Abstract
Intracellular antibody fragments such as nanobodies and scFvs are powerful tools for imaging and for modulating and neutralizing endogenous target proteins. Optogenetically activated intracellular antibodies (optobodies) constitute a light-inducible system to directly control intrabody activities in cells, with greater spatial and temporal resolution than intracellular antibodies alone. Here, we describe optogenetic and microscopic methods to activate optobodies in cells using a confocal microscope and an automated fluorescence microscope. In the protocol, we use the examples of an optobody targeting green fluorescent protein and an optobody that inhibits the endogenous gelsolin protein.
Collapse
Affiliation(s)
- Daseuli Yu
- Life Science Research Institute, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Heo Won Do
- Life Science Research Institute, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
- KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
| |
Collapse
|
37
|
Ziegler MJ, Yserentant K, Dunsing V, Middel V, Gralak AJ, Pakari K, Bargstedt J, Kern C, Petrich A, Chiantia S, Strähle U, Herten DP, Wombacher R. Mandipropamid as a chemical inducer of proximity for in vivo applications. Nat Chem Biol 2022; 18:64-69. [PMID: 34934192 PMCID: PMC8709788 DOI: 10.1038/s41589-021-00922-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 10/13/2021] [Indexed: 11/26/2022]
Abstract
Direct control of protein interactions by chemically induced protein proximity holds great potential for both cell and synthetic biology as well as therapeutic applications. Low toxicity, orthogonality and excellent cell permeability are important criteria for chemical inducers of proximity (CIPs), in particular for in vivo applications. Here, we present the use of the agrochemical mandipropamid (Mandi) as a highly efficient CIP in cell culture systems and living organisms. Mandi specifically induces complex formation between a sixfold mutant of the plant hormone receptor pyrabactin resistance 1 (PYR1) and abscisic acid insensitive (ABI). It is orthogonal to other plant hormone-based CIPs and rapamycin-based CIP systems. We demonstrate the applicability of the Mandi system for rapid and efficient protein translocation in mammalian cells and zebrafish embryos, protein network shuttling and manipulation of endogenous proteins.
Collapse
Affiliation(s)
- Michael J Ziegler
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg, Germany
- Department of Chemical Biology, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Klaus Yserentant
- Institute of Physical Chemistry, Heidelberg University, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Institute of Cardiovascular Sciences & School of Chemistry, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Valentin Dunsing
- Institute of Biology and Biochemistry, University of Potsdam, Potsdam, Germany
| | - Volker Middel
- Institute of Biological and Chemical Systems (IBCS)-Biological Information Processing (BIP), Karlsruhe Institute of Technology (KIT), Eggenstein-Leopoldshafen, Germany
| | - Antoni J Gralak
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg, Germany
- Institute of Physical Chemistry, Heidelberg University, Heidelberg, Germany
| | - Kaisa Pakari
- Institute of Physical Chemistry, Heidelberg University, Heidelberg, Germany
| | - Jörn Bargstedt
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg, Germany
| | - Christoph Kern
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg, Germany
- Institute of Physical Chemistry, Heidelberg University, Heidelberg, Germany
| | - Annett Petrich
- Institute of Biology and Biochemistry, University of Potsdam, Potsdam, Germany
| | - Salvatore Chiantia
- Institute of Biology and Biochemistry, University of Potsdam, Potsdam, Germany
| | - Uwe Strähle
- Institute of Biological and Chemical Systems (IBCS)-Biological Information Processing (BIP), Karlsruhe Institute of Technology (KIT), Eggenstein-Leopoldshafen, Germany
| | - Dirk-Peter Herten
- Institute of Physical Chemistry, Heidelberg University, Heidelberg, Germany
- Institute of Cardiovascular Sciences & School of Chemistry, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Midlands, UK
| | - Richard Wombacher
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg, Germany.
- Department of Chemical Biology, Max Planck Institute for Medical Research, Heidelberg, Germany.
| |
Collapse
|
38
|
Wagner TR, Rothbauer U. Nanobodies - Little helpers unravelling intracellular signaling. Free Radic Biol Med 2021; 176:46-61. [PMID: 34536541 DOI: 10.1016/j.freeradbiomed.2021.09.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/26/2021] [Accepted: 09/08/2021] [Indexed: 11/21/2022]
Abstract
The identification of diagnostic and therapeutic targets requires a comprehensive understanding of cellular processes, for which advanced technologies in biomedical research are needed. The emergence of nanobodies (Nbs) derived from antibody fragments of camelid heavy chain-only antibodies as intracellular research tools offers new possibilities to study and modulate target antigens in living cells. Here we summarize this rapidly changing field, beginning with a brief introduction of Nbs, followed by an overview of how target-specific Nbs can be generated, and introduce the selection of intrabodies as research tools. Intrabodies, by definition, are intracellular functional Nbs that target ectopic or endogenous intracellular antigens within living cells. Such binders can be applied in various formats, e.g. as chromobodies for live cell microscopy or as biosensors to decipher complex intracellular signaling pathways. In addition, protein knockouts can be achieved by target-specific Nbs, while modulating Nbs have the potential as future therapeutics. The development of fine-tunable and switchable Nb-based systems that simultaneously provide spatial and temporal control has recently taken the application of these binders to the next level.
Collapse
Affiliation(s)
- Teresa R Wagner
- Pharmaceutical Biotechnology, Eberhard Karls University, Tübingen, Germany; NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Ulrich Rothbauer
- Pharmaceutical Biotechnology, Eberhard Karls University, Tübingen, Germany; NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany; Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany.
| |
Collapse
|
39
|
Wang R, Xu L, Wang J, Sun L, Jiao Y, Meng Y, Chen S, Chang C, Fan C. Electric Fano resonance-based terahertz metasensors. NANOSCALE 2021; 13:18467-18472. [PMID: 34726683 DOI: 10.1039/d1nr04477j] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
An ultra-sensitive THz metasensor is presented based on quasi-BIC Fano resonance, which can distinguish extremely dilute concentrations (nM) of solutions. It provides a nondestructive sensing approach for disease prevention and diagnosis. However, the main drawback limiting the performance of THz-based bio-chemical sensors is the weak interaction between the optical field and the analyte, the characteristic scale of which is mismatched with the THz wavelength, leading to low sensitivity. Herein, we present an ultra-sensitive THz metasensor based on an electric Fano resonant metasurface which consists of three gold microrods arranged periodically. The designed electric Fano resonance provides a strong near-field enhancement near the surface of the microstructure, significantly boosting the light-analyte interactions and thus the sensitivity. Such an electric Fano resonance is formed by the interference between a leaky electric dipole resonance and a bound toroidal dipole mode which is a symmetry-protected bound state in the continuum supported by the sub-diffractive periodic system here. Owing to the strong electric fields generated near the interface of our microstructure around the toroidal dipole BIC, the proposed structure can distinguish extremely dilute concentrations (nM) of solutions. Importantly, by controlling the degree of geometrical asymmetry, the BIC-inspired mechanism provides an important and simple tool to engineer and tailor the linewidth and Q-factor of our proposed electric Fano resonance, indicating the ability to realize different biosensors for different optical regimes. Our results open new possibilities to realize a non-destructive and non-contact quantitative inspection of low-concentration solutions, providing a useful sensing approach for disease prevention and diagnosis.
Collapse
Affiliation(s)
- Ride Wang
- Innovation Laboratory of Terahertz Biophysics, National Innovation Institute of Defense Technology, Beijing, 100071, China.
| | - Lei Xu
- Advanced Optics and Photonics Laboratory, Department of Engineering, School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, UK
| | - Jiayi Wang
- Key Laboratory of Weak-Light Nonlinear Photonics, Ministry of Education, TEDA Institute of Applied Physics and School of Physics, Nankai University, Tianjin 300457, China
| | - Lang Sun
- Innovation Laboratory of Terahertz Biophysics, National Innovation Institute of Defense Technology, Beijing, 100071, China.
| | - Yanan Jiao
- Department of General Surgery, First Medical Center of Chinese PLA General Hospital, Beijing 100853, People's Republic of China
| | - Yuan Meng
- Key Laboratory of Photonics Control Technology of the Ministry of Education, Tsinghua University, China
| | - Shuo Chen
- Innovation Laboratory of Terahertz Biophysics, National Innovation Institute of Defense Technology, Beijing, 100071, China.
| | - Chao Chang
- Innovation Laboratory of Terahertz Biophysics, National Innovation Institute of Defense Technology, Beijing, 100071, China.
- School of Physics, Peking University, Beijing, 100871, China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
40
|
Akkapeddi P, Teng KW, Koide S. Monobodies as tool biologics for accelerating target validation and druggable site discovery. RSC Med Chem 2021; 12:1839-1853. [PMID: 34820623 PMCID: PMC8597423 DOI: 10.1039/d1md00188d] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/26/2021] [Indexed: 12/21/2022] Open
Abstract
Despite increased investment and technological advancement, new drug approvals have not proportionally increased. Low drug approval rates, particularly for new targets, are linked to insufficient target validation at early stages. Thus, there remains a strong need for effective target validation techniques. Here, we review the use of synthetic binding proteins as tools for drug target validation, with focus on the monobody platform among several advanced synthetic binding protein platforms. Monobodies with high affinity and high selectivity can be rapidly developed against challenging targets, such as KRAS mutants, using protein engineering technologies. They have strong tendency to bind to functional sites and thus serve as drug-like molecules, and they can serve as targeting ligands for constructing bio-PROTACs. Genetically encoded monobodies are effective "tool biologics" for validating intracellular targets. They promote crystallization and help reveal the atomic structures of the monobody-target interface, which can inform drug design. Using case studies, we illustrate the potential of the monobody technology in accelerating target validation and small-molecule drug discovery.
Collapse
Affiliation(s)
- Padma Akkapeddi
- Perlmutter Cancer Center, New York University Langone Medical Center New York NY USA
| | - Kai Wen Teng
- Perlmutter Cancer Center, New York University Langone Medical Center New York NY USA
| | - Shohei Koide
- Perlmutter Cancer Center, New York University Langone Medical Center New York NY USA
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine New York NY USA
| |
Collapse
|
41
|
Levic DS, Yamaguchi N, Wang S, Knaut H, Bagnat M. Knock-in tagging in zebrafish facilitated by insertion into non-coding regions. Development 2021; 148:dev199994. [PMID: 34495314 PMCID: PMC8513609 DOI: 10.1242/dev.199994] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 08/27/2021] [Indexed: 11/20/2022]
Abstract
Zebrafish provide an excellent model for in vivo cell biology studies because of their amenability to live imaging. Protein visualization in zebrafish has traditionally relied on overexpression of fluorescently tagged proteins from heterologous promoters, making it difficult to recapitulate endogenous expression patterns and protein function. One way to circumvent this problem is to tag the proteins by modifying their endogenous genomic loci. Such an approach is not widely available to zebrafish researchers because of inefficient homologous recombination and the error-prone nature of targeted integration in zebrafish. Here, we report a simple approach for tagging proteins in zebrafish on their N or C termini with fluorescent proteins by inserting PCR-generated donor amplicons into non-coding regions of the corresponding genes. Using this approach, we generated endogenously tagged alleles for several genes that are crucial for epithelial biology and organ development, including the tight junction components ZO-1 and Cldn15la, the trafficking effector Rab11a, the apical polarity protein aPKC and the ECM receptor Integrin β1b. Our approach facilitates the generation of knock-in lines in zebrafish, opening the way for accurate quantitative imaging studies.
Collapse
Affiliation(s)
- Daniel S. Levic
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Naoya Yamaguchi
- Skirball Institute of Biomolecular Medicine, New York University Grossman School of Medicine, Department of Cell Biology, New York, NY 10016, USA
| | - Siyao Wang
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Holger Knaut
- Skirball Institute of Biomolecular Medicine, New York University Grossman School of Medicine, Department of Cell Biology, New York, NY 10016, USA
| | - Michel Bagnat
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| |
Collapse
|
42
|
Choe CP, Choi SY, Kee Y, Kim MJ, Kim SH, Lee Y, Park HC, Ro H. Transgenic fluorescent zebrafish lines that have revolutionized biomedical research. Lab Anim Res 2021; 37:26. [PMID: 34496973 PMCID: PMC8424172 DOI: 10.1186/s42826-021-00103-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/26/2021] [Indexed: 12/22/2022] Open
Abstract
Since its debut in the biomedical research fields in 1981, zebrafish have been used as a vertebrate model organism in more than 40,000 biomedical research studies. Especially useful are zebrafish lines expressing fluorescent proteins in a molecule, intracellular organelle, cell or tissue specific manner because they allow the visualization and tracking of molecules, intracellular organelles, cells or tissues of interest in real time and in vivo. In this review, we summarize representative transgenic fluorescent zebrafish lines that have revolutionized biomedical research on signal transduction, the craniofacial skeletal system, the hematopoietic system, the nervous system, the urogenital system, the digestive system and intracellular organelles.
Collapse
Affiliation(s)
- Chong Pyo Choe
- Division of Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea.,Division of Applied Life Science, Plant Molecular Biology and Biotechnology Research Center, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Seok-Yong Choi
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - Yun Kee
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| | - Min Jung Kim
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Seok-Hyung Kim
- Department of Marine Life Sciences and Fish Vaccine Research Center, Jeju National University, Jeju, 63243, Republic of Korea
| | - Yoonsung Lee
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan, 44919, Republic of Korea
| | - Hae-Chul Park
- Department of Biomedical Sciences, College of Medicine, Korea University, Ansan, 15355, Republic of Korea
| | - Hyunju Ro
- Department of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University, Daejeon, 34134, Republic of Korea
| |
Collapse
|
43
|
Vo AA, Levenson MT, Ragle JM, Ward JD. Efficient generation of a single-copy eft-3p::TIR1::F2A:: BFP::AID*::NLS allele in the C. elegans ttTi5605 insertion site through recombination-mediated cassette exchange. MICROPUBLICATION BIOLOGY 2021; 2021. [PMID: 34355140 PMCID: PMC8335552 DOI: 10.17912/micropub.biology.000425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 11/17/2022]
Abstract
The auxin-inducible degron (AID) system is a widely used system to conditionally deplete proteins. Using CRISPR/Cas9-based genome editing in C. elegans, we recently generated a set of single-copy, tissue-specific and pan-somatic TIR1-expressing strains carrying a BFP reporter inserted in single-copy into two commonly used, well-characterized genetic loci. However, we were unable to obtain a strain carrying a pan-somatic eft-3p::TIR1::F2A::BFP::AID*::NLS transgene inserted into the chromosome II ttTi5605 insertion site. Using recombination-mediated cassette exchange (RMCE) we were able to efficiently obtain this knock-in. The resulting strain displayed equivalent depletion of an AID*::GFP reporter compared to our previously generated eft-3p::TIR1::F2A::BFP::AID*::NLS transgene knocked into the chromosome I ttTi4348 insertion site. This work highlights the power of RMCE for generating new reagents for the AID system and provides an eft-3p::TIR1::F2A::BFP::AID*::NLS allele on chromosome II which will simplify genetic crossing schemes when using the AID system.
Collapse
Affiliation(s)
- An A Vo
- Department of Molecular, Cell, and Developmental Biology, University of California - Santa Cruz, Santa Cruz, CA, USA
| | - Max T Levenson
- Department of Molecular, Cell, and Developmental Biology, University of California - Santa Cruz, Santa Cruz, CA, USA
| | - James Matthew Ragle
- Department of Molecular, Cell, and Developmental Biology, University of California - Santa Cruz, Santa Cruz, CA, USA
| | - Jordan D Ward
- Department of Molecular, Cell, and Developmental Biology, University of California - Santa Cruz, Santa Cruz, CA, USA
| |
Collapse
|
44
|
Chemical tools for dissecting cell division. Nat Chem Biol 2021; 17:632-640. [PMID: 34035515 PMCID: PMC10157795 DOI: 10.1038/s41589-021-00798-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 04/13/2021] [Indexed: 02/03/2023]
Abstract
Components of the cell division machinery typically function at varying cell cycle stages and intracellular locations. To dissect cellular mechanisms during the rapid division process, small-molecule probes act as complementary approaches to genetic manipulations, with advantages of temporal and in some cases spatial control and applicability to multiple model systems. This Review focuses on recent advances in chemical probes and applications to address select questions in cell division. We discuss uses of both enzyme inhibitors and chemical inducers of dimerization, as well as emerging techniques to promote future investigations. Overall, these concepts may open new research directions for applying chemical probes to advance cell biology.
Collapse
|
45
|
Mangeat T, Labouesse S, Allain M, Negash A, Martin E, Guénolé A, Poincloux R, Estibal C, Bouissou A, Cantaloube S, Vega E, Li T, Rouvière C, Allart S, Keller D, Debarnot V, Wang XB, Michaux G, Pinot M, Le Borgne R, Tournier S, Suzanne M, Idier J, Sentenac A. Super-resolved live-cell imaging using random illumination microscopy. CELL REPORTS METHODS 2021; 1:100009. [PMID: 35474693 PMCID: PMC9017237 DOI: 10.1016/j.crmeth.2021.100009] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/12/2021] [Accepted: 04/08/2021] [Indexed: 12/11/2022]
Abstract
Current super-resolution microscopy (SRM) methods suffer from an intrinsic complexity that might curtail their routine use in cell biology. We describe here random illumination microscopy (RIM) for live-cell imaging at super-resolutions matching that of 3D structured illumination microscopy, in a robust fashion. Based on speckled illumination and statistical image reconstruction, easy to implement and user-friendly, RIM is unaffected by optical aberrations on the excitation side, linear to brightness, and compatible with multicolor live-cell imaging over extended periods of time. We illustrate the potential of RIM on diverse biological applications, from the mobility of proliferating cell nuclear antigen (PCNA) in U2OS cells and kinetochore dynamics in mitotic S. pombe cells to the 3D motion of myosin minifilaments deep inside Drosophila tissues. RIM's inherent simplicity and extended biological applicability, particularly for imaging at increased depths, could help make SRM accessible to biology laboratories.
Collapse
Affiliation(s)
- Thomas Mangeat
- LITC Core Facility, Centre de Biologie Integrative, Université de Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Simon Labouesse
- Institut Fresnel, Aix Marseille Université, CNRS, Centrale Marseille, Marseille, France
| | - Marc Allain
- Institut Fresnel, Aix Marseille Université, CNRS, Centrale Marseille, Marseille, France
| | - Awoke Negash
- Institut Fresnel, Aix Marseille Université, CNRS, Centrale Marseille, Marseille, France
| | - Emmanuel Martin
- Molecular, Cellular & Developmental Biology (MCD), Center of Integrative Biology (CBI), Toulouse University, CNRS, UPS, Toulouse, France
| | - Aude Guénolé
- Molecular, Cellular & Developmental Biology (MCD), Center of Integrative Biology (CBI), Toulouse University, CNRS, UPS, Toulouse, France
| | - Renaud Poincloux
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Claire Estibal
- LITC Core Facility, Centre de Biologie Integrative, Université de Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Anaïs Bouissou
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Sylvain Cantaloube
- LITC Core Facility, Centre de Biologie Integrative, Université de Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Elodie Vega
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Tong Li
- Molecular, Cellular & Developmental Biology (MCD), Center of Integrative Biology (CBI), Toulouse University, CNRS, UPS, Toulouse, France
| | - Christian Rouvière
- LITC Core Facility, Centre de Biologie Integrative, Université de Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Sophie Allart
- INSERM Université de Toulouse, UPS, CNRS, Centre de Physiopathologie de Toulouse Purpan (CPTP), Toulouse, France
| | - Debora Keller
- Molecular, Cellular & Developmental Biology (MCD), Center of Integrative Biology (CBI), Toulouse University, CNRS, UPS, Toulouse, France
| | - Valentin Debarnot
- LITC Core Facility, Centre de Biologie Integrative, Université de Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Xia Bo Wang
- Molecular, Cellular & Developmental Biology (MCD), Center of Integrative Biology (CBI), Toulouse University, CNRS, UPS, Toulouse, France
| | - Grégoire Michaux
- Univ Rennes, CNRS, Institut de Génétique et Développement de Rennes (IGDR) - UMR 6290, 35000 Rennes, France
| | - Mathieu Pinot
- Univ Rennes, CNRS, Institut de Génétique et Développement de Rennes (IGDR) - UMR 6290, 35000 Rennes, France
| | - Roland Le Borgne
- Univ Rennes, CNRS, Institut de Génétique et Développement de Rennes (IGDR) - UMR 6290, 35000 Rennes, France
| | - Sylvie Tournier
- Molecular, Cellular & Developmental Biology (MCD), Center of Integrative Biology (CBI), Toulouse University, CNRS, UPS, Toulouse, France
| | - Magali Suzanne
- Molecular, Cellular & Developmental Biology (MCD), Center of Integrative Biology (CBI), Toulouse University, CNRS, UPS, Toulouse, France
| | - Jérome Idier
- LS2N, CNRS UMR 6004, 1 rue de la Noë, F44321 Nantes Cedex 3, France
| | - Anne Sentenac
- Institut Fresnel, Aix Marseille Université, CNRS, Centrale Marseille, Marseille, France
| |
Collapse
|
46
|
Ashley GE, Duong T, Levenson MT, Martinez MAQ, Johnson LC, Hibshman JD, Saeger HN, Palmisano NJ, Doonan R, Martinez-Mendez R, Davidson BR, Zhang W, Ragle JM, Medwig-Kinney TN, Sirota SS, Goldstein B, Matus DQ, Dickinson DJ, Reiner DJ, Ward JD. An expanded auxin-inducible degron toolkit for Caenorhabditis elegans. Genetics 2021; 217:iyab006. [PMID: 33677541 PMCID: PMC8045686 DOI: 10.1093/genetics/iyab006] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/10/2021] [Indexed: 12/30/2022] Open
Abstract
The auxin-inducible degron (AID) system has emerged as a powerful tool to conditionally deplete proteins in a range of organisms and cell types. Here, we describe a toolkit to augment the use of the AID system in Caenorhabditis elegans. We have generated a set of single-copy, tissue-specific (germline, intestine, neuron, muscle, pharynx, hypodermis, seam cell, anchor cell) and pan-somatic TIR1-expressing strains carrying a co-expressed blue fluorescent reporter to enable use of both red and green channels in experiments. These transgenes are inserted into commonly used, well-characterized genetic loci. We confirmed that our TIR1-expressing strains produce the expected depletion phenotype for several nuclear and cytoplasmic AID-tagged endogenous substrates. We have also constructed a set of plasmids for constructing repair templates to generate fluorescent protein::AID fusions through CRISPR/Cas9-mediated genome editing. These plasmids are compatible with commonly used genome editing approaches in the C. elegans community (Gibson or SapTrap assembly of plasmid repair templates or PCR-derived linear repair templates). Together these reagents will complement existing TIR1 strains and facilitate rapid and high-throughput fluorescent protein::AID tagging of genes. This battery of new TIR1-expressing strains and modular, efficient cloning vectors serves as a platform for straightforward assembly of CRISPR/Cas9 repair templates for conditional protein depletion.
Collapse
Affiliation(s)
- Guinevere E Ashley
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Tam Duong
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Texas A&M University, Houston, TX 77030, USA
| | - Max T Levenson
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Michael A Q Martinez
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Londen C Johnson
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Jonathan D Hibshman
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hannah N Saeger
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Nicholas J Palmisano
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Ryan Doonan
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Raquel Martinez-Mendez
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Brittany R Davidson
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Wan Zhang
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - James Matthew Ragle
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Taylor N Medwig-Kinney
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Sydney S Sirota
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Bob Goldstein
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - David Q Matus
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Daniel J Dickinson
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - David J Reiner
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Texas A&M University, Houston, TX 77030, USA
| | - Jordan D Ward
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| |
Collapse
|
47
|
Bhoi A, Palladino F, Fabrizio P. Auxin confers protection against ER stress in Caenorhabditis elegans. Biol Open 2021; 10:bio.057992. [PMID: 33495210 PMCID: PMC7875485 DOI: 10.1242/bio.057992] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Auxins are plant growth regulators that influence most aspects of plant development through complex mechanisms. The development of an auxin-inducible degradation (AID) system has enabled rapid, conditional protein depletion in yeast and cultured cells. More recently, the system was successfully adapted to Caenorhabditiselegans to achieve auxin-dependent degradation of targets in all tissues and developmental stages. Whether auxin treatment alone has an impact on nematode physiology is an open question. Here we show that indole-3-acetic acid (IAA), the auxin most commonly used to trigger AID in worms, functions through the conserved IRE-1/XBP-1 branch of the Unfolded Protein Response (UPR) to promote resistance to endoplasmic reticulum (ER) stress. Because the UPR not only plays a central role in restoring ER homeostasis, but also promotes lipid biosynthesis and regulates lifespan, we suggest that extreme caution should be exercised when using the AID system to study these and related processes. Summary: Auxin, commonly used to induce conditional protein degradation, promotes ER stress resistance in C. elegans through the unfolded protein response (UPR).
Collapse
Affiliation(s)
- Anupam Bhoi
- Laboratory of Biology and Modelling of the Cell, Ecole Normale Supérieure de Lyon, CNRS UMR5239, INSERM U1210, Université de Lyon, 69007 Lyon, France
| | - Francesca Palladino
- Laboratory of Biology and Modelling of the Cell, Ecole Normale Supérieure de Lyon, CNRS UMR5239, INSERM U1210, Université de Lyon, 69007 Lyon, France
| | - Paola Fabrizio
- Laboratory of Biology and Modelling of the Cell, Ecole Normale Supérieure de Lyon, CNRS UMR5239, INSERM U1210, Université de Lyon, 69007 Lyon, France
| |
Collapse
|
48
|
Wang W, Yuan J, Jiang C. Applications of nanobodies in plant science and biotechnology. PLANT MOLECULAR BIOLOGY 2021; 105:43-53. [PMID: 33037986 PMCID: PMC7547553 DOI: 10.1007/s11103-020-01082-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 10/05/2020] [Indexed: 05/15/2023]
Abstract
Present review summarizes the current applications of nanobodies in plant science and biotechnology, including plant expression of nanobodies, plant biotechnological applications, nanobody-based immunodetection, and nanobody-mediated resistance against plant pathogens. Nanobodies (Nbs) are variable domains of heavy chain-only antibodies (HCAbs) isolated from camelids. In spite of their single domain structure, nanobodies display many unique features, such as small size, high stability, and cryptic epitopes accessibility, which make them ideal for sophisticated applications in plants and animals. In this review, we summarize the current applications of nanobodies in plant science and biotechnology, focusing on nanobody expression in plants, plant biotechnological applications, determination of plant toxins and pathogens, and nanobody-mediated resistance against plant pathogens. Prospects and challenges of nanobody applications in plants are also discussed.
Collapse
Affiliation(s)
- Wenyi Wang
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, Hubei, China.
- Precision Medicine R&D Center, Zhuhai Institute of Advanced Technology, Chinese Academy of Sciences, Zhuhai, Guangdong Province, China.
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China.
| | - Jumao Yuan
- Precision Medicine R&D Center, Zhuhai Institute of Advanced Technology, Chinese Academy of Sciences, Zhuhai, Guangdong Province, China
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
| | - Changan Jiang
- Precision Medicine R&D Center, Zhuhai Institute of Advanced Technology, Chinese Academy of Sciences, Zhuhai, Guangdong Province, China
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
| |
Collapse
|
49
|
|
50
|
Uhlemann EME, Yu CH, Patry J, Dolgova N, Lutsenko S, Muyldermans S, Dmitriev OY. Nanobodies against the metal binding domains of ATP7B as tools to study copper transport in the cell. Metallomics 2020; 12:1941-1950. [PMID: 33094790 DOI: 10.1039/d0mt00191k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Nanobodies are genetically engineered single domain antibodies derived from the unusual heavy-chain only antibodies found in llamas and camels. The small size of the nanobodies and flexible selection schemes make them uniquely versatile tools for protein biochemistry and cell biology. We have developed a panel of nanobodies against the metal binding domains of the human copper transporter ATP7B, a multidomain membrane protein with a complex regulation of enzymatic activity and intracellular localization. To enable the use of the nanobodies as tools to investigate copper transport in the cell, we characterized their binding sites and affinity by isothermal titration calorimetry and NMR. We have identified nanobodies against each of the first four metal binding domains of ATP7B, with a wide affinity range, as evidenced by dissociation constants from below 10-9 to 10-6 M. We found both the inhibitory and activating nanobodies among those tested. The diverse properties of the nanobodies make the panel useful for the structural studies of ATP7B, immunoaffinity purification of the protein, modulation of its activity in the cell, protein dynamics studies, and as mimics of copper chaperone ATOX1, the natural interaction partner of ATP7B.
Collapse
Affiliation(s)
- Eva-Maria E Uhlemann
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, Canada.
| | | | | | | | | | | | | |
Collapse
|