1
|
Benn J, Cheng S, Keeling S, Smith AE, Vaysburd MJ, Böken D, Miller LVC, Katsinelos T, Franco C, Dupré E, Danis C, Landrieu I, Buée L, Klenerman D, James LC, McEwan WA. Aggregate-selective removal of pathological tau by clustering-activated degraders. Science 2024; 385:1009-1016. [PMID: 39208111 PMCID: PMC7616837 DOI: 10.1126/science.adp5186] [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: 03/29/2024] [Accepted: 07/25/2024] [Indexed: 09/04/2024]
Abstract
Selective degradation of pathological protein aggregates while sparing monomeric forms is of major therapeutic interest. The E3 ligase tripartite motif-containing protein 21 (TRIM21) degrades antibody-bound proteins in an assembly state-specific manner due to the requirement of TRIM21 RING domain clustering for activation, yet effective targeting of intracellular assemblies remains challenging. Here, we fused the RING domain of TRIM21 to a target-specific nanobody to create intracellularly expressed constructs capable of selectively degrading assembled proteins. We evaluated this approach against green fluorescent protein-tagged histone 2B (H2B-GFP) and tau, a protein that undergoes pathological aggregation in Alzheimer's and other neurodegenerative diseases. RING-nanobody degraders prevented or reversed tau aggregation in culture and in vivo, with minimal impact on monomeric tau. This approach may have therapeutic potential for the many disorders driven by intracellular protein aggregation.
Collapse
Affiliation(s)
- Jonathan Benn
- UK Dementia Research Institute at the University of Cambridge, Cambridge CB2 0AH, UK
| | - Shi Cheng
- UK Dementia Research Institute at the University of Cambridge, Cambridge CB2 0AH, UK
| | - Sophie Keeling
- UK Dementia Research Institute at the University of Cambridge, Cambridge CB2 0AH, UK
| | - Annabel E Smith
- UK Dementia Research Institute at the University of Cambridge, Cambridge CB2 0AH, UK
| | | | - Dorothea Böken
- UK Dementia Research Institute at the University of Cambridge, Cambridge CB2 0AH, UK
| | | | - Taxiarchis Katsinelos
- UK Dementia Research Institute at the University of Cambridge, Cambridge CB2 0AH, UK
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | | | - Elian Dupré
- CNRS EMR9002-BSI-Integrative Structural Biology, LabEx DISTALZ, F-59000 Lille, France
- Université Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE, Risk Factors and Molecular Determinants of Aging-Related Diseases, F-59000 Lille, France
| | - Clément Danis
- CNRS EMR9002-BSI-Integrative Structural Biology, LabEx DISTALZ, F-59000 Lille, France
- Université Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE, Risk Factors and Molecular Determinants of Aging-Related Diseases, F-59000 Lille, France
- Université Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, LabEx DISTALZ, F-59000 Lille, France
| | - Isabelle Landrieu
- CNRS EMR9002-BSI-Integrative Structural Biology, LabEx DISTALZ, F-59000 Lille, France
- Université Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE, Risk Factors and Molecular Determinants of Aging-Related Diseases, F-59000 Lille, France
| | - Luc Buée
- Université Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, LabEx DISTALZ, F-59000 Lille, France
| | - David Klenerman
- UK Dementia Research Institute at the University of Cambridge, Cambridge CB2 0AH, UK
| | - Leo C James
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - William A McEwan
- UK Dementia Research Institute at the University of Cambridge, Cambridge CB2 0AH, UK
| |
Collapse
|
2
|
Ogasawara S. Replication-competent influenza virus with a protein-responsive multiplication ability. N Biotechnol 2023; 77:100-110. [PMID: 37586547 DOI: 10.1016/j.nbt.2023.08.001] [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: 02/26/2023] [Revised: 07/30/2023] [Accepted: 08/12/2023] [Indexed: 08/18/2023]
Abstract
Applications of influenza A viruses (IAV) for virotherapy and biotechnology have accelerated substantially with the development of reverse genetic technology and advances in the understanding of packaging signals. While the use of a replication-competent IAV is particularly promising, owing to its efficient transmission to organ depths with high infectivity, there is also a risk that its multiplication cannot be controlled in a cell-type-specific manner, causing an infectious disease. Therefore, here a simple and effective replication-competent IAV-based cell-targeting system has been developed. It was demonstrated that the activity of the ribonucleoprotein complex (RNP) of IAV could be regulated by the interaction between the endogenous protein and a nanobody fused to the subunit of RNA-dependent RNA polymerase (RdRp). To validate the feasibility of the method, it was demonstrated that RNP containing RdRp fused with Nb139, a nanobody against p53, is inactive in HEK293T cells expressing endogenous p53, but active in p53-defective Saos-2 cells. Finally, a replication-competent IAV was successfully generated that multiplies only in p53-defective tumor cells and an IAV vector was developed that can deliver a foreign gene in cell type-specific manner. The method is flexible because the nanobody can be easily altered to target a different cell type, offering a valuable platform for virotherapy and biotechnology.
Collapse
Affiliation(s)
- Shinzi Ogasawara
- Department of Biology, Faculty of Science, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan.
| |
Collapse
|
3
|
Jayanthi BE, Jayanthi S, Segatori L. Design of Oscillatory Networks through Post-Translational Control of Network Components. SYNTHETIC BIOLOGY AND ENGINEERING 2023; 1:10004. [PMID: 38590452 PMCID: PMC11000592 DOI: 10.35534/sbe.2023.10004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Many essential functions in biological systems, including cell cycle progression and circadian rhythm regulation, are governed by the periodic behaviors of specific molecules. These periodic behaviors arise from the precise arrangement of components in biomolecular networks that generate oscillatory output signals. The dynamic properties of individual components of these networks, such as maturation delays and degradation rates, often play a key role in determining the network's oscillatory behavior. In this study, we explored the post-translational modulation of network components as a means to generate genetic circuits with oscillatory behaviors and perturb the oscillation features. Specifically, we used the NanoDeg platform-A bifunctional molecule consisting of a target-specific nanobody and a degron tag-to control the degradation rates of the circuit's components and predicted the effect of NanoDeg-mediated post-translational depletion of a key circuit component on the behavior of a series of proto-oscillating network topologies. We modeled the behavior of two main classes of oscillators, namely relaxation oscillator topologies (the activator-repressor and the Goodwin oscillator) and ring oscillator topologies (repressilators). We identified two main mechanisms by which non-oscillating networks could be induced to oscillate through post-translational modulation of network components: an increase in the separation of timescales of network components and mitigation of the leaky expression of network components. These results are in agreement with previous findings describing the effect of timescale separation and mitigation of leaky expression on oscillatory behaviors. This work thus validates the use of tools to control protein degradation rates as a strategy to modulate existing oscillatory signals and construct oscillatory networks. In addition, this study provides the design rules to implement such an approach based on the control of protein degradation rates using the NanoDeg platform, which does not require genetic manipulation of the network components and can be adapted to virtually any cellular protein. This work also establishes a framework to explore the use of tools for post-translational perturbations of biomolecular networks and generates desired behaviors of the network output.
Collapse
Affiliation(s)
- Brianna E.K. Jayanthi
- Systems, Synthetic, and Physical Biology Graduate Program, Rice University, Houston, TX 77005, USA
| | - Shridhar Jayanthi
- Department of Bioengineering, Rice University, Houston, TX 77005, USA
| | - Laura Segatori
- Systems, Synthetic, and Physical Biology Graduate Program, Rice University, Houston, TX 77005, USA
- Department of Bioengineering, Rice University, Houston, TX 77005, USA
- Department of Chemical & Biomolecular Engineering, Rice University, Houston, TX 77005, USA
- Department of BioSciences, Rice University, Houston, TX 77005, USA
| |
Collapse
|
4
|
Wang H, Guo M, Wei H, Chen Y. Targeting p53 pathways: mechanisms, structures, and advances in therapy. Signal Transduct Target Ther 2023; 8:92. [PMID: 36859359 PMCID: PMC9977964 DOI: 10.1038/s41392-023-01347-1] [Citation(s) in RCA: 233] [Impact Index Per Article: 116.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 12/19/2022] [Accepted: 02/07/2023] [Indexed: 03/03/2023] Open
Abstract
The TP53 tumor suppressor is the most frequently altered gene in human cancers, and has been a major focus of oncology research. The p53 protein is a transcription factor that can activate the expression of multiple target genes and plays critical roles in regulating cell cycle, apoptosis, and genomic stability, and is widely regarded as the "guardian of the genome". Accumulating evidence has shown that p53 also regulates cell metabolism, ferroptosis, tumor microenvironment, autophagy and so on, all of which contribute to tumor suppression. Mutations in TP53 not only impair its tumor suppressor function, but also confer oncogenic properties to p53 mutants. Since p53 is mutated and inactivated in most malignant tumors, it has been a very attractive target for developing new anti-cancer drugs. However, until recently, p53 was considered an "undruggable" target and little progress has been made with p53-targeted therapies. Here, we provide a systematic review of the diverse molecular mechanisms of the p53 signaling pathway and how TP53 mutations impact tumor progression. We also discuss key structural features of the p53 protein and its inactivation by oncogenic mutations. In addition, we review the efforts that have been made in p53-targeted therapies, and discuss the challenges that have been encountered in clinical development.
Collapse
Affiliation(s)
- Haolan Wang
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Ming Guo
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Hudie Wei
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Yongheng Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
5
|
Koulgi S, Achalere A, Sonavane U, Joshi R. Markov State Modeling Analysis Captures Changes in the Temperature-Sensitive N-Terminal and β-Turn Regions of the p53 DNA-Binding Domain. J Chem Inf Model 2022; 62:6449-6461. [PMID: 35614540 DOI: 10.1021/acs.jcim.2c00380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The transcription factor p53 is one of the most widely studied cancer proteins. Its temperature-sensitive nature suggests reduction in functionality at physiological temperatures. Temperature-induced conformational variations and their impact on its functional ability still remain unexplored. A total of 20.8 μs molecular dynamics simulations of wildtype p53 in the apo and the DNA-bound states have been performed at 300 K and 310 K. Further, Markov State Modeling (MSM) analyses were performed, considering Cα-Cα distances as reaction coordinates. Filtering of these distances based on correlation with the time-independent components (tICs) resulted in 16 and 32 distances for apo and DNA-bound systems, respectively. Individual MSM analyses using these filtered distances were performed for both p53 systems. These Cα-Cα residue pairs belonged to the N-terminal, S6/7 β-turn, loop L2, loop L3, and hydrophobic core residues. At physiological temperatures, apo-p53 exhibits exposure of its hydrophobic core, where the temperature-sensitive hotspot residues were also located. This exposure was the result of the S6/7 β-turn and N-terminal moving apart. In the DNA-bound p53 system, loop L1 attains an open conformation at physiological temperatures, which weakens the DNA binding. It is already known that p53 mutants that lack DNA binding also tend to show similar conformational variations. The S6/7 β-turn along with the already known functionally important loop L2 may pose as regions to be targeted to overcome the loss in binding of temperature-sensitive wildtype p53. Rescue strategies directed toward these temperature-sensitive regions may be useful to recuperate its strong binding at physiological temperatures.
Collapse
Affiliation(s)
- Shruti Koulgi
- High Performance Computing - Medical and Bioinformatics Applications Group, Centre for Development for Advanced Computing (C-DAC), Panchawati, Pashan, Pune 411 008, India
| | - Archana Achalere
- High Performance Computing - Medical and Bioinformatics Applications Group, Centre for Development for Advanced Computing (C-DAC), Panchawati, Pashan, Pune 411 008, India
| | - Uddhavesh Sonavane
- High Performance Computing - Medical and Bioinformatics Applications Group, Centre for Development for Advanced Computing (C-DAC), Panchawati, Pashan, Pune 411 008, India
| | - Rajendra Joshi
- High Performance Computing - Medical and Bioinformatics Applications Group, Centre for Development for Advanced Computing (C-DAC), Panchawati, Pashan, Pune 411 008, India
| |
Collapse
|
6
|
Optogenetic technologies in translational cancer research. Biotechnol Adv 2022; 60:108005. [PMID: 35690273 DOI: 10.1016/j.biotechadv.2022.108005] [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: 01/17/2022] [Revised: 05/07/2022] [Accepted: 06/04/2022] [Indexed: 11/23/2022]
Abstract
Gene and cell therapies are widely recognized as future cancer therapeutics but poor controllability limits their clinical applications. Optogenetics, the use of light-controlled proteins to precisely spatiotemporally regulate the activity of genes and cells, opens up new possibilities for cancer treatment. Light of specific wavelength can activate the immune response, oncolytic activity and modulate cell signaling in tumor cells non-invasively, in dosed manner, with tissue confined action and without side effects of conventional therapies. Here, we review optogenetic approaches in cancer research, their clinical potential and challenges of incorporating optogenetics in cancer therapy. We critically discuss beneficial combinations of optogenetic technologies with therapeutic nanobodies, T-cell activation and CAR-T cell approaches, genome editors and oncolytic viruses. We consider viral vectors and nanoparticles for delivering optogenetic payloads and activating light to tumors. Finally, we highlight herein the prospects for integrating optogenetics into immunotherapy as a novel, fast, reversible and safe approach to cancer treatment.
Collapse
|
7
|
Cancer-related Mutations with Local or Long-range Effects on an Allosteric Loop of p53. J Mol Biol 2022; 434:167663. [PMID: 35659507 DOI: 10.1016/j.jmb.2022.167663] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/19/2022] [Accepted: 05/25/2022] [Indexed: 12/31/2022]
Abstract
The tumor protein 53 (p53) is involved in transcription-dependent and independent processes. Several p53 variants related to cancer have been found to impact protein stability. Other variants, on the contrary, might have little impact on structural stability and have local or long-range effects on the p53 interactome. Our group previously identified a loop in the DNA binding domain (DBD) of p53 (residues 207-213) which can recruit different interactors. Experimental structures of p53 in complex with other proteins strengthen the importance of this interface for protein-protein interactions. We here characterized with structure-based approaches somatic and germline variants of p53 which could have a marginal effect in terms of stability and act locally or allosterically on the region 207-213 with consequences on the cytosolic functions of this protein. To this goal, we studied 1132 variants in the p53 DBD with structure-based approaches, accounting also for protein dynamics. We focused on variants predicted with marginal effects on structural stability. We then investigated each of these variants for their impact on DNA binding, dimerization of the p53 DBD, and intramolecular contacts with the 207-213 region. Furthermore, we identified variants that could modulate long-range the conformation of the region 207-213 using a coarse-grain model for allostery and all-atom molecular dynamics simulations. Our predictions have been further validated using enhanced sampling methods for 15 variants. The methodologies used in this study could be more broadly applied to other p53 variants or cases where conformational changes of loop regions are essential in the function of disease-related proteins.
Collapse
|
8
|
Protease-controlled secretion and display of intercellular signals. Nat Commun 2022; 13:912. [PMID: 35177637 PMCID: PMC8854555 DOI: 10.1038/s41467-022-28623-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/03/2022] [Indexed: 02/07/2023] Open
Abstract
To program intercellular communication for biomedicine, it is crucial to regulate the secretion and surface display of signaling proteins. If such regulations are at the protein level, there are additional advantages, including compact delivery and direct interactions with endogenous signaling pathways. Here we create a modular, generalizable design called Retained Endoplasmic Cleavable Secretion (RELEASE), with engineered proteins retained in the endoplasmic reticulum and displayed/secreted in response to specific proteases. The design allows functional regulation of multiple synthetic and natural proteins by synthetic protease circuits to realize diverse signal processing capabilities, including logic operation and threshold tuning. By linking RELEASE to additional sensing and processing circuits, we can achieve elevated protein secretion in response to "undruggable" oncogene KRAS mutants. RELEASE should enable the local, programmable delivery of intercellular cues for a broad variety of fields such as neurobiology, cancer immunotherapy and cell transplantation.
Collapse
|
9
|
D'Agostino S, Mazzega E, Praček K, Piccinin S, Pivetta F, Armellin M, Fortuna S, Maestro R, de Marco A. Interference of p53:Twist1 interaction through competing nanobodies. Int J Biol Macromol 2022; 194:24-31. [PMID: 34863830 DOI: 10.1016/j.ijbiomac.2021.11.160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 10/25/2021] [Accepted: 11/22/2021] [Indexed: 12/16/2022]
Abstract
Twist1 promote the bypass of p53 response by interacting with p53 and facilitating its MDM2-mediated degradation. We reasoned that reagents able to interfere with the p53:Twist1 complex might alleviate Twist1 inhibitory effect over p53, thus representing potential therapeutic tools in p53 wild type tumors. From a pre-immune library of llama nanobodies (VHH), we isolated binders targeting the p53 C-terminal region (p53-CTD) involved in the interaction with Twist1 by using recombinant Twist1 as an epitope-specific competitor during elution. Positive hits were validated by proving their capacity to immunoprecipitate p53 and to inhibit Twist1:p53 binding in vitro. Molecular modeling confirmed a preferential docking of positive hits with p53-CTD. D11 VHH activity was validated in human cell models, succeeded in immunoprecipitating endogenous p53 and, similarly to Twist1 knock-down, interfered with p53 turnover, p53 phosphorylation at Serine 392 and affected cell viability. Despite the limited functional effect determined by D11 expression in target cells, our results provide the proof of principle that nanobodies ectopically expressed within a cell, have the capacity to target the assembly of the pro-tumorigenic Twist1:p53 complex. These results disclose novel tools for dissecting p53 biology and lay down the grounds for the development of innovative targeted therapeutic approaches.
Collapse
Affiliation(s)
- Serena D'Agostino
- Unit of Oncogenetics and Functional Oncogenomics, Centro di Riferimento Oncologico di Aviano (CRO Aviano) IRCCS, National Cancer Institute, Via Gallini 2, 33081 Aviano, PN, Italy
| | - Elisa Mazzega
- Lab of Environmental and Life Sciences, University of Nova Gorica, Vipavska cesta 13, 5000 Rožna Dolina, Nova Gorica, Slovenia
| | - Katja Praček
- Lab of Environmental and Life Sciences, University of Nova Gorica, Vipavska cesta 13, 5000 Rožna Dolina, Nova Gorica, Slovenia; Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via L. Giorgieri 1, 34127 Trieste, Italy
| | - Sara Piccinin
- Unit of Oncogenetics and Functional Oncogenomics, Centro di Riferimento Oncologico di Aviano (CRO Aviano) IRCCS, National Cancer Institute, Via Gallini 2, 33081 Aviano, PN, Italy
| | - Flavia Pivetta
- Unit of Oncogenetics and Functional Oncogenomics, Centro di Riferimento Oncologico di Aviano (CRO Aviano) IRCCS, National Cancer Institute, Via Gallini 2, 33081 Aviano, PN, Italy
| | - Michela Armellin
- Unit of Oncogenetics and Functional Oncogenomics, Centro di Riferimento Oncologico di Aviano (CRO Aviano) IRCCS, National Cancer Institute, Via Gallini 2, 33081 Aviano, PN, Italy
| | - Sara Fortuna
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via L. Giorgieri 1, 34127 Trieste, Italy
| | - Roberta Maestro
- Unit of Oncogenetics and Functional Oncogenomics, Centro di Riferimento Oncologico di Aviano (CRO Aviano) IRCCS, National Cancer Institute, Via Gallini 2, 33081 Aviano, PN, Italy
| | - Ario de Marco
- Lab of Environmental and Life Sciences, University of Nova Gorica, Vipavska cesta 13, 5000 Rožna Dolina, Nova Gorica, Slovenia.
| |
Collapse
|
10
|
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
|
11
|
Papaleo E. Investigating Conformational Dynamics and Allostery in the p53 DNA-Binding Domain Using Molecular Simulations. Methods Mol Biol 2021; 2253:221-244. [PMID: 33315226 DOI: 10.1007/978-1-0716-1154-8_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The p53 tumor suppressor is a multifaceted context-dependent protein, which is involved in multiple cellular pathways, with the ability to either keep the cells alive or to kill them through mechanisms such as apoptosis. To complicate this picture, cancer cells that express mutant p53 becomes addicted to the mutant activity, so that the mutant variant features a myriad of gain-of-function activities, opening different venues for therapy. This makes essential to think outside the box and apply new approaches to the study of p53 structure-(mis)function relationship to find new critical components of its pathway or to understand how known parts are interconnected, compete, or cooperate. In this context, I will here illustrate how to integrate different computational methods to the identification of possible allosteric effects transmitted from the DNA binding interface of p53 to regions for cofactor recruitment. The protocol can be extended to any other cases of study. Indeed, it does not necessarily apply only to the study of DNA-induced effects, but more broadly to the investigation of long-range effects induced by a biological partner that binds to a biomolecule of interest.
Collapse
Affiliation(s)
- Elena Papaleo
- Computational Biology Laboratory, Danish Cancer Society Research Center, Copenhagen, Denmark.
| |
Collapse
|
12
|
Moradi A, Pourseif MM, Jafari B, Parvizpour S, Omidi Y. Nanobody-based therapeutics against colorectal cancer: Precision therapies based on the personal mutanome profile and tumor neoantigens. Pharmacol Res 2020; 156:104790. [DOI: 10.1016/j.phrs.2020.104790] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/07/2020] [Accepted: 03/31/2020] [Indexed: 12/19/2022]
|
13
|
Gangarapu NR, Ranganatham A, Reddy EK, Yellappa S, Chandrasekhar KB. 2-Aminoaryl-3,5-diaryl pyrazines: Synthesis, biological evaluation against Mycobacterium tuberculosis and docking studies. Arch Pharm (Weinheim) 2020; 353:e1900368. [PMID: 32399980 DOI: 10.1002/ardp.201900368] [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: 12/14/2019] [Revised: 04/16/2020] [Accepted: 04/22/2020] [Indexed: 11/10/2022]
Abstract
Rationally designed Mycobacterium tuberculosis (Mtb) inhibitors were synthesized under Buchwald conditions using Pd2 (dba)3 /xantphos and the compounds were investigated for their biological activity against the Mtb standard strain H37Rv and two other clinically isolated multidrug-resistant strains with different drug resistance patterns. Compounds 5e, 6e, 7e, and 8e exhibited excellent antituberculosis activity against H37Rv with a minimum inhibitory concentration (MIC) value of 15 μg/ml. Compounds 5a, 6c, 7b, 8a, 8b, and 8d also displayed their potency with a MIC value in the range of 15-25 μg/ml. In addition to the Mtb studies, compounds 4e, 5e, 7e, and 8e were tested for cytotoxicity on HEK-293 cells and compounds 7e and 8e were identified to have low toxicities of up to 200 and 300 μM, respectively. The synthesized compounds docked with the 2FUM protein of Mtb and the docking studies revealed that compounds 5e, 6e, 7e, and 8e can bind strongly in the active site of the enzyme and showed binding energies of -9.62, -10.7, -11.48, and -12.06 kcal/mol, respectively. Compound 7e forms four hydrogen bonds, whereas compound 8e forms five hydrogen bonds with amino acids, respectively. Based on these results, compounds 7e and 8e might be considered potential lead compounds with good anti-Mtb potency.
Collapse
Affiliation(s)
- Nagaraja Reddy Gangarapu
- Department of Chemistry, Bangalore University, Bengaluru, Karnataka, India.,Department of Chemistry, Jawaharlal Nehru Technological University, Ananthapuramu, Andhra Pradesh, India
| | - Archakam Ranganatham
- Laboratory Division, National Tuberculosis Institute, Bangalore, Karnataka, India
| | - Eeda Koti Reddy
- Department of Science and Humanities, Division of Chemistry, Vignan's Foundation for Science, Technology and Research-VFSTR (Deemed to be University), Guntur, Andhra Pradesh, India
| | - Shivaraj Yellappa
- Department of Chemistry, Bangalore University, Bengaluru, Karnataka, India
| | | |
Collapse
|
14
|
Optogenetic activation of intracellular antibodies for direct modulation of endogenous proteins. Nat Methods 2019; 16:1095-1100. [PMID: 31611691 DOI: 10.1038/s41592-019-0592-7] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 09/06/2019] [Indexed: 01/13/2023]
Abstract
Intracellular antibodies have become powerful tools for imaging, modulating and neutralizing endogenous target proteins. Here, we describe an optogenetically activated intracellular antibody (optobody) consisting of split antibody fragments and blue-light inducible heterodimerization domains. We expanded this optobody platform by generating several optobodies from previously developed intracellular antibodies, and demonstrated that photoactivation of gelsolin and β2-adrenergic receptor (β2AR) optobodies suppressed endogenous gelsolin activity and β2AR signaling, respectively.
Collapse
|
15
|
Steels A, Vannevel L, Zwaenepoel O, Gettemans J. Nb-induced stabilisation of p53 in HPV-infected cells. Sci Rep 2019; 9:12680. [PMID: 31481667 PMCID: PMC6722090 DOI: 10.1038/s41598-019-49061-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 08/13/2019] [Indexed: 11/23/2022] Open
Abstract
Cervical cancer is caused by a persistent infection of the mucosal epithelia with high-risk human papilloma viruses (HPVs). The viral oncoprotein E6 is responsible for the inactivation of the tumour suppressor p53 and thus plays a crucial role in HPV-induced tumorigenesis. The viral E6 protein forms a trimeric complex with the endogenous E3 ubiquitine ligase E6AP and the DNA-binding domain (DBD) of p53, which results in the polyubiquitination and proteasomal degradation of p53. We have developed nanobodies (Nbs) against the DBD of p53, which substantially stabilise p53 in HeLa cells. The observed effect is specific for HPV-infected cells, since similar effects were not seen for U2OS cells. Despite the fact that the stabilised p53 was strongly nuclear enriched, its tumour suppressive functions were hampered. We argue that the absence of a tumour suppressive effect is caused by inhibition of p53 transactivation in both HPV-infected and HPV-negative cells. The inactivation of the transcriptional activity of p53 was associated with an increased cellular proliferation and viability of HeLa cells. In conclusion, we demonstrate that p53 DBD Nbs positively affect protein stability whilst adversely affecting protein function, attesting to their ability to modulate protein properties in a very subtle manner.
Collapse
Affiliation(s)
- Anneleen Steels
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Campus Rommelaere, A. Baertsoenkaai 3, Ghent University, Ghent, Belgium
| | - Laura Vannevel
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Campus Rommelaere, A. Baertsoenkaai 3, Ghent University, Ghent, Belgium
| | - Olivier Zwaenepoel
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Campus Rommelaere, A. Baertsoenkaai 3, Ghent University, Ghent, Belgium
| | - Jan Gettemans
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Campus Rommelaere, A. Baertsoenkaai 3, Ghent University, Ghent, Belgium.
| |
Collapse
|
16
|
Almuttaqi H, Udalova IA. Advances and challenges in targeting IRF5, a key regulator of inflammation. FEBS J 2018; 286:1624-1637. [PMID: 30199605 PMCID: PMC6563445 DOI: 10.1111/febs.14654] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 07/26/2018] [Accepted: 09/07/2018] [Indexed: 12/14/2022]
Abstract
Interferon regulatory factor 5 (IRF5) belongs to a family of transcription factors, originally implicated in antiviral responses and interferon production. However, studies conducted in different laboratories over the last decade have placed IRF5 as a central regulator of the inflammatory response. It has become clear that IRF5 contributes to the pathogenesis of many inflammatory and autoimmune diseases, such as rheumatoid arthritis, inflammatory bowel disease and systemic lupus erythematosus. Given the role of IRF5 in physiology and disease, IRF5 represents a potential therapeutic target. However, despite a significant interest from the pharmaceutical industry, inhibitors that interfere with the IRF5 pathway remain elusive. Here, we review the advances made by various studies in targeting multiple steps of signalling leading to IRF5 activation with their therapeutic potential, and the possible complications of such strategies are discussed.
Collapse
|
17
|
Steels A, Verhelle A, Zwaenepoel O, Gettemans J. Intracellular displacement of p53 using transactivation domain (p53 TAD) specific nanobodies. MAbs 2018; 10:1045-1059. [PMID: 30111239 DOI: 10.1080/19420862.2018.1502025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The tumor suppressor p53 is of crucial importance in the prevention of cellular transformation. In the presence of cellular stress signals, the negative feedback loop between p53 and Mdm2, its main negative regulator, is disrupted, which results in the activation and stabilization of p53. Via a complex interplay between both transcription-dependent and - independent functions of p53, the cell will go through transient cell cycle arrest, cellular senescence or apoptosis. However, it remains difficult to completely fathom the mechanisms behind p53 regulation and its responses, considering the presence of multiple layers involved in fine-tuning them. In order to take the next step forward, novel research tools are urgently needed. We have developed single-domain antibodies, also known as nanobodies, that specifically bind with the N-terminal transactivation domain of wild type p53, but that leave the function of p53 as a transcriptional transactivator intact. When the nanobodies are equipped with a mitochondrial-outer-membrane (MOM)-tag, we can capture p53 at the mitochondria. This nanobody-induced mitochondrial delocalization of p53 is, in specific cases, associated with a decrease in cell viability and with morphological changes in the mitochondria. These findings underpin the potential of nanobodies as bona fide research tools to explore protein function and to unravel their biochemical pathways.
Collapse
Affiliation(s)
- Anneleen Steels
- a Department of Biochemistry, Faculty of Medicine and Health Sciences , Ghent University , Ghent , Belgium
| | - Adriaan Verhelle
- a Department of Biochemistry, Faculty of Medicine and Health Sciences , Ghent University , Ghent , Belgium
| | - Olivier Zwaenepoel
- a Department of Biochemistry, Faculty of Medicine and Health Sciences , Ghent University , Ghent , Belgium
| | - Jan Gettemans
- a Department of Biochemistry, Faculty of Medicine and Health Sciences , Ghent University , Ghent , Belgium
| |
Collapse
|
18
|
Yang-Hartwich Y, Tedja R, Roberts CM, Goodner-Bingham J, Cardenas C, Gurea M, Sumi NJ, Alvero AB, Glackin CA, Mor G. p53-Pirh2 Complex Promotes Twist1 Degradation and Inhibits EMT. Mol Cancer Res 2018; 17:153-164. [PMID: 30131448 DOI: 10.1158/1541-7786.mcr-18-0238] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 06/29/2018] [Accepted: 08/03/2018] [Indexed: 12/13/2022]
Abstract
Epithelial-mesenchymal transition (EMT) is a critical process involved in cancer metastasis and chemoresistance. Twist1 is a key EMT-inducing transcription factor, which is upregulated in multiple types of cancers and has been shown to promote tumor cell invasiveness and support tumor progression. Conversely, p53 is a tumor suppressor gene that is frequently mutated in cancers. This study demonstrates the ability of wild-type (WT) p53 to promote the degradation of Twist1 protein. By forming a complex with Twist1 and the E3 ligase Pirh2, WT p53 promotes the ubiquitination and proteasomal degradation of Twist1, thus inhibiting EMT and maintaining the epithelial phenotype. The ability of p53 to induce Twist1 degradation is abrogated when p53 is mutated. Consequently, the loss of p53-induced Twist1 degradation leads to EMT and the acquisition of a more invasive cancer phenotype.Implication: These data provide new insight into the metastatic process at the molecular level and suggest a signaling pathway that can potentially be used to develop new prognostic markers and therapeutic targets to curtail cancer progression.
Collapse
Affiliation(s)
- Yang Yang-Hartwich
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut
| | - Roslyn Tedja
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut
| | - Cai M Roberts
- Department of Stem Cell and Developmental Biology, City of Hope Beckman Research Institute, Duarte, California
| | - Jamie Goodner-Bingham
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut
| | - Carlos Cardenas
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut
| | - Marta Gurea
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut
| | - Natalia J Sumi
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut
| | - Ayesha B Alvero
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut
| | - Carlotta A Glackin
- Department of Stem Cell and Developmental Biology, City of Hope Beckman Research Institute, Duarte, California
| | - Gil Mor
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut.
| |
Collapse
|
19
|
Salema V, Fernández LÁ. Escherichia coli surface display for the selection of nanobodies. Microb Biotechnol 2017; 10:1468-1484. [PMID: 28772027 PMCID: PMC5658595 DOI: 10.1111/1751-7915.12819] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 07/12/2017] [Accepted: 07/13/2017] [Indexed: 12/29/2022] Open
Abstract
Nanobodies (Nbs) are the smallest functional antibody fragments known in nature and have multiple applications in biomedicine or environmental monitoring. Nbs are derived from the variable segment of camelid heavy chain-only antibodies, known as VHH. For selection, libraries of VHH gene segments from naïve, immunized animals or of synthetic origin have been traditionally cloned in E. coli phage display or yeast display systems, and clones binding the target antigen recovered, usually from plastic surfaces with the immobilized antigen (phage display) or using fluorescence-activated cell sorting (FACS; yeast display). This review briefly describes these conventional approaches and focuses on the distinct properties of an E. coli display system developed in our laboratory, which combines the benefits of both phage display and yeast display systems. We demonstrate that E. coli display using an N-terminal domain of intimin is an effective platform for the surface display of VHH libraries enabling selection of high-affinity Nbs by magnetic cell sorting and direct selection on live mammalian cells displaying the target antigen on their surface. Flow cytometry analysis of E. coli bacteria displaying the Nbs on their surface allows monitoring of the selection process, facilitates screening, characterization of antigen-binding clones, specificity, ligand competition and estimation of the equilibrium dissociation constant (KD ).
Collapse
Affiliation(s)
- Valencio Salema
- Department of Microbial BiotechnologyCentro Nacional de Biotecnología (CNB)Consejo Superior de Investigaciones Científicas (CSIC)MadridSpain
| | - Luis Ángel Fernández
- Department of Microbial BiotechnologyCentro Nacional de Biotecnología (CNB)Consejo Superior de Investigaciones Científicas (CSIC)MadridSpain
| |
Collapse
|
20
|
Beghein E, Gettemans J. Nanobody Technology: A Versatile Toolkit for Microscopic Imaging, Protein-Protein Interaction Analysis, and Protein Function Exploration. Front Immunol 2017; 8:771. [PMID: 28725224 PMCID: PMC5495861 DOI: 10.3389/fimmu.2017.00771] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 06/16/2017] [Indexed: 01/05/2023] Open
Abstract
Over the last two decades, nanobodies or single-domain antibodies have found their way in research, diagnostics, and therapy. These antigen-binding fragments, derived from Camelid heavy chain only antibodies, possess remarkable characteristics that favor their use over conventional antibodies or fragments thereof, in selected areas of research. In this review, we assess the current status of nanobodies as research tools in diverse aspects of fundamental research. We discuss the use of nanobodies as detection reagents in fluorescence microscopy and focus on recent advances in super-resolution microscopy. Second, application of nanobody technology in investigating protein–protein interactions is reviewed, with emphasis on possible uses in mass spectrometry. Finally, we discuss the potential value of nanobodies in studying protein function, and we focus on their recently reported application in targeted protein degradation. Throughout the review, we highlight state-of-the-art engineering strategies that could expand nanobody versatility and we suggest future applications of the technology in the selected areas of fundamental research.
Collapse
Affiliation(s)
- Els Beghein
- Nanobody Laboratory, Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Jan Gettemans
- Nanobody Laboratory, Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
21
|
Bertier L, Boucherie C, Zwaenepoel O, Vanloo B, Van Troys M, Van Audenhove I, Gettemans J. Inhibitory cortactin nanobodies delineate the role of NTA- and SH3-domain-specific functions during invadopodium formation and cancer cell invasion. FASEB J 2017; 31:2460-2476. [PMID: 28235780 DOI: 10.1096/fj.201600810rr] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 02/07/2017] [Indexed: 01/08/2023]
Abstract
Cancer cells exploit different strategies to escape from the primary tumor, gain access to the circulation, disseminate throughout the body, and form metastases, the leading cause of death by cancer. Invadopodia, proteolytically active plasma membrane extensions, are essential in this escape mechanism. Cortactin is involved in every phase of invadopodia formation, and its overexpression is associated with increased invadopodia formation, extracellular matrix degradation, and cancer cell invasion. To analyze endogenous cortactin domain function in these processes, we characterized the effects of nanobodies that are specific for the N-terminal acidic domain of cortactin and expected to target small epitopes within this domain. These nanobodies inhibit cortactin-mediated actin-related protein (Arp)2/3 activation, and, after their intracellular expression in cancer cells, decrease invadopodia formation, extracellular matrix degradation, and cancer cell invasion. In addition, one of the nanobodies affects Arp2/3 interaction and invadopodium stability, and a nanobody targeting the Src homology 3 domain of cortactin enabled comparison of 2 functional regions in invadopodium formation or stability. Given their common and distinct effects, we validate cortactin nanobodies as an instrument to selectively block and study distinct domains within a protein with unprecedented precision, aiding rational future generation of protein domain-selective therapeutic compounds.-Bertier, L., Boucherie, C., Zwaenepoel, O., Vanloo, B., Van Troys, M., Van Audenhove, I., Gettemans, J. Inhibitory cortactin nanobodies delineate the role of NTA- and SH3-domain-specific functions during invadopodium formation and cancer cell invasion.
Collapse
Affiliation(s)
- Laurence Bertier
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University-Campus Rommelaere, Ghent, Belgium
| | - Ciska Boucherie
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University-Campus Rommelaere, Ghent, Belgium
| | - Olivier Zwaenepoel
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University-Campus Rommelaere, Ghent, Belgium
| | - Berlinda Vanloo
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University-Campus Rommelaere, Ghent, Belgium
| | - Marleen Van Troys
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University-Campus Rommelaere, Ghent, Belgium
| | - Isabel Van Audenhove
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University-Campus Rommelaere, Ghent, Belgium
| | - Jan Gettemans
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University-Campus Rommelaere, Ghent, Belgium
| |
Collapse
|
22
|
Lambrughi M, De Gioia L, Gervasio FL, Lindorff-Larsen K, Nussinov R, Urani C, Bruschi M, Papaleo E. DNA-binding protects p53 from interactions with cofactors involved in transcription-independent functions. Nucleic Acids Res 2016; 44:9096-9109. [PMID: 27604871 PMCID: PMC5100575 DOI: 10.1093/nar/gkw770] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 08/19/2016] [Accepted: 08/23/2016] [Indexed: 12/15/2022] Open
Abstract
Binding-induced conformational changes of a protein at regions distant from the binding site may play crucial roles in protein function and regulation. The p53 tumour suppressor is an example of such an allosterically regulated protein. Little is known, however, about how DNA binding can affect distal sites for transcription factors. Furthermore, the molecular details of how a local perturbation is transmitted through a protein structure are generally elusive and occur on timescales hard to explore by simulations. Thus, we employed state-of-the-art enhanced sampling atomistic simulations to unveil DNA-induced effects on p53 structure and dynamics that modulate the recruitment of cofactors and the impact of phosphorylation at Ser215. We show that DNA interaction promotes a conformational change in a region 3 nm away from the DNA binding site. Specifically, binding to DNA increases the population of an occluded minor state at this distal site by more than 4-fold, whereas phosphorylation traps the protein in its major state. In the minor conformation, the interface of p53 that binds biological partners related to p53 transcription-independent functions is not accessible. Significantly, our study reveals a mechanism of DNA-mediated protection of p53 from interactions with partners involved in the p53 transcription-independent signalling. This also suggests that conformational dynamics is tightly related to p53 signalling.
Collapse
Affiliation(s)
- Matteo Lambrughi
- Computational Biology Laboratory, Unit of Statistics, Bioinformatics and Registry, Strandboulevarden 49, 2100, Copenhagen, Denmark
- Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Luca De Gioia
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milan, Italy
| | - Francesco Luigi Gervasio
- Department of Chemistry and Institute of Structural and Molecular Biology, University College London, London WC1H 0AJ, UK
| | - Kresten Lindorff-Larsen
- Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Ruth Nussinov
- Cancer and Inflammation Program, Leidos Biomedical Research Inc., Frederick National laboratory, National Cancer Institute, Frederick, MD 21702, USA
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Chiara Urani
- Department of Earth and Environmental Sciences, University of Milano-Bicocca, Piazza della Scienza 1, 20126, Milan, Italy
| | - Maurizio Bruschi
- Department of Earth and Environmental Sciences, University of Milano-Bicocca, Piazza della Scienza 1, 20126, Milan, Italy
| | - Elena Papaleo
- Computational Biology Laboratory, Unit of Statistics, Bioinformatics and Registry, Strandboulevarden 49, 2100, Copenhagen, Denmark
- Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
23
|
Beghein E, Van Audenhove I, Zwaenepoel O, Verhelle A, De Ganck A, Gettemans J. A new survivin tracer tracks, delocalizes and captures endogenous survivin at different subcellular locations and in distinct organelles. Sci Rep 2016; 6:31177. [PMID: 27514728 PMCID: PMC4981888 DOI: 10.1038/srep31177] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 07/13/2016] [Indexed: 01/18/2023] Open
Abstract
Survivin, the smallest member of the inhibitor of apoptosis protein family, plays a central role during mitosis and exerts a cytoprotective function. Survivin is highly expressed in most cancer types and contributes to multiple facets of carcinogenesis. The molecular mechanisms underlying its highly diverse functions need to be extensively explored, which is crucial for rational design of future personalized therapeutics. In this study, we have generated an alpaca survivin nanobody (SVVNb8) that binds with low nanomolar affinity to its target. When expressed as an intrabody in HeLa cells, SVVNb8 faithfully tracks survivin during different phases of mitosis without interfering with survivin function. Furthermore, coupling SVVNb8 with a subcellular delocalization tag efficiently redirects endogenous survivin towards the nucleus, the cytoplasm, peroxisomes and even to the intermembrane space of mitochondria where it presumably interacts with resident mitochondrial survivin. Based on our findings, we believe that SVVNb8 is an excellent instrument to further elucidate survivin biology and topography, and can serve as a model system to investigate mitochondrial and peroxisomal (survivin) protein import.
Collapse
Affiliation(s)
- Els Beghein
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Campus Rommelaere, A. Baertsoenkaai 3, Ghent University, Ghent, Belgium
| | - Isabel Van Audenhove
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Campus Rommelaere, A. Baertsoenkaai 3, Ghent University, Ghent, Belgium
| | - Olivier Zwaenepoel
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Campus Rommelaere, A. Baertsoenkaai 3, Ghent University, Ghent, Belgium
| | - Adriaan Verhelle
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Campus Rommelaere, A. Baertsoenkaai 3, Ghent University, Ghent, Belgium
| | - Ariane De Ganck
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Campus Rommelaere, A. Baertsoenkaai 3, Ghent University, Ghent, Belgium
| | - Jan Gettemans
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Campus Rommelaere, A. Baertsoenkaai 3, Ghent University, Ghent, Belgium
| |
Collapse
|
24
|
Buchfellner A, Yurlova L, Nüske S, Scholz AM, Bogner J, Ruf B, Zolghadr K, Drexler SE, Drexler GA, Girst S, Greubel C, Reindl J, Siebenwirth C, Romer T, Friedl AA, Rothbauer U. A New Nanobody-Based Biosensor to Study Endogenous PARP1 In Vitro and in Live Human Cells. PLoS One 2016; 11:e0151041. [PMID: 26950694 PMCID: PMC4780744 DOI: 10.1371/journal.pone.0151041] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 02/23/2016] [Indexed: 11/22/2022] Open
Abstract
Poly(ADP-ribose) polymerase 1 (PARP1) is a key player in DNA repair, genomic stability and cell survival and it emerges as a highly relevant target for cancer therapies. To deepen our understanding of PARP biology and mechanisms of action of PARP1-targeting anti-cancer compounds, we generated a novel PARP1-affinity reagent, active both in vitro and in live cells. This PARP1-biosensor is based on a PARP1-specific single-domain antibody fragment (~ 15 kDa), termed nanobody, which recognizes the N-terminus of human PARP1 with nanomolar affinity. In proteomic approaches, immobilized PARP1 nanobody facilitates quantitative immunoprecipitation of functional, endogenous PARP1 from cellular lysates. For cellular studies, we engineered an intracellularly functional PARP1 chromobody by combining the nanobody coding sequence with a fluorescent protein sequence. By following the chromobody signal, we were for the first time able to monitor the recruitment of endogenous PARP1 to DNA damage sites in live cells. Moreover, tracing of the sub-nuclear translocation of the chromobody signal upon treatment of human cells with chemical substances enables real-time profiling of active compounds in high content imaging. Due to its ability to perform as a biosensor at the endogenous level of the PARP1 enzyme, the novel PARP1 nanobody is a unique and versatile tool for basic and applied studies of PARP1 biology and DNA repair.
Collapse
Affiliation(s)
| | | | - Stefan Nüske
- Livestock Center of the Faculty of Veterinary Medicine, Ludwig Maximilians University, Munich, Germany
| | - Armin M. Scholz
- Livestock Center of the Faculty of Veterinary Medicine, Ludwig Maximilians University, Munich, Germany
| | | | - Benjamin Ruf
- ChromoTek GmbH, IZB, Planegg, Martinsried, Germany
| | | | - Sophie E. Drexler
- Department of Radiation Oncology, Ludwig Maximilians University, Munich, Germany
| | - Guido A. Drexler
- Department of Radiation Oncology, Ludwig Maximilians University, Munich, Germany
| | - Stefanie Girst
- Institute for Applied Physics and Metrology, Bundeswehr University Munich, Neubiberg, Germany
| | - Christoph Greubel
- Institute for Applied Physics and Metrology, Bundeswehr University Munich, Neubiberg, Germany
| | - Judith Reindl
- Institute for Applied Physics and Metrology, Bundeswehr University Munich, Neubiberg, Germany
| | - Christian Siebenwirth
- Department of Radiation Oncology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Tina Romer
- ChromoTek GmbH, IZB, Planegg, Martinsried, Germany
- * E-mail:
| | - Anna A. Friedl
- Department of Radiation Oncology, Ludwig Maximilians University, Munich, Germany
- Clinical Cooperation Group ‘Personalized Radiotherapy of Head and Neck Cancer’, Helmholtz Center Munich, Neuherberg, Germany
| | - Ulrich Rothbauer
- Natural and Medical Institute at the University of Tuebingen, Reutlingen, Germany
- Pharmaceutical Biotechnology, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
25
|
Van Audenhove I, Denert M, Boucherie C, Pieters L, Cornelissen M, Gettemans J. Fascin Rigidity and L-plastin Flexibility Cooperate in Cancer Cell Invadopodia and Filopodia. J Biol Chem 2016; 291:9148-60. [PMID: 26945069 DOI: 10.1074/jbc.m115.706937] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Indexed: 01/15/2023] Open
Abstract
Invadopodia and filopodia are dynamic, actin-based protrusions contributing to cancer cell migration, invasion, and metastasis. The force of actin bundles is essential for their protrusive activity. The bundling protein fascin is known to play a role in both invadopodia and filopodia. As it is more and more acknowledged that functionally related proteins cooperate, it is unlikely that only fascin bundles actin in these protrusions. Another interesting candidate is L-plastin, normally expressed in hematopoietic cells, but considered a common marker of many cancer types. We identified L-plastin as a new component of invadopodia, where it contributes to degradation and invasiveness. By means of specific, high-affinity nanobodies inhibiting bundling of fascin or L-plastin, we further unraveled their cooperative mode of action. We show that the bundlers cannot compensate for each other due to strikingly different bundling characteristics: L-plastin bundles are much thinner and less tightly packed. Composite bundles adopt an intermediate phenotype, with fascin delivering the rigidity and strength for protrusive force and structural stability, whereas L-plastin accounts for the flexibility needed for elongation. Consistent with this, elevated L-plastin expression promotes elongation and reduces protrusion density in cells with relatively lower L-plastin than fascin levels.
Collapse
Affiliation(s)
| | | | | | - Leen Pieters
- Basic Medical Science, Faculty of Medicine and Health Sciences, Ghent University, B-9000 Ghent, Belgium
| | - Maria Cornelissen
- Basic Medical Science, Faculty of Medicine and Health Sciences, Ghent University, B-9000 Ghent, Belgium
| | | |
Collapse
|
26
|
Bieli D, Alborelli I, Harmansa S, Matsuda S, Caussinus E, Affolter M. Development and Application of Functionalized Protein Binders in Multicellular Organisms. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 325:181-213. [DOI: 10.1016/bs.ircmb.2016.02.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
|
27
|
Desmyter A, Spinelli S, Roussel A, Cambillau C. Camelid nanobodies: killing two birds with one stone. Curr Opin Struct Biol 2015; 32:1-8. [PMID: 25614146 DOI: 10.1016/j.sbi.2015.01.001] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 12/23/2014] [Accepted: 01/05/2015] [Indexed: 01/09/2023]
Abstract
In recent years, the use of single-domain camelid immunoglobulins, termed vHHs or nanobodies, has seen increasing growth in biotechnology, pharmaceutical applications and structure/function research. The usefulness of nanobodies in structural biology is now firmly established, as they provide access to new epitopes in concave and hinge regions - and stabilize them. These sites are often associated with enzyme inhibition or receptor neutralization, and, at the same time, provide favorable surfaces for crystal packing. Remarkable results have been achieved by using nanobodies with flexible multi-domain proteins, large complexes and, last but not least, membrane proteins. While generating nanobodies is still a rather long and expensive procedure, the advent of naive libraries might be expected to facilitate the whole process.
Collapse
Affiliation(s)
- Aline Desmyter
- Aix-Marseille Université, Architecture et Fonction des Macromolécules Biologiques, France; Centre National de la Recherche Scientifique, AFMB, UMR 7257, case 932, 13288 Marseille Cedex 09, France
| | - Silvia Spinelli
- Aix-Marseille Université, Architecture et Fonction des Macromolécules Biologiques, France; Centre National de la Recherche Scientifique, AFMB, UMR 7257, case 932, 13288 Marseille Cedex 09, France
| | - Alain Roussel
- Aix-Marseille Université, Architecture et Fonction des Macromolécules Biologiques, France; Centre National de la Recherche Scientifique, AFMB, UMR 7257, case 932, 13288 Marseille Cedex 09, France
| | - Christian Cambillau
- Aix-Marseille Université, Architecture et Fonction des Macromolécules Biologiques, France; Centre National de la Recherche Scientifique, AFMB, UMR 7257, case 932, 13288 Marseille Cedex 09, France.
| |
Collapse
|