1
|
Sharrocks KL, Swaih AM, Hanyaloglu AC. Single-molecule localization microscopy as a tool to quantify di/oligomerization of receptor tyrosine kinases and G protein-coupled receptors. Mol Pharmacol 2025; 107:100033. [PMID: 40228395 DOI: 10.1016/j.molpha.2025.100033] [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: 12/19/2024] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 04/16/2025] Open
Abstract
Dimerization and oligomerization of membrane receptors, including G protein-coupled receptors and receptor tyrosine kinases, are fundamental for regulating cell signaling and diversifying downstream responses to mediate a range of physiological processes. Receptor di/oligomers play roles in diverse facets of receptor function. Changes in receptor di/oligomers have been implicated in a range of diseases; therefore, better understanding of the specific composition and interactions between receptors in complexes is essential, especially for the development of di/oligomer-specific drugs. Previously, different optical microscopy approaches and proximity-based biophysical assays have been used to demonstrate di/oligomerization of membrane receptors. However, in recent years, single-molecule super-resolution microscopy techniques have allowed researchers to quantify and uncover the precise dynamics and stoichiometry of specific receptor complexes. This allows the organization of membrane protein receptors to be mapped across the plasma membrane to explore the effects of factors such as ligands, effectors, membrane environment, and therapeutic agents. Quantification of receptor complexes is required to better understand the intricate balance of distinct receptor complexes in cells. In this brief review, we provide an overview of single-molecule approaches for the quantification of receptor di/oligomerization. We will discuss the techniques commonly employed to study membrane receptor di/oligomerization and their relative advantages and limitations. SIGNIFICANCE STATEMENT: Receptor di/oligomerization plays an important role in their function. For some receptors, di/oligomerization is essential for functional signaling, whereas for others, it acts as a mechanism to achieve signaling pleiotropy. Aberrant receptor di/oligomerization has been implicated in a wide range of diseases. Single-molecule super-resolution microscopy techniques provide convincing methods to precisely quantify receptor complexes at the plasma membrane. Understanding receptor complex organization in disease models can also influence the targeting of specific monomeric or oligomeric complexes in therapeutic strategies.
Collapse
Affiliation(s)
| | | | - Aylin C Hanyaloglu
- The Francis Crick Institute, London, UK; Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| |
Collapse
|
2
|
Li M, Gao J, Qi X, Li B, Zhao Y, Liu X, Zhang G, Wang H, Tong T. Super-Resolution Fluorescence Imaging Reveals the Mechanism of NRP1 Clustering on Non-Small-Cell Lung Cancer Membranes. Anal Chem 2025; 97:2326-2334. [PMID: 39841049 DOI: 10.1021/acs.analchem.4c05675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
Neuropilin 1 (NRP1) is upregulated in various types of malignant tumors, especially non-small-cell lung cancer (NSCLC). However, the precise mechanisms for membrane localization and regulation are not fully understood. Observations from super-resolution microscopy have revealed that NRP1 tends to form nanoscale clusters on the cell membrane, with these clusters varying significantly in size and density across different regions. Further research has shown that stimulation by hepatocyte growth factor (HGF) can reorganize the distribution of NRP1, reducing the number of small clusters while promoting the formation of larger ones. This suggests a propensity for internalization after activation. Additionally, dual-color dSTORM imaging has demonstrated a certain degree of colocalization between NRP1 and c-MET, indicating that c-MET plays an important role in stabilizing NRP1 clusters. This study provides new insights into the mechanism behind NRP1's clustered distribution on cell membranes and paves the way for developing more effective therapeutic strategies targeting NRP1 within tumors.
Collapse
Affiliation(s)
- Meng Li
- Department of Thoracic Surgery, The Second Hospital of Jilin University, Changchun, Jilin 130041, P. R. China
| | - Jing Gao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Xiao Qi
- Department of Thoracic Surgery, The Second Hospital of Jilin University, Changchun, Jilin 130041, P. R. China
| | - Baofeng Li
- Department of Thoracic Surgery, The Second Hospital of Jilin University, Changchun, Jilin 130041, P. R. China
| | - Yinghao Zhao
- Department of Thoracic Surgery, The Second Hospital of Jilin University, Changchun, Jilin 130041, P. R. China
| | - Xiangyu Liu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Guangxin Zhang
- Department of Thoracic Surgery, The Second Hospital of Jilin University, Changchun, Jilin 130041, P. R. China
| | - Hongda Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
- University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| | - Ti Tong
- Department of Thoracic Surgery, The Second Hospital of Jilin University, Changchun, Jilin 130041, P. R. China
| |
Collapse
|
3
|
Li Y, Arghittu SM, Dietz MS, Hella GJ, Haße D, Ferraris DM, Freund P, Barth HD, Iamele L, de Jonge H, Niemann HH, Covino R, Heilemann M. Single-molecule imaging and molecular dynamics simulations reveal early activation of the MET receptor in cells. Nat Commun 2024; 15:9486. [PMID: 39488533 PMCID: PMC11531568 DOI: 10.1038/s41467-024-53772-7] [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: 05/08/2024] [Accepted: 10/22/2024] [Indexed: 11/04/2024] Open
Abstract
Embedding of cell-surface receptors into a membrane defines their dynamics but also complicates experimental characterization of their signaling complexes. The hepatocyte growth factor receptor MET is a receptor tyrosine kinase involved in cellular processes such as proliferation, migration, and survival. It is also targeted by the pathogen Listeria monocytogenes, whose invasion protein, internalin B (InlB), binds to MET, forming a signaling dimer that triggers pathogen internalization. Here we use an integrative structural biology approach, combining molecular dynamics simulations and single-molecule Förster resonance energy transfer (smFRET) in cells, to investigate the early stages of MET activation. Our simulations show that InlB binding stabilizes MET in a conformation that promotes dimer formation. smFRET reveals that the in situ dimer structure closely resembles one of two previously published crystal structures, though with key differences. This study refines our understanding of MET activation and provides a methodological framework for studying other plasma membrane receptors.
Collapse
Affiliation(s)
- Yunqing Li
- Institute of Physical and Theoretical Chemistry, Goethe-University Frankfurt, Max-von-Laue-Str. 7, Frankfurt am Main, Germany
| | - Serena M Arghittu
- Frankfurt Institute for Advanced Studies, Ruth-Moufang-Str. 1, Frankfurt am Main, Germany
- IMPRS on Cellular Biophysics, Max-von-Laue-Str. 3, Frankfurt am Main, Germany
| | - Marina S Dietz
- Institute of Physical and Theoretical Chemistry, Goethe-University Frankfurt, Max-von-Laue-Str. 7, Frankfurt am Main, Germany
| | - Gabriel J Hella
- Frankfurt Institute for Advanced Studies, Ruth-Moufang-Str. 1, Frankfurt am Main, Germany
| | - Daniel Haße
- Department of Chemistry, Bielefeld University, Universitaetsstr. 25, Bielefeld, Germany
| | - Davide M Ferraris
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2, Novara, Italy
| | - Petra Freund
- Institute of Physical and Theoretical Chemistry, Goethe-University Frankfurt, Max-von-Laue-Str. 7, Frankfurt am Main, Germany
| | - Hans-Dieter Barth
- Institute of Physical and Theoretical Chemistry, Goethe-University Frankfurt, Max-von-Laue-Str. 7, Frankfurt am Main, Germany
| | - Luisa Iamele
- Department of Molecular Medicine, University of Pavia, Immunology and General Pathology Section, Via Ferrata 9, Pavia, Italy
| | - Hugo de Jonge
- Department of Molecular Medicine, University of Pavia, Immunology and General Pathology Section, Via Ferrata 9, Pavia, Italy
| | - Hartmut H Niemann
- Department of Chemistry, Bielefeld University, Universitaetsstr. 25, Bielefeld, Germany
| | - Roberto Covino
- Frankfurt Institute for Advanced Studies, Ruth-Moufang-Str. 1, Frankfurt am Main, Germany.
- IMPRS on Cellular Biophysics, Max-von-Laue-Str. 3, Frankfurt am Main, Germany.
- Institute of Computer Science, Goethe-University Frankfurt, Robert-Mayer-Str. 11-15, Frankfurt am Main, Germany.
| | - Mike Heilemann
- Institute of Physical and Theoretical Chemistry, Goethe-University Frankfurt, Max-von-Laue-Str. 7, Frankfurt am Main, Germany.
- IMPRS on Cellular Biophysics, Max-von-Laue-Str. 3, Frankfurt am Main, Germany.
| |
Collapse
|
4
|
Kessler L, Balakrishnan A, Menche T, Wang D, Li Y, Mantel M, Glogger M, Dietz MS, Heilemann M. Self-Quenched Fluorophore-DNA Labels for Super-Resolution Fluorescence Microscopy. J Phys Chem B 2024; 128:6751-6759. [PMID: 38955346 PMCID: PMC11264260 DOI: 10.1021/acs.jpcb.4c02065] [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] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 07/04/2024]
Abstract
Protein labeling through transient and repetitive hybridization of short, fluorophore-labeled DNA oligonucleotides has become widely applied in various optical super-resolution microscopy methods. The main advantages are multitarget imaging and molecular quantification. A challenge is the high background signal originating from the presence of unbound fluorophore-DNA labels in solution. Here, we report the self-quenching of fluorophore dimers conjugated to DNA oligonucleotides as a general concept to reduce the fluorescence background. Upon hybridization, the fluorescence signals of both fluorophores are restored. We expand the toolbox of fluorophores suitable for self-quenching and report their spectra and hybridization equilibria. We apply self-quenched fluorophore-DNA labels to stimulated emission depletion microscopy and single-molecule localization microscopy and report improved imaging performances.
Collapse
Affiliation(s)
- Laurell
F. Kessler
- Institute
of Physical and Theoretical Chemistry, Goethe-University
Frankfurt, Max-von-Laue-Str. 7, Frankfurt 60438, Germany
| | - Ashwin Balakrishnan
- Institute
of Physical and Theoretical Chemistry, Goethe-University
Frankfurt, Max-von-Laue-Str. 7, Frankfurt 60438, Germany
| | - Tanja Menche
- Institute
of Physical and Theoretical Chemistry, Goethe-University
Frankfurt, Max-von-Laue-Str. 7, Frankfurt 60438, Germany
| | - Dongni Wang
- Institute
of Physical and Theoretical Chemistry, Goethe-University
Frankfurt, Max-von-Laue-Str. 7, Frankfurt 60438, Germany
| | - Yunqing Li
- Institute
of Physical and Theoretical Chemistry, Goethe-University
Frankfurt, Max-von-Laue-Str. 7, Frankfurt 60438, Germany
| | - Maximilian Mantel
- Institute
of Physical and Theoretical Chemistry, Goethe-University
Frankfurt, Max-von-Laue-Str. 7, Frankfurt 60438, Germany
| | - Marius Glogger
- Optical
Imaging Competence Centre, Universität
Erlangen-Nürnberg, Cauerstraße 3, Erlangen 91058, Germany
| | - Marina S. Dietz
- Institute
of Physical and Theoretical Chemistry, Goethe-University
Frankfurt, Max-von-Laue-Str. 7, Frankfurt 60438, Germany
| | - Mike Heilemann
- Institute
of Physical and Theoretical Chemistry, Goethe-University
Frankfurt, Max-von-Laue-Str. 7, Frankfurt 60438, Germany
| |
Collapse
|
5
|
Zou Q, Zhang Q, Du B, Wang H, Yang X, Wang Q, Wang K. Atomic Force Microscopy Lifetime Analysis: An Intuitive Method for Evaluating Receptor Tyrosine Kinase Dimer-Targeting Inhibitors. Anal Chem 2024; 96:10962-10968. [PMID: 38925633 DOI: 10.1021/acs.analchem.4c01353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
Overexpression of receptor tyrosine kinases (RTKs) or binding to ligands can lead to the formation of specific unliganded and liganded RTK dimers, and these two RTK dimers are potential targets for preventing tumor metastasis. Traditional RTK dimer inhibitor analysis was mostly based on end point assays, which required cumbersome cell handling and behavior monitoring. There are still challenges in developing intuitive process-based analytical methods to study RTK dimer inhibitors, especially those used to visually distinguish between unliganded and liganded RTK dimer inhibitors. Herein, taking the mesenchymal-epithelial transition factor (MET) receptor, an intuitive method for evaluating MET inhibitors has been developed based on atomic force microscopy (AFM) lifetime analysis. The time interval between the start of the force and the bond break point was regarded as the bond lifetime, which could reflect the stability of the MET dimer. The results showed that there was a significant difference in the lifetime (τ) of unliganded MET dimers (τ1 = 207.87 ± 4.69 ms) and liganded MET dimers (τ2 = 330.58 ± 15.60 ms) induced by the hepatocyte growth factor, and aptamer SL1 could decrease τ1 and τ2, suggesting that SL1 could inhibit both unliganded and liganded MET dimers. However, heparin only decreased τ2, suggesting that it could inhibit only the liganded MET dimer. AFM-based lifetime analysis methods could monitor RTK dimer status rather than provide overall average results, allowing for intuitive process-based analysis and evaluation of RTK dimers and related inhibitors at the single-molecule level. This study provides a novel complementary strategy for simple and intuitive RTK inhibitor research.
Collapse
Affiliation(s)
- Qingqing Zou
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha 410082, P. R. China
| | - Qianqian Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha 410082, P. R. China
| | - Bin Du
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha 410082, P. R. China
| | - Hongqiang Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha 410082, P. R. China
| | - Xiaohai Yang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha 410082, P. R. China
| | - Qing Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha 410082, P. R. China
| | - Kemin Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha 410082, P. R. China
| |
Collapse
|
6
|
Abouelkheir M, Roy T, Krzyscik MA, Özdemir E, Hristova K. Investigations of membrane protein interactions in cells using fluorescence microscopy. Curr Opin Struct Biol 2024; 86:102816. [PMID: 38648680 PMCID: PMC11141325 DOI: 10.1016/j.sbi.2024.102816] [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: 10/30/2023] [Revised: 03/06/2024] [Accepted: 03/26/2024] [Indexed: 04/25/2024]
Abstract
The interactions between proteins in membranes govern many cellular functions. Our ability to probe for such interactions has greatly evolved in recent years due to the introduction of new fluorescence techniques. As a result, we currently have a choice of methods that can be used to assess the spatial distribution of a membrane protein, its association state, and the thermodynamic stability of the oligomers in the native milieu. These biophysical measurements have revealed new insights into important biological processes in cellular membranes.
Collapse
Affiliation(s)
- Mahmoud Abouelkheir
- Department of Materials Science and Engineering and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore MD 21218, USA; Chemistry-Biology Interface Program, Johns Hopkins University, Baltimore MD 21218, USA
| | - Tanaya Roy
- Department of Materials Science and Engineering and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore MD 21218, USA
| | - Mateusz A Krzyscik
- Department of Materials Science and Engineering and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore MD 21218, USA
| | - Ece Özdemir
- Department of Materials Science and Engineering and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore MD 21218, USA
| | - Kalina Hristova
- Department of Materials Science and Engineering and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore MD 21218, USA; Chemistry-Biology Interface Program, Johns Hopkins University, Baltimore MD 21218, USA.
| |
Collapse
|
7
|
Chen H, Yan G, Wen MH, Brooks KN, Zhang Y, Huang PS, Chen TY. Advancements and Practical Considerations for Biophysical Research: Navigating the Challenges and Future of Super-resolution Microscopy. CHEMICAL & BIOMEDICAL IMAGING 2024; 2:331-344. [PMID: 38817319 PMCID: PMC11134610 DOI: 10.1021/cbmi.4c00019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 04/06/2024] [Accepted: 04/10/2024] [Indexed: 06/01/2024]
Abstract
The introduction of super-resolution microscopy (SRM) has significantly advanced our understanding of cellular and molecular dynamics, offering a detailed view previously beyond our reach. Implementing SRM in biophysical research, however, presents numerous challenges. This review addresses the crucial aspects of utilizing SRM effectively, from selecting appropriate fluorophores and preparing samples to analyzing complex data sets. We explore recent technological advancements and methodological improvements that enhance the capabilities of SRM. Emphasizing the integration of SRM with other analytical methods, we aim to overcome inherent limitations and expand the scope of biological insights achievable. By providing a comprehensive guide for choosing the most suitable SRM methods based on specific research objectives, we aim to empower researchers to explore complex biological processes with enhanced precision and clarity, thereby advancing the frontiers of biophysical research.
Collapse
Affiliation(s)
- Huanhuan Chen
- Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Guangjie Yan
- Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Meng-Hsuan Wen
- Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Kameron N. Brooks
- Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Yuteng Zhang
- Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Pei-San Huang
- Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Tai-Yen Chen
- Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| |
Collapse
|
8
|
Tannoo RM, Richert L, Koschut D, Tomishige N, Treffert SM, Kobayashi T, Mély Y, Orian-Rousseau V. Quantitative live imaging reveals a direct interaction between CD44v6 and MET in membrane domains upon activation with both MET ligands, HGF and internalin B. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2024; 1866:184236. [PMID: 37793560 DOI: 10.1016/j.bbamem.2023.184236] [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: 01/23/2023] [Revised: 09/24/2023] [Accepted: 09/27/2023] [Indexed: 10/06/2023]
Abstract
Deregulation of the receptor tyrosine kinase MET/hepatocyte growth factor (HGF) pathway results in several pathological processes involved in tumor progression and metastasis. In a different context, MET can serve as an entry point for the bacterium Listeria monocytogenes, when activated by the internalin B (InlB) protein during infection of non-phagocytic cells. We have previously demonstrated that MET requires CD44v6 for its ligand-induced activation. However, the stoichiometry and the steps required for the formation of this complex, are still unknown. In this work, we studied the dynamics of the ligand-induced interaction of CD44v6 with MET at the plasma membrane. Using Förster resonance energy transfer-based fluorescence lifetime imaging microscopy in T-47D cells, we evidenced a direct interaction between MET and CD44v6 promoted by HGF and InlB in live cells. In the absence of MET, fluorescence correlation spectroscopy experiments further showed the dimerization of CD44v6 and the increase of its diffusion induced by HGF and InlB. In the presence of MET, stimulation of the cells by HGF or InlB significantly decreased the diffusion of CD44v6, in line with the formation of a ternary complex of MET with CD44v6 and HGF/InlB. Finally, similarly to HGF/InlB, disruption of liquid-ordered domains (Lo) by methyl-β-cyclodextrin increased CD44v6 mobility suggesting that these factors induce the exit of CD44v6 from the Lo domains. Our data led us to propose a model for MET activation, where CD44v6 dimerizes and diffuses rapidly out of Lo domains to form an oligomeric MET/ligand/CD44v6 complex that is instrumental for MET activation.
Collapse
Affiliation(s)
- Ryshtee Mary Tannoo
- Laboratory of Bioimaging and Pathologies (LBP), University of Strasbourg (UNISTRA), France; Institute of Biological and Chemical systems-Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), Germany
| | - Ludovic Richert
- Laboratory of Bioimaging and Pathologies (LBP), University of Strasbourg (UNISTRA), France.
| | - David Koschut
- Institute of Biological and Chemical systems-Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), Germany; Disease Intervention Technology Lab (DITL), Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology, and Research (A*STAR), Singapore
| | - Nario Tomishige
- Laboratory of Bioimaging and Pathologies (LBP), University of Strasbourg (UNISTRA), France
| | - Sven Máté Treffert
- Institute of Biological and Chemical systems-Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), Germany
| | - Toshihide Kobayashi
- Laboratory of Bioimaging and Pathologies (LBP), University of Strasbourg (UNISTRA), France
| | - Yves Mély
- Laboratory of Bioimaging and Pathologies (LBP), University of Strasbourg (UNISTRA), France.
| | - Véronique Orian-Rousseau
- Institute of Biological and Chemical systems-Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), Germany.
| |
Collapse
|
9
|
Zalejski J, Sun J, Sharma A. Unravelling the Mystery inside Cells by Using Single-Molecule Fluorescence Imaging. J Imaging 2023; 9:192. [PMID: 37754956 PMCID: PMC10532472 DOI: 10.3390/jimaging9090192] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/01/2023] [Accepted: 09/14/2023] [Indexed: 09/28/2023] Open
Abstract
Live-cell imaging is a powerful technique to study the dynamics and mechanics of various biological molecules like proteins, organelles, DNA, and RNA. With the rapid evolution of optical microscopy, our understanding of how these molecules are implicated in the cells' most critical physiological roles deepens. In this review, we focus on how spatiotemporal nanoscale live-cell imaging at the single molecule level allows for profound contributions towards new discoveries in life science. This review will start by summarizing how single-molecule tracking has been used to analyze membrane dynamics, receptor-ligand interactions, protein-protein interactions, inner- and extra-cellular transport, gene expression/transcription, and whole organelle tracking. We then move on to how current authors are trying to improve single-molecule tracking and overcome current limitations by offering new ways of labeling proteins of interest, multi-channel/color detection, improvements in time-lapse imaging, and new methods and programs to analyze the colocalization and movement of targets. We later discuss how single-molecule tracking can be a beneficial tool used for medical diagnosis. Finally, we wrap up with the limitations and future perspectives of single-molecule tracking and total internal reflection microscopy.
Collapse
Affiliation(s)
| | | | - Ashutosh Sharma
- Department of Chemistry, University of Illinois Chicago, Chicago, IL 60607, USA; (J.Z.); (J.S.)
| |
Collapse
|
10
|
Linzer J, Phelps Z, Vummidi S, Lee BYE, Coant N, Haley JD. Mass Spectrometry and Pharmacological Approaches to Measuring Cooption and Reciprocal Activation of Receptor Tyrosine Kinases. Proteomes 2023; 11:20. [PMID: 37368466 PMCID: PMC10304582 DOI: 10.3390/proteomes11020020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/11/2023] [Accepted: 05/26/2023] [Indexed: 06/28/2023] Open
Abstract
Receptor tyrosine kinases (RTKs) can show extensive crosstalk, directly and indirectly. Elucidating RTK crosstalk remains an important goal in the clinical combination of anti-cancer therapies. Here, we present mass spectrometry and pharmacological approaches showing the hepatocyte growth factor receptor (MET)-promoting tyrosine phosphorylation of the epidermal growth factor receptor (EGFR) and other membrane receptors in MET-amplified H1993 NSCLC cells. Conversely, in H292 wt-EGFR NSCLC cells, EGFR promotes the tyrosine phosphorylation of MET. Reciprocal regulation of the EGFR and insulin receptor (IR) was observed in the GEO CRC cells, where inhibition of the EGFR drives tyrosine phosphorylation of the insulin receptor. Similarly, in platelet-derived growth factor receptor (PDGFR)-amplified H1703 NSCLC cells, inhibition of the EGFR promotes the tyrosine phosphorylation of the PDGFR. These RTK interactions are used to illustrate basic principles applicable to other RTK signaling networks. More specifically, we focus on two types of RTK interaction: (1) co-option of one RTK by another and (2) reciprocal activation of one receptor following the inhibition of a distinct receptor.
Collapse
Affiliation(s)
| | | | | | | | | | - John D. Haley
- Department of Pathology and Cancer Center, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
11
|
Stoneman MR, Raicu V. Fluorescence-Based Detection of Proteins and Their Interactions in Live Cells. J Phys Chem B 2023. [PMID: 37205844 DOI: 10.1021/acs.jpcb.3c01419] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Recent advances in fluorescence-based microscopy techniques, such as single molecule fluorescence, Förster resonance energy transfer (FRET), fluorescence intensity fluctuations analysis, and super-resolution microscopy have expanded our ability to study proteins in greater detail within their native cellular environment and to investigate the roles that protein interactions play in biological functions, such as inter- and intracellular signaling and cargo transport. In this Perspective, we provide an up-to-date overview of the current state of the art in fluorescence-based detection of proteins and their interactions in living cells with an emphasis on recent developments that have facilitated the characterization of the spatial and temporal organization of proteins into oligomeric complexes in the presence and absence of natural and artificial ligands. Further advancements in this field will only deepen our understanding of the underlying mechanisms of biological processes and help develop new therapeutic targets.
Collapse
Affiliation(s)
- Michael R Stoneman
- Department of Physics, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, United States
| | - Valerică Raicu
- Department of Physics, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, United States
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, United States
| |
Collapse
|
12
|
Sobhanan J, Anas A, Biju V. Nanomaterials for Fluorescence and Multimodal Bioimaging. CHEM REC 2023; 23:e202200253. [PMID: 36789795 DOI: 10.1002/tcr.202200253] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/27/2023] [Indexed: 02/16/2023]
Abstract
Bioconjugated nanomaterials replace molecular probes in bioanalysis and bioimaging in vitro and in vivo. Nanoparticles of silica, metals, semiconductors, polymers, and supramolecular systems, conjugated with contrast agents and drugs for image-guided (MRI, fluorescence, PET, Raman, SPECT, photodynamic, photothermal, and photoacoustic) therapy infiltrate into preclinical and clinical settings. Small bioactive molecules like peptides, proteins, or DNA conjugated to the surfaces of drugs or probes help us to interface them with cells and tissues. Nevertheless, the toxicity and pharmacokinetics of nanodrugs, nanoprobes, and their components become the clinical barriers, underscoring the significance of developing biocompatible next-generation drugs and contrast agents. This account provides state-of-the-art advancements in the preparation and biological applications of bioconjugated nanomaterials and their molecular, cell, and in vivo applications. It focuses on the preparation, bioimaging, and bioanalytical applications of monomodal and multimodal nanoprobes composed of quantum dots, quantum clusters, iron oxide nanoparticles, and a few rare earth metal ion complexes.
Collapse
Affiliation(s)
- Jeladhara Sobhanan
- Graduate School of Environmental Science, Hokkaido University, N10 W5, Sapporo, Hokkaido, 060-0810, Japan.,Department of Chemistry, Rice University, Houston, Texas 77005, United States
| | - Abdulaziz Anas
- CSIR-National Institute of Oceanography, Regional Centre Kochi, Kerala, 682 018, India
| | - Vasudevanpillai Biju
- Graduate School of Environmental Science, Hokkaido University, N10 W5, Sapporo, Hokkaido, 060-0810, Japan.,Research Institute for Electronic Science, Hokkaido University, Sapporo, 001-0020, Japan
| |
Collapse
|
13
|
Verzelli P, Nold A, Sun C, Heilemann M, Schuman EM, Tchumatchenko T. Unbiased choice of global clustering parameters for single-molecule localization microscopy. Sci Rep 2022. [PMID: 36581654 DOI: 10.1101/2021.02.22.432198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023] Open
Abstract
Single-molecule localization microscopy resolves objects below the diffraction limit of light via sparse, stochastic detection of target molecules. Single molecules appear as clustered detection events after image reconstruction. However, identification of clusters of localizations is often complicated by the spatial proximity of target molecules and by background noise. Clustering results of existing algorithms often depend on user-generated training data or user-selected parameters, which can lead to unintentional clustering errors. Here we suggest an unbiased algorithm (FINDER) based on adaptive global parameter selection and demonstrate that the algorithm is robust to noise inclusion and target molecule density. We benchmarked FINDER against the most common density based clustering algorithms in test scenarios based on experimental datasets. We show that FINDER can keep the number of false positive inclusions low while also maintaining a low number of false negative detections in densely populated regions.
Collapse
Affiliation(s)
- Pietro Verzelli
- Institute of Experimental Epileptology and Cognition Research, University of Bonn Medical Center, Bonn, Germany
| | - Andreas Nold
- Institute of Experimental Epileptology and Cognition Research, University of Bonn Medical Center, Bonn, Germany
- Theory of Neural Dynamics Group, Max Planck Institute for Brain Research, Frankfurt, Germany
| | - Chao Sun
- Department of Synaptic Plasticity, Max Planck Institute for Brain Research, Frankfurt, Germany
| | - Mike Heilemann
- Institute of Physical and Theoretical Chemistry, Goethe-University Frankfurt, Frankfurt, Germany
| | - Erin M Schuman
- Department of Synaptic Plasticity, Max Planck Institute for Brain Research, Frankfurt, Germany
| | - Tatjana Tchumatchenko
- Institute of Experimental Epileptology and Cognition Research, University of Bonn Medical Center, Bonn, Germany.
- Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.
- Theory of Neural Dynamics Group, Max Planck Institute for Brain Research, Frankfurt, Germany.
| |
Collapse
|
14
|
Verzelli P, Nold A, Sun C, Heilemann M, Schuman EM, Tchumatchenko T. Unbiased choice of global clustering parameters for single-molecule localization microscopy. Sci Rep 2022; 12:22561. [PMID: 36581654 PMCID: PMC9800574 DOI: 10.1038/s41598-022-27074-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022] Open
Abstract
Single-molecule localization microscopy resolves objects below the diffraction limit of light via sparse, stochastic detection of target molecules. Single molecules appear as clustered detection events after image reconstruction. However, identification of clusters of localizations is often complicated by the spatial proximity of target molecules and by background noise. Clustering results of existing algorithms often depend on user-generated training data or user-selected parameters, which can lead to unintentional clustering errors. Here we suggest an unbiased algorithm (FINDER) based on adaptive global parameter selection and demonstrate that the algorithm is robust to noise inclusion and target molecule density. We benchmarked FINDER against the most common density based clustering algorithms in test scenarios based on experimental datasets. We show that FINDER can keep the number of false positive inclusions low while also maintaining a low number of false negative detections in densely populated regions.
Collapse
Affiliation(s)
- Pietro Verzelli
- Institute of Experimental Epileptology and Cognition Research, University of Bonn Medical Center, Bonn, Germany
| | - Andreas Nold
- Institute of Experimental Epileptology and Cognition Research, University of Bonn Medical Center, Bonn, Germany
- Theory of Neural Dynamics Group, Max Planck Institute for Brain Research, Frankfurt, Germany
| | - Chao Sun
- Department of Synaptic Plasticity, Max Planck Institute for Brain Research, Frankfurt, Germany
| | - Mike Heilemann
- Institute of Physical and Theoretical Chemistry, Goethe-University Frankfurt, Frankfurt, Germany
| | - Erin M Schuman
- Department of Synaptic Plasticity, Max Planck Institute for Brain Research, Frankfurt, Germany
| | - Tatjana Tchumatchenko
- Institute of Experimental Epileptology and Cognition Research, University of Bonn Medical Center, Bonn, Germany.
- Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.
- Theory of Neural Dynamics Group, Max Planck Institute for Brain Research, Frankfurt, Germany.
| |
Collapse
|
15
|
Li X, Sun B, Zhu J, Qian M, Chen Y. Construction of a Mass-Tagged Oligo Probe Set for Revealing Protein Ratiometric Relationship Associated with EGFR-HER2 Heterodimerization in Living Cells. Anal Chem 2022; 94:8838-8846. [PMID: 35709389 DOI: 10.1021/acs.analchem.1c04989] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Protein dimerization, as the most common form of protein-protein interaction, can manifest more significant roles in cellular signaling than individual monomers. For example, excessive formation of EGFR-HER2 dimer has been implicated in cancer development and therapeutic resistance in addition to the overexpression of EGFR and HER2 proteins. Thus, quantitative evaluation of these heterodimers in living cells and revelation of their ratiometric relationship with protein monomers in dimerization may provide insights into clinical cancer management. To achieve this goal, the prerequisite is protein heterodimer quantification. Given the current lack of quantitative methods, we constructed a mass-tagged oligo nanoprobe set for quantification of EGFR-HER2 dimer in living cells. The mass-tagged oligo nanoprobe set contained two targeting probes (nucleic acid aptamers), a connector probe, a hairpin probe, and a photocleavable mass-tagged probe. Two distinct aptamers can recognize target protein monomers and initiate the subsequent hybridization cascade involving binding to the connector probe, formation of an initiator strand, opening of a hairpin probe, and ensuing hybridization with a photocleavable mass-tagged probe. Ultimately, the mass tag was released under ultraviolet light and then subjected to mass spectrometric analysis. In this way, the information regarding the interaction between two protein monomers was successfully converted to the quantitative signal of the mass tag. Using the assay, the expression level of EGFR-HER2 dimer and its relationship with individual protein monomers were determined in four breast cancer cell lines. We are among the first to obtain the absolute level of protein heterodimer, and this quantitative information may be vital in understanding the molecular basis of cancer.
Collapse
Affiliation(s)
- Xiaoxu Li
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Bo Sun
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China.,Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang 222002, China
| | - Jianhua Zhu
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Moting Qian
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yun Chen
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China.,State Key Laboratory of Reproductive Medicine, Nanjing 210029, China.,Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Nanjing 211166, China
| |
Collapse
|
16
|
Wollman AJM, Fournier C, Llorente-Garcia I, Harriman O, Payne-Dwyer AL, Shashkova S, Zhou P, Liu TC, Ouaret D, Wilding J, Kusumi A, Bodmer W, Leake MC. Critical roles for EGFR and EGFR-HER2 clusters in EGF binding of SW620 human carcinoma cells. J R Soc Interface 2022; 19:20220088. [PMID: 35612280 PMCID: PMC9131850 DOI: 10.1098/rsif.2022.0088] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Epidermal growth factor (EGF) signalling regulates normal epithelial and other cell growth, with EGF receptor (EGFR) overexpression reported in many cancers. However, the role of EGFR clusters in cancer and their dependence on EGF binding is unclear. We present novel single-molecule total internal reflection fluorescence microscopy of (i) EGF and EGFR in living cancer cells, (ii) the action of anti-cancer drugs that separately target EGFR and human EGFR2 (HER2) on these cells and (iii) EGFR–HER2 interactions. We selected human epithelial SW620 carcinoma cells for their low level of native EGFR expression, for stable transfection with fluorescent protein labelled EGFR, and imaged these using single-molecule localization microscopy to quantify receptor architectures and dynamics upon EGF binding. Prior to EGF binding, we observe pre-formed EGFR clusters. Unexpectedly, clusters likely contain both EGFR and HER2, consistent with co-diffusion of EGFR and HER2 observed in a different model CHO-K1 cell line, whose stoichiometry increases following EGF binding. We observe a mean EGFR : EGF stoichiometry of approximately 4 : 1 for plasma membrane-colocalized EGFR–EGF that we can explain using novel time-dependent kinetics modelling, indicating preferential ligand binding to monomers. Our results may inform future cancer drug developments.
Collapse
Affiliation(s)
- Adam J M Wollman
- Department of Physics, University of York, York, UK.,Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Charlotte Fournier
- Department of Physics, Clarendon Laboratory, University of Oxford, Oxford OX1 3PU, UK.,Science and Technology Group, Okinawa Institute of Science and Technology Graduate University (OIST), 1919 Tancha, Onna-son, Okinawa 904-0495, Japan
| | | | - Oliver Harriman
- Department of Physics, Clarendon Laboratory, University of Oxford, Oxford OX1 3PU, UK
| | | | | | - Peng Zhou
- Membrane Cooperativity Unit, OIST, 1919 Tancha, Onna-son, Okinawa 904-0495, Japan
| | - Ta-Chun Liu
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | - Djamila Ouaret
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | - Jenny Wilding
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | - Akihiro Kusumi
- Membrane Cooperativity Unit, OIST, 1919 Tancha, Onna-son, Okinawa 904-0495, Japan
| | - Walter Bodmer
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | - Mark C Leake
- Department of Physics, University of York, York, UK.,Department of Biology, University of York, York, UK.,Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| |
Collapse
|
17
|
Malkusch S, Rahm JV, Dietz MS, Heilemann M, Sibarita JB, Lötsch J. Receptor tyrosine kinase MET ligand-interaction classified via machine learning from single-particle tracking data. Mol Biol Cell 2022; 33:ar60. [PMID: 35171646 PMCID: PMC9265154 DOI: 10.1091/mbc.e21-10-0496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/09/2022] [Accepted: 02/11/2022] [Indexed: 11/11/2022] Open
Abstract
Internalin B-mediated activation of the membrane-bound receptor tyrosine kinase MET is accompanied by a change in receptor mobility. Conversely, it should be possible to infer from receptor mobility whether a cell has been treated with internalin B. Here, we propose a method based on hidden Markov modeling and explainable artificial intelligence that machine-learns the key differences in MET mobility between internalin B-treated and -untreated cells from single-particle tracking data. Our method assigns receptor mobility to three diffusion modes (immobile, slow, and fast). It discriminates between internalin B-treated and -untreated cells with a balanced accuracy of >99% and identifies three parameters that are most affected by internalin B treatment: a decrease in the mobility of slow molecules (1) and a depopulation of the fast mode (2) caused by an increased transition of fast molecules to the slow mode (3). Our approach is based entirely on free software and is readily applicable to the analysis of other membrane receptors.
Collapse
Affiliation(s)
- Sebastian Malkusch
- Institute of Clinical Pharmacology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Johanna V. Rahm
- Institute of Physical and Theoretical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 7, 60438 Frankfurt am Main, Germany
| | - Marina S. Dietz
- Institute of Physical and Theoretical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 7, 60438 Frankfurt am Main, Germany
| | - Mike Heilemann
- Institute of Physical and Theoretical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 7, 60438 Frankfurt am Main, Germany
| | - Jean-Baptiste Sibarita
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000 Bordeaux, France
| | - Jörn Lötsch
- Institute of Clinical Pharmacology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| |
Collapse
|
18
|
MacNeil IA, Khan SA, Sen A, Soltani SM, Burns DJ, Sullivan BF, Laing LG. Functional signaling test identifies HER2 negative breast cancer patients who may benefit from c-Met and pan-HER combination therapy. Cell Commun Signal 2022; 20:4. [PMID: 34998412 PMCID: PMC8742957 DOI: 10.1186/s12964-021-00798-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 11/01/2021] [Indexed: 11/29/2022] Open
Abstract
Background Research is revealing the complex coordination between cell signaling systems as they adapt to genetic and epigenetic changes. Tools to uncover these highly complex functional linkages will play an important role in advancing more efficacious disease treatments. Current tumor cell signal transduction research is identifying coordination between receptor types, receptor families, and transduction pathways to maintain tumor cell viability despite challenging tumor microenvironment conditions. Methods In this report, coactivated abnormal levels of signaling activity for c-Met and HER family receptors in live tumor cells were measured by a new clinical test to identify a subpopulation of breast cancer patients that could be responsive to combined targeted therapies. The CELsignia Multi-Pathway Signaling Function (CELsignia) Test uses an impedance biosensor to quantify an individual patient’s ex vivo live tumor cell signaling response in real-time to specific HER family and c-Met co-stimulation and targeted therapies. Results The test identified breast tumors with hyperactive HER1, HER2, HER3/4, and c-Met coordinated signaling that express otherwise normal amounts of these receptors. The supporting data of the pre-clinical verification of this test included analyses of 79 breast cancer patients’ cell response to HER and c-Met agonists. The signaling results were confirmed using clinically approved matching targeted drugs, and combinations of targeted drugs in addition to correlative mouse xenograft tumor response to HER and c-Met targeted therapies. Conclusions The results of this study demonstrated the potential benefit of a functional test for identifying a subpopulation of breast cancer patients with coordinated abnormal HER and c-Met signaling for a clinical trial testing combination targeted therapy. Video Abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-021-00798-9.
Collapse
Affiliation(s)
- Ian A MacNeil
- Celcuity, Inc., 16305 36th Ave N, Suite 100, Minneapolis, MN, 55446, USA
| | - Salmaan A Khan
- Celcuity, Inc., 16305 36th Ave N, Suite 100, Minneapolis, MN, 55446, USA
| | - Adrish Sen
- Celcuity, Inc., 16305 36th Ave N, Suite 100, Minneapolis, MN, 55446, USA
| | - Sajjad M Soltani
- Celcuity, Inc., 16305 36th Ave N, Suite 100, Minneapolis, MN, 55446, USA
| | - David J Burns
- Celcuity, Inc., 16305 36th Ave N, Suite 100, Minneapolis, MN, 55446, USA
| | - Brian F Sullivan
- Celcuity, Inc., 16305 36th Ave N, Suite 100, Minneapolis, MN, 55446, USA
| | - Lance G Laing
- Celcuity, Inc., 16305 36th Ave N, Suite 100, Minneapolis, MN, 55446, USA.
| |
Collapse
|
19
|
Franco Nitta C, Green EW, Jhamba ED, Keth JM, Ortiz-Caraveo I, Grattan RM, Schodt DJ, Gibson AC, Rajput A, Lidke KA, Wilson BS, Steinkamp MP, Lidke DS. EGFR transactivates RON to drive oncogenic crosstalk. eLife 2021; 10:63678. [PMID: 34821550 PMCID: PMC8654365 DOI: 10.7554/elife.63678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 11/24/2021] [Indexed: 12/22/2022] Open
Abstract
Crosstalk between different receptor tyrosine kinases (RTKs) is thought to drive oncogenic signaling and allow therapeutic escape. EGFR and RON are two such RTKs from different subfamilies, which engage in crosstalk through unknown mechanisms. We combined high-resolution imaging with biochemical and mutational studies to ask how EGFR and RON communicate. EGF stimulation promotes EGFR-dependent phosphorylation of RON, but ligand stimulation of RON does not trigger EGFR phosphorylation – arguing that crosstalk is unidirectional. Nanoscale imaging reveals association of EGFR and RON in common plasma membrane microdomains. Two-color single particle tracking captured formation of complexes between RON and EGF-bound EGFR. Our results further show that RON is a substrate for EGFR kinase, and that transactivation of RON requires formation of a signaling competent EGFR dimer. These results support a role for direct EGFR/RON interactions in propagating crosstalk, such that EGF-stimulated EGFR phosphorylates RON to activate RON-directed signaling.
Collapse
Affiliation(s)
| | - Ellen W Green
- Department of Pathology, University of New Mexico, Albuquerque, United States
| | - Elton D Jhamba
- Department of Pathology, University of New Mexico, Albuquerque, United States
| | - Justine M Keth
- Department of Pathology, University of New Mexico, Albuquerque, United States
| | - Iraís Ortiz-Caraveo
- Department of Pathology, University of New Mexico, Albuquerque, United States
| | - Rachel M Grattan
- Department of Pathology, University of New Mexico, Albuquerque, United States
| | - David J Schodt
- Department of Physics & Astronomy, University of New Mexico, Albuquerque, United States
| | - Aubrey C Gibson
- Department of Pathology, University of New Mexico, Albuquerque, United States
| | - Ashwani Rajput
- Department of Surgery, University of New Mexico, Albuquerque, United States.,Comprehensive Cancer Center, University of New Mexico, Albuquerque, United States
| | - Keith A Lidke
- Department of Physics & Astronomy, University of New Mexico, Albuquerque, United States.,Comprehensive Cancer Center, University of New Mexico, Albuquerque, United States
| | - Bridget S Wilson
- Department of Pathology, University of New Mexico, Albuquerque, United States.,Comprehensive Cancer Center, University of New Mexico, Albuquerque, United States
| | - Mara P Steinkamp
- Department of Pathology, University of New Mexico, Albuquerque, United States.,Comprehensive Cancer Center, University of New Mexico, Albuquerque, United States
| | - Diane S Lidke
- Department of Pathology, University of New Mexico, Albuquerque, United States.,Comprehensive Cancer Center, University of New Mexico, Albuquerque, United States
| |
Collapse
|
20
|
Yan Q, Cai M, Jing Y, Li H, Xu H, Sun J, Gao J, Wang H. Quantitatively mapping the interaction of HER2 and EGFR on cell membranes with peptide probes. NANOSCALE 2021; 13:17629-17637. [PMID: 34664051 DOI: 10.1039/d1nr02684d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Human epidermal growth factor receptor-2 (HER2) is a member of the epidermal growth factor receptor (HER) family that is involved in various biological processes such as cell proliferation, survival, differentiation, migration and invasion. It generally functions in the form of homo-/hetero-dimers or oligomers with other HER family members. Although its essential roles in cellular activities have been widely recognized, questions concerning the spatial distribution of HER2 on the membranes and the interactions between it and other ErbB family members remain obscure. Here, we obtained a high-quality dSTORM image of HER2 nanoscale clusters recognized by peptide probes, and found that HER2 forms clusters containing different numbers of molecules on cell membranes. Moreover, we found that HER2 and EGFR formed hetero-oligomers on non-stimulated cell membranes, whereas EGF stimulation reduced the degree of heteromerization, suggesting that HER2 and EGFR hetero-oligomers may inhibit the activation of EGFR. Collectively, our work revealed the clustered distribution of HER2 and quantified the changes of the interaction between HER2 and EGFR in the resting and active states at the single molecular level, which promotes a deeper understanding of the protein-protein interaction on cell membranes.
Collapse
Affiliation(s)
- Qiuyan Yan
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Mingjun Cai
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China.
| | - Yingying Jing
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China.
| | - Hongru Li
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China.
- University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Haijiao Xu
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China.
| | - Jiayin Sun
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China.
| | - Jing Gao
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China.
| | - Hongda Wang
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China.
- University of Science and Technology of China, Hefei, Anhui 230027, China
- Laboratory for Marine Biology and Biotechnology, Qing Dao National Laboratory for Marine Science and Technology, Wenhai Road, Aoshanwei, Jimo, Qingdao, Shandong, 266237, China
| |
Collapse
|
21
|
Tada S, Ren X, Mao H, Heo Y, Park S, Isoshima T, Zhu L, Zhou X, Ito R, Kurata S, Osaki M, Kobatake E, Ito Y. Versatile Mitogenic and Differentiation-Inducible Layer Formation by Underwater Adhesive Polypeptides. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100961. [PMID: 34174166 PMCID: PMC8373149 DOI: 10.1002/advs.202100961] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/14/2021] [Indexed: 05/06/2023]
Abstract
Artificial materials have no biological functions, but they are important for medical devices such as artificial organs and matrices for regenerative medicine. In this study, mitogenic and differentiation-inducible materials are devised via the simple coating of polypeptides, which contain the sequence of epidermal growth factor or insulin-like growth factor with a key amino acid (3,4-dihydroxyphenylalanine) of underwater adhesive proteins. The adhesive polypeptides prepared via solid-phase synthesis form layers on various substrates involving organic and inorganic materials to provide biological surfaces. Through the direct activation of cognate receptors on interactive surfaces, the materials enable increased cell growth and differentiation compared to that achieved by soluble growth factors. This superior growth and differentiation are attributed to the long-lasting signal transduction (triggered by the bound growth factors), which do not cause receptor internalization and subsequent downregulation.
Collapse
Affiliation(s)
- Seiichi Tada
- Emergent Bioengineering Materials Research TeamRIKEN Center for Emergent Matter Science2‐1 HirosawaWakoSaitama351‐0198Japan
| | - Xueli Ren
- Nano Medical Engineering LaboratoryRIKEN Cluster for Pioneering Research2‐1 HirosawaWakoSaitama351‐0198Japan
| | - Hongli Mao
- Nano Medical Engineering LaboratoryRIKEN Cluster for Pioneering Research2‐1 HirosawaWakoSaitama351‐0198Japan
| | - Yun Heo
- Emergent Bioengineering Materials Research TeamRIKEN Center for Emergent Matter Science2‐1 HirosawaWakoSaitama351‐0198Japan
- Department of Environmental Chemistry and EngineeringInterdisciplinary Graduate School of Science and EngineeringTokyo Institute of TechnologyMidori‐kuYokohama226–8502Japan
| | - Shin‐Hye Park
- Nano Medical Engineering LaboratoryRIKEN Cluster for Pioneering Research2‐1 HirosawaWakoSaitama351‐0198Japan
| | - Takashi Isoshima
- Nano Medical Engineering LaboratoryRIKEN Cluster for Pioneering Research2‐1 HirosawaWakoSaitama351‐0198Japan
| | - Liping Zhu
- Nano Medical Engineering LaboratoryRIKEN Cluster for Pioneering Research2‐1 HirosawaWakoSaitama351‐0198Japan
| | - Xiaoyue Zhou
- Nano Medical Engineering LaboratoryRIKEN Cluster for Pioneering Research2‐1 HirosawaWakoSaitama351‐0198Japan
| | - Reiko Ito
- Support Unit for Bio‐Material AnalysisResearch Resources DivisionRIKEN Center for Brain Science2‐1 HirosawaWakoSaitama351‐0198Japan
| | - Shino Kurata
- Support Unit for Bio‐Material AnalysisResearch Resources DivisionRIKEN Center for Brain Science2‐1 HirosawaWakoSaitama351‐0198Japan
| | - Megumi Osaki
- Support Unit for Bio‐Material AnalysisResearch Resources DivisionRIKEN Center for Brain Science2‐1 HirosawaWakoSaitama351‐0198Japan
| | - Eiry Kobatake
- Department of Environmental Chemistry and EngineeringInterdisciplinary Graduate School of Science and EngineeringTokyo Institute of TechnologyMidori‐kuYokohama226–8502Japan
| | - Yoshihiro Ito
- Emergent Bioengineering Materials Research TeamRIKEN Center for Emergent Matter Science2‐1 HirosawaWakoSaitama351‐0198Japan
- Nano Medical Engineering LaboratoryRIKEN Cluster for Pioneering Research2‐1 HirosawaWakoSaitama351‐0198Japan
| |
Collapse
|
22
|
Narayanasamy KK, Stojic A, Li Y, Sass S, Hesse MR, Deussner-Helfmann NS, Dietz MS, Kuner T, Klevanski M, Heilemann M. Visualizing Synaptic Multi-Protein Patterns of Neuronal Tissue With DNA-Assisted Single-Molecule Localization Microscopy. Front Synaptic Neurosci 2021; 13:671288. [PMID: 34220481 PMCID: PMC8247585 DOI: 10.3389/fnsyn.2021.671288] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/25/2021] [Indexed: 02/04/2023] Open
Abstract
The development of super-resolution microscopy (SRM) has widened our understanding of biomolecular structure and function in biological materials. Imaging multiple targets within a single area would elucidate their spatial localization relative to the cell matrix and neighboring biomolecules, revealing multi-protein macromolecular structures and their functional co-dependencies. SRM methods are, however, limited to the number of suitable fluorophores that can be imaged during a single acquisition as well as the loss of antigens during antibody washing and restaining for organic dye multiplexing. We report the visualization of multiple protein targets within the pre- and postsynapse in 350–400 nm thick neuronal tissue sections using DNA-assisted single-molecule localization microscopy (SMLM). In a single labeling step, antibodies conjugated with short DNA oligonucleotides visualized multiple targets by sequential exchange of fluorophore-labeled complementary oligonucleotides present in the imaging buffer. This approach avoids potential effects on structural integrity when using multiple rounds of immunolabeling and eliminates chromatic aberration, because all targets are imaged using a single excitation laser wavelength. This method proved robust for multi-target imaging in semi-thin tissue sections with a lateral resolution better than 25 nm, paving the way toward structural cell biology with single-molecule SRM.
Collapse
Affiliation(s)
- Kaarjel K Narayanasamy
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany.,Institute of Physical and Theoretical Chemistry, Goethe-University Frankfurt, Frankfurt, Germany
| | - Aleksandar Stojic
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Yunqing Li
- Institute of Physical and Theoretical Chemistry, Goethe-University Frankfurt, Frankfurt, Germany
| | - Steffen Sass
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Marina R Hesse
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Nina S Deussner-Helfmann
- Institute of Physical and Theoretical Chemistry, Goethe-University Frankfurt, Frankfurt, Germany
| | - Marina S Dietz
- Institute of Physical and Theoretical Chemistry, Goethe-University Frankfurt, Frankfurt, Germany
| | - Thomas Kuner
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Maja Klevanski
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Mike Heilemann
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany.,Institute of Physical and Theoretical Chemistry, Goethe-University Frankfurt, Frankfurt, Germany
| |
Collapse
|
23
|
Zhu M, Wang DD, Yan H. Genotype-determined EGFR-RTK heterodimerization and its effects on drug resistance in lung Cancer treatment revealed by molecular dynamics simulations. BMC Mol Cell Biol 2021; 22:34. [PMID: 34112110 PMCID: PMC8191231 DOI: 10.1186/s12860-021-00358-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 03/10/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) and its signaling pathways play a vital role in pathogenesis of lung cancer. By disturbing EGFR signaling, mutations of EGFR may lead to progression of cancer or the emergence of resistance to EGFR-targeted drugs. RESULTS We investigated the correlation between EGFR mutations and EGFR-receptor tyrosine kinase (RTK) crosstalk in the signaling network, in order to uncover the drug resistance mechanism induced by EGFR mutations. For several EGFR wild type (WT) or mutated proteins, we measured the EGFR-RTK interactions using several computational methods based on molecular dynamics (MD) simulations, including geometrical characterization of the interfaces and conventional estimation of free energy of binding. Geometrical properties, namely the matching rate of atomic solid angles in the interfaces and center-of-mass distances between interacting atoms, were extracted relying on Alpha Shape modeling. For a couple of RTK partners (c-Met, ErbB2 and IGF-1R), results have shown a looser EGFR-RTK crosstalk for the drug-sensitive EGFR mutant while a tighter crosstalk for the drug-resistant mutant. It guarantees the genotype-determined EGFR-RTK crosstalk, and further proposes a potential drug resistance mechanism by amplified EGFR-RTK crosstalk induced by EGFR mutations. CONCLUSIONS This study will lead to a deeper understanding of EGFR mutation-induced drug resistance mechanisms and promote the design of innovative drugs.
Collapse
Affiliation(s)
- Mengxu Zhu
- Department of Electrical Engineering, City University of Hong Kong, Kowloon, Hong Kong.
| | - Debby D Wang
- Department of Electrical Engineering, City University of Hong Kong, Kowloon, Hong Kong
| | - Hong Yan
- Department of Electrical Engineering, City University of Hong Kong, Kowloon, Hong Kong
| |
Collapse
|
24
|
Malkomes P, Lunger I, Oppermann E, Abou-El-Ardat K, Oellerich T, Günther S, Canbulat C, Bothur S, Schnütgen F, Yu W, Wingert S, Haetscher N, Catapano C, Dietz MS, Heilemann M, Kvasnicka HM, Holzer K, Serve H, Bechstein WO, Rieger MA. Transglutaminase 2 promotes tumorigenicity of colon cancer cells by inactivation of the tumor suppressor p53. Oncogene 2021; 40:4352-4367. [PMID: 34103685 PMCID: PMC8225513 DOI: 10.1038/s41388-021-01847-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 04/30/2021] [Accepted: 05/17/2021] [Indexed: 02/05/2023]
Abstract
Despite a high clinical need for the treatment of colorectal carcinoma (CRC) as the second leading cause of cancer-related deaths, targeted therapies are still limited. The multifunctional enzyme Transglutaminase 2 (TGM2), which harbors transamidation and GTPase activity, has been implicated in the development and progression of different types of human cancers. However, the mechanism and role of TGM2 in colorectal cancer are poorly understood. Here, we present TGM2 as a promising drug target.In primary patient material of CRC patients, we detected an increased expression and enzymatic activity of TGM2 in colon cancer tissue in comparison to matched normal colon mucosa cells. The genetic ablation of TGM2 in CRC cell lines using shRNAs or CRISPR/Cas9 inhibited cell expansion and tumorsphere formation. In vivo, tumor initiation and growth were reduced upon genetic knockdown of TGM2 in xenotransplantations. TGM2 ablation led to the induction of Caspase-3-driven apoptosis in CRC cells. Functional rescue experiments with TGM2 variants revealed that the transamidation activity is critical for the pro-survival function of TGM2. Transcriptomic and protein-protein interaction analyses applying various methods including super-resolution and time-lapse microscopy showed that TGM2 directly binds to the tumor suppressor p53, leading to its inactivation and escape of apoptosis induction.We demonstrate here that TGM2 is an essential survival factor in CRC, highlighting the therapeutic potential of TGM2 inhibitors in CRC patients with high TGM2 expression. The inactivation of p53 by TGM2 binding indicates a general anti-apoptotic function, which may be relevant in cancers beyond CRC.
Collapse
Affiliation(s)
- Patrizia Malkomes
- Goethe University Hospital Frankfurt, Department of General, Visceral and Transplant Surgery, Frankfurt am Main, Germany
| | - Ilaria Lunger
- Goethe University Hospital Frankfurt, Department of Medicine, Hematology/Oncology, Frankfurt am Main, Germany
| | - Elsie Oppermann
- Goethe University Hospital Frankfurt, Department of General, Visceral and Transplant Surgery, Frankfurt am Main, Germany
| | - Khalil Abou-El-Ardat
- Goethe University Hospital Frankfurt, Department of Medicine, Hematology/Oncology, Frankfurt am Main, Germany
- German Cancer Consortium and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thomas Oellerich
- Goethe University Hospital Frankfurt, Department of Medicine, Hematology/Oncology, Frankfurt am Main, Germany
- German Cancer Consortium and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Frankfurt Cancer Institute, Frankfurt am Main, Germany
| | - Stefan Günther
- Max Planck Institute for Heart and Lung Research, Department I Cardiac Development and Remodelling, Bad Nauheim, Germany
| | - Can Canbulat
- Goethe University Hospital Frankfurt, Department of General, Visceral and Transplant Surgery, Frankfurt am Main, Germany
| | - Sabrina Bothur
- Goethe University Hospital Frankfurt, Department of Medicine, Hematology/Oncology, Frankfurt am Main, Germany
| | - Frank Schnütgen
- Goethe University Hospital Frankfurt, Department of Medicine, Hematology/Oncology, Frankfurt am Main, Germany
- German Cancer Consortium and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Frankfurt Cancer Institute, Frankfurt am Main, Germany
| | - Weijia Yu
- Goethe University Hospital Frankfurt, Department of Medicine, Hematology/Oncology, Frankfurt am Main, Germany
| | - Susanne Wingert
- Goethe University Hospital Frankfurt, Department of Medicine, Hematology/Oncology, Frankfurt am Main, Germany
| | - Nadine Haetscher
- Goethe University Hospital Frankfurt, Department of Medicine, Hematology/Oncology, Frankfurt am Main, Germany
| | - Claudia Catapano
- Single Molecule Biophysics, Institute of Physical and Theoretical Chemistry, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Marina S Dietz
- Single Molecule Biophysics, Institute of Physical and Theoretical Chemistry, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Mike Heilemann
- Single Molecule Biophysics, Institute of Physical and Theoretical Chemistry, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Hans-Michael Kvasnicka
- Goethe University Frankfurt, Senckenberg Institute for Pathology, Frankfurt am Main, Germany
| | - Katharina Holzer
- Goethe University Hospital Frankfurt, Department of General, Visceral and Transplant Surgery, Frankfurt am Main, Germany
- Philipps University of Marburg, Department of Visceral-, Thoracic- and Vascular Surgery, Marburg, Germany
| | - Hubert Serve
- Goethe University Hospital Frankfurt, Department of Medicine, Hematology/Oncology, Frankfurt am Main, Germany
- German Cancer Consortium and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Frankfurt Cancer Institute, Frankfurt am Main, Germany
| | - Wolf Otto Bechstein
- Goethe University Hospital Frankfurt, Department of General, Visceral and Transplant Surgery, Frankfurt am Main, Germany
| | - Michael A Rieger
- Goethe University Hospital Frankfurt, Department of Medicine, Hematology/Oncology, Frankfurt am Main, Germany.
- German Cancer Consortium and German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Frankfurt Cancer Institute, Frankfurt am Main, Germany.
- Cardio-Pulmonary Institute, Frankfurt am Main, Germany.
| |
Collapse
|
25
|
A quantitative view on multivalent nanomedicine targeting. Adv Drug Deliv Rev 2021; 169:1-21. [PMID: 33264593 DOI: 10.1016/j.addr.2020.11.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 11/11/2020] [Accepted: 11/21/2020] [Indexed: 12/17/2022]
Abstract
Although the concept of selective delivery has been postulated over 100 years ago, no targeted nanomedicine has been clinically approved so far. Nanoparticles modified with targeting ligands to promote the selective delivery of therapeutics towards a specific cell population have been extensively reported. However, the rational design of selective particles is still challenging. One of the main reasons for this is the lack of quantitative theoretical and experimental understanding of the interactions involved in cell targeting. In this review, we discuss new theoretical models and experimental methods that provide a quantitative view of targeting. We show the new advancements in multivalency theory enabling the rational design of super-selective nanoparticles. Furthermore, we present the innovative approaches to obtain key targeting parameters at the single-cell and single molecule level and their role in the design of targeting nanoparticles. We believe that the combination of new theoretical multivalent design and experimental methods to quantify receptors and ligands aids in the rational design and clinical translation of targeted nanomedicines.
Collapse
|
26
|
Baldering TN, Karathanasis C, Harwardt MLIE, Freund P, Meurer M, Rahm JV, Knop M, Dietz MS, Heilemann M. CRISPR/Cas12a-mediated labeling of MET receptor enables quantitative single-molecule imaging of endogenous protein organization and dynamics. iScience 2020; 24:101895. [PMID: 33364584 PMCID: PMC7753144 DOI: 10.1016/j.isci.2020.101895] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 11/12/2020] [Accepted: 12/02/2020] [Indexed: 12/31/2022] Open
Abstract
Single-molecule localization microscopy (SMLM) reports on protein organization in cells with near-molecular resolution and in combination with stoichiometric labeling enables protein counting. Fluorescent proteins allow stoichiometric labeling of cellular proteins; however, most methods either lead to overexpression or are complex and time demanding. We introduce CRISPR/Cas12a for simple and efficient tagging of endogenous proteins with a photoactivatable protein for quantitative SMLM and single-particle tracking. We constructed a HEK293T cell line with the receptor tyrosine kinase MET tagged with mEos4b and demonstrate full functionality. We determine the oligomeric state of MET with quantitative SMLM and find a reorganization from monomeric to dimeric MET upon ligand stimulation. In addition, we measured the mobility of single MET receptors in vivo in resting and ligand-treated cells. The combination of CRISPR/Cas12a-assisted endogenous protein labeling and super-resolution microscopy represents a powerful tool for cell biological research with molecular resolution. CRISPR/Cas12a enables endogenous protein labeling for super-resolution microscopy HEK293T cells were generated with MET endogenously labeled with mEos4b Quantitative PALM microscopy reports efficient dimerization of MET receptor Single-particle tracking shows increased MET immobilization upon ligand treatment
Collapse
Affiliation(s)
- Tim N Baldering
- Single Molecule Biophysics, Institute of Physical and Theoretical Chemistry, Goethe University Frankfurt, Max-von-Laue Str. 7, 60438 Frankfurt, Germany
| | - Christos Karathanasis
- Single Molecule Biophysics, Institute of Physical and Theoretical Chemistry, Goethe University Frankfurt, Max-von-Laue Str. 7, 60438 Frankfurt, Germany
| | - Marie-Lena I E Harwardt
- Single Molecule Biophysics, Institute of Physical and Theoretical Chemistry, Goethe University Frankfurt, Max-von-Laue Str. 7, 60438 Frankfurt, Germany
| | - Petra Freund
- Single Molecule Biophysics, Institute of Physical and Theoretical Chemistry, Goethe University Frankfurt, Max-von-Laue Str. 7, 60438 Frankfurt, Germany
| | - Matthias Meurer
- Center for Molecular Biology of Heidelberg University (ZMBH), 69120 Heidelberg, Germany
| | - Johanna V Rahm
- Single Molecule Biophysics, Institute of Physical and Theoretical Chemistry, Goethe University Frankfurt, Max-von-Laue Str. 7, 60438 Frankfurt, Germany
| | - Michael Knop
- Center for Molecular Biology of Heidelberg University (ZMBH), 69120 Heidelberg, Germany.,German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Marina S Dietz
- Single Molecule Biophysics, Institute of Physical and Theoretical Chemistry, Goethe University Frankfurt, Max-von-Laue Str. 7, 60438 Frankfurt, Germany
| | - Mike Heilemann
- Single Molecule Biophysics, Institute of Physical and Theoretical Chemistry, Goethe University Frankfurt, Max-von-Laue Str. 7, 60438 Frankfurt, Germany
| |
Collapse
|
27
|
Schueder F, Unterauer EM, Ganji M, Jungmann R. DNA-Barcoded Fluorescence Microscopy for Spatial Omics. Proteomics 2020; 20:e1900368. [PMID: 33030780 DOI: 10.1002/pmic.201900368] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/24/2020] [Indexed: 12/18/2022]
Abstract
Innovation in genomics, transcriptomics, and proteomics research has created a plethora of state-of-the-art techniques such as nucleic acid sequencing and mass-spectrometry-based proteomics with paramount impact in the life sciences. While current approaches yield quantitative abundance analysis of biomolecules on an almost routine basis, coupling this high content to spatial information in a single cell and tissue context is challenging. Here, current implementations of spatial omics are discussed and recent developments in the field of DNA-barcoded fluorescence microscopy are reviewed. Light is shed on the potential of DNA-based imaging techniques to provide a comprehensive toolbox for spatial genomics and transcriptomics and discuss current challenges, which need to be overcome on the way to spatial proteomics using high-resolution fluorescence microscopy.
Collapse
Affiliation(s)
- Florian Schueder
- Department of Physics and Center for Nanoscience, Ludwig Maximilian University, Geschwister-Scholl-Platz 1, Munich, 80539, Germany.,Max Planck Institute of Biochemistry, Am Klopferspitz 18, Martinsried, 82152, Germany
| | - Eduard M Unterauer
- Department of Physics and Center for Nanoscience, Ludwig Maximilian University, Geschwister-Scholl-Platz 1, Munich, 80539, Germany.,Max Planck Institute of Biochemistry, Am Klopferspitz 18, Martinsried, 82152, Germany
| | - Mahipal Ganji
- Department of Physics and Center for Nanoscience, Ludwig Maximilian University, Geschwister-Scholl-Platz 1, Munich, 80539, Germany.,Max Planck Institute of Biochemistry, Am Klopferspitz 18, Martinsried, 82152, Germany
| | - Ralf Jungmann
- Department of Physics and Center for Nanoscience, Ludwig Maximilian University, Geschwister-Scholl-Platz 1, Munich, 80539, Germany.,Max Planck Institute of Biochemistry, Am Klopferspitz 18, Martinsried, 82152, Germany
| |
Collapse
|
28
|
Schröder MS, Harwardt MLIE, Rahm JV, Li Y, Freund P, Dietz MS, Heilemann M. Imaging the fibroblast growth factor receptor network on the plasma membrane with DNA-assisted single-molecule super-resolution microscopy. Methods 2020; 193:38-45. [PMID: 32389748 DOI: 10.1016/j.ymeth.2020.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 04/17/2020] [Accepted: 05/05/2020] [Indexed: 12/13/2022] Open
Abstract
Fibroblast growth factor receptors (FGFRs) are a subfamily of receptor tyrosine kinases and central players in health and disease. Following ligand binding and the formation of homo- and heteromeric complexes, FGFRs initiate a cellular response. Challenges in studying FGFR activation are inner-subfamily interactions and a complex heterogeneity of these in the cell membrane, which demand for observation techniques that can resolve individual protein complexes and that are compatible with endogenous protein levels. Here, we established an imaging and analysis pipeline for multiplexed single-molecule localization microscopy (SMLM) of the FGFR network at the plasma membrane. Using DNA-labeled primary antibodies, we visualize all four FGFRs in the same cell with near-molecular spatial resolution. From the super-resolution imaging data, we extract information on FGFR density, spatial distribution, and inner-subfamily colocalization. Our approach is straightforward and easily adaptable to other multiplexed SMLM data of membrane proteins.
Collapse
Affiliation(s)
- Mark S Schröder
- Institute of Physical and Theoretical Chemistry, Goethe-University Frankfurt, Max-von-Laue-Str. 7, 60438 Frankfurt, Germany
| | - Marie-Lena I E Harwardt
- Institute of Physical and Theoretical Chemistry, Goethe-University Frankfurt, Max-von-Laue-Str. 7, 60438 Frankfurt, Germany
| | - Johanna V Rahm
- Institute of Physical and Theoretical Chemistry, Goethe-University Frankfurt, Max-von-Laue-Str. 7, 60438 Frankfurt, Germany
| | - Yunqing Li
- Institute of Physical and Theoretical Chemistry, Goethe-University Frankfurt, Max-von-Laue-Str. 7, 60438 Frankfurt, Germany
| | - Petra Freund
- Institute of Physical and Theoretical Chemistry, Goethe-University Frankfurt, Max-von-Laue-Str. 7, 60438 Frankfurt, Germany
| | - Marina S Dietz
- Institute of Physical and Theoretical Chemistry, Goethe-University Frankfurt, Max-von-Laue-Str. 7, 60438 Frankfurt, Germany
| | - Mike Heilemann
- Institute of Physical and Theoretical Chemistry, Goethe-University Frankfurt, Max-von-Laue-Str. 7, 60438 Frankfurt, Germany.
| |
Collapse
|