151
|
Ultrafast data mining of molecular assemblies in multiplexed high-density super-resolution images. Nat Commun 2019; 10:119. [PMID: 30631072 PMCID: PMC6328550 DOI: 10.1038/s41467-018-08048-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 12/13/2018] [Indexed: 12/29/2022] Open
Abstract
Multicolor single-molecule localization super-resolution microscopy has enabled visualization of ultrafine spatial organizations of molecular assemblies within cells. Despite many efforts, current approaches for distinguishing and quantifying such organizations remain limited, especially when these are contained within densely distributed super-resolution data. In theory, higher-order correlation such as the Triple-Correlation function is capable of obtaining the spatial configuration of individual molecular assemblies masked within seemingly discorded dense distributions. However, due to their enormous computational cost such analyses are impractical, even for high-end computers. Here, we developed a fast algorithm for Triple-Correlation analyses of high-content multiplexed super-resolution data. This algorithm computes the probability density of all geometric configurations formed by every triple-wise single-molecule localization from three different channels, circumventing impractical 4D Fourier Transforms of the entire megapixel image. This algorithm achieves 102-folds enhancement in computational speed, allowing for high-throughput Triple-Correlation analyses and robust quantification of molecular complexes in multiplexed super-resolution microscopy. Analyzing the organization of molecular complexes in multi-color single-molecule localization microscopy data requires heavy computation resources that are impractical for laboratory computers. Here the authors develop a coordinate-based Triple-Correlation algorithm with improved speed and reduced computational cost.
Collapse
|
152
|
Maity PC, Datta M, Nicolò A, Jumaa H. Isotype Specific Assembly of B Cell Antigen Receptors and Synergism With Chemokine Receptor CXCR4. Front Immunol 2019. [PMID: 30619343 DOI: 10.3389/fimmu.2018.02988.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Expression of the membrane-bound form of the immunoglobulin (Ig) as part of the antigen receptor is indispensable for both the development and the effector function of B cells. Among five known isotypes, IgM and IgD are the common B cell antigen receptors (BCRs) that are co-expressed in naïve B cells. Despite having identical antigen specificity and being associated with the same signaling heterodimer Igα/Igβ (CD79a/CD79b), IgM and IgD-BCR isotypes functionally differ from each other in the manner of antigen binding, the formation of isolated nanoclusters and in their interaction with co-receptors such as CD19 and CXCR4 on the plasma membrane. With recent developments in experimental techniques, it is now possible to investigate the nanoscale organization of the BCR and better understand early events of BCR engagement. Interestingly, the cytoskeleton network beneath the membrane controls the BCR isotype-specific organization and its interaction with co-receptors. BCR triggering results in reorganization of the cytoskeleton network, which is further modulated by isotype-specific signals from co-receptors. For instance, IgD-BCR is closely associated with CXCR4 on mature B cells and this close proximity allows CXCR4 to employ the BCR machinery as signaling hub. In this review, we discuss the functional specificity and nanocluster assembly of BCR isotypes and the consequences of cross-talk between CXCR4 and IgD-BCR. Furthermore, given the role of BCR and CXCR4 signaling in the development and survival of leukemic B cells, we discuss the consequences of the cross-talk between CXCR4 and the BCR for controlling the growth of transformed B cells.
Collapse
Affiliation(s)
| | - Moumita Datta
- Institute of Immunology, Ulm University, Ulm, Germany
| | | | - Hassan Jumaa
- Institute of Immunology, Ulm University, Ulm, Germany
| |
Collapse
|
153
|
Maity PC, Datta M, Nicolò A, Jumaa H. Isotype Specific Assembly of B Cell Antigen Receptors and Synergism With Chemokine Receptor CXCR4. Front Immunol 2019; 9:2988. [PMID: 30619343 PMCID: PMC6305424 DOI: 10.3389/fimmu.2018.02988] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 12/04/2018] [Indexed: 12/20/2022] Open
Abstract
Expression of the membrane-bound form of the immunoglobulin (Ig) as part of the antigen receptor is indispensable for both the development and the effector function of B cells. Among five known isotypes, IgM and IgD are the common B cell antigen receptors (BCRs) that are co-expressed in naïve B cells. Despite having identical antigen specificity and being associated with the same signaling heterodimer Igα/Igβ (CD79a/CD79b), IgM and IgD-BCR isotypes functionally differ from each other in the manner of antigen binding, the formation of isolated nanoclusters and in their interaction with co-receptors such as CD19 and CXCR4 on the plasma membrane. With recent developments in experimental techniques, it is now possible to investigate the nanoscale organization of the BCR and better understand early events of BCR engagement. Interestingly, the cytoskeleton network beneath the membrane controls the BCR isotype-specific organization and its interaction with co-receptors. BCR triggering results in reorganization of the cytoskeleton network, which is further modulated by isotype-specific signals from co-receptors. For instance, IgD-BCR is closely associated with CXCR4 on mature B cells and this close proximity allows CXCR4 to employ the BCR machinery as signaling hub. In this review, we discuss the functional specificity and nanocluster assembly of BCR isotypes and the consequences of cross-talk between CXCR4 and IgD-BCR. Furthermore, given the role of BCR and CXCR4 signaling in the development and survival of leukemic B cells, we discuss the consequences of the cross-talk between CXCR4 and the BCR for controlling the growth of transformed B cells.
Collapse
Affiliation(s)
| | - Moumita Datta
- Institute of Immunology, Ulm University, Ulm, Germany
| | | | - Hassan Jumaa
- Institute of Immunology, Ulm University, Ulm, Germany
| |
Collapse
|
154
|
Reismann AWAF, Atanasova L, Schrangl L, Zeilinger S, Schütz GJ. Temporal Filtering to Improve Single Molecule Identification in High Background Samples. Molecules 2018; 23:molecules23123338. [PMID: 30562966 PMCID: PMC6321103 DOI: 10.3390/molecules23123338] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 12/06/2018] [Accepted: 12/13/2018] [Indexed: 11/16/2022] Open
Abstract
Single molecule localization microscopy is currently revolutionizing the life sciences as it offers, for the first time, insights into the organization of biological samples below the classical diffraction limit of light microscopy. While there have been numerous examples of new biological findings reported in the last decade, the technique could not reach its full potential due to a set of limitations immanent to the samples themselves. Particularly, high background signals impede the proper performance of most single-molecule identification and localization algorithms. One option is to exploit the characteristic blinking of single molecule signals, which differs substantially from the residual brightness fluctuations of the fluorescence background. To pronounce single molecule signals, we used a temporal high-pass filtering in Fourier space on a pixel-by-pixel basis. We evaluated the performance of temporal filtering by assessing statistical parameters such as true positive rate and false discovery rate. For this, ground truth signals were generated by simulations and overlaid onto experimentally derived movies of samples with high background signals. Compared to the nonfiltered case, we found an improvement of the sensitivity by up to a factor 3.5 while no significant change in the localization accuracy was observable.
Collapse
Affiliation(s)
| | - Lea Atanasova
- Institute of Applied Physics, TU Wien, Getreidemarkt 9, A-1060 Vienna, Austria.
- Department of Microbiology, University of Innsbruck, Technikerstrasse 25, A-6020 Innsbruck, Austria.
| | - Lukas Schrangl
- Institute of Applied Physics, TU Wien, Getreidemarkt 9, A-1060 Vienna, Austria.
| | - Susanne Zeilinger
- Department of Microbiology, University of Innsbruck, Technikerstrasse 25, A-6020 Innsbruck, Austria.
- Institute of Chemical, Environmental and Bioscience Engineering, TU Wien, Gumpendorferstrasse 1a, A-1060 Vienna, Austria.
| | - Gerhard J Schütz
- Institute of Applied Physics, TU Wien, Getreidemarkt 9, A-1060 Vienna, Austria.
| |
Collapse
|
155
|
Staszowska AD, Fox-Roberts P, Hirvonen LM, Peddie CJ, Collinson LM, Jones GE, Cox S. The Rényi divergence enables accurate and precise cluster analysis for localization microscopy. Bioinformatics 2018; 34:4102-4111. [PMID: 29868717 PMCID: PMC6247934 DOI: 10.1093/bioinformatics/bty403] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 05/30/2018] [Indexed: 11/18/2022] Open
Abstract
Motivation Clustering analysis is a key technique for quantitatively characterizing structures in localization microscopy images. To build up accurate information about biological structures, it is critical that the quantification is both accurate (close to the ground truth) and precise (has small scatter and is reproducible). Results Here, we describe how the Rényi divergence can be used for cluster radius measurements in localization microscopy data. We demonstrate that the Rényi divergence can operate with high levels of background and provides results which are more accurate than Ripley’s functions, Voronoi tesselation or DBSCAN. Availability and implementation The data supporting this research and the software described are accessible at the following site: https://dx.doi.org/10.18742/RDM01-316. Correspondence and requests for materials should be addressed to the corresponding author. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Adela D Staszowska
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, UK
| | - Patrick Fox-Roberts
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, UK
| | - Liisa M Hirvonen
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, UK
| | - Christopher J Peddie
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, London, UK
| | - Lucy M Collinson
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, London, UK
| | - Gareth E Jones
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, UK
| | - Susan Cox
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, UK
| |
Collapse
|
156
|
Song X, Liu W, Yuan X, Jiang J, Wang W, Mullen M, Zhao X, Zhang Y, Liu F, Du S, Rehman A, Tian R, Li J, Frost A, Song Z, Green HN, Henry C, Liu X, Ding X, Wang D, Yao X. Acetylation of ACAP4 regulates CCL18-elicited breast cancer cell migration and invasion. J Mol Cell Biol 2018; 10:559-572. [PMID: 30395269 PMCID: PMC6692856 DOI: 10.1093/jmcb/mjy058] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 08/03/2018] [Accepted: 08/20/2018] [Indexed: 01/03/2023] Open
Abstract
Tumor metastasis represents the main causes of cancer-related death. Our recent study showed that chemokine CCL18 secreted from tumor-associated macrophages regulates breast tumor metastasis, but the underlying mechanisms remain less clear. Here, we show that ARF6 GTPase-activating protein ACAP4 regulates CCL18-elicited breast cancer cell migration via the acetyltransferase PCAF-mediated acetylation. CCL18 stimulation elicited breast cancer cell migration and invasion via PCAF-dependent acetylation. ACAP4 physically interacts with PCAF and is a cognate substrate of PCAF during CCL18 stimulation. The acetylation site of ACAP4 by PCAF was mapped to Lys311 by mass spectrometric analyses. Importantly, dynamic acetylation of ACAP4 is essential for CCL18-induced breast cancer cell migration and invasion, as overexpression of the persistent acetylation-mimicking or non-acetylatable ACAP4 mutant blocked CCL18-elicited cell migration and invasion. Mechanistically, the acetylation of ACAP4 at Lys311 reduced the lipid-binding activity of ACAP4 to ensure a robust and dynamic cycling of ARF6-ACAP4 complex with plasma membrane in response to CCL18 stimulation. Thus, these results present a previously undefined mechanism by which CCL18-elicited acetylation of the PH domain controls dynamic interaction between ACAP4 and plasma membrane during breast cancer cell migration and invasion.
Collapse
Affiliation(s)
- Xiaoyu Song
- Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, Hefei National Science Center for Physical Sciences at Nanoscale, CAS Center of Excellence in Molecular Cell Sciences, University of Science & Technology of China, Hefei, China
- Keck Center for Cellular Dynamics & Department of Physiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Wei Liu
- Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, Hefei National Science Center for Physical Sciences at Nanoscale, CAS Center of Excellence in Molecular Cell Sciences, University of Science & Technology of China, Hefei, China
- Keck Center for Cellular Dynamics & Department of Physiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Xiao Yuan
- Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, Hefei National Science Center for Physical Sciences at Nanoscale, CAS Center of Excellence in Molecular Cell Sciences, University of Science & Technology of China, Hefei, China
- Department of Chemistry, Southern University of Science & Technology, Shenzhen, China
| | - Jiying Jiang
- Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, Hefei National Science Center for Physical Sciences at Nanoscale, CAS Center of Excellence in Molecular Cell Sciences, University of Science & Technology of China, Hefei, China
- Keck Center for Cellular Dynamics & Department of Physiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Wanjuan Wang
- School of Basic Medical Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - McKay Mullen
- Keck Center for Cellular Dynamics & Department of Physiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Xuannv Zhao
- Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, Hefei National Science Center for Physical Sciences at Nanoscale, CAS Center of Excellence in Molecular Cell Sciences, University of Science & Technology of China, Hefei, China
| | - Yin Zhang
- Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, Hefei National Science Center for Physical Sciences at Nanoscale, CAS Center of Excellence in Molecular Cell Sciences, University of Science & Technology of China, Hefei, China
- School of Basic Medical Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Fusheng Liu
- Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, Hefei National Science Center for Physical Sciences at Nanoscale, CAS Center of Excellence in Molecular Cell Sciences, University of Science & Technology of China, Hefei, China
- School of Basic Medical Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Shihao Du
- Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, Hefei National Science Center for Physical Sciences at Nanoscale, CAS Center of Excellence in Molecular Cell Sciences, University of Science & Technology of China, Hefei, China
- School of Basic Medical Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Adeel Rehman
- Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, Hefei National Science Center for Physical Sciences at Nanoscale, CAS Center of Excellence in Molecular Cell Sciences, University of Science & Technology of China, Hefei, China
| | - Ruijun Tian
- Department of Chemistry, Southern University of Science & Technology, Shenzhen, China
| | - Jian Li
- Keck Center for Cellular Dynamics & Department of Physiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Andra Frost
- Department of Pathology, University of Alabama School of Medicine, Birmingham, AL, USA
| | - Zhenwei Song
- Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, Hefei National Science Center for Physical Sciences at Nanoscale, CAS Center of Excellence in Molecular Cell Sciences, University of Science & Technology of China, Hefei, China
| | - Hadiyah-Nicole Green
- Keck Center for Cellular Dynamics & Department of Physiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Calmour Henry
- Keck Center for Cellular Dynamics & Department of Physiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Xing Liu
- Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, Hefei National Science Center for Physical Sciences at Nanoscale, CAS Center of Excellence in Molecular Cell Sciences, University of Science & Technology of China, Hefei, China
- Keck Center for Cellular Dynamics & Department of Physiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Xia Ding
- School of Basic Medical Sciences, Beijing University of Chinese Medicine, Beijing, China
- Keck Center for Cellular Dynamics & Department of Physiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Dongmei Wang
- Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, Hefei National Science Center for Physical Sciences at Nanoscale, CAS Center of Excellence in Molecular Cell Sciences, University of Science & Technology of China, Hefei, China
| | - Xuebiao Yao
- Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, Hefei National Science Center for Physical Sciences at Nanoscale, CAS Center of Excellence in Molecular Cell Sciences, University of Science & Technology of China, Hefei, China
| |
Collapse
|
157
|
Mohapatra S, Weisshaar JC. Modified Pearson correlation coefficient for two-color imaging in spherocylindrical cells. BMC Bioinformatics 2018; 19:428. [PMID: 30445904 PMCID: PMC6240329 DOI: 10.1186/s12859-018-2444-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 10/22/2018] [Indexed: 11/10/2022] Open
Abstract
The revolution in fluorescence microscopy enables sub-diffraction-limit ("superresolution") localization of hundreds or thousands of copies of two differently labeled proteins in the same live cell. In typical experiments, fluorescence from the entire three-dimensional (3D) cell body is projected along the z-axis of the microscope to form a 2D image at the camera plane. For imaging of two different species, here denoted "red" and "green", a significant biological question is the extent to which the red and green spatial distributions are positively correlated, anti-correlated, or uncorrelated. A commonly used statistic for assessing the degree of linear correlation between two image matrices R and G is the Pearson Correlation Coefficient (PCC). PCC should vary from - 1 (perfect anti-correlation) to 0 (no linear correlation) to + 1 (perfect positive correlation). However, in the special case of spherocylindrical bacterial cells such as E. coli or B. subtilis, we show that the PCC fails both qualitatively and quantitatively. PCC returns the same + 1 value for 2D projections of distributions that are either perfectly correlated in 3D or completely uncorrelated in 3D. The PCC also systematically underestimates the degree of anti-correlation between the projections of two perfectly anti-correlated 3D distributions. The problem is that the projection of a random spatial distribution within the 3D spherocylinder is non-random in 2D, whereas PCC compares every matrix element of R or G with the constant mean value [Formula: see text] or [Formula: see text]. We propose a modified Pearson Correlation Coefficient (MPCC) that corrects this problem for spherocylindrical cell geometry by using the proper reference matrix for comparison with R and G. Correct behavior of MPCC is confirmed for a variety of numerical simulations and on experimental distributions of HU and RNA polymerase in live E. coli cells. The MPCC concept should be generalizable to other cell shapes.
Collapse
Affiliation(s)
- Sonisilpa Mohapatra
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA. .,Present Address: Department of Biophysics and Biophysical Chemistry, Johns Hopkins School of Medicine, Baltimore, 21205, USA.
| | - James C Weisshaar
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| |
Collapse
|
158
|
Tonzi P, Yin Y, Lee CWT, Rothenberg E, Huang TT. Translesion polymerase kappa-dependent DNA synthesis underlies replication fork recovery. eLife 2018; 7:41426. [PMID: 30422114 PMCID: PMC6251625 DOI: 10.7554/elife.41426] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 11/12/2018] [Indexed: 12/31/2022] Open
Abstract
DNA replication stress is often defined by the slowing or stalling of replication fork progression leading to local or global DNA synthesis inhibition. Failure to resolve replication stress in a timely manner contribute toward cell cycle defects, genome instability and human disease; however, the mechanism for fork recovery remains poorly defined. Here, we show that the translesion DNA polymerase (Pol) kappa, a DinB orthologue, has a unique role in both protecting and restarting stalled replication forks under conditions of nucleotide deprivation. Importantly, Pol kappa-mediated DNA synthesis during hydroxyurea (HU)-dependent fork restart is regulated by both the Fanconi Anemia (FA) pathway and PCNA polyubiquitination. Loss of Pol kappa prevents timely rescue of stalled replication forks, leading to replication-associated genomic instability, and a p53-dependent cell cycle defect. Taken together, our results identify a previously unanticipated role for Pol kappa in promoting DNA synthesis and replication stress recovery at sites of stalled forks.
Collapse
Affiliation(s)
- Peter Tonzi
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, United States
| | - Yandong Yin
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, United States
| | - Chelsea Wei Ting Lee
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, United States
| | - Eli Rothenberg
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, United States
| | - Tony T Huang
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, United States
| |
Collapse
|
159
|
Baumgart F, Arnold AM, Rossboth BK, Brameshuber M, Schütz GJ. What we talk about when we talk about nanoclusters. Methods Appl Fluoresc 2018; 7:013001. [PMID: 30412469 DOI: 10.1088/2050-6120/aaed0f] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Superresolution microscopy results have sparked the idea that many membrane proteins are not randomly distributed across the plasma membrane but are instead arranged in nanoclusters. Frequently, these new results seemed to confirm older data based on biochemical and electron microscopy experiments. Recently, however, it was recognized that multiple countings of the very same fluorescently labeled protein molecule can be easily confused with true protein clusters. Various strategies have been developed, which are intended to solve the problem of discriminating true protein clusters from imaging artifacts. We believe that there is currently no perfect algorithm for this problem; instead, different approaches have different strengths and weaknesses. In this review, we discuss single molecule localization microscopy in view of its ability to detect nanoclusters of membrane proteins. To capture the different views on nanoclustering, we chose an unconventional style for this article: we placed its scientific content in the setting of a fictive conference, where five researchers from different fields discuss the problem of detecting and quantifying nanoclusters. Using this style, we feel that the different approaches common for different research areas can be well illustrated. Similarities to a short story by Raymond Carver are not unintentional.
Collapse
|
160
|
Semino C, Carta S, Gattorno M, Sitia R, Rubartelli A. Progressive waves of IL-1β release by primary human monocytes via sequential activation of vesicular and gasdermin D-mediated secretory pathways. Cell Death Dis 2018; 9:1088. [PMID: 30352992 PMCID: PMC6199333 DOI: 10.1038/s41419-018-1121-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 09/28/2018] [Indexed: 12/20/2022]
Abstract
IL-1β is an essential cytokine, but its release needs to be strictly controlled to avoid severe inflammatory manifestations. Lacking a signal sequence, IL-1β does not follow the endoplasmic reticulum-Golgi route. Several pathways have been proposed to mediate its release. One involves the translocation of pro-IL-1β into intracellular vesicles of lysosomal origin that eventually fuse with the plasma membrane. Another exploits pores formed on the plasma membrane upon proteolytic cleavage of gasdermin D (GSDMD). Here we investigated how primary monocytes-the main source of IL-1β in humans-control IL-1β release in response to pro-inflammatory stimuli of increasing intensity and found that two different routes are induced depending on the strength of activation. Triggering of Toll-like receptor 4 (TLR4) by LPS induces slow IL-1β release through LAMP2A+ vesicles. In contrast, the simultaneous stimulation of TLR2, TLR4 and TLR7/8 drives high levels of ROS, GSDMD cleavage and faster IL-1β secretion. Drugs blocking ROS production prevent GSDMD cleavage supporting a role of oxidative stress in GSDMD-mediated secretion. Singly stimulated monocytes undergo apoptosis, whereas triple stimulation triggers pyroptosis, which might amplify inflammation. In both cases, however, IL-1β secretion precedes cell death. Inhibition of caspases 4/5 prevents GSDMD cleavage and pore-mediated secretion, but not vesicular release. The two pathways also display other distinct pharmacologic sensitivities that reflect the underlying mechanisms. Remarkably, single TLR4 stimulation is sufficient to activate massive, GSDMD-mediated IL-1β secretion in monocytes from patients affected by Cryopyrin Associated Periodic Syndrome (CAPS), an autoinflammatory disease linked to NLRP3 mutations. The exaggerated sensitivity to activation correlates with high basal ROS levels in CAPS monocytes. In conclusion, the vesicular pathway limits IL-1β release upon low pathogen load while stronger stimulation or concomitant cell stress induce instead uncontrolled secretion via GSDMD leading to detrimental inflammatory manifestations.
Collapse
Affiliation(s)
- Claudia Semino
- Protein Transport and Secretion Unit, IRCCS Ospedale San Raffaele/Università Vita-Salute San Raffaele, 20132, Milan, Italy
| | - Sonia Carta
- Cell Biology Unit, Ospedale Policlinico San Martino, 16132, Genoa, Italy
| | - Marco Gattorno
- Clinica Pediatrica e Reumatologia, "G. Gaslini" Scientific Institute, 16147, Genoa, Italy
| | - Roberto Sitia
- Protein Transport and Secretion Unit, IRCCS Ospedale San Raffaele/Università Vita-Salute San Raffaele, 20132, Milan, Italy
| | - Anna Rubartelli
- Cell Biology Unit, Ospedale Policlinico San Martino, 16132, Genoa, Italy.
| |
Collapse
|
161
|
Wijesooriya CS, Nyamekye CKA, Smith EA. Optical Imaging of the Nanoscale Structure and Dynamics of Biological Membranes. Anal Chem 2018; 91:425-440. [DOI: 10.1021/acs.analchem.8b04755] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
| | - Charles K. A. Nyamekye
- Department of Chemistry, Iowa State University, Ames, Iowa 50011, United States
- The Ames Laboratory, U.S. Department of Energy, Ames, Iowa 50011, United States
| | - Emily A. Smith
- Department of Chemistry, Iowa State University, Ames, Iowa 50011, United States
- The Ames Laboratory, U.S. Department of Energy, Ames, Iowa 50011, United States
| |
Collapse
|
162
|
Tobin SJ, Wakefield DL, Jones V, Liu X, Schmolze D, Jovanović-Talisman T. Single molecule localization microscopy coupled with touch preparation for the quantification of trastuzumab-bound HER2. Sci Rep 2018; 8:15154. [PMID: 30310083 PMCID: PMC6181918 DOI: 10.1038/s41598-018-33225-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 09/25/2018] [Indexed: 12/11/2022] Open
Abstract
All breast cancers are assessed for levels of human epidermal growth factor receptor 2 (HER2). Fluorescence in situ hybridization (FISH) and immunohistochemistry are currently used to determine if a patient is eligible for anti-HER2 therapy. Limitations of both tests include variability and relatively long processing times. Additionally, neither test determines whether HER2 contains the extracellular domain. While truncated in some tumors, this domain is required for binding of the therapeutic antibody trastuzumab. Here, trastuzumab was used to directly detect HER2 with quantitative single molecule localization microscopy (qSMLM). In proof of concept studies, our new method rapidly quantified both HER2 density and features of nano-organization. In cultured cells, the method was sensitive to subtle variations in HER2 expression. To assess patient samples, we combined qSMLM with tissue touch preparation (touch prep-qSMLM) and examined large areas of intact membranes. For cell lines and patient samples, HER2 copy numbers from FISH showed a significant positive correlation with detected densities from qSMLM and trended with HER2 cluster occupancy.
Collapse
Affiliation(s)
- Steven J Tobin
- Department of Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Devin L Wakefield
- Department of Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Veronica Jones
- Department of Surgery, City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Xueli Liu
- Division of Biostatistics, City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Daniel Schmolze
- Department of Pathology, City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Tijana Jovanović-Talisman
- Department of Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA.
| |
Collapse
|
163
|
Golfetto O, Wakefield DL, Cacao EE, Avery KN, Kenyon V, Jorand R, Tobin SJ, Biswas S, Gutierrez J, Clinton R, Ma Y, Horne DA, Williams JC, Jovanović-Talisman T. A Platform To Enhance Quantitative Single Molecule Localization Microscopy. J Am Chem Soc 2018; 140:12785-12797. [PMID: 30256630 PMCID: PMC6187371 DOI: 10.1021/jacs.8b04939] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Quantitative single molecule localization microscopy (qSMLM) is a powerful approach to study in situ protein organization. However, uncertainty regarding the photophysical properties of fluorescent reporters can bias the interpretation of detected localizations and subsequent quantification. Furthermore, strategies to efficiently detect endogenous proteins are often constrained by label heterogeneity and reporter size. Here, a new surface assay for molecular isolation (SAMI) was developed for qSMLM and used to characterize photophysical properties of fluorescent proteins and dyes. SAMI-qSMLM afforded robust quantification. To efficiently detect endogenous proteins, we used fluorescent ligands that bind to a specific site on engineered antibody fragments. Both the density and nano-organization of membrane-bound epidermal growth factor receptors (EGFR, HER2, and HER3) were determined by a combination of SAMI, antibody engineering, and pair-correlation analysis. In breast cancer cell lines, we detected distinct differences in receptor density and nano-organization upon treatment with therapeutic agents. This new platform can improve molecular quantification and can be developed to study the local protein environment of intact cells.
Collapse
Affiliation(s)
- Ottavia Golfetto
- Department of Molecular Medicine , Beckman Research Institute, City of Hope , 1500 East Duarte Road , Duarte , California 91010 , United States
| | - Devin L Wakefield
- Department of Molecular Medicine , Beckman Research Institute, City of Hope , 1500 East Duarte Road , Duarte , California 91010 , United States
| | - Eliedonna E Cacao
- Department of Molecular Medicine , Beckman Research Institute, City of Hope , 1500 East Duarte Road , Duarte , California 91010 , United States
| | - Kendra N Avery
- Department of Molecular Medicine , Beckman Research Institute, City of Hope , 1500 East Duarte Road , Duarte , California 91010 , United States
| | - Victor Kenyon
- Department of Molecular Medicine , Beckman Research Institute, City of Hope , 1500 East Duarte Road , Duarte , California 91010 , United States
| | - Raphael Jorand
- Department of Molecular Medicine , Beckman Research Institute, City of Hope , 1500 East Duarte Road , Duarte , California 91010 , United States
| | - Steven J Tobin
- Department of Molecular Medicine , Beckman Research Institute, City of Hope , 1500 East Duarte Road , Duarte , California 91010 , United States
| | - Sunetra Biswas
- Department of Molecular Medicine , Beckman Research Institute, City of Hope , 1500 East Duarte Road , Duarte , California 91010 , United States
| | - Jennifer Gutierrez
- Department of Molecular Medicine , Beckman Research Institute, City of Hope , 1500 East Duarte Road , Duarte , California 91010 , United States
| | - Ronald Clinton
- Department of Molecular Medicine , Beckman Research Institute, City of Hope , 1500 East Duarte Road , Duarte , California 91010 , United States
| | - Yuelong Ma
- Department of Molecular Medicine , Beckman Research Institute, City of Hope , 1500 East Duarte Road , Duarte , California 91010 , United States
| | - David A Horne
- Department of Molecular Medicine , Beckman Research Institute, City of Hope , 1500 East Duarte Road , Duarte , California 91010 , United States
| | - John C Williams
- Department of Molecular Medicine , Beckman Research Institute, City of Hope , 1500 East Duarte Road , Duarte , California 91010 , United States
| | - Tijana Jovanović-Talisman
- Department of Molecular Medicine , Beckman Research Institute, City of Hope , 1500 East Duarte Road , Duarte , California 91010 , United States
| |
Collapse
|
164
|
Zhao M, Nicovich PR, Janco M, Deng Q, Yang Z, Ma Y, Böcking T, Gaus K, Gooding JJ. Ultralow- and Low-Background Surfaces for Single-Molecule Localization Microscopy of Multistep Biointerfaces for Single-Molecule Sensing. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:10012-10018. [PMID: 30067032 DOI: 10.1021/acs.langmuir.8b01487] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Single-molecule localization microscopy (SMLM) has created the opportunity of pushing fluorescence microscopy from being a biological imaging tool to a surface characterization and possibly even a quantitative analytical tool. The latter could be achieved by molecular counting using pointillist SMLM data sets. However, SMLM is especially sensitive to background fluorescent signals, which influences any subsequent analysis. Therefore, fabricating sensing surfaces that resist nonspecific adsorption of proteins, even after multiple modification steps, has become paramount. Herein is reported two different ways to modify surfaces: dichlorodimethylsilane-biotinylated bovine serum albumin-Tween-20 (DbT20) and poly-l-lysine grafted polyethylene glycol (PLL-PEG) mixed with biotinylated PLL-PEG (PLL-PEG/PEGbiotin). The results show that the ability to resist nonspecific adsorption of DbT20 surfaces deteriorates with an increase in the number of modification steps required after the addition of the DbT20, which limits the applicability of this surface for SMLM. As such, a new surface for SMLM that employs PLL-PEG/PEGbiotin was developed that exhibits ultralow amounts of nonspecific protein adsorption even after many modification steps. The utility of the surface was demonstrated for human influenza hemagglutinin-tagged mEos2, which was directly pulled down from cell lysates onto the PLL-PEG/PEGbiotin surface. The results strongly indicated that the PLL-PEG/PEGbiotin surface satisfies the criteria of SMLM imaging of a negligible background signal and negligible nonspecific adsorption.
Collapse
|
165
|
Delcanale P, Miret-Ontiveros B, Arista-Romero M, Pujals S, Albertazzi L. Nanoscale Mapping Functional Sites on Nanoparticles by Points Accumulation for Imaging in Nanoscale Topography (PAINT). ACS NANO 2018; 12:7629-7637. [PMID: 30048592 DOI: 10.1021/acsnano.7b09063] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
The ability of nanoparticles to selectively recognize a molecular target constitutes the key toward nanomedicine applications such as drug delivery and diagnostics. The activity of such devices is mediated by the presence of multiple copies of functional molecules on the nanostructure surface. Therefore, understanding the number and the distribution of nanoparticle functional groups is of utmost importance for the rational design of effective materials. Analytical methods are available, but to obtain quantitative information at the level of single particles and single functional sites, i. e., going beyond the ensemble, remains highly challenging. Here we introduce the use of an optical nanoscopy technique, DNA points accumulation for imaging in nanoscale topography (DNA-PAINT), to address this issue. Combining subdiffraction spatial resolution with molecular selectivity and sensitivity, DNA-PAINT provides both geometrical and functional information at the level of a single nanostructure. We show how DNA-PAINT can be used to image and quantify relevant functional proteins such as antibodies and streptavidin on nanoparticles and microparticles with nanometric accuracy in 3D and multiple colors. The generality and the applicability of our method without the need for fluorescent labeling hold great promise for the robust quantitative nanocharacterization of functional nanomaterials.
Collapse
Affiliation(s)
- Pietro Delcanale
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology , Baldiri Reixac 15-21 , 08028 Barcelona , Spain
| | - Bernat Miret-Ontiveros
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology , Baldiri Reixac 15-21 , 08028 Barcelona , Spain
| | - Maria Arista-Romero
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology , Baldiri Reixac 15-21 , 08028 Barcelona , Spain
| | - Silvia Pujals
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology , Baldiri Reixac 15-21 , 08028 Barcelona , Spain
| | - Lorenzo Albertazzi
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology , Baldiri Reixac 15-21 , 08028 Barcelona , Spain
- Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS) , Eindhoven University of Technology , 5612AZ Eindhoven , The Netherlands
| |
Collapse
|
166
|
Monolayer surface chemistry enables 2-colour single molecule localisation microscopy of adhesive ligands and adhesion proteins. Nat Commun 2018; 9:3320. [PMID: 30127420 PMCID: PMC6102261 DOI: 10.1038/s41467-018-05837-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 07/30/2018] [Indexed: 12/20/2022] Open
Abstract
Nanofabricated and nanopatterned surfaces have revealed the sensitivity of cell adhesion to nanoscale variations in the spacing of adhesive ligands such as the tripeptide arginine-glycine-aspartic acid (RGD). To date, surface characterisation and cell adhesion are often examined in two separate experiments so that the localisation of ligands and adhesion proteins cannot be combined in the same image. Here we developed self-assembled monolayer chemistry for indium tin oxide (ITO) surfaces for single molecule localisation microscopy (SMLM). Cell adhesion and spreading were sensitive to average RGD spacing. At low average RGD spacing, a threshold exists of 0.8 RGD peptides per µm2 that tether cells to the substratum but this does not enable formation of focal adhesions. These findings suggest that cells can sense and engage single adhesive ligands but ligand clustering is required for cell spreading. Thus, our data reveal subtle differences in adhesion biology that may be obscured in ensemble measurements. To date, the precise localisation of ligands and adhesion proteins are determined in two parallel characterization setups. Here, the authors report a self-assembled monolayer chemistry for indium tin oxide surfaces allowing single molecule localisation microscopy (SMLM) imaging of ligands and adhesion proteins in a single experiment.
Collapse
|
167
|
Brandsma AM, Schwartz SL, Wester MJ, Valley CC, Blezer GLA, Vidarsson G, Lidke KA, Ten Broeke T, Lidke DS, Leusen JHW. Mechanisms of inside-out signaling of the high-affinity IgG receptor FcγRI. Sci Signal 2018; 11:11/540/eaaq0891. [PMID: 30042128 DOI: 10.1126/scisignal.aaq0891] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Fc receptors (FcRs) are an important bridge between the innate and adaptive immune system. Fc gamma receptor I (FcγRI; CD64), the high-affinity receptor for immunoglobulin G (IgG), plays roles in inflammation, autoimmune responses, and immunotherapy. Stimulation of myeloid cells with cytokines, such as tumor necrosis factor-α ( TNFα) and interferon-γ ( IFNγ), increases the binding of FcγRI to immune complexes (ICs), such as antibody-opsonized pathogens or tumor cells, through a process known as "inside-out" signaling. Using super-resolution imaging, we found that stimulation of cells with IL-3 also enhanced the clustering of FcγRI both before and after exposure to ICs. This increased clustering was dependent on an intact actin cytoskeleton. We found that chemical inhibition of the activity of the phosphatase PP1 reduced FcγRI inside-out signaling, although the phosphorylation of FcγRI itself was unaffected. Furthermore, the antibody-dependent cytotoxic activity of human neutrophils toward CD20-expressing tumor cells was increased after stimulation with TNFα and IFNγ. These results suggest that nanoscale reorganization of FcγRI, stimulated by cytokine-induced, inside-out signaling, enhances FcγRI cellular effector functions.
Collapse
Affiliation(s)
- Arianne M Brandsma
- Immunotherapy Laboratory, Laboratory for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Samantha L Schwartz
- Department of Pathology and Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87102, USA
| | - Michael J Wester
- Department of Pathology and Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87102, USA
| | - Christopher C Valley
- Department of Pathology and Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87102, USA
| | - Gittan L A Blezer
- Immunotherapy Laboratory, Laboratory for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Gestur Vidarsson
- Sanquin Research and Landsteiner Laboratory, Department of Experimental Hematology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Keith A Lidke
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM 87131, USA
| | - Toine Ten Broeke
- Immunotherapy Laboratory, Laboratory for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Diane S Lidke
- Department of Pathology and Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87102, USA
| | - Jeanette H W Leusen
- Immunotherapy Laboratory, Laboratory for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands.
| |
Collapse
|
168
|
Lebreton S, Zurzolo C, Paladino S. Organization of GPI-anchored proteins at the cell surface and its physiopathological relevance. Crit Rev Biochem Mol Biol 2018; 53:403-419. [PMID: 30040489 DOI: 10.1080/10409238.2018.1485627] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Glycosylphosphatidylinositol (GPI)-anchored proteins (GPI-APs) are a class of proteins attached to the extracellular leaflet of the plasma membrane via a post-translational modification, the glycolipid anchor. The presence of both glycolipid anchor and protein portion confers them unique features. GPI-APs are expressed in all eukaryotes, from fungi to plants and animals. They display very diverse functions ranging from enzymatic activity, signaling, cell adhesion, cell wall metabolism, neuritogenesis, and immune response. Likewise other plasma membrane proteins, the spatio-temporal organization of GPI-APs is critical for their biological activities in physiological conditions. In this review, we will summarize the latest findings on plasma membrane organization of GPI-APs and the mechanism of its regulation in different cell types. We will also examine the involvement of specific GPI-APs namely the prion protein PrPC, the Folate Receptor alpha and the urokinase plasminogen activator receptor in human diseases focusing on neurodegenerative diseases and cancer.
Collapse
Affiliation(s)
- Stéphanie Lebreton
- a Unité de Trafic Membranaire et Pathogénèse, Institut Pasteur , Paris , France
| | - Chiara Zurzolo
- a Unité de Trafic Membranaire et Pathogénèse, Institut Pasteur , Paris , France
| | - Simona Paladino
- b Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II , Napoli , Italy.,c CEINGE Biotecnologie Avanzate , Napoli , Italy
| |
Collapse
|
169
|
Rossboth B, Arnold AM, Ta H, Platzer R, Kellner F, Huppa JB, Brameshuber M, Baumgart F, Schütz GJ. TCRs are randomly distributed on the plasma membrane of resting antigen-experienced T cells. Nat Immunol 2018; 19:821-827. [PMID: 30013143 PMCID: PMC6071872 DOI: 10.1038/s41590-018-0162-7] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 06/14/2018] [Indexed: 12/29/2022]
Abstract
The main function of T cells is to identify harmful antigens as quickly and precisely as possible. Super-resolution microscopy data have indicated that global clustering of T cell antigen receptors (TCRs) occurs before T cell activation. Such pre-activation clustering has been interpreted as representing a potential regulatory mechanism that fine tunes the T cell response. We found here that apparent TCR nanoclustering could be attributed to overcounting artifacts inherent to single-molecule-localization microscopy. Using complementary super-resolution approaches and statistical image analysis, we found no indication of global nanoclustering of TCRs on antigen-experienced CD4+ T cells under non-activating conditions. We also used extensive simulations of super-resolution images to provide quantitative limits for the degree of randomness of the TCR distribution. Together our results suggest that the distribution of TCRs on the plasma membrane is optimized for fast recognition of antigen in the first phase of T cell activation.
Collapse
Affiliation(s)
| | | | - Haisen Ta
- Department of NanoBiophotonics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - René Platzer
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Florian Kellner
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Johannes B Huppa
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | | | | | | |
Collapse
|
170
|
Peters R, Griffié J, Burn GL, Williamson DJ, Owen DM. Quantitative fibre analysis of single-molecule localization microscopy data. Sci Rep 2018; 8:10418. [PMID: 29991683 PMCID: PMC6039472 DOI: 10.1038/s41598-018-28691-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 06/21/2018] [Indexed: 11/18/2022] Open
Abstract
Single molecule localization microscopy (SMLM) methods produce data in the form of a spatial point pattern (SPP) of all localized emitters. Whilst numerous tools exist to quantify molecular clustering in SPP data, the analysis of fibrous structures has remained understudied. Taking the SMLM localization coordinates as input, we present an algorithm capable of tracing fibrous structures in data generated by SMLM. Based upon a density parameter tracing routine, the algorithm outputs several fibre descriptors, such as number of fibres, length of fibres, area of enclosed regions and locations and angles of fibre branch points. The method is validated in a variety of simulated conditions and experimental data acquired using the image reconstruction by integrating exchangeable single-molecule localization (IRIS) technique. For this, the nanoscale architecture of F-actin at the T cell immunological synapse in both untreated and pharmacologically treated cells, designed to perturb actin structure, was analysed.
Collapse
Affiliation(s)
- Ruby Peters
- Department of Physics and Randall Division of Cell and Molecular Biophysics, King's College London, London, UK.
| | - Juliette Griffié
- Department of Physics and Randall Division of Cell and Molecular Biophysics, King's College London, London, UK
| | - Garth L Burn
- Cellular Microbiology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - David J Williamson
- Department of Physics and Randall Division of Cell and Molecular Biophysics, King's College London, London, UK
| | - Dylan M Owen
- Department of Physics and Randall Division of Cell and Molecular Biophysics, King's College London, London, UK
| |
Collapse
|
171
|
Roobala C, Ilanila IP, Basu JK. Applications of STED fluorescence nanoscopy in unravelling nanoscale structure and dynamics of biological systems. J Biosci 2018; 43:471-484. [PMID: 30002267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Fluorescence microscopy, especially confocal microscopy, has revolutionized the field of biological imaging. Breaking the optical diffraction barrier of conventional light microscopy, through the advent of super-resolution microscopy, has ushered in the potential for a second revolution through unprecedented insight into nanoscale structure and dynamics in biological systems. Stimulated emission depletion (STED) microscopy is one such super-resolution microscopy technique which provides real-time enhanced-resolution imaging capabilities. In addition, it can be easily integrated with well-established fluorescence-based techniques such as fluorescence correlation spectroscopy (FCS) in order to capture the structure of cellular membranes at the nanoscale with high temporal resolution. In this review, we discuss the theory of STED and different modalities of operation in order to achieve the best resolution. Various applications of this technique in cell imaging, especially that of neuronal cell imaging, are discussed as well as examples of application of STED imaging in unravelling structure formation on biological membranes. Finally, we have discussed examples from some of our recent studies on nanoscale structure and dynamics of lipids in model membranes, due to interaction with proteins, as revealed by combination of STED and FCS techniques.
Collapse
Affiliation(s)
- C Roobala
- Department of Physics, Indian Institute of Science, Bengaluru 560 012, India
| | | | | |
Collapse
|
172
|
Baddeley D, Bewersdorf J. Biological Insight from Super-Resolution Microscopy: What We Can Learn from Localization-Based Images. Annu Rev Biochem 2018; 87:965-989. [PMID: 29272143 DOI: 10.1146/annurev-biochem-060815-014801] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Super-resolution optical imaging based on the switching and localization of individual fluorescent molecules [photoactivated localization microscopy (PALM), stochastic optical reconstruction microscopy (STORM), etc.] has evolved remarkably over the last decade. Originally driven by pushing technological limits, it has become a tool of biological discovery. The initial demand for impressive pictures showing well-studied biological structures has been replaced by a need for quantitative, reliable data providing dependable evidence for specific unresolved biological hypotheses. In this review, we highlight applications that showcase this development, identify the features that led to their success, and discuss remaining challenges and difficulties. In this context, we consider the complex topic of defining resolution for this imaging modality and address some of the more common analytical methods used with this data.
Collapse
Affiliation(s)
- David Baddeley
- Department of Cell Biology, Yale School of Medicine, New Haven, Connecticut 06520, USA; , .,Auckland Bioengineering Institute, University of Auckland, Auckland 1010, New Zealand
| | - Joerg Bewersdorf
- Department of Cell Biology, Yale School of Medicine, New Haven, Connecticut 06520, USA; , .,Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06520, USA
| |
Collapse
|
173
|
Khater IM, Meng F, Wong TH, Nabi IR, Hamarneh G. Super Resolution Network Analysis Defines the Molecular Architecture of Caveolae and Caveolin-1 Scaffolds. Sci Rep 2018; 8:9009. [PMID: 29899348 PMCID: PMC5998020 DOI: 10.1038/s41598-018-27216-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 05/24/2018] [Indexed: 12/04/2022] Open
Abstract
Quantitative approaches to analyze the large data sets generated by single molecule localization super-resolution microscopy (SMLM) are limited. We developed a computational pipeline and applied it to analyzing 3D point clouds of SMLM localizations (event lists) of the caveolar coat protein, caveolin-1 (Cav1), in prostate cancer cells differentially expressing CAVIN1 (also known as PTRF), that is also required for caveolae formation. High degree (strongly-interacting) points were removed by an iterative blink merging algorithm and Cav1 network properties were compared with randomly generated networks to retain a sub-network of geometric structures (or blobs). Machine-learning based classification extracted 28 quantitative features describing the size, shape, topology and network characteristics of ∼80,000 blobs. Unsupervised clustering identified small S1A scaffolds corresponding to SDS-resistant Cav1 oligomers, as yet undescribed larger hemi-spherical S2 scaffolds and, only in CAVIN1-expressing cells, spherical, hollow caveolae. Multi-threshold modularity analysis suggests that S1A scaffolds interact to form larger scaffolds and that S1A dimers group together, in the presence of CAVIN1, to form the caveolae coat.
Collapse
Affiliation(s)
- Ismail M Khater
- Medical Image Analysis Lab, School of Computing Science, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Fanrui Meng
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Timothy H Wong
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Ivan Robert Nabi
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| | - Ghassan Hamarneh
- Medical Image Analysis Lab, School of Computing Science, Simon Fraser University, Burnaby, BC V5A 1S6, Canada.
| |
Collapse
|
174
|
Rogacki MK, Golfetto O, Tobin SJ, Li T, Biswas S, Jorand R, Zhang H, Radoi V, Ming Y, Svenningsson P, Ganjali D, Wakefield DL, Sideris A, Small AR, Terenius L, Jovanović‐Talisman T, Vukojević V. Dynamic lateral organization of opioid receptors (kappa, mu wt and mu N40D ) in the plasma membrane at the nanoscale level. Traffic 2018; 19:690-709. [PMID: 29808515 PMCID: PMC6120469 DOI: 10.1111/tra.12582] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 05/06/2018] [Accepted: 05/18/2018] [Indexed: 02/06/2023]
Abstract
Opioid receptors are important pharmacological targets for the management of numerous medical conditions (eg, severe pain), but they are also the gateway to the development of deleterious side effects (eg, opiate addiction). Opioid receptor signaling cascades are well characterized. However, quantitative information regarding their lateral dynamics and nanoscale organization in the plasma membrane remains limited. Since these dynamic properties are important determinants of receptor function, it is crucial to define them. Herein, the nanoscale lateral dynamics and spatial organization of kappa opioid receptor (KOP), wild type mu opioid receptor (MOPwt ), and its naturally occurring isoform (MOPN40D ) were quantitatively characterized using fluorescence correlation spectroscopy and photoactivated localization microscopy. Obtained results, supported by ensemble-averaged Monte Carlo simulations, indicate that these opioid receptors dynamically partition into different domains. In particular, significant exclusion from GM1 ganglioside-enriched domains and partial association with cholesterol-enriched domains was observed. Nanodomain size, receptor population density and the fraction of receptors residing outside of nanodomains were receptor-specific. KOP-containing domains were the largest and most densely populated, with the smallest fraction of molecules residing outside of nanodomains. The opposite was true for MOPN40D . Moreover, cholesterol depletion dynamically regulated the partitioning of KOP and MOPwt , whereas this effect was not observed for MOPN40D .
Collapse
Affiliation(s)
- Maciej K. Rogacki
- Department of Clinical NeuroscienceCenter for Molecular Medicine, Karolinska InstituteStockholmSweden
| | - Ottavia Golfetto
- Department of Molecular Medicine, Beckman Research Institute, City of HopeDuarteCalifornia
| | - Steven J. Tobin
- Department of Molecular Medicine, Beckman Research Institute, City of HopeDuarteCalifornia
| | - Tianyi Li
- Department of Clinical NeuroscienceCenter for Molecular Medicine, Karolinska InstituteStockholmSweden
| | - Sunetra Biswas
- Department of Molecular Medicine, Beckman Research Institute, City of HopeDuarteCalifornia
| | - Raphael Jorand
- Department of Molecular Medicine, Beckman Research Institute, City of HopeDuarteCalifornia
| | - Huiying Zhang
- Department of Molecular Medicine, Beckman Research Institute, City of HopeDuarteCalifornia
| | - Vlad Radoi
- Department of Clinical NeuroscienceCenter for Molecular Medicine, Karolinska InstituteStockholmSweden
| | - Yu Ming
- Department of Clinical NeuroscienceCenter for Molecular Medicine, Karolinska InstituteStockholmSweden
| | - Per Svenningsson
- Department of Clinical NeuroscienceCenter for Molecular Medicine, Karolinska InstituteStockholmSweden
| | - Daniel Ganjali
- Department of Mechanical and Aerospace EngineeringThe Henry Samueli School of Engineering, University of CaliforniaIrvineCalifornia
| | - Devin L. Wakefield
- Department of Molecular Medicine, Beckman Research Institute, City of HopeDuarteCalifornia
| | - Athanasios Sideris
- Department of Mechanical and Aerospace EngineeringThe Henry Samueli School of Engineering, University of CaliforniaIrvineCalifornia
| | - Alexander R. Small
- Department of Physics and AstronomyCalifornia State Polytechnic UniversityPomonaCalifornia
| | - Lars Terenius
- Department of Clinical NeuroscienceCenter for Molecular Medicine, Karolinska InstituteStockholmSweden
- Department of Molecular and Cellular NeurosciencesThe Scripps Research InstituteLa JollaCalifornia
| | | | - Vladana Vukojević
- Department of Clinical NeuroscienceCenter for Molecular Medicine, Karolinska InstituteStockholmSweden
| |
Collapse
|
175
|
Applications of STED fluorescence nanoscopy in unravelling nanoscale structure and dynamics of biological systems. J Biosci 2018. [DOI: 10.1007/s12038-018-9764-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
176
|
Harwardt MLIE, Dietz MS, Heilemann M, Wohland T. SPT and Imaging FCS Provide Complementary Information on the Dynamics of Plasma Membrane Molecules. Biophys J 2018; 114:2432-2443. [PMID: 29650369 PMCID: PMC6129459 DOI: 10.1016/j.bpj.2018.03.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 02/10/2018] [Accepted: 03/12/2018] [Indexed: 12/27/2022] Open
Abstract
The dynamics of biomolecules in the plasma membrane is of fundamental importance to understanding cellular processes. Cellular signaling often starts with extracellular ligand binding to a membrane receptor, which then transduces an intracellular signal. Ligand binding and receptor-complex activation often involve a complex rearrangement of proteins in the membrane, which results in changes in diffusion properties. Two widely used methods to characterize biomolecular diffusion are single-particle tracking (SPT) and imaging total internal reflection fluorescence correlation spectroscopy (ITIR-FCS). Here, we compare the results of recovered diffusion coefficients and mean-square displacements of the two methods by simulations of free, domain-confined, or meshwork diffusion. We introduce, to our knowledge, a new method for the determination of confinement radii from ITIR-FCS data. We further establish and demonstrate simultaneous SPT/ITIR-FCS for direct comparison within living cells. Finally, we compare the results obtained by SPT and ITIR-FCS for the receptor tyrosine kinase MET. Our results show that SPT and ITIR-FCS yield complementary information on diffusion properties of biomolecules in cell membranes.
Collapse
Affiliation(s)
- Marie-Lena I E Harwardt
- Institute of Physical and Theoretical Chemistry, Johann Wolfgang Goethe-University, Frankfurt, Germany
| | - Marina S Dietz
- Institute of Physical and Theoretical Chemistry, Johann Wolfgang Goethe-University, Frankfurt, Germany
| | - Mike Heilemann
- Institute of Physical and Theoretical Chemistry, Johann Wolfgang Goethe-University, Frankfurt, Germany.
| | - Thorsten Wohland
- Departments of Biological Sciences and Chemistry, Center for Bioimaging Sciences, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
177
|
Fülöp G, Brameshuber M, Arnold AM, Schütz GJ, Sevcsik E. Determination of the Membrane Environment of CD59 in Living Cells. Biomolecules 2018; 8:E28. [PMID: 29772810 PMCID: PMC6023084 DOI: 10.3390/biom8020028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 04/24/2018] [Accepted: 05/14/2018] [Indexed: 11/24/2022] Open
Abstract
The organization and dynamics of proteins and lipids in the plasma membrane, and their role in membrane functionality, have been subject of a long-lasting debate. Specifically, it is unclear to what extent membrane proteins are affected by their immediate lipid environment and vice versa. Studies on model membranes and plasma membrane vesicles indicated preferences of proteins for lipid phases characterized by different acyl chain order; however, whether such phases do indeed exist in live cells is still not known. Here, we refine a previously developed micropatterning approach combined with single molecule tracking to quantify the influence of the glycosylphosphatidylinositol-anchored (GPI-anchored) protein CD59 on its molecular environment directly in the live cell plasma membrane. We find that locally enriched and immobilized CD59 presents obstacles to the diffusion of fluorescently labeled lipids with a different phase-partitioning behavior independent of cell cholesterol levels and type of lipid. Our results give no evidence for either specific binding of the lipids to CD59 or the existence of nanoscopic ordered membrane regions associated with CD59.
Collapse
Affiliation(s)
- Gergő Fülöp
- Institute of Applied Physics, TU Wien, Wiedner Hauptstrasse 8-10, Vienna 1040, Austria.
| | - Mario Brameshuber
- Institute of Applied Physics, TU Wien, Wiedner Hauptstrasse 8-10, Vienna 1040, Austria.
| | - Andreas M Arnold
- Institute of Applied Physics, TU Wien, Wiedner Hauptstrasse 8-10, Vienna 1040, Austria.
| | - Gerhard J Schütz
- Institute of Applied Physics, TU Wien, Wiedner Hauptstrasse 8-10, Vienna 1040, Austria.
| | - Eva Sevcsik
- Institute of Applied Physics, TU Wien, Wiedner Hauptstrasse 8-10, Vienna 1040, Austria.
| |
Collapse
|
178
|
qSR: a quantitative super-resolution analysis tool reveals the cell-cycle dependent organization of RNA Polymerase I in live human cells. Sci Rep 2018; 8:7424. [PMID: 29743503 PMCID: PMC5943247 DOI: 10.1038/s41598-018-25454-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 04/13/2018] [Indexed: 12/31/2022] Open
Abstract
We present qSR, an analytical tool for the quantitative analysis of single molecule based super-resolution data. The software is created as an open-source platform integrating multiple algorithms for rigorous spatial and temporal characterizations of protein clusters in super-resolution data of living cells. First, we illustrate qSR using a sample live cell data of RNA Polymerase II (Pol II) as an example of highly dynamic sub-diffractive clusters. Then we utilize qSR to investigate the organization and dynamics of endogenous RNA Polymerase I (Pol I) in live human cells, throughout the cell cycle. Our analysis reveals a previously uncharacterized transient clustering of Pol I. Both stable and transient populations of Pol I clusters co-exist in individual living cells, and their relative fraction vary during cell cycle, in a manner correlating with global gene expression. Thus, qSR serves to facilitate the study of protein organization and dynamics with very high spatial and temporal resolutions directly in live cell.
Collapse
|
179
|
Bernhem K, Blom H, Brismar H. Quantification of endogenous and exogenous protein expressions of Na,K-ATPase with super-resolution PALM/STORM imaging. PLoS One 2018; 13:e0195825. [PMID: 29694368 PMCID: PMC5918999 DOI: 10.1371/journal.pone.0195825] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 03/30/2018] [Indexed: 01/19/2023] Open
Abstract
Transient transfection of fluorescent fusion proteins is a key enabling technology in fluorescent microscopy to spatio-temporally map cellular protein distributions. Transient transfection of proteins may however bypass normal regulation of expression, leading to overexpression artefacts like misallocations and excess amounts. In this study we investigate the use of STORM and PALM microscopy to quantitatively monitor endogenous and exogenous protein expression. Through incorporation of an N-terminal hemagglutinin epitope to a mMaple3 fused Na,K-ATPase (α1 isoform), we analyze the spatial and quantitative changes of plasma membrane Na,K-ATPase localization during competitive transient expression. Quantification of plasma membrane protein density revealed a time dependent increase of Na,K-ATPase, but no increase in size of protein clusters. Results show that after 41h transfection, the total plasma membrane density of Na,K-ATPase increased by 63% while the endogenous contribution was reduced by 16%.
Collapse
Affiliation(s)
- Kristoffer Bernhem
- Science for Life Laboratory, Department of Applied Physics, Royal Institute of Technology, Stockholm, Sweden
| | - Hans Blom
- Science for Life Laboratory, Department of Applied Physics, Royal Institute of Technology, Stockholm, Sweden
| | - Hjalmar Brismar
- Science for Life Laboratory, Department of Applied Physics, Royal Institute of Technology, Stockholm, Sweden
- Science for Life Laboratory, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
180
|
James CC, Zeitz MJ, Calhoun PJ, Lamouille S, Smyth JW. Altered translation initiation of Gja1 limits gap junction formation during epithelial-mesenchymal transition. Mol Biol Cell 2018; 29:797-808. [PMID: 29467255 PMCID: PMC5905293 DOI: 10.1091/mbc.e17-06-0406] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is activated during development, wound healing, and pathologies including fibrosis and cancer metastasis. Hallmarks of EMT are remodeling of intercellular junctions and adhesion proteins, including gap junctions. The GJA1 mRNA transcript encoding the gap junction protein connexin43 (Cx43) has been demonstrated to undergo internal translation initiation, yielding truncated isoforms that modulate gap junctions. The PI3K/Akt/mTOR pathway is central to translation regulation and is activated during EMT, leading us to hypothesize that altered translation initiation would contribute to gap junction loss. Using TGF-β-induced EMT as a model, we find reductions in Cx43 gap junctions despite increased transcription and stabilization of Cx43 protein. Biochemical experiments reveal suppression of the internally translated Cx43 isoform, GJA1-20k in a Smad3 and ERK-dependent manner. Ectopic expression of GJA1-20k does not halt EMT, but is sufficient to rescue gap junction formation. GJA1-20k localizes to the Golgi apparatus, and using superresolution localization microscopy we find retention of GJA1-43k at the Golgi in mesenchymal cells lacking GJA1-20k. NativePAGE demonstrates that levels of GJA1-20k regulate GJA1-43k hexamer oligomerization, a limiting step in Cx43 trafficking. These findings reveal alterations in translation initiation as an unexplored mechanism by which the cell regulates Cx43 gap junction formation during EMT.
Collapse
Affiliation(s)
- Carissa C James
- Virginia Tech Carilion Research Institute and School of Medicine, Roanoke, VA 24016.,Graduate Program in Translational Biology, Medicine, and Health, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061
| | - Michael J Zeitz
- Virginia Tech Carilion Research Institute and School of Medicine, Roanoke, VA 24016
| | - Patrick J Calhoun
- Virginia Tech Carilion Research Institute and School of Medicine, Roanoke, VA 24016.,Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061
| | - Samy Lamouille
- Virginia Tech Carilion Research Institute and School of Medicine, Roanoke, VA 24016
| | - James W Smyth
- Virginia Tech Carilion Research Institute and School of Medicine, Roanoke, VA 24016.,Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061
| |
Collapse
|
181
|
Schnitzbauer J, Wang Y, Zhao S, Bakalar M, Nuwal T, Chen B, Huang B. Correlation analysis framework for localization-based superresolution microscopy. Proc Natl Acad Sci U S A 2018; 115:3219-3224. [PMID: 29531072 PMCID: PMC5879654 DOI: 10.1073/pnas.1711314115] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Superresolution images reconstructed from single-molecule localizations can reveal cellular structures close to the macromolecular scale and are now being used routinely in many biomedical research applications. However, because of their coordinate-based representation, a widely applicable and unified analysis platform that can extract a quantitative description and biophysical parameters from these images is yet to be established. Here, we propose a conceptual framework for correlation analysis of coordinate-based superresolution images using distance histograms. We demonstrate the application of this concept in multiple scenarios, including image alignment, tracking of diffusing molecules, as well as for quantification of colocalization, showing its superior performance over existing approaches.
Collapse
Affiliation(s)
- Joerg Schnitzbauer
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94143
| | - Yina Wang
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94143
| | - Shijie Zhao
- School of Life Sciences, Peking University, Beijing 100871, China
| | - Matthew Bakalar
- UC Berkeley-UCSF Joint Graduate Group in Bioengineering, University of California, Berkeley, CA 94720
| | - Tulip Nuwal
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94143
| | - Baohui Chen
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94143
| | - Bo Huang
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94143;
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94143
- Chan Zuckerberg Biohub, San Francisco, CA 94158
| |
Collapse
|
182
|
Wang H, Feng Z, Del Signore SJ, Rodal AA, Xu B. Active Probes for Imaging Membrane Dynamics of Live Cells with High Spatial and Temporal Resolution over Extended Time Scales and Areas. J Am Chem Soc 2018; 140:3505-3509. [PMID: 29481071 PMCID: PMC5858877 DOI: 10.1021/jacs.7b13307] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Despite the advancement of molecular imaging techniques, there is an unmet need for probes for direct imaging of membrane dynamics of live cells. Here we report a novel type of active (or enzyme responsive) probes to directly image membrane dynamics of live cells with high spatial and temporal resolution over extended time scales and areas. Because lipid rafts enrich cholesterols and GPI-anchored enzymes (e.g., ectophosphatases), we design probes that consist of an enzymatic trigger, a fluorophore, and a cholesterol that are affinitive to the cell membrane. Being water-soluble and as the substrate of ectophosphatase, these cell compatible probes preferentially and rapidly assemble in plasma membrane, exhibit strong fluorescence, work at micromolar concentrations, and easily achieve high resolution monitoring of nanoscale heterogeneity in membranes of live cells, the release of exosomes, and the membrane dynamics of live cells. This work provides a facile means to link membrane dynamics and heterogeneity to cellular processes for understanding the interactions between membranes and proteins.
Collapse
Affiliation(s)
- Huaimin Wang
- Department of Chemistry , Brandeis University , 415 South Street , Waltham , Massachusetts 02453 , United States
| | - Zhaoqianqi Feng
- Department of Chemistry , Brandeis University , 415 South Street , Waltham , Massachusetts 02453 , United States
| | - Steven J Del Signore
- Department of Biology , Brandeis University , 415 South Street , Waltham , Massachusetts 02454 , United States
| | - Avital A Rodal
- Department of Biology , Brandeis University , 415 South Street , Waltham , Massachusetts 02454 , United States
| | - Bing Xu
- Department of Chemistry , Brandeis University , 415 South Street , Waltham , Massachusetts 02453 , United States
| |
Collapse
|
183
|
Zhang F, Jing W, Hunt A, Yu H, Yang Y, Wang S, Chen HY, Tao N. Label-Free Quantification of Small-Molecule Binding to Membrane Proteins on Single Cells by Tracking Nanometer-Scale Cellular Membrane Deformation. ACS NANO 2018; 12:2056-2064. [PMID: 29397682 PMCID: PMC5851003 DOI: 10.1021/acsnano.8b00235] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Measuring molecular binding to membrane proteins is critical for understanding cellular functions, validating biomarkers, and screening drugs. Despite the importance, developing such a capability has been a difficult challenge, especially for small-molecule binding to membrane proteins in their native cellular environment. Here we show that the binding of both large and small molecules to membrane proteins can be quantified on single cells by trapping single cells with a microfluidic device and detecting binding-induced cellular membrane deformation on the nanometer scale with label-free optical imaging. We develop a thermodynamic model to describe the binding-induced membrane deformation, validate the model by examining the dependence of membrane deformation on cell stiffness, membrane protein expression level, and binding affinity, and study four major types of membrane proteins, including glycoproteins, ion channels, G-protein coupled receptors, and tyrosine kinase receptors. The single-cell detection capability reveals the importance of local membrane environment on molecular binding and variability in the binding kinetics of different cell lines and heterogeneity of different cells within the same cell line.
Collapse
Affiliation(s)
- Fenni Zhang
- Center for Bioelectronics and Biosensors, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
- School of Electrical Computer and Energy Engineering, Arizona State University, Tempe, Arizona 85287, USA
| | - Wenwen Jing
- Center for Bioelectronics and Biosensors, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Ashley Hunt
- Center for Bioelectronics and Biosensors, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Hui Yu
- Center for Bioelectronics and Biosensors, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Yunze Yang
- Center for Bioelectronics and Biosensors, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
- School of Electrical Computer and Energy Engineering, Arizona State University, Tempe, Arizona 85287, USA
| | - Shaopeng Wang
- Center for Bioelectronics and Biosensors, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Hong-Yuan Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210093, China
| | - Nongjian Tao
- Center for Bioelectronics and Biosensors, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210093, China
- School of Electrical Computer and Energy Engineering, Arizona State University, Tempe, Arizona 85287, USA
| |
Collapse
|
184
|
Lu SM, Fairn GD. Mesoscale organization of domains in the plasma membrane - beyond the lipid raft. Crit Rev Biochem Mol Biol 2018; 53:192-207. [PMID: 29457544 DOI: 10.1080/10409238.2018.1436515] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The plasma membrane is compartmentalized into several distinct regions or domains, which show a broad diversity in both size and lifetime. The segregation of lipids and membrane proteins is thought to be driven by the lipid composition itself, lipid-protein interactions and diffusional barriers. With regards to the lipid composition, the immiscibility of certain classes of lipids underlies the "lipid raft" concept of plasmalemmal compartmentalization. Historically, lipid rafts have been described as cholesterol and (glyco)sphingolipid-rich regions of the plasma membrane that exist as a liquid-ordered phase that are resistant to extraction with non-ionic detergents. Over the years the interest in lipid rafts grew as did the challenges with studying these nanodomains. The term lipid raft has fallen out of favor with many scientists and instead the terms "membrane raft" or "membrane nanodomain" are preferred as they connote the heterogeneity and dynamic nature of the lipid-protein assemblies. In this article, we will discuss the classical lipid raft hypothesis and its limitations. This review will also discuss alternative models of lipid-protein interactions, annular lipid shells, and larger membrane clusters. We will also discuss the mesoscale organization of plasmalemmal domains including visible structures such as clathrin-coated pits and caveolae.
Collapse
Affiliation(s)
- Stella M Lu
- a Keenan Research Centre for Biomedical Science, St. Michael's Hospital , Toronto , Canada.,b Department of Biochemistry , University of Toronto , Toronto , Canada
| | - Gregory D Fairn
- a Keenan Research Centre for Biomedical Science, St. Michael's Hospital , Toronto , Canada.,b Department of Biochemistry , University of Toronto , Toronto , Canada.,c Department of Surgery , University of Toronto , Toronto , Canada
| |
Collapse
|
185
|
Caldieri G, Barbieri E, Nappo G, Raimondi A, Bonora M, Conte A, Verhoef LGGC, Confalonieri S, Malabarba MG, Bianchi F, Cuomo A, Bonaldi T, Martini E, Mazza D, Pinton P, Tacchetti C, Polo S, Di Fiore PP, Sigismund S. Reticulon 3-dependent ER-PM contact sites control EGFR nonclathrin endocytosis. Science 2018; 356:617-624. [PMID: 28495747 DOI: 10.1126/science.aah6152] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 02/21/2017] [Accepted: 03/24/2017] [Indexed: 11/02/2022]
Abstract
The integration of endocytic routes is critical to regulate receptor signaling. A nonclathrin endocytic (NCE) pathway of the epidermal growth factor receptor (EGFR) is activated at high ligand concentrations and targets receptors to degradation, attenuating signaling. Here we performed an unbiased molecular characterization of EGFR-NCE. We identified NCE-specific regulators, including the endoplasmic reticulum (ER)-resident protein reticulon 3 (RTN3) and a specific cargo, CD147. RTN3 was critical for EGFR/CD147-NCE, promoting the creation of plasma membrane (PM)-ER contact sites that were required for the formation and/or maturation of NCE invaginations. Ca2+ release at these sites, triggered by inositol 1,4,5-trisphosphate (IP3)-dependent activation of ER Ca2+ channels, was needed for the completion of EGFR internalization. Thus, we identified a mechanism of EGFR endocytosis that relies on ER-PM contact sites and local Ca2+ signaling.
Collapse
Affiliation(s)
- Giusi Caldieri
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Via Adamello 16, 20139 Milan, Italy
| | - Elisa Barbieri
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Via Adamello 16, 20139 Milan, Italy
| | - Gilda Nappo
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Via Adamello 16, 20139 Milan, Italy
| | - Andrea Raimondi
- Centro Imaging Sperimentale, Istituto Scientifico San Raffaele, Via Olgettina 52, 20132 Milan, Italy
| | - Massimo Bonora
- Section of Pathology, Oncology and Experimental Biology and Laboratory for Technologies of Advanced Therapies Center, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Alexia Conte
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Via Adamello 16, 20139 Milan, Italy
| | - Lisette G G C Verhoef
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Via Adamello 16, 20139 Milan, Italy
| | - Stefano Confalonieri
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Via Adamello 16, 20139 Milan, Italy
| | - Maria Grazia Malabarba
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Via Adamello 16, 20139 Milan, Italy.,Dipartimento di Oncologia ed Emato-Oncologia (DiPO)-Università degli Studi di Milano, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Fabrizio Bianchi
- Istituto Europeo di Oncologia, Via Ripamonti 435, 20141 Milan, Italy
| | - Alessandro Cuomo
- Istituto Europeo di Oncologia, Via Ripamonti 435, 20141 Milan, Italy
| | - Tiziana Bonaldi
- Istituto Europeo di Oncologia, Via Ripamonti 435, 20141 Milan, Italy
| | - Emanuele Martini
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Via Adamello 16, 20139 Milan, Italy
| | - Davide Mazza
- Centro Imaging Sperimentale, Istituto Scientifico San Raffaele, Via Olgettina 52, 20132 Milan, Italy
| | - Paolo Pinton
- Section of Pathology, Oncology and Experimental Biology and Laboratory for Technologies of Advanced Therapies Center, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Carlo Tacchetti
- Centro Imaging Sperimentale, Istituto Scientifico San Raffaele, Via Olgettina 52, 20132 Milan, Italy.,Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, Genoa, Italy
| | - Simona Polo
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Via Adamello 16, 20139 Milan, Italy.,Dipartimento di Oncologia ed Emato-Oncologia (DiPO)-Università degli Studi di Milano, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Pier Paolo Di Fiore
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Via Adamello 16, 20139 Milan, Italy. .,Dipartimento di Oncologia ed Emato-Oncologia (DiPO)-Università degli Studi di Milano, Via Festa del Perdono 7, 20122 Milan, Italy.,Istituto Europeo di Oncologia, Via Ripamonti 435, 20141 Milan, Italy
| | - Sara Sigismund
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Via Adamello 16, 20139 Milan, Italy.
| |
Collapse
|
186
|
Siebert S, Ickstadt K, Schäfer M, Radon Y, Verveer PJ. Comparison of clustering approaches with application to dual colour protein data. IET Syst Biol 2018; 12:7-17. [PMID: 29337285 PMCID: PMC8687232 DOI: 10.1049/iet-syb.2017.0019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Cells communicate with their environment via proteins, located at the plasma membrane separating the interior of a cell from its surroundings. The spatial distribution of these proteins in the plasma membrane under different physiological conditions is of importance, since this may influence their signal transmission properties. In this study, the authors compare different methods such as hierarchical clustering, extensible Markov models and the gammics method for analysing such a spatial distribution. The methods are examined in a simulation study to determine their optimal use. Afterwards, they analyse experimental imaging data and extend these methods to simulate dual colour data.
Collapse
Affiliation(s)
- Sabrina Siebert
- Faculty of Statistics, TU Dortmund University, Dortmund, Germany.
| | - Katja Ickstadt
- Faculty of Statistics, TU Dortmund University, Dortmund, Germany
| | - Martin Schäfer
- Chair of Mathematical Optimization, Mathematical Institute, Heinrich Heine University, Düsseldorf, Germany
| | - Yvonne Radon
- Max-Planck-Institute Dortmund, Dortmund, Germany
| | | |
Collapse
|
187
|
Pan L, Yan R, Li W, Xu K. Super-Resolution Microscopy Reveals the Native Ultrastructure of the Erythrocyte Cytoskeleton. Cell Rep 2018; 22:1151-1158. [DOI: 10.1016/j.celrep.2017.12.107] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Revised: 11/23/2017] [Accepted: 12/29/2017] [Indexed: 11/30/2022] Open
|
188
|
Renz M, Wunder C. Internal rulers to assess fluorescent protein photoactivation efficiency. Cytometry A 2017; 93:411-419. [DOI: 10.1002/cyto.a.23319] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 11/08/2017] [Accepted: 12/12/2017] [Indexed: 11/09/2022]
Affiliation(s)
- Malte Renz
- Division of Gynecologic OncologyStanford University School of Medicine, 300 Pasteur Drive, Stanford California 94305
| | - Christian Wunder
- Institut Curie, PSL Research University, Cellular and Chemical Biology, INSERM U 1143, CNRS UMR 3666, 26 rue d'Ulm75248 Paris Cedex 05 France
| |
Collapse
|
189
|
Sakamoto H, Ariyoshi T, Kimpara N, Sugao K, Taiko I, Takikawa K, Asanuma D, Namiki S, Hirose K. Synaptic weight set by Munc13-1 supramolecular assemblies. Nat Neurosci 2017; 21:41-49. [DOI: 10.1038/s41593-017-0041-9] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 11/07/2017] [Indexed: 01/03/2023]
|
190
|
Deschout H, Platzman I, Sage D, Feletti L, Spatz JP, Radenovic A. Investigating Focal Adhesion Substructures by Localization Microscopy. Biophys J 2017; 113:2508-2518. [PMID: 29212004 PMCID: PMC5768516 DOI: 10.1016/j.bpj.2017.09.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 09/04/2017] [Accepted: 09/29/2017] [Indexed: 11/22/2022] Open
Abstract
Cells rely on focal adhesions (FAs) to carry out a variety of important tasks, including motion, environmental sensing, and adhesion to the extracellular matrix. Although attaining a fundamental characterization of FAs is a compelling goal, their extensive complexity and small size, which can be below the diffraction limit, have hindered a full understanding. In this study we have used single-molecule localization microscopy (SMLM) to investigate integrin β3 and paxillin in rat embryonic fibroblasts growing on two different extracellular matrix-representing substrates (i.e., fibronectin-coated substrates and specifically biofunctionalized nanopatterned substrates). To quantify the substructure of FAs, we developed a clustering method based on expectation maximization of a Gaussian mixture that accounts for localization uncertainty and background. Analysis of our SMLM data indicates that the structures within FAs, characterized as a Gaussian mixture, typically have areas between 0.01 and 1 μm2, contain 10-100 localizations, and can exhibit substantial eccentricity. Our approach based on SMLM opens new avenues for studying structural and functional biology of molecular assemblies that display substantial varieties in size, shape, and density.
Collapse
Affiliation(s)
- Hendrik Deschout
- Laboratory of Nanoscale Biology, Institute of Bioengineering, School of Engineering, EPFL, Lausanne, Switzerland
| | - Ilia Platzman
- Department of Cellular Biophysics, Max-Planck-Institute for Medical Research and the Department of Biophysical Chemistry, University of Heidelberg, Heidelberg, Germany
| | - Daniel Sage
- Biomedical Imaging Group, School of Engineering, EPFL, Lausanne, Switzerland
| | - Lely Feletti
- Laboratory of Nanoscale Biology, Institute of Bioengineering, School of Engineering, EPFL, Lausanne, Switzerland
| | - Joachim P Spatz
- Department of Cellular Biophysics, Max-Planck-Institute for Medical Research and the Department of Biophysical Chemistry, University of Heidelberg, Heidelberg, Germany
| | - Aleksandra Radenovic
- Laboratory of Nanoscale Biology, Institute of Bioengineering, School of Engineering, EPFL, Lausanne, Switzerland.
| |
Collapse
|
191
|
GPI-anchored proteins are confined in subdiffraction clusters at the apical surface of polarized epithelial cells. Biochem J 2017; 474:4075-4090. [PMID: 29046391 PMCID: PMC5712066 DOI: 10.1042/bcj20170582] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/19/2017] [Accepted: 10/17/2017] [Indexed: 11/28/2022]
Abstract
Spatio-temporal compartmentalization of membrane proteins is critical for the regulation of diverse vital functions in eukaryotic cells. It was previously shown that, at the apical surface of polarized MDCK cells, glycosylphosphatidylinositol (GPI)-anchored proteins (GPI-APs) are organized in small cholesterol-independent clusters of single GPI-AP species (homoclusters), which are required for the formation of larger cholesterol-dependent clusters formed by multiple GPI-AP species (heteroclusters). This clustered organization is crucial for the biological activities of GPI-APs; hence, understanding the spatio-temporal properties of their membrane organization is of fundamental importance. Here, by using direct stochastic optical reconstruction microscopy coupled to pair correlation analysis (pc-STORM), we were able to visualize and measure the size of these clusters. Specifically, we show that they are non-randomly distributed and have an average size of 67 nm. We also demonstrated that polarized MDCK and non-polarized CHO cells have similar cluster distribution and size, but different sensitivity to cholesterol depletion. Finally, we derived a model that allowed a quantitative characterization of the cluster organization of GPI-APs at the apical surface of polarized MDCK cells for the first time. Experimental FRET (fluorescence resonance energy transfer)/FLIM (fluorescence-lifetime imaging microscopy) data were correlated to the theoretical predictions of the model.
Collapse
|
192
|
Dahmke IN, Verch A, Hermannsdörfer J, Peckys DB, Weatherup RS, Hofmann S, de Jonge N. Graphene Liquid Enclosure for Single-Molecule Analysis of Membrane Proteins in Whole Cells Using Electron Microscopy. ACS NANO 2017; 11:11108-11117. [PMID: 29023096 DOI: 10.1021/acsnano.7b05258] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Membrane proteins govern many important functions in cells via dynamic oligomerization into active complexes. However, analytical methods to study their distribution and functional state in relation to the cellular structure are currently limited. Here, we introduce a technique for studying single-membrane proteins within their native context of the intact plasma membrane. SKBR3 breast cancer cells were grown on silicon microchips with thin silicon nitride windows. The cells were fixed, and the epidermal growth factor receptor ErbB2 was specifically labeled with quantum dot (QD) nanoparticles. For correlative fluorescence- and liquid-phase electron microscopy, we enclosed the liquid samples by chemical vapor deposited (CVD) graphene films. Depending on the local cell thickness, QD labels were imaged with a spatial resolution of 2 nm at a low electron dose. The distribution and stoichiometric assembly of ErbB2 receptors were determined at several different cellular locations, including tunneling nanotubes, where we found higher levels of homodimerization at the connecting sites. This experimental approach is applicable to a wide range of cell lines and membrane proteins and particularly suitable for studies involving both inter- and intracellular heterogeneity in protein distribution and expression.
Collapse
Affiliation(s)
- Indra N Dahmke
- INM - Leibniz Institute for New Materials , D-66123 Saarbrücken, Germany
| | - Andreas Verch
- INM - Leibniz Institute for New Materials , D-66123 Saarbrücken, Germany
| | | | - Diana B Peckys
- Department of Biophysics, Saarland University , D-66421 Homburg, Germany
| | - Robert S Weatherup
- Engineering Department, University of Cambridge , Cambridge CB3 0FA, United Kingdom
| | - Stephan Hofmann
- Engineering Department, University of Cambridge , Cambridge CB3 0FA, United Kingdom
| | - Niels de Jonge
- INM - Leibniz Institute for New Materials , D-66123 Saarbrücken, Germany
- Department of Physics, Saarland University , D-66123 Saarbrücken, Germany
| |
Collapse
|
193
|
Quantifying protein densities on cell membranes using super-resolution optical fluctuation imaging. Nat Commun 2017; 8:1731. [PMID: 29170394 PMCID: PMC5700985 DOI: 10.1038/s41467-017-01857-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 10/20/2017] [Indexed: 12/03/2022] Open
Abstract
Quantitative approaches for characterizing molecular organization of cell membrane molecules under physiological and pathological conditions profit from recently developed super-resolution imaging techniques. Current tools employ statistical algorithms to determine clusters of molecules based on single-molecule localization microscopy (SMLM) data. These approaches are limited by the ability of SMLM techniques to identify and localize molecules in densely populated areas and experimental conditions of sample preparation and image acquisition. We have developed a robust, model-free, quantitative clustering analysis to determine the distribution of membrane molecules that excels in densely labeled areas and is tolerant to various experimental conditions, i.e. multiple-blinking or high blinking rates. The method is based on a TIRF microscope followed by a super-resolution optical fluctuation imaging (SOFI) analysis. The effectiveness and robustness of the method is validated using simulated and experimental data investigating nanoscale distribution of CD4 glycoprotein mutants in the plasma membrane of T cells. The ability to quantify the organization of cell membrane molecules is limited by the density of labeling and experimental conditions. Here, the authors use super-resolution optical fluctuation (SOFI) for molecular density and clustering analyses, and investigate nanoscale distribution of CD4 glycoprotein.
Collapse
|
194
|
Peksel B, Gombos I, Péter M, Vigh L, Tiszlavicz Á, Brameshuber M, Balogh G, Schütz GJ, Horváth I, Vigh L, Török Z. Mild heat induces a distinct "eustress" response in Chinese Hamster Ovary cells but does not induce heat shock protein synthesis. Sci Rep 2017; 7:15643. [PMID: 29142280 PMCID: PMC5688065 DOI: 10.1038/s41598-017-15821-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 11/02/2017] [Indexed: 11/16/2022] Open
Abstract
The current research on cellular heat stress management focuses on the roles of heat shock proteins (HSPs) and the proteostasis network under severe stress conditions. The mild, fever-type stress and the maintenance of membrane homeostasis are less well understood. Herein, we characterized the acute effect of mild, fever-range heat shock on membrane organization, and HSP synthesis and localization in two mammalian cell lines, to delineate the role of membranes in the sensing and adaptation to heat. A multidisciplinary approach combining ultrasensitive fluorescence microscopy and lipidomics revealed the molecular details of novel cellular “eustress”, when cells adapt to mild heat by maintaining membrane homeostasis, activating lipid remodeling, and redistributing chaperone proteins. Notably, this leads to acquired thermotolerance in the complete absence of the induction of HSPs. At higher temperatures, additional defense mechanisms are activated, including elevated expression of molecular chaperones, contributing to an extended stress memory and acquired thermotolerance.
Collapse
Affiliation(s)
- Begüm Peksel
- Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, H-6726, Hungary
| | - Imre Gombos
- Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, H-6726, Hungary
| | - Mária Péter
- Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, H-6726, Hungary
| | - László Vigh
- Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, H-6726, Hungary
| | - Ádám Tiszlavicz
- Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, H-6726, Hungary
| | - Mario Brameshuber
- Institute of Applied Physics - Biophysics, TU Wien, 1040, Vienna, Austria
| | - Gábor Balogh
- Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, H-6726, Hungary
| | - Gerhard J Schütz
- Institute of Applied Physics - Biophysics, TU Wien, 1040, Vienna, Austria
| | - Ibolya Horváth
- Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, H-6726, Hungary
| | - László Vigh
- Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, H-6726, Hungary
| | - Zsolt Török
- Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, H-6726, Hungary.
| |
Collapse
|
195
|
Gao J, He L, Shi Y, Cai M, Xu H, Jiang J, Zhang L, Wang H. Cell contact and pressure control of YAP localization and clustering revealed by super-resolution imaging. NANOSCALE 2017; 9:16993-17003. [PMID: 29082393 DOI: 10.1039/c7nr05818g] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Yes-associated protein (YAP) is well known for being an effecter of the Hippo signaling cascade that plays a critical role in organ size control, tumorigenesis, and regeneration. As YAP is a transcriptional coactivator, nuclear accumulation is a crucial determinant of its function. Numerous investigations have provided insights into the regulation of YAP, such as upstream molecules of the Hippo pathway, cell contact inhibition, and mechanical forces. However, detailed information regarding YAP spatial localization and organization in cells remains uncertain, and how mechanical signals control YAP distribution and function is not fully known. Therefore, we used one of the super-resolution imaging techniques, direct stochastic optical reconstruction microscopy (dSTORM), combined with confocal microscopy, to solve these problems. We found that YAP is mainly distributed in clusters in the cells, and that both cell contact and pressure on cell surfaces promote the nuclear-to-cytoplasm translocation of YAP and its phosphorylation, but weaken the clustering of nuclear YAP and its transcriptional activity. Moreover, we found that pressure regulation may be more effective on YAP from cancer cells as compared to normal cells, which could help open a door to target YAP for anticancer drug design.
Collapse
Affiliation(s)
- Jing Gao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5626 Renmin Street, Changchun, Jilin 130022, P.R. China.
| | | | | | | | | | | | | | | |
Collapse
|
196
|
Feiner-Gracia N, Beck M, Pujals S, Tosi S, Mandal T, Buske C, Linden M, Albertazzi L. Super-Resolution Microscopy Unveils Dynamic Heterogeneities in Nanoparticle Protein Corona. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2017; 13:1701631. [PMID: 28922574 DOI: 10.1002/smll.201701631] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 08/01/2017] [Indexed: 05/18/2023]
Abstract
The adsorption of serum proteins, leading to the formation of a biomolecular corona, is a key determinant of the biological identity of nanoparticles in vivo. Therefore, gaining knowledge on the formation, composition, and temporal evolution of the corona is of utmost importance for the development of nanoparticle-based therapies. Here, it is shown that the use of super-resolution optical microscopy enables the imaging of the protein corona on mesoporous silica nanoparticles with single protein sensitivity. Particle-by-particle quantification reveals a significant heterogeneity in protein absorption under native conditions. Moreover, the diversity of the corona evolves over time depending on the surface chemistry and degradability of the particles. This paper investigates the consequences of protein adsorption for specific cell targeting by antibody-functionalized nanoparticles providing a detailed understanding of corona-activity relations. The methodology is widely applicable to a variety of nanostructures and complements the existing ensemble approaches for protein corona study.
Collapse
Affiliation(s)
- Natalia Feiner-Gracia
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 15-21, 08028, Barcelona, Spain
| | - Michaela Beck
- Inorganic Chemistry II, Ulm University, Albert-Einstein-Allee 11, D-89081, Ulm, Germany
| | - Sílvia Pujals
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 15-21, 08028, Barcelona, Spain
| | - Sébastien Tosi
- Advanced Digital Microscopy Core Facility (ADMCF), Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028, Barcelona, Spain
| | - Tamoghna Mandal
- Institute of Experimental Cancer Research, University Hospital Ulm, Albert-Einstein-Allee 11, D-89081, Ulm, Germany
| | - Christian Buske
- Institute of Experimental Cancer Research, University Hospital Ulm, Albert-Einstein-Allee 11, D-89081, Ulm, Germany
| | - Mika Linden
- Inorganic Chemistry II, Ulm University, Albert-Einstein-Allee 11, D-89081, Ulm, Germany
| | - Lorenzo Albertazzi
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 15-21, 08028, Barcelona, Spain
| |
Collapse
|
197
|
Bermudez-Hernandez K, Keegan S, Whelan DR, Reid DA, Zagelbaum J, Yin Y, Ma S, Rothenberg E, Fenyö D. A Method for Quantifying Molecular Interactions Using Stochastic Modelling and Super-Resolution Microscopy. Sci Rep 2017; 7:14882. [PMID: 29093506 PMCID: PMC5665986 DOI: 10.1038/s41598-017-14922-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 10/19/2017] [Indexed: 02/05/2023] Open
Abstract
We introduce the Interaction Factor (IF), a measure for quantifying the interaction of molecular clusters in super-resolution microscopy images. The IF is robust in the sense that it is independent of cluster density, and it only depends on the extent of the pair-wise interaction between different types of molecular clusters in the image. The IF for a single or a collection of images is estimated by first using stochastic modelling where the locations of clusters in the images are repeatedly randomized to estimate the distribution of the overlaps between the clusters in the absence of interaction (IF = 0). Second, an analytical form of the relationship between IF and the overlap (which has the random overlap as its only parameter) is used to estimate the IF for the experimentally observed overlap. The advantage of IF compared to conventional methods to quantify interaction in microscopy images is that it is insensitive to changing cluster density and is an absolute measure of interaction, making the interpretation of experiments easier. We validate the IF method by using both simulated and experimental data and provide an ImageJ plugin for determining the IF of an image.
Collapse
Affiliation(s)
- Keria Bermudez-Hernandez
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
- Institute for Systems Genetics, New York University School of Medicine, New York, NY, USA
| | - Sarah Keegan
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
- Institute for Systems Genetics, New York University School of Medicine, New York, NY, USA
| | - Donna R Whelan
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Dylan A Reid
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Jennifer Zagelbaum
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Yandong Yin
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Sisi Ma
- Institute for Systems Genetics, New York University School of Medicine, New York, NY, USA
| | - Eli Rothenberg
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA.
| | - David Fenyö
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA.
- Institute for Systems Genetics, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
198
|
Bagam P, Singh DP, Inda ME, Batra S. Unraveling the role of membrane microdomains during microbial infections. Cell Biol Toxicol 2017; 33:429-455. [PMID: 28275881 PMCID: PMC7088210 DOI: 10.1007/s10565-017-9386-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 02/06/2017] [Indexed: 01/06/2023]
Abstract
Infectious diseases pose major socioeconomic and health-related threats to millions of people across the globe. Strategies to combat infectious diseases derive from our understanding of the complex interactions between the host and specific bacterial, viral, and fungal pathogens. Lipid rafts are membrane microdomains that play important role in life cycle of microbes. Interaction of microbial pathogens with host membrane rafts influences not only their initial colonization but also their spread and the induction of inflammation. Therefore, intervention strategies aimed at modulating the assembly of membrane rafts and/or regulating raft-directed signaling pathways are attractive approaches for the. management of infectious diseases. The current review discusses the latest advances in terms of techniques used to study the role of membrane microdomains in various pathological conditions and provides updated information regarding the role of membrane rafts during bacterial, viral and fungal infections.
Collapse
Affiliation(s)
- Prathyusha Bagam
- Laboratory of Pulmonary Immuno-Toxicology, Department of Environmental Toxicology, Health Research Center, Southern University and A&M College, Baton Rouge, LA, 70813, USA
| | - Dhirendra P Singh
- Laboratory of Pulmonary Immuno-Toxicology, Department of Environmental Toxicology, Health Research Center, Southern University and A&M College, Baton Rouge, LA, 70813, USA
| | - Maria Eugenia Inda
- Departamento de Microbiología, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Suipacha, Rosario, Argentina
| | - Sanjay Batra
- Laboratory of Pulmonary Immuno-Toxicology, Department of Environmental Toxicology, Health Research Center, Southern University and A&M College, Baton Rouge, LA, 70813, USA.
| |
Collapse
|
199
|
Boening D, Gauthier-Kemper A, Gmeiner B, Klingauf J. Cluster Recognition by Delaunay Triangulation of Synaptic Proteins in 3D. ACTA ACUST UNITED AC 2017; 1:e1700091. [PMID: 32646194 DOI: 10.1002/adbi.201700091] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 06/17/2017] [Indexed: 11/06/2022]
Abstract
The advent of super-resolution microscopy opens up the opportunity to study biological structures in unprecedented detail. However, revealing quantitative information about the spatial organization of a set of labeled proteins requires sophisticated analysis. This study introduces a novel robust cluster recognition algorithm based on Delaunay triangulation (CRADT), which can handle complex datasets generated by 3D super-resolution microscopy. This algorithm allows determining volume and shape of protein clusters in 3D. The study demonstrates its performance by applying this algorithm on dual-color 3D super-resolved measurements of mouse hippocampal synapses, stained against the presynaptic active zone marker protein Bassoon and the opposing postsynaptic density protein Homer as well as the exo- and endocytosis machinery proteins Synaptobrevin and Clathrin.
Collapse
Affiliation(s)
- Daniel Boening
- Department of Cellular Biophysics, Institute of Medical Physics and Biophysics, University of Münster, 48149, Münster, Germany.,Max Planck Institute for the Science of Light, Nano-Optics Division, 91058, Erlangen, Germany
| | - Anne Gauthier-Kemper
- Department of Cellular Biophysics, Institute of Medical Physics and Biophysics, University of Münster, 48149, Münster, Germany
| | - Benjamin Gmeiner
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Jürgen Klingauf
- Department of Cellular Biophysics, Institute of Medical Physics and Biophysics, University of Münster, 48149, Münster, Germany.,Cluster of Excellence EXC 1003, Cells in Motion, CiM, 48149, Münster, Germany
| |
Collapse
|
200
|
Schaus TE, Woo S, Xuan F, Chen X, Yin P. A DNA nanoscope via auto-cycling proximity recording. Nat Commun 2017; 8:696. [PMID: 28947733 PMCID: PMC5612940 DOI: 10.1038/s41467-017-00542-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 06/27/2017] [Indexed: 11/27/2022] Open
Abstract
Analysis of the spatial arrangement of molecular features enables the engineering of synthetic nanostructures and the understanding of natural ones. The ability to acquire a comprehensive set of pairwise proximities between components would satisfy an increasing interest in investigating individual macromolecules and their interactions, but current biochemical techniques detect only a single proximity partner per probe. Here, we present a biochemical DNA nanoscopy method that records nanostructure features in situ and in detail for later readout. Based on a conceptually novel auto-cycling proximity recording (APR) mechanism, it continuously and repeatedly produces proximity records of any nearby pairs of DNA-barcoded probes, at physiological temperature, without altering the probes themselves. We demonstrate the production of dozens of records per probe, decode the spatial arrangements of 7 unique probes in a homogeneous sample, and repeatedly sample the same probes in different states. The spatial organisation of nanostructures is fundamental to their function. Here, the authors develop a non-destructive, proximity-based method to record extensive spatial organization information in DNA molecules for later readout.
Collapse
Affiliation(s)
- Thomas E Schaus
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Circle, 5th Floor, Boston, MA, 02115, USA.,Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Sungwook Woo
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Circle, 5th Floor, Boston, MA, 02115, USA.,Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Feng Xuan
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Circle, 5th Floor, Boston, MA, 02115, USA.,Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Xi Chen
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Circle, 5th Floor, Boston, MA, 02115, USA.,Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Peng Yin
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Circle, 5th Floor, Boston, MA, 02115, USA. .,Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|