Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Elife ; 92020 07 21.
Article in English | MEDLINE | ID: mdl-32690136

ABSTRACT

Cancer clone evolution takes place within tissue ecosystem habitats. But, how exactly tumors arise from a few malignant cells within an intact epithelium is a central, yet unanswered question. This is mainly due to the inaccessibility of this process to longitudinal imaging together with a lack of systems that model the progression of a fraction of transformed cells within a tissue. Here, we developed a new methodology based on primary mouse mammary epithelial acini, where oncogenes can be switched on in single cells within an otherwise normal epithelial cell layer. We combine this stochastic breast tumor induction model with inverted light-sheet imaging to study single-cell behavior for up to four days and analyze cell fates utilizing a newly developed image-data analysis workflow. The power of this integrated approach is illustrated by us finding that small local clusters of transformed cells form tumors while isolated transformed cells do not.


There are now drugs to treat many types of cancer, but questions still remain around how these diseases start in the first place. Researchers think that tumor growth begins when a single cell suffers damage to certain sites in its DNA that eventually cause it to divide uncontrollably. That damaged cell, and its descendants, go on to form a lump, or tumor. The trouble with proving this theory is that it is hard to watch it happening in real time. Doctors usually only meet people with cancer when their tumors start to cause health problems. By this point, the tumors contain millions of cells. A way to watch the very beginnings of a cancer could reveal risk factors within a tissue that foster the growth of a tumor. But first, researchers need to test their theory about how the disease begins in the first place. One way to do this is to surround a single cancer cell with healthy cells and watch what happens next. To do this, Alladin, Chaible et al. took healthy cells from the breast tissue of mice and grew them in the laboratory into mini-organs called organoids. These organoids share a lot of features with actual mouse breast tissue; they can even make milk if given the right hormones. Once the organoids were ready, Alladin, Chaible et al then started modifying a small number of single cells inside them by switching on genes called oncogenes, which are known to drive cancer formation in humans. Using fluorescent proteins and a sheet of laser light it was possible to watch what happened to the cells over time. This revealed that, even though all the oncogene-driven single cells received the same signals, not all of them started to divide uncontrollably. In fact, a single modified cell had a low chance of forming a tumor on its own. The more oncogene-driven cells there were near to each other, the more likely they were to form tumors. Alladin, Chaible et al. think that this is because the healthy tissue interacts with the modified, oncogene-driven cells to suppress tumor formation. It is only when a larger number of modified cells group together and start to communicate with each other that they can override the inhibitory messages of the healthy tissue. How healthy tissue stops single modified cells from forming tumors is not yet clear. But, with this new mini-organ system, researchers now have the tools to investigate. In the future, this could lead to new strategies to stop cancer before it has a chance to get started.


Subject(s)
Acinar Cells/cytology , Breast Neoplasms/genetics , Breast Neoplasms/physiopathology , Cell Tracking/methods , Epithelial Cells/cytology , Mammary Glands, Human/cytology , Neoplastic Stem Cells/cytology , Animals , Female , Humans , Mice , Microscopy/methods , Models, Animal
2.
Dev Cell ; 52(4): 492-508.e10, 2020 02 24.
Article in English | MEDLINE | ID: mdl-32059773

ABSTRACT

How tissues migrate robustly through changing guidance landscapes is poorly understood. Here, quantitative imaging is combined with inducible perturbation experiments to investigate the mechanisms that ensure robust tissue migration in vivo. We show that tissues exposed to acute "chemokine floods" halt transiently before they perfectly adapt, i.e., return to the baseline migration behavior in the continued presence of elevated chemokine levels. A chemokine-triggered phosphorylation of the atypical chemokine receptor Cxcr7b reroutes it from constitutive ubiquitination-regulated degradation to plasma membrane recycling, thus coupling scavenging capacity to extracellular chemokine levels. Finally, tissues expressing phosphorylation-deficient Cxcr7b migrate normally in the presence of physiological chemokine levels but show delayed recovery when challenged with elevated chemokine concentrations. This work establishes that adaptation to chemokine fluctuations can be "outsourced" from canonical GPCR signaling to an autonomously acting scavenger receptor that both senses and dynamically buffers chemokine levels to increase the robustness of tissue migration.


Subject(s)
Cell Movement , Chemokines/metabolism , Embryo, Nonmammalian/metabolism , Receptors, CXCR4/metabolism , Receptors, CXCR/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Cell Communication , Chemokines/genetics , Embryo, Nonmammalian/cytology , Phosphorylation , Receptors, CXCR/genetics , Receptors, CXCR4/genetics , Signal Transduction , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish Proteins/genetics
3.
Nature ; 561(7723): 411-415, 2018 09.
Article in English | MEDLINE | ID: mdl-30202089

ABSTRACT

Essential biological functions, such as mitosis, require tight coordination of hundreds of proteins in space and time. Localization, the timing of interactions and changes in cellular structure are all crucial to ensure the correct assembly, function and regulation of protein complexes1-4. Imaging of live cells can reveal protein distributions and dynamics but experimental and theoretical challenges have prevented the collection of quantitative data, which are necessary for the formulation of a model of mitosis that comprehensively integrates information and enables the analysis of the dynamic interactions between the molecular parts of the mitotic machinery within changing cellular boundaries. Here we generate a canonical model of the morphological changes during the mitotic progression of human cells on the basis of four-dimensional image data. We use this model to integrate dynamic three-dimensional concentration data of many fluorescently knocked-in mitotic proteins, imaged by fluorescence correlation spectroscopy-calibrated microscopy5. The approach taken here to generate a dynamic protein atlas of human cell division is generic; it can be applied to systematically map and mine dynamic protein localization networks that drive cell division in different cell types, and can be conceptually transferred to other cellular functions.


Subject(s)
Cell Cycle Proteins/analysis , Cell Cycle Proteins/metabolism , Mitosis , Gene Editing , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Imaging, Three-Dimensional , Microscopy, Fluorescence , Molecular Imaging , Time Factors
4.
Nat Protoc ; 13(6): 1445-1464, 2018 06.
Article in English | MEDLINE | ID: mdl-29844523

ABSTRACT

The ability to tag a protein at its endogenous locus with a fluorescent protein (FP) enables quantitative understanding of protein dynamics at the physiological level. Genome-editing technology has now made this powerful approach routinely applicable to mammalian cells and many other model systems, thereby opening up the possibility to systematically and quantitatively map the cellular proteome in four dimensions. 3D time-lapse confocal microscopy (4D imaging) is an essential tool for investigating spatial and temporal protein dynamics; however, it lacks the required quantitative power to make the kind of absolute and comparable measurements required for systems analysis. In contrast, fluorescence correlation spectroscopy (FCS) provides quantitative proteomic and biophysical parameters such as protein concentration, hydrodynamic radius, and oligomerization but lacks the capability for high-throughput application in 4D spatial and temporal imaging. Here we present an automated experimental and computational workflow that integrates both methods and delivers quantitative 4D imaging data in high throughput. These data are processed to yield a calibration curve relating the fluorescence intensities (FIs) of image voxels to the absolute protein abundance. The calibration curve allows the conversion of the arbitrary FIs to protein amounts for all voxels of 4D imaging stacks. Using our workflow, users can acquire and analyze hundreds of FCS-calibrated image series to map their proteins of interest in four dimensions. Compared with other protocols, the current protocol does not require additional calibration standards and provides an automated acquisition pipeline for FCS and imaging data. The protocol can be completed in 1 d.


Subject(s)
Cells/chemistry , Imaging, Three-Dimensional/methods , Optical Imaging/methods , Proteins/analysis , Proteome/analysis , Proteomics/methods , Staining and Labeling/methods , Automation, Laboratory/methods , Gene Editing/methods , Spatio-Temporal Analysis
5.
Article in English | MEDLINE | ID: mdl-28035242

ABSTRACT

BACKGROUND: The dynamic three-dimensional chromatin architecture of genomes and its co-evolutionary connection to its function-the storage, expression, and replication of genetic information-is still one of the central issues in biology. Here, we describe the much debated 3D architecture of the human and mouse genomes from the nucleosomal to the megabase pair level by a novel approach combining selective high-throughput high-resolution chromosomal interaction capture (T2C), polymer simulations, and scaling analysis of the 3D architecture and the DNA sequence. RESULTS: The genome is compacted into a chromatin quasi-fibre with ~5 ± 1 nucleosomes/11 nm, folded into stable ~30-100 kbp loops forming stable loop aggregates/rosettes connected by similar sized linkers. Minor but significant variations in the architecture are seen between cell types and functional states. The architecture and the DNA sequence show very similar fine-structured multi-scaling behaviour confirming their co-evolution and the above. CONCLUSIONS: This architecture, its dynamics, and accessibility, balance stability and flexibility ensuring genome integrity and variation enabling gene expression/regulation by self-organization of (in)active units already in proximity. Our results agree with the heuristics of the field and allow "architectural sequencing" at a genome mechanics level to understand the inseparable systems genomic properties.

6.
Article in English | MEDLINE | ID: mdl-28035241

ABSTRACT

BACKGROUND: Genome organization into subchromosomal topologically associating domains (TADs) is linked to cell-type-specific gene expression programs. However, dynamic properties of such domains remain elusive, and it is unclear how domain plasticity modulates genomic accessibility for soluble factors. RESULTS: Here, we combine and compare a high-resolution topology analysis of interacting chromatin loci with fluorescence correlation spectroscopy measurements of domain dynamics in single living cells. We identify topologically and dynamically independent chromatin domains of ~1 Mb in size that are best described by a loop-cluster polymer model. Hydrodynamic relaxation times and gyration radii of domains are larger for open (161 ± 15 ms, 297 ± 9 nm) than for dense chromatin (88 ± 7 ms, 243 ± 6 nm) and increase globally upon chromatin hyperacetylation or ATP depletion. CONCLUSIONS: Based on the domain structure and dynamics measurements, we propose a loop-cluster model for chromatin domains. It suggests that the regulation of chromatin accessibility for soluble factors displays a significantly stronger dependence on factor concentration than search processes within a static network.

7.
Elife ; 52016 09 29.
Article in English | MEDLINE | ID: mdl-27684073

ABSTRACT

Formation of the three embryonic germ layers is a fundamental developmental process that initiates differentiation. How the zebrafish pluripotency factor Pou5f3 (homologous to mammalian Oct4) drives lineage commitment is unclear. Here, we introduce fluorescence lifetime imaging microscopy and fluorescence correlation spectroscopy to assess the formation of Pou5f3 complexes with other transcription factors in real-time in gastrulating zebrafish embryos. We show, at single-cell resolution in vivo, that Pou5f3 complexes with Nanog to pattern mesendoderm differentiation at the blastula stage. Later, during gastrulation, Sox32 restricts Pou5f3-Nanog complexes to the ventrolateral mesendoderm by binding Pou5f3 or Nanog in prospective dorsal endoderm. In the ventrolateral endoderm, the Elabela / Aplnr pathway limits Sox32 levels, allowing the formation of Pou5f3-Nanog complexes and the activation of downstream BMP signaling. This quantitative model shows that a balance in the spatiotemporal distribution of Pou5f3-Nanog complexes, modulated by Sox32, regulates mesendoderm specification along the dorsoventral axis.


Subject(s)
Mesoderm/embryology , Nanog Homeobox Protein/analysis , Octamer Transcription Factor-3/analysis , Zebrafish Proteins/analysis , Zebrafish/embryology , Animals , Intravital Microscopy , Mesoderm/chemistry , Microscopy, Fluorescence , Protein Binding , Spatio-Temporal Analysis , Spectrometry, Fluorescence
8.
Mol Cell ; 59(4): 564-75, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-26295961

ABSTRACT

In Drosophila, two Piwi proteins, Aubergine (Aub) and Argonaute-3 (Ago3), localize to perinuclear "nuage" granules and use guide piRNAs to target and destroy transposable element transcripts. We find that Aub and Ago3 are recruited to nuage by two different mechanisms. Aub requires a piRNA guide for nuage recruitment, indicating that its localization depends on recognition of RNA targets. Ago3 is recruited to nuage independently of a piRNA cargo and relies on interaction with Krimper, a stable component of nuage that is able to aggregate in the absence of other nuage proteins. We show that Krimper interacts directly with Aub and Ago3 to coordinate the assembly of the ping-pong piRNA processing (4P) complex. Symmetrical dimethylated arginines are required for Aub to interact with Krimper, but they are dispensable for Ago3 to bind Krimper. Our study reveals a multi-step process responsible for the assembly and function of nuage complexes in piRNA-guided transposon repression.


Subject(s)
Argonaute Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/physiology , Drosophila melanogaster/metabolism , Peptide Initiation Factors/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , Drosophila Proteins/chemistry , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Female , Kinetics , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Transport , RNA, Small Interfering/metabolism
9.
Nat Biotechnol ; 33(4): 384-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25774713

ABSTRACT

To understand the function of cellular protein networks, spatial and temporal context is essential. Fluorescence correlation spectroscopy (FCS) is a single-molecule method to study the abundance, mobility and interactions of fluorescence-labeled biomolecules in living cells. However, manual acquisition and analysis procedures have restricted live-cell FCS to short-term experiments of a few proteins. Here, we present high-throughput (HT)-FCS, which automates screening and time-lapse acquisition of FCS data at specific subcellular locations and subsequent data analysis. We demonstrate its utility by studying the dynamics of 53 nuclear proteins. We made 60,000 measurements in 10,000 living human cells, to obtain biophysical parameters that allowed us to classify proteins according to their chromatin binding and complex formation. We also analyzed the cell-cycle-dependent dynamics of the mitotic kinase complex Aurora B/INCENP and showed how a rise in Aurora concentration triggers two-step complex formation. We expect that throughput and robustness will make HT-FCS a broadly applicable technology for characterizing protein network dynamics in cells.


Subject(s)
High-Throughput Screening Assays/methods , Protein Interaction Mapping/methods , Proteome/metabolism , Spectrometry, Fluorescence/methods , Subcellular Fractions/metabolism , Molecular Imaging/methods
10.
Mol Syst Biol ; 10: 756, 2014 Nov 03.
Article in English | MEDLINE | ID: mdl-25366307

ABSTRACT

Clathrin-mediated endocytosis is a highly conserved intracellular trafficking pathway that depends on dynamic protein-protein interactions between up to 60 different proteins. However, little is known about the spatio-temporal regulation of these interactions. Using fluorescence (cross)-correlation spectroscopy in yeast, we tested 41 previously reported interactions in vivo and found 16 to exist in the cytoplasm. These detected cytoplasmic interactions included the self-interaction of Ede1, homolog of mammalian Eps15. Ede1 is the crucial scaffold for the organization of the early stages of endocytosis. We show that oligomerization of Ede1 through its central coiled coil domain is necessary for its localization to the endocytic site and we link the oligomerization of Ede1 to its function in locally concentrating endocytic adaptors and organizing the endocytic machinery. Our study sheds light on the importance of the regulation of protein-protein interactions in the cytoplasm for the assembly of the endocytic machinery in vivo.


Subject(s)
Endocytosis , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Cytosol/physiology , Gene Expression Regulation, Fungal , Genome, Fungal , Protein Interaction Domains and Motifs , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae Proteins/chemistry , Spectrometry, Fluorescence
11.
Mol Biol Cell ; 25(22): 3610-8, 2014 Nov 05.
Article in English | MEDLINE | ID: mdl-25232003

ABSTRACT

Fluorescence tagging of proteins is a widely used tool to study protein function and dynamics in live cells. However, the extent to which different mammalian transgene methods faithfully report on the properties of endogenous proteins has not been studied comparatively. Here we use quantitative live-cell imaging and single-molecule spectroscopy to analyze how different transgene systems affect imaging of the functional properties of the mitotic kinase Aurora B. We show that the transgene method fundamentally influences level and variability of expression and can severely compromise the ability to report on endogenous binding and localization parameters, providing a guide for quantitative imaging studies in mammalian cells.


Subject(s)
Aurora Kinase B/genetics , Green Fluorescent Proteins/genetics , Molecular Imaging , Plasmids/metabolism , Transfection/methods , Transgenes , Aurora Kinase B/metabolism , Base Sequence , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Microscopy, Fluorescence , Mitosis , Molecular Sequence Data , Plasmids/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
12.
Nat Commun ; 5: 4494, 2014 Jul 24.
Article in English | MEDLINE | ID: mdl-25058002

ABSTRACT

In living cells, most proteins diffuse over distances of micrometres within seconds. Protein translocation is constrained due to the cellular organization into subcompartments that impose diffusion barriers and guide enzymatic activities to their targets. Here, we introduce an approach to retrieve structural features from the scale-dependent mobility of green fluorescent protein monomer and multimers in human cells. We measure protein transport simultaneously between hundreds of positions by multi-scale fluorescence cross-correlation spectroscopy using a line-illuminating confocal microscope. From these data we derive a quantitative model of the intracellular architecture that resembles a random obstacle network for diffusing proteins. This topology partitions the cellular content and increases the dwell time of proteins in their local environment. The accessibility of obstacle surfaces depends on protein size. Our method links multi-scale mobility measurements with a quantitative description of intracellular structure that can be applied to evaluate how drug-induced perturbations affect protein transport and interactions.


Subject(s)
Proteins/analysis , Proteins/metabolism , Spectrometry, Fluorescence/methods , Cell Line , Chromatin/metabolism , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Cytoplasm/metabolism , Cytoskeleton/metabolism , Diffusion , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Image Processing, Computer-Assisted , Protein Transport , Quantum Dots , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/metabolism , Spectrometry, Fluorescence/instrumentation
13.
J Cell Biol ; 204(5): 697-712, 2014 Mar 03.
Article in English | MEDLINE | ID: mdl-24590172

ABSTRACT

SAS-6 proteins are thought to impart the ninefold symmetry of centrioles, but the mechanisms by which their assembly occurs within cells remain elusive. In this paper, we provide evidence that the N-terminal, coiled-coil, and C-terminal domains of HsSAS-6 are each required for procentriole formation in human cells. Moreover, the coiled coil is necessary and sufficient to mediate HsSAS-6 centrosomal targeting. High-resolution imaging reveals that GFP-tagged HsSAS-6 variants localize in a torus around the base of the parental centriole before S phase, perhaps indicative of an initial loading platform. Moreover, fluorescence recovery after photobleaching analysis demonstrates that HsSAS-6 is immobilized progressively at centrosomes during cell cycle progression. Using fluorescence correlation spectroscopy and three-dimensional stochastic optical reconstruction microscopy, we uncover that HsSAS-6 is present in the cytoplasm primarily as a homodimer and that its oligomerization into a ninefold symmetrical ring occurs at centrioles. Together, our findings lead us to propose a mechanism whereby HsSAS-6 homodimers are targeted to centrosomes where the local environment and high concentration of HsSAS-6 promote oligomerization, thus initiating procentriole formation.


Subject(s)
Cell Cycle Proteins/metabolism , Centrioles/metabolism , Cell Cycle , Cell Cycle Proteins/analysis , Cell Cycle Proteins/physiology , Cell Line, Tumor , Centrioles/ultrastructure , Dimerization , Fluorescence Recovery After Photobleaching , Humans , Models, Biological , Protein Transport
14.
Protoplasma ; 251(2): 373-82, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24390250

ABSTRACT

Intracellular molecular transport and localization are crucial for cells (plant cells as much as mammalian cells) to proliferate and to adapt to diverse environmental conditions. Here, some aspects of the microscopy-based method of fluorescence recovery after photobleaching (FRAP) are introduced. In the course of the last years, this has become a very powerful tool to study dynamic processes in living cells and tissue, and it is expected to experience further increasing demand because quantitative information on biological systems becomes more and more important. This review introduces the FRAP methodology, including some theoretical background, describes challenges and pitfalls, and presents some recent advanced applications.


Subject(s)
Fluorescence Recovery After Photobleaching/methods , Microscopy, Fluorescence/methods , Fluorescence Recovery After Photobleaching/instrumentation , Molecular Dynamics Simulation
15.
Cytometry A ; 83(9): 876-89, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23847177

ABSTRACT

To quantify more precisely and more reliably diffusion and reaction properties of biomolecules in living cells, a novel closed description in 3D of both the bleach and the post-bleach segment of fluorescence recovery after photobleaching (FRAP) data acquired at a point, i.e., a diffraction-limited observation area, termed point FRAP, is presented. It covers a complete coupled reaction-diffusion scheme for mobile molecules undergoing transient or long-term immobilization because of binding. We assess and confirm the feasibility with numerical solutions of the differential equations. By applying this model to free EYFP expressed in HeLa cells using a customized confocal laser scanning microscope that integrates point FRAP and fluorescence correlation spectroscopy (FCS), the applicability is validated by comparison with results from FCS. We show that by taking diffusion during bleaching into consideration and/or by employing a global analysis of series of bleach times, the results can be improved significantly. As the point FRAP approach allows to obtain data with diffraction-limited positioning accuracy, diffusion and binding properties of the exon-exon junction complex (EJC) components REF2-II and Magoh are obtained at different localizations in the nucleus of MCF7 cells and refine our view on the position-dependent association of the EJC factors with a maturating mRNP complex. Our findings corroborate the concept of combining point FRAP and FCS for a better understanding of the underlying diffusion and binding processes.


Subject(s)
Fluorescence Recovery After Photobleaching/methods , Spectrometry, Fluorescence/methods , Bacterial Proteins/chemistry , Cell Line, Tumor , HeLa Cells , Histones/chemistry , Humans , Luminescent Proteins/chemistry , MCF-7 Cells , Protein Binding , Recombinant Fusion Proteins/chemistry
16.
Proc Natl Acad Sci U S A ; 109(38): E2533-42, 2012 Sep 18.
Article in English | MEDLINE | ID: mdl-22927393

ABSTRACT

Dynamic actin filaments are a crucial component of clathrin-mediated endocytosis when endocytic proteins cannot supply enough energy for vesicle budding. Actin cytoskeleton is thought to provide force for membrane invagination or vesicle scission, but how this force is transmitted to the plasma membrane is not understood. Here we describe the molecular mechanism of plasma membrane-actin cytoskeleton coupling mediated by cooperative action of epsin Ent1 and the HIP1R homolog Sla2 in yeast Saccharomyces cerevisiae. Sla2 anchors Ent1 to a stable endocytic coat by an unforeseen interaction between Sla2's ANTH and Ent1's ENTH lipid-binding domains. The ANTH and ENTH domains bind each other in a ligand-dependent manner to provide critical anchoring of both proteins to the membrane. The C-terminal parts of Ent1 and Sla2 bind redundantly to actin filaments via a previously unknown phospho-regulated actin-binding domain in Ent1 and the THATCH domain in Sla2. By the synergistic binding to the membrane and redundant interaction with actin, Ent1 and Sla2 form an essential molecular linker that transmits the force generated by the actin cytoskeleton to the plasma membrane, leading to membrane invagination and vesicle budding.


Subject(s)
Actin Cytoskeleton/metabolism , Clathrin/metabolism , Cytoskeleton/metabolism , Endocytosis , Saccharomyces cerevisiae/metabolism , Actins/metabolism , Cell Membrane/metabolism , Cytoskeletal Proteins/metabolism , Gene Deletion , Gene Expression Regulation , Glutathione Transferase/metabolism , Lipids/chemistry , Models, Biological , Phenotype , Protein Binding , Protein Structure, Tertiary , Saccharomyces cerevisiae Proteins/metabolism , Vesicular Transport Proteins/metabolism
17.
Nat Biotechnol ; 30(7): 708-14, 2012 Jun 24.
Article in English | MEDLINE | ID: mdl-22729030

ABSTRACT

The functional state of a cell is largely determined by the spatiotemporal organization of its proteome. Technologies exist for measuring particular aspects of protein turnover and localization, but comprehensive analysis of protein dynamics across different scales is possible only by combining several methods. Here we describe tandem fluorescent protein timers (tFTs), fusions of two single-color fluorescent proteins that mature with different kinetics, which we use to analyze protein turnover and mobility in living cells. We fuse tFTs to proteins in yeast to study the longevity, segregation and inheritance of cellular components and the mobility of proteins between subcellular compartments; to measure protein degradation kinetics without the need for time-course measurements; and to conduct high-throughput screens for regulators of protein turnover. Our experiments reveal the stable nature and asymmetric inheritance of nuclear pore complexes and identify regulators of N-end rule­mediated protein degradation.


Subject(s)
Green Fluorescent Proteins/chemistry , High-Throughput Screening Assays , Proteins/metabolism , Subcellular Fractions , Kinetics , Nuclear Pore/metabolism , Nuclear Pore/ultrastructure , Protein Stability , Proteolysis , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/ultrastructure , Subcellular Fractions/metabolism , Subcellular Fractions/ultrastructure
18.
Nat Biotechnol ; 29(9): 835-9, 2011 Aug 07.
Article in English | MEDLINE | ID: mdl-21822256

ABSTRACT

Diffusion processes and local dynamic equilibria inside cells lead to nonuniform spatial distributions of molecules, which are essential for processes such as nuclear organization and signaling in cell division, differentiation and migration. To understand these mechanisms, spatially resolved quantitative measurements of protein abundance, mobilities and interactions are needed, but current methods have limited capabilities to study dynamic parameters. Here we describe a microscope based on light-sheet illumination that allows massively parallel fluorescence correlation spectroscopy (FCS) measurements and use it to visualize the diffusion and interactions of proteins in mammalian cells and in isolated fly tissue. Imaging the mobility of heterochromatin protein HP1α (ref. 4) in cell nuclei we could provide high-resolution diffusion maps that reveal euchromatin areas with heterochromatin-like HP1α-chromatin interactions. We expect that FCS imaging will become a useful method for the precise characterization of cellular reaction-diffusion processes.


Subject(s)
Microscopy, Fluorescence/methods , Protein Interaction Mapping/methods , Spectrometry, Fluorescence/methods , 3T3 Cells , Animals , Cell Line , Cell Nucleus , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Diffusion , Dogs , Drosophila/cytology , Euchromatin/genetics , Euchromatin/metabolism , Gene Expression Regulation , Genetic Vectors/genetics , Heterochromatin/genetics , Heterochromatin/metabolism , Humans , Kidney/cytology , Lentivirus/genetics , Mice , Microscopy, Confocal , Proteins/genetics , Proteins/metabolism
19.
Chromosome Res ; 19(1): 99-115, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20848178

ABSTRACT

The genome of eukaryotes is organized into a dynamic nucleoprotein complex referred to as chromatin, which can adopt different functional states. Both the DNA and the protein component of chromatin are subject to various post-translational modifications that define the cell's gene expression program. Their readout and establishment occurs in a spatio-temporally coordinated manner that is controlled by numerous chromatin-interacting proteins. Binding to chromatin in living cells can be measured by a spatially resolved analysis of protein mobility using fluorescence microscopy based approaches. Recent advancements in the acquisition of protein mobility data using fluorescence bleaching and correlation methods provide data sets on diffusion coefficients, binding kinetics, and cellular concentrations on different time and length scales. The combination of different techniques is needed to dissect the complex interplay of diffusive translocations, binding events, and mobility constraints of the chromatin environment. While bleaching techniques have their strength in the characterization of particles that are immobile on the second/minute time scale, a correlation analysis is advantageous to characterize transient binding events with millisecond residence time. The application and synergy effects of the different approaches to obtain protein mobility and interaction maps in the nucleus are illustrated for the analysis of heterochromatin protein 1.


Subject(s)
Chromatin/metabolism , Protein Interaction Mapping/methods , Protein Processing, Post-Translational , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/analysis , DNA , Diffusion , Fluorescence Recovery After Photobleaching/methods , Gene Expression , Microscopy, Fluorescence , Models, Biological , Molecular Dynamics Simulation , Protein Binding
20.
Opt Express ; 18(23): 23818-28, 2010 Nov 08.
Article in English | MEDLINE | ID: mdl-21164726

ABSTRACT

We present an implementation of fluorescence correlation spectroscopy with spectrally resolved detection based on a combined commercial confocal laser scanning/fluorescence correlation spectroscopy microscope. We have replaced the conventional detection scheme by a prism-based spectrometer and an electron-multiplying charge-coupled device camera used to record the photons. This allows us to read out more than 80,000 full spectra per second with a signal-to-noise ratio and a quantum efficiency high enough to allow single photon counting. We can identify up to four spectrally different quantum dots in vitro and demonstrate that spectrally resolved detection can be used to characterize photophysical properties of fluorophores by measuring the spectral dependence of quantum dot fluorescence emission intermittence. Moreover, we can confirm intracellular cross-correlation results as acquired with a conventional setup and show that spectral flexibility can help to optimize the choice of the detection windows.

SELECTION OF CITATIONS
SEARCH DETAIL