Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
1.
iScience ; 27(7): 110268, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39036041

ABSTRACT

The outcome of cell signaling depends not only on signal strength but also on temporal progression. We use Fluorescence Lifetime Imaging of Resonance Energy Transfer (FLIM/FRET) biosensors to investigate intracellular signaling dynamics. We examined the ß1 receptor-Gαs-cAMP signaling axis using both widefield frequency domain FLIM (fdFLIM) and fast confocal time-correlated single photon counting (TCSPC) setups. Unexpectedly, we observed that fdFLIM revealed transient cAMP responses in HeLa and Cos7 cells, contrasting with sustained responses as detected with TCSPC. Investigation revealed no light-induced effects on cAMP generation or breakdown. Rather, folic acid present in the imaging medium appeared to be the culprit, as its excitation with blue light sensitized degradation of ß1 agonists. Our findings highlight the impact of subtle phototoxicity on experimental outcomes, advocating confocal TCSPC for reliable analysis of response kinetics and stressing the need for full disclosure of chemical formulations by scientific vendors.

2.
Sci Adv ; 9(22): eadf4409, 2023 06 02.
Article in English | MEDLINE | ID: mdl-37256941

ABSTRACT

DNA interstrand crosslinks (ICLs) pose a major obstacle for DNA replication and transcription if left unrepaired. The cellular response to ICLs requires the coordination of various DNA repair mechanisms. Homologous recombination (HR) intermediates generated in response to ICLs, require efficient and timely conversion by structure-selective endonucleases. Our knowledge on the precise coordination of this process remains incomplete. Here, we designed complementary genetic screens to map the machinery involved in the response to ICLs and identified FIRRM/C1orf112 as an indispensable factor in maintaining genome stability. FIRRM deficiency leads to hypersensitivity to ICL-inducing compounds, accumulation of DNA damage during S-G2 phase of the cell cycle, and chromosomal aberrations, and elicits a unique mutational signature previously observed in HR-deficient tumors. In addition, FIRRM is recruited to ICLs, controls MUS81 chromatin loading, and thereby affects resolution of HR intermediates. FIRRM deficiency in mice causes early embryonic lethality and accelerates tumor formation. Thus, FIRRM plays a critical role in the response to ICLs encountered during DNA replication.


Subject(s)
DNA Damage , DNA Repair , Animals , Mice , DNA Replication , Homologous Recombination , DNA
3.
Nat Commun ; 13(1): 6442, 2022 10 28.
Article in English | MEDLINE | ID: mdl-36307419

ABSTRACT

The experimental need to engineer the genome both in time and space, has led to the development of several photoactivatable Cre recombinase systems. However, the combination of inefficient and non-intentional background recombination has prevented thus far the wide application of these systems in biological and biomedical research. Here, we engineer an optimized photoactivatable Cre recombinase system that we refer to as doxycycline- and light-inducible Cre recombinase (DiLiCre). Following extensive characterization in cancer cell and organoid systems, we generate a DiLiCre mouse line, and illustrated the biological applicability of DiLiCre for light-induced mutagenesis in vivo and positional cell-tracing by intravital microscopy. These experiments illustrate how newly formed HrasV12 mutant cells follow an unnatural movement towards the interfollicular dermis. Together, we develop an efficient photoactivatable Cre recombinase mouse model and illustrate how this model is a powerful genome-editing tool for biological and biomedical research.


Subject(s)
Doxycycline , Optogenetics , Mice , Animals , Doxycycline/pharmacology , Mice, Transgenic , Gene Editing , Integrases/genetics , Integrases/metabolism , Mice, Inbred Strains
4.
STAR Protoc ; 3(2): 101246, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35368806

ABSTRACT

Tumor Treating Fields (TTFields) are an FDA-approved anticancer treatment using alternating electric fields. Here, we present a protocol to perform live-cell imaging (LCI) of cells during TTFields treatment with the Inovitro LiveTM system. The setup we describe dissipates TTFields-related heat production and can be used in conjunction with any LCI-compatible microscope setup. This approach will enable further elucidation of TTFields' mechanism of action at the molecular level and facilitate the development of promising combination strategies.


Subject(s)
Electric Stimulation Therapy , Neoplasms , Combined Modality Therapy , Electric Stimulation Therapy/methods , Humans , Neoplasms/diagnostic imaging
5.
Methods Mol Biol ; 2483: 105-116, 2022.
Article in English | MEDLINE | ID: mdl-35286672

ABSTRACT

Second messenger molecules in eukaryotic cells relay the signals from activated cell surface receptors to intracellular effector proteins. FRET-based sensors are ideal to visualize and measure the often rapid changes of second messenger concentrations in time and place. Fluorescence Lifetime Imaging (FLIM) is an intrinsically quantitative technique for measuring FRET. Given the recent development of commercially available, sensitive and photon-efficient FLIM instrumentation, it is becoming the method of choice for FRET detection in signaling studies. Here, we describe a detailed protocol for time domain FLIM, using the EPAC-based FRET sensor to measure changes in cellular cAMP levels with high spatiotemporal resolution as an example.


Subject(s)
Fluorescence Resonance Energy Transfer , Optical Imaging , Fluorescence Resonance Energy Transfer/methods , Microscopy, Fluorescence/methods , Photons , Second Messenger Systems
6.
Sci Rep ; 11(1): 20711, 2021 10 20.
Article in English | MEDLINE | ID: mdl-34671065

ABSTRACT

Fluorescence Lifetime Imaging (FLIM) is an intrinsically quantitative method to screen for protein-protein interactions and is frequently used to record the outcome of signal transduction events. With new highly sensitive and photon efficient FLIM instrumentation, the technique also becomes attractive to screen, with high temporal resolution, for fast changes in Förster Resonance Energy Transfer (FRET), such as those occurring upon activation of cell signaling. The second messenger cyclic adenosine monophosphate (cAMP) is rapidly formed following activation of certain cell surface receptors. cAMP is subsequently degraded by a set of phosphodiesterases (PDEs) which display cell-type specific expression and may also affect baseline levels of the messenger. To study which specific PDEs contribute most to cAMP regulation, we knocked down individual PDEs and recorded breakdown rates of cAMP levels following transient stimulation in HeLa cells stably expressing the FRET/FLIM sensor, Epac-SH189. Many hundreds of cells were recorded at 5 s intervals for each condition. FLIM time traces were calculated for every cell, and decay kinetics were obtained. cAMP clearance was significantly slower when PDE3A and, to a lesser amount, PDE10A were knocked down, identifying these isoforms as dominant in HeLa cells. However, taking advantage of the quantitative FLIM data, we found that knockdown of individual PDEs has a very limited effect on baseline cAMP levels. By combining photon-efficient FLIM instrumentation with optimized sensors, systematic gene knockdown and an automated open-source analysis pipeline, our study demonstrates that dynamic screening of transient cell signals has become feasible. The quantitative platform described here provides detailed kinetic analysis of cellular signals in individual cells with unprecedented throughput.


Subject(s)
Signal Transduction/physiology , Cell Line, Tumor , Cyclic AMP/metabolism , Fluorescence , Fluorescence Resonance Energy Transfer/methods , HeLa Cells , Humans , Microscopy, Fluorescence/methods , Optical Imaging/methods , Phosphoric Diester Hydrolases/metabolism , Photons , Proteins/metabolism
7.
J Cell Sci ; 133(3)2020 02 10.
Article in English | MEDLINE | ID: mdl-31932507

ABSTRACT

GDE2 (also known as GDPD5) is a multispanning membrane phosphodiesterase with phospholipase D-like activity that cleaves select glycosylphosphatidylinositol (GPI)-anchored proteins and thereby promotes neuronal differentiation both in vitro and in vivo GDE2 is a prognostic marker in neuroblastoma, while loss of GDE2 leads to progressive neurodegeneration in mice; however, its regulation remains unclear. Here, we report that, in immature neuronal cells, GDE2 undergoes constitutive endocytosis and travels back along both fast and slow recycling routes. GDE2 trafficking is directed by C-terminal tail sequences that determine the ability of GDE2 to cleave GPI-anchored glypican-6 (GPC6) and induce a neuronal differentiation program. Specifically, we define a GDE2 truncation mutant that shows aberrant recycling and is dysfunctional, whereas a consecutive deletion results in cell-surface retention and gain of GDE2 function, thus uncovering distinctive regulatory sequences. Moreover, we identify a C-terminal leucine residue in a unique motif that is essential for GDE2 internalization. These findings establish a mechanistic link between GDE2 neuronal function and sequence-dependent trafficking, a crucial process gone awry in neurodegenerative diseases.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Neuroblastoma , Phospholipases , Animals , Cell Differentiation/genetics , Glycosylphosphatidylinositols/genetics , Mice , Phosphoric Diester Hydrolases/genetics
8.
Nat Commun ; 10(1): 5171, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31729386

ABSTRACT

Basement membrane transmigration during embryonal development, tissue homeostasis and tumor invasion relies on invadosomes, a collective term for invadopodia and podosomes. An adequate structural framework for this process is still missing. Here, we reveal the modular actin nano-architecture that enables podosome protrusion and mechanosensing. The podosome protrusive core contains a central branched actin module encased by a linear actin module, each harboring specific actin interactors and actin isoforms. From the core, two actin modules radiate: ventral filaments bound by vinculin and connected to the plasma membrane and dorsal interpodosomal filaments crosslinked by myosin IIA. On stiff substrates, the actin modules mediate long-range substrate exploration, associated with degradative behavior. On compliant substrates, the vinculin-bound ventral actin filaments shorten, resulting in short-range connectivity and a focally protrusive, non-degradative state. Our findings redefine podosome nanoscale architecture and reveal a paradigm for how actin modularity drives invadosome mechanosensing in cells that breach tissue boundaries.


Subject(s)
Actins/chemistry , Actins/metabolism , Podosomes/metabolism , Actins/genetics , Animals , Cell Adhesion , Cell Membrane/genetics , Cell Membrane/metabolism , Cell Movement , Cells, Cultured , Dendritic Cells/chemistry , Dendritic Cells/cytology , Dendritic Cells/metabolism , Humans , Mechanotransduction, Cellular , Mice , Podosomes/chemistry , Podosomes/genetics
9.
ACS Comb Sci ; 21(8): 598-607, 2019 08 12.
Article in English | MEDLINE | ID: mdl-31269394

ABSTRACT

We report the identification of high-affinity and selectivity integrin α5ß1-binding bicyclic peptides via "designed random libraries", that is, the screening of libraries comprising the universal integrin-binding sequence Arg-Gly-Asp (RGD) in the first loop in combination with a randomized sequence (XXX) in the second loop. Screening of first-generation libraries for α5ß1-binding peptides yielded a triple-digit nanomolar bicyclic α5ß1-binder (CT3RGDcT3AYGCT3, IC50 = 406 nM). Next-generation libraries were designed by partially varying the structure of the strongest first-generation lead inhibitor and screened for improved affinities and selectivities for this receptor. In this way, we identified three high-affinity α5ß1-binders (CT3RGDcT3AYJCT3, J = d-Leu, IC50 = 90 nM; CT3RGDcT3AYaCT3, IC50 = 156 nM; CT3RGDcT3AWGCT3, IC50 = 173 nM), of which one even showed a higher α5ß1-affinity than the 32 amino acid benchmark peptide knottin-RGD (IC50 = 114 nM). Affinity for α5ß1-integrin was confirmed by SPFS analysis showing a Kd of 4.1 nM for Cy5-labeled RGD-bicycle CT3RGDcT3AYJCT3 (J = d-Leu) and a somewhat higher Kd (9.0 nM) for Cy5-labeled knottin-RGD. The α5ß1-bicycles, for example, CT3RGDcT3AYJCT3 (J = d-Leu), showed excellent selectivities over αvß5 (IC50 ratio α5ß1/αvß5 between <0.009 and 0.039) and acceptable selectivities over αvß3 (IC50 ratios α5ß1/αvß3 between 0.090 and 0.157). In vitro staining of adipose-derived stem cells with Cy5-labeled peptides using confocal microscopy revealed strong binding of the α5ß1-selective bicycle CT3RGDcT3AWGCT3 to integrins in their natural environment, illustrating the high potential of these RGD bicycles as markers for α5ß1-integrin expression.


Subject(s)
Oligopeptides/analysis , Peptide Library , Receptors, Vitronectin/chemistry , Combinatorial Chemistry Techniques , Oligopeptides/chemical synthesis
10.
ACS Comb Sci ; 21(3): 198-206, 2019 03 11.
Article in English | MEDLINE | ID: mdl-30624885

ABSTRACT

We describe the identification of bicyclic RGD peptides with high affinity and selectivity for integrin αvß3 via high-throughput screening of partially randomized libraries. Peptide libraries (672 different compounds) comprising the universal integrin-binding sequence Arg-Gly-Asp (RGD) in the first loop and a randomized sequence XXX (X being one of 18 canonical l-amino acids) in the second loop, both enclosed by either an l- or d-Cys residue, were converted to bicyclic peptides via reaction with 1,3,5-tris(bromomethyl)benzene (T3). Screening of first-generation libraries yielded lead bicyclic inhibitors displaying submicromolar affinities for integrin αvß3 (e.g., CT3HEQcT3RGDcT3, IC50 = 195 nM). Next generation (second and third) libraries were obtained by partially varying the structure of the strongest lead inhibitors and screening for improved affinities and selectivities. In this way, we identified the highly selective bicyclic αvß3-binders CT3HPQcT3RGDcT3 (IC50 = 30 nM), CT3HPQCT3RGDcT3 (IC50 = 31 nM), and CT3HSQCT3RGDcT3 (IC50 = 42 nM) with affinities comparable to that of a knottin-RGD-type peptide (32 amino acids, IC50 = 38 nM) and outstanding selectivities over integrins αvß5 (IC50 > 10000 nM) and α5ß1 (IC50 > 10000 nM). Affinity measurements using surface plasmon-enhanced fluorescence spectroscopy (SPFS) yielded Kd values of 0.4 and 0.6 nM for the Cy5-labeled bicycle CT3HPQcT3RGDcT3 and RGD "knottin" peptide, respectively. In vitro staining of HT29 cells with Cy5-labeled bicycles using confocal microscopy revealed strong binding to integrins in their natural environment, which highlights the high potential of these peptides as markers of integrin expression.


Subject(s)
Integrin alphaVbeta3/chemistry , Oligopeptides/chemistry , Amino Acid Sequence , Amino Acids/chemistry , Gene Expression Regulation , HT29 Cells , High-Throughput Screening Assays/methods , Humans , Integrin alphaVbeta3/genetics , Optical Imaging/methods , Peptide Library , Protein Binding , Protein Conformation , Structure-Activity Relationship , Surface Properties
11.
PLoS One ; 14(1): e0209563, 2019.
Article in English | MEDLINE | ID: mdl-30615643

ABSTRACT

The nonspecific divalent cation channel TRPM7 (transient receptor potential-melastatin-like 7) is involved in many Ca2+ and Mg2+-dependent cellular processes, including survival, proliferation and migration. TRPM7 expression predicts metastasis and recurrence in breast cancer and several other cancers. In cultured cells, it can induce an invasive phenotype by promoting Ca2+-mediated epithelial-mesenchymal transition. We previously showed that in neuroblastoma cells that overexpress TRPM7 moderately, stimulation with Ca2+-mobilizing agonists leads to a characteristic sustained influx of Ca2+. Here we report that sustained influx through TRPM7 is abruptly abrogated by elevating intracellular levels of cyclic adenosine monophosphate (cAMP). Using pharmacological inhibitors and overexpression studies we show that this blockage is mediated by the cAMP effector Protein Kinase A (PKA). Mutational analysis demonstrates that the Serine residue S1269, which is present proximal to the coiled-coil domain within the protein c-terminus, is responsible for sensitivity to cAMP.


Subject(s)
Calcium/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Neurons/metabolism , TRPM Cation Channels/metabolism , Animals , Calcium Signaling/drug effects , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Isoquinolines/pharmacology , Mice , Neurons/drug effects , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Sulfonamides/pharmacology
12.
World J Urol ; 37(2): 309-315, 2019 Feb.
Article in English | MEDLINE | ID: mdl-29936567

ABSTRACT

PURPOSE: To determine the correlation of preoperative fascia thickness (FT) and intraoperative fascia preservation (FP) with erectile function (EF) after nerve-sparing robot-assisted radical prostatectomy (RARP). METHODS: Our analysis included 106 patients, with localized prostate cancer and no erectile dysfunction (ED) before RARP, assessed with preoperative 3 Tesla (3 T) multiparametric magnetic resonance imaging (MRI). FP score was defined as the extent of FP from the base to the apex of the prostate, quantitatively assessed by the surgeon. Median fascia thickness (MFT) per patient was defined as the sum of the median FT of 12 MRI regions. Preserved MFT (pMFT) was the sum of the saved MFT. The percentage of pFMT (ppMFT) was also calculated. Fascia surface (FS) was measured on MRI and it was combined with FP score resulting in preserved FS (pFS) and percentage of pFS (ppFS). RESULTS: FP score, pMFT, ppMFT, pFS and ppFS were significantly lower (p < 0.0001) in patients with ED. In the multivariate regression analysis, lower FP score [odds ratio (OR) 0.721, p = 0.03] and lower ppMFT (OR 0.001, p = 0.027) were independent predictors of ED. ROC analysis showed the highest area under the curve for ppMFT (0.787) and FP score (0.767) followed by pMFT (0.755) and ppFS (0.743). CONCLUSIONS: MRI-determined periprostatic FT combined with intraoperative FP score are correlated to postprostatectomy EF. Based on the hypothesis that a thicker fascia forms a protective layer for the nerves, we recommend assessing FT preoperatively to counsel men for the odds of preserving EF after RARP.


Subject(s)
Erectile Dysfunction/epidemiology , Fascia/diagnostic imaging , Peripheral Nerves , Postoperative Complications/epidemiology , Prostatectomy/methods , Prostatic Neoplasms/surgery , Robotic Surgical Procedures/methods , Area Under Curve , Fascia/anatomy & histology , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Odds Ratio , Organ Size , Organ Sparing Treatments , Prognosis , Prostatic Neoplasms/pathology , ROC Curve
13.
Glia ; 67(1): 68-77, 2019 01.
Article in English | MEDLINE | ID: mdl-30453391

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory disorder of the central nervous system (CNS), characterized by inflammation-mediated demyelination, axonal injury and neurodegeneration. The mechanisms underlying impaired neuronal function are not fully understood, but evidence is accumulating that the presence of the gliotic scar produced by reactive astrocytes play a critical role in these detrimental processes. Here, we identified astrocytic Transient Receptor Potential cation channel, subfamily M, member 7 (TRPM7), a Ca2+ -permeable nonselective cation channel, as a novel player in the formation of a gliotic scar. TRPM7 was found to be highly expressed in reactive astrocytes within well-characterized MS lesions and upregulated in primary astrocytes under chronic inflammatory conditions. TRPM7 overexpressing astrocytes impaired neuronal outgrowth in vitro by increasing the production of chondroitin sulfate proteoglycans, a key component of the gliotic scar. These findings indicate that astrocytic TRPM7 is a critical regulator of the formation of a gliotic scar and provide a novel mechanism by which reactive astrocytes affect neuronal outgrowth.


Subject(s)
Astrocytes/metabolism , Chondroitin Sulfate Proteoglycans/biosynthesis , Multiple Sclerosis/metabolism , Neurons/metabolism , Protein Serine-Threonine Kinases/biosynthesis , TRPM Cation Channels/biosynthesis , Adult , Aged , Aged, 80 and over , Animals , Cells, Cultured , Chondroitin Sulfate Proteoglycans/genetics , Female , Humans , Male , Middle Aged , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , Neurons/pathology , Protein Serine-Threonine Kinases/genetics , Rats , TRPM Cation Channels/genetics
14.
J Biol Chem ; 293(50): 19161-19176, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30381396

ABSTRACT

Chloride intracellular channel 4 (CLIC4) is a cytosolic protein implicated in diverse actin-based processes, including integrin trafficking, cell adhesion, and tubulogenesis. CLIC4 is rapidly recruited to the plasma membrane by RhoA-activating agonists and then partly colocalizes with ß1 integrins. Agonist-induced CLIC4 translocation depends on actin polymerization and requires conserved residues that make up a putative binding groove. However, the mechanism and significance of CLIC4 trafficking have been elusive. Here, we show that RhoA activation by either lysophosphatidic acid (LPA) or epidermal growth factor is necessary and sufficient for CLIC4 translocation to the plasma membrane and involves regulation by the RhoA effector mDia2, a driver of actin polymerization and filopodium formation. We found that CLIC4 binds the G-actin-binding protein profilin-1 via the same residues that are required for CLIC4 trafficking. Consistently, shRNA-induced profilin-1 silencing impaired agonist-induced CLIC4 trafficking and the formation of mDia2-dependent filopodia. Conversely, CLIC4 knockdown increased filopodium formation in an integrin-dependent manner, a phenotype rescued by wild-type CLIC4 but not by the trafficking-incompetent mutant CLIC4(C35A). Furthermore, CLIC4 accelerated LPA-induced filopodium retraction. We conclude that through profilin-1 binding, CLIC4 functions in a RhoA-mDia2-regulated signaling network to integrate cortical actin assembly and membrane protrusion. We propose that agonist-induced CLIC4 translocation provides a feedback mechanism that counteracts formin-driven filopodium formation.


Subject(s)
Carrier Proteins/metabolism , Chloride Channels/metabolism , Chlorides/metabolism , Profilins/metabolism , Pseudopodia/metabolism , Signal Transduction , rhoA GTP-Binding Protein/metabolism , Cell Membrane/metabolism , Chloride Channels/chemistry , Conserved Sequence , Crystallography, X-Ray , Enzyme Activation , Formins , HeLa Cells , Humans , Integrins/metabolism , Models, Molecular , Profilins/chemistry , Protein Binding , Protein Conformation , Protein Transport
15.
Cytometry A ; 93(10): 1029-1038, 2018 10.
Article in English | MEDLINE | ID: mdl-30176184

ABSTRACT

Cyclic AMP is a ubiquitous second messenger that orchestrates a variety of cellular functions over different timescales. The mechanisms underlying specificity within this signaling pathway are still not well understood. Several lines of evidence suggest the existence of spatial cAMP gradients within cells, and that compartmentalization underlies specificity within the cAMP signaling pathway. However, to date, no studies have visualized cAMP gradients in three spatial dimensions (3D: x, y, z).This is in part due to the limitations of FRET-based cAMP sensors, specifically the low signal-to-noise ratio intrinsic to all intracellular FRET probes. Here, we overcome this limitation, at least in part, by implementing spectral imaging approaches to estimate FRET efficiency when multiple fluorescent labels are used and when signals are measured from weakly expressed fluorescent proteins in the presence of background autofluorescence and stray light. Analysis of spectral image stacks in two spatial dimensions (2D) from single confocal slices indicates little or no cAMP gradients formed within pulmonary microvascular endothelial cells (PMVECs) under baseline conditions or following 10 min treatment with the adenylyl cyclase activator forskolin. However, analysis of spectral image stacks in 3D demonstrates marked cAMP gradients from the apical to basolateral face of PMVECs. Results demonstrate that spectral imaging approaches can be used to assess cAMP gradients-and in general gradients in fluorescence and FRET-within intact cells. Results also demonstrate that 2D imaging studies of localized fluorescence signals and, in particular, cAMP signals, whether using epifluorescence or confocal microscopy, may lead to erroneous conclusions about the existence and/or magnitude of gradients in either FRET or the underlying cAMP signals. Thus, with the exception of cellular structures that can be considered in one spatial dimension, such as neuronal processes, 3D measurements are required to assess mechanisms underlying compartmentalization and specificity within intracellular signaling pathways.


Subject(s)
Biosensing Techniques/instrumentation , Biosensing Techniques/methods , Cyclic AMP/metabolism , Fluorescence Resonance Energy Transfer/instrumentation , Fluorescence Resonance Energy Transfer/methods , Imaging, Three-Dimensional/instrumentation , Imaging, Three-Dimensional/methods , Animals , Cell Line , Endothelial Cells/metabolism , Male , Microscopy, Confocal/instrumentation , Microscopy, Confocal/methods , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Signal-To-Noise Ratio
16.
Nat Genet ; 50(8): 1151-1160, 2018 08.
Article in English | MEDLINE | ID: mdl-29988121

ABSTRACT

Chromatin folding contributes to the regulation of genomic processes such as gene activity. Existing conformation capture methods characterize genome topology through analysis of pairwise chromatin contacts in populations of cells but cannot discern whether individual interactions occur simultaneously or competitively. Here we present multi-contact 4C (MC-4C), which applies Nanopore sequencing to study multi-way DNA conformations of individual alleles. MC-4C distinguishes cooperative from random and competing interactions and identifies previously missed structures in subpopulations of cells. We show that individual elements of the ß-globin superenhancer can aggregate into an enhancer hub that can simultaneously accommodate two genes. Neighboring chromatin domain loops can form rosette-like structures through collision of their CTCF-bound anchors, as seen most prominently in cells lacking the cohesin-unloading factor WAPL. Here, massive collision of CTCF-anchored chromatin loops is believed to reflect 'cohesin traffic jams'. Single-allele topology studies thus help us understand the mechanisms underlying genome folding and functioning.


Subject(s)
Chromatin/genetics , Enhancer Elements, Genetic/genetics , Alleles , Animals , CCCTC-Binding Factor/genetics , Mice , Nucleic Acid Conformation , Regulatory Sequences, Nucleic Acid/genetics , beta-Globins/genetics
17.
Biochim Biophys Acta Mol Basis Dis ; 1864(7): 2409-2419, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29684587

ABSTRACT

Mechanically induced signaling pathways are important drivers of tumor progression. However, if and how mechanical signals affect metastasis or therapy response remains poorly understood. We previously found that the channel-kinase TRPM7, a regulator of cellular tension implicated in mechano-sensory processes, is required for breast cancer metastasis in vitro and in vivo. Here, we show that TRPM7 contributes to maintaining a mesenchymal phenotype in breast cancer cells by tensional regulation of the EMT transcription factor SOX4. The functional consequences of SOX4 knockdown closely mirror those produced by TRPM7 knockdown. By traction force measurements, we demonstrate that TRPM7 reduces cytoskeletal tension through inhibition of myosin II activity. Moreover, we show that SOX4 expression and downstream mesenchymal markers are inversely regulated by cytoskeletal tension and matrix rigidity. Overall, our results identify SOX4 as a transcription factor that is uniquely sensitive to cellular tension and indicate that TRPM7 may contribute to breast cancer progression by tensional regulation of SOX4.


Subject(s)
Breast Neoplasms/metabolism , Neoplasm Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , SOXC Transcription Factors/metabolism , TRPM Cation Channels/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cytoskeleton/genetics , Cytoskeleton/metabolism , Cytoskeleton/pathology , Female , Gene Knockdown Techniques , Humans , Myosin Type II/genetics , Myosin Type II/metabolism , Neoplasm Proteins/genetics , Protein Serine-Threonine Kinases/genetics , SOXC Transcription Factors/genetics , TRPM Cation Channels/genetics , Tensile Strength
18.
Elife ; 62017 08 29.
Article in English | MEDLINE | ID: mdl-28849762

ABSTRACT

The urokinase receptor (uPAR) is a glycosylphosphatidylinositol (GPI)-anchored protein that promotes tissue remodeling, tumor cell adhesion, migration and invasion. uPAR mediates degradation of the extracellular matrix through protease recruitment and enhances cell adhesion, migration and signaling through vitronectin binding and interactions with integrins. Full-length uPAR is released from the cell surface, but the mechanism and significance of uPAR shedding remain obscure. Here we identify transmembrane glycerophosphodiesterase GDE3 as a GPI-specific phospholipase C that cleaves and releases uPAR with consequent loss of function, whereas its homologue GDE2 fails to attack uPAR. GDE3 overexpression depletes uPAR from distinct basolateral membrane domains in breast cancer cells, resulting in a less transformed phenotype, it slows tumor growth in a xenograft model and correlates with prolonged survival in patients. Our results establish GDE3 as a negative regulator of the uPAR signaling network and, furthermore, highlight GPI-anchor hydrolysis as a cell-intrinsic mechanism to alter cell behavior.


Subject(s)
Breast Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Phosphoric Diester Hydrolases/genetics , Receptors, Urokinase Plasminogen Activator/genetics , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Adhesion , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Clustered Regularly Interspaced Short Palindromic Repeats , Female , Gene Knockout Techniques/methods , HEK293 Cells , Humans , Hydrolysis , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Mice, Nude , Models, Molecular , Neoplasm Transplantation , Phosphoric Diester Hydrolases/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Urokinase Plasminogen Activator/antagonists & inhibitors , Receptors, Urokinase Plasminogen Activator/metabolism , Signal Transduction , Tumor Burden , Vitronectin/genetics , Vitronectin/metabolism
19.
Nat Commun ; 8: 16068, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28703125

ABSTRACT

Clathrin lattices at the plasma membrane coat both invaginated and flat regions forming clathrin-coated pits and clathrin plaques, respectively. The function and regulation of clathrin-coated pits in endocytosis are well understood but clathrin plaques remain enigmatic nanodomains. Here we use super-resolution microscopy, molecular genetics and cell biology to show that clathrin plaques contain the machinery for clathrin-mediated endocytosis and cell adhesion, and associate with both clathrin-coated pits and filamentous actin. We also find that actin polymerization promoted by N-WASP through the Arp2/3 complex is crucial for the regulation of plaques but not pits. Clathrin plaques oppose cell migration and undergo actin- and N-WASP-dependent disassembly upon activation of LPA receptor 1, but not EGF receptor. Most importantly, plaque disassembly correlates with the endocytosis of LPA receptor 1 and down-modulation of AKT activity. Thus, clathrin plaques serve as dynamic actin-controlled hubs for clathrin-mediated endocytosis and signalling that exhibit receptor specificity.


Subject(s)
Actin Cytoskeleton/metabolism , Clathrin-Coated Vesicles/metabolism , Clathrin/physiology , Endocytosis , Actin-Related Protein 2-3 Complex/metabolism , HeLa Cells , Humans , Receptors, Lysophosphatidic Acid/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism
20.
Dis Model Mech ; 10(5): 597-603, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28067631

ABSTRACT

The neuromuscular junction is exposed to different types of insult, including mechanical trauma, toxins and autoimmune antibodies and, accordingly, has retained through evolution a remarkable ability to regenerate. Regeneration is driven by multiple signals that are exchanged among the cellular components of the junction. These signals are largely unknown. Miller Fisher syndrome is a variant of Guillain-Barré syndrome caused by autoimmune antibodies specific for epitopes of peripheral axon terminals. Using an animal model of Miller Fisher syndrome, we recently reported that a monoclonal anti-polysialoganglioside GQ1b antibody plus complement damages nerve terminals with production of mitochondrial hydrogen peroxide, which activates Schwann cells. Several additional signaling molecules are likely to be involved in the activation of the regeneration program in these cells. Using an in vitro cellular model consisting of co-cultured primary neurons and Schwann cells, we found that ATP is released by neurons injured by the anti-GQ1b antibody plus complement. Neuron-derived ATP acts as an alarm messenger for Schwann cells, where it induces the activation of intracellular pathways, including calcium signaling, cAMP and CREB, which, in turn, produce signals that promote nerve regeneration. These results contribute to defining the cross-talk taking place at the neuromuscular junction when it is attacked by anti-gangliosides autoantibodies plus complement, which is crucial for nerve regeneration and is also likely to be important in other peripheral neuropathies.


Subject(s)
Adenosine Triphosphate/metabolism , Miller Fisher Syndrome/pathology , Models, Biological , Neurons/metabolism , Schwann Cells/cytology , Animals , Coculture Techniques , In Vitro Techniques , Miller Fisher Syndrome/metabolism , Rats , Schwann Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...