Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 13437, 2024 06 11.
Article in English | MEDLINE | ID: mdl-38862601

ABSTRACT

The primary hurdles for small interference RNA (siRNA) in clinical use are targeted and cytosolic delivery. To overcome both challenges, we have established a novel platform based on phage display, called NNJA. In this approach, a lysosomal cathepsin substrate is engineered within the flexible loops of PIII, that is displaying a unique random sequence at its N-terminus. NNJA library selection targeting cell-expressed targets should yield specific peptides localized in the cytoplasm. That is because phage internalization and subsequent localization to lysosome, upon peptide binding to the cell expressed target, will result in cleavage of PIII, rendering phage non-infective. Such phage will be eliminated from the selected pool and only peptide-phage that escapes lysosomes will advance to the next round. Proof of concept studies with the NNJA library demonstrated cytosolic localization of selected peptide-phage and peptide-siRNA, confirmed through confocal microscopy. More importantly, conjugation of siHPRT to monomeric or multimeric NNJA peptides resulted in significant reduction in HPRT mRNA in various cell types without significant cytotoxicity. Sequence similarity and clustering analysis from NGS dataset provide insights into sequence composition facilitating cell penetration. NNJA platform offers a highly efficient peptide discovery engine for targeted delivery of oligonucleotides to cytosol.


Subject(s)
Cell-Penetrating Peptides , Peptide Library , RNA, Small Interfering , Cell-Penetrating Peptides/metabolism , Cell-Penetrating Peptides/chemistry , Humans , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Lysosomes/metabolism , Cell Surface Display Techniques/methods , Cytosol/metabolism
2.
J Exp Bot ; 75(10): 2829-2847, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38436428

ABSTRACT

Cell plate formation during cytokinesis entails multiple stages occurring concurrently and requiring orchestrated vesicle delivery, membrane remodelling, and timely deposition of polysaccharides, such as callose. Understanding such a dynamic process requires dissection in time and space; this has been a major hurdle in studying cytokinesis. Using lattice light sheet microscopy (LLSM), we studied cell plate development in four dimensions, through the behavior of yellow fluorescent protein (YFP)-tagged cytokinesis-specific GTPase RABA2a vesicles. We monitored the entire duration of cell plate development, from its first emergence, with the aid of YFP-RABA2a, in both the presence and absence of cytokinetic callose. By developing a robust cytokinetic vesicle volume analysis pipeline, we identified distinct behavioral patterns, allowing the identification of three easily trackable cell plate developmental phases. Notably, the phase transition between phase I and phase II is striking, indicating a switch from membrane accumulation to the recycling of excess membrane material. We interrogated the role of callose using pharmacological inhibition with LLSM and electron microscopy. Loss of callose inhibited the phase transitions, establishing the critical role and timing of the polysaccharide deposition in cell plate expansion and maturation. This study exemplifies the power of combining LLSM with quantitative analysis to decode and untangle such a complex process.


Subject(s)
Arabidopsis , Cytokinesis , Glucans , Arabidopsis/growth & development , Arabidopsis/metabolism , Glucans/metabolism , Microscopy
4.
Small Methods ; 6(6): e2200149, 2022 06.
Article in English | MEDLINE | ID: mdl-35344286

ABSTRACT

Quantifying molecular dynamics within the context of complex cellular morphologies is essential toward understanding the inner workings and function of cells. Fluorescence recovery after photobleaching (FRAP) is one of the most broadly applied techniques to measure the reaction diffusion dynamics of molecules in living cells. FRAP measurements typically restrict themselves to single-plane image acquisition within a subcellular-sized region of interest due to the limited temporal resolution and undesirable photobleaching induced by 3D fluorescence confocal or widefield microscopy. Here, an experimental and computational pipeline combining lattice light sheet microscopy, FRAP, and numerical simulations, offering rapid and minimally invasive quantification of molecular dynamics with respect to 3D cell morphology is presented. Having the opportunity to accurately measure and interpret the dynamics of molecules in 3D with respect to cell morphology has the potential to reveal unprecedented insights into the function of living cells.


Subject(s)
Molecular Dynamics Simulation , Diffusion , Fluorescence Recovery After Photobleaching/methods , Photobleaching
5.
Front Cell Dev Biol ; 9: 706126, 2021.
Article in English | MEDLINE | ID: mdl-34552926

ABSTRACT

The importance of mechanical force in biology is evident across diverse length scales, ranging from tissue morphogenesis during embryo development to mechanotransduction across single adhesion proteins at the cell surface. Consequently, many force measurement techniques rely on optical microscopy to measure forces being applied by cells on their environment, to visualize specimen deformations due to external forces, or even to directly apply a physical perturbation to the sample via photoablation or optogenetic tools. Recent developments in advanced microscopy offer improved approaches to enhance spatiotemporal resolution, imaging depth, and sample viability. These advances can be coupled with already existing force measurement methods to improve sensitivity, duration and speed, amongst other parameters. However, gaining access to advanced microscopy instrumentation and the expertise necessary to extract meaningful insights from these techniques is an unavoidable hurdle. In this Live Cell Imaging special issue Review, we survey common microscopy-based force measurement techniques and examine how they can be bolstered by emerging microscopy methods. We further explore challenges related to the accompanying data analysis in biomechanical studies and discuss the various resources available to tackle the global issue of technology dissemination, an important avenue for biologists to gain access to pre-commercial instruments that can be leveraged for biomechanical studies.

6.
Elife ; 102021 08 25.
Article in English | MEDLINE | ID: mdl-34431475

ABSTRACT

Upon inflammation, leukocytes rapidly transmigrate across the endothelium to enter the inflamed tissue. Evidence accumulates that leukocytes use preferred exit sites, alhough it is not yet clear how these hotspots in the endothelium are defined and how they are recognized by the leukocyte. Using lattice light sheet microscopy, we discovered that leukocytes prefer endothelial membrane protrusions at cell junctions for transmigration. Phenotypically, these junctional membrane protrusions are present in an asymmetric manner, meaning that one endothelial cell shows the protrusion and the adjacent one does not. Consequently, leukocytes cross the junction by migrating underneath the protruding endothelial cell. These protrusions depend on Rac1 activity and by using a photo-activatable Rac1 probe, we could artificially generate local exit-sites for leukocytes. Overall, we have discovered a new mechanism that uses local induced junctional membrane protrusions to facilitate/steer the leukocyte escape/exit from inflamed vessel walls.


Subject(s)
Gene Expression Regulation/physiology , Intercellular Junctions/physiology , Neutrophils/physiology , Animals , Cell Line , Green Fluorescent Proteins , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Muscle, Skeletal/physiology , Muscle, Skeletal/ultrastructure
8.
J Cell Sci ; 134(13)2021 07 01.
Article in English | MEDLINE | ID: mdl-34060624

ABSTRACT

The shuttling of transcription factors and transcriptional regulators into and out of the nucleus is central to the regulation of many biological processes. Here we describe a new method for studying the rates of nuclear entry and exit of transcriptional regulators. A photo-responsive LOV (light-oxygen-voltage) domain from Avena sativa is used to sequester fluorescently labelled transcriptional regulators YAP1 and TAZ (also known as WWTR1) on the surface of mitochondria and to reversibly release them upon blue light illumination. After dissociation, fluorescent signals from the mitochondria, cytoplasm and nucleus are extracted by a bespoke app and used to generate rates of nuclear entry and exit. Using this method, we demonstrate that phosphorylation of YAP1 on canonical sites enhances its rate of nuclear export. Moreover, we provide evidence that, despite high intercellular variability, YAP1 import and export rates correlate within the same cell. By simultaneously releasing YAP1 and TAZ from sequestration, we show that their rates of entry and exit are correlated. Furthermore, combining the optogenetic release of YAP1 with lattice light-sheet microscopy reveals high heterogeneity of YAP1 dynamics within different cytoplasmic regions, demonstrating the utility and versatility of our tool to study protein dynamics. This article has an associated First Person interview with Anna M. Dowbaj, joint first author of the paper.


Subject(s)
Cell Nucleus , Optogenetics , Active Transport, Cell Nucleus , Adaptor Proteins, Signal Transducing , Cell Nucleus/metabolism , Cytoplasm/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins
9.
J Cell Sci ; 134(6)2021 03 30.
Article in English | MEDLINE | ID: mdl-33785608

ABSTRACT

Recent technological advances have made microscopy indispensable in life science research. Its ubiquitous use, in turn, underscores the importance of ensuring that microscopy-based experiments are replicable and that the resulting data comparable. While there has been a wealth of review articles, practical guides and conferences devoted to the topic of maintaining standard instrument operating conditions, the paucity of attention dedicated to properly documenting microscopy experiments is undeniable. This lack of emphasis on accurate reporting extends beyond life science researchers themselves, to the review panels and editorial boards of many journals. Such oversight at the final step of communicating a scientific discovery can unfortunately negate the many valiant efforts made to ensure experimental quality control in the name of scientific reproducibility. This Review aims to enumerate the various parameters that should be reported in an imaging experiment by illustrating how their inconsistent application can lead to irreconcilable results.


Subject(s)
Microscopy , Reproducibility of Results
10.
Nature ; 591(7851): 659-664, 2021 03.
Article in English | MEDLINE | ID: mdl-33658713

ABSTRACT

Symmetric cell division requires the even partitioning of genetic information and cytoplasmic contents between daughter cells. Whereas the mechanisms coordinating the segregation of the genome are well known, the processes that ensure organelle segregation between daughter cells remain less well understood1. Here we identify multiple actin assemblies with distinct but complementary roles in mitochondrial organization and inheritance in mitosis. First, we find a dense meshwork of subcortical actin cables assembled throughout the mitotic cytoplasm. This network scaffolds the endoplasmic reticulum and organizes three-dimensional mitochondrial positioning to ensure the equal segregation of mitochondrial mass at cytokinesis. Second, we identify a dynamic wave of actin filaments reversibly assembling on the surface of mitochondria during mitosis. Mitochondria sampled by this wave are enveloped within actin clouds that can spontaneously break symmetry to form elongated comet tails. Mitochondrial comet tails promote randomly directed bursts of movement that shuffle mitochondrial position within the mother cell to randomize inheritance of healthy and damaged mitochondria between daughter cells. Thus, parallel mechanisms mediated by the actin cytoskeleton ensure both equal and random inheritance of mitochondria in symmetrically dividing cells.


Subject(s)
Actins/chemistry , Actins/metabolism , Mitochondria/metabolism , Mitosis , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/metabolism , Animals , Cell Division , Cell Line , Cytokinesis , Endoplasmic Reticulum/metabolism , Hippocampus/cytology , Hippocampus/embryology , Humans , Mitochondria/chemistry , Neurons , Rats
11.
Commun Biol ; 4(1): 142, 2021 01 29.
Article in English | MEDLINE | ID: mdl-33514834

ABSTRACT

The genetic and metabolic heterogeneity of RAS-driven cancers has confounded therapeutic strategies in the clinic. To address this, rapid and genetically tractable animal models are needed that recapitulate the heterogeneity of RAS-driven cancers in vivo. Here, we generate a Drosophila melanogaster model of Ras/Lkb1 mutant carcinoma. We show that low-level expression of oncogenic Ras (RasLow) promotes the survival of Lkb1 mutant tissue, but results in autonomous cell cycle arrest and non-autonomous overgrowth of wild-type tissue. In contrast, high-level expression of oncogenic Ras (RasHigh) transforms Lkb1 mutant tissue resulting in lethal malignant tumors. Using simultaneous multiview light-sheet microcopy, we have characterized invasion phenotypes of Ras/Lkb1 tumors in living larvae. Our molecular analysis reveals sustained activation of the AMPK pathway in malignant Ras/Lkb1 tumors, and demonstrate the genetic and pharmacologic dependence of these tumors on CaMK-activated Ampk. We further show that LKB1 mutant human lung adenocarcinoma patients with high levels of oncogenic KRAS exhibit worse overall survival and increased AMPK activation. Our results suggest that high levels of oncogenic KRAS is a driving event in the malignant transformation of LKB1 mutant tissue, and uncovers a vulnerability that may be used to target this aggressive genetic subset of RAS-driven tumors.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Genes, ras , Mutation , Neoplasms, Experimental/genetics , Protein Kinases/genetics , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases/metabolism , Adenocarcinoma of Lung/enzymology , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/mortality , Animals , Animals, Genetically Modified , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Death , Cell Movement , Databases, Genetic , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/enzymology , Enzyme Activation , Genetic Predisposition to Disease , Humans , Larva/enzymology , Larva/genetics , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Neoplasm Invasiveness , Neoplasms, Experimental/enzymology , Phenotype , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins p21(ras)/genetics
12.
J Leukoc Biol ; 108(2): 455-468, 2020 08.
Article in English | MEDLINE | ID: mdl-32323898

ABSTRACT

Neutrophil and macrophage (Mϕ) migration underpin the inflammatory response. However, the fast velocity, multidirectional instantaneous movement, and plastic, ever-changing shape of phagocytes confound high-resolution intravital imaging. Lattice lightsheet microscopy (LLSM) captures highly dynamic cell morphology at exceptional spatiotemporal resolution. We demonstrate the first extensive application of LLSM to leukocytes in vivo, utilizing optically transparent zebrafish, leukocyte-specific reporter lines that highlighted subcellular structure, and a wounding assay for leukocyte migration. LLSM revealed details of migrating leukocyte morphology, and permitted intricate, volumetric interrogation of highly dynamic activities within their native physiological setting. Very thin, recurrent uropod extensions must now be considered a characteristic feature of migrating neutrophils. LLSM resolved trailing uropod extensions, demonstrating their surprising length, and permitting quantitative assessment of cytoskeletal contributions to their evanescent form. Imaging leukocytes in blood vessel microenvironments at LLSM's spatiotemporal resolution displayed blood-flow-induced neutrophil dynamics and demonstrated unexpected leukocyte-endothelial interactions such as leukocyte-induced endothelial deformation against the intravascular pressure. LLSM of phagocytosis and cell death provided subcellular insights and uncovered novel behaviors. Collectively, we provide high-resolution LLSM examples of leukocyte structures (filopodia lamellipodia, uropod extensions, vesicles), and activities (interstitial and intravascular migration, leukocyte rolling, phagocytosis, cell death, and cytoplasmic ballooning). Application of LLSM to intravital leukocyte imaging sets the stage for transformative studies into the cellular and subcellular complexities of phagocyte biology.


Subject(s)
Chemotaxis, Leukocyte/physiology , Intravital Microscopy , Leukocytes/cytology , Leukocytes/physiology , Animals , Animals, Genetically Modified , Biomarkers , Cell Adhesion , Cell Death , Endothelium, Vascular/metabolism , Fluorescent Antibody Technique , Intravital Microscopy/methods , Macrophages/cytology , Macrophages/physiology , Models, Biological , Neutrophils/cytology , Neutrophils/physiology , Phagocytosis , Zebrafish
13.
J Cell Biol ; 219(5)2020 05 04.
Article in English | MEDLINE | ID: mdl-32294157

ABSTRACT

Filopodia are peripheral F-actin-rich structures that enable cell sensing of the microenvironment. Fascin is an F-actin-bundling protein that plays a key role in stabilizing filopodia to support efficient adhesion and migration. Fascin is also highly up-regulated in human cancers, where it increases invasive cell behavior and correlates with poor patient prognosis. Previous studies have shown that fascin phosphorylation can regulate F-actin bundling, and that this modification can contribute to subcellular fascin localization and function. However, the factors that regulate fascin dynamics within filopodia remain poorly understood. In the current study, we used advanced live-cell imaging techniques and a fascin biosensor to demonstrate that fascin phosphorylation, localization, and binding to F-actin are highly dynamic and dependent on local cytoskeletal architecture in cells in both 2D and 3D environments. Fascin dynamics within filopodia are under the control of formins, and in particular FMNL2, that binds directly to dephosphorylated fascin. Our data provide new insight into control of fascin dynamics at the nanoscale and into the mechanisms governing rapid cytoskeletal adaptation to environmental changes. This filopodia-driven exploration stage may represent an essential regulatory step in the transition from static to migrating cancer cells.


Subject(s)
Actins/genetics , Carrier Proteins/genetics , Formins/genetics , Microfilament Proteins/genetics , Neoplasms/genetics , Pseudopodia/genetics , Biosensing Techniques , Carrier Proteins/isolation & purification , Cell Adhesion/genetics , Cell Movement/genetics , Cellular Microenvironment/genetics , HeLa Cells , Humans , Microfilament Proteins/isolation & purification , Molecular Imaging , Neoplasms/pathology , Phosphorylation , Protein Binding/genetics , Pseudopodia/metabolism
14.
Development ; 146(19)2019 09 30.
Article in English | MEDLINE | ID: mdl-31570370

ABSTRACT

Histone post-translational modifications are key gene expression regulators, but their rapid dynamics during development remain difficult to capture. We applied a Fab-based live endogenous modification labeling technique to monitor the changes in histone modification levels during zygotic genome activation (ZGA) in living zebrafish embryos. Among various histone modifications, H3 Lys27 acetylation (H3K27ac) exhibited most drastic changes, accumulating in two nuclear foci in the 64- to 1k-cell-stage embryos. The elongating form of RNA polymerase II, which is phosphorylated at Ser2 in heptad repeats within the C-terminal domain (RNAP2 Ser2ph), and miR-430 transcripts were also concentrated in foci closely associated with H3K27ac. When treated with α-amanitin to inhibit transcription or JQ-1 to inhibit binding of acetyl-reader proteins, H3K27ac foci still appeared but RNAP2 Ser2ph and miR-430 morpholino were not concentrated in foci, suggesting that H3K27ac precedes active transcription during ZGA. We anticipate that the method presented here could be applied to a variety of developmental processes in any model and non-model organisms.


Subject(s)
Gene Expression Regulation, Developmental , Genome , Histones/metabolism , Lysine/metabolism , Transcription, Genetic , Zebrafish/embryology , Zebrafish/genetics , Zygote/metabolism , Acetylation/drug effects , Alpha-Amanitin/pharmacology , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental/drug effects , Histone Code/drug effects , RNA Polymerase II/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic/drug effects , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Zygote/drug effects
15.
J Cell Biol ; 218(9): 3153-3160, 2019 09 02.
Article in English | MEDLINE | ID: mdl-31444239

ABSTRACT

Lattice light-sheet microscopy (LLSM) is valuable for its combination of reduced photobleaching and outstanding spatiotemporal resolution in 3D. Using LLSM to image biosensors in living cells could provide unprecedented visualization of rapid, localized changes in protein conformation or posttranslational modification. However, computational manipulations required for biosensor imaging with LLSM are challenging for many software packages. The calculations require processing large amounts of data even for simple changes such as reorientation of cell renderings or testing the effects of user-selectable settings, and lattice imaging poses unique challenges in thresholding and ratio imaging. We describe here a new software package, named ImageTank, that is specifically designed for practical imaging of biosensors using LLSM. To demonstrate its capabilities, we use a new biosensor to study the rapid 3D dynamics of the small GTPase Rap1 in vesicles and cell protrusions.


Subject(s)
Biosensing Techniques , Fluorescence Resonance Energy Transfer , Human Umbilical Vein Endothelial Cells/metabolism , Image Processing, Computer-Assisted , Signal Transduction , Software , Telomere-Binding Proteins/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Humans , Microscopy, Fluorescence , Shelterin Complex , Telomere-Binding Proteins/genetics
16.
Mol Biol Cell ; 30(17): 2254-2267, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31242090

ABSTRACT

Macrophage fusion resulting in the formation of multinucleated giant cells occurs in a variety of chronic inflammatory diseases, yet the mechanism responsible for initiating this process is unknown. Here, we used live cell imaging to show that actin-based protrusions at the leading edge initiate macrophage fusion. Phase-contrast video microscopy demonstrated that in the majority of events, short protrusions (∼3 µm) between two closely apposed cells initiated fusion, but occasionally we observed long protrusions (∼12 µm). Using macrophages isolated from LifeAct mice and imaging with lattice light sheet microscopy, we further found that fusion-competent protrusions formed at sites enriched in podosomes. Inducing fusion in mixed populations of GFP- and mRFP-LifeAct macrophages showed rapid spatial overlap between GFP and RFP signal at the site of fusion. Cytochalasin B strongly reduced fusion and when rare fusion events occurred, protrusions were not observed. Fusion of macrophages deficient in Wiskott-Aldrich syndrome protein and Cdc42, key molecules involved in the formation of actin-based protrusions and podosomes, was also impaired both in vitro and in vivo. Finally, inhibiting the activity of the Arp2/3 complex decreased fusion and podosome formation. Together these data suggest that an actin-based protrusion formed at the leading edge initiates macrophage fusion.


Subject(s)
Actins/metabolism , Macrophages/metabolism , Podosomes/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Animals , Cell Communication , Cell Movement , Cytochalasin B/metabolism , Female , Male , Membrane Fusion/physiology , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence/methods , Wiskott-Aldrich Syndrome Protein/metabolism , cdc42 GTP-Binding Protein/metabolism
17.
Sci Immunol ; 4(33)2019 03 22.
Article in English | MEDLINE | ID: mdl-30902904

ABSTRACT

Cytotoxic T lymphocytes (CTLs) kill by forming immunological synapses with target cells and secreting toxic proteases and the pore-forming protein perforin into the intercellular space. Immunological synapses are highly dynamic structures that boost perforin activity by applying mechanical force against the target cell. Here, we used high-resolution imaging and microfabrication to investigate how CTLs exert synaptic forces and coordinate their mechanical output with perforin secretion. Using micropatterned stimulatory substrates that enable synapse growth in three dimensions, we found that perforin release occurs at the base of actin-rich protrusions that extend from central and intermediate locations within the synapse. These protrusions, which depended on the cytoskeletal regulator WASP and the Arp2/3 actin nucleation complex, were required for synaptic force exertion and efficient killing. They also mediated physical deformation of the target cell surface during CTL-target cell interactions. Our results reveal the mechanical basis of cellular cytotoxicity and highlight the functional importance of dynamic, three-dimensional architecture in immune cell-cell interfaces.


Subject(s)
Immunological Synapses/immunology , Perforin/immunology , T-Lymphocytes, Cytotoxic/immunology , Actin-Related Protein 2-3 Complex/immunology , Actins/immunology , Animals , Mice , Wiskott-Aldrich Syndrome Protein/immunology
18.
Nat Commun ; 10(1): 1249, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30890704

ABSTRACT

Phagocytosis of invading pathogens or cellular debris requires a dramatic change in cell shape driven by actin polymerization. For antibody-covered targets, phagocytosis is thought to proceed through the sequential engagement of Fc-receptors on the phagocyte with antibodies on the target surface, leading to the extension and closure of the phagocytic cup around the target. We find that two actin-dependent molecular motors, class 1 myosins myosin 1e and myosin 1f, are specifically localized to Fc-receptor adhesions and required for efficient phagocytosis of antibody-opsonized targets. Using primary macrophages lacking both myosin 1e and myosin 1f, we find that without the actin-membrane linkage mediated by these myosins, the organization of individual adhesions is compromised, leading to excessive actin polymerization, slower adhesion turnover, and deficient phagocytic internalization. This work identifies a role for class 1 myosins in coordinated adhesion turnover during phagocytosis and supports a mechanism involving membrane-cytoskeletal crosstalk for phagocytic cup closure.


Subject(s)
Actins/metabolism , Cell Adhesion/physiology , Myosin Type I/metabolism , Myosins/metabolism , Phagocytosis/physiology , Actins/ultrastructure , Animals , Bone Marrow Cells , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Female , Intravital Microscopy , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Microscopy, Fluorescence , Myosin Type I/genetics , Myosins/genetics , Primary Cell Culture , RAW 264.7 Cells , Receptors, Fc/metabolism , Receptors, Fc/ultrastructure , Time-Lapse Imaging
19.
J Neurosci Methods ; 312: 154-161, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30529411

ABSTRACT

BACKGROUND: Unbiased screening studies have repeatedly identified actin-related proteins as one of the families of proteins most influenced by neurotrauma. Nevertheless, the status quo model of cytoskeletal reorganization after neurotrauma excludes actin and incorporates only changes in microtubules and intermediate filaments. Actin is excluded in part because it is difficult to image with conventional techniques. However, recent innovations in fluorescent microscopy provide an opportunity to image the actin cytoskeleton at super-resolution resolution in living cells. This study applied these innovations to an in vitro model of neurotrauma. NEW METHOD: New methods are introduced for traumatizing neurons before imaging them with high speed structured illumination microscopy or lattice light sheet microscopy. Also, methods for analyzing structured illumination microscopy images to quantify post-traumatic neurite dystrophy are presented. RESULTS: Human induced pluripotent stem cell-derived neurons exhibited actin organization typical of immature neurons. Neurite dystrophy increased after trauma but was not influenced by jasplakinolide treatment. The F-actin content of dystrophies varied greatly from one dystrophy to another. COMPARISON WITH EXISTING METHODS: In contrast to fixation dependent methods, these methods capture the evolution of the actin cytoskeleton over time in a living cell. In contrast to prior methods based on counting dystrophies, this quantification scheme parameterizes the severity of a given dystrophy as it evolves from a local swelling to an almost-perfect spheroid that threatens to transect the neurite. CONCLUSIONS: These methods can be used to investigate genetic factors and therapeutic interventions that modulate the course of neurite dystrophy after trauma.


Subject(s)
Brain Injuries/diagnostic imaging , Microscopy, Fluorescence/instrumentation , Microscopy, Fluorescence/methods , Neurites/pathology , Neurons/pathology , Actin Cytoskeleton/pathology , Brain Injuries/pathology , Humans , Induced Pluripotent Stem Cells
20.
J Cell Biol ; 217(11): 3873-3885, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30150290

ABSTRACT

Pathogen-mediated activation of macrophages arms innate immune responses that include enhanced surface ruffling and macropinocytosis for environmental sampling and receptor internalization and signaling. Activation of macrophages with bacterial lipopolysaccharide (LPS) generates prominent dorsal ruffles, which are precursors for macropinosomes. Very rapid, high-resolution imaging of live macrophages with lattice light sheet microscopy (LLSM) reveals new features and actions of dorsal ruffles, which redefine the process of macropinosome formation and closure. We offer a new model in which ruffles are erected and supported by F-actin tent poles that cross over and twist to constrict the forming macropinosomes. This process allows for formation of large macropinosomes induced by LPS. We further describe the enrichment of active Rab13 on tent pole ruffles and show that CRISPR deletion of Rab13 results in aberrant tent pole ruffles and blocks the formation of large LPS-induced macropinosomes. Based on the exquisite temporal and spatial resolution of LLSM, we can redefine the ruffling and macropinosome processes that underpin innate immune responses.


Subject(s)
Actins/metabolism , Cell Membrane Structures/metabolism , Macrophages/metabolism , rab GTP-Binding Proteins/metabolism , Actins/genetics , Animals , CRISPR-Cas Systems , Cell Membrane Structures/genetics , Gene Deletion , Lipopolysaccharides/pharmacology , Mice , RAW 264.7 Cells , rab GTP-Binding Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...