Curated Optogenetic Publication Database

Search precisely and efficiently by using the advantage of the hand-assigned publication tags that allow you to search for papers involving a specific trait, e.g. a particular optogenetic switch or a host organism.

Showing 101 - 125 of 203 results
101.

Golgi-associated microtubules are fast cargo tracks and required for persistent cell migration.

blue AsLOV2 human retinal pigment epithelium cells Control of cytoskeleton / cell motility / cell shape
EMBO Rep, 27 Jan 2020 DOI: 10.15252/embr.201948385 Link to full text
Abstract: Microtubules derived from the Golgi (Golgi MTs) have been implicated to play critical roles in persistent cell migration, but the underlying mechanisms remain elusive, partially due to the lack of direct observation of Golgi MT-dependent vesicular trafficking. Here, using super-resolution stochastic optical reconstruction microscopy (STORM), we discovered that post-Golgi cargos are more enriched on Golgi MTs and also surprisingly move much faster than on non-Golgi MTs. We found that, compared to non-Golgi MTs, Golgi MTs are morphologically more polarized toward the cell leading edge with significantly fewer inter-MT intersections. In addition, Golgi MTs are more stable and contain fewer lattice repair sites than non-Golgi MTs. Our STORM/live-cell imaging demonstrates that cargos frequently pause at the sites of both MT intersections and MT defects. Furthermore, by optogenetic maneuvering of cell direction, we demonstrate that Golgi MTs are essential for persistent cell migration but not for cells to change direction. Together, our study unveils the role of Golgi MTs in serving as a group of "fast tracks" for anterograde trafficking of post-Golgi cargos.
102.

Optogenetics reveals Cdc42 local activation by scaffold-mediated positive feedback and Ras GTPase.

blue CRY2/CIB1 S. pombe Control of cytoskeleton / cell motility / cell shape
PLoS Biol, 24 Jan 2020 DOI: 10.1371/journal.pbio.3000600 Link to full text
Abstract: Local activity of the small GTPase Cdc42 is critical for cell polarization. Whereas scaffold-mediated positive feedback was proposed to break symmetry of budding yeast cells and produce a single zone of Cdc42 activity, the existence of similar regulation has not been probed in other organisms. Here, we address this problem using rod-shaped cells of fission yeast Schizosaccharomyces pombe, which exhibit zones of active Cdc42-GTP at both cell poles. We implemented the CRY2-CIB1 optogenetic system for acute light-dependent protein recruitment to the plasma membrane, which allowed to directly demonstrate positive feedback. Indeed, optogenetic recruitment of constitutively active Cdc42 leads to co-recruitment of the guanine nucleotide exchange factor (GEF) Scd1 and endogenous Cdc42, in a manner dependent on the scaffold protein Scd2. We show that Scd2 function is dispensable when the positive feedback operates through an engineered interaction between the GEF and a Cdc42 effector, the p21-activated kinase 1 (Pak1). Remarkably, this rewired positive feedback confers viability and allows cells to form 2 zones of active Cdc42 even when otherwise essential Cdc42 activators are lacking. These cells further revealed that the small GTPase Ras1 plays a role in both localizing the GEF Scd1 and promoting its activity, which potentiates the positive feedback. We conclude that scaffold-mediated positive feedback, gated by Ras activity, confers robust polarization for rod-shape formation.
103.

Optogenetic control of spine-head JNK reveals a role in dendritic spine regression.

blue AsLOV2 rat hippocampal neurons Control of cytoskeleton / cell motility / cell shape
eNeuro, 14 Jan 2020 DOI: 10.1523/eneuro.0303-19.2019 Link to full text
Abstract: In this study, we use an optogenetic inhibitor of JNK in dendritic spine sub-compartments of rat hippocampal neurons. JNK inhibition exerts rapid (within seconds) reorganisation of actin in the spine-head. Using real-time FRET to measure JNK activity, we find that either excitotoxic insult (NMDA) or endocrine stress (corticosterone), activate spine-head JNK causing internalization of AMPARs and spine retraction. Both events are prevented upon optogenetic inhibition of JNK, and rescued by JNK inhibition even 2 h after insult. Moreover, we identify that the fast-acting anti-depressant ketamine reduces JNK activity in hippocampal neurons suggesting that JNK inhibition may be a downstream mediator of its anti-depressant effect. In conclusion, we show that JNK activation plays a role in triggering spine elimination by NMDA or corticosterone stress, whereas inhibition of JNK facilitates regrowth of spines even in the continued presence of glucocorticoid. This identifies that JNK acts locally in the spine-head to promote AMPAR internalization and spine shrinkage following stress, and reveals a protective function for JNK inhibition in preventing spine regression.SIGNIFICANCE STATEMENT Identifying mechanisms that underlie dendritic spine elimination is important if we are to understand maladaptive changes that contribute to psychiatric disease. Compartment-specific, fast-acting tools can expedite this endeavor. Here we use a light-activated inhibitor of JNK to control kinase activity specifically in dendritic spines. Light-activation of the JNK inhibitor reduces AMPA receptor removal and spine regression in response to corticosterone and NMDA stress. Furthermore, we find that the anti-depressant drug ketamine lowers JNK activity in hippocampal neurons and prevents spine regression, though direct JNK inhibition is more effective. This study identifies a role for JNK in spine regression and may be relevant for endocrine control of synaptic strength and for conditions where chronic glucocorticoid stress leads to spine elimination.
104.

Dynamic organelle distribution initiates actin-based spindle migration in mouse oocytes.

blue iLID mouse oocytes Control of cytoskeleton / cell motility / cell shape
Nat Commun, 14 Jan 2020 DOI: 10.1038/s41467-019-14068-3 Link to full text
Abstract: Migration of meiosis-I (MI) spindle from the cell center to a sub-cortical location is a critical step for mouse oocytes to undergo asymmetric meiotic cell division. In this study, we investigate the mechanism by which formin-2 (FMN2) orchestrates the initial movement of MI spindle. By defining protein domains responsible for targeting FMN2, we show that spindle-periphery localized FMN2 is required for spindle migration. The spindle-peripheral FMN2 nucleates short actin bundles from vesicles derived likely from the endoplasmic reticulum (ER) and concentrated in a layer outside the spindle. This layer is in turn surrounded by mitochondria. A model based on polymerizing actin filaments pushing against mitochondria, thus generating a counter force on the spindle, demonstrated an inherent ability of this system to break symmetry and evolve directional spindle motion. The model is further supported through experiments involving spatially biasing actin nucleation via optogenetics and disruption of mitochondrial distribution and dynamics.
105.

Stick-slip dynamics of cell adhesion triggers spontaneous symmetry breaking and directional migration of mesenchymal cells on one-dimensional lines.

blue CRY2/CIB1 NIH/3T3 Control of cytoskeleton / cell motility / cell shape
Sci Adv, 3 Jan 2020 DOI: 10.1126/sciadv.aau5670 Link to full text
Abstract: Directional cell motility relies on the ability of single cells to establish a front-rear polarity and can occur in the absence of external cues. The initiation of migration has often been attributed to the spontaneous polarization of cytoskeleton components, while the spatiotemporal evolution of cell-substrate interaction forces has yet to be resolved. Here, we establish a one-dimensional microfabricated migration assay that mimics the complex in vivo fibrillar environment while being compatible with high-resolution force measurements, quantitative microscopy, and optogenetics. Quantification of morphometric and mechanical parameters of NIH-3T3 fibroblasts and RPE1 epithelial cells reveals a generic stick-slip behavior initiated by contractility-dependent stochastic detachment of adhesive contacts at one side of the cell, which is sufficient to trigger cell motility in 1D in the absence of pre-established polarity. A theoretical model validates the crucial role of adhesion dynamics, proposing that front-rear polarity can emerge independently of a complex self-polarizing system.
106.

RhoA Mediates Epithelial Cell Shape Changes via Mechanosensitive Endocytosis.

blue TULIP Caco-2 Signaling cascade control Control of cytoskeleton / cell motility / cell shape
Dev Cell, 26 Dec 2019 DOI: 10.1016/j.devcel.2019.12.002 Link to full text
Abstract: Epithelial remodeling involves ratcheting behavior whereby periodic contractility produces transient changes in cell-cell contact lengths, which stabilize to produce lasting morphogenetic changes. Pulsatile RhoA activity is thought to underlie morphogenetic ratchets, but how RhoA governs transient changes in junction length, and how these changes are rectified to produce irreversible deformation, remains poorly understood. Here, we use optogenetics to characterize responses to pulsatile RhoA in model epithelium. Short RhoA pulses drive reversible junction contractions, while longer pulses produce irreversible junction length changes that saturate with prolonged pulse durations. Using an enhanced vertex model, we show this is explained by two effects: thresholded tension remodeling and continuous strain relaxation. Our model predicts that structuring RhoA into multiple pulses overcomes the saturation of contractility and confirms this experimentally. Junction remodeling also requires formin-mediated E-cadherin clustering and dynamin-dependent endocytosis. Thus, irreversible junction deformations are regulated by RhoA-mediated contractility, membrane trafficking, and adhesion receptor remodeling.
107.

Mapping the proximity interaction network of the Rho-family GTPases reveals signalling pathways and regulatory mechanisms.

blue AsLOV2 HeLa Signaling cascade control Control of cytoskeleton / cell motility / cell shape
Nat Cell Biol, 23 Dec 2019 DOI: 10.1038/s41556-019-0438-7 Link to full text
Abstract: Guanine nucleotide exchange factors (RhoGEFs) and GTPase-activating proteins (RhoGAPs) coordinate the activation state of the Rho family of GTPases for binding to effectors. Here, we exploited proximity-dependent biotinylation to systematically define the Rho family proximity interaction network from 28 baits to produce 9,939 high-confidence proximity interactions in two cell lines. Exploiting the nucleotide states of Rho GTPases, we revealed the landscape of interactions with RhoGEFs and RhoGAPs. We systematically defined effectors of Rho proteins to reveal candidates for classical and atypical Rho proteins. We used optogenetics to demonstrate that KIAA0355 (termed GARRE here) is a RAC1 interactor. A functional screen of RHOG candidate effectors identified PLEKHG3 as a promoter of Rac-mediated membrane ruffling downstream of RHOG. We identified that active RHOA binds the kinase SLK in Drosophila and mammalian cells to promote Ezrin-Radixin-Moesin phosphorylation. Our proximity interactions data pave the way for dissecting additional Rho signalling pathways, and the approaches described here are applicable to the Ras family.
108.

Primary Cilia Signaling Promotes Axonal Tract Development and Is Disrupted in Joubert Syndrome-Related Disorders Models.

blue bPAC (BlaC) CRY2/CIB1 primary mouse deep cerebellar nuclei neurons Control of cytoskeleton / cell motility / cell shape Immediate control of second messengers
Dev Cell, 16 Dec 2019 DOI: 10.1016/j.devcel.2019.11.005 Link to full text
Abstract: Appropriate axonal growth and connectivity are essential for functional wiring of the brain. Joubert syndrome-related disorders (JSRD), a group of ciliopathies in which mutations disrupt primary cilia function, are characterized by axonal tract malformations. However, little is known about how cilia-driven signaling regulates axonal growth and connectivity. We demonstrate that the deletion of related JSRD genes, Arl13b and Inpp5e, in projection neurons leads to de-fasciculated and misoriented axonal tracts. Arl13b deletion disrupts the function of its downstream effector, Inpp5e, and deregulates ciliary-PI3K/AKT signaling. Chemogenetic activation of ciliary GPCR signaling and cilia-specific optogenetic modulation of downstream second messenger cascades (PI3K, AKT, and AC3) commonly regulated by ciliary signaling receptors induce rapid changes in axonal dynamics. Further, Arl13b deletion leads to changes in transcriptional landscape associated with dysregulated PI3K/AKT signaling. These data suggest that ciliary signaling acts to modulate axonal connectivity and that impaired primary cilia signaling underlies axonal tract defects in JSRD.
109.

Optogenetic control of cofilin and αTAT in living cells using Z-lock.

blue LOVTRAP HEK293T HeLa MTLn3 Control of cytoskeleton / cell motility / cell shape
Nat Chem Biol, 18 Nov 2019 DOI: 10.1038/s41589-019-0405-4 Link to full text
Abstract: Here we introduce Z-lock, an optogenetic approach for reversible, light-controlled steric inhibition of protein active sites. The light oxygen voltage (LOV) domain and Zdk, a small protein that binds LOV selectively in the dark, are appended to the protein of interest where they sterically block the active site. Irradiation causes LOV to change conformation and release Zdk, exposing the active site. Computer-assisted protein design was used to optimize linkers and Zdk-LOV affinity, for both effective binding in the dark, and effective light-induced release of the intramolecular interaction. Z-lock cofilin was shown to have actin severing ability in vitro, and in living cancer cells it produced protrusions and invadopodia. An active fragment of the tubulin acetylase αTAT was similarly modified and shown to acetylate tubulin on irradiation.
110.

Locally Activating TrkB Receptor Generates Actin Waves and Specifies Axonal Fate.

blue AsLOV2 CRY2/CRY2 rat hippocampal neurons Control of cytoskeleton / cell motility / cell shape
Cell Chem Biol, 24 Oct 2019 DOI: 10.1016/j.chembiol.2019.10.006 Link to full text
Abstract: Actin waves are filamentous actin (F-actin)-rich structures that initiate in the somato-neuritic area and move toward neurite ends. The upstream cues that initiate actin waves are poorly understood. Here, using an optogenetic approach (Opto-cytTrkB), we found that local activation of the TrkB receptor around the neurite end initiates actin waves and triggers neurite elongation. During actin wave generation, locally activated TrkB signaling in the distal neurite was functionally connected with preferentially localized Rac1 and its signaling pathways in the proximal region. Moreover, TrkB activity changed the location of ankyrinG--the master organizer of the axonal initial segment-and initiated the stimulated neurite to acquire axonal characteristics. Taken together, these findings suggest that local Opto-cytTrkB activation switches the fate from minor to major axonal neurite during neuronal polarization by generating actin waves.
111.

Chemokine Receptor CCR7 Triggers an Endomembrane Signaling Complex for Spatial Rac Activation.

blue AsLOV2 HEK293 Control of cytoskeleton / cell motility / cell shape
Cell Rep, 22 Oct 2019 DOI: 10.1016/j.celrep.2019.09.031 Link to full text
Abstract: Chemokine-guided cell migration is pivotal for many immunological and developmental processes. How chemokine receptor signaling persists to guarantee sustained directional migration despite receptor desensitization and internalization remains poorly understood. Here, we uncover a function for an intracellular pool of the chemokine receptor CCR7 present in human dendritic cells and cellular model systems. We find that CCR7 signaling, initiated at the plasma membrane, is translocated by joint trafficking of β-arrestin and Src kinase to endomembrane-residing CCR7. There, Src tyrosine phosphorylates CCR7, required for the recruitment of Vav1 to form an endomembrane-residing multi-protein signaling complex comprising CCR7, the RhoGEF Vav1, and its effector, Rac1. Interfering with vesicular trafficking affects CCR7-driven cell migration, whereas CCR7:Vav1 interaction at endomembranes is essential for local Rac1 recruitment to CCR7. Photoactivation of Rac1 at endomembranes leads to lamellipodia formation at the cell's leading edge, supporting the role of sustained endomembrane signaling in guiding cell migration.
112.

Mechanosensitive junction remodelling promotes robust epithelial morphogenesis.

blue TULIP Caco-2 Control of cytoskeleton / cell motility / cell shape
Lancet Diabetes Endocrinol, 28 Sep 2019 DOI: 10.1016/j.bpj.2019.09.027 Link to full text
Abstract: Morphogenesis of epithelial tissues requires tight spatiotemporal coordination of cell shape changes. In vivo, many tissue-scale shape changes are driven by pulsatile contractions of intercellular junctions, which are rectified to produce irreversible deformations. The functional role of this pulsatory ratchet and its mechanistic basis remain unknown. Here we combine theory and biophysical experiments to show that mechanosensitive tension remodelling of epithelial cell junctions promotes robust epithelial shape changes via ratcheting. Using optogenetic control of actomyosin contractility, we find that epithelial junctions show elastic behaviour under low contractile stress, returning to their original lengths after contraction, but undergo irreversible deformation under higher magnitudes of contractile stress. Existing vertex-based models for the epithelium are unable to capture these results, with cell junctions displaying purely elastic or fluid-like behaviours, depending on the choice of model parameters. To describe the experimental results, we propose a modified vertex model with two essential ingredients for junction mechanics: thresholded tension remodelling and continuous strain relaxation. First, a critical strain threshold for tension remodelling triggers irreversible junction length changes for sufficiently strong contractions, making the system robust to small fluctuations in contractile activity. Second, continuous strain relaxation allows for mechanical memory removal, enabling frequency-dependent modulation of cell shape changes via mechanical ratcheting. Taken together, the combination of mechanosensitive tension remodelling and junctional strain relaxation provides a robust mechanism for large-scale morphogenesis.
113.

Photocleavable Cadherin Inhibits Cell-to-Cell Mechanotransduction by Light.

violet PhoCl MCF7 MDCK Control of cytoskeleton / cell motility / cell shape
ACS Chem Biol, 20 Sep 2019 DOI: 10.1021/acschembio.9b00460 Link to full text
Abstract: Precise integration of individual cell behaviors is indispensable for collective tissue morphogenesis and maintenance of tissue integrity. Organized multicellular behavior is achieved via mechanical coupling of individual cellular contractility, mediated by cell adhesion molecules at the cell-cell interface. Conventionally, gene depletion or laser microsurgery has been used for functional analysis of intercellular mechanotransduction. Nevertheless, these methods are insufficient to investigate either the spatiotemporal dynamics or the biomolecular contribution in cell-cell mechanical coupling within collective multicellular behaviors. Herein, we present our effort in adaption of PhoCl for attenuation of cell-to-cell tension transmission mediated by E-cadherin. To release intercellular contractile tension applied on E-cadherin molecules with external light, a genetically encoded photocleavable module called PhoCl was inserted into the intracellular domain of E-cadherin, thereby creating photocleavable cadherin (PC-cadherin). In response to light illumination, the PC-cadherin cleaved into two fragments inside cells, resulting in attenuating mechanotransduction at intercellular junctions in living epithelial cells. Light-induced perturbation of the intercellular tension balance with surrounding cells changed the cell shape in an epithelial cell sheet. The method is expected to enable optical manipulation of force-mediated cell-to-cell communications in various multicellular behaviors, which contributes to a deeper understanding of embryogenesis and oncogenesis.
114.

Secretory Vesicle Clustering in Fungal Filamentous Cells Does Not Require Directional Growth.

blue CRY2/CIB1 C. albicans Control of cytoskeleton / cell motility / cell shape
Cell Rep, 20 Aug 2019 DOI: 10.1016/j.celrep.2019.07.062 Link to full text
Abstract: During symmetry breaking, the highly conserved Rho GTPase Cdc42 becomes stabilized at a defined site via an amplification process. However, little is known about how a new polarity site is established in an already asymmetric cell-a critical process in a changing environment. The human fungal pathogen Candida albicans switches from budding to filamentous growth in response to external cues, a transition controlled by Cdc42. Here, we have used optogenetic manipulation of cell polarity to reset growth in asymmetric filamentous C. albicans cells. We show that increasing the level of active Cdc42 on the plasma membrane results in disruption of the exocyst subunit Sec3 localization and a striking de novo clustering of secretory vesicles. This new cluster of secretory vesicles is highly dynamic, moving by hops and jumps, until a new growth site is established. Our results reveal that secretory vesicle clustering can occur in the absence of directional growth.
115.

Transient Activations of Rac1 at the Lamellipodium Tip Trigger Membrane Protrusion.

blue CRY2/CIB1 MEF-1 Control of cytoskeleton / cell motility / cell shape
Curr Biol, 8 Aug 2019 DOI: 10.1016/j.cub.2019.07.035 Link to full text
Abstract: The spatiotemporal coordination of actin regulators in the lamellipodium determines the dynamics and architecture of branched F-actin networks during cell migration. The WAVE regulatory complex (WRC), an effector of Rac1 during cell protrusion, is concentrated at the lamellipodium tip. Thus, activated Rac1 should operate at this location to activate WRC and trigger membrane protrusion. Yet correlation of Rho GTPase activation with cycles of membrane protrusion previously revealed complex spatiotemporal patterns of Rac1 and RhoA activation in the lamellipodium. Combining single protein tracking (SPT) and super-resolution imaging with loss- or gain-of-function mutants of Rho GTPases, we show that Rac1 immobilizations at the lamellipodium tip correlate with its activation, in contrast to RhoA. Using Rac1 effector loop mutants and wild-type versus mutant variants of WRC, we show that selective immobilizations of activated Rac1 at the lamellipodium tip depend on effector binding, including WRC. In contrast, wild-type Rac1 only displays slower diffusion at the lamellipodium tip, suggesting transient activations. Local optogenetic activation of Rac1, triggered by membrane recruitment of Tiam1, shows that Rac1 activation must occur close to the lamellipodium tip and not behind the lamellipodium to trigger efficient membrane protrusion. However, coupling tracking with optogenetic activation of Rac1 demonstrates that diffusive properties of wild-type Rac1 are unchanged despite enhanced lamellipodium protrusion. Taken together, our results support a model whereby transient activations of Rac1 occurring close to the lamellipodium tip trigger WRC binding. This short-lived activation ensures a local and rapid control of Rac1 actions on its effectors to trigger actin-based protrusion.
116.

Coordination of protrusion dynamics within and between collectively migrating border cells by myosin II.

blue AsLOV2 D. melanogaster in vivo Control of cytoskeleton / cell motility / cell shape
Mol Biol Cell, 7 Aug 2019 DOI: 10.1091/mbc.e19-02-0124 Link to full text
Abstract: Collective cell migration is emerging as a major driver of embryonic development, organogenesis, tissue homeostasis, and tumor dissemination. In contrast to individually migrating cells, collectively migrating cells maintain cell-cell adhesions and coordinate direction-sensing as they move. While non-muscle myosin II has been studied extensively in the context of cells migrating individually in vitro, its roles in cells migrating collectively in three-dimensional, native environments are not fully understood. Here we use genetics, Airyscan microscopy, live imaging, optogenetics, and Förster resonance energy transfer to probe the localization, dynamics, and functions of myosin II in migrating border cells of the Drosophila ovary. We find that myosin accumulates transiently at the base of protrusions, where it functions to retract them. E-cadherin and myosin co-localize at border cell-border cell contacts and cooperate to transmit directional information. A phosphomimetic form of myosin is sufficient to convert border cells to a round morphology and blebbing migration mode. Together these studies demonstrate that distinct and dynamic pools of myosin II regulate protrusion dynamics within and between collectively migrating cells and suggest a new model for the role of protrusions in collective direction sensing in vivo. Movie S1 Movie S1 Live imaging of border cell specification and delamination from anterior epithelium From Figure 1D-I. Slbo promoter driving Lifeact-GFP (green) marks border cells, Upd-Gal4, UAS-DsRed.nls (red) mark polar cell nuclei. Hoechst 33342 (blue) marks DNA. Time resolution is 4 min. Movie S2 Movie S2 Representative Z-projected and registered live imaging of Sqh-mCherry accumulating in cortical junctions (flashing arrows) during border cell migration. From Figure 3J-K. Time resolution is 25 sec. Movie S3 Movie S3 Representative Z-projected and registered live imaging of E-cad-GFP during border cell migration. From Figure 3M-N. Time resolution is 60 sec. Movie S4 Movie S4 Representative Z-projection of control flpout cells from hs-Flp;, Slbo>Lifeact-GFP; AyGal4, UAS-RFP. Clonal cells are marked by magenta nuclei (nls-RFP). Time resolution is 2.5 min. From Supp. Figure 3 A-D. Movie S5 Movie S5 Representative Z-projection of Sqh-RNAi flpout cells from hs-Flp;, Slbo>Lifeact-GFP; AyGal4, UAS-RFP, UAS-sqh-RNAi. Clonal cells are marked by magenta nuclei (nls-RFP). Time resolution is 2.5 min. From Supp. Figure 3 E-H. Movie S6 Movie S6 Representative Z-projected c306-Gal4; tub-GAL80ts driving UAS-Lifeact-GFP and UAS-white RNAi. Time resolution is 2 min. From Supp. Figure 4 A-D. Movie S7 Movie S7 Representative Z-projected c306-Gal4; tub-GAL80ts driving UAS-Lifeact-GFP and UAS-sqh-RNAi showing frequent side protrusions. Time resolution is 2 min. From Supp. Figure 4 E-H. White arrows indicate ectopic side and rear protrusions. Movie S8 Movie S8 Representative Z-projected c306-Gal4; tub-GAL80ts driving UAS-Lifeact-GFP and UAS-sqh-RNAi showing long lived side protrusions. Time resolution is 2 min. From Supp. Figure 4 I-L. Movie S9 Movie S9 Representative Z-projected live imaging of c306-Gal4 driving UAS-white-RNAi in clusters co-expressing Lifeact-GFP under the control of the slbo enhancer and Sqh-mCherry from its endogenous promoter during periods of protrusive and round migration phases. From Figure 6A-D. 25 min corresponds to 6A and B and 1hr:25 min corresponds to 6C and D. Time resolution is 2.5 min. Movie S10 Movie S10 Sqh-mCherry (magenta) channel from Supplementary Movie 9. From Figure 6A-D. 25 min corresponds to 6A and B and 1hr:25 min corresponds to 6C and D. Time resolution is 2.5 min. Movie S11 Movie S11 Representative Z-projected live imaging of c306-Gal4 driving UAS-Ecad-RNAi in clusters co-expressing Lifeact-GFP under the control of the slbo enhancer and Sqh-mCherry from its endogenous promoter during a protrusive phase of migration. From Figure 6E-F. Time resolution is 2.5 min. Movie S12 Movie S12 Sqh-mCherry (magenta) channel from Supplementary Movie 11. From Figure 6E-F. Time resolution is 2.5 min. Movie S13 Movie S13 Representative Z-projected live imaging of c306-Gal4 driving UAS-Ecad-RNAi in clusters co-expressing Lifeact-GFP under the control of the slbo enhancer and Sqh-mCherry from its endogenous promoter during a rounded phase of migration. From Figure 6G-H. Time resolution is 2.5 min. Movie S14 Movie S14 Sqh-mCherry (magenta) channel from Supplementary Movie 13. From Figure 6G-H. Time resolution is 2.5 min. Movie S15 Movie S15 Example segmentation analysis from a representative Z-projected time lapse of a cluster expressing c306-Gal4 driving UAS-white-RNAi in clusters co-expressing Lifeact-GFP under the control of the slbo enhancer and Sqh-mCherry from its endogenous promoter during migration. Time lapse analyzed in Imaris by 1. segmentation of the cluster using Lifeact-GFP, 2. Rendering of Sqh-mCherry by masking the inside of the Life-act surface, 3. performing a distance transformation using the masked Sqh-mCherry that is color coded for distance from membrane (dark colors are short distances and bright/white colors are more distant), 4. combining the distance transformation with the Sqh-mCherry mask to only include the cortical 2 μm of the original Sqh-mCherry signal for quantification in Figure 6I. Movie S16 Movie S16 Representative Z-projected time lapse of Lifeact-GFP and Sqh-mCherry expressing clusters used for quantification of Figure 7B-C during protrusion/retractions cycles. Time resolution is 2 min. Movie S17 Movie S17 Sqh-mCherry channel from Supplementary movie 16. Time resolution is 2 min. Movie S18 Movie S18 Representative Z-projections of Lifeact-GFP (green) in c306-Gal4; tub-GAL80ts driving UAS-Lifeact-GFP and UAS-Sqh-E20E21 migrating border cells clusters that split. Time resolution is 2 min. Movie S19 Movie S19 Representative Z-projections of Lifeact-GFP (green) in c306-Gal4; tub-GAL80ts driving UAS-LifeactGFP and UAS-Sqh-E20E21 migrating border cells clusters during protrusive phase. Time resolution is 2 min. Movie S20 Movie S20 Representative Z-projection of Lifeact-GFP (green) in c306-Gal4; tub-GAL80ts driving UAS-Lifeact-GFP and UAS-Sqh-E20E21 border cells cluster at the oocyte border during a blebbing phase. Time resolution is 2 min. Movie S21 Movie S21 Representative Z-projection of control cluster expressing slbo-Gal4; UAS-PLCδ1-PH-GFP. Time resolution is 2 min. Movie S22 Movie S22 Representative Z-projection of cluster expressing slbo-Gal4; UAS-PLCδ1-PH-GFP, UAS-Rho1V14. Blebs are marked by white arrows. Time resolution is 2 min.
117.

VIEW-MOD: a versatile illumination engine with a modular optical design for fluorescence microscopy.

blue AsLOV2 MEF-1 Control of cytoskeleton / cell motility / cell shape
Opt Express, 8 Jul 2019 DOI: 10.1364/oe.27.019950 Link to full text
Abstract: We developed VIEW-MOD (Versatile Illumination Engine with a Modular Optical Design): a compact, multi-modality microscope, which accommodates multiple illumination schemes including variable angle total internal reflection, point scanning and vertical/horizontal light sheet. This system allows combining and flexibly switching between different illuminations and imaging modes by employing three electrically tunable lenses and two fast-steering mirrors. This versatile optics design provides control of 6 degrees of freedom of the illumination source (3 translation, 2 tilt, and beam shape) plus the axial position of the imaging plane. We also developed standalone software with an easy-to-use GUI to calibrate and control the microscope. We demonstrate the applications of this system and software in biosensor imaging, optogenetics and fast 3D volume imaging. This system is ready to fit into complex imaging circumstances requiring precise control of illumination and detection paths, and has a broad scope of usability for a myriad of biological applications.
118.

Cross-linker-mediated regulation of actin network organization controls tissue morphogenesis.

blue CRY2/CIB1 D. melanogaster in vivo Control of cytoskeleton / cell motility / cell shape
J Cell Biol, 28 Jun 2019 DOI: 10.1083/jcb.201811127 Link to full text
Abstract: Contraction of cortical actomyosin networks driven by myosin activation controls cell shape changes and tissue morphogenesis during animal development. In vitro studies suggest that contractility also depends on the geometrical organization of actin filaments. Here we analyze the function of actomyosin network topology in vivo using optogenetic stimulation of myosin-II in Drosophila embryos. We show that early during cellularization, hexagonally arrayed actomyosin fibers are resilient to myosin-II activation. Actomyosin fibers then acquire a ring-like conformation and become contractile and sensitive to myosin-II. This transition is controlled by Bottleneck, a Drosophila unique protein expressed for only a short time during early cellularization, which we show regulates actin bundling. In addition, it requires two opposing actin cross-linkers, Filamin and Fimbrin. Filamin acts synergistically with Bottleneck to facilitate hexagonal patterning, while Fimbrin controls remodeling of the hexagonal network into contractile rings. Thus, actin cross-linking regulates the spatio-temporal organization of actomyosin contraction in vivo, which is critical for tissue morphogenesis.
119.

Cyclic Nucleotide-Specific Optogenetics Highlights Compartmentalization of the Sperm Flagellum into cAMP Microdomains.

blue red bPAC (BlaC) LAPD HEK293 mouse sperm cells Signaling cascade control Control of cytoskeleton / cell motility / cell shape Immediate control of second messengers
Cells, 27 Jun 2019 DOI: 10.3390/cells8070648 Link to full text
Abstract: Inside the female genital tract, mammalian sperm undergo a maturation process called capacitation, which primes the sperm to navigate across the oviduct and fertilize the egg. Sperm capacitation and motility are controlled by 3',5'-cyclic adenosine monophosphate (cAMP). Here, we show that optogenetics, the control of cellular signaling by genetically encoded light-activated proteins, allows to manipulate cAMP dynamics in sperm flagella and, thereby, sperm capacitation and motility by light. To this end, we used sperm that express the light-activated phosphodiesterase LAPD or the photo-activated adenylate cyclase bPAC. The control of cAMP by LAPD or bPAC combined with pharmacological interventions provides spatiotemporal precision and allows to probe the physiological function of cAMP compartmentalization in mammalian sperm.
120.

Self-Organized Nuclear Positioning Synchronizes the Cell Cycle in Drosophila Embryos.

blue CRY2/CIB1 D. melanogaster in vivo Control of cytoskeleton / cell motility / cell shape Organelle manipulation
Cell, 9 Apr 2019 DOI: 10.1016/j.cell.2019.03.007 Link to full text
Abstract: The synchronous cleavage divisions of early embryogenesis require coordination of the cell-cycle oscillator, the dynamics of the cytoskeleton, and the cytoplasm. Yet, it remains unclear how spatially restricted biochemical signals are integrated with physical properties of the embryo to generate collective dynamics. Here, we show that synchronization of the cell cycle in Drosophila embryos requires accurate nuclear positioning, which is regulated by the cell-cycle oscillator through cortical contractility and cytoplasmic flows. We demonstrate that biochemical oscillations are initiated by local Cdk1 inactivation and spread through the activity of phosphatase PP1 to generate cortical myosin II gradients. These gradients cause cortical and cytoplasmic flows that control proper nuclear positioning. Perturbations of PP1 activity and optogenetic manipulations of cortical actomyosin disrupt nuclear spreading, resulting in loss of cell-cycle synchrony. We conclude that mitotic synchrony is established by a self-organized mechanism that integrates the cell-cycle oscillator and embryo mechanics.
121.

Cortical mitochondria regulate insulin secretion by local Ca2+ buffering.

blue CRY2/CIB1 INS-1E MIN6 Control of cytoskeleton / cell motility / cell shape Organelle manipulation
J Cell Sci, 29 Mar 2019 DOI: 10.1242/jcs.228544 Link to full text
Abstract: Mitochondria play an essential role in regulating insulin secretion from beta cells by providing ATP needed for the membrane depolarization that results in voltage-dependent Ca2+ influx and subsequent insulin granule exocytosis. Ca2+, in turn, is also rapidly taken up by the mitochondria and exerts important feedback regulation of metabolism. The aim of this study was to determine if the distribution of mitochondria within beta cells is important for the secretory capacity of these cells. We find that cortically localized mitochondria are abundant in beta cells, and that these mitochondria redistribute towards the cell interior following depolarization. The redistribution requires Ca2+-induced remodeling of the cortical F-actin network. Using light-regulated motor proteins, we increased the cortical density of mitochondria 2-fold and found that this blunted the voltage-dependent increase in cytosolic Ca2+ concentration and suppressed insulin secretion. The activity-dependent changes in mitochondria distribution are likely important for the generation of Ca2+ microdomains required for efficient insulin granule release.
122.

Optogenetic control of cell morphogenesis on protein micropatterns.

blue AsLOV2 HFF-1 Control of cytoskeleton / cell motility / cell shape
bioRxiv, 4 Mar 2019 DOI: 10.1101/563353 Link to full text
Abstract: Cell morphogenesis is critical for embryonic development, tissue formation, and wound healing. Our ability to manipulate endogenous mechanisms to control cell shape, however, remains limited. Here we combined surface micropatterning of adhesion molecules with optogenetic activation of intracellular signaling pathways to control the nature and morphology of cellular protrusions. We employed geometry-dependent pre-organization of cytoskeletal structures together with acute activation of signaling pathways that control actin assembly to create a tool capable of generating membrane protrusions at defined cellular locations. Further, we find that the size of microfabricated patterns of adhesion molecules influences the molecular mechanism of cell protrusion: larger patterns enable cells to create actin-filled lamellipodia while smaller patterns promote formation of spherical blebs. Optogenetic perturbation of signaling pathways in these cells changes the size of blebs and convert them into lamellipodia. Our results demonstrate how the coordinated manipulation of adhesion geometry and cytoskeletal dynamics can be used to control membrane protrusion and cell morphogenesis.
123.

Physical Plasma Membrane Perturbation Using Subcellular Optogenetics Drives Integrin-Activated Cell Migration.

blue CRY2/CIB1 iLID RAW264.7 Control of cytoskeleton / cell motility / cell shape
ACS Synth Biol, 22 Feb 2019 DOI: 10.1021/acssynbio.8b00356 Link to full text
Abstract: Cells experience physical deformations to the plasma membrane that can modulate cell behaviors like migration. Understanding the molecular basis for how physical cues affect dynamic cellular responses requires new approaches that can physically perturb the plasma membrane with rapid, reversible, subcellular control. Here we present an optogenetic approach based on light-inducible dimerization that alters plasma membrane properties by recruiting cytosolic proteins at high concentrations to a target site. Surprisingly, this polarized accumulation of proteins in a cell induces directional amoeboid migration in the opposite direction. Consistent with known effects of constraining high concentrations of proteins to a membrane in vitro, there is localized curvature and tension decrease in the plasma membrane. Integrin activity, sensitive to mechanical forces, is activated in this region. Localized mechanical activation of integrin with optogenetics allowed simultaneous imaging of the molecular and cellular response, helping uncover a positive feedback loop comprising SFK- and ERK-dependent RhoA activation, actomyosin contractility, rearward membrane flow, and membrane tension decrease underlying this mode of cell migration.
124.

Intensiometric biosensors visualize the activity of multiple small GTPases in vivo.

blue CRY2/CRY2 MDA-MB-231 rat hippocampal neurons Signaling cascade control Control of cytoskeleton / cell motility / cell shape
Nat Commun, 14 Jan 2019 DOI: 10.1038/s41467-018-08217-3 Link to full text
Abstract: Ras and Rho small GTPases are critical for numerous cellular processes including cell division, migration, and intercellular communication. Despite extensive efforts to visualize the spatiotemporal activity of these proteins, achieving the sensitivity and dynamic range necessary for in vivo application has been challenging. Here, we present highly sensitive intensiometric small GTPase biosensors visualizing the activity of multiple small GTPases in single cells in vivo. Red-shifted sensors combined with blue light-controllable optogenetic modules achieved simultaneous monitoring and manipulation of protein activities in a highly spatiotemporal manner. Our biosensors revealed spatial dynamics of Cdc42 and Ras activities upon structural plasticity of single dendritic spines, as well as a broad range of subcellular Ras activities in the brains of freely behaving mice. Thus, these intensiometric small GTPase sensors enable the spatiotemporal dissection of complex protein signaling networks in live animals.
125.

Optogenetic dissection of Rac1 and Cdc42 gradient shaping.

blue CRY2/CIB1 CRY2/CRY2 HeLa Control of cytoskeleton / cell motility / cell shape
Nat Commun, 16 Nov 2018 DOI: 10.1038/s41467-018-07286-8 Link to full text
Abstract: During cell migration, Rho GTPases spontaneously form spatial gradients that define the front and back of cells. At the front, active Cdc42 forms a steep gradient whereas active Rac1 forms a more extended pattern peaking a few microns away. What are the mechanisms shaping these gradients, and what is the functional role of the shape of these gradients? Here we report, using a combination of optogenetics and micropatterning, that Cdc42 and Rac1 gradients are set by spatial patterns of activators and deactivators and not directly by transport mechanisms. Cdc42 simply follows the distribution of Guanine nucleotide Exchange Factors, whereas Rac1 shaping requires the activity of a GTPase-Activating Protein, β2-chimaerin, which is sharply localized at the tip of the cell through feedbacks from Cdc42 and Rac1. Functionally, the spatial extent of Rho GTPases gradients governs cell migration, a sharp Cdc42 gradient maximizes directionality while an extended Rac1 gradient controls the speed.
Submit a new publication to our database