Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Microbiol Spectr ; 12(1): e0356723, 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38078717

ABSTRACT

IMPORTANCE: The bacterial pathogen Pseudomonas aeruginosa is responsible for a variety of chronic human infections. Even in the absence of identifiable resistance mutations, this pathogen can tolerate lethal antibiotic doses through phenotypic strategies like biofilm formation and metabolic quiescence. In this study, we determined that P. aeruginosa maintains greater metabolic activity in the stationary phase compared to the model organism, Escherichia coli, which has traditionally been used to study fluoroquinolone antibiotic tolerance. We demonstrate that hallmarks of E. coli fluoroquinolone tolerance are not conserved in P. aeruginosa, including the timing of cell death and necessity of the SOS DNA damage response for survival. The heightened sensitivity of stationary-phase P. aeruginosa to fluoroquinolones is attributed to maintained transcriptional and reductase activity. Our data suggest that perturbations that suppress transcription and respiration in P. aeruginosa may actually protect the pathogen against this important class of antibiotics.


Subject(s)
Levofloxacin , Pseudomonas Infections , Humans , Levofloxacin/pharmacology , Levofloxacin/metabolism , Pseudomonas aeruginosa/metabolism , Escherichia coli/genetics , Anti-Bacterial Agents/therapeutic use , Pseudomonas Infections/drug therapy , Fluoroquinolones/pharmacology , Microbial Sensitivity Tests
2.
Nat Commun ; 8: 15831, 2017 06 21.
Article in English | MEDLINE | ID: mdl-28635959

ABSTRACT

During bone remodelling, osteoclasts induce chemotaxis of osteoblasts and yet maintain spatial segregation. We show that osteoclasts express the repulsive guidance factor Semaphorin 4D and induce contact inhibition of locomotion (CIL) in osteoblasts through its receptor Plexin-B1. To examine causality and elucidate how localized Plexin-B1 stimulation may spatiotemporally coordinate its downstream targets in guiding cell migration, we develop an optogenetic tool for Plexin-B1 designated optoPlexin. Precise optoPlexin activation at the leading edge of migrating osteoblasts readily induces local retraction and, unexpectedly, distal protrusions to steer cells away. These morphological changes are accompanied by reorganization of Myosin II, PIP3, adhesion and active Cdc42. We attribute the resultant repolarization to RhoA/ROCK-mediated redistribution of ß-Pix, which activates Cdc42 and promotes protrusion. Thus, our data demonstrate a causal role of Plexin-B1 for CIL in osteoblasts and reveals a previously unknown effect of Semaphorin signalling on spatial distribution of an activator of cell migration.


Subject(s)
Nerve Tissue Proteins/metabolism , Osteoblasts/metabolism , Osteoclasts/metabolism , Receptors, Cell Surface/metabolism , Animals , Cell Movement/radiation effects , Cell Polarity/radiation effects , Light , Male , Mice , Mice, Inbred C57BL , Myosin Type II/genetics , Myosin Type II/metabolism , Nerve Tissue Proteins/genetics , Optogenetics , Osteoblasts/cytology , Osteoblasts/radiation effects , Osteoclasts/cytology , Osteoclasts/radiation effects , Receptors, Cell Surface/genetics , Semaphorins/metabolism , Signal Transduction/radiation effects , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
3.
ACS Chem Neurosci ; 8(7): 1554-1569, 2017 07 19.
Article in English | MEDLINE | ID: mdl-28418645

ABSTRACT

Kalirin7 (Kal7), a postsynaptic Rho GDP/GTP exchange factor (RhoGEF), plays a crucial role in long-term potentiation and in the effects of cocaine on behavior and spine morphology. The KALRN gene has been linked to schizophrenia and other disorders of synaptic function. Mass spectrometry was used to quantify phosphorylation at 26 sites in Kal7 from individual adult rat nucleus accumbens and prefrontal cortex before and after exposure to acute or chronic cocaine. Region- and isoform-specific phosphorylation was observed along with region-specific effects of cocaine on Kal7 phosphorylation. Evaluation of the functional significance of multisite phosphorylation in a complex protein like Kalirin is difficult. With the identification of five tyrosine phosphorylation (pY) sites, a panel of 71 SH2 domains was screened, identifying subsets that interacted with multiple pY sites in Kal7. In addition to this type of reversible interaction, endoproteolytic cleavage by calpain plays an essential role in long-term potentiation. Calpain cleaved Kal7 at two sites, separating the N-terminal domain, which affects spine length, and the PDZ binding motif from the GEF domain. Mutations preventing phosphorylation did not affect calpain sensitivity or GEF activity; phosphomimetic mutations at specific sites altered protein stability, increased calpain sensitivity, and reduced GEF activity.


Subject(s)
Calpain/metabolism , Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Guanine Nucleotide Exchange Factors/metabolism , Nucleus Accumbens/metabolism , Prefrontal Cortex/metabolism , Animals , Binding Sites , Guanine Nucleotide Exchange Factors/genetics , HEK293 Cells , Humans , Male , Mass Spectrometry , Nucleus Accumbens/drug effects , PDZ Domains , Phosphorylation , Prefrontal Cortex/drug effects , Protein Isoforms , Rats, Sprague-Dawley , Tyrosine/metabolism , rac1 GTP-Binding Protein/metabolism , src Homology Domains
4.
Mol Biol Cell ; 28(10): 1347-1360, 2017 May 15.
Article in English | MEDLINE | ID: mdl-28356423

ABSTRACT

Breast carcinoma cells use specialized, actin-rich protrusions called invadopodia to degrade and invade through the extracellular matrix. Phosphorylation of the actin nucleation-promoting factor and actin-stabilizing protein cortactin downstream of the epidermal growth factor receptor-Src-Arg kinase cascade is known to be a critical trigger for invadopodium maturation and subsequent cell invasion in breast cancer cells. The functions of cortactin phosphorylation in this process, however, are not completely understood. We identify the Rho-family guanine nucleotide exchange factor Vav2 in a comprehensive screen for human SH2 domains that bind selectively to phosphorylated cortactin. We demonstrate that the Vav2 SH2 domain binds selectively to phosphotyrosine-containing peptides corresponding to cortactin tyrosines Y421 and Y466 but not to Y482. Mutation of the Vav2 SH2 domain disrupts its recruitment to invadopodia, and an SH2-domain mutant form of Vav2 cannot support efficient matrix degradation in invasive MDA-MB-231 breast cancer cells. We show that Vav2 function is required for promoting invadopodium maturation and consequent actin polymerization, matrix degradation, and invasive migratory behavior. Using biochemical assays and a novel Rac3 biosensor, we show that Vav2 promotes Rac3 activation at invadopodia. Rac3 knockdown reduces matrix degradation by invadopodia, whereas a constitutively active Rac3 can rescue the deficits in invadopodium function in Vav2-knockdown cells. Together these data indicate that phosphorylated cortactin recruits Vav2 to activate Rac3 and promote invadopodial maturation in invasive breast cancer cells.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cortactin/metabolism , Podosomes/metabolism , Proto-Oncogene Proteins c-vav/metabolism , Actins/metabolism , Cell Line, Tumor , Extracellular Matrix/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Humans , Neoplasm Invasiveness , Phosphorylation , Phosphotyrosine/metabolism , Podosomes/physiology , Protein-Tyrosine Kinases/metabolism , Pseudopodia/metabolism , rac GTP-Binding Proteins/metabolism
5.
Sci Rep ; 6: 36825, 2016 11 11.
Article in English | MEDLINE | ID: mdl-27833100

ABSTRACT

Rho GTPases are master regulators of the eukaryotic cytoskeleton. The activation of Rho GTPases is governed by Rho guanine nucleotide exchange factors (GEFs). Three RhoGEF isoforms are produced by the gene ARHGEF25; p63RhoGEF580, GEFT and a recently discovered longer isoform of 619 amino acids (p63RhoGEF619). The subcellular distribution of p63RhoGEF580 and p63RhoGEF619 is strikingly different in unstimulated cells, p63RhoGEF580 is located at the plasma membrane and p63RhoGEF619 is confined to the cytoplasm. Interestingly, we find that both P63RhoGEF580 and p63RhoGEF619 activate RhoGTPases to a similar extent after stimulation of Gαq coupled GPCRs. Furthermore, we show that p63RhoGEF619 relocates to the plasma membrane upon activation of Gαq coupled GPCRs, resembling the well-known activation mechanism of RhoGEFs activated by Gα12/13. Synthetic recruitment of p63RhoGEF619 to the plasma membrane increases RhoGEF activity towards RhoA, but full activation requires allosteric activation via Gαq. Together, these findings reveal a dual role for Gαq in RhoGEF activation, as it both recruits and allosterically activates cytosolic ARHGEF25 isoforms.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Rho Guanine Nucleotide Exchange Factors/metabolism , Allosteric Regulation , Amino Acid Sequence , Cell Membrane/metabolism , HeLa Cells , Humans , Kinetics , Protein Isoforms/metabolism , Protein Transport , Signal Transduction
6.
Sci Rep ; 6: 25502, 2016 05 05.
Article in English | MEDLINE | ID: mdl-27147504

ABSTRACT

Endothelial cells line the vasculature and are important for the regulation of blood pressure, vascular permeability, clotting and transendothelial migration of leukocytes and tumor cells. A group of proteins that that control the endothelial barrier function are the RhoGTPases. This study focuses on three homologous (>88%) RhoGTPases: RhoA, RhoB, RhoC of which RhoB and RhoC have been poorly characterized. Using a RhoGTPase mRNA expression analysis we identified RhoC as the highest expressed in primary human endothelial cells. Based on an existing RhoA FRET sensor we developed new RhoB/C FRET sensors to characterize their spatiotemporal activation properties. We found all these RhoGTPase sensors to respond to physiologically relevant agonists (e.g. Thrombin), reaching transient, localized FRET ratio changes up to 200%. These RhoA/B/C FRET sensors show localized GEF and GAP activity and reveal spatial activation differences between RhoA/C and RhoB. Finally, we used these sensors to monitor GEF-specific differential activation of RhoA/B/C. In summary, this study adds high-contrast RhoB/C FRET sensors to the currently available FRET sensor toolkit and uncover new insights in endothelial and RhoGTPase cell biology. This allows us to study activation and signaling by these closely related RhoGTPases with high spatiotemporal resolution in primary human cells.


Subject(s)
Antigens, CD/genetics , Biosensing Techniques/methods , Cadherins/genetics , Human Umbilical Vein Endothelial Cells/enzymology , rhoA GTP-Binding Protein/genetics , rhoB GTP-Binding Protein/genetics , rhoC GTP-Binding Protein/genetics , Antigens, CD/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cadherins/metabolism , Enzyme Activation , Fluorescence Resonance Energy Transfer , Gap Junctions , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/ultrastructure , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Models, Molecular , Nocodazole/pharmacology , Primary Cell Culture , Protein Structure, Secondary , Signal Transduction , Thrombin/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , rhoA GTP-Binding Protein/metabolism , rhoB GTP-Binding Protein/metabolism , rhoC GTP-Binding Protein/metabolism
7.
J Biol Chem ; 291(9): 4323-33, 2016 Feb 26.
Article in English | MEDLINE | ID: mdl-26740622

ABSTRACT

Invadosomes are actin-rich membrane protrusions that degrade the extracellular matrix to drive tumor cell invasion. Key players in invadosome formation are c-Src and Rho family GTPases. Invadosomes can reassemble into circular rosette-like superstructures, but the underlying signaling mechanisms remain obscure. Here we show that Src-induced invadosomes in human melanoma cells (A375M and MDA-MB-435) undergo rapid remodeling into dynamic extracellular matrix-degrading rosettes by distinct G protein-coupled receptor agonists, notably lysophosphatidic acid (LPA; acting through the LPA1 receptor) and endothelin. Agonist-induced rosette formation is blocked by pertussis toxin, dependent on PI3K activity and accompanied by localized production of phosphatidylinositol 3,4,5-trisphosphate, whereas MAPK and Ca(2+) signaling are dispensable. Using FRET-based biosensors, we show that LPA and endothelin transiently activate Cdc42 through Gi, concurrent with a biphasic decrease in Rac activity and differential effects on RhoA. Cdc42 activity is essential for rosette formation, whereas G12/13-mediated RhoA-ROCK signaling suppresses the remodeling process. Our results reveal a Gi-mediated Cdc42 signaling axis by which G protein-coupled receptors trigger invadosome remodeling, the degree of which is dictated by the Cdc42-RhoA activity balance.


Subject(s)
Endothelins/metabolism , Lysophospholipids/metabolism , Melanoma/metabolism , Podosomes/metabolism , Receptors, Lysophosphatidic Acid/agonists , cdc42 GTP-Binding Protein/agonists , rac1 GTP-Binding Protein/metabolism , Biomarkers/metabolism , Cell Line, Tumor , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fluorescence Resonance Energy Transfer , Humans , Hydrolysis , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Melanoma/enzymology , Melanoma/pathology , Microscopy, Confocal , Microscopy, Fluorescence , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Podosomes/enzymology , Podosomes/pathology , RNA Interference , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Receptors, Lysophosphatidic Acid/genetics , Receptors, Lysophosphatidic Acid/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Time-Lapse Imaging , cdc42 GTP-Binding Protein/antagonists & inhibitors , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/agonists , rac1 GTP-Binding Protein/antagonists & inhibitors , rac1 GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
8.
Mol Biol Cell ; 27(2): 247-59, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26609071

ABSTRACT

The Nck adaptor protein recruits cytosolic effectors such as N-WASP that induce localized actin polymerization. Experimental aggregation of Nck SH3 domains at the membrane induces actin comet tails--dynamic, elongated filamentous actin structures similar to those that drive the movement of microbial pathogens such as vaccinia virus. Here we show that experimental manipulation of the balance between unbranched/branched nucleation altered the morphology and dynamics of Nck-induced actin comets. Inhibition of linear, formin-based nucleation with the small-molecule inhibitor SMIFH2 or overexpression of the formin FH1 domain resulted in formation of predominantly circular-shaped actin structures with low mobility (actin blobs). These results indicate that formin-based linear actin polymerization is critical for the formation and maintenance of Nck-dependent actin comet tails. Consistent with this, aggregation of an exclusively branched nucleation-promoting factor (the VCA domain of N-WASP), with density and turnover similar to those of N-WASP in Nck comets, did not reconstitute dynamic, elongated actin comets. Furthermore, enhancement of branched Arp2/3-mediated nucleation by N-WASP overexpression caused loss of the typical actin comet tail shape induced by Nck aggregation. Thus the ratio of linear to dendritic nucleation activity may serve to distinguish the properties of actin structures induced by various viral and bacterial pathogens.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Oncogene Proteins/metabolism , Animals , Dendrites/metabolism , Fetal Proteins/metabolism , Formins , HeLa Cells , Humans , Mice , Microfilament Proteins/metabolism , NIH 3T3 Cells , Nuclear Proteins/metabolism , Structure-Activity Relationship , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , src Homology Domains
9.
Sci Rep ; 5: 14693, 2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26435194

ABSTRACT

The small GTPase RhoA is involved in cell morphology and migration. RhoA activity is tightly regulated in time and space and depends on guanine exchange factors (GEFs). However, the kinetics and subcellular localization of GEF activity towards RhoA are poorly defined. To study the mechanism underlying the spatiotemporal control of RhoA activity by GEFs, we performed single cell imaging with an improved FRET sensor reporting on the nucleotide loading state of RhoA. By employing the FRET sensor we show that a plasma membrane located RhoGEF, p63RhoGEF, can rapidly activate RhoA through endogenous GPCRs and that localized RhoA activity at the cell periphery correlates with actin polymerization. Moreover, synthetic recruitment of the catalytic domain derived from p63RhoGEF to the plasma membrane, but not to the Golgi apparatus, is sufficient to activate RhoA. The synthetic system enables local activation of endogenous RhoA and effectively induces actin polymerization and changes in cellular morphology. Together, our data demonstrate that GEF activity at the plasma membrane is sufficient for actin polymerization via local RhoA signaling.


Subject(s)
Actins/metabolism , Cell Membrane/enzymology , rhoA GTP-Binding Protein/physiology , Cell Nucleus , Enzyme Activation , HeLa Cells , Humans , Protein Multimerization , Protein Structure, Tertiary , Protein Transport , Rho Guanine Nucleotide Exchange Factors , Transcription Factors/metabolism
10.
Nature ; 525(7569): 333-8, 2015 Sep 17.
Article in English | MEDLINE | ID: mdl-26352471

ABSTRACT

Dendritic spines are the major loci of synaptic plasticity and are considered as possible structural correlates of memory. Nonetheless, systematic manipulation of specific subsets of spines in the cortex has been unattainable, and thus, the link between spines and memory has been correlational. We developed a novel synaptic optoprobe, AS-PaRac1 (activated synapse targeting photoactivatable Rac1), that can label recently potentiated spines specifically, and induce the selective shrinkage of AS-PaRac1-containing spines. In vivo imaging of AS-PaRac1 revealed that a motor learning task induced substantial synaptic remodelling in a small subset of neurons. The acquired motor learning was disrupted by the optical shrinkage of the potentiated spines, whereas it was not affected by the identical manipulation of spines evoked by a distinct motor task in the same cortical region. Taken together, our results demonstrate that a newly acquired motor skill depends on the formation of a task-specific dense synaptic ensemble.


Subject(s)
Memory/physiology , Memory/radiation effects , Motor Cortex/physiology , Motor Cortex/radiation effects , Neuronal Plasticity/physiology , Neuronal Plasticity/radiation effects , Synapses/physiology , Synapses/radiation effects , Animals , Dendritic Spines/physiology , Dendritic Spines/radiation effects , Hippocampus/cytology , Hippocampus/physiology , Hippocampus/radiation effects , In Vitro Techniques , Light , Long-Term Potentiation/physiology , Long-Term Potentiation/radiation effects , Male , Mice , Molecular Probes , Motor Cortex/cytology , Motor Skills/physiology , Motor Skills/radiation effects , Rotarod Performance Test , Spatio-Temporal Analysis
11.
Nat Commun ; 6: 6619, 2015 Apr 08.
Article in English | MEDLINE | ID: mdl-25851023

ABSTRACT

Directed cell migration in native environments is influenced by multiple migratory cues. These cues may include simultaneously occurring attractive soluble growth factor gradients and repulsive effects arising from cell-cell contact, termed contact inhibition of locomotion (CIL). How single cells reconcile potentially conflicting cues remains poorly understood. Here we show that a dynamic crosstalk between epidermal growth factor (EGF)-mediated chemotaxis and CIL guides metastatic breast cancer cell motility, whereby cells become progressively insensitive to CIL in a chemotactic input-dependent manner. This balance is determined via integration of protrusion-enhancing signalling from EGF gradients and protrusion-suppressing signalling induced by CIL, mediated in part through EphB. Our results further suggest that EphB and EGF signalling inputs control protrusion formation by converging onto regulation of phosphatidylinositol 3-kinase (PI3K). We propose that this intricate interplay may enhance the spread of loose cell ensembles in pathophysiological conditions such as cancer, and possibly other physiological settings.


Subject(s)
Cell Movement/genetics , Chemotaxis/genetics , Contact Inhibition/physiology , Mammary Neoplasms, Animal/genetics , Receptor, EphB3/genetics , Animals , Blotting, Western , Cell Line, Tumor , Epidermal Growth Factor/metabolism , Female , Gene Knockdown Techniques , Image Processing, Computer-Assisted , Immunohistochemistry , Mammary Neoplasms, Animal/metabolism , Rats , Receptors, Eph Family/genetics , Reverse Transcriptase Polymerase Chain Reaction
12.
Proc Natl Acad Sci U S A ; 112(3): E267-76, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25561548

ABSTRACT

Polarized Rac1 signaling is a hallmark of many cellular functions, including cell adhesion, motility, and cell division. The two steps of Rac1 activation are its translocation to the plasma membrane and the exchange of nucleotide from GDP to GTP. It is, however, unclear whether these two processes are regulated independent of each other and what their respective roles are in polarization of Rac1 signaling. We designed a single-particle tracking (SPT) method to quantitatively analyze the kinetics of Rac1 membrane translocation in living cells. We found that the rate of Rac1 translocation was significantly elevated in protrusions during cell spreading on collagen. Furthermore, combining FRET sensor imaging with SPT measurements in the same cell, the recruitment of Rac1 was found to be polarized to an extent similar to that of the nucleotide exchange process. Statistical analysis of single-molecule trajectories and optogenetic manipulation of membrane lipids revealed that Rac1 membrane translocation precedes nucleotide exchange, and is governed primarily by interactions with phospholipids, particularly PI(3,4,5)P3, instead of protein factors. Overall, the study highlights the significance of membrane translocation in spatial Rac1 signaling, which is in addition to the traditional view focusing primarily on GEF distribution and exchange reaction.


Subject(s)
Signal Transduction , rac1 GTP-Binding Protein/metabolism , Humans , Kinetics , MCF-7 Cells , Microscopy, Fluorescence , Protein Transport
13.
Methods Mol Biol ; 1251: 277-89, 2015.
Article in English | MEDLINE | ID: mdl-25391805

ABSTRACT

Recent developments in optogenetics have extended optical control of signaling to intracellular proteins, including Rac, a small G protein in the Rho family. A blue light-sensing LOV (light, oxygen, or voltage) domain derived from Avena sativa (oat) phototropin was fused to the N-terminus of a constitutively active mutant of Rac, via an α-helix (Jα) that is conserved among plant phototropins. The fused LOV domain occluded binding of downstream effectors to Rac in the dark. Exposure to blue light caused a conformational change of the LOV domain and unwinding of the Jα helix, relieving steric inhibition. The LOV domain incorporates a flavin as the photon-absorbing cofactor and can be activated by light in a reversible and repeatable fashion. In cultured cells, global illumination with blue light rapidly activated Rac and led to cell spreading and membrane ruffling. Localized and pulsed illumination generated a gradient of Rac activity and induced directional migration. In this chapter, we will describe the techniques in detail and present some examples of applications of using photoactivatable Rac (PA-Rac) in living cells.


Subject(s)
Cytological Techniques/methods , Microscopy, Fluorescence/methods , Optogenetics/methods , Signal Transduction/physiology , rac GTP-Binding Proteins/ultrastructure , Animals , Avena/chemistry , HeLa Cells , Humans , Light , Lighting , Mice , Microscopy, Fluorescence/instrumentation , Phototropins/analysis , Phototropins/genetics , Protein Conformation , Protein Structure, Tertiary
14.
Biochemistry ; 52(40): 7082-90, 2013 Oct 08.
Article in English | MEDLINE | ID: mdl-24011356

ABSTRACT

The short cytoplasmic tails of the α- and ß-chains of integrin adhesion receptors regulate integrin activation and cell signaling. Significantly less is known about proteins that bind to α-integrin cytoplasmic tails (CTs) as opposed to ß-CTs to regulate integrins. Calcium and integrin binding protein 1 (CIB1) was previously identified as an αIIb binding partner that inhibits agonist-induced activation of the platelet-specific integrin, αIIbß3. A sequence alignment of all α-integrin CTs revealed that key residues in the CIB1 binding site of αIIb are well-conserved, and was used to delineate a consensus binding site (I/L-x-x-x-L/M-W/Y-K-x-G-F-F). Because the CIB1 binding site of αIIb is conserved in all α-integrins and CIB1 expression is ubiquitous, we asked if CIB1 could interact with other α-integrin CTs. We predicted that multiple α-integrin CTs were capable of binding to the same hydrophobic binding pocket on CIB1 with docking models generated by all-atom replica exchange discrete molecular dynamics. After demonstrating novel in vivo interactions between CIB1 and other whole integrin complexes with co-immunoprecipitations, we validated the modeled predictions with solid-phase competitive binding assays, which showed that other α-integrin CTs compete with the αIIb CT for binding to CIB1 in vitro. Isothermal titration calorimetry measurements indicated that this binding is driven by hydrophobic interactions and depends on residues in the CIB1 consensus binding site. These new mechanistic details of CIB1-integrin binding imply that CIB1 could bind to all integrin complexes and act as a broad regulator of integrin function.


Subject(s)
Calcium-Binding Proteins/metabolism , Integrin alpha Chains/metabolism , Amino Acid Sequence , Binding Sites , Calcium-Binding Proteins/genetics , Hydrophobic and Hydrophilic Interactions , Molecular Dynamics Simulation , Platelet Membrane Glycoprotein IIb/metabolism , Protein Binding , Sequence Alignment
15.
Pflugers Arch ; 465(3): 397-408, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23417571

ABSTRACT

Optogenetics arises from the innovative application of microbial opsins in mammalian neurons and has since been a powerful technology that fuels the advance of our knowledge in neuroscience. In recent years, there has been growing interest in designing optogenetic tools extendable to broader cell types and biochemical signals. To date, a variety of photoactivatable proteins (refers to induction of protein activity in contrast to fluorescence) have been developed based on the understanding of plant and microbial photoreceptors including phototropins, blue light sensors using flavin adenine dinucleotide proteins, cryptochromes, and phytochromes. Such tools offered researchers reversible, quantitative, and precise spatiotemporal control of enzymatic activity, protein-protein interaction, protein translocation, as well as gene transcription in cells and in whole animals. In this review, we will briefly introduce these photosensory proteins, describe recent developments in optogenetics, and compare and contrast different methods based on their advantages and limitations.


Subject(s)
Optogenetics , Signal Transduction , Allosteric Regulation , Animals , Bacteria , Cryptochromes/genetics , Cryptochromes/metabolism , Humans , Photoreceptors, Microbial/genetics , Photoreceptors, Microbial/metabolism , Phototropins/genetics , Phototropins/metabolism , Plants , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
16.
Nat Cell Biol ; 14(8): 874-81, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22750946

ABSTRACT

Many of the more than 20 mammalian proteins with N-BAR domains control cell architecture and endocytosis by associating with curved sections of the plasma membrane. It is not well understood whether N-BAR proteins are recruited directly by processes that mechanically curve the plasma membrane or indirectly by plasma-membrane-associated adaptor proteins that recruit proteins with N-BAR domains that then induce membrane curvature. Here, we show that externally induced inward deformation of the plasma membrane by cone-shaped nanostructures (nanocones) and internally induced inward deformation by contracting actin cables both trigger recruitment of isolated N-BAR domains to the curved plasma membrane. Markedly, live-cell imaging in adherent cells showed selective recruitment of full-length N-BAR proteins and isolated N-BAR domains to plasma membrane sub-regions above nanocone stripes. Electron microscopy confirmed that N-BAR domains are recruited to local membrane sites curved by nanocones. We further showed that N-BAR domains are periodically recruited to curved plasma membrane sites during local lamellipodia retraction in the front of migrating cells. Recruitment required myosin-II-generated force applied to plasma-membrane-connected actin cables. Together, our results show that N-BAR domains can be directly recruited to the plasma membrane by external push or internal pull forces that locally curve the plasma membrane.


Subject(s)
Actins/physiology , Cell Membrane/metabolism , Cytoskeletal Proteins/metabolism , Mechanical Phenomena , Nanostructures , Protein Interaction Domains and Motifs/physiology , 3T3 Cells , Animals , Cell Membrane/ultrastructure , Cytoskeletal Proteins/genetics , HeLa Cells , Humans , Mice , Microscopy, Electron, Scanning , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism
17.
J Mol Biol ; 413(2): 513-22, 2011 Oct 21.
Article in English | MEDLINE | ID: mdl-21888918

ABSTRACT

The inhibitory switch (IS) domain of p21-activated kinase 1 (PAK1) stabilizes full-length PAK1 in an inactive conformation by binding to the PAK1 kinase domain. Competitive binding of small guanosine triphosphatases to the IS domain disrupts the autoinhibitory interactions and exposes the IS domain binding site on the surface of the kinase domain. To build an affinity reagent that selectively binds the activated state of PAK1, we used molecular modeling to reengineer the isolated IS domain so that it was soluble and stable, did not bind to guanosine triphosphatases and bound more tightly to the PAK1 kinase domain. Three design strategies were tested: in the first and second cases, extension and redesign of the N-terminus were used to expand the hydrophobic core of the domain, and in the third case, the termini were redesigned to be adjacent in space so that the domain could be stabilized by insertion into a loop in a host cyan fluorescent protein (CFP). The best-performing design, called CFP-PAcKer, was based on the third strategy and bound the kinase domain of PAK1 with an affinity of 400 nM. CFP-PAcKer binds more tightly to a full-length variant of PAK1 that is stabilized in the "open" state (K(d)=3.3 µM) than to full-length PAK1 in the "closed" state (undetectable affinity), and binding can be monitored with fluorescence by placing an environmentally sensitive fluorescence dye on CFP-PAcKer adjacent to the binding site.


Subject(s)
Biosensing Techniques , Computer-Aided Design , Green Fluorescent Proteins/metabolism , Molecular Dynamics Simulation , p21-Activated Kinases/antagonists & inhibitors , p21-Activated Kinases/metabolism , Amino Acid Sequence , Binding Sites , Circular Dichroism , Enzyme Activation , Fluorescence Polarization , Green Fluorescent Proteins/chemistry , Humans , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Conformation , Sequence Homology, Amino Acid , Software
18.
Methods Enzymol ; 497: 393-407, 2011.
Article in English | MEDLINE | ID: mdl-21601095

ABSTRACT

Signaling networks in living systems are coordinated through subcellular compartmentalization and precise timing of activation. These spatiotemporal aspects ensure the fidelity of signaling while contributing to the diversity and specificity of downstream events. This is studied through development of molecular tools that generate localized and precisely timed protein activity in living systems. To study the molecular events responsible for cytoskeletal changes in real time, we generated versions of Rho family GTPases whose interactions with downstream effectors is controlled by light. GTPases were grafted to the phototropin LOV (light, oxygen, or voltage) domain (Huala, E., Oeller, P. W., Liscum, E., Han, I., Larsen, E., and Briggs, W. R. (1997). Arabidopsis NPH1: A protein kinase with a putative redox-sensing domain. Science278, 2120-2123.) via an alpha helix on the LOV C-terminus (Wu, Y. I., Frey, D., Lungu, O. I., Jaehrig, A., Schlichting, I., Kuhlman, B., and Hahn, K. M. (2009). A genetically encoded photoactivatable Rac controls the motility of living cells. Nature461, 104-108.). The LOV domain sterically blocked the GTPase active site until it was irradiated. Exposure to 400-500nm light caused unwinding of the helix linking the LOV domain to the GTPase, relieving steric inhibition. The change was reversible and repeatable, and the protein could be returned to its inactive state simply by turning off the light. The LOV domain incorporates a flavin as the active chromophore. This naturally occurring molecule is incorporated simply upon expression of the LOV fusion in cells or animals, permitting ready control of GTPase function in different systems. In cultured single cells, light-activated Rac leads to membrane ruffling, protrusion, and migration. In collectively migrating border cells in the Drosophila ovary, focal activation of photoactivatable Rac (PA-Rac) in a single cell is sufficient to redirect the entire group. PA-Rac in a single cell also rescues the phenotype caused by loss of endogenous guidance receptor signaling in the whole group. These findings demonstrate that cells within the border cell cluster communicate and are guided collectively. Here, we describe optimization and application of PA-Rac using detailed examples that we hope will help others apply the approach to different proteins and in a variety of different cells, tissues, and organisms.


Subject(s)
Light , Monomeric GTP-Binding Proteins/chemistry , Monomeric GTP-Binding Proteins/metabolism , Phototropins/chemistry , Phototropins/metabolism , Protein Structure, Tertiary , Animals , Cell Movement , Cells, Cultured , Drosophila melanogaster/anatomy & histology , Female , Microscopy/instrumentation , Microscopy/methods , Monomeric GTP-Binding Proteins/genetics , Ovary/cytology , Phototropins/genetics , Signal Transduction/physiology , rac GTP-Binding Proteins/chemistry , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism
19.
Mol Biol Cell ; 22(8): 1274-89, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21346197

ABSTRACT

Vimentin intermediate filaments (VIF) extend throughout the rear and perinuclear regions of migrating fibroblasts, but only nonfilamentous vimentin particles are present in lamellipodial regions. In contrast, VIF networks extend to the entire cell periphery in serum-starved or nonmotile fibroblasts. Upon serum addition or activation of Rac1, VIF are rapidly phosphorylated at Ser-38, a p21-activated kinase phosphorylation site. This phosphorylation of vimentin is coincident with VIF disassembly at and retraction from the cell surface where lamellipodia form. Furthermore, local induction of photoactivatable Rac1 or the microinjection of a vimentin mimetic peptide (2B2) disassemble VIF at sites where lamellipodia subsequently form. When vimentin organization is disrupted by a dominant-negative mutant or by silencing, there is a loss of polarity, as evidenced by the formation of lamellipodia encircling the entire cell, as well as reduced cell motility. These findings demonstrate an antagonistic relationship between VIF and the formation of lamellipodia.


Subject(s)
Cell Movement , Neuropeptides/metabolism , Peptide Fragments/metabolism , Pseudopodia/metabolism , Vimentin/metabolism , rac GTP-Binding Proteins/metabolism , Animals , Cell Polarity , Escherichia coli , Gene Expression , Gene Silencing , Humans , Intermediate Filaments/metabolism , Mice , Mice, Knockout , Microinjections , NIH 3T3 Cells , Neuropeptides/genetics , Peptide Fragments/genetics , Phosphorylation , Pseudopodia/genetics , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serine/metabolism , Serum/metabolism , Vimentin/genetics , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism , rac GTP-Binding Proteins/genetics , rac1 GTP-Binding Protein
20.
Nat Cell Biol ; 12(6): 591-7, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20473296

ABSTRACT

The small GTPase Rac induces actin polymerization, membrane ruffling and focal contact formation in cultured single cells but can either repress or stimulate motility in epithelial cells depending on the conditions. The role of Rac in collective epithelial cell movements in vivo, which are important for both morphogenesis and metastasis, is therefore difficult to predict. Recently, photoactivatable analogues of Rac (PA-Rac) have been developed, allowing rapid and reversible activation or inactivation of Rac using light. In cultured single cells, light-activated Rac leads to focal membrane ruffling, protrusion and migration. Here we show that focal activation of Rac is also sufficient to polarize an entire group of cells in vivo, specifically the border cells of the Drosophila ovary. Moreover, activation or inactivation of Rac in one cell of the cluster caused a dramatic response in the other cells, suggesting that the cells sense direction as a group according to relative levels of Rac activity. Communication between cells of the cluster required Jun amino-terminal kinase (JNK) but not guidance receptor signalling. These studies further show that photoactivatable proteins are effective tools in vivo.


Subject(s)
Cell Movement/genetics , Proteins/metabolism , Animals , Cell Surface Extensions/genetics , Cell Surface Extensions/metabolism , Cells, Cultured , Cricetinae , Drosophila/genetics , Drosophila/metabolism , Epithelial Cells/metabolism , Female , Humans , Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...