Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
bioRxiv ; 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38464100

ABSTRACT

Doublecortin (DCX) is a microtubule-associated protein critical for brain development. Although most highly expressed in the developing central nervous system, the molecular function of DCX in neuron morphogenesis remains unknown and controversial. We demonstrate that DCX function is intimately linked to its microtubule-binding activity. By using human induced pluripotent stem cell (hiPSC)- derived cortical i 3 Neurons genome engineered to express mEmerald-tagged DCX from the endogenous locus, we find that DCX-MT interactions become highly polarized very early during neuron morphogenesis. DCX becomes enriched only on straight microtubules in advancing growth cones with approximately 120 DCX molecules bound per micrometer of growth cone microtubule. At a similar saturation, microtubule-bound DCX molecules begin to impede lysosome transport, and thus can potentially control growth cone organelle entry. In addition, by comparing control, DCX-mEmerald and knockout DCX -/Y i 3 Neurons, we find that DCX stabilizes microtubules in the growth cone peripheral domain by reducing the microtubule catastrophe frequency and the depolymerization rate. DCX -/Y i 3 Neuron morphogenesis was inhibited in soft microenvironments that mimic the viscoelasticity of brain tissue and DCX -/Y neurites failed to grow toward brain-derived neurotrophic factor (BDNF) gradients. Together with high resolution traction force microscopy data, we propose a model in which DCX-decorated, rigid growth cone microtubules provide intracellular mechanical resistance to actomyosin generated contractile forces in soft physiological environments in which weak and transient adhesion-mediated forces in the growth cone periphery may be insufficient for productive growth cone advance. These data provide a new mechanistic understanding of how DCX mutations cause lissencephaly-spectrum brain malformations by impacting growth cone dynamics during neuron morphogenesis in physiological environments.

2.
bioRxiv ; 2024 Feb 07.
Article in English | MEDLINE | ID: mdl-38370785

ABSTRACT

Stress granules (SGs) are macromolecular assemblies that form under cellular stress. Formation of these condensates is driven by the condensation of RNA and RNA-binding proteins such as G3BPs. G3BPs condense into SGs following stress-induced translational arrest. Three G3BP paralogs (G3BP1, G3BP2A, and G3BP2B) have been identified in vertebrates. However, the contribution of different G3BP paralogs to stress granule formation and stress-induced gene expression changes is incompletely understood. Here, we identified key residues for G3BP condensation such as V11. This conserved amino acid is required for formation of the G3BP-Caprin-1 complex, hence promoting SG assembly. Total RNA sequencing and ribosome profiling revealed that disruption of G3BP condensation corresponds to changes in mRNA levels and ribosome engagement during the integrated stress response (ISR). Moreover, we found that G3BP2B preferentially condenses and promotes changes in mRNA expression under endoplasmic reticulum (ER) stress. Together, this work suggests that stress granule assembly promotes changes in gene expression under cellular stress, which is differentially regulated by G3BP paralogs.

3.
Elife ; 122023 01 30.
Article in English | MEDLINE | ID: mdl-36715499

ABSTRACT

A challenge in analyzing dynamic intracellular cell biological processes is the dearth of methodologies that are sufficiently fast and specific to perturb intracellular protein activities. We previously developed a light-sensitive variant of the microtubule plus end-tracking protein EB1 by inserting a blue light-controlled protein dimerization module between functional domains. Here, we describe an advanced method to replace endogenous EB1 with this light-sensitive variant in a single genome editing step, thereby enabling this approach in human induced pluripotent stem cells (hiPSCs) and hiPSC-derived neurons. We demonstrate that acute and local optogenetic EB1 inactivation in developing cortical neurons induces microtubule depolymerization in the growth cone periphery and subsequent neurite retraction. In addition, advancing growth cones are repelled from areas of blue light exposure. These phenotypes were independent of the neuronal EB1 homolog EB3, revealing a direct dynamic role of EB1-mediated microtubule plus end interactions in neuron morphogenesis and neurite guidance.


Subject(s)
Induced Pluripotent Stem Cells , Microtubule-Associated Proteins , Humans , Genomics , Growth Cones/metabolism , Induced Pluripotent Stem Cells/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Protein Binding
4.
Methods Mol Biol ; 2430: 467-481, 2022.
Article in English | MEDLINE | ID: mdl-35476350

ABSTRACT

Micropatterning of extracellular matrix proteins enables defining cell position and shape in experiments investigating intracellular dynamics and organization. While such standardization is advantageous in automated and quantitative analysis of many cells, the original methods generating such patterns are cumbersome and inflexible. However, recent development of contact-less methods that allow photochemical generation of protein patterns robustly and rapidly is boosting the broader availability of micropatterning approaches. Here, we describe an optimized protocol to achieve large micropatterned areas with high fidelity using a commercially available microscope-mounted UV projection system.


Subject(s)
Extracellular Matrix Proteins , Extracellular Matrix , Cell Shape , Extracellular Matrix Proteins/metabolism , Microtubules/metabolism
5.
J Cell Biol ; 221(4)2022 04 04.
Article in English | MEDLINE | ID: mdl-35266954

ABSTRACT

Missense mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common cause of familial Parkinson's disease (PD); however, pathways regulating LRRK2 subcellular localization, function, and turnover are not fully defined. We performed quantitative mass spectrometry-based interactome studies to identify 48 novel LRRK2 interactors, including the microtubule-associated E3 ubiquitin ligase TRIM1 (tripartite motif family 1). TRIM1 recruits LRRK2 to the microtubule cytoskeleton for ubiquitination and proteasomal degradation by binding LRRK2911-919, a nine amino acid segment within a flexible interdomain region (LRRK2853-981), which we designate the "regulatory loop" (RL). Phosphorylation of LRRK2 Ser910/Ser935 within LRRK2 RL influences LRRK2's association with cytoplasmic 14-3-3 versus microtubule-bound TRIM1. Association with TRIM1 modulates LRRK2's interaction with Rab29 and prevents upregulation of LRRK2 kinase activity by Rab29 in an E3-ligase-dependent manner. Finally, TRIM1 rescues neurite outgrowth deficits caused by PD-driving mutant LRRK2 G2019S. Our data suggest that TRIM1 is a critical regulator of LRRK2, controlling its degradation, localization, binding partners, kinase activity, and cytotoxicity.


Subject(s)
Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Parkinson Disease , Protein Serine-Threonine Kinases , Tripartite Motif Proteins , Cytoskeleton , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Microtubule-Associated Proteins , Microtubules , Mutation , Parkinson Disease/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Transcription Factors , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , rab GTP-Binding Proteins/metabolism
6.
Curr Biol ; 32(5): 1197-1205.e4, 2022 03 14.
Article in English | MEDLINE | ID: mdl-35090591

ABSTRACT

Chromosome segregation is accomplished by the mitotic spindle, a bipolar micromachine built primarily from microtubules. Different microtubule populations contribute to spindle function: kinetochore microtubules attach and transmit forces to chromosomes, antiparallel interpolar microtubules support spindle structure, and astral microtubules connect spindle poles to the cell cortex.1,2 In mammalian cells, end-binding (EB) proteins associate with all growing microtubule plus ends throughout the cell cycle and serve as adaptors for diverse +TIPs that control microtubule dynamics and interactions with other intracellular structures.3 Because binding of many +TIPs to EB1 and thus microtubule-end association is switched off by mitotic phosphorylation,4-6 the mitotic function of EBs remains poorly understood. To analyze how EB1 and associated +TIPs on different spindle microtubule populations contribute to mitotic spindle dynamics, we use a light-sensitive EB1 variant, π-EB1, that allows local, acute, and reversible inactivation of +TIP association with growing microtubule ends in live cells.7 We find that acute π-EB1 photoinactivation results in rapid and reversible metaphase spindle shortening and transient relaxation of tension across the central spindle. However, in contrast to interphase, π-EB1 photoinactivation does not inhibit microtubule growth in metaphase but instead increases astral microtubule length and number. Yet in the absence of EB1 activity, astral microtubules fail to engage the cortical dynein/dynactin machinery, and spindle poles move away from regions of π-EB1 photoinactivation. In conclusion, our optogenetic approach reveals mitotic EB1 functions that remain hidden in genetic experiments, likely due to compensatory molecular systems regulating vertebrate spindle dynamics.


Subject(s)
Microtubule-Associated Proteins , Optogenetics , Animals , Mammals , Metaphase , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Spindle Apparatus/metabolism
7.
Mol Biol Cell ; 32(2): 131-142, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33237838

ABSTRACT

Many lysosome functions are determined by a lumenal pH of ∼5.0, including the activity of resident acid-activated hydrolases. Lysosome pH (pHlys) is often increased in neurodegenerative disorders and predicted to be decreased in cancers, making it a potential target for therapeutics to limit the progression of these diseases. Accurately measuring pHlys, however, is limited by currently used dyes that accumulate in multiple intracellular compartments and cannot be propagated in clonal cells for longitudinal studies or used for in vivo determinations. To resolve this limitation, we developed a genetically encoded ratiometric pHlys biosensor, pHLARE (pH Lysosomal Activity REporter), which localizes predominantly in lysosomes, has a dynamic range of pH 4.0 to 6.5, and can be stably expressed in cells. Using pHLARE we show decreased pHlys with inhibiting activity of the mammalian target of rapamycin complex 1 (mTORC1). Also, cancer cells from different tissue origins have a lower pHlys than untransformed cells, and stably expressing oncogenic RasV12 in untransformed cells is sufficient to decrease pHlys. pHLARE is a new tool to accurately measure pHlys for improved understanding of lysosome dynamics, which is increasingly considered a therapeutic target.


Subject(s)
Biosensing Techniques , Lysosomes/metabolism , Neoplasms/metabolism , Animals , Calibration , Cell Line, Tumor , Cell Transformation, Neoplastic/pathology , Humans , Hydrogen-Ion Concentration , Rats , Reproducibility of Results
8.
Nature ; 588(7838): 459-465, 2020 12.
Article in English | MEDLINE | ID: mdl-32866962

ABSTRACT

Aberrant aggregation of the RNA-binding protein TDP-43 in neurons is a hallmark of frontotemporal lobar degeneration caused by haploinsufficiency in the gene encoding progranulin1,2. However, the mechanism leading to TDP-43 proteinopathy remains unclear. Here we use single-nucleus RNA sequencing to show that progranulin deficiency promotes microglial transition from a homeostatic to a disease-specific state that causes endolysosomal dysfunction and neurodegeneration in mice. These defects persist even when Grn-/- microglia are cultured ex vivo. In addition, single-nucleus RNA sequencing reveals selective loss of excitatory neurons at disease end-stage, which is characterized by prominent nuclear and cytoplasmic TDP-43 granules and nuclear pore defects. Remarkably, conditioned media from Grn-/- microglia are sufficient to promote TDP-43 granule formation, nuclear pore defects and cell death in excitatory neurons via the complement activation pathway. Consistent with these results, deletion of the genes encoding C1qa and C3 mitigates microglial toxicity and rescues TDP-43 proteinopathy and neurodegeneration. These results uncover previously unappreciated contributions of chronic microglial toxicity to TDP-43 proteinopathy during neurodegeneration.


Subject(s)
Microglia/metabolism , Microglia/pathology , Neurons/metabolism , Neurons/pathology , Progranulins/deficiency , TDP-43 Proteinopathies/metabolism , TDP-43 Proteinopathies/pathology , Aging/genetics , Aging/pathology , Animals , Cell Nucleus/genetics , Cell Nucleus/pathology , Complement Activation/drug effects , Complement Activation/immunology , Complement C1q/antagonists & inhibitors , Complement C1q/immunology , Complement C3b/antagonists & inhibitors , Complement C3b/immunology , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/pharmacology , DNA-Binding Proteins/metabolism , Disease Models, Animal , Female , Male , Mice , Nuclear Pore/metabolism , Nuclear Pore/pathology , Progranulins/genetics , RNA-Seq , Single-Cell Analysis , TDP-43 Proteinopathies/drug therapy , TDP-43 Proteinopathies/genetics , Thalamus/metabolism , Thalamus/pathology , Transcriptome
9.
Curr Opin Cell Biol ; 66: 1-10, 2020 10.
Article in English | MEDLINE | ID: mdl-32371345

ABSTRACT

Cell biology is moving from observing molecules to controlling them in real time, a critical step towards a mechanistic understanding of how cells work. Initially developed from light-gated ion channels to control neuron activity, optogenetics now describes any genetically encoded protein system designed to accomplish specific light-mediated tasks. Recent photosensitive switches use many ingenious designs that bring spatial and temporal control within reach for almost any protein or pathway of interest. This next generation optogenetics includes light-controlled protein-protein interactions and shape-shifting photosensors, which in combination with live microscopy enable acute modulation and analysis of dynamic protein functions in living cells. We provide a brief overview of various types of optogenetic switches. We then discuss how diverse approaches have been used to control cytoskeleton dynamics with light through Rho GTPase signaling, microtubule and actin assembly, mitotic spindle positioning and intracellular transport and highlight advantages and limitations of different experimental strategies.


Subject(s)
Cytoskeleton/metabolism , Optogenetics , Animals , Cytoskeleton/radiation effects , Humans , Light , Signal Transduction/radiation effects , rho GTP-Binding Proteins/metabolism
10.
Methods Mol Biol ; 2101: 211-234, 2020.
Article in English | MEDLINE | ID: mdl-31879907

ABSTRACT

Light can be controlled with high spatial and temporal accuracy. Therefore, optogenetics is an attractive experimental approach to modulate intracellular cytoskeleton dynamics at much faster timescales than by genetic modification. For example, in mammalian cells, microtubules (MTs) grow tens of micrometers per minute and many intracellular MT functions are mediated by a complex of +TIP proteins that dynamically associate with growing MT plus ends. EB1 is a central component of this +TIP protein network, and we recently developed a photo-inactivated π-EB1 by inserting a blue light-sensitive LOV2/Zdk1 module between the EB1 MT-binding domain and the +TIP adaptor domain. Blue light-induced π-EB1 photodissociation results in disassembly of the +TIP complex and strongly attenuates MT growth in mammalian cells.In this chapter, we discuss theoretical and practical aspects of how to perform high-resolution live-cell microscopy in combination with π-EB1 photodissociation. However, these techniques are broadly applicable to other LOV2-based and likely other blue light-sensitive optogenetics. In addition to being a tool to investigate +TIP functions acutely and with subcellular resolution, because of its dramatic and rapid change in intracellular localization, π-EB1 can serve as a powerful tool to test and characterize optogenetic illumination setups. We describe protocols on how to achieve micrometer-scale intracellular control of π-EB1 activity using patterned illumination, and we introduce a do-it-yourself LED cube design compatible with transmitted light microscopy in multiwell plates.


Subject(s)
Microscopy, Fluorescence , Microtubules/metabolism , Optogenetics , Animals , Cell Line , Humans , Microscopy, Fluorescence/instrumentation , Microscopy, Fluorescence/methods , Microtubule-Associated Proteins/metabolism , Optogenetics/instrumentation , Optogenetics/methods , Protein Binding , Protein Interaction Domains and Motifs , Time-Lapse Imaging
11.
J Biol Chem ; 294(22): 8779-8790, 2019 05 31.
Article in English | MEDLINE | ID: mdl-30992364

ABSTRACT

Tau, a member of the MAP2/tau family of microtubule-associated proteins, stabilizes and organizes axonal microtubules in healthy neurons. In neurodegenerative tauopathies, tau dissociates from microtubules and forms neurotoxic extracellular aggregates. MAP2/tau family proteins are characterized by three to five conserved, intrinsically disordered repeat regions that mediate electrostatic interactions with the microtubule surface. Here, we used molecular dynamics, microtubule-binding experiments, and live-cell microscopy, revealing that highly-conserved histidine residues near the C terminus of each microtubule-binding repeat are pH sensors that can modulate tau-microtubule interaction strength within the physiological intracellular pH range. We observed that at low pH (<7.5), these histidines are positively charged and interact with phenylalanine residues in a hydrophobic cleft between adjacent tubulin dimers. At higher pH (>7.5), tau deprotonation decreased binding to microtubules both in vitro and in cells. Electrostatic and hydrophobic characteristics of histidine were both required for tau-microtubule binding, as substitutions with constitutively and positively charged nonaromatic lysine or uncharged alanine greatly reduced or abolished tau-microtubule binding. Consistent with these findings, tau-microtubule binding was reduced in a cancer cell model with increased intracellular pH but was rapidly restored by decreasing the pH to normal levels. These results add detailed insights into the intracellular regulation of tau activity that may be relevant in both normal and pathological conditions.


Subject(s)
Histidine/metabolism , Microtubules/metabolism , tau Proteins/metabolism , Amino Acid Sequence , Binding Sites , Cell Line, Tumor , Humans , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Kinetics , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Protein Binding , Protein Structure, Tertiary , Sequence Alignment , Static Electricity , tau Proteins/genetics
12.
Bioessays ; 41(3): e1800194, 2019 03.
Article in English | MEDLINE | ID: mdl-30730055

ABSTRACT

Microtubules form a highly dynamic filament network in all eukaryotic cells. Individual microtubules grow by tubulin dimer subunit addition and frequently switch between phases of growth and shortening. These unique dynamics are powered by GTP hydrolysis and drive microtubule network remodeling, which is central to eukaryotic cell biology and morphogenesis. Yet, our knowledge of the molecular events at growing microtubule ends remains incomplete. Here, recent ultrastructural, biochemical and cell biological data are integrated to develop a realistic model of growing microtubule ends comprised of structurally distinct but biochemically overlapping zones. Proteins that recognize microtubule lattice conformations associated with specific tubulin guanosine nucleotide states may independently control major structural transitions at growing microtubule ends. A model is proposed in which tubulin dimer addition and subsequent closure of the MT wall are optimized in cells to achieve rapid physiological microtubule growth.


Subject(s)
Microtubules/metabolism , Tubulin/chemistry , Animals , Cell Line, Tumor , Cryoelectron Microscopy , Doublecortin Domain Proteins , Guanosine/chemistry , Guanosine Triphosphate/chemistry , Humans , Hydrolysis , Mammals , Microtubule-Associated Proteins/metabolism , Microtubules/ultrastructure , Models, Molecular , Morphogenesis , Neuropeptides/metabolism , Polymerization , Protein Binding , Protein Conformation , Tubulin/ultrastructure , Tubulin Modulators/metabolism
13.
Nat Cell Biol ; 20(3): 252-261, 2018 03.
Article in English | MEDLINE | ID: mdl-29379139

ABSTRACT

End-binding proteins (EBs) are adaptors that recruit functionally diverse microtubule plus-end-tracking proteins (+TIPs) to growing microtubule plus ends. To test with high spatial and temporal accuracy how, when and where +TIP complexes contribute to dynamic cell biology, we developed a photo-inactivated EB1 variant (π-EB1) by inserting a blue-light-sensitive protein-protein interaction module between the microtubule-binding and +TIP-binding domains of EB1. π-EB1 replaces endogenous EB1 function in the absence of blue light. By contrast, blue-light-mediated π-EB1 photodissociation results in rapid +TIP complex disassembly, and acutely and reversibly attenuates microtubule growth independent of microtubule end association of the microtubule polymerase CKAP5 (also known as ch-TOG and XMAP215). Local π-EB1 photodissociation allows subcellular control of microtubule dynamics at the second and micrometre scale, and elicits aversive turning of migrating cancer cells. Importantly, light-mediated domain splitting can serve as a template to optically control other intracellular protein activities.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Cell Movement , Lung Neoplasms/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Optogenetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Microscopy, Fluorescence , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/radiation effects , Microtubules/genetics , Microtubules/pathology , Microtubules/radiation effects , Photolysis , Protein Binding , Protein Interaction Domains and Motifs , Signal Transduction , Time Factors , Time-Lapse Imaging
14.
J Cell Biol ; 216(8): 2305-2313, 2017 08 07.
Article in English | MEDLINE | ID: mdl-28646105

ABSTRACT

Despite abundant knowledge of the regulation and biochemistry of glycolytic enzymes, we have limited understanding on how they are spatially organized in the cell. Emerging evidence indicates that nonglycolytic metabolic enzymes regulating diverse pathways can assemble into polymers. We now show tetramer- and substrate-dependent filament assembly by phosphofructokinase-1 (PFK1), which is considered the "gatekeeper" of glycolysis because it catalyzes the step committing glucose to breakdown. Recombinant liver PFK1 (PFKL) isoform, but not platelet PFK1 (PFKP) or muscle PFK1 (PFKM) isoforms, assembles into filaments. Negative-stain electron micrographs reveal that filaments are apolar and made of stacked tetramers oriented with exposed catalytic sites positioned along the edge of the polymer. Electron micrographs and biochemical data with a PFKL/PFKP chimera indicate that the PFKL regulatory domain mediates filament assembly. Quantified live-cell imaging shows dynamic properties of localized PFKL puncta that are enriched at the plasma membrane. These findings reveal a new behavior of a key glycolytic enzyme with insights on spatial organization and isoform-specific glucose metabolism in cells.


Subject(s)
Glucose/metabolism , Liver/enzymology , Phosphofructokinase-1, Liver Type/metabolism , Blood Platelets/enzymology , Cell Membrane/enzymology , Glycolysis , HEK293 Cells , Humans , Kinetics , Microscopy, Confocal , Microscopy, Electron, Transmission , Microscopy, Video , Muscle, Skeletal/enzymology , Phosphofructokinase-1, Liver Type/genetics , Phosphofructokinase-1, Liver Type/ultrastructure , Phosphofructokinase-1, Muscle Type/metabolism , Phosphofructokinase-1, Muscle Type/ultrastructure , Phosphofructokinase-1, Type C/metabolism , Phosphofructokinase-1, Type C/ultrastructure , Protein Multimerization , Protein Structure, Quaternary , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure , Structure-Activity Relationship , Substrate Specificity , Time-Lapse Imaging
15.
J Cell Sci ; 130(8): 1404-1412, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28232523

ABSTRACT

Error-free chromosome segregation requires dynamic control of microtubule attachment to kinetochores, but how kinetochore-microtubule interactions are spatially and temporally controlled during mitosis remains incompletely understood. In addition to the NDC80 microtubule-binding complex, other proteins with demonstrated microtubule-binding activities localize to kinetochores. One such protein is the cytoplasmic linker-associated protein 2 (CLASP2). Here, we show that global GSK3-mediated phosphorylation of the longest isoform, CLASP2α, largely abolishes CLASP2α-microtubule association in metaphase. However, it does not directly control localization of CLASP2α to kinetochores. Using dominant phosphorylation-site variants, we find that CLASP2α phosphorylation weakens kinetochore-microtubule interactions as evidenced by decreased tension between sister kinetochores. Expression of CLASP2α phosphorylation-site mutants also resulted in increased chromosome segregation defects, indicating that GSK3-mediated control of CLASP2α-microtubule interactions contributes to correct chromosome dynamics. Because of global inhibition of CLASP2α-microtubule interactions, we propose a model in which only kinetochore-bound CLASP2α is dephosphorylated, locally engaging its microtubule-binding activity.


Subject(s)
Keratinocytes/physiology , Kinetochores/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Mitosis/physiology , CDC2 Protein Kinase , Cell Line , Chromosome Segregation/genetics , Cyclin-Dependent Kinases/metabolism , Cytoskeletal Proteins , Glycogen Synthase Kinase 3/metabolism , Humans , Microtubule-Associated Proteins/genetics , Mutation/genetics , Nuclear Proteins/metabolism , Phosphorylation/genetics , Protein Binding , Protein Engineering
16.
J Cell Sci ; 129(20): 3685-3693, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27672021

ABSTRACT

Autophagy, a pathway for lysosomal-mediated cellular degradation, has recently been described as a regulator of cell migration. Although the molecular mechanisms underlying autophagy-dependent motility are only beginning to emerge, new work demonstrates that selective autophagy mediated by the autophagy cargo receptor, NBR1, specifically promotes the dynamic turnover of integrin-based focal adhesion sites during motility. Here, we discuss the detailed mechanisms through which NBR1-dependent selective autophagy supports focal adhesion remodeling, and we describe the interconnections between this pathway and other established regulators of focal adhesion turnover, such as microtubules. We also highlight studies that examine the contribution of autophagy to selective degradation of proteins that mediate cellular tension and to integrin trafficking; these findings hint at further roles for autophagy in supporting adhesion and migration. Given the recently appreciated importance of selective autophagy in diverse cellular processes, we propose that further investigation into autophagy-mediated focal adhesion turnover will not only shed light onto how focal adhesions are regulated but will also unveil new mechanisms regulating selective autophagy.


Subject(s)
Autophagy , Cell Movement , Animals , Cell Adhesion , Focal Adhesions/metabolism , Humans , Integrins/metabolism , Protein Transport
17.
Curr Biol ; 26(12): 1549-1555, 2016 06 20.
Article in English | MEDLINE | ID: mdl-27238282

ABSTRACT

Many microtubule (MT) functions are mediated by a diverse class of proteins (+TIPs) at growing MT plus ends that control intracellular MT interactions and dynamics and depend on end-binding proteins (EBs) [1]. Cryoelectron microscopy has recently identified the EB binding site as the interface of four tubulin dimers that undergoes a conformational change in response to ß-tubulin GTP hydrolysis [2, 3]. Doublecortin (DCX), a MT-associated protein (MAP) required for neuronal migration during cortical development [4, 5], binds to the same site as EBs [6], and recent in vitro studies proposed DCX localization to growing MT ends independent of EBs [7]. Because this conflicts with observations in neurons [8, 9] and the molecular function of DCX is not well understood, we revisited intracellular DCX dynamics at low expression levels. Here, we report that DCX is not a +TIP in cells but, on the contrary, is excluded from the EB1 domain. In addition, we find that DCX-MT interactions are highly sensitive to MT geometry. In cells, DCX binding was greatly reduced at MT segments with high local curvature. Remarkably, this geometry-dependent binding to MTs was completely reversed in the presence of taxanes, which reconciles incompatible observations in cells [9] and in vitro [10]. We propose a model explaining DCX specificity for different MT geometries based on structural changes induced by GTP hydrolysis that decreases the spacing between adjacent tubulin dimers [11]. Our data are consistent with a unique mode of MT interaction in which DCX specifically recognizes this compacted GDP-like MT lattice.


Subject(s)
Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Neuropeptides/metabolism , Cell Line, Tumor , Doublecortin Domain Proteins , Doublecortin Protein , Guanosine Diphosphate/metabolism , Humans
18.
J Cell Biol ; 212(5): 577-90, 2016 Feb 29.
Article in English | MEDLINE | ID: mdl-26903539

ABSTRACT

Autophagy is a catabolic pathway involving the sequestration of cellular contents into a double-membrane vesicle, the autophagosome. Although recent studies have demonstrated that autophagy supports cell migration, the underlying mechanisms remain unknown. Using live-cell imaging, we uncover that autophagy promotes optimal migratory rate and facilitates the dynamic assembly and disassembly of cell-matrix focal adhesions (FAs), which is essential for efficient motility. Additionally, our studies reveal that autophagosomes associate with FAs primarily during disassembly, suggesting autophagy locally facilitates the destabilization of cell-matrix contact sites. Furthermore, we identify the selective autophagy cargo receptor neighbor of BRCA1 (NBR1) as a key mediator of autophagy-dependent FA remodeling. NBR1 depletion impairs FA turnover and decreases targeting of autophagosomes to FAs, whereas ectopic expression of autophagy-competent, but not autophagy-defective, NBR1 enhances FA disassembly and reduces FA lifetime during migration. Our findings provide mechanistic insight into how autophagy promotes migration by revealing a requirement for NBR1-mediated selective autophagy in enabling FA disassembly in motile cells.


Subject(s)
Autophagy , Focal Adhesions , Proteins/metabolism , Animals , Cells, Cultured , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred C57BL
19.
Dev Cell ; 35(6): 713-24, 2015 Dec 21.
Article in English | MEDLINE | ID: mdl-26702830

ABSTRACT

The proper positioning of organs during development is essential, yet little is known about the regulation of this process in mammals. Using murine tooth development as a model, we have found that cell migration plays a central role in positioning of the organ primordium. By combining lineage tracing, genetic cell ablation, and confocal live imaging, we identified a migratory population of Fgf8-expressing epithelial cells in the embryonic mandible. These Fgf8-expressing progenitors furnish the epithelial cells required for tooth development, and the progenitor population migrates toward a Shh-expressing region in the mandible, where the tooth placode will initiate. Inhibition of Fgf and Shh signaling disrupted the oriented migration of cells, leading to a failure of tooth development. These results demonstrate the importance of intraepithelial cell migration in proper positioning of an initiating organ.


Subject(s)
Cell Movement/physiology , Gene Expression Regulation, Developmental/physiology , Molar/embryology , Morphogenesis/physiology , Tooth/cytology , Animals , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fibroblast Growth Factors/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Mice , Molar/cytology , Molar/metabolism , Odontogenesis/physiology , Tooth/embryology
20.
Dev Cell ; 34(3): 373-8, 2015 Aug 10.
Article in English | MEDLINE | ID: mdl-26212133

ABSTRACT

CRISPR-based technologies have emerged as powerful tools to alter genomes and mark chromosomal loci, but an inexpensive method for generating large numbers of RNA guides for whole genome screening and labeling is lacking. Using a method that permits library construction from any source of DNA, we generated guide libraries that label repetitive loci or a single chromosomal locus in Xenopus egg extracts and show that a complex library can target the E. coli genome at high frequency.


Subject(s)
Bacterial Proteins , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Endonucleases , Gene Library , RNA/genetics , Animals , CRISPR-Associated Protein 9 , Cloning, Molecular , Ovum/cytology , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL
...