Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.790
Filter
Add more filters

Publication year range
1.
Annu Rev Cell Dev Biol ; 35: 29-54, 2019 10 06.
Article in English | MEDLINE | ID: mdl-31394046

ABSTRACT

Microtubules are core components of the cytoskeleton and serve as tracks for motor protein-based intracellular transport. Microtubule networks are highly diverse across different cell types and are believed to adapt to cell type-specific transport demands. Here we review how the spatial organization of different subsets of microtubules into higher-order networks determines the traffic rules for motor-based transport in different animal cell types. We describe the interplay between microtubule network organization and motor-based transport within epithelial cells, oocytes, neurons, cilia, and the spindle apparatus.


Subject(s)
Microtubules/metabolism , Animals , Cell Polarity , Cytoskeleton/chemistry , Cytoskeleton/metabolism , Humans , Microtubule-Associated Proteins/metabolism , Microtubules/chemistry , Molecular Motor Proteins/metabolism , Protein Transport , Spindle Apparatus/chemistry , Spindle Apparatus/metabolism
2.
Nat Rev Mol Cell Biol ; 21(6): 307-326, 2020 06.
Article in English | MEDLINE | ID: mdl-32107477

ABSTRACT

Microtubules are core components of the eukaryotic cytoskeleton with essential roles in cell division, shaping, motility and intracellular transport. Despite their functional heterogeneity, microtubules have a highly conserved structure made from almost identical molecular building blocks: the tubulin proteins. Alternative tubulin isotypes and a variety of post-translational modifications control the properties and functions of the microtubule cytoskeleton, a concept known as the 'tubulin code'. Here we review the current understanding of the molecular components of the tubulin code and how they impact microtubule properties and functions. We discuss how tubulin isotypes and post-translational modifications control microtubule behaviour at the molecular level and how this translates into physiological functions at the cellular and organism levels. We then go on to show how fine-tuning of microtubule function by some tubulin modifications can affect homeostasis and how perturbation of this fine-tuning can lead to a range of dysfunctions, many of which are linked to human disease.


Subject(s)
Microtubules/metabolism , Protein Processing, Post-Translational , Tubulin/genetics , Tubulin/metabolism , Animals , Cell Division , Cell Movement , Cytoskeleton/chemistry , Cytoskeleton/metabolism , Humans , Microtubule-Associated Proteins/metabolism , Microtubules/chemistry , Protein Isoforms , Tubulin/chemistry
3.
Annu Rev Cell Dev Biol ; 34: 1-28, 2018 10 06.
Article in English | MEDLINE | ID: mdl-30059630

ABSTRACT

Intermediate filaments (IFs) are one of the three major elements of the cytoskeleton. Their stability, intrinsic mechanical properties, and cell type-specific expression patterns distinguish them from actin and microtubules. By providing mechanical support, IFs protect cells from external forces and participate in cell adhesion and tissue integrity. IFs form an extensive and elaborate network that connects the cell cortex to intracellular organelles. They act as a molecular scaffold that controls intracellular organization. However, IFs have been revealed as much more than just rigid structures. Their dynamics is regulated by multiple signaling cascades and appears to contribute to signaling events in response to cell stress and to dynamic cellular functions such as mitosis, apoptosis, and migration.


Subject(s)
Cell Biology/trends , Cytoplasm/genetics , Intermediate Filaments/genetics , Microtubules/genetics , Actins/chemistry , Actins/genetics , Cytoplasm/chemistry , Cytoskeleton/chemistry , Cytoskeleton/genetics , Glial Fibrillary Acidic Protein/genetics , Humans , Intermediate Filaments/chemistry , Microtubules/chemistry , Mitosis/genetics , Signal Transduction/genetics
4.
Annu Rev Biochem ; 85: 685-713, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-26865532

ABSTRACT

Autophagy is a conserved intracellular pathway that delivers cytoplasmic contents to lysosomes for degradation via double-membrane autophagosomes. Autophagy substrates include organelles such as mitochondria, aggregate-prone proteins that cause neurodegeneration and various pathogens. Thus, this pathway appears to be relevant to the pathogenesis of diverse diseases, and its modulation may have therapeutic value. Here, we focus on the cell and molecular biology of mammalian autophagy and review the key proteins that regulate the process by discussing their roles and how these may be modulated by posttranslational modifications. We consider the membrane-trafficking events that impact autophagy and the questions relating to the sources of autophagosome membrane(s). Finally, we discuss data from structural studies and some of the insights these have provided.


Subject(s)
Autophagy-Related Proteins/metabolism , Autophagy/genetics , Class III Phosphatidylinositol 3-Kinases/metabolism , Protein Processing, Post-Translational , SNARE Proteins/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Autophagy-Related Proteins/genetics , Class III Phosphatidylinositol 3-Kinases/genetics , Cytoskeleton/chemistry , Cytoskeleton/metabolism , Endocytosis , Humans , Lysosomes/metabolism , Mammals , Models, Molecular , Phagosomes/metabolism , SNARE Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Signal Transduction , rab GTP-Binding Proteins/genetics
5.
Cell ; 166(2): 522-522.e1, 2016 Jul 14.
Article in English | MEDLINE | ID: mdl-27419875

ABSTRACT

Most bacteria and archaea contain filamentous proteins and filament systems that are collectively known as the bacterial cytoskeleton, though not all of them are cytoskeletal, affect cell shape, or maintain intracellular organization. To view this SnapShot, open or download the PDF.


Subject(s)
Bacteria/cytology , Cytoskeleton/chemistry , Archaea/chemistry , Archaea/cytology , Bacteria/chemistry , Bacterial Proteins/analysis
6.
Cell ; 162(2): 403-411, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26165941

ABSTRACT

Small molecules that interfere with microtubule dynamics, such as Taxol and the Vinca alkaloids, are widely used in cell biology research and as clinical anticancer drugs. However, their activity cannot be restricted to specific target cells, which also causes severe side effects in chemotherapy. Here, we introduce the photostatins, inhibitors that can be switched on and off in vivo by visible light, to optically control microtubule dynamics. Photostatins modulate microtubule dynamics with a subsecond response time and control mitosis in living organisms with single-cell spatial precision. In longer-term applications in cell culture, photostatins are up to 250 times more cytotoxic when switched on with blue light than when kept in the dark. Therefore, photostatins are both valuable tools for cell biology, and are promising as a new class of precision chemotherapeutics whose toxicity may be spatiotemporally constrained using light.


Subject(s)
Antimitotic Agents/chemistry , Cell Death , Microtubules/drug effects , Mitosis , Stilbenes/chemistry , Animals , Antimitotic Agents/toxicity , Cell Line, Tumor , Cytoskeleton/chemistry , Humans , Light , Mice , Polymerization , Stilbenes/toxicity
7.
Proc Natl Acad Sci U S A ; 119(3)2022 01 18.
Article in English | MEDLINE | ID: mdl-35042781

ABSTRACT

The precise assembly and disassembly of actin filaments is required for several cellular processes, and their regulation has been scrutinized for decades. Twenty years ago, a handful of studies marked the advent of a new type of experiment to study actin dynamics: using optical microscopy to look at individual events, taking place on individual filaments in real time. Here, we summarize the main characteristics of this approach and how it has changed our ability to understand actin assembly dynamics. We also highlight some of its caveats and reflect on what we have learned over the past 20 years, leading us to propose a set of guidelines, which we hope will contribute to a better exploitation of this powerful tool.


Subject(s)
Actin Cytoskeleton/chemistry , Actin Cytoskeleton/metabolism , Actins/chemistry , Actins/metabolism , Biochemistry , Biophysics , Cytoskeleton/chemistry , Cytoskeleton/metabolism , Hydrogen-Ion Concentration , Kinetics , Microscopy, Fluorescence , Temperature
8.
Nat Immunol ; 13(7): 674-80, 2012 Jun 03.
Article in English | MEDLINE | ID: mdl-22660579

ABSTRACT

The binding of T cell antigen receptors (TCRs) to specific complexes of peptide and major histocompatibility complex (pMHC) is typically of very low affinity, which necessitates the use of multimeric pMHC complexes to label T lymphocytes stably. We report here the development of pMHC complexes able to be crosslinked by ultraviolet irradiation; even as monomers, these efficiently and specifically stained cognate T cells. We also used this reagent to probe T cell activation and found that a covalently bound pMHC was more stimulatory than an agonist pMHC on lipid bilayers. This finding suggested that serial engagement of TCRs is dispensable for activation when a substantial fraction of TCRs are stably engaged. Finally, pMHC-bound TCRs were 'preferentially' transported into the central supramolecular activation cluster after activation, which suggested that ligand engagement enabled linkage of the TCR and its associated CD3 signaling molecules to the cytoskeleton.


Subject(s)
Cross-Linking Reagents/chemistry , Major Histocompatibility Complex/immunology , Receptors, Antigen, T-Cell/chemistry , T-Lymphocytes/chemistry , Animals , CD3 Complex/chemistry , CD3 Complex/immunology , Cells, Cultured , Coloring Agents/chemistry , Cytoskeleton/chemistry , Cytoskeleton/immunology , Lymphocyte Activation , Mice , Mice, Transgenic , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology
9.
Nat Rev Mol Cell Biol ; 13(3): 183-94, 2012 Feb 08.
Article in English | MEDLINE | ID: mdl-22314400

ABSTRACT

Septins belong to a family of proteins that is highly conserved in eukaryotes and is increasingly recognized as a novel component of the cytoskeleton. All septins are GTP-binding proteins that form hetero-oligomeric complexes and higher-order structures, including filaments and rings. Recent studies have provided structural information about the different levels of septin organization; however, the crucial structural determinants and factors responsible for septin assembly remain unclear. Investigations on the molecular functions of septins have highlighted their roles as scaffolds for protein recruitment and as diffusion barriers for subcellular compartmentalization in numerous biological processes, including cell division and host-microorganism interactions.


Subject(s)
Cytoskeleton/metabolism , Septins/physiology , Animals , Bacterial Infections/immunology , Bacterial Infections/metabolism , Cytoskeleton/chemistry , Host-Pathogen Interactions , Humans , Immunity, Innate , Permeability , Protein Structure, Quaternary , Protein Structure, Tertiary , Septins/chemistry , Septins/metabolism
10.
EMBO J ; 38(11)2019 06 03.
Article in English | MEDLINE | ID: mdl-31036554

ABSTRACT

To characterize keratin intermediate filament assembly mechanisms at atomic resolution, we determined the crystal structure of wild-type human keratin-1/keratin-10 helix 1B heterotetramer at 3.0 Å resolution. It revealed biochemical determinants for the A11 mode of axial alignment in keratin filaments. Four regions on a hydrophobic face of the K1/K10-1B heterodimer dictated tetramer assembly: the N-terminal hydrophobic pocket (defined by L227K1, Y230K1, F231K1, and F234K1), the K10 hydrophobic stripe, K1 interaction residues, and the C-terminal anchoring knob (formed by F314K1 and L318K1). Mutation of both knob residues to alanine disrupted keratin 1B tetramer and full-length filament assembly. Individual knob residue mutant F314AK1, but not L318AK1, abolished 1B tetramer formation. The K1-1B knob/pocket mechanism is conserved across keratins and many non-keratin intermediate filaments. To demonstrate how pathogenic mutations cause skin disease by altering filament assembly, we additionally determined the 2.39 Å structure of K1/10-1B containing a S233LK1 mutation linked to epidermolytic palmoplantar keratoderma. Light scattering and circular dichroism measurements demonstrated enhanced aggregation of K1S233L/K10-1B in solution without affecting secondary structure. The K1S233L/K10-1B octamer structure revealed S233LK1 causes aberrant hydrophobic interactions between 1B tetramers.


Subject(s)
Intermediate Filament Proteins/chemistry , Intermediate Filament Proteins/metabolism , Keratin-10 , Keratin-1 , Protein Interaction Domains and Motifs , Protein Multimerization/physiology , Amino Acid Substitution , Circular Dichroism , Crystallography, X-Ray , Cytoskeleton/chemistry , Cytoskeleton/metabolism , Dynamic Light Scattering , Humans , Hydrophobic and Hydrophilic Interactions , Intermediate Filament Proteins/genetics , Keratin-1/chemistry , Keratin-1/genetics , Keratin-1/metabolism , Keratin-10/chemistry , Keratin-10/genetics , Keratin-10/metabolism , Models, Molecular , Mutation, Missense , Protein Folding , Protein Interaction Domains and Motifs/genetics , Protein Structure, Quaternary , Protein Structure, Secondary , Skin Diseases/genetics , Skin Diseases/metabolism , Skin Diseases/pathology
11.
PLoS Pathog ; 17(8): e1009902, 2021 08.
Article in English | MEDLINE | ID: mdl-34460869

ABSTRACT

The p21-activated kinase (PAK) family regulate a multitude of cellular processes, including actin cytoskeleton remodelling. Numerous bacterial pathogens usurp host signalling pathways that regulate actin reorganisation in order to promote Infection. Salmonella and pathogenic Escherichia coli drive actin-dependent forced uptake and intimate attachment respectively. We demonstrate that the pathogen-driven generation of both these distinct actin structures relies on the recruitment and activation of PAK. We show that the PAK kinase domain is dispensable for this actin remodelling, which instead requires the GTPase-binding CRIB and the central poly-proline rich region. PAK interacts with and inhibits the guanine nucleotide exchange factor ß-PIX, preventing it from exerting a negative effect on cytoskeleton reorganisation. This kinase-independent function of PAK may be usurped by other pathogens that modify host cytoskeleton signalling and helps us better understand how PAK functions in normal and diseased eukaryotic cells.


Subject(s)
Actins/chemistry , Cytoskeleton/chemistry , Salmonella Infections/microbiology , Salmonella enterica/physiology , p21-Activated Kinases/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Phosphorylation , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , Salmonella Infections/metabolism , Salmonella Infections/pathology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , p21-Activated Kinases/genetics
12.
Nat Chem Biol ; 17(5): 608-614, 2021 05.
Article in English | MEDLINE | ID: mdl-33686294

ABSTRACT

Many RNA-binding proteins undergo liquid-liquid phase separation, which underlies the formation of membraneless organelles, such as stress granules and P-bodies. Studies of the molecular mechanism of phase separation in vitro are hampered by the coalescence and sedimentation of organelle-sized droplets interacting with glass surfaces. Here, we demonstrate that liquid droplets of fused in sarcoma (FUS)-a protein found in cytoplasmic aggregates of amyotrophic lateral sclerosis and frontotemporal dementia patients-can be stabilized in vitro using an agarose hydrogel that acts as a cytoskeleton mimic. This allows their spectroscopic characterization by liquid-phase NMR and electron paramagnetic resonance spectroscopy. Protein signals from both dispersed and condensed phases can be observed simultaneously, and their respective proportions can be quantified precisely. Furthermore, the agarose hydrogel acts as a cryoprotectant during shock-freezing, which facilitates pulsed electron paramagnetic resonance measurements at cryogenic temperatures. Surprisingly, double electron-electron resonance measurements revealed a compaction of FUS in the condensed phase.


Subject(s)
Cryoprotective Agents/chemistry , Hydrogels/chemistry , RNA-Binding Protein FUS/chemistry , Sepharose/chemistry , Biomimetic Materials/chemistry , Cloning, Molecular , Cytoskeleton/chemistry , Electron Spin Resonance Spectroscopy , Escherichia coli/genetics , Escherichia coli/metabolism , Eukaryotic Cells/chemistry , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation , Recombinant Proteins/chemistry
13.
Soft Matter ; 19(29): 5430-5442, 2023 Jul 26.
Article in English | MEDLINE | ID: mdl-37272768

ABSTRACT

Cytoskeletal gels are prototyped to reproduce the mechanical contraction of the cytoskeleton in vitro. They are composed of a polymer network (backbone), swollen by the presence of a liquid solvent, and active molecules (molecular motors, MMs) that transduce chemical energy into the mechanical work of contraction. These motors attach to the polymer chains to shorten them and/or act as dynamic crosslinks, thereby constraining the thermal fluctuations of the chains. We describe both mechanisms thermodynamically as a microstructural reconfiguration, where the backbone stiffens to motivate solvent (out)flow and accommodate contraction. Via simple steady-state energetic analysis, under the simplest case of isotropic deformation, we quantify the mechanical energy required to achieve contraction as a function of polymer chain density and molecular motor density. We identify two limit regimes, namely, fast MM activation (FM), and slow MM activation (SM). FM assumes that MMs provide all the available mechanical energy 'instantaneously' and leave the polymer in a stiffened state, i.e. the MM activity occurs at a time scale that is much smaller than that of solvent diffusion. SM assumes that the timescale for MM activation is much longer than that of solvent diffusion. To achieve the same final contracted state, FM requires the largest amount of work per unit reference volume, while SM requires the least. For all intermediate cases where the timescale of MM activation is comparable with that of solvent diffusion, the required work ranges between these two limits. We provide all these quantities as a function of chain density and MM density. Finally, we compare our results on contraction energetics with experiments and observe good agreement.


Subject(s)
Cytoskeleton , Polymers , Cytoskeleton/chemistry , Gels/chemistry , Polymers/chemistry , Solvents , Microtubules
14.
Nature ; 543(7644): 261-264, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28241138

ABSTRACT

The nuclear lamina is a fundamental constituent of metazoan nuclei. It is composed mainly of lamins, which are intermediate filament proteins that assemble into a filamentous meshwork, bridging the nuclear envelope and chromatin. Besides providing structural stability to the nucleus, the lamina is involved in many nuclear activities, including chromatin organization, transcription and replication. However, the structural organization of the nuclear lamina is poorly understood. Here we use cryo-electron tomography to obtain a detailed view of the organization of the lamin meshwork within the lamina. Data analysis of individual lamin filaments resolves a globular-decorated fibre appearance and shows that A- and B-type lamins assemble into tetrameric filaments of 3.5 nm thickness. Thus, lamins exhibit a structure that is remarkably different from the other canonical cytoskeletal elements. Our findings define the architecture of the nuclear lamin meshworks at molecular resolution, providing insights into their role in scaffolding the nuclear lamina.


Subject(s)
Lamins/chemistry , Lamins/ultrastructure , Nuclear Lamina/chemistry , Nuclear Lamina/ultrastructure , Animals , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Chromatin/ultrastructure , Cryoelectron Microscopy , Cytoskeleton/chemistry , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Humans , Intermediate Filament Proteins/chemistry , Intermediate Filament Proteins/metabolism , Intermediate Filament Proteins/ultrastructure , Lamins/metabolism , Mice , Nuclear Lamina/metabolism , Tomography
15.
Proc Natl Acad Sci U S A ; 117(40): 24670-24678, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32958682

ABSTRACT

Cell crawling requires the generation of intracellular forces by the cytoskeleton and their transmission to an extracellular substrate through specific adhesion molecules. Crawling cells show many features of excitable systems, such as spontaneous symmetry breaking and crawling in the absence of external cues, and periodic and propagating waves of activity. Mechanical instabilities in the active cytoskeleton network and feedback loops in the biochemical network of activators and repressors of cytoskeleton dynamics have been invoked to explain these dynamical features. Here, I show that the interplay between the dynamics of cell-substrate adhesion and linear cellular mechanics is sufficient to reproduce many nonlinear dynamical patterns observed in spreading and crawling cells. Using an analytical formalism of the molecular clutch model of cell adhesion, regulated by local mechanical forces, I show that cellular traction forces exhibit stick-slip dynamics resulting in periodic waves of protrusion/retraction and propagating waves along the cell edge. This can explain spontaneous symmetry breaking and polarization of spreading cells, leading to steady crawling or bipedal motion, and bistability, where persistent cell motion requires a sufficiently strong transient external stimulus. The model also highlights the role of membrane tension in providing the long-range mechanical communication across the cell required for symmetry breaking.


Subject(s)
Actins/metabolism , Cell Polarity , Cells/cytology , Actins/chemistry , Biomechanical Phenomena , Cell Adhesion , Cell Movement , Cell Surface Extensions , Cells/chemistry , Cells/metabolism , Cytoskeleton/chemistry , Cytoskeleton/metabolism , Kinetics , Models, Biological
16.
Proc Natl Acad Sci U S A ; 117(11): 5861-5872, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32123101

ABSTRACT

The cytoskeletal protein actin polymerizes into filaments that are essential for the mechanical stability of mammalian cells. In vitro experiments showed that direct interactions between actin filaments and lipid bilayers are possible and that the net charge of the bilayer as well as the presence of divalent ions in the buffer play an important role. In vivo, colocalization of actin filaments and divalent ions are suppressed, and cells rely on linker proteins to connect the plasma membrane to the actin network. Little is known, however, about why this is the case and what microscopic interactions are important. A deeper understanding is highly beneficial, first, to obtain understanding in the biological design of cells and, second, as a possible basis for the building of artificial cortices for the stabilization of synthetic cells. Here, we report the results of coarse-grained molecular dynamics simulations of monomeric and filamentous actin in the vicinity of differently charged lipid bilayers. We observe that charges on the lipid head groups strongly determine the ability of actin to adsorb to the bilayer. The inclusion of divalent ions leads to a reversal of the binding affinity. Our in silico results are validated experimentally by reconstitution assays with actin on lipid bilayer membranes and provide a molecular-level understanding of the actin-membrane interaction.


Subject(s)
Actin Cytoskeleton/chemistry , Actin Cytoskeleton/metabolism , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Actins/chemistry , Artificial Cells , Cell Membrane/chemistry , Cell Membrane/metabolism , Chemical Phenomena , Computational Biology , Computer Simulation , Cytoskeleton/chemistry , Cytoskeleton/metabolism , Ions/chemistry , Ions/metabolism , Models, Molecular , Molecular Dynamics Simulation , Protein Binding , Protein Conformation , Static Electricity
17.
Proc Natl Acad Sci U S A ; 117(4): 1902-1909, 2020 01 28.
Article in English | MEDLINE | ID: mdl-31932440

ABSTRACT

Executing gene circuits by cell-free transcription-translation into cell-sized compartments, such as liposomes, is one of the major bottom-up approaches to building minimal cells. The dynamic synthesis and proper self-assembly of macromolecular structures inside liposomes, the cytoskeleton in particular, stands as a central limitation to the development of cell analogs genetically programmed. In this work, we express the Escherichia coli gene mreB inside vesicles with bilayers made of lipid-polyethylene glycol (PEG). We demonstrate that two-dimensional molecular crowding, emulated by the PEG molecules at the lipid bilayer, is enough to promote the polymerization of the protein MreB at the inner membrane into a sturdy cytoskeleton capable of transforming spherical liposomes into elongated shapes, such as rod-like compartments. We quantitatively describe this mechanism with respect to the size of liposomes, lipid composition of the membrane, crowding at the membrane, and strength of MreB synthesis. So far unexplored, molecular crowding at the surface of synthetic cells emerges as an additional development with potential broad applications. The symmetry breaking observed could be an important step toward compartment self-reproduction.


Subject(s)
Artificial Cells/metabolism , Cell Membrane/metabolism , Cell Shape , Cytoskeleton/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli/metabolism , Liposomes/metabolism , Cell Membrane/chemistry , Cytoskeleton/chemistry , Escherichia coli/cytology , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Liposomes/chemistry , Polymerization , Protein Biosynthesis , Protein Conformation
18.
Proc Natl Acad Sci U S A ; 117(33): 19904-19913, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32747565

ABSTRACT

Asgard archaea genomes contain potential eukaryotic-like genes that provide intriguing insight for the evolution of eukaryotes. The eukaryotic actin polymerization/depolymerization cycle is critical for providing force and structure in many processes, including membrane remodeling. In general, Asgard genomes encode two classes of actin-regulating proteins from sequence analysis, profilins and gelsolins. Asgard profilins were demonstrated to regulate actin filament nucleation. Here, we identify actin filament severing, capping, annealing and bundling, and monomer sequestration activities by gelsolin proteins from Thorarchaeota (Thor), which complete a eukaryotic-like actin depolymerization cycle, and indicate complex actin cytoskeleton regulation in Asgard organisms. Thor gelsolins have homologs in other Asgard archaea and comprise one or two copies of the prototypical gelsolin domain. This appears to be a record of an initial preeukaryotic gene duplication event, since eukaryotic gelsolins are generally comprise three to six domains. X-ray structures of these proteins in complex with mammalian actin revealed similar interactions to the first domain of human gelsolin or cofilin with actin. Asgard two-domain, but not one-domain, gelsolins contain calcium-binding sites, which is manifested in calcium-controlled activities. Expression of two-domain gelsolins in mammalian cells enhanced actin filament disassembly on ionomycin-triggered calcium release. This functional demonstration, at the cellular level, provides evidence for a calcium-controlled Asgard actin cytoskeleton, indicating that the calcium-regulated actin cytoskeleton predates eukaryotes. In eukaryotes, dynamic bundled actin filaments are responsible for shaping filopodia and microvilli. By correlation, we hypothesize that the formation of the protrusions observed from Lokiarchaeota cell bodies may involve the gelsolin-regulated actin structures.


Subject(s)
Actin Depolymerizing Factors/metabolism , Actins/metabolism , Archaea/metabolism , Archaeal Proteins/metabolism , Gelsolin/metabolism , Actin Depolymerizing Factors/chemistry , Actin Depolymerizing Factors/genetics , Actins/chemistry , Actins/genetics , Amino Acid Sequence , Archaea/chemistry , Archaea/genetics , Archaeal Proteins/chemistry , Archaeal Proteins/genetics , Cytoskeleton/chemistry , Cytoskeleton/genetics , Cytoskeleton/metabolism , Evolution, Molecular , Gelsolin/chemistry , Gelsolin/genetics , Genome, Archaeal , Polymerization , Protein Conformation, alpha-Helical , Sequence Alignment
19.
Biophys J ; 121(3): 502-514, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34954156

ABSTRACT

The sizes of filamentous structures in a cell are often regulated for many physiological processes. A key question in cell biology is how such size control is achieved. Here, we theoretically study the length distributions of multiple filaments, growing by stochastic assembly and disassembly of subunits from a limiting subunit pool. Importantly, we consider a chemical switching of subunits (hydrolysis) prevalent in many biofilaments like microtubules (MTs). We show by simulations of different models that hydrolysis leads to a skewed unimodal length distribution for a single MT. In contrast, hydrolysis can lead to bimodal distributions of individual lengths for two MTs, where individual filaments toggle stochastically between bigger and smaller sizes. For more than two MTs, length distributions are also bimodal, although the bimodality becomes less prominent. We further show that this collective phenomenon is connected with the nonequilibrium nature of hydrolysis, and the bimodality disappears for reversible dynamics. Consistent with earlier theoretical studies, a homogeneous subunit pool, without hydrolysis, cannot control filament lengths. We thus elucidate the role of hydrolysis as a control mechanism on MT length diversity.


Subject(s)
Cytoskeleton , Microtubules , Cytoskeleton/chemistry , Hydrolysis , Microtubules/chemistry
20.
J Biol Chem ; 296: 100232, 2021.
Article in English | MEDLINE | ID: mdl-33372034

ABSTRACT

Myosin VI ensembles on endocytic cargo facilitate directed transport through a dense cortical actin network. Myosin VI is recruited to clathrin-coated endosomes via the cargo adaptor Dab2. Canonically, it has been assumed that the interactions between a motor and its cargo adaptor are stable. However, it has been demonstrated that the force generated by multiple stably attached motors disrupts local cytoskeletal architecture, potentially compromising transport. In this study, we demonstrate that dynamic multimerization of myosin VI-Dab2 complexes facilitates cargo processivity without significant reorganization of cortical actin networks. Specifically, we find that Dab2 myosin interacting region (MIR) binds myosin VI with a moderate affinity (184 nM) and single-molecule kinetic measurements demonstrate a high rate of turnover (1 s-1) of the Dab2 MIR-myosin VI interaction. Single-molecule motility shows that saturating Dab2-MIR concentration (2 µM) promotes myosin VI homodimerization and processivity with run lengths comparable with constitutive myosin VI dimers. Cargo-mimetic DNA origami scaffolds patterned with Dab2 MIR-myosin VI complexes are weakly processive, displaying sparse motility on single actin filaments and "stop-and-go" motion on a cellular actin network. On a minimal actin cortex assembled on lipid bilayers, unregulated processive movement by either constitutive myosin V or VI dimers results in actin remodeling and foci formation. In contrast, Dab2 MIR-myosin VI interactions preserve the integrity of a minimal cortical actin network. Taken together, our study demonstrates the importance of dynamic motor-cargo association in enabling cargo transportation without disrupting cytoskeletal organization.


Subject(s)
Actin Cytoskeleton/genetics , Adaptor Proteins, Signal Transducing/chemistry , Apoptosis Regulatory Proteins/chemistry , Multiprotein Complexes/chemistry , Myosin Heavy Chains/chemistry , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/ultrastructure , Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/ultrastructure , Clathrin-Coated Vesicles/chemistry , Clathrin-Coated Vesicles/genetics , Cytoskeleton/chemistry , Cytoskeleton/genetics , Cytoskeleton/ultrastructure , Endocytosis/genetics , Endosomes/genetics , Humans , Kinetics , Multiprotein Complexes/genetics , Multiprotein Complexes/ultrastructure , Myosin Heavy Chains/genetics , Myosin Heavy Chains/ultrastructure , Phosphatidylserines/genetics , Protein Binding/genetics , Protein Multimerization/genetics , Single Molecule Imaging
SELECTION OF CITATIONS
SEARCH DETAIL