Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 37
1.
J Phys Chem B ; 128(19): 4590-4601, 2024 May 16.
Article En | MEDLINE | ID: mdl-38701111

Cofilin, a key actin-binding protein, orchestrates the dynamics of the actomyosin network through its actin-severing activity and by promoting the recycling of actin monomers. Recent experiments suggest that cofilin forms functionally distinct oligomers via thiol post-translational modifications (PTMs) that promote actin nucleation and assembly. Despite these advances, the structural conformations of cofilin oligomers that modulate actin activity remain elusive because there are combinatorial ways to oxidize thiols in cysteines to form disulfide bonds rapidly. This study employs molecular dynamics simulations to investigate human cofilin 1 as a case study for exploring cofilin dimers via disulfide bond formation. Utilizing a biasing scheme in simulations, we focus on analyzing dimer conformations conducive to disulfide bond formation. Additionally, we explore potential PTMs arising from the examined conformational ensemble. Using the free energy profiling, our simulations unveil a range of probable cofilin dimer structures not represented in current Protein Data Bank entries. These candidate dimers are characterized by their distinct population distributions and relative free energies. Of particular note is a dimer featuring an interface between cysteines 139 and 147 residues, which demonstrates stable free energy characteristics and intriguingly symmetrical geometry. In contrast, the experimentally proposed dimer structure exhibits a less stable free energy profile. We also evaluate frustration quantification based on the energy landscape theory in the protein-protein interactions at the dimer interfaces. Notably, the 39-39 dimer configuration emerges as a promising candidate for forming cofilin tetramers, as substantiated by frustration analysis. Additionally, docking simulations with actin filaments further evaluate the stability of these cofilin dimer-actin complexes. Our findings thus offer a computational framework for understanding the role of thiol PTM of cofilin proteins in regulating oligomerization, and the subsequent cofilin-mediated actin dynamics in the actomyosin network.


Actin Cytoskeleton , Disulfides , Molecular Dynamics Simulation , Disulfides/chemistry , Humans , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/metabolism , Cofilin 1/chemistry , Cofilin 1/metabolism , Protein Multimerization , Actins/chemistry , Actins/metabolism , Actin Depolymerizing Factors/chemistry , Actin Depolymerizing Factors/metabolism , Thermodynamics
2.
Gene ; 819: 146206, 2022 Apr 20.
Article En | MEDLINE | ID: mdl-35092861

The cofilin-1 protein, encoded by CFL1, is an actin-binding protein that regulates F-actin depolymerization and nucleation activity through phosphorylation and dephosphorylation. CFL1 has been implicated in the development of neurodegenerative diseases (Alzheimer's disease and Huntington's disease), neuronal migration disorders (lissencephaly, epilepsy, and schizophrenia), and neural tube closure defects. Mutations in CFL1 have been associated with impaired neural crest cell migration and neural tube closure defects. In our study, various computational approaches were utilized to explore single-nucleotide polymorphisms (SNPs) in CFL1. The Variation Viewer and gnomAD databases were used to retrieve CFL1 SNPs, including 46 nonsynonymous SNPs (nsSNPs). The functional and structural annotation of SNPs was performed using 12 sequence-based web applications, which identified 20 nsSNPs as being the most likely to be deleterious or disease-causing. The conservation of cofilin-1 protein structures was illustrated using the ConSurf and PROSITE web servers, which projected the 12 most deleterious nsSNPs onto conserved domains, with the potential to disrupt the protein's functionality. These 12 nsSNPs were selected for protein structure construction, and the DynaMut/DUET servers predicted that the protein variants V7G, L84P, and L99A were the most likely to be damaging to the cofilin-1 protein structure or function. The evaluation of molecular docking studies demonstrated that the L99A and L84P cofilin-1 variants reduce the binding affinity for actin compared with the native cofilin-1 structure, and molecular dynamic simulation studies confirmed that these variants might destabilize the protein structure. The consequences of putative mutations on protein-protein interactions and post-translational modification sites in the cofilin-1 protein structure were analyzed. This study represents the first complete approach to understanding the effects of nsSNPs within the actin-depolymerizing factor/cofilin family, which suggested that SNPs resulting in L84P (rs199716082) and L99A (rs267603119) variants represent significant CFL1 mutations associated with disease development.


Cofilin 1/genetics , Cofilin 1/metabolism , Polymorphism, Single Nucleotide , Amino Acid Sequence , Cofilin 1/chemistry , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Mutant Proteins/metabolism , Mutation , Phylogeny , Protein Conformation , Protein Domains/genetics
3.
Biochem Biophys Res Commun ; 569: 187-192, 2021 09 10.
Article En | MEDLINE | ID: mdl-34256187

Cofilin-1, an actin dynamizing protein, forms actin-cofilin rods, which is one of the major events that exacerbates the pathophysiology of amyloidogenic diseases. Cysteine oxidation in cofilin-1 under oxidative stress plays a crucial role in the formation of these rods. Others and we have reported that cofilin-1 possesses a self-oligomerization property in vitro and in vivo under physiological conditions. However, it remains elusive if cofilin-1 itself forms amyloid-like structures. We, therefore, hypothesized that cofilin-1 might form amyloid-like assemblies, with a potential to intensify the pathophysiology of amyloid-linked diseases. We used various in silico and in vitro techniques and examined the amyloid-forming propensity of cofilin-1. The study confirms that cofilin-1 possesses an intrinsic tendency of aggregation and forms amyloid-like structures in vitro. Further, we studied the effect of cysteine oxidation on the stability and structural features of cofilin-1. Our data show that oxidation at Cys-80 renders cofilin-1 unstable, leading to a partial loss of protein structure. The results substantiate our hypothesis and establish a strong possibility that cofilin-1 aggregation might play a role in cofilin-mediated pathology and the progression of several amyloid-linked diseases.


Amyloidogenic Proteins/metabolism , Cofilin 1/metabolism , Cysteine/metabolism , Neurodegenerative Diseases/metabolism , Alzheimer Disease/diagnosis , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amino Acid Sequence , Amyloid/chemistry , Amyloid/metabolism , Amyloidogenic Proteins/chemistry , Amyloidogenic Proteins/genetics , Cofilin 1/chemistry , Cofilin 1/genetics , Computer Simulation , Cysteine/chemistry , Cysteine/genetics , Humans , Models, Molecular , Mutation , Neurodegenerative Diseases/diagnosis , Neurodegenerative Diseases/genetics , Oxidation-Reduction , Propensity Score , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/metabolism , Protein Stability , Protein Unfolding , Sequence Homology, Amino Acid
4.
PLoS Biol ; 18(11): e3000925, 2020 11.
Article En | MEDLINE | ID: mdl-33216759

Lifeact is a short actin-binding peptide that is used to visualize filamentous actin (F-actin) structures in live eukaryotic cells using fluorescence microscopy. However, this popular probe has been shown to alter cellular morphology by affecting the structure of the cytoskeleton. The molecular basis for such artefacts is poorly understood. Here, we determined the high-resolution structure of the Lifeact-F-actin complex using electron cryo-microscopy (cryo-EM). The structure reveals that Lifeact interacts with a hydrophobic binding pocket on F-actin and stretches over 2 adjacent actin subunits, stabilizing the DNase I-binding loop (D-loop) of actin in the closed conformation. Interestingly, the hydrophobic binding site is also used by actin-binding proteins, such as cofilin and myosin and actin-binding toxins, such as the hypervariable region of TccC3 (TccC3HVR) from Photorhabdus luminescens and ExoY from Pseudomonas aeruginosa. In vitro binding assays and activity measurements demonstrate that Lifeact indeed competes with these proteins, providing an explanation for the altering effects of Lifeact on cell morphology in vivo. Finally, we demonstrate that the affinity of Lifeact to F-actin can be increased by introducing mutations into the peptide, laying the foundation for designing improved actin probes for live cell imaging.


Actins/chemistry , Microfilament Proteins/chemistry , Actins/metabolism , Actins/ultrastructure , Animals , Bacterial Toxins/chemistry , Binding Sites , Binding, Competitive , Cofilin 1/chemistry , Cofilin 1/ultrastructure , Cryoelectron Microscopy , Fluorescent Dyes/chemistry , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , In Vitro Techniques , Microfilament Proteins/metabolism , Microfilament Proteins/ultrastructure , Microscopy, Confocal , Models, Molecular , Myosins/chemistry , Peptide Fragments/chemistry , Peptide Fragments/ultrastructure , Protein Engineering , Protein Interaction Domains and Motifs , Rabbits , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/ultrastructure , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/ultrastructure
5.
Arch Biochem Biophys ; 682: 108280, 2020 03 30.
Article En | MEDLINE | ID: mdl-31996302

Tropomyosin and cofilin are involved in the regulation of actin filament dynamic polymerization and depolymerization. Binding of cofilin changes actin filaments structure, leading to their severing and depolymerization. Non-muscle tropomyosin isoforms were shown before to differentially regulate the activity of cofilin 1; products of TPM1 gene stabilized actin filaments, but products of TPM3 gene promoted cofilin-dependent severing and depolymerization. Here, conformational changes at the longitudinal and lateral interface between actin subunits resulting from tropomyosin and cofilin 1 binding were studied using skeletal actin and yeast wild type and mutant Q41C and S265C actins. Cross-linking of F-actin and fluorescence changes in F-actin labeled with acrylodan at Cys41 (in D-loop) or Cys265 (in H-loop) showed that tropomyosin isoforms differentially regulated cofilin-induced conformational rearrangements at longitudinal and lateral filament interfaces. Tryptic digestion of F-Mg-actin confirmed the differences between tropomyosin isoforms in their regulation of cofilin-dependent changes at actin-actin interfaces. Changes in the fluorescence of AEDANS attached to C-terminal Cys of actin, as well as FRET between Trp residues in actin subdomain 1 and AEDANS, did not show differences in the conformation of the C-terminal segment of F-actin in the presence of different tropomyosins ± cofilin 1. Therefore, actin's D- and H-loop are the sites involved in regulation of cofilin activity by tropomyosin isoforms.


Actin Cytoskeleton/chemistry , Actins/chemistry , Cofilin 1/chemistry , Tropomyosin/chemistry , Animals , Cytoskeleton/chemistry , Humans , Mice , Models, Molecular , Mutation , Polymerization , Protein Binding , Protein Domains , Protein Isoforms , Rabbits , Saccharomyces cerevisiae , Trypsin/chemistry
6.
Biochemistry ; 58(1): 40-47, 2019 01 08.
Article En | MEDLINE | ID: mdl-30499293

Actin depolymerizing factor (ADF)/cofilin is the main protein family promoting the disassembly of actin filaments, which is essential for numerous cellular functions. ADF/cofilin proteins disassemble actin filaments through different reactions, as they bind to their sides, sever them, and promote the depolymerization of the resulting ADF/cofilin-saturated filaments. Moreover, the efficiency of ADF/cofilin is known to be very sensitive to pH. ADF/cofilin thus illustrates two challenges in actin biochemistry: separating the different regulatory actions of a single protein and characterizing them as a function of specific biochemical conditions. Here, we investigate the different reactions of ADF/cofilin on actin filaments, at four different pH values ranging from 6.6 to 7.8, using single-filament microfluidics techniques. We show that decreasing the pH decreases the effective filament severing rate by increasing the rate at which filaments become saturated by ADF/cofilin, thereby reducing the number of ADF/cofilin domain boundaries, where severing can occur. The severing rate per domain boundary, however, remains unchanged at different pH values. The ADF/cofilin-decorated filaments ("cofilactin" filaments) depolymerize from both ends. We show here that, at physiological pH (7.0-7.4), the pointed end depolymerization of cofilactin filaments is barely faster than that of bare filaments. In contrast, cofilactin barbed ends undergo an "unstoppable" depolymerization (depolymerizing for minutes despite the presence of free actin monomers and capping protein in solution), throughout our pH range. We thus show that, at physiological pH, the main contribution of ADF/cofilin to filament depolymerization is at the barbed end.


Actin Cytoskeleton/metabolism , Actin Depolymerizing Factors/metabolism , Actins/metabolism , Cofilin 1/metabolism , Actin Cytoskeleton/chemistry , Actin Depolymerizing Factors/chemistry , Actins/chemistry , Animals , Cofilin 1/chemistry , Humans , Hydrogen-Ion Concentration , Mice , Rabbits
7.
Mol Cell Proteomics ; 18(3): 504-519, 2019 03.
Article En | MEDLINE | ID: mdl-30587509

The fumarate ester dimethyl fumarate (DMF) has been introduced recently as a treatment for relapsing remitting multiple sclerosis (RRMS), a chronic inflammatory condition that results in neuronal demyelination and axonal loss. DMF is known to act by depleting intracellular glutathione and modifying thiols on Keap1 protein, resulting in the stabilization of the transcription factor Nrf2, which in turn induces the expression of antioxidant response element genes. We have previously shown that DMF reacts with a wide range of protein thiols, suggesting that the complete mechanisms of action of DMF are unknown. Here, we investigated other intracellular thiol residues that may also be irreversibly modified by DMF in neurons and astrocytes. Using mass spectrometry, we identified 24 novel proteins that were modified by DMF in neurons and astrocytes, including cofilin-1, tubulin and collapsin response mediator protein 2 (CRMP2). Using an in vitro functional assay, we demonstrated that DMF-modified cofilin-1 loses its activity and generates less monomeric actin, potentially inhibiting its cytoskeletal remodeling activity, which could be beneficial in the modulation of myelination during RRMS. DMF modification of tubulin did not significantly impact axonal lysosomal trafficking. We found that the oxygen consumption rate of N1E-115 neurons and the levels of proteins related to mitochondrial energy production were only slightly affected by the highest doses of DMF, confirming that DMF treatment does not impair cellular respiratory function. In summary, our work provides new insights into the mechanisms supporting the neuroprotective and remyelination benefits associated with DMF treatment in addition to the antioxidant response by Nrf2.


Astrocytes/metabolism , Cysteine/drug effects , Dimethyl Fumarate/pharmacology , NF-E2-Related Factor 2/metabolism , Neurons/metabolism , 3T3-L1 Cells , Animals , Astrocytes/cytology , Astrocytes/drug effects , Cells, Cultured , Cofilin 1/chemistry , Cofilin 1/metabolism , Intercellular Signaling Peptides and Proteins , Mass Spectrometry , Mice , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/drug effects , Rats , Tubulin/chemistry , Tubulin/metabolism
8.
Biochim Biophys Acta Proteins Proteom ; 1865(1): 88-98, 2017 Jan.
Article En | MEDLINE | ID: mdl-27693909

Tropomyosin and cofilin are actin-binding proteins which control dynamics of actin assembly and disassembly. Tropomyosin isoforms can either inhibit or enhance cofilin activity, but the mechanism of this diverse regulation is not well understood. In this work mechanisms of actin dynamics regulation by four cytoskeletal tropomyosin isoforms and cofilin-1 were studied with the use of biochemical and fluorescent microscopy assays. The recombinant tropomyosin isoforms were products of two genes: TPM1 (Tpm1.6 and Tpm1.8) and TPM3 (Tpm3.2 and Tpm3.4). Tpm1.6/1.8 bound to F-actin with higher apparent binding constants and lower cooperativities than Tpm3.2/3.4. In consequence, subsaturating concentrations of cofilin-1 removed 50% of Tpm3.2/3.4 from F-actin. By contrast, 2 and 5.5 molar excess of cofilin-1 over actin was required to dissociate 50% of Tpm1.6/1.8. All tropomyosins inhibited the rate of spontaneous polymerization of actin, which was reversed by cofilin-1. Products of TPM1 favored longer filaments and protected them from cofilin-induced depolymerization. This was in contrast to the isoforms derived from TPM3, which facilitated depolymerization. Tpm3.4 was the only isoform, which increased frequency of the filament severing by cofilin-1. Tpm1.6/1.8 inhibited, but Tpm3.2/3.4 enhanced cofilin-induced conformational changes leading to accelerated release of rhodamine-phalloidin from the filament. We concluded that the effects were executed through different actin affinities of tropomyosin isoforms and cooperativities of tropomyosin and cofilin-1 binding. The results obtained in vitro were in good agreement with localization of tropomyosin isoforms in stable or highly dynamic filaments demonstrated before in various cells.


Actin Cytoskeleton/metabolism , Cofilin 1/metabolism , Tropomyosin/metabolism , Actin Cytoskeleton/chemistry , Amino Acid Sequence , Animals , Cofilin 1/chemistry , Cytoplasm/metabolism , Humans , Mice , Phalloidine/analogs & derivatives , Phalloidine/chemistry , Polymerization , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Rats , Rhodamines/chemistry , Tropomyosin/chemistry
9.
Mol Cell ; 62(3): 397-408, 2016 05 05.
Article En | MEDLINE | ID: mdl-27153537

Cofilin/actin-depolymerizing factor (ADF) proteins are critical nodes that relay signals from protein kinase cascades to the actin cytoskeleton, in particular through site-specific phosphorylation at residue Ser3. This is important for regulation of the roles of cofilin in severing and stabilizing actin filaments. Consequently, cofilin/ADF Ser3 phosphorylation is tightly controlled as an almost exclusive substrate for LIM kinases. Here we determine the LIMK1:cofilin-1 co-crystal structure. We find an interface that is distinct from canonical kinase-substrate interactions. We validate this previously unobserved mechanism for high-fidelity kinase-substrate recognition by in vitro kinase assays, examination of cofilin phosphorylation in mammalian cells, and functional analysis in S. cerevisiae. The interface is conserved across all LIM kinases. Remarkably, we also observe both pre- and postphosphotransfer states in the same crystal lattice. This study therefore provides a molecular understanding of how kinase-substrate recognition acts as a gatekeeper to regulate actin cytoskeletal dynamics.


Cofilin 1/metabolism , Lim Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Binding Sites , Catalytic Domain , Cofilin 1/chemistry , Cofilin 1/genetics , Crystallography, X-Ray , HEK293 Cells , Humans , Lim Kinases/chemistry , Lim Kinases/genetics , Models, Molecular , Mutation , Phosphorylation , Protein Binding , Protein Conformation , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Serine , Structure-Activity Relationship , Substrate Specificity , Transfection
10.
J Mol Biol ; 428(8): 1604-16, 2016 Apr 24.
Article En | MEDLINE | ID: mdl-26996939

Dynamic remodeling and turnover of cellular actin networks requires actin filament severing by actin-depolymerizing factor (ADF)/Cofilin proteins. Mammals express three different ADF/Cofilins (Cof1, Cof2, and ADF), and genetic studies suggest that in vivo they perform both overlapping and unique functions. To gain mechanistic insights into their different roles, we directly compared their G-actin and F-actin binding affinities, and quantified the actin filament severing activities of human Cof1, Cof2, and ADF using in vitro total internal reflection fluorescence microscopy. All three ADF/Cofilins had similar affinities for G-actin and F-actin. However, Cof2 and ADF severed filaments much more efficiently than Cof1 at both lower and higher concentrations and using either muscle or platelet actin. Furthermore, Cof2 and ADF were more effective than Cof1 in promoting "enhanced disassembly" when combined with actin disassembly co-factors Coronin-1B and actin-interacting protein 1 (AIP1), and these differences were observed on both preformed and actively growing filaments. To probe the mechanism underlying these differences, we used multi-wavelength total internal reflection fluorescence microscopy to directly observe Cy3-Cof1 and Cy3-Cof2 interacting with actin filaments in real time during severing. Cof1 and Cof2 each bound to filaments with similar kinetics, yet Cof2 induced severing much more rapidly than Cof1, decreasing the time interval between initial binding on a filament and severing at the same location. These differences in ADF/Cofilin activities and mechanisms may be used in cells to tune filament turnover rates, which can vary widely for different actin structures.


Actin Cytoskeleton/chemistry , Cofilin 1/chemistry , Cofilin 2/chemistry , Destrin/chemistry , Microscopy/methods , Actin Depolymerizing Factors/metabolism , Actins/chemistry , Animals , Dose-Response Relationship, Drug , Escherichia coli/metabolism , Humans , Nucleotides/chemistry , Plasmids/metabolism , Protein Binding , Rabbits
11.
Shock ; 42(6): 554-61, 2014 Dec.
Article En | MEDLINE | ID: mdl-25243430

The purpose of this study is to understand the role of rho-kinase (ROCK-2) in the regulation of liver microcirculation after inflammatory stress. Endothelin-1 (ET-1)-induced nitric oxide (NO) is essential in the regulation of blood flow in hepatic sinusoids. Lipopolysaccharide (LPS) inhibits this ET-1-induced NO production and disrupts liver microcirculation; however, the exact molecular mechanism is unknown. Liver sinusoidal endothelial cells were isolated, pretreated with 10 ng/mL LPS for 6 h, and treated with 10 µM Y27632 (ROCK-2 inhibitor) for 30 min and 10 nM ET-1 for 30 min. Lipopolysaccharide induced RhoA membrane translocation that was attenuated by methyl-ß-cyclodextrin (cholesterol sequester) or targeted mutation of caveolin-1. Lipopolysaccharide increased ROCK-2 expressions (+60%) and ROCK-2 activity (+36%). Endothelin-1 increased endothelial NO synthase (eNOS) activity (+70%), but LPS inhibited this ET-1-mediated eNOS response. Treatment with Y27632 restored ET-1-mediated eNOS activity (+61%) and stimulated NO production in the perinuclear region after LPS pretreatment. This treatment reduced cofilin-Ser3 phosphorylation (-73%), increased vasodilator-stimulated phosphoprotein-Ser239 phosphorylation (+88%), and stimulated globular actin/eNOS association. Lipopolysaccharide induces Rho/ROCKs signaling pathway to disrupt the ET-1-mediated eNOS activation in liver sinusoidal endothelial cells. Rho-kinase ROCK-2 inhibition restores ET-1-mediated NO production after the LPS pretreatment, in part, through an increase in actin depolymerization.


Endothelial Cells/cytology , Endothelin-1/metabolism , Liver/metabolism , Nitric Oxide Synthase Type III/metabolism , rho-Associated Kinases/metabolism , Actins/metabolism , Amides/chemistry , Animals , Caveolin 1/metabolism , Cells, Cultured , Cofilin 1/chemistry , Enzyme Activation , Inflammation , Lipopolysaccharides , Liver/blood supply , Male , Microcirculation , Nitric Oxide/metabolism , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Pyridines/chemistry , Rats , Rats, Sprague-Dawley , Sepsis , Serine/metabolism
12.
Angew Chem Int Ed Engl ; 53(41): 10904-7, 2014 Oct 06.
Article En | MEDLINE | ID: mdl-25156888

Genetically encoded, light-activatable proteins provide the means to probe biochemical pathways at specific subcellular locations with exquisite temporal control. However, engineering these systems in order to provide a dramatic jump in localized activity, while retaining a low dark-state background remains a significant challenge. When placed within the framework of a genetically encodable, light-activatable heterodimerizer system, the actin-remodelling protein cofilin induces dramatic changes in the F-actin network and consequent cell motility upon illumination. We demonstrate that the use of a partially impaired mutant of cofilin is critical for maintaining low background activity in the dark. We also show that light-directed recruitment of the reduced activity cofilin mutants to the cytoskeleton is sufficient to induce F-actin remodeling, formation of filopodia, and directed cell motility.


Light , Optogenetics , Actin Cytoskeleton/metabolism , Actins/metabolism , Amino Acid Substitution , Animals , Cell Line, Tumor , Cell Movement , Cofilin 1/chemistry , Cofilin 1/genetics , Cofilin 1/metabolism , Cryptochromes/chemistry , Cryptochromes/genetics , Cryptochromes/metabolism , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Microscopy, Confocal , Pseudopodia/metabolism , Red Fluorescent Protein
13.
J Biol Chem ; 289(37): 25699-710, 2014 Sep 12.
Article En | MEDLINE | ID: mdl-25096578

Cell death by necrosis is emerging not merely as a passive phenomenon but as a cell-regulated process. Here, by using different necrotic triggers, we prove the existence of two distinct necrotic pathways. The mitochondrial reactive oxygen species generator 2,3-dimethoxy-1,4-naphthoquinone elicits necrosis characterized by the involvement of RIP1 and Drp1. However, G5, a non-selective isopeptidase inhibitor, triggers a distinct necrotic pathway that depends on the protein phosphatase PP2A and the actin cytoskeleton. PP2A catalytic subunit is stabilized by G5 treatment, and its activity is increased. Furthermore, PP2Ac accumulates into the cytoplasm during necrosis similarly to HMGB1. We have also defined in the actin-binding protein cofilin-1 a link between PP2A, actin cytoskeleton, and necrotic death. Cofilin-1-severing/depolymerization activity is negatively regulated by phosphorylation of serine 3. PP2A contributes to the dephosphorylation of serine 3 elicited by G5. Finally, a cofilin mutant that mimics phosphorylated Ser-3 can partially rescue necrosis in response to G5.


Actin Cytoskeleton/metabolism , Cofilin 1/metabolism , Nuclear Pore Complex Proteins/metabolism , Protein Phosphatase 2/metabolism , RNA-Binding Proteins/metabolism , Actin Cytoskeleton/ultrastructure , Actin Depolymerizing Factors/chemistry , Cell Membrane Structures/chemistry , Cell Membrane Structures/drug effects , Cofilin 1/chemistry , HT29 Cells , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Necrosis/genetics , Necrosis/metabolism , Nuclear Pore Complex Proteins/chemistry , Protein Phosphatase 2/antagonists & inhibitors , Protein Phosphatase 2/genetics , Proteolysis , Pyrans/pharmacology , RNA-Binding Proteins/chemistry , Reactive Oxygen Species/metabolism , Sulfhydryl Compounds/pharmacology
14.
J Biol Chem ; 289(7): 4043-54, 2014 Feb 14.
Article En | MEDLINE | ID: mdl-24371134

Actin depolymerizing factor (ADF)/cofilins are essential regulators of actin turnover in eukaryotic cells. These multifunctional proteins facilitate both stabilization and severing of filamentous (F)-actin in a concentration-dependent manner. At high concentrations ADF/cofilins bind stably to F-actin longitudinally between two adjacent actin protomers forming what is called a decorative interaction. Low densities of ADF/cofilins, in contrast, result in the optimal severing of the filament. To date, how these two contrasting modalities are achieved by the same protein remains uncertain. Here, we define the proximate amino acids between the actin filament and the malaria parasite ADF/cofilin, PfADF1 from Plasmodium falciparum. PfADF1 is unique among ADF/cofilins in being able to sever F-actin but do so without stable filament binding. Using chemical cross-linking and mass spectrometry (XL-MS) combined with structure reconstruction we describe a previously overlooked binding interface on the actin filament targeted by PfADF1. This site is distinct from the known binding site that defines decoration. Furthermore, total internal reflection fluorescence (TIRF) microscopy imaging of single actin filaments confirms that this novel low affinity site is required for F-actin severing. Exploring beyond malaria parasites, selective blocking of the decoration site with human cofilin (HsCOF1) using cytochalasin D increases its severing rate. HsCOF1 may therefore also use a decoration-independent site for filament severing. Thus our data suggest that a second, low affinity actin-binding site may be universally used by ADF/cofilins for actin filament severing.


Destrin/chemistry , Plasmodium falciparum/chemistry , Protozoan Proteins/chemistry , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Actins/chemistry , Actins/genetics , Actins/metabolism , Binding Sites , Cofilin 1/chemistry , Cofilin 1/genetics , Cofilin 1/metabolism , Cytochalasin D/chemistry , Destrin/genetics , Destrin/metabolism , Humans , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
15.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 9): 1780-8, 2013 Sep.
Article En | MEDLINE | ID: mdl-23999301

The actin cytoskeleton is the chassis that gives a cell its shape and structure, and supplies the power for numerous dynamic processes including motility, endocytosis, intracellular transport and division. To perform these activities, the cytoskeleton undergoes constant remodelling and reorganization. One of the major actin-remodelling families are the cofilin proteins, made up of cofilin 1, cofilin 2 and actin-depolymerizing factor (ADF), which sever aged ADP-associated actin filaments to reduce filament length and provide new potential nucleation sites. Despite the significant interest in cofilin as a central node in actin-cytoskeleton dynamics, to date the only forms of cofilin for which crystal structures have been solved are from the yeast, Chromalveolata and plant kingdoms; none have previously been reported for an animal cofilin protein. Two distinct regions in animal cofilin are significantly larger than in the forms previously crystallized, suggesting that they would be uniquely organized. Therefore, it was sought to determine the structure of human cofilin 1 by X-ray crystallography to elucidate how it could interact with and regulate dynamic actin-cytoskeletal structures. Although wild-type human cofilin 1 proved to be recalcitrant, a C147A point mutant yielded crystals that diffracted to 2.8 Šresolution. These studies revealed how the actin-binding helix undergoes a conformational change that increases the number of potential hydrogen bonds available for substrate binding.


Actins/metabolism , Cofilin 1/chemistry , Actins/chemistry , Amino Acid Sequence , Cofilin 1/genetics , Cofilin 1/metabolism , Crystallography, X-Ray , Humans , Hydrogen Bonding , Molecular Sequence Data , Point Mutation , Protein Binding/genetics , Protein Conformation , Protein Structure, Secondary/genetics
16.
Biochim Biophys Acta ; 1834(10): 2010-9, 2013 Oct.
Article En | MEDLINE | ID: mdl-23845993

The main goal of the work was to uncover the dynamical changes in actin induced by the binding of cofilin and profilin. The change in the structure and flexibility of the small domain and its function in the thermodynamic stability of the actin monomer were examined with fluorescence spectroscopy and differential scanning calorimetry (DSC). The structure around the C-terminus of actin is slightly affected by the presence of cofilin and profilin. Temperature dependent fluorescence resonance energy transfer measurements indicated that both actin binding proteins decreased the flexibility of the protein matrix between the subdomains 1 and 2. Time resolved anisotropy decay measurements supported the idea that cofilin and profilin changed similarly the dynamics around the fluorescently labeled Cys-374 and Lys-61 residues in subdomains 1 and 2, respectively. DSC experiments indicated that the thermodynamic stability of actin increased by cofilin and decreased in the presence of profilin. Based on the information obtained it is possible to conclude that while the small domain of actin acts uniformly in the presence of cofilin and profilin the overall stability of actin changes differently in the presence of the studied actin binding proteins. The results support the idea that the small domain of actin behaves as a rigid unit during the opening and closing of the nucleotide binding pocket in the presence of profilin and cofilin as well. The structural arrangement of the nucleotide binding cleft mainly influences the global stability of actin while the dynamics of the different segments can change autonomously.


Actins/chemistry , Adenosine Triphosphate/chemistry , Cofilin 1/chemistry , Profilins/chemistry , Actins/isolation & purification , Animals , Calorimetry, Differential Scanning , Cofilin 1/genetics , Escherichia coli/genetics , Kinetics , Mice , Molecular Dynamics Simulation , Muscle, Skeletal/chemistry , Profilins/genetics , Protein Binding , Protein Structure, Tertiary , Rabbits , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Spectrometry, Fluorescence , Thermodynamics
17.
Int J Biochem Cell Biol ; 45(8): 1720-9, 2013 Aug.
Article En | MEDLINE | ID: mdl-23702034

The role of cytoskeleton-associated proteins during TNF-induced apoptosis is not fully understood. A potential candidate kinase that might connect TNF signaling to actin reorganization is the death-associated protein kinase (DAPK). To identify new DAPK interaction partners in TNF-induced apoptosis, we performed a peptide array screen. We show that TNF-treatment enhanced the phosphorylation of LIMK at threonine508 and its downstream target cofilin at serine3 (p-cofilin(Ser3)). Modulation of DAPK activity and expression by DAPK inhibitor treatment, siRNA knockdown, and overexpression affected the phosphorylation of both proteins. We propose a 3D structural model where DAPK functions as a scaffold for the LIMK/cofilin complex and triggers a closer interaction of both proteins under TNF stimulation. Upon TNF a striking redistribution of LIMK, DAPK, and cofilin to the perinuclear compartment was observed. The pro-apoptotic DAPK/LIMK/cofilin multiprotein complex was abrogated in detached cells, indicating that its signaling was no longer needed if cells committed to apoptosis. P-cofilin(Ser3) was strongly accumulated in cells with condensed chromatin, pronounced membrane blebs and Annexin V up-regulation. From studying different cofilin(Ser3) mutants we suggest that p-cofilin(Ser3) is an indicator of TNF-induced apoptosis. Collectively, our findings identify a novel molecular cytoskeleton-associated mechanism in TNF-induced DAPK-dependent apoptosis.


Apoptosis/drug effects , Cofilin 1/metabolism , Death-Associated Protein Kinases/metabolism , Lim Kinases/metabolism , Multiprotein Complexes/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Amino Acid Sequence , Cell Adhesion/drug effects , Cofilin 1/chemistry , HCT116 Cells , Humans , Models, Biological , Models, Molecular , Molecular Sequence Data , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Transport/drug effects , Signal Transduction/drug effects
18.
Mol Biosyst ; 9(3): 447-56, 2013 Mar.
Article En | MEDLINE | ID: mdl-23340498

Doxorubicin is an anticancer drug used in a wide range of cancer therapies, yet some side effects have been reported. One of these is cardiotoxicity, including cardiomyopathy and ultimately congestive heart failure. This damage to the heart has been shown to result from doxorubicin-induced reactive oxygen species. However, the cellular targets of doxorubicin-induced oxidative damage on cardiomyocytes are largely unknown. For this, a cysteine-labeling-based two-dimensional difference gel electrophoresis (2D-DIGE) combined with MALDI-TOF/TOF mass spectrometry (MALDI-TOF/TOF MS) were employed to analyze the impact of doxorubicin treatment on the redox regulation in rat cardiomyocytes. This study demonstrated 25 unique protein features that had significantly changed in their thiol reactivity and revealed that doxorubicin-induced cardiotoxicity involves dysregulation of protein folding, translational regulation and cytoskeleton regulation. Our work shows that this combined proteomic strategy provides a rapid method to study the molecular mechanisms of doxorubicin-induced cytotoxicity in the heart. The identified targets may be useful for further evaluation as potential cardiotoxic biomarkers during damage to the heart induced by doxorubicin, as well as possible diagnostic or therapeutic applications.


Antibiotics, Antineoplastic/toxicity , Cysteine/metabolism , Doxorubicin/toxicity , Myocytes, Cardiac/metabolism , Proteome/metabolism , Amino Acid Sequence , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Cell Line , Cell Shape/drug effects , Cell Survival/drug effects , Cofilin 1/chemistry , Cofilin 1/metabolism , Cytoskeleton/drug effects , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/metabolism , Metabolic Networks and Pathways , Molecular Sequence Data , Myocytes, Cardiac/drug effects , NM23 Nucleoside Diphosphate Kinases/chemistry , NM23 Nucleoside Diphosphate Kinases/metabolism , Oxidation-Reduction , Peptide Fragments/chemistry , Protein Processing, Post-Translational , Proteome/chemistry , Rats , Reactive Oxygen Species/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Two-Dimensional Difference Gel Electrophoresis
19.
Mol Biol Cell ; 24(1): 31-41, 2013 Jan.
Article En | MEDLINE | ID: mdl-23135996

Actin filament severing is critical for the dynamic turnover of cellular actin networks. Cofilin severs filaments, but additional factors may be required to increase severing efficiency in vivo. Srv2/cyclase-associated protein (CAP) is a widely expressed protein with a role in binding and recycling actin monomers ascribed to domains in its C-terminus (C-Srv2). In this paper, we report a new biochemical and cellular function for Srv2/CAP in directly catalyzing cofilin-mediated severing of filaments. This function is mediated by its N-terminal half (N-Srv2), and is physically and genetically separable from C-Srv2 activities. Using dual-color total internal reflection fluorescence microscopy, we determined that N-Srv2 stimulates filament disassembly by increasing the frequency of cofilin-mediated severing without affecting cofilin binding to filaments. Structural analysis shows that N-Srv2 forms novel hexameric star-shaped structures, and disrupting oligomerization impairs N-Srv2 activities and in vivo function. Further, genetic analysis shows that the combined activities of N-Srv2 and Aip1 are essential in vivo. These observations define a novel mechanism by which the combined activities of cofilin and Srv2/CAP lead to enhanced filament severing and support an emerging view that actin disassembly is controlled not by cofilin alone, but by a more complex set of factors working in concert.


Actin Cytoskeleton/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cofilin 1/metabolism , Cytoskeletal Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/ultrastructure , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Carbocyanines/chemistry , Catalysis , Cofilin 1/chemistry , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Microscopy, Electron , Microscopy, Fluorescence/methods , Models, Molecular , Mutation , Protein Binding , Protein Multimerization , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics
20.
J Cell Sci ; 125(Pt 17): 3977-88, 2012 Sep 01.
Article En | MEDLINE | ID: mdl-22623727

Cofilin protein is involved in regulating the actin cytoskeleton during typical steady state conditions, as well as during cell stress conditions where cofilin saturates F-actin, forming cofilin-actin rods. Cofilin can enter the nucleus through an active nuclear localization signal (NLS), accumulating in nuclear actin rods during stress. Here, we characterize the active nuclear export of cofilin through a leptomycin-B-sensitive, CRM1-dependent, nuclear export signal (NES). We also redefine the NLS of cofilin as a bipartite NLS, with an additional basic epitope required for nuclear localization. Using fluorescence lifetime imaging microscopy (FLIM) and Förster resonant energy transfer (FRET) between cofilin moieties and actin, as well as automated image analysis in live cells, we have defined subtle mutations in the cofilin NLS that allow cofilin to bind actin in vivo and affect cofilin dynamics during stress. We further define the requirement of cofilin-actin rod formation in a system of cell stress by temporal live-cell imaging. We propose that cofilin nuclear shuttling is critical for the cofilin-actin rod stress response with cofilin dynamically communicating between the nucleus and cytoplasm during cell stress.


Actins/metabolism , Cell Nucleus/metabolism , Cofilin 1/metabolism , Stress, Physiological , Amino Acid Sequence , Animals , Cell Line , Cofilin 1/chemistry , Conserved Sequence , Gene Knockdown Techniques , Humans , Karyopherins/metabolism , Mice , Molecular Sequence Data , Mutant Proteins/metabolism , Mutation , Nuclear Export Signals , Nuclear Localization Signals/chemistry , Nuclear Localization Signals/metabolism , Phenotype , Protein Binding , Protein Transport , Receptors, Cytoplasmic and Nuclear/metabolism , Exportin 1 Protein
...