Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
1.
J Phys Chem B ; 128(19): 4590-4601, 2024 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-38701111

RESUMO

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.


Assuntos
Citoesqueleto de Actina , Dissulfetos , Simulação de Dinâmica Molecular , Dissulfetos/química , Humanos , Citoesqueleto de Actina/química , Citoesqueleto de Actina/metabolismo , Cofilina 1/química , Cofilina 1/metabolismo , Multimerização Proteica , Actinas/química , Actinas/metabolismo , Fatores de Despolimerização de Actina/química , Fatores de Despolimerização de Actina/metabolismo , Termodinâmica
2.
Chem Commun (Camb) ; 60(37): 4910-4913, 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38623638

RESUMO

Several natural cytotoxic C2-symmetric bis-lactones, such as swinholide A and rhizopodin, sequester actin dimer from the actin network and potently inhibit actin dynamics. To develop new protein-protein interaction (PPI) modulators, we synthesized structurally simplified actin-binding side-chain dimers of antitumor macrolide aplyronine A. By fixing the two side-chains closer than those of rhizopodin, the C4 linker analog depolymerized filamentous actin more potently than natural aplyronines. Cross-link experiments revealed that actin dimer was formed by treatment with the C4 linker analog. Molecular dynamics simulations showed that this analog significantly changed the interaction and spatial arrangement of the two actins compared to those in rhizopodin to provide a highly distorted and twisted orientation in the complex. Our study may promote the development of PPI-based anticancer and other drug leads related to cytoskeletal dynamics.


Assuntos
Actinas , Macrolídeos , Multimerização Proteica , Fatores de Despolimerização de Actina/química , Fatores de Despolimerização de Actina/farmacologia , Actinas/metabolismo , Actinas/química , Antineoplásicos/farmacologia , Antineoplásicos/química , Antineoplásicos/síntese química , Dimerização , Macrolídeos/química , Macrolídeos/farmacologia , Macrolídeos/síntese química , Simulação de Dinâmica Molecular , Multimerização Proteica/efeitos dos fármacos
3.
J Mol Biol ; 435(22): 168295, 2023 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-37783285

RESUMO

A variety of biological roles of mechanical forces have been proposed in cell biology, such as cell signaling pathways for survival, development, growth, and differentiation. Mechanical forces alter the mechanical conditions within cells and their environment, which strongly influences the reorganization of the actin cytoskeleton. Single-molecule imaging studies of actin filaments have led to the hypothesis that the actin filament acts as a mechanosensor; e.g., increases in actin filament tension alter their conformation and affinity for regulatory proteins. However, our understanding of the molecular mechanisms underlying how tension modulates the mechanical behavior of a single actin filament is still incomplete. In this study, a direct measurement of the twisting and bending of a fluorescently labeled single actin filament under different tension levels by force application (0.8-3.4 pN) was performed using single-molecule fluorescence polarization (SMFP) microscopy. The results showed that the amplitude of twisting and bending fluctuations of a single actin filament decreased with increasing tension. Electron micrograph analysis of tensed filaments also revealed that the fluctuations in the crossover length of actin filaments decreased with increasing filament tension. Possible molecular mechanisms underlying these results involving the binding of actin-binding proteins, such as cofilin, to the filament are discussed.


Assuntos
Citoesqueleto de Actina , Estresse Mecânico , Citoesqueleto de Actina/química , Fatores de Despolimerização de Actina/química , Imagem Individual de Molécula , Resistência à Tração , Torção Mecânica
4.
PLoS Comput Biol ; 17(11): e1009171, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34843456

RESUMO

Predictive approaches such as virtual screening have been used in drug discovery with the objective of reducing developmental time and costs. Current machine learning and network-based approaches have issues related to generalization, usability, or model interpretability, especially due to the complexity of target proteins' structure/function, and bias in system training datasets. Here, we propose a new method "DRUIDom" (DRUg Interacting Domain prediction) to identify bio-interactions between drug candidate compounds and targets by utilizing the domain modularity of proteins, to overcome problems associated with current approaches. DRUIDom is composed of two methodological steps. First, ligands/compounds are statistically mapped to structural domains of their target proteins, with the aim of identifying their interactions. As such, other proteins containing the same mapped domain or domain pair become new candidate targets for the corresponding compounds. Next, a million-scale dataset of small molecule compounds, including those mapped to domains in the previous step, are clustered based on their molecular similarities, and their domain associations are propagated to other compounds within the same clusters. Experimentally verified bioactivity data points, obtained from public databases, are meticulously filtered to construct datasets of active/interacting and inactive/non-interacting drug/compound-target pairs (~2.9M data points), and used as training data for calculating parameters of compound-domain mappings, which led to 27,032 high-confidence associations between 250 domains and 8,165 compounds, and a finalized output of ~5 million new compound-protein interactions. DRUIDom is experimentally validated by syntheses and bioactivity analyses of compounds predicted to target LIM-kinase proteins, which play critical roles in the regulation of cell motility, cell cycle progression, and differentiation through actin filament dynamics. We showed that LIMK-inhibitor-2 and its derivatives significantly block the cancer cell migration through inhibition of LIMK phosphorylation and the downstream protein cofilin. One of the derivative compounds (LIMKi-2d) was identified as a promising candidate due to its action on resistant Mahlavu liver cancer cells. The results demonstrated that DRUIDom can be exploited to identify drug candidate compounds for intended targets and to predict new target proteins based on the defined compound-domain relationships. Datasets, results, and the source code of DRUIDom are fully-available at: https://github.com/cansyl/DRUIDom.


Assuntos
Quinases Lim/antagonistas & inibidores , Quinases Lim/química , Fatores de Despolimerização de Actina/química , Fatores de Despolimerização de Actina/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Biologia Computacional , Simulação por Computador , Desenvolvimento de Medicamentos , Descoberta de Drogas , Avaliação Pré-Clínica de Medicamentos , Interações Medicamentosas , Humanos , Técnicas In Vitro , Ligantes , Quinases Lim/metabolismo , Aprendizado de Máquina , Simulação de Acoplamento Molecular , Invasividade Neoplásica/prevenção & controle , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Farmacologia em Rede/estatística & dados numéricos , Fosforilação/efeitos dos fármacos , Domínios Proteicos , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Interface Usuário-Computador
5.
Cells ; 10(10)2021 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-34685706

RESUMO

Proteins of the actin depolymerizing factor (ADF)/cofilin family are ubiquitous among eukaryotes and are essential regulators of actin dynamics and function. Mammalian neurons express cofilin-1 as the major isoform, but ADF and cofilin-2 are also expressed. All isoforms bind preferentially and cooperatively along ADP-subunits in F-actin, affecting the filament helical rotation, and when either alone or when enhanced by other proteins, promotes filament severing and subunit turnover. Although self-regulating cofilin-mediated actin dynamics can drive motility without post-translational regulation, cells utilize many mechanisms to locally control cofilin, including cooperation/competition with other proteins. Newly identified post-translational modifications function with or are independent from the well-established phosphorylation of serine 3 and provide unexplored avenues for isoform specific regulation. Cofilin modulates actin transport and function in the nucleus as well as actin organization associated with mitochondrial fission and mitophagy. Under neuronal stress conditions, cofilin-saturated F-actin fragments can undergo oxidative cross-linking and bundle together to form cofilin-actin rods. Rods form in abundance within neurons around brain ischemic lesions and can be rapidly induced in neurites of most hippocampal and cortical neurons through energy depletion or glutamate-induced excitotoxicity. In ~20% of rodent hippocampal neurons, rods form more slowly in a receptor-mediated process triggered by factors intimately connected to disease-related dementias, e.g., amyloid-ß in Alzheimer's disease. This rod-inducing pathway requires a cellular prion protein, NADPH oxidase, and G-protein coupled receptors, e.g., CXCR4 and CCR5. Here, we will review many aspects of cofilin regulation and its contribution to synaptic loss and pathology of neurodegenerative diseases.


Assuntos
Fatores de Despolimerização de Actina/metabolismo , Actinas/metabolismo , Degeneração Neural/patologia , Neurônios/metabolismo , Fatores de Despolimerização de Actina/química , Sequência de Aminoácidos , Animais , Humanos , Neuritos/metabolismo , Neurogênese
6.
J Mol Biol ; 433(7): 166833, 2021 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-33524412

RESUMO

Cofilin is an essential actin filament severing protein that accelerates the assembly dynamics and turnover of actin networks by increasing the number of filament ends where subunits add and dissociate. It binds filament subunits stoichiometrically and cooperatively, forming clusters of contiguously-bound cofilin at sub-saturating occupancies. Filaments partially occupied with cofilin sever at boundaries between bare and cofilin-decorated segments. Imaging studies concluded that bound clusters must reach a critical size (Cc) of 13-100 cofilins to sever filaments. In contrast, structural and modeling studies suggest that a few or even a single cofilin can sever filaments, possibly with different severing rate constants. How clusters grow through the cooperative incorporation of additional cofilin molecules, specifically if they elongate asymmetrically or uniformly from both ends and if they are modulated by filament shape and external force, also lacks consensus. Here, using hydrodynamic flow to visualize individual actin filaments with TIRF microscopy, we found that neither flow-induced filament bending, tension, nor surface attachment conditions substantially affected the kinetics of cofilin binding to actin filaments. Clusters of bound cofilin preferentially extended toward filament pointed ends and displayed severing competency at small sizes (Cc < 3), with no detectable severing dependence on cluster size. These data support models in which small clusters of cofilin introduce local, but asymmetric, structural changes in actin filaments that promote filament severing with a rate constant that depends weakly on the size of the cluster.


Assuntos
Citoesqueleto de Actina/ultraestrutura , Fatores de Despolimerização de Actina/ultraestrutura , Actinas/ultraestrutura , Citoesqueleto/ultraestrutura , Citoesqueleto de Actina/química , Citoesqueleto de Actina/genética , Fatores de Despolimerização de Actina/química , Fatores de Despolimerização de Actina/genética , Actinas/química , Actinas/genética , Fenômenos Biofísicos , Microscopia Crioeletrônica , Humanos , Cinética , Ligação Proteica/genética
7.
J Biol Chem ; 296: 100337, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33508320

RESUMO

Members of the ADF/cofilin family of regulatory proteins bind actin filaments cooperatively, locally change actin subunit conformation and orientation, and sever filaments at "boundaries" between bare and cofilin-occupied segments. A cluster of bound cofilin introduces two distinct classes of boundaries due to the intrinsic polarity of actin filaments, one at the "pointed" end side and the other at the "barbed" end-side of the cluster; severing occurs more readily at the pointed end side of the cluster ("fast-severing" boundary) than the barbed end side ("slow-severing" boundary). A recent electron-cryomicroscopy (cryo-EM) model of the slow-severing boundary revealed structural "defects" at the interface that potentially contribute to severing. However, the structure of the fast-severing boundary remains uncertain. Here, we use extensive molecular dynamics simulations to produce atomic resolution models of both severing boundaries. Our equilibrated simulation model of the slow-severing boundary is consistent with the cryo-EM structural model. Simulations indicate that actin subunits at both boundaries adopt structures intermediate between those of bare and cofilin-bound actin subunits. These "intermediate" states have compromised intersubunit contacts, but those at the slow-severing boundary are stabilized by cofilin bridging interactions, accounting for its lower fragmentation probability. Simulations where cofilin proteins are removed from cofilactin filaments favor a mechanism in which a cluster of two contiguously bound cofilins is needed to fully stabilize the cofilactin conformation, promote cooperative binding interactions, and accelerate filament severing. Together, these studies provide a molecular-scale foundation for developing coarse-grained and theoretical descriptions of cofilin-mediated actin filament severing.


Assuntos
Fatores de Despolimerização de Actina/metabolismo , Actinas/metabolismo , Citoesqueleto de Actina/química , Citoesqueleto de Actina/metabolismo , Fatores de Despolimerização de Actina/química , Actinas/química , Animais , Galinhas , Humanos , Simulação de Dinâmica Molecular , Conformação Proteica , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Coelhos
8.
Open Biol ; 10(9): 200157, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32873155

RESUMO

The actin cytoskeleton has the particularity of being assembled into many functionally distinct filamentous networks from a common reservoir of monomeric actin. Each of these networks has its own geometrical, dynamical and mechanical properties, because they are capable of recruiting specific families of actin-binding proteins (ABPs), while excluding the others. This review discusses our current understanding of the underlying molecular mechanisms that cells have developed over the course of evolution to segregate ABPs to appropriate actin networks. Segregation of ABPs requires the ability to distinguish actin networks as different substrates for ABPs, which is regulated in three different ways: (1) by the geometrical organization of actin filaments within networks, which promotes or inhibits the accumulation of ABPs; (2) by the identity of the networks' filaments, which results from the decoration of actin filaments with additional proteins such as tropomyosin, from the use of different actin isoforms or from covalent modifications of actin; (3) by the existence of collaborative or competitive binding to actin filaments between two or multiple ABPs. This review highlights that all these effects need to be taken into account to understand the proper localization of ABPs in cells, and discusses what remains to be understood in this field of research.


Assuntos
Citoesqueleto de Actina/metabolismo , Citoesqueleto/metabolismo , Proteínas dos Microfilamentos/metabolismo , Citoesqueleto de Actina/química , Citoesqueleto de Actina/genética , Fatores de Despolimerização de Actina/química , Fatores de Despolimerização de Actina/metabolismo , Sequência de Aminoácidos , Animais , Fenômenos Bioquímicos , Proteínas de Transporte/metabolismo , Citoesqueleto/química , Humanos , Proteínas dos Microfilamentos/química , Filogenia , Ligação Proteica , Isoformas de Proteínas , Processamento de Proteína Pós-Traducional , Transporte Proteico , Tropomiosina/química , Tropomiosina/genética , Tropomiosina/metabolismo
9.
Proc Natl Acad Sci U S A ; 117(33): 19904-19913, 2020 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-32747565

RESUMO

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.


Assuntos
Fatores de Despolimerização de Actina/metabolismo , Actinas/metabolismo , Archaea/metabolismo , Proteínas Arqueais/metabolismo , Gelsolina/metabolismo , Fatores de Despolimerização de Actina/química , Fatores de Despolimerização de Actina/genética , Actinas/química , Actinas/genética , Sequência de Aminoácidos , Archaea/química , Archaea/genética , Proteínas Arqueais/química , Proteínas Arqueais/genética , Citoesqueleto/química , Citoesqueleto/genética , Citoesqueleto/metabolismo , Evolução Molecular , Gelsolina/química , Gelsolina/genética , Genoma Arqueal , Polimerização , Conformação Proteica em alfa-Hélice , Alinhamento de Sequência
10.
J Biol Chem ; 295(38): 13299-13313, 2020 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-32723865

RESUMO

Cofilin is an actin filament severing protein necessary for fast actin turnover dynamics. Coronin and Aip1 promote cofilin-mediated actin filament disassembly, but the mechanism is somewhat controversial. An early model proposed that the combination of cofilin, coronin, and Aip1 disassembled filaments in bursts. A subsequent study only reported severing. Here, we used EM to show that actin filaments convert directly into globular material. A monomer trap assay also shows that the combination of all three factors produces actin monomers faster than any two factors alone. We show that coronin accelerates the release of Pi from actin filaments and promotes highly cooperative cofilin binding to actin to create long stretches of polymer with a hypertwisted morphology. Aip1 attacks these hypertwisted regions along their sides, disintegrating them into monomers or short oligomers. The results are consistent with a catastrophic mode of disassembly, not enhanced severing alone.


Assuntos
4-Butirolactona/análogos & derivados , Citoesqueleto de Actina/química , Fatores de Despolimerização de Actina/química , Proteínas dos Microfilamentos/química , 4-Butirolactona/química , Citoesqueleto de Actina/ultraestrutura , Humanos
11.
Proc Natl Acad Sci U S A ; 117(3): 1478-1484, 2020 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-31900364

RESUMO

Members of the cofilin/ADF family of proteins sever actin filaments, increasing the number of filament ends available for polymerization or depolymerization. Cofilin binds actin filaments with positive cooperativity, forming clusters of contiguously bound cofilin along the filament lattice. Filament severing occurs preferentially at boundaries between bare and cofilin-decorated (cofilactin) segments and is biased at 1 side of a cluster. A molecular understanding of cooperative binding and filament severing has been impeded by a lack of structural data describing boundaries. Here, we apply methods for analyzing filament cryo-electron microscopy (cryo-EM) data at the single subunit level to directly investigate the structure of boundaries within partially decorated cofilactin filaments. Subnanometer resolution maps of isolated, bound cofilin molecules and an actin-cofilactin boundary indicate that cofilin-induced actin conformational changes are local and limited to subunits directly contacting bound cofilin. An isolated, bound cofilin compromises longitudinal filament contacts of 1 protofilament, consistent with a single cofilin having filament-severing activity. An individual, bound phosphomimetic (S3D) cofilin with weak severing activity adopts a unique binding mode that does not perturb actin structure. Cofilin clusters disrupt both protofilaments, consistent with a higher severing activity at boundaries compared to single cofilin. Comparison of these structures indicates that this disruption is substantially greater at pointed end sides of cofilactin clusters than at the barbed end. These structures, with the distribution of bound cofilin clusters, suggest that maximum binding cooperativity is achieved when 2 cofilins occupy adjacent sites. These results reveal the structural origins of cooperative cofilin binding and actin filament severing.


Assuntos
Citoesqueleto de Actina/química , Fatores de Despolimerização de Actina/química , Citoesqueleto de Actina/metabolismo , Fatores de Despolimerização de Actina/metabolismo , Animais , Sítios de Ligação , Microscopia Crioeletrônica , Humanos , Fosforilação , Ligação Proteica , Coelhos
12.
Soft Matter ; 16(8): 2017-2024, 2020 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-31996875

RESUMO

The fracture and severing of polymer chains plays a critical role in the failure of fibrous materials and the regulated turnover of intracellular filaments. Using continuum wormlike chain models, we investigate the fracture of semiflexible polymers via thermal bending fluctuations, focusing on the role of filament flexibility and dynamics. Our results highlight a previously unappreciated consequence of mechanical heterogeneity in the filament, which enhances the rate of thermal fragmentation particularly in cases where constraints hinder the movement of the chain ends. Although generally applicable to semiflexible chains with regions of different bending stiffness, the model is motivated by a specific biophysical system: the enhanced severing of actin filaments at the boundary between stiff bare regions and mechanically softened regions that are coated with cofilin regulatory proteins. The results presented here point to a potential mechanism for disassembly of polymeric materials in general and cytoskeletal actin networks in particular by the introduction of locally softened chain regions, as occurs with cofilin binding.


Assuntos
Polímeros/química , Citoesqueleto de Actina/química , Fatores de Despolimerização de Actina/química , Fenômenos Biofísicos , Temperatura Alta , Cinética , Modelos Moleculares
13.
Nat Chem Biol ; 15(12): 1183-1190, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31740825

RESUMO

Here we introduce Z-lock, an optogenetic approach for reversible, light-controlled steric inhibition of protein active sites. The light oxygen voltage (LOV) domain and Zdk, a small protein that binds LOV selectively in the dark, are appended to the protein of interest where they sterically block the active site. Irradiation causes LOV to change conformation and release Zdk, exposing the active site. Computer-assisted protein design was used to optimize linkers and Zdk-LOV affinity, for both effective binding in the dark, and effective light-induced release of the intramolecular interaction. Z-lock cofilin was shown to have actin severing ability in vitro, and in living cancer cells it produced protrusions and invadopodia. An active fragment of the tubulin acetylase αTAT was similarly modified and shown to acetylate tubulin on irradiation.


Assuntos
Acetilesterase/química , Fatores de Despolimerização de Actina/química , Optogenética , Tubulina (Proteína)/química , Acetilação
14.
Biochemistry ; 58(1): 40-47, 2019 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-30499293

RESUMO

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.


Assuntos
Citoesqueleto de Actina/metabolismo , Fatores de Despolimerização de Actina/metabolismo , Actinas/metabolismo , Cofilina 1/metabolismo , Citoesqueleto de Actina/química , Fatores de Despolimerização de Actina/química , Actinas/química , Animais , Cofilina 1/química , Humanos , Concentração de Íons de Hidrogênio , Camundongos , Coelhos
15.
PLoS One ; 13(12): e0208979, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30550596

RESUMO

LIM kinases are located at a strategic crossroad, downstream of several signaling pathways and upstream of effectors such as microtubules and the actin cytoskeleton. Cofilin is the only LIM kinases substrate that is well described to date, and its phosphorylation on serine 3 by LIM kinases controls cofilin actin-severing activity. Consequently, LIM kinases inhibition leads to actin cytoskeleton disorganization and blockade of cell motility, which makes this strategy attractive in anticancer treatments. LIMK has also been reported to be involved in pathways that are deregulated in hematologic malignancies, with little information regarding cofilin phosphorylation status. We have used proteomic approaches to investigate quantitatively and in detail the phosphorylation status of cofilin in myeloid tumor cell lines of murine and human origin. Our results show that under standard conditions, only a small fraction (10 to 30% depending on the cell line) of cofilin is phosphorylated (including serine 3 phosphorylation). In addition, after a pharmacological inhibition of LIM kinases, a residual cofilin phosphorylation is observed on serine 3. Interestingly, this 2D gel based proteomic study identified new phosphorylation sites on cofilin, such as threonine 63, tyrosine 82 and serine 108.


Assuntos
Fatores de Despolimerização de Actina/metabolismo , Carbazóis/farmacologia , Quinases Lim/antagonistas & inibidores , Células Mieloides/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteômica , Fatores de Despolimerização de Actina/química , Actinas/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Linhagem Celular , Humanos , Células Mieloides/efeitos dos fármacos , Fosforilação/efeitos dos fármacos
16.
PLoS Biol ; 16(7): e2005380, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29985916

RESUMO

Cofilin is an essential actin remodeling protein promoting depolymerization and severing of actin filaments. To address the relevance of cofilin for the development and function of T cells in vivo, we generated knock-in mice in which T-cell-specific nonfunctional (nf) cofilin was expressed instead of wild-type (WT) cofilin. Nf cofilin mice lacked peripheral αß T cells and showed a severe thymus atrophy. This was caused by an early developmental arrest of thymocytes at the double negative (DN) stage. Importantly, even though DN thymocytes expressed the TCRß chain intracellularly, they completely lacked TCRß surface expression. In contrast, nf cofilin mice possessed normal numbers of γδ T cells. Their functionality was confirmed in the γδ T-cell-driven, imiquimod (IMQ)-induced, psoriasis-like murine model. Overall, this study not only highlights the importance of cofilin for early αß T-cell development but also shows for the first time that an actin-binding protein is differentially involved in αß versus γδ T-cell development.


Assuntos
Fatores de Despolimerização de Actina/metabolismo , Actinas/metabolismo , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Timo/metabolismo , Fatores de Despolimerização de Actina/química , Animais , Movimento Celular , Técnicas de Introdução de Genes , Humanos , Células Jurkat , Camundongos , Mutação/genética , Prolina/metabolismo , Subpopulações de Linfócitos T/metabolismo , Linfócitos T/metabolismo , Timócitos/metabolismo
17.
Proc Natl Acad Sci U S A ; 115(22): E5000-E5007, 2018 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-29760064

RESUMO

The complex interplay between actin regulatory proteins facilitates the formation of diverse cellular actin structures. Formin homology proteins (formins) play an essential role in the formation of actin stress fibers and yeast actin cables, to which the major actin depolymerizing factor cofilin barely associates. In vitro, F-actin decorated with cofilin exhibits a marked increase in the filament twist. On the other hand, a mammalian formin mDia1 rotates along the long-pitch actin helix during processive actin elongation (helical rotation). Helical rotation may impose torsional force on F-actin in the opposite direction of the cofilin-induced twisting. Here, we show that helical rotation of mDia1 converts F-actin resistant to cofilin both in vivo and in vitro. F-actin assembled by mDia1 without rotational freedom became more resistant to the severing and binding activities of cofilin than freely rotatable F-actin. Electron micrographic analysis revealed untwisting of the long-pitch helix of F-actin elongating from mDia1 on tethering of both mDia1 and the pointed end side of the filament. In cells, single molecules of mDia1ΔC63, an activated mutant containing N-terminal regulatory domains, showed tethering to cell structures more frequently than autoinhibited wild-type mDia1 and mDia1 devoid of N-terminal domains. Overexpression of mDia1ΔC63 induced the formation of F-actin, which has prolonged lifetime and accelerates dissociation of cofilin. Helical rotation of formins may thus serve as an F-actin stabilizing mechanism by which a barbed end-bound molecule can enhance the stability of a filament over a long range.


Assuntos
Citoesqueleto de Actina/metabolismo , Fatores de Despolimerização de Actina/metabolismo , Proteínas de Transporte/metabolismo , Proteínas Fetais/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas Nucleares/metabolismo , Citoesqueleto de Actina/química , Fatores de Despolimerização de Actina/química , Animais , Proteínas de Transporte/química , Proteínas de Transporte/genética , Linhagem Celular , Proteínas Fetais/química , Forminas , Camundongos , Proteínas dos Microfilamentos/química , Mutação , Proteínas Nucleares/química , Conformação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Rotação , Xenopus laevis
18.
J Biol Chem ; 293(15): 5377-5383, 2018 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-29463680

RESUMO

Cofilin/ADF proteins are actin-remodeling proteins, essential for actin disassembly in various cellular processes, including cell division, intracellular transport, and motility. Cofilins bind actin filaments cooperatively and sever them preferentially at boundaries between bare and cofilin-decorated (cofilactin) segments. The cooperative binding to actin has been proposed to originate from conformational changes that propagate allosterically from clusters of bound cofilin to bare actin segments. Estimates of the lengths over which these cooperative conformational changes propagate vary dramatically, ranging from 2 to >100 subunits. Here, we present a general, structure-based method for detecting from cryo-EM micrographs small variations in filament geometry (i.e. twist) with single-subunit precision. How these variations correlate with regulatory protein occupancy reveals how far allosteric, conformational changes propagate along filaments. We used this method to determine the effects of cofilin on the actin filament twist. Our results indicate that cofilin-induced changes in filament twist propagate only 1-2 subunits from the boundary into the bare actin segment, independently of the boundary polarity (i.e. irrespective of whether or not the bare actin segment flanks the pointed or barbed-end side of the boundary) and the pyrene fluorophore labeling of actin. These observations indicate that the filament twist changes abruptly at boundaries between bare and cofilin-decorated segments, thereby constraining mechanistic models of cooperative actin filament interactions and severing by cofilin. The methods presented here extend the capability of cryo-EM to analyze biologically relevant deviations from helical symmetry in actin as well as other classes of linear polymers.


Assuntos
Citoesqueleto de Actina/química , Citoesqueleto de Actina/ultraestrutura , Fatores de Despolimerização de Actina/química , Animais , Microscopia Crioeletrônica , Estrutura Quaternária de Proteína , Coelhos
19.
Poult Sci ; 97(2): 684-694, 2018 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-29121270

RESUMO

In order to study the tenderization mechanism of ATP treatments by depolymerizing actin filaments, breast muscles of Eastern Zhejiang White Geese were randomly divided into 3 groups: control, 10 and 20 mM groups. Shear force (SF), sarcomere length (SL) and myofibrillar fraction index (MFI), the content of F-actin and G-actin, the expression of actin associated proteins (cofilins and tropomodulins) were investigated during conditioning. In 20 mM group, cofilins content increased from 48 to 168 h, while tropomodulins decreased; the content of F-actin decreased from 24 to 168 h, while the increased G-actin was observed upto 48 h. In the control, the degraded tropomodulins were observed at 168 h, and the increased cofilins and G-actin were detected at the same time; the increase of MFI and decrease of F-actin content were shown at 96 and 168 h. Compared to control group, 20 mM group accelerated the transformation of F-actin into G-actin; it showed higher SL and MFI, and lower SF at 48, 96 and 168 h, respectively. We concluded that depolymerization of actin filaments, which was regulated by cofilins and tropomodulins, contributed to myofibrillar fraction and low SF during conditioning.


Assuntos
Citoesqueleto de Actina/química , Trifosfato de Adenosina/química , Gansos , Carne/análise , Músculo Esquelético/química , Sarcômeros/fisiologia , Fatores de Despolimerização de Actina/química , Actinas/química , Animais , Masculino , Resistência ao Cisalhamento , Tropomodulina/química
20.
Immunobiology ; 223(2): 246-251, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29102047

RESUMO

House dust mite (HDM) allergen is a major cause of allergic disease. In this study, two-dimensional immunoblot and Matrix-Assisted Laser Desorption Ionization tandem Time-of-flight mass spectrometry (MALDI-TOF-MS) were used to identify Der f 31. After Der f 31 was cloned, expressed and purified, skin prick test (SPT), Immune inhibitory assays, Western blot, ELISA and asthmatic mouse model were employed to examine the allergenicity of recombinant Der f 31. The gene of Der f 31 includes 447 bps, and encoded 148 amino acids. Positive responses of SPT to r-Der f 31 were 32.5% in 43 HDM-allergic patients. r-Der f 31 can induce allergic pulmonary inflammation in the mouse model. In conclusion, Der f 31 is a novel subtype of dust mite allergens.


Assuntos
Alérgenos/imunologia , Antígenos de Dermatophagoides/imunologia , Asma/imunologia , Dermatophagoides farinae/imunologia , Hipersensibilidade/imunologia , Fatores de Despolimerização de Actina/química , Fatores de Despolimerização de Actina/imunologia , Adolescente , Adulto , Idoso , Animais , Criança , Modelos Animais de Doenças , Feminino , Humanos , Imunoglobulina E/sangue , Masculino , Camundongos , Pessoa de Meia-Idade , Testes Cutâneos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...