Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
Add more filters











Publication year range
1.
Dev Cell ; 57(1): 5-18.e8, 2022 01 10.
Article in English | MEDLINE | ID: mdl-34883065

ABSTRACT

Tubulin dimers assemble into dynamic microtubules, which are used by molecular motors as tracks for intracellular transport. Organization and dynamics of the microtubule network are commonly thought to be regulated at the polymer ends, where tubulin dimers can be added or removed. Here, we show that molecular motors running on microtubules cause exchange of dimers along the shaft in vitro and in cells. These sites of dimer exchange act as rescue sites where depolymerizing microtubules stop shrinking and start re-growing. Consequently, the average length of microtubules increases depending on how frequently they are used as motor tracks. An increase of motor activity densifies the cellular microtubule network and enhances cell polarity. Running motors leave marks in the shaft, serving as traces of microtubule usage to organize the polarity landscape of the cell.


Subject(s)
Kinesins/physiology , Microtubules/physiology , Actin Depolymerizing Factors/metabolism , Actin Depolymerizing Factors/physiology , Cell Polarity/physiology , HeLa Cells , Humans , Kinesins/metabolism , Molecular Motor Proteins/metabolism , Molecular Motor Proteins/physiology , Tubulin/physiology
2.
Mol Biol Cell ; 32(9): 869-879, 2021 04 19.
Article in English | MEDLINE | ID: mdl-33439671

ABSTRACT

Microtubule plus-end depolymerization rate is a potentially important target of physiological regulation, but it has been challenging to measure, so its role in spatial organization is poorly understood. Here we apply a method for tracking plus ends based on time difference imaging to measure depolymerization rates in large interphase asters growing in Xenopus egg extract. We observed strong spatial regulation of depolymerization rates, which were higher in the aster interior compared with the periphery, and much less regulation of polymerization or catastrophe rates. We interpret these data in terms of a limiting component model, where aster growth results in lower levels of soluble tubulin and microtubule-associated proteins (MAPs) in the interior cytosol compared with that at the periphery. The steady-state polymer fraction of tubulin was ∼30%, so tubulin is not strongly depleted in the aster interior. We propose that the limiting component for microtubule assembly is a MAP that inhibits depolymerization, and that egg asters are tuned to low microtubule density.


Subject(s)
Actin Depolymerizing Factors/metabolism , Microtubules/physiology , Actin Depolymerizing Factors/physiology , Actins/metabolism , Animals , Interphase/physiology , Kinesins/metabolism , Microscopy, Fluorescence/methods , Microtubules/metabolism , Spatio-Temporal Analysis , Spindle Apparatus/metabolism , Tubulin/metabolism , Xenopus laevis
3.
Shock ; 55(5): 660-665, 2021 05 01.
Article in English | MEDLINE | ID: mdl-32433215

ABSTRACT

INTRODUCTION: We recently demonstrated that fibrinogen stabilizes syndecan-1 on the endothelial cell (EC) surface and contributes to EC barrier protection, though the intracellular signaling pathway remains unclear. P21 (Rac1) activated kinase 1 (PAK1) is a protein kinase involved in intracellular signaling leading to actin cytoskeleton rearrangement and plays an important role in maintaining endothelial barrier integrity. We therefore hypothesized that fibrinogen binding to syndecan-1 activated the PAK1 pathway. METHODS: Primary human lung microvascular endothelial cells were incubated in 10% lactated Ringers (LR) solution or 10% fibrinogen saline solution (5 mg/mL). Protein phosphorylation was determined by Western blot analysis and endothelial permeability measured by fluorescein isothiocyanate (FITC)-dextran. Cells were silenced by siRNA transfection. Protein concentration was measured in the lung lavages of mice. RESULTS: Fibrinogen treatment resulted in increased syndecan-1, PAK1 activation (phosphorylation), cofilin activation (dephosphorylation), as well as decreased stress fibers and permeability when compared with LR treatment. Cofilin is an actin-binding protein that depolymerizes F-actin to decrease stress fiber formation. Notably, fibrinogen did not influence myosin light chain activation (phosphorylation), a mediator of EC tension. Silencing of PAK1 prevented fibrinogen-induced dephosphorylation of cofilin and barrier integrity. Moreover, to confirm the in vitro findings, mice underwent hemorrhagic shock and were resuscitated with either LR or fibrinogen. Hemorrhage shock decreased lung p-PAK1 levels and caused significant lung vascular leakage. However, fibrinogen administration increased p-PAK1 expression to near sham levels and remarkably prevented the lung leakage. CONCLUSION: We have identified a novel pathway by which fibrinogen activates PAK1 signaling to stimulate/dephosphorylate cofilin, leading to disassembly of stress fibers and reduction of endothelial permeability.


Subject(s)
Actin Depolymerizing Factors/physiology , Endothelial Cells/physiology , Fibrinogen/physiology , Signal Transduction , p21-Activated Kinases/physiology , Animals , Male , Mice , Mice, Inbred C57BL
4.
J Cell Sci ; 133(8)2020 04 28.
Article in English | MEDLINE | ID: mdl-32152181

ABSTRACT

The mechanisms that control intrinsic axon growth potential, and thus axon regeneration following injury, are not well understood. Developmental axon regrowth of Drosophila mushroom body γ-neurons during neuronal remodeling offers a unique opportunity to study the molecular mechanisms controlling intrinsic growth potential. Motivated by the recently uncovered developmental expression atlas of γ-neurons, we here focus on the role of the actin-severing protein cofilin during axon regrowth. We show that Twinstar (Tsr), the fly cofilin, is a crucial regulator of both axon growth and branching during developmental remodeling of γ-neurons. tsr mutant axons demonstrate growth defects both in vivo and in vitro, and also exhibit actin-rich filopodial-like structures at failed branch points in vivo Our data is inconsistent with Tsr being important for increasing G-actin availability. Furthermore, analysis of microtubule localization suggests that Tsr is required for microtubule infiltration into the axon tips and branch points. Taken together, we show that Tsr promotes axon growth and branching, likely by clearing F-actin to facilitate protrusion of microtubules.


Subject(s)
Actin Depolymerizing Factors , Drosophila Proteins/physiology , Drosophila , Microfilament Proteins/physiology , Neurons/physiology , Actin Depolymerizing Factors/physiology , Actins/genetics , Animals , Axons , Microtubules , Nerve Regeneration
5.
Cell Rep ; 26(13): 3493-3501.e4, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30917306

ABSTRACT

Environmental stress threatens the fidelity of embryonic morphogenesis. Heat, for example, is a teratogen. Yet how heat affects morphogenesis is poorly understood. Here, we identify a heat-inducible actin stress response (ASR) in Drosophila embryos that is mediated by the activation of the actin regulator Cofilin. Similar to ASR in adult mammalian cells, heat stress in fly embryos triggers the assembly of intra-nuclear actin rods. Rods measure up to a few microns in length, and their assembly depends on elevated free nuclear actin concentration and Cofilin. Outside the nucleus, heat stress causes Cofilin-dependent destabilization of filamentous actin (F-actin) in actomyosin networks required for morphogenesis. F-actin destabilization increases the chance of morphogenesis mistakes. Blocking the ASR by reducing Cofilin dosage improves the viability of heat-stressed embryos. However, improved viability correlates with restoring F-actin stability, not rescuing morphogenesis. Thus, ASR endangers embryos, perhaps by shifting actin from cytoplasmic filaments to an elevated nuclear pool.


Subject(s)
Actin Depolymerizing Factors/physiology , Actins/physiology , Heat-Shock Response , Morphogenesis/physiology , Adaptation, Physiological , Animals , Cytoplasm , Drosophila/embryology , Embryo, Nonmammalian , Up-Regulation
6.
Oncogene ; 37(11): 1485-1502, 2018 03.
Article in English | MEDLINE | ID: mdl-29321664

ABSTRACT

Mitochondria fission and mitophagy are fundamentally crucial to cellular physiology and play important roles in cancer progression. Developing a comprehensive understanding of the molecular mechanism underlying mitochondrial fission and mitophagy will provide novel strategies for cancer prevention and treatment. Actin has been shown to participate in mitochondrial fission and mitophagy regulation. Cofilin is best known as an actin-depolymerizing factor. However, the molecular mechanism by which cofilin regulates mitochondrial fission and mitophagy remains largely unknown. Here we report that knockdown of cofilin attenuates and overexpression of cofilin potentiates mitochondrial fission as well as PINK1/PARK2-dependent mitophagy induced by staurosporine (STS), etoposide (ETO), and carbonyl cyanide 3-chlorophenylhydrazone (CCCP). Cofilin-mediated-PINK1 (PTEN-induced putative kinase 1) accumulation mainly depends on its regulation of mitochondrial proteases, including peptidase mitochondrial processing beta (MPPß), presenilin-associated rhomboid-like protease (PARL), and ATPase family gene 3-like 2 (AFG3L2), via mitochondrial membrane potential activity. We also found that the interaction and colocalization of G-actin/F-actin with cofilin at mitochondrial fission sites undergo constriction after CCCP treatment. Pretreatment with the actin polymerization inhibitor latrunculin B (LatB) increased and actin-depolymerization inhibitor jasplakinolide (Jas) decreased mitochondrial translocation of actin induced by STS, ETO, and CCCP. Both LatB and Jas abrogated CCCP-mediated mitochondrial fission and mitophagy. Our data suggest that G-actin is the actin form that is translocated to mitochondria, and the actin-depolymerization activity regulated by cofilin at the mitochondrial fission site is crucial for inducing mitochondrial fission and mitophagy.


Subject(s)
Actin Depolymerizing Factors/physiology , Actins/metabolism , Mitochondrial Dynamics/genetics , Mitophagy/genetics , Protein Multimerization/genetics , Actin Depolymerizing Factors/metabolism , Binding Sites , Cells, Cultured , Humans , Protein Kinases/physiology , Protein Transport , Ubiquitin-Protein Ligases/physiology
7.
Curr Genet ; 64(3): 619-634, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29170805

ABSTRACT

Aspergillus fumigatus is a major fungal pathogen that is responsible for approximately 90% of human aspergillosis. Cofilin is an actin depolymerizing factor that plays crucial roles in multiple cellular functions in many organisms. However, the functions of cofilin in A. fumigatus are still unknown. In this study, we constructed an A. fumigatus strain overexpressing cofilin (cofilin OE). The cofilin OE strain displayed a slightly different growth phenotype, significantly increased resistance against H2O2 and diamide, and increased activation of the high osmolarity glycerol pathway compared to the wild-type strain (WT). The cofilin OE strain internalized more efficiently into lung epithelial A549 cells, and induced increased transcription of inflammatory factors (MCP-1, TNF-α and IL-8) compared to WT. Cofilin overexpression also resulted in increased polysaccharides including ß-1, 3-glucan and chitin, and increased transcription of genes related to oxidative stress responses and polysaccharide synthesis in A. fumigatus. However, the cofilin OE strain exhibited similar virulence to the wild-type strain in murine and Galleria mellonella infection models. These results demonstrated for the first time that cofilin, a regulator of actin cytoskeleton dynamics, might play a critical role in the regulation of oxidative stress responses and cell wall polysaccharide synthesis in A. fumigatus.


Subject(s)
Actin Depolymerizing Factors/physiology , Actins/metabolism , Aspergillus fumigatus/metabolism , Oxidative Stress , A549 Cells , Actin Depolymerizing Factors/genetics , Actin Depolymerizing Factors/metabolism , Aspergillus fumigatus/growth & development , Aspergillus fumigatus/pathogenicity , Blotting, Western , Cell Wall/metabolism , Endocytosis , Humans , Hydrogen Peroxide/metabolism , Inflammation Mediators/metabolism , Interleukin-8/genetics , Monocyte Chemoattractant Proteins/genetics , Polymerization , Polysaccharides/biosynthesis , Polysaccharides/metabolism , Real-Time Polymerase Chain Reaction , Transcription, Genetic , Tumor Necrosis Factor-alpha/genetics , Virulence
8.
PLoS One ; 12(2): e0171491, 2017.
Article in English | MEDLINE | ID: mdl-28152068

ABSTRACT

PURPOSE: The role of exercise to prevent or reverse aging-induced cognitive decline has been widely reported. This neuroprotection is associated with changes in the synaptic structure plasticity. However, the mechanisms of exercise-induced synaptic plasticity in the aging brain are still unclear. Thus, the aim of the present study is to investigate the aging-related alterations of Rho-GTPase and the modulatory influences of exercise training. METHODS: Young and old rats were used in this study. Old rats were subjected to different schedules of aerobic exercise (12 m/min, 60 min/d, 3d/w or 5d/w) or kept sedentary for 12 w. After 12 w of aerobic exercise, the synapse density in the cortex and hippocampus was detected with immunofluorescent staining using synaptophysin as a marker. The total protein levels of RhoA, Rac1, Cdc42 and cofilin in the cortex and hippocampus were detected with Western Blot. The activities of RhoA, Rac1 and Cdc42 were determined using a pull down assay. RESULTS: We found that synapse loss occurred in aging rats. However, the change of expression and activity of RhoA, Rac1 and Cdc42 was different in the cortex and hippocampus. In the cortex, the expression and activity of Rac1 and Cdc42 was greatly increased with aging, whereas there were no changes in the expression and activity of RhoA. In the hippocampus, the expression and activity of Rac1 and Cdc42 was greatly decreased and there were no changes in the expression and activity of RhoA. As a major downstream substrate of the Rho GTPase family, the increased expression of cofilin was only observed in the cortex. High frequency exercise ameliorated all aging-related changes in the cortex and hippocampus. CONCLUSIONS: These data suggest that aerobic exercise reverses synapse loss in the cortex and hippocampus in aging rats, which might be related to the regulation of Rho GTPases.


Subject(s)
Aging/physiology , Physical Conditioning, Animal/physiology , Signal Transduction/physiology , Synapses/physiology , rac1 GTP-Binding Protein/physiology , rhoA GTP-Binding Protein/physiology , Actin Depolymerizing Factors/analysis , Actin Depolymerizing Factors/physiology , Animals , Blotting, Western , Cerebral Cortex/chemistry , Cerebral Cortex/physiology , Fluorescent Antibody Technique , Hippocampus/chemistry , Hippocampus/physiology , Male , Rats , Rats, Wistar , cdc42 GTP-Binding Protein/analysis , cdc42 GTP-Binding Protein/physiology , rac1 GTP-Binding Protein/analysis , rhoA GTP-Binding Protein/analysis
9.
Plant Cell ; 29(2): 395-408, 2017 02.
Article in English | MEDLINE | ID: mdl-28123105

ABSTRACT

Functional divergence in paralogs is an important genetic source of evolutionary innovation. Actin-depolymerizing factors (ADFs) are among the most important actin binding proteins and are involved in generating and remodeling actin cytoskeletal architecture via their conserved F-actin severing or depolymerizing activity. In plants, ADFs coevolved with actin, but their biochemical properties are diverse. Unfortunately, the biochemical function of most plant ADFs and the potential mechanisms of their functional divergence remain unclear. Here, in vitro biochemical analyses demonstrated that all 11 ADF genes in Arabidopsis thaliana exhibit opposing biochemical properties. Subclass III ADFs evolved F-actin bundling (B-type) function from conserved F-actin depolymerizing (D-type) function, and subclass I ADFs have enhanced D-type function. By tracking historical mutation sites on ancestral proteins, several fundamental amino acid residues affecting the biochemical functions of these proteins were identified in Arabidopsis and various plants, suggesting that the biochemical divergence of ADFs has been conserved during the evolution of angiosperm plants. Importantly, N-terminal extensions on subclass III ADFs that arose from intron-sliding events are indispensable for the alteration of D-type to B-type function. We conclude that the evolution of these N-terminal extensions and several conserved mutations produced the diverse biochemical functions of plant ADFs from a putative ancestor.


Subject(s)
Actin Depolymerizing Factors/physiology , Arabidopsis Proteins/physiology , Arabidopsis/metabolism , Biological Evolution , Actin Depolymerizing Factors/genetics , Actin Depolymerizing Factors/metabolism , Actins/metabolism , Arabidopsis/chemistry , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Models, Molecular , Phylogeny
10.
Neuropharmacology ; 112(Pt A): 66-75, 2017 01.
Article in English | MEDLINE | ID: mdl-27543417

ABSTRACT

Long lasting synaptic plasticity involves both functional and morphological changes, but how these processes are molecularly linked to achieve coordinated plasticity remains poorly understood. Cofilin is a common target of multiple signaling pathways at the synapse and is required for both functional and spine plasticity, but how it is regulated is unclear. In this study, we investigate whether the involvement of cofilin in plasticity is developmentally regulated by examining the role of cofilin in hippocampal long-term depression (LTD) in both young (2 weeks) and mature (2 months) mice. We show that both total protein level of cofilin and its activation undergo significant changes as the brain matures, so that although the amount of cofilin decreases significantly in mature mice, its regulation by protein phosphorylation becomes increasingly important. Consistent with these biochemical data, we show that cofilin-mediated actin reorganization is essential for LTD in mature, but not in young mice. In contrast to cofilin, the GluA2 interactions with NSF and PICK1 appear to be required in both young and mature mice, indicating that AMPAR internalization is a common key mechanism for LTD expression regardless of the developmental stages. These results establish the temporal specificity of cofilin in LTD regulation and suggest that cofilin-mediated actin reorganization may serve as a key mechanism underlying developmental regulation of synaptic and spine plasticity. This article is part of the Special Issue entitled 'Ionotropic glutamate receptors'.


Subject(s)
Actin Depolymerizing Factors/physiology , Actins/physiology , Hippocampus/physiology , Long-Term Synaptic Depression , Actin Depolymerizing Factors/metabolism , Actins/metabolism , Animals , Carrier Proteins/metabolism , Carrier Proteins/physiology , Cell Cycle Proteins , Excitatory Postsynaptic Potentials , Hippocampus/growth & development , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , N-Ethylmaleimide-Sensitive Proteins/metabolism , N-Ethylmaleimide-Sensitive Proteins/physiology , Nuclear Proteins/metabolism , Nuclear Proteins/physiology
11.
Biophys Chem ; 218: 27-35, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27589672

ABSTRACT

Cofilin is one of the most essential regulatory proteins and participates in the process of disassembling actin filaments. Cofilin induces conformational changes to actin filaments, and both the bending and torsional rigidity of the filament. In this study, we investigate the effects of cofilin on the mechanical properties of actin filaments using computational methods. Three models defined by their number of bound cofilins are constructed using coarse-grained MARTINI force field, and they are then extended with steered molecular dynamics simulation. After obtaining the stress-strain curves of the models, we calculate their Young's moduli and other mechanical properties that have not yet been determined for actin filaments. We analyze the cause of the different behaviors of the three models based on their atomistic geometrical differences. Finally, it is demonstrated that cofilin binding causes changes in the distances, angles, and stabilities of the residues in actin filaments.


Subject(s)
Actin Cytoskeleton/physiology , Actin Depolymerizing Factors/physiology , Molecular Dynamics Simulation , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/metabolism , Actin Depolymerizing Factors/metabolism , Biomechanical Phenomena , Models, Chemical , Molecular Conformation
12.
J Biomech ; 49(9): 1831-1835, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27143106

ABSTRACT

Cofilin makes the actin filament flexible and thermally unstable by disassembling the filament and inducing bending and torsional compliance. Actin monomers bound to cofilin are able to chemically and mechanically interact in response to external forces. In this study, we performed two molecular dynamics tensile tests for actin and cofilactin filaments under identical conditions. Surprisingly, cofilactin filaments were found to be twisted, generating shear stress caused by torsion. Additionally, analysis by plane stress assumption indicated that the extension-torsion coupling effect increases the amount of principal stress by 10%. Using elasticity and solid mechanics theories, our study elucidates the role of cofilin in the disassembly of actin filaments under tensile forces.


Subject(s)
Actin Cytoskeleton/physiology , Actin Depolymerizing Factors/physiology , Biophysical Phenomena , Elasticity , Molecular Dynamics Simulation , Stress, Mechanical
13.
Sci Signal ; 8(399): ra105, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26486174

ABSTRACT

Smooth muscle cells (SMCs) and the extracellular matrix (ECM) are intimately associated in the aortic wall. Fbln4(SMKO) mice with an SMC-specific deletion of the Fbln4 gene, which encodes the vascular ECM component fibulin-4, develop ascending aortic aneurysms that have increased abundance of angiotensin-converting enzyme (ACE); inhibiting angiotensin II signaling within the first month of life prevents aneurysm development. We used comparative proteomics analysis of Fbln4(SMKO) aortas from postnatal day (P) 1 to P30 mice to identify key molecules involved in aneurysm initiation and expansion. At P14, the actin depolymerizing factor cofilin was dephosphorylated and thus activated, and at P7, the abundance of slingshot-1 (SSH1) phosphatase, an activator of cofilin, was increased, leading to actin cytoskeletal remodeling. Also, by P7, biomechanical changes and underdeveloped elastic lamina-SMC connections were evident, and the abundance of early growth response 1 (Egr1), a mechanosensitive transcription factor that stimulates ACE expression, was increased, which was before the increases in ACE abundance and cofilin activation. Postnatal deletion of Fbln4 in SMCs at P7 prevented cofilin activation and aneurysm formation, suggesting that these processes required disruption of elastic lamina-SMC connections. Phosphoinositide 3-kinase (PI3K) is involved in the angiotensin II-mediated activation of SSH1, and administration of PI3K inhibitors from P7 to P30 decreased SSH1 abundance and prevented aneurysms. These results suggest that aneurysm formation arises from abnormal mechanosensing of SMCs resulting from the loss of elastic lamina-SMC connections and from increased SSH1 and cofilin activity, which may be potential therapeutic targets for treating ascending aortic aneurysms.


Subject(s)
Actin Depolymerizing Factors/physiology , Aortic Aneurysm/physiopathology , Animals , Disease Progression , Mice
14.
Dev Growth Differ ; 57(4): 275-90, 2015 May.
Article in English | MEDLINE | ID: mdl-25864508

ABSTRACT

Reorganization of the actin cytoskeleton is essential for cellular processes during animal development. Cofilin and actin depolymerizing factor (ADF) are potent actin-binding proteins that sever and depolymerize actin filaments, acting to generate the dynamics of the actin cytoskeleton. The activity of cofilin is spatially and temporally regulated by a variety of intracellular molecular mechanisms. Cofilin is regulated by cofilin binding molecules, is phosphorylated at Ser-3 (inactivation) by LIM-kinases (LIMKs) and testicular protein kinases (TESKs), and is dephosphorylated (reactivation) by slingshot protein phosphatases (SSHs). Although studies of the molecular mechanisms of cofilin-induced reorganization of the actin cytoskeleton have been ongoing for decades, the multicellular functions of cofilin and its regulation in development are just becoming apparent. This review describes the molecular mechanisms of generating actin dynamics by cofilin and the intracellular signaling pathways for regulating cofilin activity. Furthermore, recent findings of the roles of cofilin in the development of several tissues and organs, especially neural tissues and cells, in model animals are described. Recent developmental studies have indicated that cofilin and its regulatory mechanisms are involved in cellular proliferation and migration, the establishment of cellular polarity, and the dynamic regulation of organ morphology.


Subject(s)
Actin Depolymerizing Factors/physiology , Actin Depolymerizing Factors/chemistry , Actin Depolymerizing Factors/genetics , Animals , Gene Expression , Humans , Protein Conformation , Signal Transduction
16.
Plant Cell ; 26(1): 340-52, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24464292

ABSTRACT

Conserved microbe-associated molecular patterns (MAMPs) are sensed by pattern recognition receptors (PRRs) on cells of plants and animals. MAMP perception typically triggers rearrangements to actin cytoskeletal arrays during innate immune signaling. However, the signaling cascades linking PRR activation by MAMPs to cytoskeleton remodeling are not well characterized. Here, we developed a system to dissect, at high spatial and temporal resolution, the regulation of actin dynamics during innate immune signaling in plant cells. Within minutes of MAMP perception, we detected changes to single actin filament turnover in epidermal cells treated with bacterial and fungal MAMPs. These MAMP-induced alterations phenocopied an ACTIN DEPOLYMERIZING FACTOR4 (ADF4) knockout mutant. Moreover, actin arrays in the adf4 mutant were unresponsive to a bacterial MAMP, elf26, but responded normally to the fungal MAMP, chitin. Together, our data provide strong genetic and cytological evidence for the inhibition of ADF activity regulating actin remodeling during innate immune signaling. This work is the first to directly link an ADF/cofilin to the cytoskeletal rearrangements elicited directly after pathogen perception in plant or mammalian cells.


Subject(s)
Actin Depolymerizing Factors/physiology , Actins/metabolism , Arabidopsis Proteins/physiology , Arabidopsis/metabolism , Signal Transduction/immunology , Actin Cytoskeleton/metabolism , Actin Depolymerizing Factors/genetics , Actin Depolymerizing Factors/metabolism , Arabidopsis/genetics , Arabidopsis/immunology , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Chitin/immunology , Gene Knockout Techniques , Immunity, Innate/genetics , Phenotype , Receptors, Pattern Recognition/genetics , Receptors, Pattern Recognition/metabolism , Receptors, Pattern Recognition/physiology
18.
J Neurosci ; 33(48): 18836-48, 2013 Nov 27.
Article in English | MEDLINE | ID: mdl-24285890

ABSTRACT

Axonal injury generates growth inert retraction bulbs with dynamic cytoskeletal properties that are severely compromised. Conversion of "frozen" retraction bulbs into actively progressing growth cones is a major aim in axon regeneration. Here we report that murine serum response factor (SRF), a gene regulator linked to the actin cytoskeleton, modulates growth cone actin dynamics during axon regeneration. In regeneration-competent facial motoneurons, Srf deletion inhibited axonal regeneration. In wild-type mice after nerve injury, SRF translocated from the nucleus to the cytoplasm, suggesting a cytoplasmic SRF function in axonal regeneration. Indeed, adenoviral overexpression of cytoplasmic SRF (SRF-ΔNLS-GFP) stimulated axonal sprouting and facial nerve regeneration in vivo. In primary central and peripheral neurons, SRF-ΔNLS-GFP stimulated neurite outgrowth, branch formation, and growth cone morphology. Furthermore, we uncovered a link between SRF and the actin-severing factor cofilin during axonal regeneration in vivo. Facial nerve axotomy increased the total cofilin abundance and also nuclear localization of phosphorylated cofilin in a subpopulation of lesioned motoneurons. This cytoplasmic-to-nucleus translocation of P-cofilin upon axotomy was reduced in motoneurons expressing SRF-ΔNLS-GFP. Finally, we demonstrate that cytoplasmic SRF and cofilin formed a reciprocal regulatory unit. Overexpression of cytoplasmic SRF reduced cofilin phosphorylation and vice versa: overexpression of cofilin inhibited SRF phosphorylation. Therefore, a regulatory loop consisting of SRF and cofilin might take part in reactivating actin dynamics in growth-inert retraction bulbs and facilitating axon regeneration.


Subject(s)
Actin Depolymerizing Factors/physiology , Axons/drug effects , Cytoplasm/metabolism , Nerve Regeneration/drug effects , Serum Response Factor/pharmacology , Actins/metabolism , Animals , Axotomy , Cytoplasm/drug effects , Facial Nerve/physiology , Female , Green Fluorescent Proteins , Male , Mice , Peripheral Nerves/cytology , Peripheral Nerves/drug effects , Phosphorylation , Polymerase Chain Reaction , Subcellular Fractions/metabolism
19.
J Neurochem ; 127(2): 199-208, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23895321

ABSTRACT

We and others have previously shown that the neuropeptide galanin modulates neurite outgrowth from adult sensory neurons via activation of the second galanin receptor; however, the intracellular signalling pathways that mediate this neuritogenic effect have yet to be elucidated. Here, we demonstrate that galanin decreases the activation state in adult sensory neurons and PC12 cells of Rho and Cdc42 GTPases, both known regulators of filopodial and growth cone motility. Consistent with this, activated levels of Rho and Cdc42 levels are increased in the dorsal root ganglion of adult galanin knockout animals compared with wildtype controls. Furthermore, galanin markedly increases the activation state of cofilin, a downstream effector of many of the small GTPases, in the cell bodies and growth cones of sensory neurons and in PC12 cells. We also demonstrate a reduction in the activation of cofilin, and alteration in growth cone motility, in cultured galanin knockout neurons compared with wildtype controls. These data provide the first evidence that galanin regulates the Rho family of GTPases and cofilin to stimulate growth cone dynamics and neurite outgrowth in sensory neurons. These findings have important therapeutic implications for the treatment of peripheral sensory neuropathies.


Subject(s)
Actin Depolymerizing Factors/physiology , Galanin/pharmacology , Neurites/drug effects , Sensory Receptor Cells/drug effects , cdc42 GTP-Binding Protein/antagonists & inhibitors , rho GTP-Binding Proteins/antagonists & inhibitors , Animals , Blotting, Western , Enzyme Activation/drug effects , Female , Ganglia, Spinal/cytology , Growth Cones/drug effects , Mice , Mice, Knockout , Microscopy, Video , PC12 Cells , Pseudopodia/drug effects , Rats , Stimulation, Chemical , rac GTP-Binding Proteins/metabolism
20.
Nat Rev Mol Cell Biol ; 14(7): 405-15, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23778968

ABSTRACT

Recently, a consensus has emerged that cofilin severing activity can generate free actin filament ends that are accessible for F-actin polymerization and depolymerization without changing the rate of G-actin association and dissociation at either filament end. The structural basis of actin filament severing by cofilin is now better understood. These results have been integrated with recently discovered mechanisms for cofilin activation in migrating cells, which led to new models for cofilin function that provide insights into how cofilin regulation determines the temporal and spatial control of cell behaviour.


Subject(s)
Actin Depolymerizing Factors/physiology , Cell Movement , Actin Depolymerizing Factors/chemistry , Actins/metabolism , Animals , Cell Surface Extensions/metabolism , Humans , Models, Molecular , Phosphorylation , Protein Multimerization , Protein Processing, Post-Translational , Protein Structure, Tertiary , Protein Transport
SELECTION OF CITATIONS
SEARCH DETAIL