Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 22
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
bioRxiv ; 2024 Mar 04.
Article de Anglais | MEDLINE | ID: mdl-38496404

RÉSUMÉ

Replication stress describes various types of endogenous and exogenous challenges to DNA replication in S-phase. Stress during this critical process results in helicase-polymerase decoupling at replication forks, triggering the S-phase checkpoint, which orchestrates global replication fork stalling and delayed entry into G2. The replication stressor most often used to induce the checkpoint response is hydroxyurea (HU), a chemotherapeutic agent. The primary mechanism of S-phase checkpoint activation by HU has thus far been considered to be a reduction of dNTP synthesis by inhibition of ribonucleotide reductase (RNR), leading to helicase-polymerase decoupling and subsequent activation of the checkpoint, mediated by the replisome associated effector kinase Mrc1. In contrast, we observe that HU causes cell cycle arrest in budding yeast independent of both the Mrc1-mediated replication checkpoint response and the Psk1-Mrc1 oxidative signaling pathway. We demonstrate a direct relationship between HU incubation and reactive oxygen species (ROS) production in yeast nuclei. We further observe that ROS strongly inhibits the in vitro polymerase activity of replicative polymerases (Pols), Pol α, Pol δ, and Pol ε, causing polymerase complex dissociation and subsequent loss of DNA substrate binding, likely through oxidation of their integral iron sulfur Fe-S clusters. Finally, we present "RNR-deg," a genetically engineered alternative to HU in yeast with greatly increased specificity of RNR inhibition, allowing researchers to achieve fast, nontoxic, and more readily reversible checkpoint activation compared to HU, avoiding harmful ROS generation and associated downstream cellular effects that may confound interpretation of results.

2.
Cell Rep ; 43(3): 113914, 2024 Mar 26.
Article de Anglais | MEDLINE | ID: mdl-38451813

RÉSUMÉ

Stroke, trauma, and neurodegenerative disorders cause loss of neurites (axons and dendrites) in addition to neuronal death. Neurite loss may result directly from a primary insult, secondary to parental neuron death, or secondary to a post-injury inflammatory response. Here, we use lipopolysaccharide and the alarmin S100ß to selectively evaluate neurite loss caused by the inflammatory response. Activation of microglia and infiltrating macrophages by these stimuli causes neurite loss that far exceeds neuronal death, both in vitro and in vivo. Neurite loss is accompanied by the formation of cofilactin rods and aggregates (CARs), which are polymers of cofilin-1 and actin induced by oxidative stress and other factors. Mice deficient in either cofilin-1 or the superoxide-generating enzyme NADPH oxidase-2 show reduced CAR formation, neurite loss, and motor impairment. The findings identify a mechanism by which inflammation leads to neurite loss via CAR formation and highlight the relevance of neurite loss to functional impairment.


Sujet(s)
Neurites , Maladies neurodégénératives , Souris , Animaux , Neurones , Axones , Inflammation
3.
Biomedicines ; 12(1)2024 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-38255199

RÉSUMÉ

Synapse loss is the principal cause of cognitive decline in Alzheimer's disease (AD) and related disorders (ADRD). Synapse development depends on the intricate dynamics of the neuronal cytoskeleton. Cofilin, the major protein regulating actin dynamics, can be sequestered into cofilactin rods, intra-neurite bundles of cofilin-saturated actin filaments that can disrupt vesicular trafficking and cause synaptic loss. Rods are a brain pathology in human AD and mouse models of AD and ADRD. Eliminating rods is the focus of this paper. One pathway for rod formation is triggered in ~20% of rodent hippocampal neurons by disease-related factors (e.g., soluble oligomers of Amyloid-ß (Aß)) and requires cellular prion protein (PrPC), active NADPH oxidase (NOX), and cytokine/chemokine receptors (CCRs). FDA-approved antagonists of CXCR4 and CCR5 inhibit Aß-induced rods in both rodent and human neurons with effective concentrations for 50% rod reduction (EC50) of 1-10 nM. Remarkably, two D-amino acid receptor-active peptides (RAP-103 and RAP-310) inhibit Aß-induced rods with an EC50 of ~1 pM in mouse neurons and ~0.1 pM in human neurons. These peptides are analogs of D-Ala-Peptide T-Amide (DAPTA) and share a pentapeptide sequence (TTNYT) antagonistic to several CCR-dependent responses. RAP-103 does not inhibit neuritogenesis or outgrowth even at 1 µM, >106-fold above its EC50. N-terminal methylation, or D-Thr to D-Ser substitution, decreases the rod-inhibiting potency of RAP-103 by 103-fold, suggesting high target specificity. Neither RAP peptide inhibits neuronal rod formation induced by excitotoxic glutamate, but both inhibit rods induced in human neurons by several PrPC/NOX pathway activators (Aß, HIV-gp120 protein, and IL-6). Significantly, RAP-103 completely protects against Aß-induced loss of mature and developing synapses and, at 0.1 nM, reverses rods in both rodent and human neurons (T½ ~ 3 h) even in the continuous presence of Aß. Thus, this orally available, brain-permeable peptide should be highly effective in reducing rod pathology in multifactorial neurological diseases with mixed proteinopathies acting through PrPC/NOX.

4.
Methods Enzymol ; 673: 191-225, 2022.
Article de Anglais | MEDLINE | ID: mdl-35965008

RÉSUMÉ

R-loop proteins present a stable and robust blockade to the progression of a DNA replication fork during S-phase. The consequences of this block can include mutagenesis and other irreversible chromosomal catastrophes, causing genomic instability and disease. As such, further investigation into the molecular mechanisms underlying R-loop protein resolution is warranted. The critical role of non-replicative accessory helicases in R-loop protein resolution has increasingly come into light in recent years. Such helicases include the Pif1-family, monomeric helicases that have been studied in many different contexts and that have been ascribed to a multitude of separable protective functions in the cell. In this chapter, we present protocols to study R-loop protein resolution by Pif1 helicase at stalled replication forks using purified proteins, both at the biochemical and single-molecule level. Our system uses recombinant proteins expressed in Saccharomyces cerevisiae but could apply to practically any organism of interest due to the high interspecies homology of the proteins involved in DNA replication. The methods we outline are extensible to many systems and should be applicable to studying R-loop clearance by any Superfamily (SF) 1B helicase. These techniques will further enable mechanistic research on these critical but understudied components of the genomic maintenance program.


Sujet(s)
Structures en boucle R , Protéines de Saccharomyces cerevisiae , Helicase/composition chimique , Réplication de l'ADN , Saccharomyces cerevisiae/génétique , Saccharomyces cerevisiae/métabolisme , Protéines de Saccharomyces cerevisiae/génétique , Protéines de Saccharomyces cerevisiae/métabolisme
5.
PLoS One ; 16(3): e0248309, 2021.
Article de Anglais | MEDLINE | ID: mdl-33705493

RÉSUMÉ

Nearly 50% of individuals with long-term HIV infection are affected by the onset of progressive HIV-associated neurocognitive disorders (HAND). HIV infiltrates the central nervous system (CNS) early during primary infection where it establishes persistent infection in microglia (resident macrophages) and astrocytes that in turn release inflammatory cytokines, small neurotoxic mediators, and viral proteins. While the molecular mechanisms underlying pathology in HAND remain poorly understood, synaptodendritic damage has emerged as a hallmark of HIV infection of the CNS. Here, we report that the HIV viral envelope glycoprotein gp120 induces the formation of aberrant, rod-shaped cofilin-actin inclusions (rods) in cultured mouse hippocampal neurons via a signaling pathway common to other neurodegenerative stimuli including oligomeric, soluble amyloid-ß and proinflammatory cytokines. Previous studies showed that synaptic function is impaired preferentially in the distal proximity of rods within dendrites. Our studies demonstrate gp120 binding to either chemokine co-receptor CCR5 or CXCR4 is capable of inducing rod formation, and signaling through this pathway requires active NADPH oxidase presumably through the formation of superoxide (O2-) and the expression of cellular prion protein (PrPC). These findings link gp120-mediated oxidative stress to the generation of rods, which may underlie early synaptic dysfunction observed in HAND.


Sujet(s)
Facteurs de dépolymérisation de l'actine/métabolisme , Actines/métabolisme , Protéine d'enveloppe gp120 du VIH/métabolisme , Infections à VIH/métabolisme , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/métabolisme , Hippocampe/métabolisme , NADPH oxidase/métabolisme , Neurones/métabolisme , Protéines PrPC/métabolisme , Récepteurs CCR5/métabolisme , Récepteurs CXCR4/métabolisme , Facteurs de dépolymérisation de l'actine/génétique , Actines/génétique , Animaux , Protéine d'enveloppe gp120 du VIH/génétique , Infections à VIH/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , Souris , Souris knockout , NADPH oxidase/génétique , Stress oxydatif/génétique , Protéines PrPC/génétique , Récepteurs CCR5/génétique , Récepteurs CXCR4/génétique
6.
J Neurosci ; 41(3): 392-407, 2021 01 20.
Article de Anglais | MEDLINE | ID: mdl-33268543

RÉSUMÉ

Neuroligins (NLGNs) are a class of postsynaptic cell adhesion molecules that interact with presynaptic neurexins (NRXNs) and regulate synapse function. NLGN4 is a member of the NLGN family and consists of a unique amino acid sequence in humans that is not evolutionarily well conserved in rodents. The human-specific NLGN4 gene has been reported to be mutated in many patients with autism and other neurodevelopmental disorders. However, it remained unclear how these mutations might alter the molecular properties of NLGN4 and affect synaptic transmission in human neurons. Here, we describe a severely autistic male patient carrying a single amino acid substitution (R101Q) in the NLGN4 gene. When expressed in HEK293 cells, the R101Q mutation in NLGN4 did not affect its binding affinity for NRXNs or its capacity to form homodimers. This mutation, however, impaired the maturation of NLGN4 protein by inhibiting N-linked glycosylation at an adjacent residue (N102), which is conserved in all NLGNs. As a result, the R101Q substitution significantly decreased the surface trafficking of NLGN4 and increased its retention in the endoplasmic reticulum and Golgi apparatus. In human neurons derived from male stem cell lines, the R101Q mutation also similarly reduced the synaptic localization of NLGN4, resulting in a loss-of-function phenotype. This mutation-induced trafficking defect substantially diminished the ability of NLGN4 to form excitatory synapses and modulate their functional properties. Viewed together, our findings suggest that the R101Q mutation is pathogenic for NLGN4 and can lead to synaptic dysfunction in autism.


Sujet(s)
Trouble autistique/génétique , Molécules d'adhérence cellulaire neuronale/génétique , Potentiels post-synaptiques excitateurs/physiologie , Mutation/génétique , Transmission synaptique/physiologie , Substitution d'acide aminé , Trouble autistique/psychologie , Enfant , Réticulum endoplasmique/métabolisme , Appareil de Golgi/métabolisme , Cellules HEK293 , Humains , Mâle , Mutation faux-sens/génétique , Cellules souches neurales , Tests neuropsychologiques , Techniques de patch-clamp , Synapses/métabolisme
7.
PLoS One ; 13(10): e0198709, 2018.
Article de Anglais | MEDLINE | ID: mdl-30325927

RÉSUMÉ

Functional impairment after brain ischemia results in part from loss of neuronal spines and dendrites, independent of neuronal death. Cofilin-actin rods are covalently linked aggregates of cofilin-1 and actin that form in neuronal processes (neurites) under conditions of ATP depletion and oxidative stress, and which cause neurite degeneration if not disassembled. ATP depletion and oxidative stress occur with differing severity, duration, and time course in different ischemic conditions. Here we evaluated four mouse models of brain ischemia to define the conditions that drive formation of cofilin-actin rods. Three of the models provide early reperfusion: transient middle cerebral artery occlusion (MCAo), transient bilateral common carotid artery occlusion (CCAo), and cardiac arrest / cardiopulmonary resuscitation (CA/CPR). Early reperfusion restores ATP generating capacity, but also induces oxidative stress. The fourth model, photothrombotic cortical infarction, does not provide reperfusion. Cofilin-actin rods were formed in each of these models, but with differing patterns. Where acute reperfusion occurred, rod formation was maximal within 4 hours after reperfusion. Where infarction occurred, rods continued to form for at least 24 hours after ischemic onset, and extended into the adjacent non-ischemic tissue. Interventions that limit cofilin-actin rod formation may help to preserve integrity of neuronal processes in permanent ischemia.


Sujet(s)
Actines/métabolisme , Encéphalopathie ischémique/métabolisme , Cofiline-1/métabolisme , Agrégation pathologique de protéines/métabolisme , Actines/analyse , Actines/ultrastructure , Animaux , Encéphalopathie ischémique/anatomopathologie , Cellules cultivées , Cofiline-1/analyse , Cofiline-1/ultrastructure , Modèles animaux de maladie humaine , Mâle , Souris de lignée C57BL , Neurones/métabolisme , Neurones/anatomopathologie , Stress oxydatif , Agrégation pathologique de protéines/anatomopathologie
8.
Pharmacol Ther ; 175: 17-27, 2017 Jul.
Article de Anglais | MEDLINE | ID: mdl-28232023

RÉSUMÉ

Cofilin is a ubiquitous protein which cooperates with many other actin-binding proteins in regulating actin dynamics. Cofilin has essential functions in nervous system development including neuritogenesis, neurite elongation, growth cone pathfinding, dendritic spine formation, and the regulation of neurotransmission and spine function, components of synaptic plasticity essential for learning and memory. Cofilin's phosphoregulation is a downstream target of many transmembrane signaling processes, and its misregulation in neurons has been linked in rodent models to many different neurodegenerative and neurological disorders including Alzheimer disease (AD), aggression due to neonatal isolation, autism, manic/bipolar disorder, and sleep deprivation. Cognitive and behavioral deficits of these rodent models have been largely abrogated by modulation of cofilin activity using viral-mediated, genetic, and/or small molecule or peptide therapeutic approaches. Neuropathic pain in rats from sciatic nerve compression has also been reduced by modulating the cofilin pathway within neurons of the dorsal root ganglia. Neuroinflammation, which occurs following cerebral ischemia/reperfusion, but which also accompanies many other neurodegenerative syndromes, is markedly reduced by peptides targeting specific chemokine receptors, which also modulate cofilin activity. Thus, peptide therapeutics offer potential for cost-effective treatment of a wide variety of neurological disorders. Here we discuss some recent results from rodent models using therapeutic peptides with a surprising ability to cross the rodent blood brain barrier and alter cofilin activity in brain. We also offer suggestions as to how neuronal-specific cofilin regulation might be achieved.


Sujet(s)
Cofiline-1/métabolisme , Maladies du système nerveux/métabolisme , Peptides/pharmacologie , Animaux , Encéphale/métabolisme , Humains , Maladies du système nerveux/traitement médicamenteux , Peptides/administration et posologie , Peptides/usage thérapeutique , Espèces réactives de l'oxygène/métabolisme
9.
J Vis Exp ; (130)2017 12 28.
Article de Anglais | MEDLINE | ID: mdl-29364208

RÉSUMÉ

Cultured rodent brain slices are useful for studying the cellular and molecular behavior of neurons and glia in an environment that maintains many of their normal in vivo interactions. Slices obtained from a variety of transgenic mouse lines or use of viral vectors for expression of fluorescently tagged proteins or reporters in wild type brain slices allow for high-resolution imaging by fluorescence microscopy. Although several methods have been developed for imaging brain slices, combining slice culture with the ability to perform repetitive high-resolution imaging of specific cells in live slices over long time periods has posed problems. This is especially true when viral vectors are used for expression of exogenous proteins since this is best done in a closed system to protect users and prevent cross contamination. Simple modifications made to the roller tube brain slice culture method that allow for repetitive high-resolution imaging of slices over many weeks in an enclosed system are reported. Culturing slices on photoetched coverslips permits the use of fiducial marks to rapidly and precisely reposition the stage to image the identical field over time before and after different treatments. Examples are shown for the use of this method combined with specific neuronal staining and expression to observe changes in hippocampal slice architecture, viral-mediated neuronal expression of fluorescent proteins, and the development of cofilin pathology, which was previously observed in the hippocampus of Alzheimer's disease (AD) in response to slice treatment with oligomers of amyloid-ß (Aß) peptide.


Sujet(s)
Encéphale/cytologie , Techniques de culture de tissus/méthodes , Maladie d'Alzheimer/anatomopathologie , Animaux , Encéphale/anatomopathologie , Encéphale/chirurgie , Hippocampe/cytologie , Hippocampe/anatomopathologie , Hippocampe/chirurgie , Humains , Souris , Microscopie confocale
10.
PLoS One ; 9(4): e95995, 2014.
Article de Anglais | MEDLINE | ID: mdl-24760020

RÉSUMÉ

Neurites of neurons under acute or chronic stress form bundles of filaments (rods) containing 1∶1 cofilin∶actin, which impair transport and synaptic function. Rods contain disulfide cross-linked cofilin and are induced by treatments resulting in oxidative stress. Rods form rapidly (5-30 min) in >80% of cultured hippocampal or cortical neurons treated with excitotoxic levels of glutamate or energy depleted (hypoxia/ischemia or mitochondrial inhibitors). In contrast, slow rod formation (50% of maximum response in ∼6 h) occurs in a subpopulation (∼20%) of hippocampal neurons upon exposure to soluble human amyloid-ß dimer/trimer (Aßd/t) at subnanomolar concentrations. Here we show that proinflammatory cytokines (TNFα, IL-1ß, IL-6) also induce rods at the same rate and within the same neuronal population as Aßd/t. Neurons from prion (PrP(C))-null mice form rods in response to glutamate or antimycin A, but not in response to proinflammatory cytokines or Aßd/t. Two pathways inducing rod formation were confirmed by demonstrating that NADPH-oxidase (NOX) activity is required for prion-dependent rod formation, but not for rods induced by glutamate or energy depletion. Surprisingly, overexpression of PrP(C) is by itself sufficient to induce rods in over 40% of hippocampal neurons through the NOX-dependent pathway. Persistence of PrP(C)-dependent rods requires the continuous activity of NOX. Removing inducers or inhibiting NOX activity in cells containing PrP(C)-dependent rods causes rod disappearance with a half-life of about 36 min. Cofilin-actin rods provide a mechanism for synapse loss bridging the amyloid and cytokine hypotheses for Alzheimer disease, and may explain how functionally diverse Aß-binding membrane proteins induce synaptic dysfunction.


Sujet(s)
Peptides bêta-amyloïdes/métabolisme , Hippocampe/cytologie , Hippocampe/métabolisme , Neurites/métabolisme , Protéines PrPC/métabolisme , Transduction du signal , Facteur de nécrose tumorale alpha/métabolisme , Facteurs de dépolymérisation de l'actine/métabolisme , Actines/métabolisme , Animaux , Cellules cultivées , Dactinomycine/pharmacologie , Femelle , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Acide glutamique/pharmacologie , Humains , Inflammation/métabolisme , Souris , NADPH oxidase/métabolisme , Protéines PrPC/génétique , Rats , Transmission synaptique/effets des médicaments et des substances chimiques
11.
BMC Cell Biol ; 14: 45, 2013 Oct 05.
Article de Anglais | MEDLINE | ID: mdl-24093776

RÉSUMÉ

BACKGROUND: ADF/cofilin proteins are key modulators of actin dynamics in metastasis and invasion of cancer cells. Here we focused on the roles of ADF and cofilin-1 individually in the development of polarized migration of rat mammary adenocarcinoma (MTLn3) cells, which express nearly equal amounts of each protein. Small interference RNA (siRNA) technology was used to knockdown (KD) the expression of ADF and cofilin-1 independently. RESULTS: Either ADF KD or cofilin KD caused cell elongation, a reduction in cell area, a decreased ability to form invadopodia, and a decreased percentage of polarized cells after 180 s of epidermal growth factor stimulation. Moreover, ADF KD or cofilin KD increased the rate of cell migration and the time of lamellipodia protrusion but through different mechanisms: lamellipodia protrude more frequently in ADF KD cells and are more persistent in cofilin KD cells. ADF KD cells showed a significant increase in F-actin aggregates, whereas cofilin KD cells showed a significant increase in prominent F-actin bundles and increased cell adhesion. Focal adhesion area and cell adhesion in cofilin KD cells were returned to control levels by expressing exogenous cofilin but not ADF. Return to control rates of cell migration in ADF KD cells was achieved by expression of exogenous ADF but not cofilin, whereas in cofilin KD cells, expression of cofilin efficiently rescued control migration rates. CONCLUSION: Although ADF and cofilin have many redundant functions, each of these isoforms has functional differences that affect F-actin structures, cell adhesion and lamellipodial dynamics, all of which are important determinants of cell migration.


Sujet(s)
Actines/métabolisme , Adénocarcinome/métabolisme , Cofiline-1/génétique , Destrine/génétique , Régulation de l'expression des gènes tumoraux , Tumeurs mammaires de l'animal/métabolisme , Cytosquelette d'actine/effets des médicaments et des substances chimiques , Cytosquelette d'actine/génétique , Cytosquelette d'actine/métabolisme , Actines/génétique , Adénocarcinome/génétique , Adénocarcinome/anatomopathologie , Animaux , Adhérence cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire , Cofiline-1/antagonistes et inhibiteurs , Cofiline-1/métabolisme , Destrine/antagonistes et inhibiteurs , Destrine/métabolisme , Facteur de croissance épidermique/pharmacologie , Femelle , Contacts focaux/effets des médicaments et des substances chimiques , Contacts focaux/métabolisme , Contacts focaux/ultrastructure , Tumeurs mammaires de l'animal/génétique , Tumeurs mammaires de l'animal/anatomopathologie , Métastase tumorale , Petit ARN interférent/génétique , Petit ARN interférent/métabolisme , Rats , Transduction du signal , Cellules cancéreuses en culture
12.
PLoS One ; 8(12): e83609, 2013.
Article de Anglais | MEDLINE | ID: mdl-24391794

RÉSUMÉ

Filament bundles (rods) of cofilin and actin (1:1) form in neurites of stressed neurons where they inhibit synaptic function. Live-cell imaging of rod formation is hampered by the fact that overexpression of a chimera of wild type cofilin with a fluorescent protein causes formation of spontaneous and persistent rods, which is exacerbated by the photostress of imaging. The study of rod induction in living cells calls for a rod reporter that does not cause spontaneous rods. From a study in which single cofilin surface residues were mutated, we identified a mutant, cofilinR21Q, which when fused with monomeric Red Fluorescent Protein (mRFP) and expressed several fold above endogenous cofilin, does not induce spontaneous rods even during the photostress of imaging. CofilinR21Q-mRFP only incorporates into rods when they form from endogenous proteins in stressed cells. In neurons, cofilinR21Q-mRFP reports on rods formed from endogenous cofilin and induced by all modes tested thus far. Rods have a half-life of 30-60 min upon removal of the inducer. Vesicle transport in neurites is arrested upon treatments that form rods and recovers as rods disappear. CofilinR21Q-mRFP is a genetically encoded rod reporter that is useful in live cell imaging studies of induced rod formation, including rod dynamics, and kinetics of rod elimination.


Sujet(s)
Facteurs de dépolymérisation de l'actine/métabolisme , Actines/métabolisme , Neurones/métabolisme , Facteurs de dépolymérisation de l'actine/composition chimique , Facteurs de dépolymérisation de l'actine/génétique , Actines/composition chimique , Actines/génétique , Animaux , Lignée cellulaire tumorale , Systèmes informatiques , Gènes rapporteurs , Cellules HeLa , Humains , Cellules LLC-PK1 , Protéines luminescentes/composition chimique , Protéines luminescentes/génétique , Protéines luminescentes/métabolisme , Modèles moléculaires , Complexes multiprotéiques/composition chimique , Complexes multiprotéiques/génétique , Complexes multiprotéiques/métabolisme , Mutagenèse dirigée , Structure quaternaire des protéines , Protéines de fusion recombinantes/composition chimique , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/métabolisme , Analyse sur cellule unique , Suidae , Synapses/métabolisme ,
13.
Neuron ; 76(6): 1091-107, 2012 Dec 20.
Article de Anglais | MEDLINE | ID: mdl-23259946

RÉSUMÉ

Neurites are the characteristic structural element of neurons that will initiate brain connectivity and elaborate information. Early in development, neurons are spherical cells but this symmetry is broken through the initial formation of neurites. This fundamental step is thought to rely on actin and microtubule dynamics. However, it is unclear which aspects of the complex actin behavior control neuritogenesis and which molecular mechanisms are involved. Here, we demonstrate that augmented actin retrograde flow and protrusion dynamics facilitate neurite formation. Our data indicate that a single family of actin regulatory proteins, ADF/Cofilin, provides the required control of actin retrograde flow and dynamics to form neurites. In particular, the F-actin severing activity of ADF/Cofilin organizes space for the protrusion and bundling of microtubules, the backbone of neurites. Our data reveal how ADF/Cofilin organizes the cytoskeleton to drive actin retrograde flow and thus break the spherical shape of neurons.


Sujet(s)
Facteurs de dépolymérisation de l'actine/physiologie , Actines/métabolisme , Forme de la cellule/physiologie , Cortex cérébral/embryologie , Destrine/physiologie , Cônes de croissance/métabolisme , Neurites/métabolisme , Animaux , Transport biologique , Processus de croissance cellulaire/physiologie , Cellules cultivées , Cortex cérébral/cytologie , Hippocampe/cytologie , Hippocampe/embryologie , Techniques in vitro , Souris , Souris knockout , Microtubules/physiologie , Neurogenèse/physiologie
14.
J Neurosci ; 32(19): 6670-81, 2012 May 09.
Article de Anglais | MEDLINE | ID: mdl-22573689

RÉSUMÉ

Rod-shaped aggregates ("rods"), containing equimolar actin and the actin dynamizing protein cofilin, appear in neurons following a wide variety of potentially oxidative stress: simulated microischemia, cofilin overexpression, and exposure to peroxide, excess glutamate, or the dimer/trimer forms of amyloid-ß peptide (Aßd/t), the most synaptotoxic Aß species. These rods are initially reversible and neuroprotective, but if they persist in neurites, the synapses degenerate without neurons dying. Herein we report evidence that rod formation depends on the generation of intermolecular disulfide bonds in cofilin. Of four Cys-to-Ala cofilin mutations expressed in rat E18 hippocampal neurons, only the mutant incapable of forming intermolecular bonds (CC39,147AA) has significantly reduced ability to incorporate into rods. Rod regions show unusually high oxidation levels. Rods, isolated from stressed neurons, contain dithiothreitol-sensitive multimeric forms of cofilin, predominantly dimer. Oligomerization of cofilin in cells represents one more mechanism for regulating the actin dynamizing activity of cofilin and probably underlies synaptic loss.


Sujet(s)
Facteurs de dépolymérisation de l'actine/métabolisme , Actines/physiologie , Disulfures/métabolisme , Maladies neurodégénératives/métabolisme , Facteurs de dépolymérisation de l'actine/composition chimique , Facteurs de dépolymérisation de l'actine/génétique , Animaux , Lignée cellulaire tumorale , Cellules cultivées , Poulets , Disulfures/composition chimique , Femelle , Humains , Mâle , Souris , Maladies neurodégénératives/génétique , Oxydoréduction , Multimérisation de protéines/génétique , Rats , Suidae
15.
Dev Cell ; 22(3): 530-43, 2012 Mar 13.
Article de Anglais | MEDLINE | ID: mdl-22421043

RÉSUMÉ

The contractile actin cortex is important for diverse fundamental cell processes, but little is known about how the assembly of F-actin and myosin II motors is regulated. We report that depletion of actin depolymerizing factor (ADF)/cofilin proteins in human cells causes increased contractile cortical actomyosin assembly. Remarkably, our data reveal that the major cellular defects resulting from ADF/cofilin depletion, including cortical F-actin accumulation, were largely due to excessive myosin II activity. We identify that ADF/cofilins from unicellular organisms to humans share a conserved activity to inhibit myosin II binding to F-actin, indicating a mechanistic rationale for our cellular results. Our study establishes an essential requirement for ADF/cofilin proteins in the control of normal cortical contractility and in processes such as mitotic karyokinesis. We propose that ADF/cofilin proteins are necessary for controlling actomyosin assembly and intracellular contractile force generation, a function of equal physiological importance to their established roles in mediating F-actin turnover.


Sujet(s)
Facteurs de dépolymérisation de l'actine/métabolisme , Actines/métabolisme , Actomyosine/métabolisme , Myosine de type II/métabolisme , Cellules HeLa , Humains , Liaison aux protéines
16.
Mol Biol Cell ; 21(20): 3529-39, 2010 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-20739464

RÉSUMÉ

GLUT4 vesicles are actively recruited to the muscle cell surface upon insulin stimulation. Key to this process is Rac-dependent reorganization of filamentous actin beneath the plasma membrane, but the underlying molecular mechanisms have yet to be elucidated. Using L6 rat skeletal myoblasts stably expressing myc-tagged GLUT4, we found that Arp2/3, acting downstream of Rac GTPase, is responsible for the cortical actin polymerization evoked by insulin. siRNA-mediated silencing of either Arp3 or p34 subunits of the Arp2/3 complex abrogated actin remodeling and impaired GLUT4 translocation. Insulin also led to dephosphorylation of the actin-severing protein cofilin on Ser-3, mediated by the phosphatase slingshot. Cofilin dephosphorylation was prevented by strategies depolymerizing remodeled actin (latrunculin B or p34 silencing), suggesting that accumulation of polymerized actin drives severing to enact a dynamic actin cycling. Cofilin knockdown via siRNA caused overwhelming actin polymerization that subsequently inhibited GLUT4 translocation. This inhibition was relieved by reexpressing Xenopus wild-type cofilin-GFP but not the S3E-cofilin-GFP mutant that emulates permanent phosphorylation. Transferrin recycling was not affected by depleting Arp2/3 or cofilin. These results suggest that cofilin dephosphorylation is required for GLUT4 translocation. We propose that Arp2/3 and cofilin coordinate a dynamic cycle of actin branching and severing at the cell cortex, essential for insulin-mediated GLUT4 translocation in muscle cells.


Sujet(s)
Facteurs de dépolymérisation de l'actine/métabolisme , Complexe Arp-2-3/métabolisme , Actines/métabolisme , Membrane cellulaire/métabolisme , Transporteur de glucose de type 4/métabolisme , Insuline/pharmacologie , Cellules musculaires/métabolisme , Complexe Arp-2-3/déficit , Animaux , Lignée cellulaire , Membrane cellulaire/effets des médicaments et des substances chimiques , Régulation négative/effets des médicaments et des substances chimiques , Électrophorèse bidimensionnelle sur gel , Techniques de knock-down de gènes , Protéines à fluorescence verte/métabolisme , Humains , Protéines des microfilaments/métabolisme , Cellules musculaires/cytologie , Cellules musculaires/effets des médicaments et des substances chimiques , Phosphoric monoester hydrolases , Phosphorylation/effets des médicaments et des substances chimiques , Transport des protéines/effets des médicaments et des substances chimiques , Rats , Transduction du signal/effets des médicaments et des substances chimiques , Transferrine/métabolisme , Xenopus
17.
Dev Neurobiol ; 69(12): 761-79, 2009 Oct.
Article de Anglais | MEDLINE | ID: mdl-19513994

RÉSUMÉ

Axonogenesis involves a shift from uniform delivery of materials to all neurites to preferential delivery to the putative axon, supporting its more rapid extension. Waves, growth cone-like structures that propagate down the length of neurites, were shown previously to correlate with neurite growth in dissociated cultured hippocampal neurons. Waves are similar to growth cones in their structure, composition and dynamics. Here, we report that waves form in all undifferentiated neurites, but occur more frequently in the future axon during initial neuronal polarization. Moreover, wave frequency and their impact on neurite growth are altered in neurons treated with stimuli that enhance axonogenesis. Coincident with wave arrival, growth cones enlarge and undergo a marked increase in dynamics. Through their engorgement of filopodia along the neurite shaft, waves can induce de novo neurite branching. Actin in waves maintains much of its cohesiveness during transport whereas actin in nonwave regions of the neurite rapidly diffuses as measured by live cell imaging of photoactivated GFP-actin and photoconversion of Dendra-actin. Thus, waves represent an alternative axonal transport mechanism for actin. Waves also occur in neurons in organotypic hippocampal slices where they propagate along neurites in the dentate gyrus and the CA regions and induce branching. Taken together, our results indicate that waves are physiologically relevant and contribute to axon growth and branching via the transport of actin and by increasing growth cone dynamics.


Sujet(s)
Axones/physiologie , Cônes de croissance/physiologie , Neurites/physiologie , Neurones/cytologie , Actines/métabolisme , Animaux , Transport axonal/physiologie , Axones/métabolisme , Mouvement cellulaire/physiologie , Cellules cultivées , Techniques de transfert de gènes , Cônes de croissance/métabolisme , Hippocampe/cytologie , Hippocampe/métabolisme , Hippocampe/physiologie , Traitement d'image par ordinateur , Immunohistochimie , Souris , Microscopie confocale , Neurites/métabolisme , Neurones/métabolisme , Neurones/physiologie , Techniques de culture d'organes , Rats
18.
Cell Signal ; 18(9): 1501-14, 2006 Sep.
Article de Anglais | MEDLINE | ID: mdl-16442263

RÉSUMÉ

We previously showed that the transcription factor Pax3 regulates mesenchymal-to-epithelial transition (MET) in cultured osteogenic Saos-2 cells. Herein we demonstrate that Pax3 induced MET in these cells requires intact Pax3 DNA binding motifs and is associated with the altered expression and activity of numerous proteins involved in signal transduction pathways that regulate cytoskeleton remodeling, the majority of which were not previously detected by mRNA expression array analysis. Proper levels of active Rho GTPases are essential for Pax3 induced MET. Rac activity and actomyosin contractility via Rho/ROCK signaling are required for the formation of circumferential actin bundles, epithelial discoid cell shape and the regulation of membrane protrusions. Precise spatial activation of Rho GTPase signaling components is also paramount for MET. Endogenous PAK2, Rac1 and PIX, a Rac/Cdc42-GEF, localize to focal adhesions. Dynamic localization of PAK and PIX to focal adhesions is required for Pax3 induced MET and is dependent on full PAK activity because kinase dead or GTPase-binding deficient mutants of PAK sequester PIX at focal adhesions and disrupt Pax3 induced phenotypic MET. All together, our results define roles for Rho GTPases and their effectors in MET and newly identify proteins and signal transduction cascades regulated by Pax3.


Sujet(s)
Épithélium/physiologie , Mésoderme/physiologie , Morphogenèse , Facteurs de transcription PAX/métabolisme , Transduction du signal/physiologie , Protéines G rho/métabolisme , Actines/métabolisme , Animaux , Lignée cellulaire , Forme de la cellule , Cytosquelette/métabolisme , Cytosquelette/ultrastructure , Régulation de l'expression des gènes , Humains , Protéines et peptides de signalisation intracellulaire/métabolisme , Myosines/métabolisme , Facteur de transcription PAX3 , Facteurs de transcription PAX/génétique , Phénotype , Protein-Serine-Threonine Kinases/métabolisme , p21-Activated Kinases , Protéines G rho/génétique , rho-Associated Kinases
19.
J Neurobiol ; 66(2): 103-14, 2006 Feb 05.
Article de Anglais | MEDLINE | ID: mdl-16215999

RÉSUMÉ

Rho family GTPases have important roles in mediating the effects of guidance cues and growth factors on the motility of neuronal growth cones. We previously showed that the neurotrophin BDNF regulates filopodial dynamics on growth cones of retinal ganglion cell axons through activation of the actin regulatory proteins ADF and cofilin by inhibiting a RhoA-dependent pathway that phosphorylates (inactivates) ADF/cofilin. The GTPase Cdc42 has also been implicated in mediating the effects of positive guidance cues. In this article we investigated whether Cdc42 is involved in the effects of BDNF on filopodial dynamics. BDNF treatment increases Cdc42 activity in retinal neurons, and neuronal incorporation of constitutively active Cdc42 mimics the increases in filopodial number and length. Furthermore, constitutively active and dominant negative Cdc42 decreased and increased, respectively, the activity of RhoA in retinal growth cones, indicating crosstalk between these GTPases in retinal growth cones. Constitutively active Cdc42 mimicked the activation of ADF/cofilin that resulted from BDNF treatment, while dominant negative Cdc42 blocked the effects of BDNF on filopodia and ADF/cofilin. The inability of dominant negative Cdc42 to block ADF/cofilin activation and stimulation of filopodial dynamics by the ROCK inhibitor Y-27632 indicate interaction between Cdc42 and RhoA occurs upstream of ROCK. Our results demonstrate crosstalk occurs between GTPases in mediating the effects of BDNF on growth cone motility, and Cdc42 activity can promote actin dynamics via activation of ADF/cofilin.


Sujet(s)
Facteurs de dépolymérisation de l'actine/métabolisme , Facteur neurotrophique dérivé du cerveau/métabolisme , Cônes de croissance/métabolisme , Pseudopodes/métabolisme , Rétine/métabolisme , Protéine G cdc42/métabolisme , Animaux , Cellules cultivées , Embryon de poulet , Activation enzymatique/physiologie , Technique d'immunofluorescence , Traitement d'image par ordinateur
20.
J Neurosci ; 24(47): 10741-9, 2004 Nov 24.
Article de Anglais | MEDLINE | ID: mdl-15564592

RÉSUMÉ

The molecular mechanisms by which neurotrophins regulate growth cone motility are not well understood. This study investigated the signaling involved in transducing BDNF-induced increases of filopodial dynamics. Our results indicate that BDNF regulates filopodial length and number through a Rho kinase-dependent mechanism. Additionally, actin depolymerizing factor (ADF)/cofilin activity is necessary and sufficient to transduce the effects of BDNF. Our data indicate that activation of ADF/cofilin mimics the effects of BDNF on filopodial dynamics, whereas ADF/cofilin inactivity blocks the effects of BDNF. Furthermore, BDNF promotes the activation of ADF/cofilin by reducing the phosphorylation of ADF/cofilin. Although inhibition of myosin II also enhances filopodial length, our results indicate that BDNF signaling is independent of myosin II activity and that the two pathways result in additive effects on filopodial length. Thus, filopodial extension is regulated by at least two independent mechanisms. The BDNF-dependent pathway works via regulation of ADF/cofilin, independently of myosin II activity.


Sujet(s)
Facteur neurotrophique dérivé du cerveau/physiologie , Cônes de croissance/physiologie , Protéines des microfilaments/physiologie , Pseudopodes/physiologie , Rétine/ultrastructure , Protéines 14-3-3/physiologie , Facteurs de dépolymérisation de l'actine , Animaux , Facteur neurotrophique dérivé du cerveau/antagonistes et inhibiteurs , Embryon de poulet , Destrine , Cônes de croissance/ultrastructure , Composés hétérocycliques avec 4 noyaux ou plus/pharmacologie , Protéines et peptides de signalisation intracellulaire , Protéines des microfilaments/métabolisme , Myosine de type II/antagonistes et inhibiteurs , Myosine de type II/physiologie , Phosphorylation , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Protein-Serine-Threonine Kinases/physiologie , Transduction du signal/physiologie , Techniques de culture de tissus , rho-Associated Kinases
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...