14 research outputs found

    Important shapeshifter: mechanisms allowing astrocytes to respond to the changing nervous system during development, injury and disease

    Get PDF
    Astrocytes are the most prevalent glial cells in the brain. Historically considered as "merely supporting" neurons, recent research has shown that astrocytes actively participate in a large variety of central nervous system (CNS) functions including synaptogenesis, neuronal transmission and synaptic plasticity. During disease and injury, astrocytes efficiently protect neurons by various means, notably by sealing them off from neurotoxic factors and repairing the blood-brain barrier. Their ramified morphology allows them to perform diverse tasks by interacting with synapses, blood vessels and other glial cells. In this review article, we provide an overview of how astrocytes acquire their complex morphology during development. We then move from the developing to the mature brain, and review current research on perisynaptic astrocytic processes, with a particular focus on how astrocytes engage synapses and modulate their formation and activity. Comprehensive changes have been reported in astrocyte cell shape in many CNS pathologies. Factors influencing these morphological changes are summarized in the context of brain pathologies, such as traumatic injury and degenerative conditions. We provide insight into the molecular, cellular and cytoskeletal machinery behind these shape changes which drive the dynamic remodeling in astrocyte morphology during injury and the development of pathologies

    Analyzing the functional diversity of profilin1 and profilin 2a

    Get PDF
    Profiline sind kleine Aktin-bindende Proteine, die den Nukleotidaustausch der Aktin-Monomere beschleunigen und diese an die Plus-Enden der Aktin-Filamente befördern. Da diese Funktionen für die Aktin-Dynamik essentiell sind, ist die Existenz gewebsspezifischer Profilin-Isoformen weitgehend unverstanden. In dieser Arbeit wurde untersucht, ob sich das in Mammalia ubiquitäre Profilin 1 (PFN1) und das ZNS-spezifische Profilin 2a (PFN2a) redundant zueinander verhalten oder ob sie unterschiedliche Funktionen besitzen. Trotz ähnlicher biochemischer Eigenschaften lokalisieren PFN1 und PFN2a unterschiedlich in Neuronen: Während PFN1 überwiegend homogen in den Neuriten verteilt ist, akkumuliert PFN2a in synaptischen Strukturen. Zudem wurde in dieser Arbeit ein RNAi-System entwickelt, mit dem endogene Proteine gegen exogene Varianten substituiert werden können. Mit diesem „Knock down & Knock in“-System wurde der Einfluss von PFN2a auf organotypisch kultivierte CA1-Neurone untersucht. Der Verlust von PFN2a verringert die Anzahl von dendritischen Verzweigungen sowie von „dendritic spines“. Die Substitution des endogenen PFN2a mit einer Profilin-Mutante mit reduzierter Aktin-Bindung hat gezeigt, dass die dendritische Morphologie auf der Interaktion von PFN2a mit Aktin beruht. Hingegen bewirkt die Expression einer Poly-Prolin-Bindungsmutante von PFN2a eine normale dendritische Komplexität und eine signifikant erhöhte Anzahl der „dendritic spines”. Sequenzvergleiche ergaben, dass PFN2a höher konserviert ist als PFN1. Darüber hinaus belegten Expressionsanalysen von PFN1 und PFN2a, dass PFN2a im Huhn ubiquitär exprimiert wird. RNAi-Experimente haben zudem gezeigt, dass in Hühnerfibroblasten vornehmlich PFN2a anstelle von PFN1 die Zelladhäsion und Zellmotilität moduliert. Die Ergebnisse dieser Arbeit bestätigen, dass PFN2a neuro-spezifische Funktionen ausübt. Andererseits weisen die Befunde an Hühnerzellen daraufhin, dass PFN2a prinzipiell auch als Aktin-Regulator fungieren kann.Profilins are small actin-binding proteins that accelerate the nucleotide-exchange of G-actin and deliver it to plus-ends of actin-filaments. According to the substantial contribution of profilin to actin dynamics, the functions of tissue-specific profilin-isoforms are poorly understood. Here, the functional diversity of the in Mammals ubiquitously expressed profilin 1 (PFN1) and the CNS-restricted profilin 2a (PFN2a) were studied. Although possessing similar biochemical properties, PFN1 and PFN2a exhibit differential localizations in neurons. Whereas PFN1 is homogenously distributed, PFN2a accumulates at synaptic structures. Furthermore, a RNAi-system were also developed that enables the simultaneous depletion of endogenous target proteins and the expression of exogenous variants. This so-called ôKnock down & knock inö-system were utilized to study the functions of PFN2a in organotypic-cultivated CA1-neurons. RNAi-mediated gene silencing of PFN2a leads to loss of dendritic arborizations and reduces significantly the number of dendritic spines. The substitution of endogenous PFN2a against a mutant deficient in actin binding demonstrates the modulation of dendritic morphology by the direct interaction of PFN2a and G-actin. However, the expression of PFN2a with disrupted poly-proline binding site restores dendritic complexity and increases significantly the density of dendritic spines in CA1-neurons. Phylogenetic analysis revealed that PFN2a is better conserved among vertebrates than PFN1. However, analysis of expression pattern showed that PFN2a is ubiquitously expressed in chicken tissues. Additionally, knock down experiments in chicken fibroblasts revealed the predominant involvement of PFN2a in processes of actin-based motility. In summary, the results of this work demonstrate specific functions of PFN2a in the mammalian CNS, whereas experiments in chicken show that PFN2a can principally serve as regulator of actin dynamics in non-neuronal cells

    Drebrin controls scar formation and astrocyte reactivity upon traumatic brain injury by regulating membrane trafficking

    Get PDF
    The brain of mammals lacks a significant ability to regenerate neurons and is thus particularly vulnerable. To protect the brain from injury and disease, damage control by astrocytes through astrogliosis and scar formation is vital. Here, we show that brain injury in mice triggers an immediate upregulation of the actin-binding protein Drebrin (DBN) in astrocytes, which is essential for scar formation and maintenance of astrocyte reactivity. In turn, DBN loss leads to defective astrocyte scar formation and excessive neurodegeneration following brain injuries. At the cellular level, we show that DBN switches actin homeostasis from ARP2/3-dependent arrays to microtubule-compatible scaffolds, facilitating the formation of RAB8-positive membrane tubules. This injury-specific RAB8 membrane compartment serves as hub for the trafficking of surface proteins involved in astrogliosis and adhesion mediators, such as β1-integrin. Our work shows that DBN-mediated membrane trafficking in astrocytes is an important neuroprotective mechanism following traumatic brain injury in mice

    Neuronal Profilin Isoforms Are Addressed by Different Signalling Pathways

    Get PDF
    Profilins are prominent regulators of actin dynamics. While most mammalian cells express only one profilin, two isoforms, PFN1 and PFN2a are present in the CNS. To challenge the hypothesis that the expression of two profilin isoforms is linked to the complex shape of neurons and to the activity-dependent structural plasticity, we analysed how PFN1 and PFN2a respond to changes of neuronal activity. Simultaneous labelling of rodent embryonic neurons with isoform-specific monoclonal antibodies revealed both isoforms in the same synapse. Immunoelectron microscopy on brain sections demonstrated both profilins in synapses of the mature rodent cortex, hippocampus and cerebellum. Both isoforms were significantly more abundant in postsynaptic than in presynaptic structures. Immunofluorescence showed PFN2a associated with gephyrin clusters of the postsynaptic active zone in inhibitory synapses of embryonic neurons. When cultures were stimulated in order to change their activity level, active synapses that were identified by the uptake of synaptotagmin antibodies, displayed significantly higher amounts of both isoforms than non-stimulated controls. Specific inhibition of NMDA receptors by the antagonist APV in cultured rat hippocampal neurons resulted in a decrease of PFN2a but left PFN1 unaffected. Stimulation by the brain derived neurotrophic factor (BDNF), on the other hand, led to a significant increase in both synaptic PFN1 and PFN2a. Analogous results were obtained for neuronal nuclei: both isoforms were localized in the same nucleus, and their levels rose significantly in response to KCl stimulation, whereas BDNF caused here a higher increase in PFN1 than in PFN2a. Our results strongly support the notion of an isoform specific role for profilins as regulators of actin dynamics in different signalling pathways, in excitatory as well as in inhibitory synapses. Furthermore, they suggest a functional role for both profilins in neuronal nuclei

    Profilin Isoforms in Health and Disease – All the Same but Different

    Get PDF
    Profilins are small actin binding proteins, which are structurally conserved throughout evolution. They are probably best known to promote and direct actin polymerization. However, they also participate in numerous cell biological processes beyond the roles typically ascribed to the actin cytoskeleton. Moreover, most complex organisms express several profilin isoforms. Their cellular functions are far from being understood, whereas a growing number of publications indicate that profilin isoforms are involved in the pathogenesis of various diseases. In this review, we will provide an overview of the profilin family and "typical" profilin properties including the control of actin dynamics. We will then discuss the profilin isoforms of higher animals in detail. In terms of cellular functions, we will focus on the role of Profilin 1 (PFN1) and Profilin 2a (PFN2a), which are co-expressed in the central nervous system. Finally, we will discuss recent findings that link PFN1 and PFN2a to neurological diseases, such as amyotrophic lateral sclerosis (ALS), Fragile X syndrome (FXS), Huntington's disease and spinal muscular atrophy (SMA)

    Testis-expressed profilins 3 and 4 show distinct functional characteristics and localize in the acroplaxome-manchette complex in spermatids

    No full text
    Abstract Background Multiple profilin isoforms exist in mammals; at least four are expressed in the mammalian testis. The testis-specific isoforms profilin-3 (PFN3) and profilin-4 (PFN4) may have specialized roles in spermatogenic cells which are distinct from known functions fulfilled by the "somatic" profilins, profilin-1 (PFN1) and profilin-2 (PFN2). Results Ligand interactions and spatial distributions of PFN3 and PFN4 were compared by biochemical, molecular and immunological methods; PFN1 and PFN2 were employed as controls. β-actin, phosphoinositides, poly-L-proline and mDia3, but not VASP, were confirmed as in vitro interaction partners of PFN3. In parallel experiments, PFN4 bound to selected phosphoinositides but not to poly-L-proline, proline-rich proteins, or actin. Immunofluorescence microscopy of PFN3 and PFN4 revealed distinct subcellular locations in differentiating spermatids. Both were associated first with the acroplaxome and later with the transient manchette. Predicted 3D structures indicated that PFN3 has the actin-binding site conserved, but retains only approximately half of the common poly-L-proline binding site. PFN4, in comparison, has lost both, polyproline and actin binding sites completely, which is well in line with the experimental data. Conclusion The testis-specific isoform PFN3 showed major hallmarks of the well characterized "somatic" profilin isoforms, albeit with distinct binding affinities. PFN4, on the other hand, did not interact with actin or polyproline in vitro. Rather, it seemed to be specialized for phospholipid binding, possibly providing cellular functions which are distinct from actin dynamics regulation.</p

    PFN2a is enriched in postsynaptic regions of inhibitory synapses.

    No full text
    <p>(A): Confocal images of neurons with simultaneous immunostaining of PFN1 (left column), PFN2a (centre column) and gephyrin, a protein concentrated in the active zone of inhibitory postsynapses. (cf. <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0034167#pone.0034167-Jockusch1" target="_blank">[2]</a>). Note that PFN2a is frequently concentrated in gephyrin clusters, while PFN1 is rarely enriched in these structures (arrows). Right column: Higher magnification of a neuron immunostained for PFN2a, gephyrin and VGAT, a marker for the inhibitory presynapse. Note that PFN2a primarily colocalises with synaptic gephyrin clusters (arrows), whereas extra-synaptic gephyrin clusters, identified by lack of VGAT staining, are mostly negative for PFN2a (arrow heads). Bar: 10 µm. (B): Quantitative analysis of the presence of PFN2a in synaptic and extra-synaptic gephyrin clusters (at least 483 extra-synaptic and 1830 synaptic gephyrin clusters per experiment, mean errors are based on 3 independent experiments, statistical analysis by unpaired <i>t</i> test).</p

    ATM phosphorylation of the actin-binding protein drebrin controls oxidation stress-resistance in mammalian neurons and C. elegans

    No full text
    Drebrin is an actin-binding protein known to play a role in neuronal dendritic spines but its precise regulation is unclear. Here, the authors report that DBN is activated by oxidative stress in an ATM-kinase dependent manner and increases resistance to oxidative stress in mice and in C. elegans

    PFN1 and PFN2a are enriched at postsynaptic sites in the adult rat brain.

    No full text
    <p>(A): Ultrastructural localisation of PFN1 (upper row) and 2a (centre row) in the cortex (left column), CA1 region of the hippocampus (centre column) and cerebellar cortex (right column) as seen with pre-embedding immunogold labelling. Immunogold labelling of synaptophysin (bottom rows), a presynaptic marker, served to demonstrate the localisation of both profilin isoforms in synapses. Sp: dendritic spine. Bars: 50 nm. (B): Quantitation of PFN1 and PFN2a immunoreactivity in pre- and postsynaptic structures in cortex, hippocampus and cerebellum. The Y-axis represents the percentage of pre- and postsynapses positive for gold particles. Note that both isoforms are more densely concentrated in postsynaptic than in presynaptic structures. (25–50 synapses per experiment, mean errors are based on 2 independent experiments; * P<0.05, statistical analysis by paired <i>t-</i>test).</p

    Specificity of the monoclonal antibodies raised against PFN1 (2C5) or PFN2a (4H5).

    No full text
    <p>(A): Isoform-specific epitopes recognised by the monoclonal antibodies 2C5 and 4H5, as determined by pepscan analysis on overlapping 15mer amino acid sequences, as described <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0034167#pone.0034167-Schoenenberger1" target="_blank">[51]</a>. Their respective location on the surface of PFN1 or PFN2a (blue) is indicated in the structural models designed by using Pymol software (DeLano Scientific LLC, Palo Alto, USA, version 0.98). (B): PFN isoform specific reactivity of 2C5 and 4H5. (B1): Immunoblot of 2C5 with purified recombinant mouse PFN1, mouse brain PFN2a and total extract of mouse spleen. (B2): Dot blot (left) and immunoblot (right) of 4H5 with recombinant mouse PFN1, mouse brain PFN2a and total brain extract. BSA: bovine serum albumin used as control. (C): Confocal images of immunofluorescence with 2C5 (upper panels) and 4H5 (lower panels) of C2C12 mouse myoblasts transfected with GFP-PFN2a. The antibody 2C5 reveals a typical fine diffuse cytoplasmic, but also a nuclear staining for PFN1, while the PFN2a specific antibody 4H5 labels only the GFP-PFN2a transfected cells (lower left) as part of a C2C12 cell population (DIC image; lower right). Filamentous actin was stained with FITC-phalloidin. Bars: 10 µm.</p
    corecore