Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
3.
Front Cell Dev Biol ; 9: 681122, 2021.
Article in English | MEDLINE | ID: mdl-34458253

ABSTRACT

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).

4.
Nat Commun ; 12(1): 1490, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33674568

ABSTRACT

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 ß1-integrin. Our work shows that DBN-mediated membrane trafficking in astrocytes is an important neuroprotective mechanism following traumatic brain injury in mice.


Subject(s)
Astrocytes/metabolism , Brain Injuries, Traumatic/metabolism , Cicatrix/metabolism , Neuropeptides/genetics , Neuropeptides/metabolism , Actin-Related Protein 2-3 Complex , Actins/metabolism , Animals , Brain/metabolism , Brain/pathology , Brain Injuries/metabolism , Brain Injuries/pathology , Brain Injuries, Traumatic/pathology , Cell Movement , Central Nervous System/metabolism , Disease Models, Animal , Gliosis/metabolism , Gliosis/pathology , Mice , Mice, Knockout , Neuroprotection , Transcriptome , rab GTP-Binding Proteins/metabolism
5.
Nat Commun ; 10(1): 486, 2019 01 30.
Article in English | MEDLINE | ID: mdl-30700723

ABSTRACT

Drebrin (DBN) regulates cytoskeletal functions during neuronal development, and is thought to contribute to structural and functional synaptic changes associated with aging and Alzheimer's disease. Here we show that DBN coordinates stress signalling with cytoskeletal dynamics, via a mechanism involving kinase ataxia-telangiectasia mutated (ATM). An excess of reactive oxygen species (ROS) stimulates ATM-dependent phosphorylation of DBN at serine-647, which enhances protein stability and accounts for improved stress resilience in dendritic spines. We generated a humanized DBN Caenorhabditis elegans model and show that a phospho-DBN mutant disrupts the protective ATM effect on lifespan under sustained oxidative stress. Our data indicate a master regulatory function of ATM-DBN in integrating cytosolic stress-induced signalling with the dynamics of actin remodelling to provide protection from synapse dysfunction and ROS-triggered reduced lifespan. They further suggest that DBN protein abundance governs actin filament stability to contribute to the consequences of oxidative stress in physiological and pathological conditions.


Subject(s)
Actins/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Oxidative Stress , Actins/genetics , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amino Acid Motifs , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Caenorhabditis elegans , Cells, Cultured , Dendritic Spines/genetics , Dendritic Spines/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropeptides/genetics , Phosphorylation , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
6.
Front Cell Neurosci ; 12: 261, 2018.
Article in English | MEDLINE | ID: mdl-30186118

ABSTRACT

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.

7.
Mol Biol Cell ; 26(25): 4552-61, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26466675

ABSTRACT

Astrocytic tumors are the most common form of primary brain tumor. Astrocytic tumor cells infiltrate the surrounding CNS tissue, allowing them to evade removal upon surgical resection of the primary tumor. Dynamic changes to the actin cytoskeleton are crucial to cancer cell invasion, but the specific mechanisms that underlie the particularly invasive phenotype of astrocytic tumor cells are unclear. Protein interacting with C kinase 1 (PICK1) is a PDZ and BAR domain-containing protein that inhibits actin-related protein 2/3 (Arp2/3)-dependent actin polymerization and is involved in regulating the trafficking of a number of cell-surface receptors. Here we report that, in contrast to other cancers, PICK1 expression is down-regulated in grade IV astrocytic tumor cell lines and also in clinical cases of the disease in which grade IV tumors have progressed from lower-grade tumors. Exogenous expression of PICK1 in the grade IV astrocytic cell line U251 reduces their capacity for anchorage-independent growth, two-dimensional migration, and invasion through a three-dimensional matrix, strongly suggesting that low PICK1 expression plays an important role in astrocytic tumorigenesis. We propose that PICK1 negatively regulates neoplastic infiltration of astrocytic tumors and that manipulation of PICK1 is an attractive possibility for therapeutic intervention.


Subject(s)
Actin Cytoskeleton/genetics , Astrocytoma/genetics , Brain Neoplasms/genetics , Carrier Proteins/biosynthesis , Nuclear Proteins/biosynthesis , Actin-Related Protein 2-3 Complex/genetics , Astrocytes/metabolism , Astrocytes/pathology , Astrocytoma/pathology , Brain Neoplasms/pathology , Carrier Proteins/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Neoplasm Invasiveness , Nuclear Proteins/genetics
8.
Neuron ; 79(2): 293-307, 2013 Jul 24.
Article in English | MEDLINE | ID: mdl-23889934

ABSTRACT

Inhibition of Arp2/3-mediated actin polymerization by PICK1 is a central mechanism to AMPA receptor (AMPAR) internalization and long-term depression (LTD), although the signaling pathways that modulate this process in response to NMDA receptor (NMDAR) activation are unknown. Here, we define a function for the GTPase Arf1 in this process. We show that Arf1-GTP binds PICK1 to limit PICK1-mediated inhibition of Arp2/3 activity. Expression of mutant Arf1 that does not bind PICK1 leads to reduced surface levels of GluA2-containing AMPARs and smaller spines in hippocampal neurons, which occludes subsequent NMDA-induced AMPAR internalization and spine shrinkage. In organotypic slices, NMDAR-dependent LTD of AMPAR excitatory postsynaptic currents is abolished in neurons expressing mutant Arf1. Furthermore, NMDAR stimulation downregulates Arf1 activation and binding to PICK1 via the Arf-GAP GIT1. This study defines Arf1 as a critical regulator of actin dynamics and synaptic function via modulation of PICK1.


Subject(s)
ADP-Ribosylation Factor 1/physiology , Actin-Related Protein 2-3 Complex/physiology , Actins/metabolism , Carrier Proteins/physiology , Neuronal Plasticity/physiology , Nuclear Proteins/physiology , Synapses/metabolism , Actin-Related Protein 2-3 Complex/antagonists & inhibitors , Actins/physiology , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Cytoskeletal Proteins , HEK293 Cells , Humans , Organ Culture Techniques , Polymerization , Rats , Rats, Wistar
9.
J Cell Sci ; 126(Pt 17): 3873-83, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23843614

ABSTRACT

Astrocytes exhibit a complex, branched morphology, allowing them to functionally interact with numerous blood vessels, neighboring glial processes and neuronal elements, including synapses. They also respond to central nervous system (CNS) injury by a process known as astrogliosis, which involves morphological changes, including cell body hypertrophy and thickening of major processes. Following severe injury, astrocytes exhibit drastically reduced morphological complexity and collectively form a glial scar. The mechanistic details behind these morphological changes are unknown. Here, we investigate the regulation of the actin-nucleating Arp2/3 complex in controlling dynamic changes in astrocyte morphology. In contrast to other cell types, Arp2/3 inhibition drives the rapid expansion of astrocyte cell bodies and major processes. This intervention results in a reduced morphological complexity of astrocytes in both dissociated culture and in brain slices. We show that this expansion requires functional myosin II downstream of ROCK and RhoA. Knockdown of the Arp2/3 subunit Arp3 or the Arp2/3 activator N-WASP by siRNA also results in cell body expansion and reduced morphological complexity, whereas depleting WAVE2 specifically reduces the branching complexity of astrocyte processes. By contrast, knockdown of the Arp2/3 inhibitor PICK1 increases astrocyte branching complexity. Furthermore, astrocyte expansion induced by ischemic conditions is delayed by PICK1 knockdown or N-WASP overexpression. Our findings identify a new morphological outcome for Arp2/3 activation in restricting rather than promoting outwards movement of the plasma membrane in astrocytes. The Arp2/3 regulators PICK1, and N-WASP and WAVE2 function antagonistically to control the complexity of astrocyte branched morphology, and this mechanism underlies the morphological changes seen in astrocytes during their response to pathological insult.


Subject(s)
Actin-Related Protein 2-3 Complex/metabolism , Astrocytes/metabolism , Carrier Proteins/metabolism , Central Nervous System/metabolism , Nuclear Proteins/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , Actin-Related Protein 2-3 Complex/genetics , Amides/pharmacology , Animals , Astrocytes/cytology , Astrocytes/drug effects , Carrier Proteins/genetics , Cells, Cultured , Colforsin/pharmacology , Enzyme Inhibitors/pharmacology , Fibroblasts , HEK293 Cells , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Mice , Myosin Type II/antagonists & inhibitors , Myosin Type II/metabolism , Nuclear Proteins/genetics , Pyridines/pharmacology , RNA Interference , RNA, Small Interfering , Rats , Thiazoles/pharmacology , Thiones/pharmacology , Uracil/analogs & derivatives , Uracil/pharmacology , Vasodilator Agents/pharmacology , Wiskott-Aldrich Syndrome Protein, Neuronal/genetics , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/metabolism
10.
PLoS One ; 7(3): e34167, 2012.
Article in English | MEDLINE | ID: mdl-22470532

ABSTRACT

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.


Subject(s)
Neurons/metabolism , Profilins/metabolism , Signal Transduction , Animals , Antibodies, Monoclonal/immunology , Brain-Derived Neurotrophic Factor/pharmacology , Carrier Proteins/metabolism , Cell Line , Cell Nucleus/metabolism , Hippocampus/metabolism , Membrane Proteins/metabolism , Mice , Profilins/analysis , Profilins/genetics , Protein Isoforms , Rats , Synapses/drug effects , Synapses/metabolism
11.
Proc Natl Acad Sci U S A ; 107(36): 15780-5, 2010 Sep 07.
Article in English | MEDLINE | ID: mdl-20798032

ABSTRACT

Two profilin isoforms (PFN1 and PFN2a) are expressed in the mammalian brain. Although profilins are essential for regulating actin dynamics in general, the specific role of these isoforms in neurons has remained elusive. We show that knockdown of the neuron-specific PFN2a results in a significant reduction in dendrite complexity and spine numbers of hippocampal neurons. Overexpression of PFN1 in PFN2a-deficient neurons prevents the loss of spines but does not restore dendritic complexity. Furthermore, we show that profilins are involved in differentially regulating actin dynamics downstream of the pan-neurotrophin receptor (p75(NTR)), a receptor engaged in modulating neuronal morphology. Overexpression of PFN2a restores the morphological changes in dendrites caused by p75(NTR) overexpression, whereas PFN1 restores the normal spine density. Our data assign specific functions to the two PFN isoforms, possibly attributable to different affinities for potent effectors also involved in actin dynamics, and suggest that they are important for the signal-dependent fine-tuning of neuronal architecture.


Subject(s)
Neurons/cytology , Profilins/physiology , Protein Isoforms/physiology , Animals , Base Sequence , Gene Knockdown Techniques , Immunohistochemistry , Mice , Profilins/genetics , Protein Isoforms/genetics , RNA Interference , Signal Transduction
12.
BMC Cell Biol ; 10: 34, 2009 May 06.
Article in English | MEDLINE | ID: mdl-19419568

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. beta-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.


Subject(s)
Profilins/metabolism , Spermatids/metabolism , Testis/metabolism , Actins/metabolism , Animals , Binding Sites , Computer Simulation , Humans , Kinetics , Male , Peptides/metabolism , Phospholipids/metabolism , Protein Structure, Tertiary , Rats , Spermatogenesis
13.
J Cell Sci ; 122(Pt 7): 957-64, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19258389

ABSTRACT

Profilins are small actin-binding proteins expressed in all eukaryotes. They are involved in the regulation of actin filament dynamics and various signalling pathways. The identification of a variety of profilin isoforms led to the assumption that there may be isoform-specific functions. In mammals, profilin-1 (PFN1) is ubiquitously expressed and engaged in the regulation of various motility processes in all cell types. By contrast, profilin-2a (PFN2a) is mainly restricted to neuronal cells and there is evidence that it is involved in neuronal plasticity and membrane trafficking. However, the PFN2a sequence is much better conserved than PFN1 throughout different phyla, indicating that its restricted expression and specialized function in mammals might be unique. Using isoform-specific antibodies, we show that the situation is different in birds. PFN2a is ubiquitously expressed in embryonic and adult chicken tissues at equal and frequently higher amounts than in mammals. Together with PFN1, it is present in cultivated chicken fibroblasts, but differentially localized. Knockdown experiments with miRNA reveal that PFN2a is involved in cell adhesion, spreading and locomotion, and silencing this isoform has pronounced consequences on these processes. Our results indicate profilin isoform expression is differentially regulated among vertebrates.


Subject(s)
Actins/metabolism , Cell Movement , Chickens/metabolism , Profilins/metabolism , Amino Acid Sequence , Animals , Antibody Specificity , Cell Adhesion , Chick Embryo , Conserved Sequence , Epitopes/immunology , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Knockdown Techniques , Intracellular Space/metabolism , Molecular Sequence Data , Profilins/chemistry , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Transport
SELECTION OF CITATIONS
SEARCH DETAIL
...