Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
Add more filters










Publication year range
1.
J Neurosci ; 44(6)2024 02 07.
Article in English | MEDLINE | ID: mdl-38129132

ABSTRACT

The coordinated action of a plethora of factors is required for the organization and dynamics of membranous structures critically underlying the development and function of cells, organs, and organisms. The evolutionary acquisition of additional amino acid motifs allows for expansion and/or specification of protein functions. We identify a thus far unrecognized motif specific for chordata EHBP1 proteins and demonstrate that this motif is critically required for interaction with syndapin I, an F-BAR domain-containing, membrane-shaping protein predominantly expressed in neurons. Gain-of-function and loss-of-function studies in rat primary hippocampal neurons (of mixed sexes) unraveled that EHBP1 has an important role in neuromorphogenesis. Surprisingly, our analyses uncovered that this newly identified function of EHBP1 did not require the domain responsible for Rab GTPase binding but was strictly dependent on EHBP1's syndapin I binding interface and on the presence of syndapin I in the developing neurons. These findings were underscored by temporally and spatially remarkable overlapping dynamics of EHBP1 and syndapin I at nascent dendritic branch sites. In addition, rescue experiments demonstrated the necessity of two additional EHBP1 domains for dendritic arborization, the C2 and CH domains. Importantly, the additionally uncovered critical involvement of the actin nucleator Cobl in EHBP1 functions suggested that not only static association with F-actin via EHBP1's CH domain is important for dendritic arbor formation but also actin nucleation. Syndapin interactions organize ternary protein complexes composed of EHBP1, syndapin I, and Cobl, and our functional data show that only together these factors give rise to proper cell shape during neuronal development.


Subject(s)
Actins , Microfilament Proteins , Rats , Animals , Actins/metabolism , Microfilament Proteins/metabolism , Actin Cytoskeleton/metabolism , Neurons/metabolism , Protein Binding
2.
Nature ; 618(7964): 394-401, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37225996

ABSTRACT

The endoplasmic reticulum (ER) undergoes continuous remodelling via a selective autophagy pathway, known as ER-phagy1. ER-phagy receptors have a central role in this process2, but the regulatory mechanism remains largely unknown. Here we report that ubiquitination of the ER-phagy receptor FAM134B within its reticulon homology domain (RHD) promotes receptor clustering and binding to lipidated LC3B, thereby stimulating ER-phagy. Molecular dynamics (MD) simulations showed how ubiquitination perturbs the RHD structure in model bilayers and enhances membrane curvature induction. Ubiquitin molecules on RHDs mediate interactions between neighbouring RHDs to form dense receptor clusters that facilitate the large-scale remodelling of lipid bilayers. Membrane remodelling was reconstituted in vitro with liposomes and ubiquitinated FAM134B. Using super-resolution microscopy, we discovered FAM134B nanoclusters and microclusters in cells. Quantitative image analysis revealed a ubiquitin-mediated increase in FAM134B oligomerization and cluster size. We found that the E3 ligase AMFR, within multimeric ER-phagy receptor clusters, catalyses FAM134B ubiquitination and regulates the dynamic flux of ER-phagy. Our results show that ubiquitination enhances RHD functions via receptor clustering, facilitates ER-phagy and controls ER remodelling in response to cellular demands.


Subject(s)
Autophagy , Endoplasmic Reticulum Stress , Endoplasmic Reticulum , Ubiquitination , Autophagy/physiology , Endoplasmic Reticulum/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Ubiquitins/metabolism , Microtubule-Associated Proteins/metabolism , Receptors, Autocrine Motility Factor/metabolism
3.
Nature ; 618(7964): 402-410, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37225994

ABSTRACT

Membrane-shaping proteins characterized by reticulon homology domains play an important part in the dynamic remodelling of the endoplasmic reticulum (ER). An example of such a protein is FAM134B, which can bind LC3 proteins and mediate the degradation of ER sheets through selective autophagy (ER-phagy)1. Mutations in FAM134B result in a neurodegenerative disorder in humans that mainly affects sensory and autonomic neurons2. Here we report that ARL6IP1, another ER-shaping protein that contains a reticulon homology domain and is associated with sensory loss3, interacts with FAM134B and participates in the formation of heteromeric multi-protein clusters required for ER-phagy. Moreover, ubiquitination of ARL6IP1 promotes this process. Accordingly, disruption of Arl6ip1 in mice causes an expansion of ER sheets in sensory neurons that degenerate over time. Primary cells obtained from Arl6ip1-deficient mice or from patients display incomplete budding of ER membranes and severe impairment of ER-phagy flux. Therefore, we propose that the clustering of ubiquitinated ER-shaping proteins facilitates the dynamic remodelling of the ER during ER-phagy and is important for neuronal maintenance.


Subject(s)
Autophagy , Endoplasmic Reticulum Stress , Endoplasmic Reticulum , Ubiquitinated Proteins , Ubiquitination , Animals , Humans , Mice , Autophagy/genetics , Endoplasmic Reticulum/metabolism , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/metabolism , Ubiquitinated Proteins/metabolism , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/pathology , Intracellular Membranes/metabolism
4.
Commun Biol ; 6(1): 366, 2023 04 03.
Article in English | MEDLINE | ID: mdl-37012315

ABSTRACT

Synaptic plasticity involves proper establishment and rearrangement of structural and functional microdomains. Yet, visualization of the underlying lipid cues proved challenging. Applying a combination of rapid cryofixation, membrane freeze-fracturing, immunogold labeling and electron microscopy, we visualize and quantitatively determine the changes and the distribution of phosphatidylinositol-4,5-bisphosphate (PIP2) in the plasma membrane of dendritic spines and subareas thereof at ultra-high resolution. These efforts unravel distinct phases of PIP2 signals during induction of long-term depression (LTD). During the first minutes PIP2 rapidly increases in a PIP5K-dependent manner forming nanoclusters. PTEN contributes to a second phase of PIP2 accumulation. The transiently increased PIP2 signals are restricted to upper and middle spine heads. Finally, PLC-dependent PIP2 degradation provides timely termination of PIP2 cues during LTD induction. Together, this work unravels the spatial and temporal cues set by PIP2 during different phases after LTD induction and dissects the molecular mechanisms underlying the observed PIP2 dynamics.


Subject(s)
Long-Term Synaptic Depression , Neurons , Phosphatidylinositols , Neuronal Plasticity , Neurons/physiology , Phosphatidylinositol 4,5-Diphosphate/metabolism
5.
Nat Cell Biol ; 25(1): 120-133, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36543981

ABSTRACT

In response to different types and intensities of mechanical force, cells modulate their physical properties and adapt their plasma membrane (PM). Caveolae are PM nano-invaginations that contribute to mechanoadaptation, buffering tension changes. However, whether core caveolar proteins contribute to PM tension accommodation independently from the caveolar assembly is unknown. Here we provide experimental and computational evidence supporting that caveolin-1 confers deformability and mechanoprotection independently from caveolae, through modulation of PM curvature. Freeze-fracture electron microscopy reveals that caveolin-1 stabilizes non-caveolar invaginations-dolines-capable of responding to low-medium mechanical forces, impacting downstream mechanotransduction and conferring mechanoprotection to cells devoid of caveolae. Upon cavin-1/PTRF binding, doline size is restricted and membrane buffering is limited to relatively high forces, capable of flattening caveolae. Thus, caveolae and dolines constitute two distinct albeit complementary components of a buffering system that allows cells to adapt efficiently to a broad range of mechanical stimuli.


Subject(s)
Caveolae , Caveolin 1 , Caveolae/metabolism , Caveolin 1/metabolism , Mechanotransduction, Cellular , Cell Membrane/metabolism , Proteins/metabolism
6.
J Neurosci ; 2022 Jul 21.
Article in English | MEDLINE | ID: mdl-35879097

ABSTRACT

Glycine receptor-mediated inhibitory neurotransmission is key for spinal cord function. Recent observations suggested that by largely elusive mechanisms also glycinergic synapses display synaptic plasticity. We imaged receptor fields at ultra-high resolution at freeze-fractured membranes, tracked surface and internalized glycine receptors (GlyR) and studied differential regulations of GlyRß interactions with the scaffold protein gephyrin and the F-BAR domain protein syndapin I and thereby reveal key principles of this process. S403 phosphorylation of GlyRß, known to be triggered by synaptic signaling, caused a decoupling from gephyrin scaffolds but simultaneously promoted association of syndapin I with GlyRß. In line, kainate-treatments used to trigger rearrangements of glycine receptors in murine syndapin I KO spinal cords (mixed sex) showed even more severe receptor field fragmentation than already observed in untreated syndapin I KO spinal cords. Syndapin I KO furthermore resulted in more dispersed receptors and increased receptor mobility also pointing out an important contribution of syndapin I in the organization of GlyRß fields. Strikingly, syndapin I KO also led to a complete disruption of kainate-induced GlyRß internalization. Accompanying quantitative ultra-high resolution studies in dissociated spinal cord neurons strongly suggested that the observed defects in GlyR internalization observed in syndapin I KO spinal cords are directly caused by syndapin I deficiency within murine spinal cord neurons. Together our results unveiled important mechanisms organizing and altering glycine receptor fields during both steady-state and particularly upon kainate-induced synaptic rearrangement - principles organizing and fine-tuning synaptic efficacy and plasticity of glycinergic synapses in the spinal cord.SIGNIFICANCE STATEMENTInitial observations suggested that also glycinergic synapses - key for spinal cord and brain stem functions - may display some form of synaptic plasticity. Imaging receptor fields at ultra-high resolution at freeze-fractured membranes, tracking surface and internalized glycine receptors (GlyR) and studying regulations of GlyRß interactions we here reveal key principles of these kainate-inducible adaptations. A switch from gephyrin-mediated receptor scaffolding to syndapin I-mediated GlyRß scaffolding and internalization allows for modulating synaptic receptor availability. In line, kainate-induced GlyRß internalization was completely disrupted and GlyRß receptor fields were distorted upon syndapin I KO. These results unveiled important mechanisms during both steady-state and kainate-induced alterations of synaptic GlyR fields - principles underlying synaptic efficacy and plasticity of synapses in the spinal cord.

7.
Cell Mol Life Sci ; 79(6): 286, 2022 May 09.
Article in English | MEDLINE | ID: mdl-35534740

ABSTRACT

Endocytosis is controlled by a well-orchestrated molecular machinery, where the individual players as well as their precise interactions are not fully understood. We now show that syndapin I/PACSIN 1 is expressed in pancreatic ß cells and that its knockdown abrogates ß cell endocytosis leading to disturbed plasma membrane protein homeostasis, as exemplified by an elevated density of L-type Ca2+ channels. Intriguingly, inositol hexakisphosphate (InsP6) activates casein kinase 2 (CK2) that phosphorylates syndapin I/PACSIN 1, thereby promoting interactions between syndapin I/PACSIN 1 and neural Wiskott-Aldrich syndrome protein (N-WASP) and driving ß cell endocytosis. Dominant-negative interference with endogenous syndapin I/PACSIN 1 protein complexes, by overexpression of the syndapin I/PACSIN 1 SH3 domain, decreases InsP6-stimulated endocytosis. InsP6 thus promotes syndapin I/PACSIN 1 priming by CK2-dependent phosphorylation, which endows the syndapin I/PACSIN 1 SH3 domain with the capability to interact with the endocytic machinery and thereby initiate endocytosis, as exemplified in ß cells.


Subject(s)
Cytoskeletal Proteins , Phytic Acid , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cytoskeletal Proteins/metabolism , Endocytosis/physiology , Phosphorylation
8.
PLoS Biol ; 19(12): e3001399, 2021 12.
Article in English | MEDLINE | ID: mdl-34898601

ABSTRACT

Ischemic stroke is a major cause of death and long-term disability. We demonstrate that middle cerebral artery occlusion (MCAO) in mice leads to a strong decline in dendritic arborization of penumbral neurons. These defects were subsequently repaired by an ipsilateral recovery process requiring the actin nucleator Cobl. Ischemic stroke and excitotoxicity, caused by calpain-mediated proteolysis, significantly reduced Cobl levels. In an apparently unique manner among excitotoxicity-affected proteins, this Cobl decline was rapidly restored by increased mRNA expression and Cobl then played a pivotal role in poststroke dendritic arbor repair in peri-infarct areas. In Cobl knockout (KO) mice, the dendritic repair window determined to span day 2 to 4 poststroke in wild-type (WT) strikingly passed without any dendritic regrowth. Instead, Cobl KO penumbral neurons of the primary motor cortex continued to show the dendritic impairments caused by stroke. Our results thereby highlight a powerful poststroke recovery process and identified causal molecular mechanisms critical during poststroke repair.


Subject(s)
Ischemic Stroke/metabolism , Microfilament Proteins/metabolism , Neuronal Plasticity/physiology , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Cytoskeletal Proteins/metabolism , Gene Expression/genetics , Infarction, Middle Cerebral Artery , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/genetics , Microfilament Proteins/physiology , Neurons/metabolism , Neurons/physiology
9.
Elife ; 102021 07 15.
Article in English | MEDLINE | ID: mdl-34264190

ABSTRACT

Local actin filament formation is indispensable for development of the dendritic arbor of neurons. We show that, surprisingly, the action of single actin filament-promoting factors was insufficient for powering dendritogenesis. Instead, this required the actin nucleator Cobl and its only evolutionary distant ancestor Cobl-like acting interdependently. This coordination between Cobl-like and Cobl was achieved by physical linkage by syndapins. Syndapin I formed nanodomains at convex plasma membrane areas at the base of protrusive structures and interacted with three motifs in Cobl-like, one of which was Ca2+/calmodulin-regulated. Consistently, syndapin I, Cobl-like's newly identified N terminal calmodulin-binding site and the single Ca2+/calmodulin-responsive syndapin-binding motif all were critical for Cobl-like's functions. In dendritic arbor development, local Ca2+/CaM-controlled actin dynamics thus relies on regulated and physically coordinated interactions of different F-actin formation-promoting factors and only together they have the power to bring about the sophisticated neuronal morphologies required for neuronal network formation in mammals.


Subject(s)
Actins/genetics , Actins/metabolism , Microfilament Proteins/metabolism , Neurons/metabolism , Actin Cytoskeleton/metabolism , Animals , Calcium Signaling , Calmodulin/metabolism , Cell Membrane/metabolism , Cytoskeletal Proteins/metabolism , Mice , Mice, Inbred C57BL , Microfilament Proteins/genetics , Protein Binding , Rats
10.
Cells ; 10(5)2021 05 01.
Article in English | MEDLINE | ID: mdl-34062765

ABSTRACT

The brain encompasses a complex network of neurons with exceptionally elaborated morphologies of their axonal (signal-sending) and dendritic (signal-receiving) parts. De novo actin filament formation is one of the major driving and steering forces for the development and plasticity of the neuronal arbor. Actin filament assembly and dynamics thus require tight temporal and spatial control. Such control is particularly effective at the level of regulating actin nucleation-promoting factors, as these are key components for filament formation. Arginine methylation represents an important post-translational regulatory mechanism that had previously been mainly associated with controlling nuclear processes. We will review and discuss emerging evidence from inhibitor studies and loss-of-function models for protein arginine methyltransferases (PRMTs), both in cells and whole organisms, that unveil that protein arginine methylation mediated by PRMTs represents an important regulatory mechanism in neuritic arbor formation, as well as in dendritic spine induction, maturation and plasticity. Recent results furthermore demonstrated that arginine methylation regulates actin cytosolic cytoskeletal components not only as indirect targets through additional signaling cascades, but can also directly control an actin nucleation-promoting factor shaping neuronal cells-a key process for the formation of neuronal networks in vertebrate brains.


Subject(s)
Actin Cytoskeleton/metabolism , Neurons/pathology , Protein Processing, Post-Translational , Actins/metabolism , Animals , Arginine/metabolism , Axons , Cells, Cultured , Cytoskeleton/metabolism , Cytosol/metabolism , Dendrites/metabolism , Dendritic Spines/metabolism , Humans , Inflammation , Methylation , Microfilament Proteins/metabolism , Neurites , Neurogenesis , Neuronal Plasticity , Neurons/metabolism , Phosphorylation , Protein-Arginine N-Methyltransferases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Serine/metabolism , Signal Transduction
11.
Curr Opin Cell Biol ; 68: 10-19, 2021 02.
Article in English | MEDLINE | ID: mdl-32927373

ABSTRACT

An intimate interplay of the plasma membrane with curvature-sensing and curvature-inducing proteins would allow for defining specific sites or nanodomains of action at the plasma membrane, for example, for protrusion, invagination, and polarization. In addition, such connections are predestined to ensure spatial and temporal order and sequences. The combined forces of membrane shapers and the cortical actin cytoskeleton might hereby in particular be required to overcome the strong resistance against membrane rearrangements in case of high plasma membrane tension or cellular turgor. Interestingly, also the opposite might be necessary, the inhibition of both membrane shapers and cytoskeletal reinforcement structures to relieve membrane tension to protect cells from membrane damage and rupturing during mechanical stress. In this review article, we discuss recent conceptual advances enlightening the interplay of plasma membrane curvature and the cortical actin cytoskeleton during endocytosis, modulations of membrane tensions, and the shaping of entire cells.


Subject(s)
Actin Cytoskeleton/physiology , Cell Membrane/chemistry , Cell Membrane/physiology , Cell Shape , Endocytosis , Actins/metabolism , Animals , Cytoskeleton/metabolism , Humans , Yeasts
12.
Sci Rep ; 10(1): 13110, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32753644

ABSTRACT

Sepsis-associated liver dysfunction manifesting as cholestasis is common during multiple organ failure. Three hepatocytic dysfunctions are considered as major hallmarks of cholestasis in sepsis: impairments of microvilli covering canalicular membranes, disruptions of tight junctions sealing bile-collecting canaliculae and disruptions of Mrp2-mediated hepatobiliary transport. PI3Kγ loss-of-function was suggested as beneficial in early sepsis. Yet, the PI3Kγ-regulated cellular processes in hepatocytes remained largely unclear. We analysed all three sepsis hallmarks for responsiveness to massive PI3K/Akt signalling and PI3Kγ loss-of-function, respectively. Surprisingly, neither microvilli nor tight junctions were strongly modulated, as shown by electron microscopical studies of mouse liver samples. Instead, quantitative electron microscopy proved that solely Mrp2 surface availability, i.e. the third hallmark, responded strongly to PI3K/Akt signalling. Mrp2 plasma membrane levels were massively reduced upon PI3K/Akt signalling. Importantly, Mrp2 levels at the plasma membrane of PI3Kγ KO hepatocytes remained unaffected upon PI3K/Akt signalling stimulation. The effect explicitly relied on PI3Kγ's enzymatic ability, as shown by PI3Kγ kinase-dead mice. Keeping the surface availability of the biliary transporter Mrp2 therefore is a cell biological process that may underlie the observation that PI3Kγ loss-of-function protects from hepatic excretory dysfunction during early sepsis and Mrp2 should thus take center stage in pharmacological interventions.


Subject(s)
Chemokines, CC/metabolism , Cholestasis/complications , Cholestasis/pathology , Macrophage Inflammatory Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Sepsis/complications , Animals , Cell Line , Cell Membrane/metabolism , Cholestasis/metabolism , Gene Knockout Techniques , Mice , Phosphatidylinositol 3-Kinases/deficiency , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
13.
Sci Rep ; 10(1): 11156, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32636403

ABSTRACT

Brush borders of intestinal epithelial cells are mandatory for nutrient uptake. Yet, which actin nucleators are crucial for forming the F-actin bundles supporting microvilli and the actin filaments of the terminal web, in which microvilli are rooted, is unknown. We show that mice lacking the actin nucleator Cobl surprisingly did not display reduced microvilli densities or changes in microvillar F-actin bundles or microvilli diameter but particularly in the duodenum displayed increased microvillar length. Interestingly, Cobl-deficient mice furthermore showed a significant widening of the terminal web. Quantitative analyses of high-resolution cryo-scanning electron microscopy (EM) of deep-etched duodenum samples revealed that Cobl is specifically important for the formation of fine filaments in the central terminal web that connect the apical structure of the terminal web underlying the plasma membrane, the microvilli rootlets and the basal structure of the terminal web with each other. Thus, the actin nucleator Cobl is critically involved in generating one of the cellular structures of the brush border-decorated apical cortex of enterocytes representing the absorptive intestinal surface.


Subject(s)
Enterocytes/metabolism , Microfilament Proteins/physiology , Actins/metabolism , Animals , Blotting, Western , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cryoelectron Microscopy/methods , Enterocytes/ultrastructure , Intestinal Mucosa/metabolism , Intestinal Mucosa/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Scanning/methods , Microvilli/physiology , Microvilli/ultrastructure , Real-Time Polymerase Chain Reaction
14.
Cells ; 9(6)2020 06 01.
Article in English | MEDLINE | ID: mdl-32492970

ABSTRACT

A major challenge in neuroscience is how to study structural alterations in the brain. Even small changes in synaptic composition could have severe outcomes for body functions. Many neuropathological diseases are attributable to disorganization of particular synaptic proteins. Yet, to detect and comprehensively describe and evaluate such often rather subtle deviations from the normal physiological status in a detailed and quantitative manner is very challenging. Here, we have compared side-by-side several commercially available light microscopes for their suitability in visualizing synaptic components in larger parts of the brain at low resolution, at extended resolution as well as at super-resolution. Microscopic technologies included stereo, widefield, deconvolution, confocal, and super-resolution set-ups. We also analyzed the impact of adaptive optics, a motorized objective correction collar and CUDA graphics card technology on imaging quality and acquisition speed. Our observations evaluate a basic set of techniques, which allow for multi-color brain imaging from centimeter to nanometer scales. The comparative multi-modal strategy we established can be used as a guide for researchers to select the most appropriate light microscopy method in addressing specific questions in brain research, and we also give insights into recent developments such as optical aberration corrections.


Subject(s)
Brain/anatomy & histology , Imaging, Three-Dimensional , Research , Animals , Male , Mice , Microscopy, Confocal , Neurons/cytology , Rats , Single-Cell Analysis , Synapses/physiology
15.
Methods Mol Biol ; 2169: 11-25, 2020.
Article in English | MEDLINE | ID: mdl-32548815

ABSTRACT

Membrane topology information and views of membrane-embedded protein complexes promote our understanding of membrane organization and cell biological function involving membrane compartments. Freeze-fracturing of biological membranes offers both stunning views onto integral membrane proteins and perpendicular views over wide areas of the membrane at electron microscopical resolution. This information is directly assessable for 3D analyses and quantitative analyses of the distribution of components within the membrane if it were possible to specifically detect the components of interest in the membranes. Freeze-fracture replica immunolabeling (FRIL) achieves just that. In addition, FRIL preserves antigens in their genuine cellular context free of artifacts of chemical fixation, as FRIL uses chemically unfixed cellular samples that are rapidly cryofixed. In principle, the method is not limited to integral proteins spanning the membrane. Theoretically, all membrane components should be addressable as long as they are antigenic, embedded into at least one membrane leaflet, and accessible for immunolabeling from either the intracellular or the extracellular side. Consistently, integral proteins spanning both leaflets and only partially inserted membrane proteins have been successfully identified and studied for their molecular organization and distribution in the membrane and/or in relationship to specialized membrane domains. Here we describe the freeze-fracturing of both cultured cells and tissues and the sample preparations that allowed for a successful immunogold-labeling of caveolin1 and caveolin3 or even for double-immunolabelings of caveolins with members of the syndapin family of membrane-associating and -shaping BAR domain proteins as well as with cavin 1. For this purpose samples are cryopreserved, fractured, and replicated. We also describe how the obtained stabilized membrane fractures are then cleaned to remove all loosely attached material and immunogold labeled to finally be viewed by transmission electron microscopy.


Subject(s)
Caveolae/metabolism , Caveolins/metabolism , Cell Membrane/metabolism , Freeze Fracturing/methods , Immunohistochemistry/methods , Microscopy, Electron, Transmission/methods , Animals , Caveolae/ultrastructure , Cell Line , Cryopreservation/instrumentation , Cryopreservation/methods , Freeze Fracturing/instrumentation , Membrane Proteins
16.
J Neurosci ; 40(25): 4954-4969, 2020 06 17.
Article in English | MEDLINE | ID: mdl-32354853

ABSTRACT

Glycine receptors (GlyRs) are the major mediators of fast synaptic inhibition in the adult human spinal cord and brainstem. Hereditary mutations to GlyRs can lead to the rare, but potentially fatal, neuromotor disorder hyperekplexia. Most mutations located in the large intracellular domain (TM3-4 loop) of the GlyRα1 impair surface expression levels of the receptors. The novel GLRA1 mutation P366L, located in the TM3-4 loop, showed normal surface expression but reduced chloride currents, and accelerated whole-cell desensitization observed in whole-cell recordings. At the single-channel level, we observed reduced unitary conductance accompanied by spontaneous opening events in the absence of extracellular glycine. Using peptide microarrays and tandem MS-based analysis methods, we show that the proline-rich stretch surrounding P366 mediates binding to syndapin I, an F-BAR domain protein involved in membrane remodeling. The disruption of the noncanonical Src homology 3 recognition motif by P366L reduces syndapin I binding. These data suggest that the GlyRα1 subunit interacts with intracellular binding partners and may therefore play a role in receptor trafficking or synaptic anchoring, a function thus far only ascribed to the GlyRß subunit. Hence, the P366L GlyRα1 variant exhibits a unique set of properties that cumulatively affect GlyR functionality and thus might explain the neuropathological mechanism underlying hyperekplexia in the mutant carriers. P366L is the first dominant GLRA1 mutation identified within the GlyRα1 TM3-4 loop that affects GlyR physiology without altering protein expression at the whole-cell and surface levels.SIGNIFICANCE STATEMENT We show that the intracellular domain of the inhibitory glycine receptor α1 subunit contributes to trafficking and synaptic anchoring. A proline-rich stretch in this receptor domain forms a noncanonical recognition motif important for the interaction with syndapin I (PACSIN1). The disruption of this motif, as present in a human patient with hyperekplexia led to impaired syndapin I binding. Functional analysis revealed that the altered proline-rich stretch determines several functional physiological parameters of the ion channel (e.g., faster whole-cell desensitization) reduced unitary conductance and spontaneous opening events. Thus, the proline-rich stretch from the glycine receptor α1 subunit represents a multifunctional intracellular protein motif.


Subject(s)
Receptors, Glycine/genetics , Receptors, Glycine/metabolism , Stiff-Person Syndrome/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Motifs , Animals , Humans , Mutation , Protein Binding/genetics , Protein Structure, Quaternary , Protein Transport/genetics , Receptors, Glycine/chemistry
17.
Cereb Cortex ; 30(8): 4306-4324, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32147734

ABSTRACT

Schizophrenia is associated with cognitive and behavioral dysfunctions thought to reflect imbalances in neurotransmission systems. Recent screenings suggested that lack of (functional) syndapin I (PACSIN1) may be linked to schizophrenia. We therefore studied syndapin I KO mice to address the suggested causal relationship to schizophrenia and to analyze associated molecular, cellular, and neurophysiological defects. Syndapin I knockout (KO) mice developed schizophrenia-related behaviors, such as hyperactivity, reduced anxiety, reduced response to social novelty, and an exaggerated novel object response and exhibited defects in dendritic arborization in the cortex. Neuromorphogenic deficits were also observed for a schizophrenia-associated syndapin I mutant in cultured neurons and coincided with a lack of syndapin I-mediated membrane recruitment of cytoskeletal effectors. Syndapin I KO furthermore caused glutamatergic hypofunctions. Syndapin I regulated both AMPAR and NMDAR availabilities at synapses during basal synaptic activity and during synaptic plasticity-particularly striking were a complete lack of long-term potentiation and defects in long-term depression in syndapin I KO mice. These synaptic plasticity defects coincided with alterations of postsynaptic actin dynamics, synaptic GluA1 clustering, and GluA1 mobility. Both GluA1 and GluA2 were not appropriately internalized. Summarized, syndapin I KO led to schizophrenia-like behavior, and our analyses uncovered associated molecular and cellular mechanisms.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Brain/metabolism , Neuronal Plasticity/physiology , Schizophrenia/metabolism , Animals , Behavior, Animal/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism
18.
Biochem Soc Trans ; 48(1): 137-146, 2020 02 28.
Article in English | MEDLINE | ID: mdl-32104881

ABSTRACT

The formation of caveolae, bulb-shaped plasma membrane invaginations, requires the coordinated action of distinct lipid-interacting and -shaping proteins. The interdependence of caveolar structure and function has evoked substantial scientific interest given the association of human diseases with caveolar dysfunction. Model systems deficient of core components of caveolae, caveolins or cavins, did not allow for an explicit attribution of observed functional defects to the requirement of caveolar invagination as they lack both invaginated caveolae and caveolin proteins. Knockdown studies in cultured cells and recent knockout studies in mice identified an additional family of membrane-shaping proteins crucial for caveolar formation, syndapins (PACSINs) - BAR domain superfamily proteins characterized by crescent-shaped membrane binding interfaces recognizing and inducing distinct curved membrane topologies. Importantly, syndapin loss-of-function resulted exclusively in impairment of caveolar invagination without a reduction in caveolin or cavin at the plasma membrane, thereby allowing the specific role of the caveolar invagination to be unveiled. Muscle cells of syndapin III KO mice showed severe reductions of caveolae reminiscent of human caveolinopathies and were more vulnerable to membrane damage upon changes in membrane tensions. Consistent with the lack of syndapin III-dependent invaginated caveolae providing mechanoprotection by releasing membrane reservoirs through caveolar flattening, physical exercise of syndapin III KO mice resulted in pathological defects reminiscent of the clinical symptoms of human myopathies associated with caveolin 3 mutation suggesting that the ability of muscular caveolae to respond to mechanical forces is a key physiological process.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cardiomyopathies/physiopathology , Caveolae/metabolism , Muscular Diseases/physiopathology , Adaptor Proteins, Signal Transducing/genetics , Animals , Caveolins/genetics , Caveolins/metabolism , Gene Knockdown Techniques , HeLa Cells , Humans , Mice , Mice, Knockout , Mutation , NIH 3T3 Cells
19.
Cereb Cortex ; 29(10): 4263-4276, 2019 09 13.
Article in English | MEDLINE | ID: mdl-30541023

ABSTRACT

Brain functions are extremely sensitive to pH changes because of the pH-dependence of proteins involved in neuronal excitability and synaptic transmission. Here, we show that the Na+/H+ exchanger Nhe1, which uses the Na+ gradient to extrude H+, is expressed at both inhibitory and excitatory presynapses. We disrupted Nhe1 specifically in mice either in Emx1-positive glutamatergic neurons or in parvalbumin-positive cells, mainly GABAergic interneurons. While Nhe1 disruption in excitatory neurons had no effect on overall network excitability, mice with disruption of Nhe1 in parvalbumin-positive neurons displayed epileptic activity. From our electrophysiological analyses in the CA1 of the hippocampus, we conclude that the disruption in parvalbumin-positive neurons impairs the release of GABA-loaded vesicles, but increases the size of GABA quanta. The latter is most likely an indirect pH-dependent effect, as Nhe1 was not expressed in purified synaptic vesicles itself. Conclusively, our data provide first evidence that Nhe1 affects network excitability via modulation of inhibitory interneurons.


Subject(s)
CA1 Region, Hippocampal/physiology , Membrane Potentials , Presynaptic Terminals/physiology , Sodium-Hydrogen Exchanger 1/physiology , gamma-Aminobutyric Acid/physiology , Animals , Epilepsy/physiopathology , Female , GABAergic Neurons/physiology , Glutamic Acid/metabolism , Interneurons/physiology , Male , Mice, Inbred C57BL , Mice, Transgenic , Presynaptic Terminals/metabolism , Vesicular Glutamate Transport Protein 1/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism , gamma-Aminobutyric Acid/metabolism
20.
Biochem Biophys Res Commun ; 506(2): 355-360, 2018 11 25.
Article in English | MEDLINE | ID: mdl-30093111

ABSTRACT

Actin filament formation plays a pivotal role in the development, regeneration and modulation of the morphologies and physiological functions of subcellular compartments and entire cells. All of these processes require tight temporal and spatial control of F-actin assembly. Recent work has shed new light on the control of actin filament formation by Ca2+ as very fast, transient messenger allowing for defined responses to signal intensities spanning several orders of magnitude. Recent discoveries highlight that a small but rapidly growing set of actin nucleators and related proteins, i.e. factors that have the power to promote the formation of new actin filaments in cells, are tightly controlled by the Ca2+ sensor protein CaM. We here review the cellular functions and the molecular mechanisms that couple Ca2+ signaling to the cytoskeletal functions of these factors. This set of proteins currently includes one actin nucleator of the formin family (INF2), the WH2 domain-based actin nucleator Cobl and its ancestor protein Cobl-like as well as fesselin/synaptopodin-2/myopodin and myelin basic protein (MBP). Considering the mechanistic principles of Ca2+ control of actin filament formation unveiled thus far and the diverse cell biological processes involving Ca2+ signaling it is obvious that our understanding of the cell biological crosstalk of Ca2+ transients with the in part highly specialized actin cytoskeletal structures observed in different cell types is only at its infancy.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/metabolism , Calcium/metabolism , Calmodulin/metabolism , Microfilament Proteins/metabolism , Plasma Membrane Calcium-Transporting ATPases/metabolism , Actin Cytoskeleton/genetics , Actin Cytoskeleton/ultrastructure , Actins/chemistry , Actins/genetics , Animals , Calcium Signaling , Calmodulin/genetics , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Formins , Gene Expression Regulation , Humans , Ion Transport , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microfilament Proteins/genetics , Myelin Basic Protein/genetics , Myelin Basic Protein/metabolism , Plasma Membrane Calcium-Transporting ATPases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...