Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Int J Nanomedicine ; 19: 3123-3142, 2024.
Article in English | MEDLINE | ID: mdl-38585474

ABSTRACT

Purpose: To study whether the absence of laminar shear stress (LSS) enables the uptake of very small superparamagnetic iron oxide nanoparticles (VSOP) in endothelial cells by altering the composition, size, and barrier function of the endothelial surface layer (ESL). Methods and Results: A quantitative particle exclusion assay with living human umbilical endothelial cells using spinning disc confocal microscopy revealed that the dimension of the ESL was reduced in cells cultivated in the absence of LSS. By combining gene expression analysis, flow cytometry, high pressure freezing/freeze substitution immuno-transmission electron microscopy, and confocal laser scanning microscopy, we investigated changes in ESL composition. We found that increased expression of the hyaluronan receptor CD44 by absence of shear stress did not affect the uptake rate of VSOPs. We identified collagen as a previously neglected component of ESL that contributes to its barrier function. Experiments with inhibitor halofuginone and small interfering RNA (siRNA) demonstrated that suppression of collagen expression facilitates VSOP uptake in endothelial cells grown under LSS. Conclusion: The absence of laminar shear stress disturbs the barrier function of the ESL, facilitating membrane accessibility and endocytic uptake of VSOP. Collagen, a previously neglected component of ESL, contributes to its barrier function.


Subject(s)
Endothelial Cells , Magnetic Iron Oxide Nanoparticles , Humans , Endothelial Cells/metabolism , Endothelium , Gene Expression Profiling , Collagen/metabolism , Stress, Mechanical , Cells, Cultured
2.
Cardiovasc Res ; 120(6): 644-657, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38309955

ABSTRACT

AIMS: Virus infection triggers inflammation and, may impose nutrient shortage to the heart. Supported by type I interferon (IFN) signalling, cardiomyocytes counteract infection by various effector processes, with the IFN-stimulated gene of 15 kDa (ISG15) system being intensively regulated and protein modification with ISG15 protecting mice Coxsackievirus B3 (CVB3) infection. The underlying molecular aspects how the ISG15 system affects the functional properties of respective protein substrates in the heart are unknown. METHODS AND RESULTS: Based on the protective properties due to protein ISGylation, we set out a study investigating CVB3-infected mice in depth and found cardiac atrophy with lower cardiac output in ISG15-/- mice. By mass spectrometry, we identified the protein targets of the ISG15 conjugation machinery in heart tissue and explored how ISGylation affects their function. The cardiac ISGylome showed a strong enrichment of ISGylation substrates within glycolytic metabolic processes. Two control enzymes of the glycolytic pathway, hexokinase 2 (HK2) and phosphofructokinase muscle form (PFK1), were identified as bona fide ISGylation targets during infection. In an integrative approach complemented with enzymatic functional testing and structural modelling, we demonstrate that protein ISGylation obstructs the activity of HK2 and PFK1. Seahorse-based investigation of glycolysis in cardiomyocytes revealed that, by conjugating proteins, the ISG15 system prevents the infection-/IFN-induced up-regulation of glycolysis. We complemented our analysis with proteomics-based advanced computational modelling of cardiac energy metabolism. Our calculations revealed an ISG15-dependent preservation of the metabolic capacity in cardiac tissue during CVB3 infection. Functional profiling of mitochondrial respiration in cardiomyocytes and mouse heart tissue by Seahorse technology showed an enhanced oxidative activity in cells with a competent ISG15 system. CONCLUSION: Our study demonstrates that ISG15 controls critical nodes in cardiac metabolism. ISG15 reduces the glucose demand, supports higher ATP production capacity in the heart, despite nutrient shortage in infection, and counteracts cardiac atrophy and dysfunction.


Subject(s)
Coxsackievirus Infections , Cytokines , Energy Metabolism , Glycolysis , Mitochondria, Heart , Myocytes, Cardiac , Ubiquitins , Animals , Humans , Male , Coxsackievirus Infections/metabolism , Coxsackievirus Infections/virology , Coxsackievirus Infections/genetics , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Enterovirus B, Human/pathogenicity , Enterovirus B, Human/metabolism , Host-Pathogen Interactions , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/virology , Myocytes, Cardiac/pathology , Protein Processing, Post-Translational , Signal Transduction , Ubiquitins/metabolism , Ubiquitins/genetics
3.
Viruses ; 14(4)2022 04 07.
Article in English | MEDLINE | ID: mdl-35458499

ABSTRACT

Enteroviruses (EV) are implicated in an extensive range of clinical manifestations, such as pancreatic failure, cardiovascular disease, hepatitis, and meningoencephalitis. We recently reported on the biochemical properties of the highly conserved cysteine residue at position 38 (C38) of enteroviral protein 3A and demonstrated a C38-mediated homodimerization of the Coxsackievirus B3 protein 3A (CVB3-3A) that resulted in its profound stabilization. Here, we show that residue C38 of protein 3A supports the replication of CVB3, a clinically relevant member of the enterovirus genus. The infection of HeLa cells with protein 3A cysteine 38 to alanine mutants (C38A) attenuates virus replication, resulting in comparably lower virus particle formation. Consistently, in a mouse infection model, the enhanced virus propagation of CVB3-3A wt in comparison to the CVB3-3A[C38A] mutant was confirmed and found to promote severe liver tissue damage. In contrast, infection with the CVB3-3A[C38A] mutant mitigated hepatic tissue injury and ameliorated the signs of systemic inflammatory responses, such as hypoglycemia and hypothermia. Based on these data and our previous report on the C38-mediated stabilization of the CVB3-3A protein, we conclude that the highly conserved amino acid C38 in protein 3A enhances the virulence of CVB3.


Subject(s)
Coxsackievirus Infections , Enterovirus Infections , Enterovirus , Animals , Cysteine , Enterovirus B, Human/physiology , HeLa Cells , Humans , Mice , Virulence , Virus Replication
4.
PLoS Biol ; 20(3): e3001503, 2022 03.
Article in English | MEDLINE | ID: mdl-35312684

ABSTRACT

Recent advances in imaging technology have highlighted that scaffold proteins and receptors are arranged in subsynaptic nanodomains. The synaptic membrane-associated guanylate kinase (MAGUK) scaffold protein membrane protein palmitoylated 2 (MPP2) is a component of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor-associated protein complexes and also binds to the synaptic cell adhesion molecule SynCAM 1. Using superresolution imaging, we show that-like SynCAM 1-MPP2 is situated at the periphery of the postsynaptic density (PSD). In order to explore MPP2-associated protein complexes, we used a quantitative comparative proteomics approach and identified multiple γ-aminobutyric acid (GABA)A receptor subunits among novel synaptic MPP2 interactors. In line with a scaffold function for MPP2 in the assembly and/or modulation of intact GABAA receptors, manipulating MPP2 expression had effects on inhibitory synaptic transmission. We further show that GABAA receptors are found together with MPP2 in a subset of dendritic spines and thus highlight MPP2 as a scaffold that serves as an adaptor molecule, linking peripheral synaptic elements critical for inhibitory regulation to central structures at the PSD of glutamatergic synapses.


Subject(s)
Membrane Proteins , Post-Synaptic Density , Membrane Proteins/metabolism , Post-Synaptic Density/metabolism , Receptors, AMPA/metabolism , Receptors, GABA-A , Synapses/metabolism
5.
Nano Lett ; 22(7): 2682-2690, 2022 04 13.
Article in English | MEDLINE | ID: mdl-35290738

ABSTRACT

Several variants of multicolor single-molecule localization microscopy (SMLM) have been developed to resolve the spatial relationship of nanoscale structures in biological samples. The oligonucleotide-based SMLM approach "DNA-PAINT" robustly achieves nanometer localization precision and can be used to count binding sites within nanostructures. However, multicolor DNA-PAINT has primarily been realized by "Exchange-PAINT", which requires sequential exchange of the imaging solution and thus leads to extended acquisition times. To alleviate the need for fluid exchange and to speed up the acquisition of current multichannel DNA-PAINT, we here present a novel approach that combines DNA-PAINT with simultaneous multicolor acquisition using spectral demixing (SD). By using newly designed probes and a novel multichannel registration procedure, we achieve simultaneous multicolor SD-DNA-PAINT with minimal crosstalk. We demonstrate high localization precision (3-6 nm) and multicolor registration of dual- and triple-color SD-DNA-PAINT by resolving patterns on DNA origami nanostructures and cellular structures.


Subject(s)
Nanostructures , Single Molecule Imaging , DNA/chemistry , Microscopy, Fluorescence/methods , Oligonucleotides/chemistry , Single Molecule Imaging/methods
6.
FEBS J ; 289(13): 3826-3838, 2022 07.
Article in English | MEDLINE | ID: mdl-35066984

ABSTRACT

RNA viruses in the Picornaviridae family express a large 250 kDa viral polyprotein that is processed by virus-encoded proteinases into mature functional proteins with specific functions for virus replication. One of these proteins is the highly conserved enteroviral transmembrane protein 3A that assists in reorganizing cellular membranes associated with the Golgi apparatus. Here, we studied the molecular properties of the Coxsackievirus B3 (CVB3) protein 3A with regard to its dimerization and its functional stability. By applying mutational analysis and biochemical characterization, we demonstrate that protein 3A forms DTT-sensitive disulfide-linked dimers via a conserved cytosolic cysteine residue at position 38 (Cys38). Homodimerization of CVB3 protein 3A via Cys38 leads to profound stabilization of the protein, whereas a C38A mutation promotes a rapid proteasome-dependent elimination of its monomeric form. The lysosomotropic agent chloroquine (CQ) exerted only minor stabilizing effects on the 3A monomer but resulted in enrichment of the homodimer. Our experimental data demonstrate that disulfide linkages via a highly conserved Cys-residue in enteroviral protein 3A have an important role in the dimerization of this viral protein, thereby preserving its stability and functional integrity.


Subject(s)
Disulfides , Enterovirus B, Human , Dimerization , Disulfides/metabolism , Enterovirus B, Human/genetics , Enterovirus B, Human/metabolism , HeLa Cells , Humans , Viral Proteins/metabolism , Virus Replication
7.
Mol Metab ; 45: 101151, 2021 03.
Article in English | MEDLINE | ID: mdl-33359402

ABSTRACT

OBJECTIVE: Hormone secretion from metabolically active tissues, such as pancreatic islets, is governed by specific and highly regulated signaling pathways. Defects in insulin secretion are among the major causes of diabetes. The molecular mechanisms underlying regulated insulin secretion are, however, not yet completely understood. In this work, we studied the role of the GTPase ARFRP1 on insulin secretion from pancreatic ß-cells. METHODS: A ß-cell-specific Arfrp1 knockout mouse was phenotypically characterized. Pulldown experiments and mass spectrometry analysis were employed to screen for new ARFRP1-interacting proteins. Co-immunoprecipitation assays as well as super-resolution microscopy were applied for validation. RESULTS: The GTPase ARFRP1 interacts with the Golgi-associated PDZ and coiled-coil motif-containing protein (GOPC). Both proteins are co-localized at the trans-Golgi network and regulate the first and second phase of insulin secretion by controlling the plasma membrane localization of the SNARE protein SNAP25. Downregulation of both GOPC and ARFRP1 in Min6 cells interferes with the plasma membrane localization of SNAP25 and enhances its degradation, thereby impairing glucose-stimulated insulin release from ß-cells. In turn, overexpression of SNAP25 as well as GOPC restores insulin secretion in islets from ß-cell-specific Arfrp1 knockout mice. CONCLUSION: Our results identify a hitherto unrecognized pathway required for insulin secretion at the level of trans-Golgi sorting.


Subject(s)
ADP-Ribosylation Factors/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Golgi Apparatus/metabolism , Golgi Matrix Proteins/metabolism , Insulin Secretion/physiology , Insulin-Secreting Cells/metabolism , ADP-Ribosylation Factors/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Female , Golgi Matrix Proteins/genetics , HeLa Cells , Humans , Male , Mice , Mice, Knockout , Protein Transport , SNARE Proteins/metabolism , trans-Golgi Network/metabolism
8.
Cell Rep Methods ; 1(5): 100068, 2021 09 27.
Article in English | MEDLINE | ID: mdl-35474672

ABSTRACT

Advances in single-cell RNA sequencing have allowed for the identification of cellular subtypes on the basis of quantification of the number of transcripts in each cell. However, cells might also differ in the spatial distribution of molecules, including RNAs. Here, we present DypFISH, an approach to quantitatively investigate the subcellular localization of RNA and protein. We introduce a range of analytical techniques to interrogate single-molecule RNA fluorescence in situ hybridization (smFISH) data in combination with protein immunolabeling. DypFISH is suited to study patterns of clustering of molecules, the association of mRNA-protein subcellular localization with microtubule organizing center orientation, and interdependence of mRNA-protein spatial distributions. We showcase how our analytical tools can achieve biological insights by utilizing cell micropatterning to constrain cellular architecture, which leads to reduction in subcellular mRNA distribution variation, allowing for the characterization of their localization patterns. Furthermore, we show that our method can be applied to physiological systems such as skeletal muscle fibers.


Subject(s)
Muscle Fibers, Skeletal , RNA , RNA/genetics , In Situ Hybridization, Fluorescence/methods , RNA, Messenger/genetics , Muscle Fibers, Skeletal/metabolism , Protein Transport
9.
EMBO Rep ; 21(3): e48530, 2020 03 04.
Article in English | MEDLINE | ID: mdl-32003148

ABSTRACT

Pathological aggregation of amyloid-ß (Aß) is a main hallmark of Alzheimer's disease (AD). Recent genetic association studies have linked innate immune system actions to AD development, and current evidence suggests profound gender differences in AD pathogenesis. Here, we characterise gender-specific pathologies in the APP23 AD-like mouse model and find that female mice show stronger amyloidosis and astrogliosis compared with male mice. We tested the gender-specific effect of lack of IL12p40, the shared subunit of interleukin (IL)-12 and IL-23, that we previously reported to ameliorate pathology in APPPS1 mice. IL12p40 deficiency gender specifically reduces Aß plaque burden in male APP23 mice, while in female mice, a significant reduction in soluble Aß1-40 without changes in Aß plaque burden is seen. Similarly, plasma and brain cytokine levels are altered differently in female versus male APP23 mice lacking IL12p40, while glial properties are unchanged. These data corroborate the therapeutic potential of targeting IL-12/IL-23 signalling in AD, but also highlight the importance of gender considerations when studying the role of the immune system and AD.


Subject(s)
Alzheimer Disease , Interleukin-12/deficiency , Interleukin-23 Subunit p19/deficiency , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Disease Models, Animal , Female , Interleukin-12/genetics , Interleukin-12 Subunit p40/deficiency , Interleukin-12 Subunit p40/genetics , Interleukin-23 Subunit p19/genetics , Male , Mice , Mice, Transgenic , Plaque, Amyloid
10.
Sci Adv ; 5(11): eaax5775, 2019 11.
Article in English | MEDLINE | ID: mdl-31807703

ABSTRACT

Clathrin-mediated endocytosis, an essential process for plasma membrane homeostasis and cell signaling, is characterized by stunning heterogeneity in the size and lifetime of clathrin-coated endocytic pits (CCPs). If and how CCP growth and lifetime are coupled and how this relates to their physiological function are unknown. We combine computational modeling, automated tracking of CCP dynamics, electron microscopy, and functional rescue experiments to demonstrate that CCP growth and lifetime are closely correlated and mechanistically linked by the early-acting endocytic F-BAR protein FCHo2. FCHo2 assembles at the rim of CCPs to control CCP growth and lifetime by coupling the invagination of early endocytic intermediates to clathrin lattice assembly. Our data suggest a mechanism for the nanoscale control of CCP growth and stability that may similarly apply to other metastable structures in cells.


Subject(s)
Cell Membrane/metabolism , Clathrin/metabolism , Endocytosis/physiology , Animals , Biological Transport, Active/physiology , COS Cells , Cell Membrane/ultrastructure , Chlorocebus aethiops , HEK293 Cells , HeLa Cells , Humans
11.
Cell Rep ; 29(7): 2028-2040.e8, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31722215

ABSTRACT

In developing neurons, phosphoinositide 3-kinases (PI3Ks) control axon growth and branching by positively regulating PI3K/PI(3,4,5)P3, but how neurons are able to generate sufficient PI(3,4,5)P3 in the presence of high levels of the antagonizing phosphatase PTEN is difficult to reconcile. We find that normal axon morphogenesis involves homeostasis of elongation and branch growth controlled by accumulation of PI(3,4,5)P3 through PTEN inhibition. We identify a plasma membrane-localized protein-protein interaction of PTEN with plasticity-related gene 2 (PRG2). PRG2 stabilizes membrane PI(3,4,5)P3 by inhibiting PTEN and localizes in nanoclusters along axon membranes when neurons initiate their complex branching behavior. We demonstrate that PRG2 is both sufficient and necessary to account for the ability of neurons to generate axon filopodia and branches in dependence on PI3K/PI(3,4,5)P3 and PTEN. Our data indicate that PRG2 is part of a neuronal growth program that induces collateral branch growth in axons by conferring local inhibition of PTEN.


Subject(s)
Axons/metabolism , Membrane Proteins/metabolism , PTEN Phosphohydrolase/metabolism , Animals , COS Cells , Chlorocebus aethiops , Female , Humans , Male , Membrane Proteins/genetics , Mice , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol Phosphates/genetics , Phosphatidylinositol Phosphates/metabolism
12.
Elife ; 82019 09 19.
Article in English | MEDLINE | ID: mdl-31535974

ABSTRACT

All synapses require fusion-competent vesicles and coordinated Ca2+-secretion coupling for neurotransmission, yet functional and anatomical properties are diverse across different synapse types. We show that the presynaptic protein RIM-BP2 has diversified functions in neurotransmitter release at different central murine synapses and thus contributes to synaptic diversity. At hippocampal pyramidal CA3-CA1 synapses, RIM-BP2 loss has a mild effect on neurotransmitter release, by only regulating Ca2+-secretion coupling. However, at hippocampal mossy fiber synapses, RIM-BP2 has a substantial impact on neurotransmitter release by promoting vesicle docking/priming and vesicular release probability via stabilization of Munc13-1 at the active zone. We suggest that differences in the active zone organization may dictate the role a protein plays in synaptic transmission and that differences in active zone architecture is a major determinant factor in the functional diversity of synapses.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Mossy Fibers, Hippocampal/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Synaptic Vesicles/metabolism , Animals , Mice , Neurotransmitter Agents/metabolism
13.
J Am Soc Nephrol ; 30(6): 946-961, 2019 06.
Article in English | MEDLINE | ID: mdl-31097611

ABSTRACT

BACKGROUND: Antagonists of the V1a vasopressin receptor (V1aR) are emerging as a strategy for slowing progression of CKD. Physiologically, V1aR signaling has been linked with acid-base homeostasis, but more detailed information is needed about renal V1aR distribution and function. METHODS: We used a new anti-V1aR antibody and high-resolution microscopy to investigate Va1R distribution in rodent and human kidneys. To investigate whether V1aR activation promotes urinary H+ secretion, we used a V1aR agonist or antagonist to evaluate V1aR function in vasopressin-deficient Brattleboro rats, bladder-catheterized mice, isolated collecting ducts, and cultured inner medullary collecting duct (IMCD) cells. RESULTS: Localization of V1aR in rodent and human kidneys produced a basolateral signal in type A intercalated cells (A-ICs) and a perinuclear to subapical signal in type B intercalated cells of connecting tubules and collecting ducts. Treating vasopressin-deficient Brattleboro rats with a V1aR agonist decreased urinary pH and tripled net acid excretion; we observed a similar response in C57BL/6J mice. In contrast, V1aR antagonist did not affect urinary pH in normal or acid-loaded mice. In ex vivo settings, basolateral treatment of isolated perfused medullary collecting ducts with the V1aR agonist or vasopressin increased intracellular calcium levels in ICs and decreased luminal pH, suggesting V1aR-dependent calcium release and stimulation of proton-secreting proteins. Basolateral treatment of IMCD cells with the V1aR agonist increased apical abundance of vacuolar H+-ATPase in A-ICs. CONCLUSIONS: Our results show that activation of V1aR contributes to urinary acidification via H+ secretion by A-ICs, which may have clinical implications for pharmacologic targeting of V1aR.


Subject(s)
Acid-Base Equilibrium/drug effects , Receptors, Vasopressin/drug effects , Vasopressins/pharmacology , Acid-Base Equilibrium/genetics , Animals , Cells, Cultured/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Fluorescent Antibody Technique , HEK293 Cells/drug effects , HEK293 Cells/metabolism , Humans , Hydrogen-Ion Concentration/drug effects , Immunohistochemistry , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/metabolism , Male , Mice, Inbred C57BL , Rats, Brattleboro , Rats, Wistar , Real-Time Polymerase Chain Reaction/methods , Receptors, Vasopressin/genetics , Sensitivity and Specificity , Urinalysis/methods
14.
J Cell Sci ; 132(3)2019 02 01.
Article in English | MEDLINE | ID: mdl-30709970

ABSTRACT

Compartmentalization of membrane transport and signaling processes is of pivotal importance to eukaryotic cell function. While plasma membrane compartmentalization and dynamics are well known to depend on the scaffolding function of septin GTPases, the roles of septins at intracellular membranes have remained largely elusive. Here, we show that the structural and functional integrity of the Golgi depends on its association with a septin 1 (SEPT1)-based scaffold, which promotes local microtubule nucleation and positioning of the Golgi. SEPT1 function depends on the Golgi matrix protein GM130 (also known as GOLGA2) and on centrosomal proteins, including CEP170 and components of γ-tubulin ring complex (γ-Turc), to facilitate the perinuclear concentration of Golgi membranes. Accordingly, SEPT1 depletion triggers a massive fragmentation of the Golgi ribbon, thereby compromising anterograde membrane traffic at the level of the Golgi.


Subject(s)
Autoantigens/genetics , Centrosome/metabolism , Golgi Apparatus/metabolism , Membrane Proteins/genetics , Microtubule-Associated Proteins/genetics , Microtubules/metabolism , Septins/genetics , 3T3-L1 Cells , Animals , Autoantigens/metabolism , Biological Transport , Cell Compartmentation , Cell Line , Centrosome/ultrastructure , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Gene Expression Regulation , Golgi Apparatus/ultrastructure , HEK293 Cells , HeLa Cells , Humans , Jurkat Cells/metabolism , Jurkat Cells/ultrastructure , Membrane Proteins/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Microtubules/ultrastructure , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Septins/antagonists & inhibitors , Septins/metabolism , Signal Transduction
15.
EMBO J ; 38(4)2019 02 15.
Article in English | MEDLINE | ID: mdl-30617086

ABSTRACT

Alzheimer's disease is characterized not only by extracellular amyloid plaques and neurofibrillary tangles, but also by microglia-mediated neuroinflammation. Recently, autophagy has been linked to the regulation of the inflammatory response. Thus, we investigated how an impairment of autophagy mediated by BECN1/Beclin1 reduction, as described in Alzheimer's disease patients, would influence cytokine production of microglia. Acutely stimulated microglia from Becn1+/- mice exhibited increased expression of IL-1beta and IL-18 compared to wild-type microglia. Becn1+/-APPPS1 mice also contained enhanced IL-1beta levels. The investigation of the IL-1beta/IL-18 processing pathway showed an elevated number of cells with inflammasomes and increased levels of NLRP3 and cleaved CASP1/Caspase1 in Becn1+/- microglia. Super-resolation microscopy revealed a very close association of NLRP3 aggregates and LC3-positive vesicles. Interestingly, CALCOCO2 colocalized with NLRP3 and its downregulation increased IL-1beta release. These data support the notion that selective autophagy can impact microglia activation by modulating IL-1beta and IL-18 production via NLRP3 degradation and thus present a mechanism how impaired autophagy could contribute to neuroinflammation in Alzheimer's disease.


Subject(s)
Autophagy , Beclin-1/physiology , Inflammation/immunology , Microglia/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Plaque, Amyloid/immunology , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/physiology , Animals , Autophagosomes , Cytokines/metabolism , Female , Inflammasomes , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Knockout , Microglia/metabolism , Microglia/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Presenilin-1/physiology
16.
PLoS One ; 13(4): e0193257, 2018.
Article in English | MEDLINE | ID: mdl-29617365

ABSTRACT

Inhibition of the phospholipid phosphatase and tumor suppressor PTEN leads to excessive polarized cell growth during directed cell migration and neurite outgrowth. These processes require the precise regulation of both the actin and microtubule cytoskeleton. While PTEN is known to regulate actin dynamics through phospholipid modulation, whether and how PTEN regulates microtubule dynamics is unknown. Here, we show that depletion of PTEN leads to elevated levels of stable and post-translationally modified (detyrosinated) microtubules in fibroblasts and developing neurons. Further, PTEN depletion enhanced axon outgrowth, which was rescued by reducing the level of detyrosinated microtubules. These data demonstrate a novel role of PTEN in regulating the microtubule cytoskeleton. They further show a novel function of detyrosinated microtubules in axon outgrowth. Specifically, PTEN suppresses axon outgrowth by down-regulating the level of detyrosinated microtubules. Our results suggest that PTEN's role in preventing excessive cell growth in cancerous and neurodevelopmental phenotypes is partially exerted by stabilization and detyrosination of the microtubule cytoskeleton.


Subject(s)
Microtubules/metabolism , Neuronal Outgrowth , Neurons/cytology , PTEN Phosphohydrolase/metabolism , Tyrosine/metabolism , Animals , Axons/metabolism , Down-Regulation , Fibroblasts/cytology , Fibroblasts/metabolism , Mice , NIH 3T3 Cells , Neurons/metabolism
17.
Curr Biol ; 27(19): 2999-3009.e9, 2017 Oct 09.
Article in English | MEDLINE | ID: mdl-28966089

ABSTRACT

The nucleus is the main microtubule-organizing center (MTOC) in muscle cells due to the accumulation of centrosomal proteins and microtubule (MT) nucleation activity at the nuclear envelope (NE) [1-4]. The relocalization of centrosomal proteins, including Pericentrin, Pcm1, and γ-tubulin, depends on Nesprin-1, an outer nuclear membrane (ONM) protein that connects the nucleus to the cytoskeleton via its N-terminal region [5-7]. Nesprins are also involved in the recruitment of kinesin to the NE and play a role in nuclear positioning in skeletal muscle cells [8-12]. However, a function for MT nucleation from the NE in nuclear positioning has not been established. Using the proximity-dependent biotin identification (BioID) method [13, 14], we found several centrosomal proteins, including Akap450, Pcm1, and Pericentrin, whose association with Nesprin-1α is increased in differentiated myotubes. We show that Nesprin-1α recruits Akap450 to the NE independently of kinesin and that Akap450, but not other centrosomal proteins, is required for MT nucleation from the NE. Furthermore, we demonstrate that this mechanism is disrupted in congenital muscular dystrophy patient myotubes carrying a nonsense mutation within the SYNE1 gene (23560 G>T) encoding Nesprin-1 [15, 16]. Finally, using computer simulation and cell culture systems, we provide evidence for a role of MT nucleation from the NE on nuclear spreading in myotubes. Our data thus reveal a novel function for Nesprin-1α/Nesprin-1 in nuclear positioning through recruitment of Akap450-mediated MT nucleation activity to the NE.


Subject(s)
A Kinase Anchor Proteins/genetics , Microtubule-Associated Proteins/genetics , Microtubules/metabolism , Nerve Tissue Proteins/genetics , Nuclear Envelope/metabolism , Nuclear Proteins/genetics , A Kinase Anchor Proteins/metabolism , Animals , Cell Line , Cytoskeletal Proteins , Female , HeLa Cells , Humans , Mice , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Rats
18.
Nat Commun ; 8: 15873, 2017 06 19.
Article in English | MEDLINE | ID: mdl-28627515

ABSTRACT

Clathrin-mediated endocytosis (CME) involves membrane-associated scaffolds of the bin-amphiphysin-rvs (BAR) domain protein family as well as the GTPase dynamin, and is accompanied and perhaps triggered by changes in local lipid composition. How protein recruitment, scaffold assembly and membrane deformation is spatiotemporally controlled and coupled to fission is poorly understood. We show by computational modelling and super-resolution imaging that phosphatidylinositol 3,4-bisphosphate [PI(3,4)P2] synthesis within the clathrin-coated area of endocytic intermediates triggers selective recruitment of the PX-BAR domain protein SNX9, as a result of complex interactions of endocytic proteins competing for phospholipids. The specific architecture induces positioning of SNX9 at the invagination neck where its self-assembly regulates membrane constriction, thereby providing a template for dynamin fission. These data explain how lipid conversion at endocytic pits couples local membrane constriction to fission. Our work demonstrates how computational modelling and super-resolution imaging can be combined to unravel function and mechanisms of complex cellular processes.


Subject(s)
Cell Membrane/metabolism , Phosphatidylinositol Phosphates/biosynthesis , Sorting Nexins/metabolism , Transport Vesicles/metabolism , Animals , Binding Sites , COS Cells , Cell Membrane/chemistry , Chlorocebus aethiops , Clathrin-Coated Vesicles/metabolism , Dynamins/metabolism , HeLa Cells , Humans , Models, Theoretical , Nuclear Proteins/metabolism , Phospholipids/metabolism , Protein Domains , Sorting Nexins/chemistry , Sorting Nexins/genetics , Surface Plasmon Resonance , Transcription Factors/metabolism
19.
Nat Neurosci ; 20(2): 209-218, 2017 02.
Article in English | MEDLINE | ID: mdl-27941788

ABSTRACT

The skin is equipped with specialized mechanoreceptors that allow the perception of the slightest brush. Indeed, some mechanoreceptors can detect even nanometer-scale movements. Movement is transformed into electrical signals via the gating of mechanically activated ion channels at sensory endings in the skin. The sensitivity of Piezo mechanically gated ion channels is controlled by stomatin-like protein-3 (STOML3), which is required for normal mechanoreceptor function. Here we identify small-molecule inhibitors of STOML3 oligomerization that reversibly reduce the sensitivity of mechanically gated currents in sensory neurons and silence mechanoreceptors in vivo. STOML3 inhibitors in the skin also reversibly attenuate fine touch perception in normal mice. Under pathophysiological conditions following nerve injury or diabetic neuropathy, the slightest touch can produce pain, and here STOML3 inhibitors can reverse mechanical hypersensitivity. Thus, small molecules applied locally to the skin can be used to modulate touch and may represent peripherally available drugs to treat tactile-driven pain following neuropathy.


Subject(s)
Hypersensitivity/metabolism , Ion Channels/metabolism , Mechanoreceptors/metabolism , Membrane Proteins/antagonists & inhibitors , Nerve Tissue Proteins/antagonists & inhibitors , Sensory Receptor Cells/metabolism , Animals , Ganglia, Spinal/metabolism , Hypersensitivity/drug therapy , Mechanotransduction, Cellular/drug effects , Mechanotransduction, Cellular/physiology , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Sensory Receptor Cells/drug effects , Skin/innervation , Touch/physiology
20.
J Biophotonics ; 9(1-2): 161-70, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25973835

ABSTRACT

Precise multicolor single molecule localization-based microscopy (SMLM) requires bright probes with compatible photo-chemical and spectral properties to resolve distinct molecular species at the nanoscale. The accuracy of multicolor SMLM is further challenged by color channel crosstalk and chromatic alignment errors. These constrains limit the applicability of known reversibly switchable organic dyes for optimized multicolor SMLM. Here, we tested 28 commercially available dyes for their suitability to super-resolve a known cellular nanostructure. We identified eight novel dyes in different spectral regimes that enable high quality dSTORM imaging. Among those, the spectrally close dyes CF647 and CF680 comprise an optimal dye pair for spectral demixing-based, registration free multicolor dSTORM with low crosstalk. Combining this dye pair with the separately excited CF568 we performed 3-color dSTORM to image the relative nanoscale distribution of components of the endocytic machinery and the cytoskeleton.


Subject(s)
Fluorescent Dyes/metabolism , Microscopy/methods , Animals , Biological Transport , Carbocyanines/metabolism , Cell Membrane/metabolism , Color , Cytoskeleton/metabolism , Mice , NIH 3T3 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...