Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Mol Cell ; 82(15): 2858-2870.e8, 2022 08 04.
Article in English | MEDLINE | ID: mdl-35732190

ABSTRACT

The tolerance of amino acid starvation is fundamental to robust cellular fitness. Asparagine depletion is lethal to some cancer cells, a vulnerability that can be exploited clinically. We report that resistance to asparagine starvation is uniquely dependent on an N-terminal low-complexity domain of GSK3α, which its paralog GSK3ß lacks. In response to depletion of specific amino acids, including asparagine, leucine, and valine, this domain mediates supramolecular assembly of GSK3α with ubiquitin-proteasome system components in spatially sequestered cytoplasmic bodies. This effect is independent of mTORC1 or GCN2. In normal cells, GSK3α promotes survival during essential amino acid starvation. In human leukemia, GSK3α body formation predicts asparaginase resistance, and sensitivity to asparaginase combined with a GSK3α inhibitor. We propose that GSK3α body formation provides a cellular mechanism to maximize the catalytic efficiency of proteasomal protein degradation in response to amino acid starvation, an adaptive response co-opted by cancer cells for asparaginase resistance.


Subject(s)
Asparaginase , Leukemia , Amino Acids/metabolism , Asparaginase/genetics , Asparaginase/metabolism , Asparaginase/pharmacology , Asparagine , Humans , Protein Serine-Threonine Kinases
2.
J Mol Cell Cardiol ; 185: 26-37, 2023 12.
Article in English | MEDLINE | ID: mdl-37797718

ABSTRACT

Hypertrophic cardiomyopathy (HCM) is the most prevalent inherited cardiac disease. Up to 40% of cases are associated with heterozygous mutations in myosin binding protein C (cMyBP-C, MYBPC3). Most of these mutations lead to premature termination codons (PTC) and patients show reduction of functional cMyBP-C. This so-called haploinsufficiency most likely contributes to disease development. We analyzed mechanisms underlying haploinsufficiency using cardiac tissue from HCM-patients with truncation mutations in MYBPC3 (MYBPC3trunc). We compared transcriptional activity, mRNA and protein expression to donor controls. To differentiate between HCM-specific and general hypertrophy-induced mechanisms we used patients with left ventricular hypertrophy due to aortic stenosis (AS) as an additional control. We show that cMyBP-C haploinsufficiency starts at the mRNA level, despite hypertrophy-induced increased transcriptional activity. Gene set enrichment analysis (GSEA) of RNA-sequencing data revealed an increased expression of NMD-components. Among them, Up-frameshift protein UPF3B, a regulator of NMD was upregulated in MYBPC3trunc patients and not in AS-patients. Strikingly, we show that in sarcomeres UPF3B but not UPF1 and UPF2 are localized to the Z-discs, the presumed location of sarcomeric protein translation. Our data suggest that cMyBP-C haploinsufficiency in HCM-patients is established by UPF3B-dependent NMD during the initial translation round at the Z-disc.


Subject(s)
Cardiomyopathy, Hypertrophic , Myocytes, Cardiac , Humans , Cardiomyopathy, Hypertrophic/metabolism , Haploinsufficiency , Hypertrophy/metabolism , Mutation , Myocytes, Cardiac/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism
3.
J Cell Sci ; 134(4)2021 02 19.
Article in English | MEDLINE | ID: mdl-33536244

ABSTRACT

Morphological remodeling of dendritic spines is critically involved in memory formation and depends on adhesion molecules. Serotonin receptors are also implicated in this remodeling, though the underlying mechanisms remain enigmatic. Here, we uncovered a signaling pathway involving the adhesion molecule L1CAM (L1) and serotonin receptor 5-HT4 (5-HT4R, encoded by HTR4). Using Förster resonance energy transfer (FRET) imaging, we demonstrated a physical interaction between 5-HT4R and L1, and found that 5-HT4R-L1 heterodimerization facilitates mitogen-activated protein kinase activation in a Gs-dependent manner. We also found that 5-HT4R-L1-mediated signaling is involved in G13-dependent modulation of cofilin-1 activity. In hippocampal neurons in vitro, the 5-HT4R-L1 pathway triggers maturation of dendritic spines. Thus, the 5-HT4R-L1 signaling module represents a previously unknown molecular pathway regulating synaptic remodeling.


Subject(s)
Neural Cell Adhesion Molecule L1 , Hippocampus , Neural Cell Adhesion Molecule L1/genetics , Neurons , Serotonin , Signal Transduction
4.
J Biol Chem ; 295(18): 5970-5983, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32184353

ABSTRACT

Barttin is the accessory subunit of the human ClC-K chloride channels, which are expressed in both the kidney and inner ear. Barttin promotes trafficking of the complex it forms with ClC-K to the plasma membrane and is involved in activating this channel. Barttin undergoes post-translational palmitoylation that is essential for its functions, but the enzyme(s) catalyzing this post-translational modification is unknown. Here, we identified zinc finger DHHC-type containing 7 (DHHC7) protein as an important barttin palmitoyl acyltransferase, whose depletion affected barttin palmitoylation and ClC-K-barttin channel activation. We investigated the functional role of barttin palmitoylation in vivo in Zdhhc7-/- mice. Although palmitoylation of barttin in kidneys of Zdhhc7-/- animals was significantly decreased, it did not pathologically alter kidney structure and functions under physiological conditions. However, when Zdhhc7-/- mice were fed a low-salt diet, they developed hyponatremia and mild metabolic alkalosis, symptoms characteristic of human Bartter syndrome (BS) type IV. Of note, we also observed decreased palmitoylation of the disease-causing R8L barttin variant associated with human BS type IV. Our results indicate that dysregulated DHHC7-mediated barttin palmitoylation appears to play an important role in chloride channel dysfunction in certain BS variants, suggesting that targeting DHHC7 activity may offer a potential therapeutic strategy for reducing hypertension.


Subject(s)
Acyltransferases/metabolism , Chloride Channels/metabolism , Palmitic Acid/metabolism , Protein Processing, Post-Translational , Acyltransferases/deficiency , Acyltransferases/genetics , Animals , Dogs , Gene Knockout Techniques , HEK293 Cells , Humans , Kidney/cytology , Kidney/metabolism , Madin Darby Canine Kidney Cells , Mice , Mutation , Phenotype
5.
Glia ; 69(12): 2798-2811, 2021 12.
Article in English | MEDLINE | ID: mdl-34388285

ABSTRACT

Recent achievements in indicator optimization and imaging techniques promote the advancement of functional imaging to decipher complex signaling processes in living cells, such as Ca2+ activity patterns. Astrocytes are important regulators of the brain network and well known for their highly complex morphology and spontaneous Ca2+ activity. However, the astrocyte community is lacking standardized methods to analyze and interpret Ca2+ activity recordings, hindering global comparisons. Here, we present a biophysically-based analytical concept for deciphering the complex spatio-temporal changes of Ca2+ biosensor fluorescence for understanding the underlying signaling mechanisms. We developed a pixel-based multi-threshold event detection (MTED) analysis of multidimensional data, which accounts for signal strength as an additional signaling dimension and provides the experimenter with a comprehensive toolbox for a differentiated and in-depth characterization of fluorescence signals. MTED was validated by analyzing astrocytic Ca2+ activity across Ca2+ indicators, imaging setups, and model systems from primary cell culture to awake, head-fixed mice. We identified extended Ca2+ activity at 25°C compared to 37°C physiological body temperature and dissected how neuronal activity shapes long-lasting astrocytic Ca2+ activity. Our MTED strategy, as a parameter-free approach, is easily transferrable to other fluorescent indicators and biosensors and embraces the additional dimensionality of signaling activity strength. It will also advance the definition of standardized procedures and parameters to improve comparability of research data and reports.


Subject(s)
Astrocytes , Calcium Signaling , Animals , Astrocytes/metabolism , Brain/diagnostic imaging , Brain/metabolism , Calcium/metabolism , Calcium Signaling/physiology , Mice , Neurons/metabolism
6.
Glia ; 69(4): 872-889, 2021 04.
Article in English | MEDLINE | ID: mdl-33156956

ABSTRACT

Astrocytes are an important component of the multipartite synapse and crucial for proper neuronal network function. Although small GTPases of the Rho family are powerful regulators of cellular morphology, the signaling modules of Rho-mediated pathways in astrocytes remain enigmatic. Here we demonstrated that the serotonin receptor 4 (5-HT4 R) is expressed in hippocampal astrocytes, both in vitro and in vivo. Through fluorescence microscopy, we established that 5-HT4 R activation triggered RhoA activity via Gα13 -mediated signaling, which boosted filamentous actin assembly, leading to morphological changes in hippocampal astrocytes. We investigated the effects of these 5-HT4 R-mediated changes in mixed cultures and in acute slices, in which 5-HT4 R was expressed exclusively in astrocytes. In both systems, 5-HT4 R-RhoA signaling changed glutamatergic synaptic transmission: It increased the frequency of miniature excitatory postsynaptic currents (mEPSCs) in mixed cultures and reduced the paired-pulse-ratio (PPR) of field excitatory postsynaptic potentials (fEPSPs) in acute slices. Overall, our present findings demonstrate that astrocytic 5-HT4 R-Gα13 -RhoA signaling is a previously unrecognized molecular pathway involved in the functional regulation of excitatory synaptic circuits.


Subject(s)
Astrocytes , Serotonin , Excitatory Postsynaptic Potentials , Hippocampus , Receptors, Serotonin/genetics , Synaptic Transmission
7.
J Cell Sci ; 132(16)2019 08 23.
Article in English | MEDLINE | ID: mdl-31371490

ABSTRACT

Protein-protein interaction is often investigated using quantitative molecular microscopy with Förster resonant energy transfer (FRET). Here, we combined 'linear unmixing FRET' (lux-FRET) with the simultaneous application of a FRET-based biosensor for cAMP to investigate the oligomerization between the 5-HT7 receptor (5-HT7R, also known as HTR7) and the 5-HT1A receptor (5-HT1AR, also known as HTR1A) and its importance for cAMP signaling. We found that the 5-HT7R not only stimulates cAMP production, but also forms hetero-oligomers with 5-HT1AR, which blocks the inhibitory effect of the latter. 5-HT7R signaling, however, is not affected by this hetero-oligomerization. By modeling the kinetics of intracellular cAMP level changes in relation to the 5-HT7R:5-HT1AR stoichiometry, we were able to decipher the complex signaling characteristics of endogenous serotonin receptors in cultured hippocampal neurons. Our findings indicate that serotonergic signaling is not only modulated by the concentration of an individual receptor but also by its specific interaction with other receptors in endogenous systems. We conclude that the regulated ratio of serotonin receptors in immature and mature neurons may be critically involved in both the onset and response to treatments of psychiatric diseases, such as anxiety and depression.


Subject(s)
Cyclic AMP/metabolism , Protein Multimerization , Receptor, Serotonin, 5-HT1A/metabolism , Receptors, Serotonin/metabolism , Second Messenger Systems , Animals , Cell Line, Tumor , Cyclic AMP/genetics , Mice , Receptor, Serotonin, 5-HT1A/genetics , Receptors, Serotonin/genetics
8.
J Mol Cell Cardiol ; 135: 79-89, 2019 10.
Article in English | MEDLINE | ID: mdl-31419438

ABSTRACT

The identification of spatiotemporally restricted Ca2+ signals, Ca2+ sparks, was instrumental for our understanding of cardiac Ca2+ homeostasis. High-speed 2D confocal imaging enables acquisition of such Ca2+ sparks with high-content information but their full appreciation is constrained by the lack of unbiased and easy-to-use analysis tools. We developed a software toolset for unbiased and automatic Ca2+ spark analysis for huge data sets of subcellular Ca2+ signals. iSpark was developed to be scanner and detector independent. In myocytes from hearts subjected to various degrees of hypertrophy we acquired >5.000.000 Ca2+ sparks from 14 mice. The iSpark-enabled analysis of this large Ca2+ spark data set showed that the highly organized distribution of Ca2+ sparks present in healthy cells disarrayed concomitant with the development of aberrant transverse tubules and disease severity. Thus, iSpark represents a versatile and universal tool for analyzing local Ca2+ signaling in healthy as well as diseased, aberrant local Ca2+ signal transduction. The results from the unbiased analysis of large data sets provide a deeper insight into possible mechanisms contributing to the onset and progression of cardiac diseases such as hypertrophy.


Subject(s)
Calcium Signaling , Image Processing, Computer-Assisted , Myocytes, Cardiac/metabolism , Software , Animals , Mice , Microscopy, Fluorescence , Myocytes, Cardiac/cytology
9.
Glia ; 67(2): 376-392, 2019 02.
Article in English | MEDLINE | ID: mdl-30450647

ABSTRACT

The tightly controlled processes of myelination and remyelination require the participation of the cytoskeleton. The reorganization of the cytoskeleton is controlled by small GTPases of the RhoA family. Here, we report that Vav3, a Rho GTPase regulating guanine nucleotide exchange factor (GEF) is involved in oligodendrocyte maturation, myelination and remyelination. When Vav3 was eliminated by genetic recombination, oligodendrocyte precursor cell (OPC) differentiation toward mature oligodendrocytes was accelerated. In contrast, Vav3-deficient oligodendrocytes displayed a reduced capacity to myelinate synthetic microfibers in vitro. Furthermore, remyelination was impaired in Vav3 knockout cerebellar slice cultures that were demyelinated by the addition of lysolecithin. In agreement with these observations, remyelination was compromised when the cuprizone model of myelin lesion was performed in Vav3-deficient mice. When Vav3-deficient oligodendrocytes were examined with Förster resonance energy transfer (FRET)-based biosensors, an altered activation profile of RhoA GTPases was revealed on the cellular level, which could be responsible for an impaired remyelination. Taken together, this study highlights Vav3 as a novel regulator of oligodendrocyte maturation and remyelination, suggesting that manipulation of the Vav3-dependent signaling pathway could help to improve myelin repair.


Subject(s)
Cell Differentiation/genetics , Leukoencephalopathies/pathology , Oligodendrocyte Precursor Cells/physiology , Proto-Oncogene Proteins c-vav/metabolism , Remyelination/genetics , Animals , Caspase 3/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Chelating Agents/toxicity , Cuprizone/toxicity , Disease Models, Animal , GTP Phosphohydrolases/metabolism , Ki-67 Antigen/metabolism , Leukoencephalopathies/chemically induced , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Basic Protein/metabolism , Nerve Fibers, Myelinated/pathology , Oligodendrocyte Precursor Cells/pathology , Organ Culture Techniques , Proto-Oncogene Proteins c-vav/genetics , Remyelination/drug effects , rhoA GTP-Binding Protein/metabolism
10.
J Cell Sci ; 125(Pt 10): 2486-99, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22357950

ABSTRACT

Serotonin receptors 5-HT(1A) and 5-HT(7) are highly coexpressed in brain regions implicated in depression. However, their functional interaction has not been established. In the present study we show that 5-HT(1A) and 5-HT(7) receptors form heterodimers both in vitro and in vivo. Foerster resonance energy transfer-based assays revealed that, in addition to heterodimers, homodimers composed either of 5-HT(1A) or 5-HT(7) receptors together with monomers coexist in cells. The highest affinity for complex formation was obtained for the 5-HT(7)-5-HT(7) homodimers, followed by the 5-HT(7)-5-HT(1A) heterodimers and 5-HT(1A)-5-HT(1A) homodimers. Functionally, heterodimerization decreases 5-HT(1A)-receptor-mediated activation of G(i) protein without affecting 5-HT(7)-receptor-mediated signalling. Moreover, heterodimerization markedly decreases the ability of the 5-HT(1A) receptor to activate G-protein-gated inwardly rectifying potassium channels in a heterologous system. The inhibitory effect on such channels was also preserved in hippocampal neurons, demonstrating a physiological relevance of heteromerization in vivo. In addition, heterodimerization is crucially involved in initiation of the serotonin-mediated 5-HT(1A) receptor internalization and also enhances the ability of the 5-HT(1A) receptor to activate the mitogen-activated protein kinases. Finally, we found that production of 5-HT(7) receptors in the hippocampus continuously decreases during postnatal development, indicating that the relative concentration of 5-HT(1A)-5-HT(7) heterodimers and, consequently, their functional importance undergoes pronounced developmental changes.


Subject(s)
Receptor, Serotonin, 5-HT1A/metabolism , Receptors, Serotonin/metabolism , Signal Transduction , Animals , Brain/metabolism , Cell Line, Tumor , Dimerization , Mice , Neurons/metabolism , Protein Binding , Protein Transport , Receptor, Serotonin, 5-HT1A/chemistry , Receptor, Serotonin, 5-HT1A/genetics , Receptors, Serotonin/chemistry , Receptors, Serotonin/genetics
11.
Redox Biol ; 73: 103170, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38692092

ABSTRACT

Adipose tissue-derived non-esterified saturated long-chain fatty acid palmitate (PA) decisively contributes to ß-cell demise in type 2 diabetes mellitus in part through the excessive generation of hydrogen peroxide (H2O2). The endoplasmic reticulum (ER) as the primary site of oxidative protein folding could represent a significant source of H2O2. Both ER-oxidoreductin-1 (ERO-1) isoenzymes, ERO-1α and ERO-1ß, catalyse oxidative protein folding within the ER, generating equimolar amounts of H2O2 for every disulphide bond formed. However, whether ERO-1-derived H2O2 constitutes a potential source of cytotoxic luminal H2O2 under lipotoxic conditions is still unknown. Here, we demonstrate that both ERO-1 isoforms are expressed in pancreatic ß-cells, but interestingly, PA only significantly induces ERO-1α. Its specific deletion significantly attenuates PA-mediated oxidative ER stress and subsequent ß-cell death by decreasing PA-mediated ER-luminal and mitochondrial H2O2 accumulation, by counteracting the dysregulation of ER Ca2+ homeostasis, and by mitigating the reduction of mitochondrial membrane potential and lowered ATP content. Moreover, ablation of ERO-1α alleviated PA-induced hyperoxidation of the ER redox milieu. Importantly, ablation of ERO-1α did not affect the insulin secretory capacity, the unfolded protein response, or ER redox homeostasis under steady-state conditions. The involvement of ERO-1α-derived H2O2 in PA-mediated ß-cell lipotoxicity was corroborated by the overexpression of a redox-active ERO-1α underscoring the proapoptotic activity of ERO-1α in pancreatic ß-cells. Overall, our findings highlight the critical role of ERO-1α-derived H2O2 in lipotoxic ER stress and ß-cell failure.


Subject(s)
Apoptosis , Endoplasmic Reticulum Stress , Hydrogen Peroxide , Insulin-Secreting Cells , Palmitates , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/drug effects , Endoplasmic Reticulum Stress/drug effects , Animals , Apoptosis/drug effects , Palmitates/metabolism , Palmitates/pharmacology , Hydrogen Peroxide/metabolism , Mice , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/drug effects , Oxidation-Reduction , Oxidative Stress/drug effects , Humans , Mitochondria/metabolism , Mitochondria/drug effects , Membrane Potential, Mitochondrial/drug effects
12.
Hum Mol Genet ; 20(24): 4865-78, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-21920940

ABSTRACT

Spinal muscular atrophy (SMA), a frequent neurodegenerative disease, is caused by reduced levels of functional survival of motoneuron (SMN) protein. SMN is involved in multiple pathways, including RNA metabolism and splicing as well as motoneuron development and function. Here we provide evidence for a major contribution of the Rho-kinase (ROCK) pathway in SMA pathogenesis. Using an in vivo protein interaction system based on SUMOylation of proteins, we found that SMN is directly interacting with profilin2a. Profilin2a binds to a stretch of proline residues in SMN, which is heavily impaired by a novel SMN2 missense mutation (S230L) derived from a SMA patient. In different SMA models, we identified differential phosphorylation of the ROCK-downstream targets cofilin, myosin-light chain phosphatase and profilin2a. We suggest that hyper-phosphorylation of profilin2a is the molecular link between SMN and the ROCK pathway repressing neurite outgrowth in neuronal cells. Finally, we found a neuron-specific increase in the F-/G-actin ratio that further support the role of actin dynamics in SMA pathogenesis.


Subject(s)
Muscular Atrophy, Spinal/metabolism , Profilins/metabolism , Signal Transduction , Survival of Motor Neuron 1 Protein/metabolism , rho-Associated Kinases/metabolism , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Disease Models, Animal , Gene Knockdown Techniques , Growth Cones/metabolism , Growth Cones/pathology , Humans , Mice , Models, Biological , Motor Neurons/metabolism , Motor Neurons/pathology , Muscular Atrophy, Spinal/pathology , Mutant Proteins/metabolism , Mutation, Missense/genetics , Neurites/metabolism , Phosphorylation , Protein Binding , Rats , Survival of Motor Neuron 1 Protein/genetics
13.
Cells ; 12(2)2023 01 15.
Article in English | MEDLINE | ID: mdl-36672265

ABSTRACT

Structural changes of astrocytes and their perisynaptic processes occur in response to various physiological and pathophysiological stimuli. They are thought to profoundly affect synaptic signalling and neuron-astrocyte communication. Understanding the causal relationship between astrocyte morphology changes and their functional consequences requires experimental tools to selectively manipulate astrocyte morphology. Previous studies indicate that RhoA-related signalling can play a major role in controlling astrocyte morphology, but the direct effect of increased RhoA activity has not been documented in vitro and in vivo. Therefore, we established a viral approach to manipulate astrocytic RhoA activity. We tested if and how overexpression of wild-type RhoA, of a constitutively active RhoA mutant (RhoA-CA), and of a dominant-negative RhoA variant changes the morphology of cultured astrocytes. We found that astrocytic expression of RhoA-CA induced robust cytoskeletal changes and a withdrawal of processes in cultured astrocytes. In contrast, overexpression of other RhoA variants led to more variable changes of astrocyte morphology. These induced morphology changes were reproduced in astrocytes of the hippocampus in vivo. Importantly, astrocytic overexpression of RhoA-CA did not alter the branching pattern of larger GFAP-positive processes of astrocytes. This indicates that a prolonged increase of astrocytic RhoA activity leads to a distinct morphological phenotype in vitro and in vivo, which is characterized by an isolated reduction of fine peripheral astrocyte processes in vivo. At the same time, we identified a promising experimental approach for investigating the functional consequences of astrocyte morphology changes.


Subject(s)
Astrocytes , Neurons , Astrocytes/metabolism , Cytoskeleton , Signal Transduction
14.
Biophys J ; 103(9): 1821-7, 2012 Nov 07.
Article in English | MEDLINE | ID: mdl-23199910

ABSTRACT

Förster resonance energy transfer (FRET) has become an important tool for analyzing different aspects of interactions among biological macromolecules in their native environments. FRET analysis has also been successfully applied to study the spatiotemporal regulation of various cellular processes using genetically encoded FRET-based biosensors. A variety of procedures have been described for measuring FRET efficiency or the relative abundance of donor-acceptor complexes, based on analysis of the donor fluorescence lifetime or the spectrally resolved fluorescence intensity. The latter methods are preferable if one wants to not only quantify the apparent FRET efficiencies but also calculate donor-acceptor stoichiometry and observe fast dynamic changes in the interactions among donor and acceptor molecules in live cells. This review focuses on a comparison of the available intensity-based approaches used to measure FRET. We discuss their strengths and weaknesses in terms of FRET quantification, and provide several examples of biological applications.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , Animals , Fluorescence , Humans , Microscopy, Confocal , Microscopy, Fluorescence , Optogenetics/methods
15.
Mol Pharmacol ; 82(3): 448-63, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22669805

ABSTRACT

Experimental evidence suggests that most members of class A G-protein coupled receptors (GPCRs) can form homomers and heteromers in addition to functioning as single monomers. In particular, serotonin (5-HT) receptors were shown to homodimerize and heterodimerize with other GPCRs, although the details and the physiological role of the oligomerization has not yet been fully elucidated. Here we used computational modeling of the 5-HT(1A) receptor monomer and dimer to predict residues important for dimerization. Based on these results, we carried out rationally designed site-directed mutagenesis. The ability of the mutants to dimerize was evaluated using different FRET-based approaches. The reduced levels of acceptor photobleaching-Förster resonance energy transfer (FRET) and the lower number of monomers participating in oligomers, as assessed by lux-FRET, confirmed the decreased ability of the mutants to dimerize and the involvement of the predicted contacts (Trp175(4.64), Tyr198(5.41), Arg151(4.40), and Arg152(4.41)) at the interface. This information was reintroduced as constraints for computational protein-protein docking to obtain a high-quality dimer model. Analysis of the refined model as well as molecular dynamics simulations of wild-type (WT) and mutant dimers revealed compensating interactions in dimers composed of WT and W175A mutant. This provides an explanation for the requirement of mutations of Trp175(4.64) in both homomers for disrupting dimerization. Our iterative computational-experimental study demonstrates that transmembrane domains TM4/TM5 can form an interaction interface in 5-HT(1A) receptor dimers and indicates that specific amino acid interactions maintain this interface. The mutants and the optimized model of the dimer structure may be used in functional studies of serotonin dimers.


Subject(s)
Membrane Proteins/chemistry , Membrane Proteins/metabolism , Receptor, Serotonin, 5-HT1A/chemistry , Receptor, Serotonin, 5-HT1A/metabolism , Animals , Cell Membrane/genetics , Cell Membrane/metabolism , Fluorescence Resonance Energy Transfer/methods , Glycosylation , Membrane Proteins/genetics , Mice , Mutagenesis, Site-Directed/methods , Mutation , Neuroblastoma/genetics , Neuroblastoma/metabolism , Photobleaching , Protein Multimerization , Protein Structure, Tertiary , Receptor, Serotonin, 5-HT1A/genetics , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Serotonin/genetics , Serotonin/metabolism , Transfection/methods , Tumor Cells, Cultured
16.
J Biol Chem ; 286(26): 23419-31, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21454618

ABSTRACT

Ratiometric measurements with FRET-based biosensors in living cells using a single fluorescence excitation wavelength are often affected by a significant ion sensitivity and the aggregation behavior of the FRET pair. This is an important problem for quantitative approaches. Here we report on the influence of physiological ion concentration changes on quantitative ratiometric measurements by comparing different FRET pairs for a cAMP-detecting biosensor. We exchanged the enhanced CFP/enhanced YFP FRET pair of an established Epac1-based biosensor by the fluorophores mCerulean/mCitrine. In the case of enhanced CFP/enhanced YFP, we showed that changes in proton, and (to a lesser extent) chloride ion concentrations result in incorrect ratiometric FRET signals, which may exceed the dynamic range of the biosensor. Calcium ions have no direct, but an indirect pH-driven effect by mobilizing protons. These ion dependences were greatly eliminated when mCerulean/mCitrine fluorophores were used. For such advanced FRET pairs the biosensor is less sensitive to changes in ion concentration and allows consistent cAMP concentration measurements under different physiological conditions, as occur in metabolically active cells. In addition, we verified that the described FRET pair exchange increased the dynamic range of the FRET efficiency response. The time window for stable experimental conditions was also prolonged by a faster biosensor expression rate in transfected cells and a greatly reduced tendency to aggregate, which reduces cytotoxicity. These properties were verified in functional tests in single cells co-expressing the biosensor and the 5-HT(1A) receptor.


Subject(s)
Biosensing Techniques/methods , Cyclic AMP/metabolism , Fluorescence Resonance Energy Transfer/methods , Animals , Calcium/analysis , Calcium/metabolism , Cell Line, Tumor , Cyclic AMP/analysis , Green Fluorescent Proteins/biosynthesis , Guanine Nucleotide Exchange Factors/biosynthesis , Mice , Receptor, Serotonin, 5-HT1A/metabolism
17.
J Biol Chem ; 286(25): 22203-10, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21502319

ABSTRACT

Voltage-dependent calcium channels constitute the main entry pathway for calcium into excitable cells. They are heteromultimers formed by an α(1) pore-forming subunit (Ca(V)α(1)) and accessory subunits. To achieve a precise coordination of calcium signals, the expression and activity of these channels is tightly controlled. The accessory ß-subunit (Ca(V)ß), a membrane associated guanylate kinase containing one guanylate kinase (ß-GK) and one Src homology 3 (ß-SH3) domain, has antagonistic effects on calcium currents by regulating different aspects of channel function. Although ß-GK binds to a conserved site within the α(1)-pore-forming subunit and facilitates channel opening, ß-SH3 binds to dynamin and promotes endocytosis. Here, we investigated the molecular switch underlying the functional duality of this modular protein. We show that ß-SH3 homodimerizes through a single disulfide bond. Substitution of the only cysteine residue abolishes dimerization and impairs internalization of L-type Ca(V)1.2 channels expressed in Xenopus oocytes while preserving dynamin binding. Covalent linkage of the ß-SH3 dimerization-deficient mutant yields a concatamer that binds to dynamin and restores endocytosis. Moreover, using FRET analysis, we show in living cells that Ca(V)ß form oligomers and that this interaction is reduced by Ca(V)α(1). Association of Ca(V)ß with a polypeptide encoding the binding motif in Ca(V)α(1) inhibited endocytosis. Together, these findings reveal that ß-SH3 dimerization is crucial for endocytosis and suggest that channel activation and internalization are two mutually exclusive functions of Ca(V)ß. We propose that a change in the oligomeric state of Ca(V)ß is the functional switch between channel activator and channel internalizer.


Subject(s)
Calcium Channels, L-Type/chemistry , Calcium Channels, L-Type/metabolism , Dynamins/metabolism , Endocytosis , Protein Multimerization , src Homology Domains , Animals , Calcium Channels, L-Type/genetics , Cell Line , Cell Membrane/metabolism , Disulfides/chemistry , Models, Molecular , Mutation , Porosity , Protein Structure, Quaternary
18.
Front Physiol ; 13: 930769, 2022.
Article in English | MEDLINE | ID: mdl-35784882

ABSTRACT

Members of the regulatory Kvß family modulate the kinetics and traffic of voltage-dependent K+ (Kv) channels. The crystal structure of Kv channels associated with Kvß peptides suggests a α4/ß4 composition. Although Kvß2 and Kvß1 form heteromers, evidence supports that only Kvß2.1 forms tetramers in the absence of α subunits. Therefore, the stoichiometry of the Kvß oligomers fine-tunes the activity of hetero-oligomeric Kv channel complexes. We demonstrate that Kvß subtypes form homo- and heterotetramers with similar affinities. The Kvß1.1/Kvß2.1 heteromer showed an altered spatial distribution in lipid rafts, recapitulating the Kvß1.1 pattern. Because Kvß2 is an active partner of the Kv1.3-TCR complex at the immunological synapse (IS), an association with Kvß1 would alter this location, shaping the immune response. Differential regulation of Kvßs influences the traffic and architecture of the Kvß heterotetramer, modulating Kvß-dependent physiological responses.

19.
Neuroinformatics ; 20(3): 679-698, 2022 07.
Article in English | MEDLINE | ID: mdl-34743262

ABSTRACT

Three-dimensional segmentation and analysis of dendritic spine morphology involve two major challenges: 1) how to segment individual spines from the dendrites and 2) how to quantitatively assess the morphology of individual spines. To address these two issues, we developed software called 3dSpAn (3-dimensional Spine Analysis), based on implementing a previously published method, 3D multi-scale opening algorithm in shared intensity space. 3dSpAn consists of four modules: a) Preprocessing and Region of Interest (ROI) selection, b) Intensity thresholding and seed selection, c) Multi-scale segmentation, and d) Quantitative morphological feature extraction. In this article, we present the results of segmentation and morphological analysis for different observation methods and conditions, including in vitro and ex vivo imaging with confocal microscopy, and in vivo observations using high-resolution two-photon microscopy. In particular, we focus on software usage, the influence of adjustable parameters on the obtained results, user reproducibility, accuracy analysis, and also include a qualitative comparison with a commercial benchmark. 3dSpAn software is freely available for non-commercial use at www.3dSpAn.org .


Subject(s)
Dendritic Spines , Imaging, Three-Dimensional , Imaging, Three-Dimensional/methods , Microscopy, Confocal/methods , Reproducibility of Results , Software
20.
Cells ; 11(15)2022 08 02.
Article in English | MEDLINE | ID: mdl-35954229

ABSTRACT

Serotonin receptor 5-HT2A and tropomyosin receptor kinase B (TrkB) strongly contribute to neuroplasticity regulation and are implicated in numerous neuronal disorders. Here, we demonstrate a physical interaction between 5-HT2A and TrkB in vitro and in vivo using co-immunoprecipitation and biophysical and biochemical approaches. Heterodimerization decreased TrkB autophosphorylation, preventing its activation with agonist 7,8-DHF, even with low 5-HT2A receptor expression. A blockade of 5-HT2A receptor with the preferential antagonist ketanserin prevented the receptor-mediated downregulation of TrkB phosphorylation without restoring the TrkB response to its agonist 7,8-DHF in vitro. In adult mice, intraperitoneal ketanserin injection increased basal TrkB phosphorylation in the frontal cortex and hippocampus, which is in accordance with our findings demonstrating the prevalence of 5-HT2A-TrkB heteroreceptor complexes in these brain regions. An expression analysis revealed strong developmental regulation of 5-HT2A and TrkB expressions in the cortex, hippocampus, and especially the striatum, demonstrating that the balance between TrkB and 5-HT2A may shift in certain brain regions during postnatal development. Our data reveal the functional role of 5-HT2A-TrkB receptor heterodimerization and suggest that the regulated expression of 5-HT2A and TrkB is a molecular mechanism for the brain-region-specific modulation of TrkB functions during development and under pathophysiological conditions.


Subject(s)
Receptor, Serotonin, 5-HT2A/metabolism , Receptor, trkB/metabolism , Serotonin , Animals , Ketanserin , Mice , Receptors, Serotonin , Serotonin/metabolism , Serotonin/pharmacology , Tropomyosin
SELECTION OF CITATIONS
SEARCH DETAIL