Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 173
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 186(4): 821-836.e13, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36750096

ABSTRACT

The low-density lipoprotein (LDL) receptor-related protein 2 (LRP2 or megalin) is representative of the phylogenetically conserved subfamily of giant LDL receptor-related proteins, which function in endocytosis and are implicated in diseases of the kidney and brain. Here, we report high-resolution cryoelectron microscopy structures of LRP2 isolated from mouse kidney, at extracellular and endosomal pH. The structures reveal LRP2 to be a molecular machine that adopts a conformation for ligand binding at the cell surface and for ligand shedding in the endosome. LRP2 forms a homodimer, the conformational transformation of which is governed by pH-sensitive sites at both homodimer and intra-protomer interfaces. A subset of LRP2 deleterious missense variants in humans appears to impair homodimer assembly. These observations lay the foundation for further understanding the function and mechanism of LDL receptors and implicate homodimerization as a conserved feature of the LRP receptor subfamily.


Subject(s)
Endocytosis , Low Density Lipoprotein Receptor-Related Protein-2 , Animals , Humans , Mice , Cryoelectron Microscopy , Kidney/metabolism , Ligands , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Low Density Lipoprotein Receptor-Related Protein-2/metabolism
2.
Cell ; 181(7): 1547-1565.e15, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32492405

ABSTRACT

Homeostasis of neural firing properties is important in stabilizing neuronal circuitry, but how such plasticity might depend on alternative splicing is not known. Here we report that chronic inactivity homeostatically increases action potential duration by changing alternative splicing of BK channels; this requires nuclear export of the splicing factor Nova-2. Inactivity and Nova-2 relocation were connected by a novel synapto-nuclear signaling pathway that surprisingly invoked mechanisms akin to Hebbian plasticity: Ca2+-permeable AMPA receptor upregulation, L-type Ca2+ channel activation, enhanced spine Ca2+ transients, nuclear translocation of a CaM shuttle, and nuclear CaMKIV activation. These findings not only uncover commonalities between homeostatic and Hebbian plasticity but also connect homeostatic regulation of synaptic transmission and neuronal excitability. The signaling cascade provides a full-loop mechanism for a classic autoregulatory feedback loop proposed ∼25 years ago. Each element of the loop has been implicated previously in neuropsychiatric disease.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channels/metabolism , Long-Term Potentiation/physiology , Nerve Tissue Proteins/metabolism , RNA-Binding Proteins/metabolism , Action Potentials/physiology , Alternative Splicing/genetics , Alternative Splicing/physiology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Female , HEK293 Cells , Homeostasis/physiology , Humans , Large-Conductance Calcium-Activated Potassium Channels/genetics , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/physiology , Neuro-Oncological Ventral Antigen , Neuronal Plasticity/physiology , Neurons/metabolism , RNA-Binding Proteins/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction , Synapses/metabolism , Synaptic Transmission/physiology
3.
Cell ; 159(2): 281-94, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25303525

ABSTRACT

Activity-dependent CREB phosphorylation and gene expression are critical for long-term neuronal plasticity. Local signaling at CaV1 channels triggers these events, but how information is relayed onward to the nucleus remains unclear. Here, we report a mechanism that mediates long-distance communication within cells: a shuttle that transports Ca(2+)/calmodulin from the surface membrane to the nucleus. We show that the shuttle protein is γCaMKII, its phosphorylation at Thr287 by ßCaMKII protects the Ca(2+)/CaM signal, and CaN triggers its nuclear translocation. Both ßCaMKII and CaN act in close proximity to CaV1 channels, supporting their dominance, whereas γCaMKII operates as a carrier, not as a kinase. Upon arrival within the nucleus, Ca(2+)/CaM activates CaMKK and its substrate CaMKIV, the CREB kinase. This mechanism resolves long-standing puzzles about CaM/CaMK-dependent signaling to the nucleus. The significance of the mechanism is emphasized by dysregulation of CaV1, γCaMKII, ßCaMKII, and CaN in multiple neuropsychiatric disorders.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Animals , Calcium/metabolism , Calcium Channels/metabolism , Calmodulin/metabolism , Cell Nucleus/metabolism , Neurons/metabolism , Phosphorylation , Rats, Sprague-Dawley , Transcription, Genetic
4.
EMBO J ; 40(23): e108428, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34661298

ABSTRACT

Mitochondrial cristae are extraordinarily crowded with proteins, which puts stress on the bilayer organization of lipids. We tested the hypothesis that the high concentration of proteins drives the tafazzin-catalyzed remodeling of fatty acids in cardiolipin, thereby reducing bilayer stress in the membrane. Specifically, we tested whether protein crowding induces cardiolipin remodeling and whether the lack of cardiolipin remodeling prevents the membrane from accumulating proteins. In vitro, the incorporation of large amounts of proteins into liposomes altered the outcome of the remodeling reaction. In yeast, the concentration of proteins involved in oxidative phosphorylation (OXPHOS) correlated with the cardiolipin composition. Genetic ablation of either remodeling or biosynthesis of cardiolipin caused a substantial drop in the surface density of OXPHOS proteins in the inner membrane of the mouse heart and Drosophila flight muscle mitochondria. Our data suggest that OXPHOS protein crowding induces cardiolipin remodelling and that remodeled cardiolipin supports the high concentration of these proteins in the inner mitochondrial membrane.


Subject(s)
Acyltransferases/physiology , Cardiolipins/metabolism , Mitochondria, Heart/metabolism , Mitochondria, Muscle/metabolism , Mitochondrial Membranes/metabolism , Oxidative Phosphorylation , Proteins/metabolism , Animals , Cardiolipins/chemistry , Cardiolipins/genetics , Drosophila melanogaster , Fatty Acids/metabolism , Female , Liposomes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction , Saccharomyces cerevisiae
5.
Nature ; 574(7779): 559-564, 2019 10.
Article in English | MEDLINE | ID: mdl-31645735

ABSTRACT

Although glucose-sensing neurons were identified more than 50 years ago, the physiological role of glucose sensing in metazoans remains unclear. Here we identify a pair of glucose-sensing neurons with bifurcated axons in the brain of Drosophila. One axon branch projects to insulin-producing cells to trigger the release of Drosophila insulin-like peptide 2 (dilp2) and the other extends to adipokinetic hormone (AKH)-producing cells to inhibit secretion of AKH, the fly analogue of glucagon. These axonal branches undergo synaptic remodelling in response to changes in their internal energy status. Silencing of these glucose-sensing neurons largely disabled the response of insulin-producing cells to glucose and dilp2 secretion, disinhibited AKH secretion in corpora cardiaca and caused hyperglycaemia, a hallmark feature of diabetes mellitus. We propose that these glucose-sensing neurons maintain glucose homeostasis by promoting the secretion of dilp2 and suppressing the release of AKH when haemolymph glucose levels are high.


Subject(s)
Brain/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Glucagon/metabolism , Glucose/metabolism , Insulin/metabolism , Neurons/metabolism , Animals , Axons/metabolism , Brain/anatomy & histology , Drosophila Proteins/metabolism , Drosophila melanogaster/anatomy & histology , Glucose/analysis , Insect Hormones/metabolism , Male , Neural Inhibition , Neural Pathways , Neuropeptides/chemistry , Neuropeptides/metabolism , Neurotransmitter Agents/metabolism , Oligopeptides/metabolism , Pyrrolidonecarboxylic Acid/analogs & derivatives , Pyrrolidonecarboxylic Acid/metabolism
6.
J Biol Chem ; 299(10): 105241, 2023 10.
Article in English | MEDLINE | ID: mdl-37690688

ABSTRACT

Respiratory complexes and cardiolipins have exceptionally long lifetimes. The fact that they co-localize in mitochondrial cristae raises the question of whether their longevities have a common cause and whether the longevity of OXPHOS proteins is dependent on cardiolipin. To address these questions, we developed a method to measure side-by-side the half-lives of proteins and lipids in wild-type Drosophila and cardiolipin-deficient mutants. We fed adult flies with stable isotope-labeled precursors (13C615N2-lysine or 13C6-glucose) and determined the relative abundance of heavy isotopomers in protein and lipid species by mass spectrometry. To minimize the confounding effects of tissue regeneration, we restricted our analysis to the thorax, the bulk of which consists of post-mitotic flight muscles. Analysis of 680 protein and 45 lipid species showed that the subunits of respiratory complexes I-V and the carriers for phosphate and ADP/ATP were among the longest-lived proteins (average half-life of 48 ± 16 days) while the molecular species of cardiolipin were the longest-lived lipids (average half-life of 27 ± 6 days). The remarkable longevity of these crista residents was not shared by all mitochondrial proteins, especially not by those residing in the matrix and the inner boundary membrane. Ablation of cardiolipin synthase, which causes replacement of cardiolipin by phosphatidylglycerol, and ablation of tafazzin, which causes partial replacement of cardiolipin by monolyso-cardiolipin, decreased the lifetimes of the respiratory complexes. Ablation of tafazzin also decreased the lifetimes of the remaining cardiolipin species. These data suggest that an important function of cardiolipin in mitochondria is to protect respiratory complexes from degradation.


Subject(s)
Cardiolipins , Animals , Cardiolipins/metabolism , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Muscles/metabolism , Drosophila melanogaster
7.
Glia ; 72(3): 625-642, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38031883

ABSTRACT

Astrocytes are a heterogeneous population of central nervous system glial cells that respond to pathological insults and injury by undergoing a transformation called "reactivity." Reactive astrocytes exhibit distinct and context-dependent cellular, molecular, and functional state changes that can either support or disturb tissue homeostasis. We recently identified a reactive astrocyte sub-state defined by interferon-responsive genes like Igtp, Ifit3, Mx1, and others, called interferon-responsive reactive astrocytes (IRRAs). To further this transcriptomic definition of IRRAs, we wanted to define the proteomic changes that occur in this reactive sub-state. We induced IRRAs in immunopanned rodent astrocytes and human iPSC-differentiated astrocytes using TNF, IL1α, C1Q, and IFNß and characterized their proteomic profile (both cellular and secreted) using unbiased quantitative proteomics. We identified 2335 unique cellular proteins, including IFIT2/3, IFITM3, OASL1/2, MX1/2/3, and STAT1. We also report that rodent and human IRRAs secrete PAI1, a serine protease inhibitor which may influence reactive states and functions of nearby cells. Finally, we evaluated how IRRAs are distinct from neurotoxic reactive astrocytes (NRAs). While NRAs are described by expression of the complement protein C3, it was not upregulated in IRRAs. Instead, we found ~90 proteins unique to IRRAs not identified in NRAs, including OAS1A, IFIT3, and MX1. Interferon signaling in astrocytes is critical for the antiviral immune response and for regulating synaptic plasticity and glutamate transport mechanisms. How IRRAs contribute to these functions is unknown. This study provides the basis for future experiments to define the functional roles of IRRAs in the context of neurodegenerative disorders.


Subject(s)
Astrocytes , Interferons , Animals , Humans , Astrocytes/metabolism , Interferons/metabolism , Rodentia/metabolism , Proteomics , Central Nervous System/metabolism , Membrane Proteins/metabolism , RNA-Binding Proteins/metabolism
9.
Mol Cell ; 61(1): 98-110, 2016 Jan 07.
Article in English | MEDLINE | ID: mdl-26687682

ABSTRACT

The molecular basis by which receptor tyrosine kinases (RTKs) recruit and phosphorylate Src Homology 2 (SH2) domain-containing substrates has remained elusive. We used X-ray crystallography, NMR spectroscopy, and cell-based assays to demonstrate that recruitment and phosphorylation of Phospholipase Cγ (PLCγ), a prototypical SH2 containing substrate, by FGF receptors (FGFR) entails formation of an allosteric 2:1 FGFR-PLCγ complex. We show that the engagement of pTyr-binding pocket of the cSH2 domain of PLCγ by the phosphorylated tail of an FGFR kinase induces a conformational change at the region past the cSH2 core domain encompassing Tyr-771 and Tyr-783 to facilitate the binding/phosphorylation of these tyrosines by another FGFR kinase in trans. Our data overturn the current paradigm that recruitment and phosphorylation of substrates are carried out by the same RTK monomer in cis and disclose an obligatory role for receptor dimerization in substrate phosphorylation in addition to its canonical role in kinase activation.


Subject(s)
Phospholipase C gamma/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , ErbB Receptors/metabolism , HEK293 Cells , Humans , Hydrolysis , Models, Molecular , Molecular Sequence Data , Multienzyme Complexes , Nuclear Magnetic Resonance, Biomolecular , Phosphatidylinositols/metabolism , Phospholipase C gamma/chemistry , Phospholipase C gamma/genetics , Phosphorylation , Protein Binding , Protein Conformation , Protein Transport , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 2/chemistry , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptors, Platelet-Derived Growth Factor/metabolism , Structure-Activity Relationship , Transfection , Vascular Endothelial Growth Factor Receptor-2/metabolism , src Homology Domains
10.
Nat Chem Biol ; 16(3): 267-277, 2020 03.
Article in English | MEDLINE | ID: mdl-31959966

ABSTRACT

A long-standing mystery shrouds the mechanism by which catalytically repressed receptor tyrosine kinase domains accomplish transphosphorylation of activation loop (A-loop) tyrosines. Here we show that this reaction proceeds via an asymmetric complex that is thermodynamically disadvantaged because of an electrostatic repulsion between enzyme and substrate kinases. Under physiological conditions, the energetic gain resulting from ligand-induced dimerization of extracellular domains overcomes this opposing clash, stabilizing the A-loop-transphosphorylating dimer. A unique pathogenic fibroblast growth factor receptor gain-of-function mutation promotes formation of the complex responsible for phosphorylation of A-loop tyrosines by eliminating this repulsive force. We show that asymmetric complex formation induces a more phosphorylatable A-loop conformation in the substrate kinase, which in turn promotes the active state of the enzyme kinase. This explains how quantitative differences in the stability of ligand-induced extracellular dimerization promotes formation of the intracellular A-loop-transphosphorylating asymmetric complex to varying extents, thereby modulating intracellular kinase activity and signaling intensity.


Subject(s)
AAA Domain/physiology , Protein-Tyrosine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , AAA Domain/genetics , Catalytic Domain , Dimerization , Enzyme Activation , Humans , Ligands , Phosphorylation , Protein Binding , Protein Conformation , Protein-Tyrosine Kinases/physiology , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/physiology , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Signal Transduction , Structure-Activity Relationship , Tyrosine/chemistry
11.
Mol Cell ; 55(5): 782-90, 2014 Sep 04.
Article in English | MEDLINE | ID: mdl-25175024

ABSTRACT

Oncogenic RAS (H-RAS(V12)) induces premature senescence in primary cells by triggering production of reactive oxygen species (ROS), but the molecular role of ROS in senescence remains elusive. We investigated whether inhibition of protein tyrosine phosphatases by ROS contributed to H-RAS(V12)-induced senescence. We identified protein tyrosine phosphatase 1B (PTP1B) as a major target of H-RAS(V12)-induced ROS. Inactivation of PTP1B was necessary and sufficient to induce premature senescence in H-RAS(V12)-expressing IMR90 fibroblasts. We identified phospho-Tyr 393 of argonaute 2 (AGO2) as a direct substrate of PTP1B. Phosphorylation of AGO2 at Tyr 393 inhibited loading with microRNAs (miRNAs) and thus miRNA-mediated gene silencing, which counteracted the function of H-RAS(V12)-induced oncogenic miRNAs. Overall, our data illustrate that premature senescence in H-RAS(V12)-transformed primary cells is a consequence of oxidative inactivation of PTP1B and inhibition of miRNA-mediated gene silencing.


Subject(s)
Argonaute Proteins/metabolism , Gene Silencing , Protein Tyrosine Phosphatase, Non-Receptor Type 1/physiology , Tyrosine/metabolism , ras Proteins/physiology , Argonaute Proteins/chemistry , Cell Line , Cellular Senescence/genetics , Humans , MicroRNAs/metabolism , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Reactive Oxygen Species/metabolism , Tyrosine/chemistry , ras Proteins/genetics , ras Proteins/metabolism
12.
Proc Natl Acad Sci U S A ; 115(12): E2829-E2838, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29507251

ABSTRACT

Sweet-insensitive Drosophila mutants are unable to readily identify sugar. In presence of wild-type (WT) flies, however, these mutant flies demonstrated a marked increase in their preference for nutritive sugar. Real-time recordings of starved WT flies revealed that these flies discharge a drop from their gut end after consuming nutritive sugars, but not nonnutritive sugars. We proposed that the drop may contain a molecule(s) named calorie-induced secreted factor (CIF), which serves as a signal to inform other flies about its nutritional value. Consistent with this, we observed a robust preference of flies for nutritive sugar containing CIF over nutritive sugar without CIF. Feeding appears to be a prerequisite for the release of CIF, given that fed flies did not produce it. Additionally, correlation analyses and pharmacological approaches suggest that the nutritional value, rather than the taste, of the consumed sugar correlates strongly with the amount (or intensity) of the released CIF. We observed that the release of this attractant signal requires the consumption of macronutrients, specifically nutritive sugars and l-enantiomer essential amino acids (l-eAAs), but it is negligibly released when flies are fed nonnutritive sugars, unnatural d-enantiomer essential amino acids (d-eAAs), fatty acids, alcohol, or salts. Finally, CIF (i) is not detected by the olfactory system, (ii) is not influenced by the sex of the fly, and (iii) is not limited to one species of Drosophila.


Subject(s)
Drosophila/physiology , Pheromones/metabolism , Sugars , Animal Communication , Animals , Drosophila/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Female , Male , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nutritive Value , Olfactory Bulb/physiology , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Species Specificity , Sugars/metabolism , Sugars/pharmacology
13.
J Lipid Res ; 61(1): 95-104, 2020 01.
Article in English | MEDLINE | ID: mdl-31712250

ABSTRACT

Lipid metabolism plays an important role in the regulation of cellular homeostasis. However, because it is difficult to measure the actual rates of synthesis and degradation of individual lipid species, lipid compositions are often used as a surrogate to evaluate lipid metabolism even though they provide only static snapshots of the lipodome. Here, we designed a simple method to determine the turnover rate of phospholipid and acylglycerol species based on the incorporation of 13C6-glucose combined with LC-MS/MS. We labeled adult Drosophila melanogaster with 13C6-glucose that incorporates into the entire lipidome, derived kinetic parameters from mass spectra, and studied effects of deletion of CG6718, the fly homolog of the calcium-independent phospholipase A2ß, on lipid metabolism. Although 13C6-glucose gave rise to a complex pattern of 13C incorporation, we were able to identify discrete isotopomers in which 13C atoms were confined to the glycerol group. With these isotopomers, we calculated turnover rate constants, half-life times, and fluxes of the glycerol backbone of multiple lipid species. To perform these calculations, we estimated the fraction of labeled molecules in glycerol-3-phosphate, the lipid precursor, by mass isotopomer distribution analysis of the spectra of phosphatidylglycerol. When we applied this method to D. melanogaster, we found a range of lipid half-lives from 2 to 200 days, demonstrated tissue-specific fluxes of individual lipid species, and identified a novel function of CG6718 in triacylglycerol metabolism. This method provides fluxomics-type data with significant potential to improve the understanding of complex lipid regulation in a variety of research models.


Subject(s)
Drosophila melanogaster/metabolism , Isotope Labeling , Lipid Metabolism , Lipidomics , Lipids/analysis , Animals , Carbon Isotopes , Chromatography, Liquid , Tandem Mass Spectrometry
14.
PLoS Biol ; 15(7): e2002457, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28719605

ABSTRACT

Extracellular phosphorylation of proteins was suggested in the late 1800s when it was demonstrated that casein contains phosphate. More recently, extracellular kinases that phosphorylate extracellular serine, threonine, and tyrosine residues of numerous proteins have been identified. However, the functional significance of extracellular phosphorylation of specific residues in the nervous system is poorly understood. Here we show that synaptic accumulation of GluN2B-containing N-methyl-D-aspartate receptors (NMDARs) and pathological pain are controlled by ephrin-B-induced extracellular phosphorylation of a single tyrosine (p*Y504) in a highly conserved region of the fibronectin type III (FN3) domain of the receptor tyrosine kinase EphB2. Ligand-dependent Y504 phosphorylation modulates the EphB-NMDAR interaction in cortical and spinal cord neurons. Furthermore, Y504 phosphorylation enhances NMDAR localization and injury-induced pain behavior. By mediating inducible extracellular interactions that are capable of modulating animal behavior, extracellular tyrosine phosphorylation of EphBs may represent a previously unknown class of mechanism mediating protein interaction and function.


Subject(s)
Pain/metabolism , Receptor, EphB2/metabolism , Receptors, N-Methyl-D-Aspartate/analysis , Animals , HEK293 Cells , Humans , Mice , Neurons/metabolism , Phosphorylation , Rats , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, N-Methyl-D-Aspartate/physiology , Sequence Analysis, Protein , Spinal Cord/metabolism , Spinal Cord/pathology , Tyrosine/metabolism
15.
Nucleic Acids Res ; 45(16): 9398-9412, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28934503

ABSTRACT

In mammals, faithful inheritance of genomic methylation patterns ensures proper gene regulation and cell behaviour, impacting normal development and fertility. Following establishment, genomic methylation patterns are transmitted through S-phase by the maintenance methyltransferase Dnmt1. Using a protein interaction screen, we identify Microprocessor component DROSHA as a novel DNMT1-interactor. Drosha-deficient embryonic stem (ES) cells display genomic hypomethylation that is not accounted for by changes in the levels of DNMT proteins. DNMT1-mediated methyltransferase activity is also reduced in these cells. We identify two transcripts that are specifically upregulated in Drosha- but not Dicer-deficient ES cells. Regions within these transcripts predicted to form stem-loop structures are processed by Microprocessor and can inhibit DNMT1-mediated methylation in vitro. Our results highlight DROSHA as a novel regulator of mammalian DNA methylation and we propose that DROSHA-mediated processing of RNA is necessary to ensure full DNMT1 activity. This adds to the DROSHA repertoire of non-miRNA dependent functions as well as implicating RNA in regulating DNMT1 activity and correct levels of genomic methylation.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Ribonuclease III/physiology , Animals , CRISPR-Cas Systems , Cells, Cultured , Cytosine/metabolism , DNA (Cytosine-5-)-Methyltransferase 1 , Embryonic Stem Cells/enzymology , HEK293 Cells , Humans , Mice , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Ribonuclease III/genetics , Ribonuclease III/metabolism , Up-Regulation
16.
Adv Exp Med Biol ; 1140: 531-539, 2019.
Article in English | MEDLINE | ID: mdl-31347069

ABSTRACT

Stable isotope labeling by amino acids in cell culture (SILAC) is a powerful approach for high-throughput quantitative proteomics. SILAC allows highly accurate protein quantitation through metabolic encoding of whole cell proteomes using stable isotope labeled amino acids. Since its introduction in 2002, SILAC has become increasingly popular. In this chapter we review the methodology and application of SILAC, with an emphasis on three research areas: dynamics of posttranslational modifications, protein-protein interactions, and protein turnover.


Subject(s)
Amino Acids/chemistry , Cell Culture Techniques , Isotope Labeling , Proteomics/methods , Proteome
17.
Biochim Biophys Acta Mol Basis Dis ; 1864(1): 208-225, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28711595

ABSTRACT

Extensive parenchymal and vascular Aß deposits are pathological hallmarks of Alzheimer's disease (AD). Besides classic full-length peptides, biochemical analyses of brain deposits have revealed high degree of Aß heterogeneity likely resulting from the action of multiple proteolytic enzymes. In spite of the numerous studies focusing in Aß, the relevance of N- and C-terminal truncated species for AD pathogenesis remains largely understudied. In the present work, using novel antibodies specifically recognizing Aß species N-terminally truncated at position 4 or C-terminally truncated at position 34, we provide a clear assessment of the differential topographic localization of these species in AD brains and transgenic models. Based on their distinct solubility, brain N- and C-terminal truncated species were extracted by differential fractionation and identified via immunoprecipitation coupled to mass spectrometry analysis. Biochemical/biophysical studies with synthetic homologues further confirmed the different solubility properties and contrasting fibrillogenic characteristics of the truncated species composing the brain Aß peptidome. Aß C-terminal degradation leads to the production of more soluble fragments likely to be more easily eliminated from the brain. On the contrary, N-terminal truncation at position 4 favors the formation of poorly soluble, aggregation prone peptides with high amyloidogenic propensity and the potential to exacerbate the fibrillar deposits, self-perpetuating the amyloidogenic loop. Detailed assessment of the molecular diversity of Aß species composing interstitial fluid and amyloid deposits at different disease stages, as well as the evaluation of the truncation profile during various pharmacologic approaches will provide a comprehensive understanding of the still undefined contribution of Aß truncations to the disease pathogenesis and their potential as novel therapeutic targets.


Subject(s)
Amyloid beta-Peptides/metabolism , Brain/metabolism , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Aged , Aged, 80 and over , Amyloid beta-Peptides/chemistry , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/pathology , Case-Control Studies , Female , Humans , Male , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Rabbits
18.
Am J Pathol ; 187(8): 1867-1878, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28606795

ABSTRACT

Optic gliomas are brain tumors characterized by slow growth, progressive loss of vision, and limited therapeutic options. Optic gliomas contain various amounts of myxoid matrix, which can represent most of the tumor mass. We sought to investigate biological function and protein structure of the myxoid matrix in optic gliomas to identify novel therapeutic targets. We reviewed histological features and clinical imaging properties, analyzed vasculature by immunohistochemistry and electron microscopy, and performed liquid chromatography-mass spectrometry on optic gliomas, which varied in the amount of myxoid matrix. We found that although subtypes of optic gliomas are indistinguishable on imaging, the microvascular network of pilomyxoid astrocytoma, a subtype of optic glioma with abundant myxoid matrix, is characterized by the presence of endothelium-free channels in the myxoid matrix. These tumors show normal perfusion by clinical imaging and lack histological evidence of hemorrhage organization or thrombosis. The myxoid matrix is composed predominantly of the proteoglycan versican and its linking protein, a vertebrate hyaluronan and proteoglycan link protein 1. We propose that pediatric optic gliomas can maintain blood supply without endothelial cells by using invertebrate-like channels, which we termed primitive myxoid vascularization. Enzymatic targeting of the proteoglycan versican/hyaluronan and proteoglycan link protein 1 rich myxoid matrix, which is in direct contact with circulating blood, can provide novel therapeutic avenues for optic gliomas of childhood.


Subject(s)
Blood Vessels/pathology , Endothelial Cells/pathology , Neovascularization, Pathologic/pathology , Optic Nerve Glioma/pathology , Endothelium/pathology , Humans , Optic Nerve Glioma/blood supply , Retrospective Studies
19.
Nat Chem Biol ; 12(8): 641-7, 2016 08.
Article in English | MEDLINE | ID: mdl-27348092

ABSTRACT

Cardiolipin is a specific mitochondrial phospholipid that has a high affinity for proteins and that stabilizes the assembly of supercomplexes involved in oxidative phosphorylation. We found that sequestration of cardiolipin in protein complexes is critical to protect it from degradation. The turnover of cardiolipin is slower by almost an order of magnitude than the turnover of other phospholipids. However, in subjects with Barth syndrome, cardiolipin is rapidly degraded via the intermediate monolyso-cardiolipin. Treatments that induce supercomplex assembly decrease the turnover of cardiolipin and the concentration of monolyso-cardiolipin, whereas dissociation of supercomplexes has the opposite effect. Our data suggest that cardiolipin is uniquely protected from normal lipid turnover by its association with proteins, but this association is compromised in subjects with Barth syndrome, leading cardiolipin to become unstable, which in turn causes the accumulation of monolyso-cardiolipin.


Subject(s)
Barth Syndrome/metabolism , Cardiolipins/metabolism , Humans
20.
Mol Cell ; 40(5): 787-97, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-21145486

ABSTRACT

Endoplasmic reticulum (ER) oxidation 1 (ERO1) transfers disulfides to protein disulfide isomerase (PDI) and is essential for oxidative protein folding in simple eukaryotes such as yeast and worms. Surprisingly, ERO1-deficient mammalian cells exhibit only a modest delay in disulfide bond formation. To identify ERO1-independent pathways to disulfide bond formation, we purified PDI oxidants with a trapping mutant of PDI. Peroxiredoxin IV (PRDX4) stood out in this list, as the related cytosolic peroxiredoxins are known to form disulfides in the presence of hydroperoxides. Mouse embryo fibroblasts lacking ERO1 were intolerant of PRDX4 knockdown. Introduction of wild-type mammalian PRDX4 into the ER rescued the temperature-sensitive phenotype of an ero1 yeast mutation. In the presence of an H(2)O(2)-generating system, purified PRDX4 oxidized PDI and reconstituted oxidative folding of RNase A. These observations implicate ER-localized PRDX4 in a previously unanticipated, parallel, ERO1-independent pathway that couples hydroperoxide production to oxidative protein folding in mammalian cells.


Subject(s)
Endoplasmic Reticulum/metabolism , Peroxiredoxins/chemistry , Animals , Catalytic Domain , Hydrogen Peroxide/metabolism , Mice , Oxidation-Reduction , Peroxiredoxins/metabolism , Protein Disulfide-Isomerases/chemistry , Protein Disulfide-Isomerases/metabolism , Protein Folding
SELECTION OF CITATIONS
SEARCH DETAIL