Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Cell ; 173(3): 720-734.e15, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29677515

ABSTRACT

Reversible phase separation underpins the role of FUS in ribonucleoprotein granules and other membrane-free organelles and is, in part, driven by the intrinsically disordered low-complexity (LC) domain of FUS. Here, we report that cooperative cation-π interactions between tyrosines in the LC domain and arginines in structured C-terminal domains also contribute to phase separation. These interactions are modulated by post-translational arginine methylation, wherein arginine hypomethylation strongly promotes phase separation and gelation. Indeed, significant hypomethylation, which occurs in FUS-associated frontotemporal lobar degeneration (FTLD), induces FUS condensation into stable intermolecular ß-sheet-rich hydrogels that disrupt RNP granule function and impair new protein synthesis in neuron terminals. We show that transportin acts as a physiological molecular chaperone of FUS in neuron terminals, reducing phase separation and gelation of methylated and hypomethylated FUS and rescuing protein synthesis. These results demonstrate how FUS condensation is physiologically regulated and how perturbations in these mechanisms can lead to disease.


Subject(s)
Arginine/chemistry , Molecular Chaperones/chemistry , RNA-Binding Protein FUS/chemistry , Amyotrophic Lateral Sclerosis/metabolism , Animals , Cations , DNA Methylation , Frontotemporal Dementia/metabolism , Frontotemporal Lobar Degeneration/metabolism , Humans , Microscopy, Atomic Force , Microscopy, Fluorescence , Protein Binding , Protein Domains , Protein Processing, Post-Translational , Protein Structure, Secondary , RNA-Binding Protein FUS/metabolism , Tyrosine/chemistry , Xenopus laevis
2.
J Neurochem ; 165(2): 230-245, 2023 04.
Article in English | MEDLINE | ID: mdl-36511154

ABSTRACT

The bank vole (BV) prion protein (PrP) can function as a universal acceptor of prions. However, the molecular details of BVPrP's promiscuity for replicating a diverse range of prion strains remain obscure. To develop a cultured cell paradigm capable of interrogating the unique properties of BVPrP, we generated monoclonal lines of CAD5 cells lacking endogenous PrP but stably expressing either hamster (Ha), mouse (Mo), or BVPrP (M109 or I109 polymorphic variants) and then challenged them with various strains of mouse or hamster prions. Cells expressing BVPrP were susceptible to both mouse and hamster prions, whereas cells expressing MoPrP or HaPrP could only be infected with species-matched prions. Propagation of mouse and hamster prions in cells expressing BVPrP resulted in strain adaptation in several instances, as evidenced by alterations in conformational stability, glycosylation, susceptibility to anti-prion small molecules, and the inability of BVPrP-adapted mouse prion strains to infect cells expressing MoPrP. Interestingly, cells expressing BVPrP containing the G127V prion gene variant, identified in individuals resistant to kuru, were unable to become infected with prions. Moreover, the G127V polymorphic variant impeded the spontaneous aggregation of recombinant BVPrP. These results demonstrate that BVPrP can facilitate cross-species prion replication in cultured cells and that a single amino acid change can override the prion-permissive nature of BVPrP. This cellular paradigm will be useful for dissecting the molecular features of BVPrP that allow it to function as a universal prion acceptor.


Subject(s)
Prion Diseases , Prions , Cricetinae , Animals , Prions/genetics , Prions/metabolism , Prion Proteins/genetics , Prion Proteins/metabolism , Prion Diseases/genetics , Arvicolinae/genetics , Arvicolinae/metabolism , Cells, Cultured
3.
PLoS Pathog ; 17(10): e1009991, 2021 10.
Article in English | MEDLINE | ID: mdl-34610054

ABSTRACT

Corruption of cellular prion protein (PrPC) function(s) at the plasma membrane of neurons is at the root of prion diseases, such as Creutzfeldt-Jakob disease and its variant in humans, and Bovine Spongiform Encephalopathies, better known as mad cow disease, in cattle. The roles exerted by PrPC, however, remain poorly elucidated. With the perspective to grasp the molecular pathways of neurodegeneration occurring in prion diseases, and to identify therapeutic targets, achieving a better understanding of PrPC roles is a priority. Based on global approaches that compare the proteome and metabolome of the PrPC expressing 1C11 neuronal stem cell line to those of PrPnull-1C11 cells stably repressed for PrPC expression, we here unravel that PrPC contributes to the regulation of the energetic metabolism by orienting cells towards mitochondrial oxidative degradation of glucose. Through its coupling to cAMP/protein kinase A signaling, PrPC tones down the expression of the pyruvate dehydrogenase kinase 4 (PDK4). Such an event favors the transfer of pyruvate into mitochondria and its conversion into acetyl-CoA by the pyruvate dehydrogenase complex and, thereby, limits fatty acids ß-oxidation and subsequent onset of oxidative stress conditions. The corruption of PrPC metabolic role by pathogenic prions PrPSc causes in the mouse hippocampus an imbalance between glucose oxidative degradation and fatty acids ß-oxidation in a PDK4-dependent manner. The inhibition of PDK4 extends the survival of prion-infected mice, supporting that PrPSc-induced deregulation of PDK4 activity and subsequent metabolic derangements contribute to prion diseases. Our study posits PDK4 as a potential therapeutic target to fight against prion diseases.


Subject(s)
Glucose/metabolism , Nerve Degeneration/metabolism , PrPSc Proteins/metabolism , Prion Diseases/metabolism , Prion Diseases/pathology , Animals , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Nerve Degeneration/pathology , Oxidative Stress/physiology , Protein Kinases/metabolism
4.
J Biol Chem ; 297(3): 101073, 2021 09.
Article in English | MEDLINE | ID: mdl-34390689

ABSTRACT

The study of prions and the discovery of candidate therapeutics for prion disease have been facilitated by the ability of prions to replicate in cultured cells. Paradigms in which prion proteins from different species are expressed in cells with low or no expression of endogenous prion protein (PrP) have expanded the range of prion strains that can be propagated. In these systems, cells stably expressing a PrP of interest are typically generated via coexpression of a selectable marker and treatment with an antibiotic. Here, we report the unexpected discovery that the aminoglycoside G418 (Geneticin) interferes with the ability of stably transfected cultured cells to become infected with prions. In G418-resistant lines of N2a or CAD5 cells, the presence of G418 reduced levels of protease-resistant PrP following challenge with the RML or 22L strains of mouse prions. G418 also interfered with the infection of cells expressing hamster PrP with the 263K strain of hamster prions. Interestingly, G418 had minimal to no effect on protease-resistant PrP levels in cells with established prion infection, arguing that G418 selectively interferes with de novo prion infection. As G418 treatment had no discernible effect on cellular PrP levels or its localization, this suggests that G418 may specifically target prion assemblies or processes involved in the earliest stages of prion infection.


Subject(s)
Gentamicins/pharmacology , Prion Proteins/drug effects , Prions/antagonists & inhibitors , Aminoglycosides/metabolism , Aminoglycosides/pharmacology , Animals , Cell Line , Cell Line, Tumor , Gentamicins/metabolism , Mice , PrPC Proteins/drug effects , PrPC Proteins/metabolism , PrPSc Proteins/drug effects , PrPSc Proteins/metabolism , Prion Diseases/prevention & control , Prion Proteins/metabolism , Prions/metabolism , Protein Synthesis Inhibitors
5.
J Biol Chem ; 296: 100118, 2021.
Article in English | MEDLINE | ID: mdl-33234594

ABSTRACT

Astrocytes can support neuronal survival through a range of secreted signals that protect against neurotoxicity, oxidative stress, and apoptotic cascades. Thus, analyzing the effects of the astrocyte secretome may provide valuable insight into these neuroprotective mechanisms. Previously, we characterized a potent neuroprotective activity mediated by retinal astrocyte conditioned media (ACM) on retinal and cortical neurons in metabolic stress models. However, the molecular mechanism underlying this complex activity in neuronal cells has remained unclear. Here, a chemical genetics screen of kinase inhibitors revealed phosphoinositide 3-kinase (PI3K) as a central player transducing ACM-mediated neuroprotection. To identify additional proteins contributing to the protective cascade, endogenous PI3K was immunoprecipitated from neuronal cells exposed to ACM or control media, followed by MS/MS proteomic analyses. These data pointed toward a relatively small number of proteins that coimmunoprecipitated with PI3K, and surprisingly only five were regulated by the ACM signal. These hits included expected PI3K interactors, such as the platelet-derived growth factor receptor A (PDGFRA), as well as novel RNA-binding protein interactors ZC3H14 (zinc finger CCCH-type containing 14) and THOC1 (THO complex protein 1). In particular, ZC3H14 has recently emerged as an important RNA-binding protein with multiple roles in posttranscriptional regulation. In validation studies, we show that PI3K recruitment of ZC3H14 is necessary for PDGF-induced neuroprotection and that this interaction is present in primary retinal ganglion cells. Thus, we identified a novel non-cell autonomous neuroprotective signaling cascade mediated through PI3K that requires recruitment of ZC3H14 and may present a promising strategy to promote astrocyte-secreted prosurvival signals.


Subject(s)
Astrocytes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Poly(A)-Binding Proteins/metabolism , RNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Immunoprecipitation , Neuroprotection/physiology , Phosphatidylinositol 3-Kinases/chemistry , Poly(A)-Binding Proteins/genetics , RNA-Binding Proteins/genetics , Tandem Mass Spectrometry
6.
PLoS Comput Biol ; 17(3): e1008771, 2021 03.
Article in English | MEDLINE | ID: mdl-33711010

ABSTRACT

Alzheimer's disease is associated with the formation of toxic aggregates of amyloid beta (Aß) peptides. Despite tremendous efforts, our understanding of the molecular mechanisms of aggregation, as well as cofactors that might influence it, remains incomplete. The small cyclic neuropeptide somatostatin-14 (SST14) was recently found to be the most selectively enriched protein in human frontal lobe extracts that binds Aß42 aggregates. Furthermore, SST14's presence was also found to promote the formation of toxic Aß42 oligomers in vitro. In order to elucidate how SST14 influences the onset of Aß oligomerization, we performed all-atom molecular dynamics simulations of model mixtures of Aß42 or Aß40 peptides with SST14 molecules and analyzed the structure and dynamics of early-stage aggregates. For comparison we also analyzed the aggregation of Aß42 in the presence of arginine vasopressin (AVP), a different cyclic neuropeptide. We observed the formation of self-assembled aggregates containing the Aß chains and small cyclic peptides in all mixtures of Aß42-SST14, Aß42-AVP, and Aß40-SST14. The Aß42-SST14 mixtures were found to develop compact, dynamically stable, but small aggregates with the highest exposure of hydrophobic residues to the solvent. Differences in the morphology and dynamics of aggregates that comprise SST14 or AVP appear to reflect distinct (1) regions of the Aß chains they interact with; (2) propensities to engage in hydrogen bonds with Aß peptides; and (3) solvent exposures of hydrophilic and hydrophobic groups. The presence of SST14 was found to impede aggregation in the Aß42-SST14 system despite a high hydrophobicity, producing a stronger "sticky surface" effect in the aggregates at the onset of Aß42-SST14 oligomerization.


Subject(s)
Amyloid beta-Peptides , Molecular Dynamics Simulation , Peptide Fragments , Somatostatin , Alzheimer Disease , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Computational Biology , Humans , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Aggregation, Pathological , Somatostatin/chemistry , Somatostatin/metabolism
7.
Int J Mol Sci ; 23(23)2022 Nov 26.
Article in English | MEDLINE | ID: mdl-36499150

ABSTRACT

Several strands of investigation have established that a reduction in the levels of the cellular prion protein (PrPC) is a promising avenue for the treatment of prion diseases. We recently described an indirect approach for reducing PrPC levels that targets Na,K-ATPases (NKAs) with cardiac glycosides (CGs), causing cells to respond with the degradation of these pumps and nearby molecules, including PrPC. Because the therapeutic window of widely used CGs is narrow and their brain bioavailability is low, we set out to identify a CG with improved pharmacological properties for this indication. Starting with the CG known as oleandrin, we combined in silico modeling of CG binding poses within human NKA folds, CG structure-activity relationship (SAR) data, and predicted blood-brain barrier (BBB) penetrance scores to identify CG derivatives with improved characteristics. Focusing on C4'-dehydro-oleandrin as a chemically accessible shortlisted CG derivative, we show that it reaches four times higher levels in the brain than in the heart one day after subcutaneous administration, exhibits promising pharmacological properties, and suppresses steady-state PrPC levels by 84% in immortalized human cells that have been differentiated to acquire neural or astrocytic characteristics. Finally, we validate that the mechanism of action of this approach for reducing cell surface PrPC levels requires C4'-dehydro-oleandrin to engage with its cognate binding pocket within the NKA α subunit. The improved brain bioavailability of C4'-dehydro-oleandrin, combined with its relatively low toxicity, make this compound an attractive lead for brain CG indications and recommends its further exploration for the treatment of prion diseases.


Subject(s)
Cardiac Glycosides , Creutzfeldt-Jakob Syndrome , Prion Diseases , Prions , Humans , Prion Proteins/metabolism , Creutzfeldt-Jakob Syndrome/metabolism , Cardiac Glycosides/therapeutic use , Prions/metabolism , Prion Diseases/drug therapy , Prion Diseases/metabolism , Brain/metabolism
8.
J Biol Chem ; 294(13): 4911-4923, 2019 03 29.
Article in English | MEDLINE | ID: mdl-30705093

ABSTRACT

Prions are infectious protein aggregates that cause several fatal neurodegenerative diseases. Prion research has been hindered by a lack of cellular paradigms for studying the replication of prions from different species. Although hamster prions have been widely used to study prion replication in animals and within in vitro amplification systems, they have proved challenging to propagate in cultured cells. Because the murine catecholaminergic cell line CAD5 is susceptible to a diverse range of mouse prion strains, we hypothesized that it might also be capable of propagating nonmouse prions. Here, using CRISPR/Cas9-mediated genome engineering, we demonstrate that CAD5 cells lacking endogenous mouse PrP expression (CAD5-PrP-/- cells) can be chronically infected with hamster prions following stable expression of hamster PrP. When exposed to the 263K, HY, or 139H hamster prion strains, these cells stably propagated high levels of protease-resistant PrP. Hamster prion replication required absence of mouse PrP, and hamster PrP inhibited the propagation of mouse prions. Cellular homogenates from 263K-infected cells exhibited prion seeding activity in the RT-QuIC assay and were infectious to naïve cells expressing hamster PrP. Interestingly, murine N2a neuroblastoma cells ablated for endogenous PrP expression were susceptible to mouse prions, but not hamster prions upon expression of cognate PrP, suggesting that CAD5 cells either possess cellular factors that enhance or lack factors that restrict the diversity of prion strains that can be propagated. We conclude that transfected CAD5-PrP-/- cells may be a useful tool for assessing the biology of prion strains and dissecting the mechanism of prion replication.


Subject(s)
Prions/metabolism , Animals , CRISPR-Cas Systems , Cell Line, Tumor , Cricetinae , Gene Editing , Mice , Prions/genetics
9.
Mol Cell Proteomics ; 14(11): 3000-14, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26269332

ABSTRACT

The tau protein is central to the etiology of several neurodegenerative diseases, including Alzheimer's disease, a subset of frontotemporal dementias, progressive supranuclear palsy and dementia following traumatic brain injury, yet the proteins it interacts with have not been studied using a systematic discovery approach. Here we employed mild in vivo crosslinking, isobaric labeling, and tandem mass spectrometry to characterize molecular interactions of human tau in a neuroblastoma cell model. The study revealed a robust association of tau with the ribonucleoproteome, including major protein complexes involved in RNA processing and translation, and documented binding of tau to several heat shock proteins, the proteasome and microtubule-associated proteins. Follow-up experiments determined the relative contribution of cellular RNA to the tau interactome and mapped interactions to N- or C-terminal tau domains. We further document that expression of P301L mutant tau disrupts interactions of the C-terminal half of tau with heat shock proteins and the proteasome. The data are consistent with a model whereby a higher propensity of P301L mutant tau to aggregate may reflect a perturbation of its chaperone-assisted stabilization and proteasome-dependent degradation. Finally, using a global proteomics approach, we show that heterologous expression of a tau construct that lacks the C-terminal domain, including the microtubule binding domain, does not cause a discernible shift of the proteome except for a significant direct correlation of steady-state levels of tau and cystatin B.


Subject(s)
Epithelial Cells/metabolism , Molecular Chaperones/metabolism , Neurons/metabolism , Proteasome Endopeptidase Complex/metabolism , Ribonucleoproteins/metabolism , tau Proteins/metabolism , Animals , Binding Sites , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , Cystatin B/genetics , Cystatin B/metabolism , Epithelial Cells/cytology , Gene Expression Regulation , HEK293 Cells , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Molecular Chaperones/genetics , Molecular Sequence Annotation , Mutation , Neurons/cytology , Protein Binding , Protein Interaction Mapping , Protein Structure, Tertiary , Ribonucleoproteins/genetics , Signal Transduction , tau Proteins/genetics
10.
Biochem J ; 473(16): 2531-44, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27274087

ABSTRACT

There is growing evidence that zinc and its transporters are involved in cell migration during development and in cancer. In the present study, we show that zinc transporter ZIP10 (SLC39A10) stimulates cell motility and proliferation, both in mammalian cells and in the zebrafish embryo. This is associated with inactivation of GSK (glycogen synthase kinase)-3α and -3ß and down-regulation of E-cadherin (CDH1). Morpholino-mediated knockdown of zip10 causes delayed epiboly and deformities of the head, eye, heart and tail. Furthermore, zip10 deficiency results in overexpression of cdh1, zip6 and stat3, the latter gene product driving transcription of both zip6 and zip10 The non-redundant requirement of Zip6 and Zip10 for epithelial to mesenchymal transition (EMT) is consistent with our finding that they exist as a heteromer. We postulate that a subset of ZIPs carrying prion protein (PrP)-like ectodomains, including ZIP6 and ZIP10, are integral to cellular pathways and plasticity programmes, such as EMT.


Subject(s)
Cation Transport Proteins/metabolism , Cell Movement , Embryonic Development , Zinc/metabolism , Animals , CHO Cells , Cation Transport Proteins/classification , Cell Adhesion , Cell Proliferation , Clustered Regularly Interspaced Short Palindromic Repeats , Cricetulus , Epithelial-Mesenchymal Transition , Female , Humans , MCF-7 Cells , Male , Phylogeny , Zebrafish/embryology
11.
Nat Genet ; 39(2): 168-77, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17220890

ABSTRACT

The recycling of the amyloid precursor protein (APP) from the cell surface via the endocytic pathways plays a key role in the generation of amyloid beta peptide (Abeta) in Alzheimer disease. We report here that inherited variants in the SORL1 neuronal sorting receptor are associated with late-onset Alzheimer disease. These variants, which occur in at least two different clusters of intronic sequences within the SORL1 gene (also known as LR11 or SORLA) may regulate tissue-specific expression of SORL1. We also show that SORL1 directs trafficking of APP into recycling pathways and that when SORL1 is underexpressed, APP is sorted into Abeta-generating compartments. These data suggest that inherited or acquired changes in SORL1 expression or function are mechanistically involved in causing Alzheimer disease.


Subject(s)
Alzheimer Disease/genetics , LDL-Receptor Related Proteins/genetics , Membrane Transport Proteins/genetics , Age of Onset , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Cell Line , Endosomes/metabolism , Genetic Variation , Haplotypes , Humans , Introns , Models, Genetic , Organ Specificity , Polymorphism, Single Nucleotide , Protease Nexins , Receptors, Cell Surface/metabolism , Vesicular Transport Proteins/metabolism
12.
J Biol Chem ; 288(21): 15352-66, 2013 May 24.
Article in English | MEDLINE | ID: mdl-23589300

ABSTRACT

γ-Secretase plays a pivotal role in the production of neurotoxic amyloid ß-peptides (Aß) in Alzheimer disease (AD) and consists of a heterotetrameric core complex that includes the aspartyl intramembrane protease presenilin (PS). The human genome codes for two presenilin paralogs. To understand the causes for distinct phenotypes of PS paralog-deficient mice and elucidate whether PS mutations associated with early-onset AD affect the molecular environment of mature γ-secretase complexes, quantitative interactome comparisons were undertaken. Brains of mice engineered to express wild-type or mutant PS1, or HEK293 cells stably expressing PS paralogs with N-terminal tandem-affinity purification tags served as biological source materials. The analyses revealed novel interactions of the γ-secretase core complex with a molecular machinery that targets and fuses synaptic vesicles to cellular membranes and with the H(+)-transporting lysosomal ATPase macrocomplex but uncovered no differences in the interactomes of wild-type and mutant PS1. The catenin/cadherin network was almost exclusively found associated with PS1. Another intramembrane protease, signal peptide peptidase, predominantly co-purified with PS2-containing γ-secretase complexes and was observed to influence Aß production.


Subject(s)
Alzheimer Disease/enzymology , Amyloid Precursor Protein Secretases/immunology , Membrane Proteins/metabolism , Presenilin-2/metabolism , Serine Endopeptidases/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Cadherins/genetics , Cadherins/metabolism , Catenins/genetics , Catenins/metabolism , HEK293 Cells , Humans , Membrane Proteins/genetics , Mice , Mice, Transgenic , Mutation , Presenilin-2/genetics , Protein Binding/genetics , Serine Endopeptidases/genetics , Vacuolar Proton-Translocating ATPases/genetics , Vacuolar Proton-Translocating ATPases/metabolism
13.
J Biol Chem ; 288(52): 37241-55, 2013 Dec 27.
Article in English | MEDLINE | ID: mdl-24225951

ABSTRACT

Widely expressed in the adult central nervous system, the cellular prion protein (PrP(C)) is implicated in a variety of processes, including neuronal excitability. Dipeptidyl aminopeptidase-like protein 6 (DPP6) was first identified as a PrP(C) interactor using in vivo formaldehyde cross-linking of wild type (WT) mouse brain. This finding was confirmed in three cell lines and, because DPP6 directs the functional assembly of K(+) channels, we assessed the impact of WT and mutant PrP(C) upon Kv4.2-based cell surface macromolecular complexes. Whereas a Gerstmann-Sträussler-Scheinker disease version of PrP with eight extra octarepeats was a loss of function both for complex formation and for modulation of Kv4.2 channels, WT PrP(C), in a DPP6-dependent manner, modulated Kv4.2 channel properties, causing an increase in peak amplitude, a rightward shift of the voltage-dependent steady-state inactivation curve, a slower inactivation, and a faster recovery from steady-state inactivation. Thus, the net impact of wt PrP(C) was one of enhancement, which plays a critical role in the down-regulation of neuronal membrane excitability and is associated with a decreased susceptibility to seizures. Insofar as previous work has established a requirement for WT PrP(C) in the Aß-dependent modulation of excitability in cholinergic basal forebrain neurons, our findings implicate PrP(C) regulation of Kv4.2 channels as a mechanism contributing to the effects of oligomeric Aß upon neuronal excitability and viability.


Subject(s)
Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Potassium Channels/metabolism , PrPC Proteins/metabolism , Prosencephalon/metabolism , Shal Potassium Channels/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Cell Membrane/genetics , Cell Membrane/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , HEK293 Cells , Humans , Membrane Potentials/physiology , Mice , Mice, Mutant Strains , Mutation , Nerve Tissue Proteins/genetics , Neurons/cytology , Potassium Channels/genetics , PrPC Proteins/genetics , Prosencephalon/cytology , Shal Potassium Channels/genetics
14.
PLoS Pathog ; 7(11): e1002391, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22114562

ABSTRACT

During prion infections of the central nervous system (CNS) the cellular prion protein, PrP(C), is templated to a conformationally distinct form, PrP(Sc). Recent studies have demonstrated that the Sprn gene encodes a GPI-linked glycoprotein Shadoo (Sho), which localizes to a similar membrane environment as PrP(C) and is reduced in the brains of rodents with terminal prion disease. Here, analyses of prion-infected mice revealed that down-regulation of Sho protein was not related to Sprn mRNA abundance at any stage in prion infection. Down-regulation was robust upon propagation of a variety of prion strains in Prnp(a) and Prnp(b) mice, with the exception of the mouse-adapted BSE strain 301 V. In addition, Sho encoded by a TgSprn transgene was down-regulated to the same extent as endogenous Sho. Reduced Sho levels were not seen in a tauopathy, in chemically induced spongiform degeneration or in transgenic mice expressing the extracellular ADan amyloid peptide of familial Danish dementia. Insofar as prion-infected Prnp hemizygous mice exhibited accumulation of PrP(Sc) and down-regulation of Sho hundreds of days prior to onset of neurologic symptoms, Sho depletion can be excluded as an important trigger for clinical disease or as a simple consequence of neuronal damage. These studies instead define a disease-specific effect, and we hypothesize that membrane-associated Sho comprises a bystander substrate for processes degrading PrP(Sc). Thus, while protease-resistant PrP detected by in vitro digestion allows post mortem diagnosis, decreased levels of endogenous Sho may trace an early response to PrP(Sc) accumulation that operates in the CNS in vivo. This cellular response may offer new insights into the homeostatic mechanisms involved in detection and clearance of the misfolded proteins that drive prion disease pathogenesis.


Subject(s)
Brain/metabolism , Nerve Tissue Proteins/genetics , PrPSc Proteins/biosynthesis , Prion Diseases/metabolism , Animals , Down-Regulation , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/genetics , Mice , Mice, Transgenic , Nerve Tissue Proteins/biosynthesis , PrPC Proteins/metabolism , RNA, Messenger/metabolism
15.
Cell Death Dis ; 14(4): 240, 2023 04 05.
Article in English | MEDLINE | ID: mdl-37019881

ABSTRACT

Transcriptional regulators encoded by the Myocyte Enhancer Factor 2 (MEF2) gene family play a fundamental role in cardiac development, homeostasis and pathology. Previous studies indicate that MEF2A protein-protein interactions serve as a network hub in several cardiomyocyte cellular processes. Based on the idea that interactions with regulatory protein partners underly the diverse roles of MEF2A in cardiomyocyte gene expression, we undertook a systematic unbiased screen of the MEF2A protein interactome in primary cardiomyocytes using an affinity purification-based quantitative mass spectrometry approach. Bioinformatic processing of the MEF2A interactome revealed protein networks involved in the regulation of programmed cell death, inflammatory responses, actin dynamics and stress signaling in primary cardiomyocytes. Further biochemical and functional confirmation of specific protein-protein interactions documented a dynamic interaction between MEF2A and STAT3 proteins. Integration of transcriptome level data from MEF2A and STAT3-depleted cardiomyocytes reveals that the balance between MEF2A and STAT3 activity exerts a level of executive control over the inflammatory response and cardiomyocyte cell survival and experimentally ameliorates Phenylephrine induced cardiomyocyte hypertrophy. Lastly, we identified several MEF2A/STAT3 co-regulated genes, including the MMP9 gene. Herein, we document the cardiomyocyte MEF2A interactome, which furthers our understanding of protein networks involved in the hierarchical control of normal and pathophysiological cardiomyocyte gene expression in the mammalian heart.


Subject(s)
Myocytes, Cardiac , Signal Transduction , Animals , MEF2 Transcription Factors/metabolism , Myocytes, Cardiac/metabolism , Mammals
16.
Sci Rep ; 13(1): 2337, 2023 02 09.
Article in English | MEDLINE | ID: mdl-36759538

ABSTRACT

The neuroendocrine peptide somatostatin (SST) has long been thought of as influencing the deposition of the amyloid ß peptide (Aß) in Alzheimer's disease (AD). Missing have been in vivo data in a relevant Aß amyloidosis model. Here we crossed AppNL-F/NL-F mice with Sst-deficient mice to assess if and how the presence of Sst influences pathological hallmarks of Aß amyloidosis. We found that Sst had no influence on whole brain neprilysin transcript, protein or activity levels, an observation that cannot be accounted for by a compensatory upregulation of the Sst paralog, cortistatin (Cort), that we observed in 15-month-old Sst-deficient mice. Sst-deficiency led to a subtle but significant increase in the density of cortical Aß amyloid plaques. Follow-on western blot analyses of whole brain extracts indicated that Sst interferes with early steps of Aß assembly that manifest in the appearance of SDS-stable smears of 55-150 kDa in Sst null brain samples. As expected, no effect of Sst on tau steady-state levels or its phosphorylation were observed. Results from this study are easier reconciled with an emerging body of data that point toward Sst affecting Aß amyloid plaque formation through direct interference with Aß aggregation rather than through its effects on neprilysin expression.


Subject(s)
Alzheimer Disease , Amyloidosis , Mice , Animals , Amyloid beta-Peptides/metabolism , Plaque, Amyloid/pathology , Neprilysin/genetics , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Amyloidosis/pathology , Somatostatin/metabolism , Mice, Transgenic , Disease Models, Animal
17.
Biomedicines ; 11(10)2023 Oct 08.
Article in English | MEDLINE | ID: mdl-37893099

ABSTRACT

Recombinant adeno-associated virus (rAAV) vectors are gene therapy delivery tools that offer a promising platform for the treatment of neurodegenerative diseases. Keeping up with developments in this fast-moving area of research is a challenge. This review was thus written with the intention to introduce this field of study to those who are new to it and direct others who are struggling to stay abreast of the literature towards notable recent studies. In ten sections, we briefly highlight early milestones within this field and its first clinical success stories. We showcase current clinical trials, which focus on gene replacement, gene augmentation, or gene suppression strategies. Next, we discuss ongoing efforts to improve the tropism of rAAV vectors for brain applications and introduce pre-clinical research directed toward harnessing rAAV vectors for gene editing applications. Subsequently, we present common genetic elements coded by the single-stranded DNA of rAAV vectors, their so-called payloads. Our focus is on recent advances that are bound to increase treatment efficacies. As needed, we included studies outside the neurodegenerative disease field that showcased improved pre-clinical designs of all-in-one rAAV vectors for gene editing applications. Finally, we discuss risks associated with off-target effects and inadvertent immunogenicity that these technologies harbor as well as the mitigation strategies available to date to make their application safer.

18.
Proteome Sci ; 10(1): 33, 2012 May 18.
Article in English | MEDLINE | ID: mdl-22607704

ABSTRACT

BACKGROUND: Signal peptide peptidase (SPP), a member of the presenilin-like intra-membrane cleaving aspartyl protease family, migrates on Blue Native (BN) gels as 100 kDa, 200 kDa and 450 kDa species. SPP has recently been implicated in other non-proteolytic functions such as retro-translocation of MHC Class I molecules and binding of misfolded proteins in the endoplasmic reticulum (ER). These high molecular weight SPP complexes might contain additional proteins that regulate the proteolytic activity of SPP or support its non-catalytic functions. RESULTS: In this study, an unbiased iTRAQ-labeling mass spectrometry approach was used to identify SPP-interacting proteins. We found that vigilin, a ubiquitous multi-KH domain containing cytoplasmic protein involved in RNA binding and protein translation control, selectively enriched with SPP. Vigilin interacted with SPP and both proteins co-localized in restricted intracellular domains near the ER, biochemically co-fractionated and were part of the same 450 kDa complex on BN gels. However, vigilin does not alter the protease activity of SPP, suggesting that the SPP-vigilin interaction might be involved in the non-proteolytic functions of SPP. CONCLUSIONS: We have identified and validated vigilin as a novel interacting partner of SPP that could play an important role in the non-proteolytic functions of SPP. This data adds further weight to the idea that intramembrane-cleaving aspartyl proteases, such as presenilin and SPPs, could have other functions besides the proteolysis of short membrane stubs.

19.
Brain ; 139(Pt 10): 2586-2589, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27671028
20.
Nature ; 440(7088): 1208-12, 2006 Apr 27.
Article in English | MEDLINE | ID: mdl-16641999

ABSTRACT

The presenilin proteins (PS1 and PS2) and their interacting partners nicastrin, aph-1 (refs 4, 5) and pen-2 (ref. 5) form a series of high-molecular-mass, membrane-bound protein complexes that are necessary for gamma-secretase and epsilon-secretase cleavage of selected type 1 transmembrane proteins, including the amyloid precursor protein, Notch and cadherins. Modest cleavage activity can be generated by reconstituting these four proteins in yeast and Spodoptera frugiperda (sf9) cells. However, a critical but unanswered question about the biology of the presenilin complexes is how their activity is modulated in terms of substrate specificity and/or relative activities at the gamma and epsilon sites. A corollary to this question is whether additional proteins in the presenilin complexes might subsume these putative regulatory functions. The hypothesis that additional proteins might exist in the presenilin complexes is supported by the fact that enzymatically active complexes have a mass that is much greater than predicted for a 1:1:1:1 stoichiometric complex (at least 650 kDa observed, compared with about 220 kDa predicted). To address these questions we undertook a search for presenilin-interacting proteins that differentially affected gamma- and epsilon-site cleavage events. Here we report that TMP21, a member of the p24 cargo protein family, is a component of presenilin complexes and differentially regulates gamma-secretase cleavage without affecting epsilon-secretase activity.


Subject(s)
Endopeptidases/metabolism , Membrane Proteins/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Amyloid Precursor Protein Secretases , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Peptides/genetics , Animals , Aspartic Acid Endopeptidases , Cell Line , Endopeptidases/chemistry , Humans , Membrane Proteins/chemistry , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Models, Biological , Nucleocytoplasmic Transport Proteins , Presenilin-1 , Presenilin-2 , Protein Binding , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL