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










Publication year range
1.
Redox Biol ; 67: 102943, 2023 11.
Article in English | MEDLINE | ID: mdl-37883843

ABSTRACT

Accumulation of misfolded proteins or perturbation of calcium homeostasis leads to endoplasmic reticulum (ER) stress and is linked to the pathogenesis of neurodegenerative diseases. Hence, understanding the ability of neuronal cells to cope with chronic ER stress is of fundamental interest. Interestingly, several brain areas uphold functions that enable them to resist challenges associated with neurodegeneration. Here, we established novel clonal mouse hippocampal (HT22) cell lines that are resistant to prolonged (chronic) ER stress induced by thapsigargin (TgR) or tunicamycin (TmR) as in vitro models to study the adaption to ER stress. Morphologically, we observed a significant increase in vesicular und autophagosomal structures in both resistant lines and 'giant lysosomes', especially striking in TgR cells. While autophagic activity increased under ER stress, lysosomal function appeared slightly impaired; in both cell lines, we observed enhanced ER-phagy. However, proteomic analyses revealed that various protein clusters and signaling pathways were differentially regulated in TgR versus TmR cells in response to chronic ER stress. Additionally, bioenergetic analyses in both resistant cell lines showed a shift toward aerobic glycolysis ('Warburg effect') and a defective complex I of the oxidative phosphorylation (OXPHOS) machinery. Furthermore, ER stress-resistant cells differentially activated the unfolded protein response (UPR) comprising IRE1α and ATF6 pathways. These findings display the wide portfolio of adaptive responses of neuronal cells to chronic ER stress. ER stress-resistant neuronal cells could be the basis to uncover molecular modulators of adaptation, resistance, and neuroprotection as potential pharmacological targets for preventing neurodegeneration.


Subject(s)
Endoribonucleases , Protein Serine-Threonine Kinases , Mice , Animals , Protein Serine-Threonine Kinases/metabolism , Endoribonucleases/genetics , Proteomics , Endoplasmic Reticulum Stress , Unfolded Protein Response , Endoplasmic Reticulum/metabolism
2.
Viruses ; 14(7)2022 07 05.
Article in English | MEDLINE | ID: mdl-35891458

ABSTRACT

Human papillomaviruses (HPVs) inflict a significant burden on the human population. The clinical manifestations caused by high-risk HPV types are cancers at anogenital sites, including cervical cancer, as well as head and neck cancers. Host cell defense mechanisms such as autophagy are initiated upon HPV entry. At the same time, the virus modulates cellular antiviral processes and structures such as promyelocytic leukemia nuclear bodies (PML NBs) to enable infection. Here, we uncover the autophagy adaptor p62, also known as p62/sequestosome-1, as a novel proviral factor in infections by the high-risk HPV type 16 (HPV16). Proteomics, imaging and interaction studies of HPV16 pseudovirus-treated HeLa cells display that p62 is recruited to virus-filled endosomes, interacts with incoming capsids, and accompanies the virus to PML NBs, the sites of viral transcription and replication. Cellular depletion of p62 significantly decreased the delivery of HPV16 viral DNA to PML NBs and HPV16 infection rate. Moreover, the absence of p62 leads to an increase in the targeting of viral components to autophagic structures and enhanced degradation of the viral capsid protein L2. The proviral role of p62 and formation of virus-p62-PML hybrid bodies have also been observed in human primary keratinocytes, the HPV target cells. Together, these findings suggest the previously unrecognized virus-induced formation of p62-PML hybrid bodies as a viral mechanism to subvert the cellular antiviral defense, thus enabling viral gene expression.


Subject(s)
Human papillomavirus 16 , Papillomavirus Infections , Antiviral Agents , HeLa Cells , Human papillomavirus 16/genetics , Human papillomavirus 16/metabolism , Humans , Papillomaviridae/metabolism , Promyelocytic Leukemia Protein/genetics
3.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Article in English | MEDLINE | ID: mdl-34782467

ABSTRACT

Nondegradative ubiquitin chains attached to specific targets via Lysine 63 (K63) residues have emerged to play a fundamental role in synaptic function. The K63-specific deubiquitinase CYLD has been widely studied in immune cells and lately also in neurons. To better understand if CYLD plays a role in brain and synapse homeostasis, we analyzed the behavioral profile of CYLD-deficient mice. We found that the loss of CYLD results in major autism-like phenotypes including impaired social communication, increased repetitive behavior, and cognitive dysfunction. Furthermore, the absence of CYLD leads to a reduction in hippocampal network excitability, long-term potentiation, and pyramidal neuron spine numbers. By providing evidence that CYLD can modulate mechanistic target of rapamycin (mTOR) signaling and autophagy at the synapse, we propose that synaptic K63-linked ubiquitination processes could be fundamental in understanding the pathomechanisms underlying autism spectrum disorder.


Subject(s)
Autophagy/physiology , Hippocampus/metabolism , Signal Transduction/physiology , TOR Serine-Threonine Kinases/metabolism , Animals , Autism Spectrum Disorder , Autistic Disorder , Deubiquitinating Enzyme CYLD , Female , Lysine/metabolism , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins , Nerve Tissue Proteins , Neurons/metabolism , Synapses/metabolism , Ubiquitin/metabolism , Ubiquitination
4.
Cell Mol Life Sci ; 78(2): 645-660, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32322926

ABSTRACT

The cellular protein homeostasis (proteostasis) network responds effectively to insults. In a functional screen in C. elegans, we recently identified the gene receptor-mediated endocytosis 8 (rme-8; human ortholog: DNAJC13) as a component of the proteostasis network. Accumulation of aggregation-prone proteins, such as amyloid-ß 42 (Aß), α-synuclein, or mutant Cu/Zn-superoxide dismutase (SOD1), were aggravated upon the knockdown of rme-8/DNAJC13 in C. elegans and in human cell lines, respectively. DNAJC13 is involved in endosomal protein trafficking and associated with the retromer and the WASH complex. As both complexes have been linked to autophagy, we investigated the role of DNAJC13 in this degradative pathway. In knockdown and overexpression experiments, DNAJC13 acts as a positive modulator of autophagy. In contrast, the overexpression of the Parkinson's disease-associated mutant DNAJC13(N855S) did not enhance autophagy. Reduced DNAJC13 levels affected ATG9A localization at and its transport from the recycling endosome. As a consequence, ATG9A co-localization at LC3B-positive puncta under steady-state and autophagy-induced conditions is impaired. These data demonstrate a novel function of RME-8/DNAJC13 in cellular homeostasis by modulating ATG9A trafficking and autophagy.


Subject(s)
Autophagy , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Molecular Chaperones/metabolism , Proteostasis , Animals , Caenorhabditis elegans/cytology , HEK293 Cells , HeLa Cells , Humans , Protein Aggregates
5.
Trends Neurosci ; 43(2): 79-81, 2020 02.
Article in English | MEDLINE | ID: mdl-31918966

ABSTRACT

Neurodegenerative diseases are linked to dysfunctional proteostasis and disturbed autophagy. Here, we discuss how the sigma-1 receptor (Sig-1R) may act at the intersection of this interaction, as loss-of-function mutations of this unique chaperone are associated with defective autophagy and its pharmacological activation induces autophagic activity.


Subject(s)
Neurodegenerative Diseases , Receptors, sigma , Autophagy , Humans , Neurodegenerative Diseases/genetics , Proteostasis , Receptors, sigma/genetics , Receptors, sigma/metabolism , Sigma-1 Receptor
6.
EMBO J ; 38(9)2019 05 02.
Article in English | MEDLINE | ID: mdl-30886048

ABSTRACT

Neurodegenerative diseases are characterized by the accumulation of misfolded proteins in the brain. Insights into protein quality control mechanisms to prevent neuronal dysfunction and cell death are crucial in developing causal therapies. Here, we report that various disease-associated protein aggregates are modified by the linear ubiquitin chain assembly complex (LUBAC). HOIP, the catalytic component of LUBAC, is recruited to misfolded Huntingtin in a p97/VCP-dependent manner, resulting in the assembly of linear polyubiquitin. As a consequence, the interactive surface of misfolded Huntingtin species is shielded from unwanted interactions, for example with the low complexity sequence domain-containing transcription factor Sp1, and proteasomal degradation of misfolded Huntingtin is facilitated. Notably, all three core LUBAC components are transcriptionally regulated by Sp1, linking defective LUBAC expression to Huntington's disease. In support of a protective activity of linear ubiquitination, silencing of OTULIN, a deubiquitinase with unique specificity for linear polyubiquitin, decreases proteotoxicity, whereas silencing of HOIP has the opposite effect. These findings identify linear ubiquitination as a protein quality control mechanism and hence a novel target for disease-modifying strategies in proteinopathies.


Subject(s)
Huntingtin Protein/metabolism , Huntington Disease/metabolism , Polyubiquitin/metabolism , Protein Processing, Post-Translational , Sp1 Transcription Factor/metabolism , Valosin Containing Protein/metabolism , Adult , Aged , Animals , Brain/metabolism , Brain/pathology , Case-Control Studies , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Huntingtin Protein/genetics , Huntington Disease/genetics , Huntington Disease/pathology , Male , Mice , Mice, Knockout , Middle Aged , NF-kappa B/genetics , NF-kappa B/metabolism , Neurons/metabolism , Neurons/pathology , Protein Binding , Protein Interaction Domains and Motifs , Signal Transduction , Sp1 Transcription Factor/genetics , Ubiquitination , Valosin Containing Protein/genetics
7.
Neurobiol Dis ; 96: 294-311, 2016 12.
Article in English | MEDLINE | ID: mdl-27629805

ABSTRACT

Peripheral or central nerve injury is a frequent cause of chronic pain and the mechanisms are not fully understood. Using newly generated transgenic mice we show that progranulin overexpression in sensory neurons attenuates neuropathic pain after sciatic nerve injury and accelerates nerve healing. A yeast-2-hybrid screen revealed putative interactions of progranulin with autophagy-related proteins, ATG12 and ATG4b. This was supported by colocalization and proteomic studies showing regulations of ATG13 and ATG4b and other members of the autophagy network, lysosomal proteins and proteins involved in endocytosis. The association of progranulin with the autophagic pathway was functionally confirmed in primary sensory neurons. Autophagy and survival were impaired in progranulin-deficient neurons and improved in progranulin overexpressing neurons. Nerve injury in vivo caused an accumulation of LC3b-EGFP positive bodies in neurons of the dorsal root ganglia and nerves suggesting an impairment of autophagic flux. Overexpression of progranulin in these neurons was associated with a reduction of the stress marker ATF3, fewer protein aggregates in the injured nerve and enhanced stump healing. At the behavioral level, further inhibition of the autophagic flux by hydroxychloroquine intensified cold and heat nociception after sciatic nerve injury and offset the pain protection provided by progranulin. We infer that progranulin may assist in removal of protein waste and thereby helps to resolve neuropathic pain after nerve injury.


Subject(s)
Autophagy/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Neuralgia/pathology , Sensory Receptor Cells/metabolism , Activating Transcription Factor 3/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Proteins/metabolism , CD11b Antigen/metabolism , Calcium-Binding Proteins/metabolism , Cysteine Endopeptidases/metabolism , Disease Models, Animal , Ganglia, Spinal/pathology , Gene Ontology , Granulins , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Lysosomal-Associated Membrane Protein 1/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Pain Measurement , Progranulins
8.
J Cereb Blood Flow Metab ; 35(7): 1112-21, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25853911

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder characterized by progressive paralysis due to the loss of primary and secondary motor neurons. Mutations in the Cu/Zn-superoxide dismutase (SOD1) gene are associated with familial ALS and to date numerous hypotheses for ALS pathology exist including impairment of the blood-spinal cord barrier. In transgenic mice carrying mutated SOD1 genes, a disrupted blood-spinal cord barrier as well as decreased levels of tight junction (TJ) proteins ZO-1, occludin, and claudin-5 were detected. Here, we examined TJ protein levels and barrier function of primary blood-spinal cord barrier endothelial cells of presymptomatic hSOD1(G93A) mice and bEnd.3 cells stably expressing hSOD1(G93A). In both cellular systems, we observed reduced claudin-5 levels and a decreased transendothelial resistance (TER) as well as an increased apparent permeability. Analysis of the ß-catenin/AKT/forkhead box protein O1 (FoxO1) pathway and the FoxO1-regulated activity of the claudin-5 promoter revealed a repression of the claudin-5 gene expression in hSOD1(G93A) cells, which was depended on the phosphorylation status of FoxO1. These results strongly indicate that mutated SOD1 affects the expression and localization of TJ proteins leading to impaired integrity and breakdown of the blood-spinal cord barrier.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Claudin-5/genetics , Gene Expression Regulation , Spinal Cord/blood supply , Spinal Cord/pathology , Superoxide Dismutase/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Cell Line , Cells, Cultured , Claudin-5/analysis , Claudin-5/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Mice , Mice, Transgenic , Signal Transduction , Spinal Cord/metabolism , Superoxide Dismutase/metabolism , Tight Junction Proteins/analysis , Tight Junction Proteins/genetics , Tight Junction Proteins/metabolism
9.
Sci Rep ; 5: 8585, 2015 Feb 26.
Article in English | MEDLINE | ID: mdl-25717019

ABSTRACT

Low-frequency magnetic fields (LF-MF) generated by power lines represent a potential environmental health risk and are classified as possibly carcinogenic by the World Health Organization. Epidemiological studies indicate that LF-MF might propagate neurodegenerative diseases like Alzheimer's disease (AD) or amyotrophic lateral sclerosis (ALS). We conducted a comprehensive analysis to determine whether long-term exposure to LF-MF (50 Hz, 1 mT) interferes with disease development in established mouse models for AD and ALS, namely APP23 mice and mice expressing mutant Cu/Zn-superoxide dismutase (SOD1), respectively. Exposure for 16 months did not aggravate learning deficit of APP23 mice. Likewise, disease onset and survival of SOD1(G85R) or SOD1(G93A) mice were not altered upon LF-MF exposure for ten or eight months, respectively. These results and an extended biochemical analysis of protein aggregation, glial activation and levels of toxic protein species suggests that LF-MF do not affect cellular processes involved in the pathogenesis of AD or ALS.


Subject(s)
Alzheimer Disease/pathology , Amyotrophic Lateral Sclerosis/pathology , Magnetic Fields/adverse effects , Alzheimer Disease/metabolism , Alzheimer Disease/psychology , Amyloid beta-Protein Precursor/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/psychology , Animals , Brain/metabolism , Brain/pathology , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Disease Progression , Female , Glial Fibrillary Acidic Protein , Male , Mice, Transgenic , Microfilament Proteins/metabolism , Microglia/physiology , Nerve Tissue Proteins/metabolism , Oxidative Stress , Protein Aggregation, Pathological/metabolism , Spatial Learning , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
10.
Autophagy ; 10(12): 2297-309, 2014.
Article in English | MEDLINE | ID: mdl-25495476

ABSTRACT

Macroautophagy is a degradative pathway that sequesters and transports cytosolic cargo in autophagosomes to lysosomes, and its deterioration affects intracellular proteostasis. Membrane dynamics accompanying autophagy are mostly elusive and depend on trafficking processes. RAB GTPase activating proteins (RABGAPs) are important factors for the coordination of cellular vesicle transport systems, and several TBC (TRE2-BUB2-CDC16) domain-containing RABGAPs are associated with autophagy. Employing C. elegans and human primary fibroblasts, we show that RAB3GAP1 and RAB3GAP2, which are components of the TBC domain-free RAB3GAP complex, influence protein aggregation and affect autophagy at basal and rapamycin-induced conditions. Correlating the activity of RAB3GAP1/2 with ATG3 and ATG16L1 and analyzing ATG5 punctate structures, we illustrate that the RAB3GAPs modulate autophagosomal biogenesis. Significant levels of RAB3GAP1/2 colocalize with members of the Atg8 family at lipid droplets, and their autophagy modulatory activity depends on the GTPase-activating activity of RAB3GAP1 but is independent of the RAB GTPase RAB3. Moreover, we analyzed RAB3GAP1/2 in relation to the previously reported suppressive autophagy modulators FEZ1 and FEZ2 and demonstrate that both reciprocally regulate autophagy. In conclusion, we identify RAB3GAP1/2 as novel conserved factors of the autophagy and proteostasis network.


Subject(s)
Autophagy/drug effects , GTPase-Activating Proteins/metabolism , Sirolimus/pharmacology , rab3 GTP-Binding Proteins/metabolism , Animals , Autophagy/physiology , Biological Transport/physiology , Caenorhabditis elegans , Humans , Lysosomes/metabolism , Phagosomes/metabolism
11.
Free Radic Biol Med ; 72: 41-54, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24742816

ABSTRACT

Redox control of proteins that form disulfide bonds upon oxidative challenge is an emerging topic in the physiological and pathophysiological regulation of protein function. We have investigated the role of the neuronal calcium sensor protein visinin-like protein 1 (VILIP-1) as a novel redox sensor in a cellular system. We have found oxidative stress to trigger dimerization of VILIP-1 within a cellular environment and identified thioredoxin reductase as responsible for facilitating the remonomerization of the dimeric protein. Dimerization is modulated by calcium and not dependent on the myristoylation of VILIP-1. Furthermore, we show by site-directed mutagenesis that dimerization is exclusively mediated by Cys187. As a functional consequence, VILIP-1 dimerization modulates the sensitivity of cells to an oxidative challenge. We have investigated whether dimerization of VILIP-1 occurs in two different animal models of amyotrophic lateral sclerosis (ALS) and detected soluble VILIP-1 dimers to be significantly enriched in the spinal cord from phenotypic disease onset onwards. Moreover, VILIP-1 is part of the ALS-specific protein aggregates. We show for the first time that the C-terminus of VILIP-1, containing Cys187, might represent a novel redox-sensitive motif and that VILIP-1 dimerization and aggregation are hallmarks of ALS. This suggests that VILIP-1 dimers play a functional role in integrating the cytosolic calcium concentration and the oxidative status of the cell. Furthermore, a loss of VILIP-1 function owing to protein aggregation in ALS could be relevant in the pathophysiology of the disease.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Calcium/metabolism , Neurocalcin/metabolism , Oxidative Stress/physiology , Protein Multimerization , Amino Acid Motifs , Animals , Blotting, Western , Disease Models, Animal , HEK293 Cells , Humans , Immunohistochemistry , Mass Spectrometry , Mice , Mice, Transgenic , Mutagenesis, Site-Directed , Oxidation-Reduction , Superoxide Dismutase/genetics , Superoxide Dismutase-1
12.
Neurobiol Dis ; 62: 479-88, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24200866

ABSTRACT

Mutations in the gene encoding Cu/Zn superoxide dismutase (SOD1) are responsible for a subset of amyotrophic lateral sclerosis cases presumably by the acquisition of as yet unknown toxic properties. Additional overexpression of wild-type SOD1 in mutant SOD1 transgenic mice did not improve but rather accelerated the disease course. Recently, it was documented that the presence of wild-type SOD1 (SOD(WT)) reduced the aggregation propensity of mutant SOD1 by the formation of heterodimers between mutant and SOD1(WT) and that these heterodimers displayed at least a similar toxicity in cellular and animal models. In this study we investigated the biochemical and biophysical properties of obligate SOD1 dimers that were connected by a peptide linker. Circular dichroism spectra indicate an increased number of unstructured residues in SOD1 mutants. However, SOD1(WT) stabilized the folding of heterodimers compared to mutant homodimers as evidenced by an increase in resistance against proteolytic degradation. Heterodimerization also reduced the affinity of mutant SOD1 to antibodies detecting misfolded SOD1. In addition, the formation of obligate dimers resulted in a detection of substantial dismutase activity even of the relatively labile SOD1(G85R) mutant. These data indicate that soluble, dismutase-active SOD1 dimers might contribute at least partially to mutant SOD1 toxicity.


Subject(s)
Superoxide Dismutase/chemistry , HEK293 Cells , Humans , Mutation , Protein Multimerization , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
13.
J Cell Sci ; 126(Pt 2): 580-92, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23178947

ABSTRACT

Aggregation of misfolded proteins and the associated loss of neurons are considered a hallmark of numerous neurodegenerative diseases. Optineurin is present in protein inclusions observed in various neurodegenerative diseases including amyotrophic lateral sclerosis (ALS), Huntington's disease, Alzheimer's disease, Parkinson's disease, Creutzfeld-Jacob disease and Pick's disease. Optineurin deletion mutations have also been described in ALS patients. However, the role of optineurin in mechanisms of protein aggregation remains unclear. In this report, we demonstrate that optineurin recognizes various protein aggregates via its C-terminal coiled-coil domain in a ubiquitin-independent manner. We also show that optineurin depletion significantly increases protein aggregation in HeLa cells and that morpholino-silencing of the optineurin ortholog in zebrafish causes the motor axonopathy phenotype similar to a zebrafish model of ALS. A more severe phenotype is observed when optineurin is depleted in zebrafish carrying ALS mutations. Furthermore, TANK1 binding kinase 1 (TBK1) is colocalized with optineurin on protein aggregates and is important in clearance of protein aggregates through the autophagy-lysosome pathway. TBK1 phosphorylates optineurin at serine 177 and regulates its ability to interact with autophagy modifiers. This study provides evidence for a ubiquitin-independent function of optineurin in autophagic clearance of protein aggregates as well as additional relevance for TBK1 as an upstream regulator of the autophagic pathway.


Subject(s)
Neurodegenerative Diseases/metabolism , Transcription Factor TFIIIA/metabolism , Ubiquitin/metabolism , Animals , Autophagy/physiology , Cell Cycle Proteins , Disease Models, Animal , HeLa Cells , Humans , Membrane Transport Proteins , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurodegenerative Diseases/genetics , Phosphorylation , Protein Binding , Zebrafish
15.
EMBO Rep ; 12(2): 149-56, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21252941

ABSTRACT

Increasing evidence indicates the existence of selective autophagy pathways, but the manner in which substrates are recognized and targeted to the autophagy system is poorly understood. One strategy is transport of a particular substrate to the aggresome, a perinuclear compartment with high autophagic activity. In this paper, we identify a new cellular pathway that uses the specificity of heat-shock protein 70 (Hsp70) to misfolded proteins as the basis for aggresome-targeting and autophagic degradation. This pathway is regulated by the stress-induced co-chaperone Bcl-2-associated athanogene 3 (BAG3), which interacts with the microtubule-motor dynein and selectively directs Hsp70 substrates to the motor and thereby to the aggresome. Notably, aggresome-targeting by BAG3 is distinct from previously described mechanisms, as it does not depend on substrate ubiquitination.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Autophagy , HSP70 Heat-Shock Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis Regulatory Proteins , COS Cells , Chlorocebus aethiops , Dyneins/metabolism , Green Fluorescent Proteins/genetics , HEK293 Cells , Humans , Inclusion Bodies/metabolism , Inclusion Bodies/ultrastructure , Mice , Mice, Transgenic , Motor Neurons/metabolism , Point Mutation , Proteasome Inhibitors , Protein Folding , Protein Transport , Recombinant Fusion Proteins/genetics , Sequence Deletion , Spinal Cord/cytology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
16.
J Biol Chem ; 285(32): 24398-403, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20530481

ABSTRACT

The human enzyme paraoxonase-2 (PON2) has two functions, an enzymatic lactonase activity and the reduction of intracellular oxidative stress. As a lactonase, it dominantly hydrolyzes bacterial signaling molecule 3OC12 and may contribute to the defense against pathogenic Pseudomonas aeruginosa. By its anti-oxidative effect, PON2 reduces cellular oxidative damage and influences redox signaling, which promotes cell survival. This may be appreciated but also deleterious given that high PON2 levels reduce atherosclerosis but may stabilize tumor cells. Here we addressed the unknown mechanisms and linkage of PON2 enzymatic and anti-oxidative function. We demonstrate that PON2 indirectly but specifically reduced superoxide release from the inner mitochondrial membrane, irrespective whether resulting from complex I or complex III of the electron transport chain. PON2 left O(2)(-) dismutase activities and cytochrome c expression unaltered, and it did not oxidize O(2)(-) but rather prevented its formation, which implies that PON2 acts by modulating quinones. To analyze linkage to hydrolytic activity, we introduced several point mutations and show that residues His(114) and His(133) are essential for PON2 activity. Further, we mapped its glycosylation sites and provide evidence that glycosylation, but not a native polymorphism Ser/Cys(311), was critical to its activity. Importantly, none of these mutations altered the anti-oxidative/anti-apoptotic function of PON2, demonstrating unrelated activities of the same protein. Collectively, our study provides detailed mechanistic insight into the functions of PON2, which is important for its role in innate immunity, atherosclerosis, and cancer.


Subject(s)
Apoptosis , Aryldialkylphosphatase/physiology , Lactones/metabolism , Mitochondria/metabolism , Superoxides/metabolism , Antioxidants/chemistry , Aryldialkylphosphatase/chemistry , Endothelium, Vascular/cytology , Glycosylation , Humans , Models, Biological , Oxidative Stress , Oxygen/chemistry , Pseudomonas aeruginosa/enzymology , Reactive Oxygen Species , Subcellular Fractions
17.
PLoS One ; 5(1): e8568, 2010 Jan 05.
Article in English | MEDLINE | ID: mdl-20052290

ABSTRACT

Protein stability under changing conditions is of vital importance for the cell and under the control of a fine-tuned network of molecular chaperones. Aging and age-related neurodegenerative diseases are directly associated with enhanced protein instability. Employing C. elegans expressing GFP-tagged luciferase as a reporter for evaluation of protein stability we show that the chaperoning strategy of body wall muscle cells and neurons is significantly different and that both are differently affected by aging. Muscle cells of young worms are largely resistant to heat stress, which is directly mediated by the stress response controlled through Heat Shock Transcription Factor 1. During recovery following heat stress the ability to refold misfolded proteins is missing. Young neurons are highly susceptible to chronic heat stress, but show a high potency to refold or disaggregate proteins during subsequent recovery. The particular proteome instability in neurons results from a delayed induction of the heat shock response. In aged neurons protein stability is increased during heat stress, whereas muscle cells show enhanced protein instability due to a deteriorated heat shock response. An efficient refolding activity is absent in both aged tissues. These results provide molecular insights into the differential protein stabilization capacity in different tissues and during aging.


Subject(s)
Aging/physiology , Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/physiology , Molecular Chaperones/physiology , Muscles/physiology , Neurons/physiology , Transcription Factors/physiology , Animals , Animals, Genetically Modified , Base Sequence , Caenorhabditis elegans Proteins/genetics , DNA Primers , Green Fluorescent Proteins/genetics , Microscopy, Fluorescence , Proteome , Transcription Factors/genetics
18.
Neurobiol Dis ; 36(2): 331-42, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19660548

ABSTRACT

Aggregation of Cu/Zn superoxide dismutase (SOD1) is a hallmark of a subset of familial amyotrophic lateral sclerosis (ALS) cases. The expression of wild-type SOD1 [SOD(hWT)] surprisingly exacerbates the phenotype of mutant SOD1 in vivo. Here we studied whether SOD1(hWT) may affect mutant SOD1 aggregation by employing fluorescence microscopy techniques combined with lifetime-based Förster resonance energy transfer (FRET). Only a very minor fraction of SOD1(hWT) was observed in aggregates induced by mutant SOD1(G37R), SOD1(G85R) or SOD1(G93C). Quite in contrast, co-expression of SOD(hWT) reduced the amount of mutant SOD1 in the aggregate fraction. Furthermore, we did not detect endogenous mouse SOD1 in aggregates formed by mutant SOD1 in two distinct mutant SOD1 mouse lines. The hypothesis that SOD1(WT) is able to keep mutant SOD1 variants in a soluble state is supported by the increased presence of heterodimers upon SOD1(hWT) co-expression. Therefore we propose that SOD1(WT) contributes to disease by heterodimerization with mutant SOD1 forms.


Subject(s)
Amyotrophic Lateral Sclerosis/enzymology , Amyotrophic Lateral Sclerosis/genetics , Mutation/physiology , Superoxide Dismutase/biosynthesis , Superoxide Dismutase/genetics , Animals , Cell Line , Humans , Mice , Mice, Transgenic , Protein Folding , Protein Multimerization/genetics , Solubility , Superoxide Dismutase/chemistry , Superoxide Dismutase-1
19.
Hum Mol Genet ; 17(10): 1373-85, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18211954

ABSTRACT

Recent studies provide evidence that wild-type Cu/Zn-superoxide dismutase (SOD1(hWT)) might be an important factor in mutant SOD1-mediated amyotrophic lateral sclerosis (ALS). In order to investigate its functional role in the pathogenesis of ALS, we designed fusion proteins of two SOD1 monomers linked by a polypeptide. We demonstrated that wild-type-like mutants, but not SOD1(G85R) homodimers, as well as mutant heterodimers including SOD1(G85R)-SOD1(hWT) display dismutase activity. Mutant homodimers showed an increased aggregation compared with the corresponding heterodimers in cell cultures and transgenic Caenorhabditis elegans, although SOD1(G85R) heterodimers are more toxic in functional assays. Our data show that (i) toxicity of mutant SOD1 is not correlated to its aggregation potential; (ii) dismutase-inactive mutants form dismutase-active heterodimers with SOD1(hWT); (iii) SOD1(hWT) can be converted to contribute to disease by forming active heterodimers. Therefore, we conclude that toxicity of mutant SOD1 is at least partially mediated through heterodimer formation with SOD1(hWT) in vivo and does not correlate with the aggregation potential of individual mutants.


Subject(s)
Amyotrophic Lateral Sclerosis/enzymology , Amyotrophic Lateral Sclerosis/genetics , Superoxide Dismutase/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans/physiology , Cell Line, Tumor , Cell Survival/drug effects , Dimerization , Humans , Hydrogen Peroxide/pharmacology , Mice , Motor Neurons/enzymology , Motor Neurons/physiology , Protein Carbonylation , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Superoxide Dismutase/chemistry , Superoxide Dismutase/genetics , Superoxide Dismutase-1
20.
J Biol Chem ; 280(41): 34924-32, 2005 Oct 14.
Article in English | MEDLINE | ID: mdl-16079146

ABSTRACT

Steroids that activate glucocorticoid receptors (GRs) and mineralocorticoid receptors have important regulatory effects on neural development, plasticity, and the body's stress response. Here, we investigated the role of corticosteroids in regulating the expression of the glial glutamate transporters glial glutamate transporter-1 (GLT-1) and glutamate-aspartate transporter (GLAST) in rat primary astrocytes. The synthetic glucocorticoid dexamethasone provoked a marked increase of GLT-1 transcription and protein levels in cortical astrocytes, whereas GLAST expression remained unaffected. Up-regulation of GLT-1 expression was accompanied by an enhanced glutamate uptake, which could be blocked by the specific GLT-1 inhibitor dihydrokainate. The promoting effect of dexamethasone on GLT-1 gene expression and function was abolished by the GR antagonist mifepristone. A predominant role of the GR was further supported by the observation that corticosterone could elevate GLT-1 expression in a dose-dependent manner, whereas aldosterone, the physiological ligand of the mineralocorticoid receptor, exerted only weak effects even when applied at high concentrations. Moreover, we monitored brain region-specific differences, since all corticosteroids used in this study failed to alter the expression of GLT-1 in midbrain and cerebellar glia, although expression levels of both corticosteroid receptor subtypes were similar in all brain regions analyzed. Dexamethasone, however, modestly enhanced GLT-1 expression in cerebellar glia in combination with the DNA methyltransferase inhibitor 5-aza-2-deoxycytidine, suggesting that suppression of GLT-1 expression in cerebellar cultures may at least in part be epigenetically mediated by a DNA methylation-dependent process. Taken together, our data highlight a potential role for glucocorticoids in regulating GLT-1 gene expression during central nervous system development or pathophysiological processes including stress.


Subject(s)
Amino Acid Transport System X-AG/biosynthesis , Brain/metabolism , Glucocorticoids/chemistry , Adrenal Cortex Hormones/metabolism , Amino Acid Transport System X-AG/metabolism , Animals , Astrocytes/cytology , Astrocytes/metabolism , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Blotting, Western , Central Nervous System/pathology , Cerebellum/pathology , Corticosterone/metabolism , Corticosterone/pharmacology , DNA Methylation , DNA Modification Methylases/metabolism , DNA Primers/chemistry , Decitabine , Detergents/pharmacology , Dexamethasone/pharmacology , Dose-Response Relationship, Drug , Fluorescent Dyes/pharmacology , Immunohistochemistry , Kainic Acid/analogs & derivatives , Kainic Acid/pharmacology , Ligands , Luciferases/metabolism , Membrane Microdomains/metabolism , Mifepristone/pharmacology , Neuroglia/pathology , Rats , Rats, Sprague-Dawley , Receptors, Mineralocorticoid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transfection , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...