Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Mol Cell Proteomics ; 20: 100065, 2021.
Article in English | MEDLINE | ID: mdl-33640490

ABSTRACT

Drosophila melanogaster has been a workhorse of genetics and cell biology for more than a century. However, proteomic-based methods have been limited due to the complexity and dynamic range of the fly proteome and the lack of efficient labeling methods. Here, we advanced a chemically defined food source into direct stable-isotope labeling of amino acids in flies (SILAF). It allows for the rapid and cost-efficient generation of a large number of larvae or flies, with full incorporation of lysine-[13C6] after six labeling days. SILAF followed by fractionation and enrichment gave proteomic insights at a depth of 7196 proteins and 8451 phosphorylation sites, which substantiated metabolic regulation on enzymatic level. We applied SILAF to quantify the mitochondrial phosphoproteome of an early-stage leucine-rich PPR motif-containing protein (LRPPRC)-knockdown fly model of mitochondrial disease that almost exclusively affects protein levels of the oxidative phosphorylation (OXPHOS) system. While the mitochondrial compartment was hypo-phosphorylated, two conserved phosphosites on OXPHOS subunits NDUFB10 and NDUFA4 were significantly upregulated upon impaired OXPHOS function. The ease and versatility of the method actuate the fruit fly as an appealing model in proteomic and posttranslational modification studies, and it enlarges potential metabolic applications based on heavy amino acid diets.


Subject(s)
Drosophila Proteins/metabolism , Mitochondrial Proteins/metabolism , Phosphoproteins/metabolism , Amino Acids/metabolism , Animals , Drosophila melanogaster , Female , Isotope Labeling , Male , Phosphorylation , Proteome
2.
Mol Psychiatry ; 26(4): 1410-1423, 2021 04.
Article in English | MEDLINE | ID: mdl-31520067

ABSTRACT

Aggregation and accumulation of amyloid beta (Aß) are believed to play a key role in the pathogenesis of Alzheimer's disease (AD). We previously reported that Thioredoxin-80 (Trx80), a truncated form of Thioredoxin-1, prevents the toxic effects of Aß and inhibits its aggregation in vitro. Trx80 levels were found to be dramatically reduced both in the human brain and cerebrospinal fluid of AD patients. In this study, we investigated the effect of Trx80 expression using in vivo and in vitro models of Aß pathology. We developed Drosophila melanogaster models overexpressing either human Trx80, human Aß42, or both Aß42/Trx80 in the central nervous system. We found that Trx80 expression prevents Aß42 accumulation in the brain and rescues the reduction in life span and locomotor impairments seen in Aß42 expressing flies. Also, we show that Trx80 induces autophagosome formation and reverses the inhibition of Atg4b-Atg8a/b autophagosome formation pathway caused by Aß42. These effects were also confirmed in human neuroblastoma cells. These results give insight into Trx80 function in vivo, suggesting its role in the autophagosome biogenesis and thus in Aß42 degradation. Our findings put Trx80 on the spotlight as an endogenous agent against Aß42-induced toxicity in the brain suggesting that strategies to enhance Trx80 levels in neurons could potentially be beneficial against AD pathology in humans.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Alzheimer Disease/genetics , Animals , Drosophila melanogaster , Humans , Lysosomes , Peptide Fragments , Thioredoxins/genetics
3.
PLoS Genet ; 15(7): e1008240, 2019 07.
Article in English | MEDLINE | ID: mdl-31365523

ABSTRACT

The RNA helicase SUV3 and the polynucleotide phosphorylase PNPase are involved in the degradation of mitochondrial mRNAs but their roles in vivo are not fully understood. Additionally, upstream processes, such as transcript maturation, have been linked to some of these factors, suggesting either dual roles or tightly interconnected mechanisms of mitochondrial RNA metabolism. To get a better understanding of the turn-over of mitochondrial RNAs in vivo, we manipulated the mitochondrial mRNA degrading complex in Drosophila melanogaster models and studied the molecular consequences. Additionally, we investigated if and how these factors interact with the mitochondrial poly(A) polymerase, MTPAP, as well as with the mitochondrial mRNA stabilising factor, LRPPRC. Our results demonstrate a tight interdependency of mitochondrial mRNA stability, polyadenylation and the removal of antisense RNA. Furthermore, disruption of degradation, as well as polyadenylation, leads to the accumulation of double-stranded RNAs, and their escape out into the cytoplasm is associated with an altered immune-response in flies. Together our results suggest a highly organised and inter-dependable regulation of mitochondrial RNA metabolism with far reaching consequences on cellular physiology.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , RNA, Mitochondrial/chemistry , RNA, Mitochondrial/metabolism , Animals , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Female , Male , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Polyadenylation , Polyribonucleotide Nucleotidyltransferase/genetics , Polyribonucleotide Nucleotidyltransferase/metabolism , RNA Stability , RNA, Antisense/chemistry , RNA, Antisense/metabolism , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/metabolism
4.
Hum Mutat ; 42(4): 378-384, 2021 04.
Article in English | MEDLINE | ID: mdl-33502047

ABSTRACT

Mutations in structural subunits and assembly factors of complex I of the oxidative phosphorylation system constitute the most common cause of mitochondrial respiratory chain defects. Such mutations can present a wide range of clinical manifestations, varying from mild deficiencies to severe, lethal disorders. We describe a patient presenting intrauterine growth restriction and anemia, which displayed postpartum hypertrophic cardiomyopathy, lactic acidosis, encephalopathy, and a severe complex I defect with fatal outcome. Whole genome sequencing revealed an intronic biallelic mutation in the NDUFB7 gene (c.113-10C>G) and splicing pattern alterations in NDUFB7 messenger RNA were confirmed by RNA Sequencing. The detected variant resulted in a significant reduction of the NDUFB7 protein and reduced complex I activity. Complementation studies with expression of wild-type NDUFB7 in patient fibroblasts normalized complex I function. Here we report a case with a primary complex I defect due to a homozygous mutation in an intron region of the NDUFB7 gene.


Subject(s)
Acidosis, Lactic , Cardiomyopathy, Hypertrophic , Mitochondrial Diseases , NADH, NADPH Oxidoreductases/genetics , Acidosis, Lactic/genetics , Cardiomyopathy, Hypertrophic/genetics , Electron Transport Complex I/genetics , Humans , Mitochondrial Diseases/genetics , Mutation
5.
Cell Mol Life Sci ; 77(13): 2483-2496, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31912194

ABSTRACT

Understanding the mechanisms behind neurodifferentiation in adults will be an important milestone in our quest to identify treatment strategies for cognitive disorders observed during our natural ageing or disease. It is now clear that the maturation of neural stem cells to neurones, fully integrated into neuronal circuits requires a complete remodelling of cellular metabolism, including switching the cellular energy source. Mitochondria are central for this transition and are increasingly seen as the regulatory hub in defining neural stem cell fate and neurodevelopment. This review explores our current knowledge of metabolism during adult neurodifferentiation.


Subject(s)
Brain/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Adult , Animals , Humans , Lipid Metabolism , Mitochondria/ultrastructure , Oxidation-Reduction , Reactive Oxygen Species/metabolism
6.
Am J Hum Genet ; 99(3): 735-743, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27545679

ABSTRACT

SQSTM1 (sequestosome 1; also known as p62) encodes a multidomain scaffolding protein involved in various key cellular processes, including the removal of damaged mitochondria by its function as a selective autophagy receptor. Heterozygous variants in SQSTM1 have been associated with Paget disease of the bone and might contribute to neurodegeneration in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Using exome sequencing, we identified three different biallelic loss-of-function variants in SQSTM1 in nine affected individuals from four families with a childhood- or adolescence-onset neurodegenerative disorder characterized by gait abnormalities, ataxia, dysarthria, dystonia, vertical gaze palsy, and cognitive decline. We confirmed absence of the SQSTM1/p62 protein in affected individuals' fibroblasts and found evidence of a defect in the early response to mitochondrial depolarization and autophagosome formation. Our findings expand the SQSTM1-associated phenotypic spectrum and lend further support to the concept of disturbed selective autophagy pathways in neurodegenerative diseases.


Subject(s)
Ataxia/genetics , Autophagy/genetics , Dystonia/genetics , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/physiopathology , Sequestosome-1 Protein/deficiency , Supranuclear Palsy, Progressive/genetics , Adolescent , Adult , Age of Onset , Ataxia/complications , Autophagosomes/metabolism , Autophagosomes/pathology , Child , Cognition Disorders/genetics , Dysarthria/complications , Dysarthria/genetics , Dystonia/complications , Female , Fibroblasts/metabolism , Gait/genetics , Humans , Male , Mitochondria/metabolism , Mitochondria/pathology , Movement Disorders/complications , Movement Disorders/genetics , Neurodegenerative Diseases/complications , Pedigree , Phenotype , RNA, Messenger/analysis , Sequestosome-1 Protein/genetics , Supranuclear Palsy, Progressive/complications , Young Adult
7.
PLoS Genet ; 12(5): e1006028, 2016 05.
Article in English | MEDLINE | ID: mdl-27176048

ABSTRACT

Polyadenylation has well characterised roles in RNA turnover and translation in a variety of biological systems. While polyadenylation on mitochondrial transcripts has been suggested to be a two-step process required to complete translational stop codons, its involvement in mitochondrial RNA turnover is less well understood. We studied knockdown and knockout models of the mitochondrial poly(A) polymerase (MTPAP) in Drosophila melanogaster and demonstrate that polyadenylation of mitochondrial mRNAs is exclusively performed by MTPAP. Further, our results show that mitochondrial polyadenylation does not regulate mRNA stability but protects the 3' terminal integrity, and that despite a lack of functioning 3' ends, these trimmed transcripts are translated, suggesting that polyadenylation is not required for mitochondrial translation. Additionally, loss of MTPAP leads to reduced steady-state levels and disturbed maturation of tRNACys, indicating that polyadenylation in mitochondria might be important for the stability and maturation of specific tRNAs.


Subject(s)
Drosophila melanogaster/genetics , Polyadenylation/genetics , Protein Biosynthesis/genetics , RNA, Messenger/genetics , Animals , Codon, Terminator , Gene Knockdown Techniques , Mitochondria/genetics , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/genetics , RNA, Mitochondrial , RNA, Transfer/genetics
8.
Traffic ; 15(11): 1235-46, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25131297

ABSTRACT

Generation and turnover of phosphatidylinositol 3-phosphate (PtdIns3P) signaling is essential for autophagosome formation and other membrane traffic processes. In both Dictyostelium discoideum and mammalian cells, autophagosomes are formed from specialized regions of the endoplasmic reticulum (ER), called omegasomes, which are enriched in the signaling lipid PtdIns3P. Vacuole membrane protein 1 (Vmp1) is a multispanning membrane protein localized at the ER that is required for autophagosome formation. There are conflicting reports in the literature as to whether Vmp1 is strictly required or not for autophagy-related PtdIns3P signaling and its hierarchical relationship with Atg1 and PI3K. We have now addressed these questions in the Dictyostelium model. We show that Dictyostelium cells lacking Vmp1 have elevated and aberrant PtdIns3P signaling on the ER, resulting in an increased and persistent recruitment of Atg18 and other autophagic proteins. This indicates that Vmp1 is not strictly essential for the generation of PtdIns3P signaling but rather suggests a role in the correct turnover or modulation of this signaling. Of interest, these PtdIns3P-enriched regions of the ER surround ubiquitinated protein aggregates but are unable to form functional autophagosomes. vmp1 null cells also have additional defects in macropinocytosis and growth, which are not shared by other autophagy mutants. Remarkably, we show that these defects and also the aberrant PtdIns3P distribution are largely suppressed by the concomitant loss of Atg1, indicating that aberrant autophagic signaling on the ER inhibits macropinocytosis. These results suggest that Atg1 functions upstream of Vmp1 in this signaling pathway and demonstrates a previously unappreciated link between abnormal autophagy signaling and macropinocytosis.


Subject(s)
Autophagy , Dictyostelium/metabolism , Membrane Proteins/metabolism , Phagosomes/metabolism , Phosphatidylinositol Phosphates/metabolism , Protozoan Proteins/metabolism , Endoplasmic Reticulum/metabolism , Membrane Proteins/genetics , Protozoan Proteins/genetics , Signal Transduction
9.
J Cell Sci ; 123(Pt 10): 1674-83, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20406883

ABSTRACT

Dictyostelium and human MidA are homologous proteins that belong to a family of proteins of unknown function called DUF185. Using yeast two-hybrid screening and pull-down experiments, we showed that both proteins interact with the mitochondrial complex I subunit NDUFS2. Consistent with this, Dictyostelium cells lacking MidA showed a specific defect in complex I activity, and knockdown of human MidA in HEK293T cells resulted in reduced levels of assembled complex I. These results indicate a role for MidA in complex I assembly or stability. A structural bioinformatics analysis suggested the presence of a methyltransferase domain; this was further supported by site-directed mutagenesis of specific residues from the putative catalytic site. Interestingly, this complex I deficiency in a Dictyostelium midA(-) mutant causes a complex phenotypic outcome, which includes phototaxis and thermotaxis defects. We found that these aspects of the phenotype are mediated by a chronic activation of AMPK, revealing a possible role of AMPK signaling in complex I cytopathology.


Subject(s)
Methyltransferases/metabolism , Mitochondria/metabolism , Protozoan Proteins/metabolism , AMP-Activated Protein Kinase Kinases , Catalytic Domain/genetics , Cell Movement/genetics , Computational Biology , Dictyostelium , Electron Transport Complex I/metabolism , Humans , Methyltransferases/genetics , Mutagenesis, Site-Directed , Mutation/genetics , NADH Dehydrogenase/metabolism , Protein Binding , Protein Kinases/metabolism , Protozoan Proteins/genetics , RNA, Small Interfering/genetics , Signal Transduction/genetics , Two-Hybrid System Techniques
10.
Nat Commun ; 13(1): 5750, 2022 09 30.
Article in English | MEDLINE | ID: mdl-36180430

ABSTRACT

Canonical RNA processing in mammalian mitochondria is defined by tRNAs acting as recognition sites for nucleases to release flanking transcripts. The relevant factors, their structures, and mechanism are well described, but not all mitochondrial transcripts are punctuated by tRNAs, and their mode of processing has remained unsolved. Using Drosophila and mouse models, we demonstrate that non-canonical processing results in the formation of 3' phosphates, and that phosphatase activity by the carbon catabolite repressor 4 domain-containing family member ANGEL2 is required for their hydrolysis. Furthermore, our data suggest that members of the FAST kinase domain-containing protein family are responsible for these 3' phosphates. Our results therefore propose a mechanism for non-canonical RNA processing in metazoan mitochondria, by identifying the role of ANGEL2.


Subject(s)
RNA Processing, Post-Transcriptional , RNA , Animals , Carbon/metabolism , Drosophila , Exoribonucleases , Mammals/genetics , Mice , Phosphates/metabolism , Phosphoric Monoester Hydrolases/metabolism , RNA/metabolism , RNA, Mitochondrial/genetics , RNA, Mitochondrial/metabolism , RNA, Transfer/metabolism
11.
STAR Protoc ; 2(2): 100528, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34027486

ABSTRACT

Here, we present a revised protocol to derive neuroepithelial stem (NES) cells from human induced pluripotent stem cells. NES cells can be further differentiated into a culture of neurons (90%) and glia (10%). We describe how to derive and maintain NES cells in culture and how to differentiate them. In addition, we show the potential use of NES cells to study the role of reactive oxygen species in neuronal differentiation and a guideline for NES cell transfection. For complete details on the use and execution of this protocol, please refer to Calvo-Garrido et al. (2019); Falk et al. (2012).


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation/physiology , Induced Pluripotent Stem Cells/cytology , Neuroepithelial Cells/cytology , Cells, Cultured , Humans , Neuroglia/cytology , Neurons/cytology
12.
Neurol Genet ; 7(2): e566, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33732874

ABSTRACT

OBJECTIVE: To investigate the pathogenicity of a novel MT-ND3 mutation identified in a patient with adult-onset sensorimotor axonal polyneuropathy and report the clinical, morphologic, and biochemical findings. METHODS: Clinical assessments and morphologic and biochemical investigations of skeletal muscle and cultured myoblasts from the patient were performed. Whole-genome sequencing (WGS) of DNA from skeletal muscle and Sanger sequencing of mitochondrial DNA (mtDNA) from both skeletal muscle and cultured myoblasts were performed. Heteroplasmic levels of mutated mtDNA in different tissues were quantified by last-cycle hot PCR. RESULTS: Muscle showed ragged red fibers, paracrystalline inclusions, a significant reduction in complex I (CI) respiratory chain (RC) activity, and decreased adenosine triphosphate (ATP) production for all substrates used by CI. Sanger sequencing of DNA from skeletal muscle detected a unique previously unreported heteroplasmic mutation in mtDNA encoded MT-ND3, coding for a subunit in CI. WGS confirmed the mtDNA mutation but did not detect any other mutation explaining the disease. Cultured myoblasts, however, did not carry the mutation, and RC activity measurements in myoblasts were normal. CONCLUSIONS: We report a case with adult-onset sensorimotor axonal polyneuropathy caused by a novel mtDNA mutation in MT-ND3. Loss of heteroplasmy in blood, cultured fibroblasts and myoblasts from the patient, and normal measurement of RC activity of the myoblasts support pathogenicity of the mutation. These findings highlight the importance of mitochondrial investigations in patients presenting with seemingly idiopathic polyneuropathy, especially if muscle also is affected.

13.
Stem Cell Reports ; 12(4): 696-711, 2019 04 09.
Article in English | MEDLINE | ID: mdl-30827875

ABSTRACT

Neurodegenerative disorders are an increasingly common and irreversible burden on society, often affecting the aging population, but their etiology and disease mechanisms are poorly understood. Studying monogenic neurodegenerative diseases with known genetic cause provides an opportunity to understand cellular mechanisms also affected in more complex disorders. We recently reported that loss-of-function mutations in the autophagy adaptor protein SQSTM1/p62 lead to a slowly progressive neurodegenerative disease presenting in childhood. To further elucidate the neuronal involvement, we studied the cellular consequences of loss of p62 in a neuroepithelial stem cell (NESC) model and differentiated neurons derived from reprogrammed p62 patient cells or by CRISPR/Cas9-directed gene editing in NESCs. Transcriptomic and proteomic analyses suggest that p62 is essential for neuronal differentiation by controlling the metabolic shift from aerobic glycolysis to oxidative phosphorylation required for neuronal maturation. This shift is blocked by the failure to sufficiently downregulate lactate dehydrogenase expression due to the loss of p62, possibly through impaired Hif-1α downregulation and increased sensitivity to oxidative stress. The findings imply an important role for p62 in neuronal energy metabolism and particularly in the regulation of the shift between glycolysis and oxidative phosphorylation required for normal neurodifferentiation.


Subject(s)
Cell Differentiation/genetics , Cellular Reprogramming/genetics , Energy Metabolism/genetics , Sequestosome-1 Protein/genetics , Gene Expression Profiling , Glycolysis , Humans , Mitophagy , Models, Biological , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurons/cytology , Neurons/metabolism , Oxidative Phosphorylation , Oxidative Stress , Oxygen/metabolism , Sequestosome-1 Protein/metabolism
14.
Mol Genet Genomic Med ; 7(6): e654, 2019 06.
Article in English | MEDLINE | ID: mdl-30920170

ABSTRACT

BACKGROUND: Mutations in mitochondrial aminoacyl tRNA synthetases form a subgroup of mitochondrial disorders often only perturbing brain function by affecting mitochondrial translation. Here we report two siblings with mitochondrial disease, due to compound heterozygous mutations in the mitochondrial tryptophanyl-tRNA synthetase (WARS2) gene, presenting with severe neurological symptoms but normal mitochondrial function in skeletal muscle biopsies and cultured skin fibroblasts. METHODS: Whole exome sequencing on genomic DNA samples from both subjects and their parents identified two compound heterozygous variants c.833T>G (p.Val278Gly) and c.938A>T (p.Lys313Met) in the WARS2 gene as potential disease-causing variants. We generated patient-derived neuroepithelial stem cells and modeled the disease in yeast and Drosophila melanogaster to confirm pathogenicity. RESULTS: Biochemical analysis of patient-derived neuroepithelial stem cells revealed a mild combined complex I and IV defect, while modeling the disease in yeast demonstrated that the reported aminoacylation defect severely affects respiration and viability. Furthermore, silencing of wild type WARS2 in Drosophila melanogaster showed that a partial defect in aminoacylation is enough to cause lethality. CONCLUSIONS: Our results establish the identified WARS2 variants as disease-causing and highlight the benefit of including human neuronal models, when investigating mutations specifically affecting the nervous system.


Subject(s)
Leukoencephalopathies/genetics , Tryptophan-tRNA Ligase/genetics , Adolescent , Adult , Amino Acid Sequence , Amino Acyl-tRNA Synthetases/genetics , Aminoacylation , Animals , Child , Disease Models, Animal , Drosophila melanogaster , Growth Disorders/genetics , Humans , Leukoencephalopathies/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Diseases/genetics , Mutation , Pedigree , Tryptophan-tRNA Ligase/metabolism , Exome Sequencing
15.
BMC Microbiol ; 8: 109, 2008 Jun 30.
Article in English | MEDLINE | ID: mdl-18590548

ABSTRACT

BACKGROUND: Pseudomonas aeruginosa is one of the most relevant human opportunistic bacterial pathogens. Two strains (PAO1 and PA14) have been mainly used as models for studying virulence of P. aeruginosa. The strain PA14 is more virulent than PAO1 in a wide range of hosts including insects, nematodes and plants. Whereas some of the differences might be attributable to concerted action of determinants encoded in pathogenicity islands present in the genome of PA14, a global analysis of the differential host responses to these P. aeruginosa strains has not been addressed. Little is known about the host response to infection with P. aeruginosa and whether or not the global host transcription is being affected as a defense mechanism or altered in the benefit of the pathogen. Since the social amoeba Dictyostelium discoideum is a suitable host to study virulence of P. aeruginosa and other pathogens, we used available genomic tools in this model system to study the transcriptional host response to P. aeruginosa infection. RESULTS: We have compared the virulence of the P. aeruginosa PAO1 and PA14 using D. discoideum and studied the transcriptional response of the amoeba upon infection. Our results showed that PA14 is more virulent in Dictyostelium than PA01using different plating assays. For studying the differential response of the host to infection by these model strains, D. discoideum cells were exposed to either P. aeruginosa PAO1 or P. aeruginosa PA14 (mixed with an excess of the non-pathogenic bacterium Klebsiella aerogenes as food supply) and after 4 hours, cellular RNA extracted. A three-way comparison was made using whole-genome D. discoideum microarrays between RNA samples from cells treated with the two different strains and control cells exposed only to K. aerogenes. The transcriptomic analyses have shown the existence of common and specific responses to infection. The expression of 364 genes changed in a similar way upon infection with one or another strain, whereas 169 genes were differentially regulated depending on whether the infecting strain was either P. aeruginosa PAO1 or PA14. Effects on metabolism, signalling, stress response and cell cycle can be inferred from the genes affected. CONCLUSION: Our results show that pathogenic Pseudomonas strains invoke both a common transcriptional response from Dictyostelium and a strain specific one, indicating that the infective process of bacterial pathogens can be strain-specific and is more complex than previously thought.


Subject(s)
Dictyostelium/microbiology , Genome, Protozoan , Protozoan Proteins/metabolism , Pseudomonas aeruginosa/pathogenicity , Animals , Dictyostelium/growth & development , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation , Humans , Protozoan Proteins/genetics , Pseudomonas aeruginosa/classification , Species Specificity , Virulence
17.
J Alzheimers Dis ; 56(2): 601-617, 2017.
Article in English | MEDLINE | ID: mdl-28035917

ABSTRACT

The major genetic risk factor for Alzheimer's disease (AD), apolipoprotein E4 (ApoE4), has been suggested to have detrimental effects on neurons, including direct toxicity via apoptosis. Thioredoxin-1 (Trx1) is an endogenous antioxidant protein important for redox regulation and participates in the regulation of apoptosis through the inhibition of apoptosis signal-regulating kinase-1 (Ask-1). In this study, we have investigated the effects of ApoE on Trx1 in the brain. Our results showed that the protein levels of Trx1 were reduced in the hippocampus of ApoE4 targeted replacement (TR) mice compared to ApoE3 TR mice. The reduction was also seen in vitro after treatment of both human primary cortical neurons and neuroblastoma cells with human recombinant ApoE4 (rApoE4). Furthermore, ApoE4 caused a disruption of lysosomal integrity and a shift in the localization of Cathepsin D, an enzyme known to degrade Trx1. ApoE4 treatment induced in addition apoptosis through translocation of Death-domain associated protein-6 (Daxx) from the nucleus to the cytosol, suggesting an activation of the Ask-1 pathway. This toxicity was prevented by overexpression of Trx1 and other endogenous Ask-1 inhibitors. Our data suggests that down-regulation of Trx1 is involved in the toxicity caused by ApoE4. An activated ASK-1 pathway might indeed make cells more vulnerable to other insults such as amyloid-ß, which could partially explain the mechanism behind the strongest genetic risk factor for AD.


Subject(s)
Apolipoprotein E4/metabolism , Apoptosis/physiology , Cathepsin D/metabolism , Lysosomes/metabolism , Thioredoxins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apolipoprotein E4/genetics , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Cell Line, Tumor , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Co-Repressor Proteins , Female , Hippocampus/metabolism , Hippocampus/pathology , Humans , MAP Kinase Kinase Kinase 5/antagonists & inhibitors , MAP Kinase Kinase Kinase 5/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Molecular Chaperones , Neurons/metabolism , Neurons/pathology , Nuclear Proteins/metabolism , Oxidative Stress/physiology , Recombinant Proteins/metabolism
18.
J Alzheimers Dis ; 53(1): 349-61, 2016 05 07.
Article in English | MEDLINE | ID: mdl-27163814

ABSTRACT

Serotonin (5-HT) plays a central role in the integrity of different brain functions. The 5-HT homeostasis is regulated by many factors, including serotonin transporter (SERT), monoamine oxidase enzyme (MAO), and several 5-HT receptors, including the 5-HT1B. There is little knowledge how the dynamics of this system is affected by the amyloid-ß (Aß) burden of Alzheimer's disease (AD) pathology. SH-SY5Y neuroblastoma cells transfected with the amyloid precursor protein (APP) gene containing the Swedish mutations causing familial AD (APPswe), were used as a model to explore the effect of Aß pathology on 5-HT1B and related molecules including the receptor adaptor protein (p11), SERT and MAOA gene expression, and MAOA activity after treatment with selective serotonin reuptake inhibitor (SSRI) (sertraline), and a 5-HT1B receptor antagonist. Sertraline led more than 70 fold increase of 5-HT1B gene expression (p < 0.001), an increased serotonin turnover in both APPswe and control cells and reduced intracellular serotonin levels by 75% in APPswe cells but not in controls (p > 0.05). Treatment with the 5-HT1B receptor antagonist increased SERT gene-expression in control cells but not in the APPswe cells. 5-HT and 5-HT1B antagonist treatment resulted in different p11 expression patterns in APPswe cells compared to controls. Although MAOA gene expression was not changed by APPswe overexpression, adding 5-HT lead to a significant increase in MAOA gene expression in APPswe but not control cells. These findings suggest that the sensitivity of the 5-HT1B receptor and related systems is affected by APPswe overexpression, with potential relevance for pharmacologic intervention in AD. This may at least partly explain the lack of effect of SSRIs in patients with AD and depression.


Subject(s)
Gene Expression Regulation/drug effects , Receptor, Serotonin, 5-HT1B/metabolism , Serotonin Agents/pharmacology , Serotonin/metabolism , Amyloid beta-Protein Precursor/genetics , Cell Line, Tumor , Chromatography, Liquid , Electrochemical Techniques , Gene Expression Regulation/genetics , Humans , Hydroxyindoleacetic Acid/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Monoamine Oxidase/metabolism , Mutation/genetics , Neuroblastoma/pathology , Phosphorylation/drug effects , Piperidones/pharmacology , RNA, Messenger/metabolism , Receptor, Serotonin, 5-HT1B/genetics , Serotonin/pharmacology , Serotonin Plasma Membrane Transport Proteins/metabolism , Spiro Compounds/pharmacology , Statistics, Nonparametric , Transfection
19.
Neurosci Lett ; 594: 137-43, 2015 May 06.
Article in English | MEDLINE | ID: mdl-25841787

ABSTRACT

Serotonergic dysfunction is implicated in Alzheimer's disease (AD). In addition, reductions in brain of both monoamine synthesis and release have been reported. Serotonin 1B receptors (5-HT1B), along with serotonin transporter (SERT) are among the regulators of extracellular 5-HT levels. We investigated the effect of the familial AD APP (Amyloid precursor protein) K670N/M671L double mutation, APP Swedish mutation (APPswe), on the expression of 5-HT1B, SERT, MAOA, p11 and 5-HT and its metabolite 5-HIAA in SH-SY5Y human neuroblastoma cell line stably transfected with APPswe mutation. In addition, hippocampal expressions of 5-HT1B and SERT were assessed in wild type and transgenic mice expressing APPswe mutation (Tg2576) at different age groups. We found a reduction of 5-HT1B as well as SERT in both APPswe in vitro and ex vivo. P11 and 5HT were also reduced, whereas 5HT turnover and MAOA were increased. Our results indicate that APPswe induced decreased 5-HT1B expression and 5-HT release, as well as increased MAOA activity and 5-HT breakdown. Further studies to explore the detailed mechanism behind reduced 5-HT1B and SERT in AD and their clinical implications are needed.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Receptor, Serotonin, 5-HT1B/metabolism , Alzheimer Disease/metabolism , Animals , Annexin A2/metabolism , Cell Line, Tumor , Female , Hippocampus/metabolism , Humans , Hydroxyindoleacetic Acid/metabolism , Mice, Transgenic , Monoamine Oxidase/metabolism , Mutation , Receptor, Serotonin, 5-HT1B/genetics , S100 Proteins/metabolism , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism
20.
Methods Mol Biol ; 983: 461-70, 2013.
Article in English | MEDLINE | ID: mdl-23494324

ABSTRACT

Autophagy is an intracellular degradation mechanism essential for cell survival and maintenance of cellular homeostasis, differentiation, and development. Recent research has highlighted the impact of autophagy in neurodegenerative diseases and aging. We are still far from fully understanding the molecular mechanism of autophagy and its regulation, essential for its future use as a therapeutic target. In the last years many different techniques have been developed to study this process and some of them have been successfully used in Dictyostelium. We describe here the use of confocal microscopy to detect the pattern of different autophagic markers and the differences expected in strains deficient in autophagy. Autophagy dysfunction might also lead to the formation of intracellular ubiquitinated protein aggregates that can be easily detected by immunofluorescence. In addition, we describe two different techniques that allow the assessment of the so-called autophagic flux, the progression of autophagosomes until their fusion with lysosomes. The first one is a proteolytic cleavage assay of cytosolic markers such as GFP-PgkA and the second the visualization of the RFP-GFP-Atg8 marker by confocal microscopy.


Subject(s)
Autophagy , Dictyostelium/cytology , Single-Cell Analysis/methods , Biomarkers/metabolism , Buffers , Dictyostelium/physiology , Green Fluorescent Proteins/metabolism , Microbial Viability , Microfilament Proteins/metabolism , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Proteolysis , Protozoan Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Ubiquitinated Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL