144 research outputs found

    TDP-43 knockdown impairs neurite outgrowth dependent on its target histone deacetylase 6

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>Trans-activation response element (TAR) DNA binding protein of 43kDa (TDP-43) is causally related to the neurodegenerative diseases frontotemporal dementia and amyotrophic lateral sclerosis being the hallmark protein in the disease-characteristic neuropathological lesions and via genetic linkage. Histone deacetylase 6 (HDAC6) is an established target of the RNA-binding protein TDP-43. HDAC6 is an unusual cytosolic deacetylase enzyme, central for a variety of pivotal cellular functions including aggregating protein turnover, microtubular dynamics and filopodia formation. All these functions are important in the context of neurodegenerative proteinopathies involving TDP-43. We have previously shown in a human embryonic kidney cell line that TDP-43 knockdown significantly impairs the removal of a toxic, aggregating polyQ ataxin-3 fusion protein in an HDAC6-dependent manner. Here we investigated the influence of TDP-43 and its target HDAC6 on neurite outgrowth.</p> <p>Results</p> <p>Human neuroblastoma SH-SY5Y cells with stably silenced TDP-43 showed a significant reduction of neurite outgrowth induced by retinoic acid and brain-derived neurotrophic factor. Re-transfection with TDP-43 as well as HDAC6 rescued retinoic acid-induced neurite outgrowth. In addition, we show that silencing of HDAC6 alone is sufficient to reduce neurite outgrowth of <it>in vitro </it>differentiated SH-SY5Y cells.</p> <p>Conclusions</p> <p>TDP-43 deficiency leads to impairment of neurite growth in an HDAC6-dependent manner, thereby contributing to neurodegenerative events in TDP-43 diseases.</p

    TDP-43 regulates global translational yield by splicing of exon junction complex component SKAR

    Get PDF
    TDP-43 is linked to neurodegenerative diseases including frontotemporal dementia and amyotrophic lateral sclerosis. Mostly localized in the nucleus, TDP-43 acts in conjunction with other ribonucleoproteins as a splicing co-factor. Several RNA targets of TDP-43 have been identified so far, but its role(s) in pathogenesis remains unclear. Using Affymetrix exon arrays, we have screened for the first time for splicing events upon TDP-43 knockdown. We found alternative splicing of the ribosomal S6 kinase 1 (S6K1) Aly/REF-like target (SKAR) upon TDP-43 knockdown in non-neuronal and neuronal cell lines. Alternative SKAR splicing depended on the first RNA recognition motif (RRM1) of TDP-43 and on 5ā€²-GA-3ā€™ and 5ā€²-UG-3ā€² repeats within the SKAR pre-mRNA. SKAR is a component of the exon junction complex, which recruits S6K1, thereby facilitating the pioneer round of translation and promoting cell growth. Indeed, we found that expression of the alternatively spliced SKAR enhanced S6K1-dependent signaling pathways and the translational yield of a splice-dependent reporter. Consistent with this, TDP-43 knockdown also increased translational yield and significantly increased cell size. This indicates a novel mechanism of deregulated translational control upon TDP-43 deficiency, which might contribute to pathogenesis of the protein aggregation diseases frontotemporal dementia and amyotrophic lateral sclerosis

    Chronicā€“Progressive Dopaminergic Deficiency Does Not Induce Midbrain Neurogenesis

    Get PDF
    Background: Consecutive adult neurogenesis is a well-known phenomenon in the ventricularā€“subventricular zone of the lateral wall of the lateral ventricles (Vā€“SVZ) and has been controversially discussed in so-called ā€œnon-neurogenicā€ brain areas such as the periventricular regions (PVRs) of the aqueduct and the fourth ventricle. Dopamine is a known modulator of adult neural stem cell (aNSC) proliferation and dopaminergic neurogenesis in the olfactory bulb, though a possible interplay between local dopaminergic neurodegeneration and induction of aNSC proliferation in mid/hindbrain PVRs is currently enigmatic. Objective/Hypothesis: To analyze the influence of chronicā€“progressive dopaminergic neurodegeneration on both consecutive adult neurogenesis in the PVRs of the Vā€“SVZ and mid/hindbrain aNSCs in two mechanistically different transgenic animal models of ParkinsonĀ“s disease (PD). Methods: We used Thy1-m[A30P]h Ī± synuclein mice and Leu9ā€²Ser hypersensitive Ī±4* nAChR mice to assess the influence of midbrain dopaminergic neuronal loss on neurogenic activity in the PVRs of the Vā€“SVZ, the aqueduct and the fourth ventricle. Results: In both animal models, overall proliferative activity in the Vā€“SVZ was not altered, though the proportion of B2/activated B1 cells on all proliferating cells was reduced in the Vā€“SVZ in Leu9ā€²Ser hypersensitive Ī±4* nAChR mice. Putative aNSCs in the mid/hindbrain PVRs are known to be quiescent in vivo in healthy controls, and dopaminergic deficiency did not induce proliferative activity in these regions in both disease models. Conclusions: Our data do not support an activation of endogenous aNSCs in mid/hindbrain PVRs after local dopaminergic neurodegeneration. Spontaneous endogenous regeneration of dopaminergic cell loss through resident aNSCs is therefore unlikely

    FAS-dependent cell death in Ī±-synuclein transgenic oligodendrocyte models of multiple system atrophy

    Get PDF
    Multiple system atrophy is a parkinsonian neurodegenerative disorder. It is cytopathologically characterized by accumulation of the protein p25Ī± in cell bodies of oligodendrocytes followed by accumulation of aggregated Ī±-synuclein in so-called glial cytoplasmic inclusions. p25Ī± is a stimulator of Ī±-synuclein aggregation, and coexpression of Ī±-synuclein and p25Ī± in the oligodendroglial OLN-t40-AS cell line causes Ī±-synuclein aggregate-dependent toxicity. In this study, we investigated whether the FAS system is involved in Ī±-synuclein aggregate dependent degeneration in oligodendrocytes and may play a role in multiple system atrophy. Using rat oligodendroglial OLN-t40-AS cells we demonstrate that the cytotoxicity caused by coexpressing Ī±-synuclein and p25Ī± relies on stimulation of the death domain receptor FAS and caspase-8 activation. Using primary oligodendrocytes derived from PLP-Ī±-synuclein transgenic mice we demonstrate that they exist in a sensitized state expressing pro-apoptotic FAS receptor, which makes them sensitive to FAS ligand-mediated apoptosis. Immunoblot analysis shows an increase in FAS in brain extracts from multiple system atrophy cases. Immunohistochemical analysis demonstrated enhanced FAS expression in multiple system atrophy brains notably in oligodendrocytes harboring the earliest stages of glial cytoplasmic inclusion formation. Oligodendroglial FAS expression is an early hallmark of oligodendroglial pathology in multiple system atrophy that mechanistically may be coupled to Ī±-synuclein dependent degeneration and thus represent a potential target for protective intervention

    Reduced Basal Autophagy and Impaired Mitochondrial Dynamics Due to Loss of Parkinson's Disease-Associated Protein DJ-1

    Get PDF
    BACKGROUND: Mitochondrial dysfunction and degradation takes a central role in current paradigms of neurodegeneration in Parkinson's disease (PD). Loss of DJ-1 function is a rare cause of familial PD. Although a critical role of DJ-1 in oxidative stress response and mitochondrial function has been recognized, the effects on mitochondrial dynamics and downstream consequences remain to be determined. METHODOLOGY/PRINCIPAL FINDINGS: Using DJ-1 loss of function cellular models from knockout (KO) mice and human carriers of the E64D mutation in the DJ-1 gene we define a novel role of DJ-1 in the integrity of both cellular organelles, mitochondria and lysosomes. We show that loss of DJ-1 caused impaired mitochondrial respiration, increased intramitochondrial reactive oxygen species, reduced mitochondrial membrane potential and characteristic alterations of mitochondrial shape as shown by quantitative morphology. Importantly, ultrastructural imaging and subsequent detailed lysosomal activity analyses revealed reduced basal autophagic degradation and the accumulation of defective mitochondria in DJ-1 KO cells, that was linked with decreased levels of phospho-activated ERK2. CONCLUSIONS/SIGNIFICANCE: We show that loss of DJ-1 leads to impaired autophagy and accumulation of dysfunctional mitochondria that under physiological conditions would be compensated via lysosomal clearance. Our study provides evidence for a critical role of DJ-1 in mitochondrial homeostasis by connecting basal autophagy and mitochondrial integrity in Parkinson's disease

    Neuropathology in Mice Expressing Mouse Alpha-Synuclein

    Get PDF
    Ī±-Synuclein (Ī±SN) in human is tightly linked both neuropathologically and genetically to Parkinson's disease (PD) and related disorders. Disease-causing properties in vivo of the wildtype mouse ortholog (mĪ±SN), which carries a threonine at position 53 like the A53T human mutant version that is genetically linked to PD, were never reported. To this end we generated mouse lines that express mĪ±SN in central neurons at levels reaching up to six-fold compared to endogenous mĪ±SN. Unlike transgenic mice expressing human wildtype or mutant forms of Ī±SN, these mĪ±SN transgenic mice showed pronounced ubiquitin immunopathology in spinal cord and brainstem. Isoelectric separation of mĪ±SN species revealed multiple isoforms including two Ser129-phosphorylated species in the most severely affected brain regions. Neuronal Ser129-phosphorylated Ī±SN occured in granular and small fibrillar aggregates and pathological staining patterns in neurites occasionally revealed a striking ladder of small alternating segments staining either for Ser129-phosphorylated Ī±SN or ubiquitin but not both. Axonal degeneration in long white matter tracts of the spinal cord, with breakdown of myelin sheaths and degeneration of neuromuscular junctions with loss of integrity of the presynaptic neurofilament network in mĪ±SN transgenic mice, was similar to what we have reported for mice expressing human Ī±SN wildtype or mutant forms. In hippocampal neurons, the mĪ±SN protein accumulated and was phosphorylated but these neurons showed no ubiquitin immunopathology. In contrast to the early-onset motor abnormalities and muscle weakness observed in mice expressing human Ī±SN, mĪ±SN transgenic mice displayed only end-stage phenotypic alterations that manifested alongside with neuropathology. Altogether these findings show that increased levels of wildtype mĪ±SN does not induce early-onset behavior changes, but drives end-stage pathophysiological changes in murine neurons that are strikingly similar to those evoked by expression of human wildtype or mutant forms

    Prominent microglial inclusions in transgenic mouse models of Ī±-synucleinopathy that are distinct from neuronal lesions.

    Get PDF
    Alpha-synucleinopathies are a group of progressive neurodegenerative disorders, characterized by intracellular deposits of aggregated Ī±-synuclein (Ī±S). The clinical heterogeneity of these diseases is thought to be attributed to conformers (or strains) of Ī±S but the contribution of inclusions in various cell types is unclear. The aim of the present work was to study Ī±S conformers among different transgenic (TG) mouse models of Ī±-synucleinopathies. To this end, four different TG mouse models were studied (Prnp-h[A53T]Ī±S; Thy1-h[A53T]Ī±S; Thy1-h[A30P]Ī±S; Thy1-mĪ±S) that overexpress human or murine Ī±S and differed in their age-of-symptom onset and subsequent disease progression. Postmortem analysis of end-stage brains revealed robust neuronal Ī±S pathology as evidenced by accumulation of Ī±S serine 129 (p-Ī±S) phosphorylation in the brainstem of all four TG mouse lines. Overall appearanceĀ of the pathology was similar and only modest differences were observed among additionally affected brain regions. To study Ī±S conformers in these mice, we used pentameric formyl thiophene acetic acid (pFTAA), a fluorescent dye with amyloid conformation-dependent spectral properties. Unexpectedly, besides the neuronal Ī±S pathology, we also found abundant pFTAA-positive inclusions in microglia of all four TG mouse lines. These microglial inclusions were also positive for Thioflavin S and showed immunoreactivity with antibodies recognizing the N-terminus of Ī±S, but were largely p-Ī±S-negative. In all four lines, spectral pFTAA analysis revealed conformational differences between microglia and neuronal inclusions but not among the different mouse models. Concomitant with neuronal lesions, microglial inclusions were already present at presymptomatic stages and could also be induced by seeded Ī±S aggregation. Although nature and significance of microglial inclusions for human Ī±-synucleinopathies remain to be clarified, the previously overlooked abundance of microglial inclusions in TG mouse models of Ī±-synucleinopathy bears importance for mechanistic and preclinical-translational studies

    Microglial inclusions and neurofilament light chain release follow neuronal Ī±-synuclein lesions in long-term brain slice cultures.

    Get PDF
    BACKGROUND: Proteopathic brain lesions are a hallmark of many age-related neurodegenerative diseases including synucleinopathies and develop at least a decade before the onset of clinical symptoms. Thus, understanding of the initiation and propagation of such lesions is key for developing therapeutics to delay or halt disease progression. METHODS: Alpha-synuclein (Ī±S) inclusions were induced in long-term murine and human slice cultures by seeded aggregation. An Ī±S seed-recognizing human antibody was tested for blocking seeding and/or spreading of the Ī±S lesions. Release of neurofilament light chain (NfL) into the culture medium was assessed. RESULTS: To study initial stages of Ī±-synucleinopathies, we induced Ī±S inclusions inĀ murine hippocampal slice cultures by seeded aggregation. Induction of Ī±S inclusions in neurons was apparent as early as 1week post-seeding, followed by the occurrence of microglial inclusions in vicinity of the neuronal lesionsĀ at 2-3ā€‰weeks. The amount of Ī±S inclusions was dependent on the type of Ī±S seed and on the culture's genetic background (wildtype vs A53T-Ī±S genotype). Formation of Ī±S inclusions could be monitored by neurofilament light chain protein release into the culture medium, a fluid biomarker of neurodegeneration commonly used in clinical settings. Local microinjection of Ī±S seeds resulted in spreading of Ī±S inclusions to neuronally connected hippocampal subregions, and seeding and spreading could be inhibited by an Ī±S seed-recognizing human antibody. We then applied parameters of the murine cultures to surgical resection-derived adult human long-term neocortical slice cultures from 22 to 61-year-old donors. Similarly, in these human slice cultures, proof-of-principle induction of Ī±S lesions was achievedĀ atĀ 1week post-seeding in combination with viral A53T-Ī±S expressions. CONCLUSION: The successful translation of these brain cultures from mouse to human with the first reported induction of human Ī±S lesions in a true adult human brain environment underlines the potential of this model to study proteopathic lesions in intact mouse and now even aged human brain environments

    TOM40 Mediates Mitochondrial Dysfunction Induced by Ī±-Synuclein Accumulation in Parkinson's Disease.

    Get PDF
    Alpha-synuclein (Ī±-Syn) accumulation/aggregation and mitochondrial dysfunction play prominent roles in the pathology of Parkinson's disease. We have previously shown that postmortem human dopaminergic neurons from PD brains accumulate high levels of mitochondrial DNA (mtDNA) deletions. We now addressed the question, whether alterations in a component of the mitochondrial import machinery -TOM40- might contribute to the mitochondrial dysfunction and damage in PD. For this purpose, we studied levels of TOM40, mtDNA deletions, oxidative damage, energy production, and complexes of the respiratory chain in brain homogenates as well as in single neurons, using laser-capture-microdissection in transgenic mice overexpressing human wildtype Ī±-Syn. Additionally, we used lentivirus-mediated stereotactic delivery of a component of this import machinery into mouse brain as a novel therapeutic strategy. We report here that TOM40 is significantly reduced in the brain of PD patients and in Ī±-Syn transgenic mice. TOM40 deficits were associated with increased mtDNA deletions and oxidative DNA damage, and with decreased energy production and altered levels of complex I proteins in Ī±-Syn transgenic mice. Lentiviral-mediated overexpression of Tom40 in Ī±-Syn-transgenic mice brains ameliorated energy deficits as well as oxidative burden. Our results suggest that alterations in the mitochondrial protein transport machinery might contribute to mitochondrial impairment in Ī±-Synucleinopathies

    The Role of Alpha-Synuclein Oligomerization and Aggregation in Cellular and Animal Models of Parkinsonā€™s Disease

    Get PDF
    Ī±-synuclein (Ī±-syn) is a synaptic protein in which four mutations (A53T, A30P, E46K and gene triplication) have been found to cause an autosomal dominant form of Parkinsonā€™s disease (PD). It is also the major component of intraneuronal protein aggregates, designated as Lewy bodies (LBs), a prominent pathological hallmark of PD. How Ī±-syn contributes to LB formation and PD is still not well-understood. It has been proposed that aggregation of Ī±-syn contributes to the formation of LBs, which then leads to neurodegeneration in PD. However, studies have also suggested that aggregates formation is a protective mechanism against more toxic Ī±-syn oligomers. In this study, we have generated Ī±-syn mutants that have increased propensity to form aggregates by attaching a CL1 peptide to the C-terminal of Ī±-syn. Data from our cellular study suggest an inverse correlation between cell viability and the amount of Ī±-syn aggregates formed in the cells. In addition, our animal model of PD indicates that attachment of CL1 to Ī±-syn enhanced its toxicity to dopaminergic neurons in an age-dependent manner and induced the formation of Lewy body-like Ī±-syn aggregates in the substantia nigra. These results provide new insights into how Ī±-syn-induced toxicity is related to its aggregation
    • ā€¦
    corecore