Spelling suggestions: "subject:"axonaler cotransport"" "subject:"axonaler detransport""
1 |
Axonal hnRNP R: regulation by Ptbp2 and functions in neurodegenerative disorders / Axonales hnRNP R: Regulation durch Ptbp2 und Funktionen bei neurodegenerativen ErkrankungenSalehi, Saeede January 2024 (has links) (PDF)
Axon growth, a fundamental process of neuron development, is regulated by both intrinsic and external guidance signals. Impairment of axon growth and maintenance is implicated in the pathogenesis of neurodegenerative disorders such as Amyotrophic Lateral Sclerosis and Alzheimer’s disease (AD). Axon growth is driven by several post-transcriptional RNA processing mechanisms, including alternative splicing, polyadenylation, subcellular localization, and translation. These mechanisms are controlled by RNA-binding proteins (RBPs) through interacting with their target RNAs in a sequence-dependent manner. In this study, we investigate the cytosolic functions of two neuronal RBPs, Ptbp2 and hnRNP R, which are essential for axon growth in motoneurons.
Polypyrimidine tract binding protein 2 (Ptbp2) contributes to neuronal differentiation and axonogenesis by modulating different splicing programs to adjust the level of proteins involved in these processes. While the nuclear functions of Ptbp2 in alternative splicing have been studied in more detail, the cytosolic roles of Ptbp2 associated with axon growth have remained elusive. In the first part of the study, we show that Ptbp2 is present in cytosolic fractions of motoneurons including axons and axon terminals. Depletion of Ptbp2 impairs axon growth and growth cone maturation in cultured embryonic mouse motoneurons. Moreover, Ptbp2 knockdown affects the level of piccolo protein in the growth cone of cultured motoneurons. We detect Ptbp2 as a top interactor of the 3' UTR of the Hnrnpr transcript encoding the RBP hnRNP R. This interaction results in axonal localization of and thereby local translation of Hnrnpr mRNA in motoneurons. Consequently, axonal synthesis of hnRNP R was diminished upon depletion of Ptbp2 in motoneurons. We present evidence that Ptbp2 through cooperation with translation factor eIF5A2 controls hnRNP R synthesis. Additionally, we observe that re-expression of hnRNP R in Ptbp2-deficient motoneurons rescued axon growth defect while Ptbp2 overexpression failed to normalize the axon elongation defect observed in hnRNP R-deficient motoneurons. Our findings pinpoint axonal synthesized hnRNP R as a mediator of Ptbp2 functions in axon growth.
In the second part of this study, we identify hnRNP R binds to the 3' UTR of microtubule-associated tau (Mapt) transcript encoding tau protein and regulates the axonal translocation and translation of Mapt mRNA. Tau protein has a central role in neuronal microtubule assembly and stability. However, in AD, the accumulation of abnormally hyperphosphorylated tau protein leads to axon outgrowth defects. Loss of hnRNP R reduces axonal tau protein but not the total level of tau. We observe that the brains of 5xFAD mice, as a mouse model of AD, deficient for hnRNP R contain lower phospho-tau and amyloid-β plaques. Likewise, Neurons treated with blocking antisense oligonucleotides (ASO) to prevent binding of hnRNP R to Mapt mRNA show reduced axonal Mapt mRNA and consequently newly synthesized tau protein levels. We show that blocking Mapt mRNA transport to axons impairs axon elongation. Our data thus suggest that reducing tau levels selectively in axons, a major subcellular site of tangle formation, might represent a novel therapeutic approach for the treatment of AD. / Axonwachstum ist ein grundlegender Prozess der Neuronenentwicklung und wird sowohl durch intrinsische als auch externe Leitsignale reguliert. Eine Beeinträchtigung des Axonwachstums und der Aufrechterhaltung von Axonen ist mit der Pathogenese neurodegenerativer Erkrankungen wie der Amyotrophen Lateralsklerose und der AlzheimerKrankheit (AD) verbunden. Mehrere posttranskriptionelle RNA-Verarbeitungsmechanismen, darunter alternatives Spleißen, Polyadenylierung, subzelluläre Lokalisierung und Translation, steuern das Axonwachstum. RNA-bindende Proteine (RBPs) steuern diese Mechanismen, indem sie sequenzabhängig mit ihren Ziel-RNAs interagieren. In dieser Studie untersuchen wir die zytosolischen Funktionen von zwei neuronalen RBPs, Ptbp2 und hnRNP R, die für das Axonwachstum in Motoneuronen essentiell sind.
Das Polypyrimidin-Trakt-Bindungsprotein 2 (Ptbp2) trägt zur neuronalen Differenzierung und Axonogenese bei, indem es verschiedene Spleißprogramme moduliert, um die Menge der an diesen Prozessen beteiligten Proteine anzupassen. Während die nukleären Funktionen von
Ptbp2 beim alternativen Spleißen detaillierter untersucht wurden, sind die zytosolischen Rollen von Ptbp2 im Zusammenhang mit dem Axonwachstum noch unklar. Im ersten Teil der Studie zeigen wir, dass Ptbp2 in zytosolischen Fraktionen von Motoneuronen einschließlich Axonen und Axonterminals vorhanden ist. Die Reduktion von Ptbp2 beeinträchtigt das Axonwachstum und die Reifung der Wachstumskegel in kultivierten embryonalen Motoneuronen von Mäusen. Darüber hinaus beeinflusst der Ptbp2-Knockdown den Gehalt an Piccolo-Protein im
Wachstumskegel kultivierter Motoneuronen. Wir identifizierten Ptbp2 als Top-Interaktor der 3'-UTR des Hnrnpr-Transkripts, welches das RBP hnRNP R kodiert. Diese Interaktion führt zur axonalen Lokalisierung und damit zur lokalen Translation der Hnrnpr-mRNA in Motoneuronen.
Folglich wurde die axonale Synthese von hnRNP R durch Depletion von Ptbp2 in Motoneuronen verringert. Wir legen Beweise dafür vor, dass Ptbp2 durch die Zusammenarbeit mit dem Translationsfaktor eIF5A2 die hnRNP R-Synthese steuert. Darüber hinaus beobachten wir, dass die erneute Expression von hnRNP R in Motoneuronen mit Ptbp2-Mangel den Axonwachstumsdefekt rettete, während die Überexpression von Ptbp2 den auch bei Motoneuronen mit hnRNP R-Mangel beobachteten Axonwachstumsdefekt nicht normalisieren konnte. Unsere Ergebnisse zeigen, dass axonal synthetisiertes hnRNP R ein Vermittler der Ptbp2-Funktionen beim Axonwachstum ist.
Im zweiten Teil dieser Studie beobachteten wir, dass hnRNP R an die 3'-UTR des Mikrotubuliassoziierten Tau-Transkripts (Mapt) bindet, welches das Protein tau kodiert. Die hnRNP RMapt Interaktion bewirkt die axonale Translokation und Translation der Mapt-mRNA. Tau spielt eine zentrale Rolle beim Aufbau und der Stabilität neuronaler Mikrotubuli. Bei AD führt die Anhäufung von abnormal hyperphosphoryliertem tau-Protein jedoch zu neuronaler
Degeneration. Der Verlust von hnRNP R verringert das axonale tau-Protein, jedoch nicht den Gesamtspiegel von tau. Wir beobachten, dass die Gehirne von 5xFAD-Mäusen, einem AD Mausmodell, durch Reduktion von hnRNP R geringere Mengen an Phospho-tau- und Amyloidβ-Plaques aufweisen. Ebenso zeigen Neuronen, die mit blockierenden ntisenseOligonukleotiden (ASO) behandelt wurden, um die Bindung von hnRNP R an Mapt-mRNA zu verhindern, verringerte axonale Mapt-mRNA Mengen und reduzierte Mengen an neu synthetisiertem tau-Protein. Unsere Daten legen daher nahe, dass die selektive Reduzierung des tau-Spiegels in Axonen, einem wichtigen subzellulären Ort der Neurofibrillenbildung, einen neuen therapeutischen Ansatz für die Behandlung der AD darstellen könnte.
|
2 |
Mechanisms of Axonal Transport Defects in ALSSeifert, Anne 21 May 2021 (has links)
Neurodegenerative diseases have become one of the most common causes of death worldwide over the last couple of decades, with increasing tendency. Amyotrophic lateral sclerosis (ALS) is the most common neurodegenerative disease affecting specifically spinal (lower) and cortical (upper) motor neurons in the spinal cord and brainstem, respectively. It is usually a late onset disorder (average age of onset in Germany is 61 years) and leads to death within 2-5 years after symptoms onset due to respiratory failure. To date, there is no cure for ALS and only two drugs have been approved for its treatment, which prolong the lifespan for up to six months or slow down disease progression in a subpopulation of patients. About 90 % of ALS cases are sporadic, while about 10 % are familial and hence caused by mutations in specific genes, among them fused in sarcoma (FUS), a DNA- and RNA-binding protein. Mutations in FUS account for roughly 5 % of familial cases and occur predominantly in its nuclear localization sequence (NLS), such as the FUS-P525L mutation. Neurons expressing this variant display a strong cytoplasmic mislocalization of FUS and hence a loss of its nuclear function. Among other pathological events, defects in axonal transport along microtubules have been observed early in disease progression in several models of FUS-ALS, indicating its role as a major hallmark of the disease. However, the mechanism of how transport is impaired within these neurons remains unknown to date. This study aimed at investigating two possible mechanisms how the FUS-P525L mutant variant affects microtubule-based axonal transport. First, it was analyzed whether FUS directly interacts with microtubules or motors and if the mislocalized, mutant variant alters this interaction. Secondly, cytoplasmic mislocalized FUS-P525L can no longer fulfil its regular role in the splicing of pre-mRNAs, among them the mRNA coding for the microtubule-associated protein tau. This reportedly leads to an increased ratio of translated tau isoforms containing four microtubule binding repeats (4R) to those containing three repeats (3R). 4R tau isoforms are known to have a stronger binding affinity towards microtubules and may hence impair transport more severely by acting as a roadblock for motor proteins. Towards this end, this study investigated whether an increase in 4R:3R tau isoform ratio is sufficient to impair microtubule based transport. Axonal transport was reconstituted in vitro using a kinesin-1-dependent microtubule gliding assays, in which microtubules are propelled by surface-immobilized kinesin-1 motors. The assay was modified and optimized to operate sensitively and robust in the presence of complex solutions such as whole cell lysates and the microtubule gliding velocity analyzed as a measure for motility of the underlying motors. To determine the direct interaction of FUS variants with kinesin-1 or microtubules, recombinant human wildtype FUS-GFP and FUS-P525L-GFP was added to the assay. In addition, ALS patient-specific induced pluripotent stem cells (iPSCs) expressing the same FUS variants were differentiated towards spinal motor neurons and their cell lysates applied to this assay in order to determine whether FUS variants need endogenous adaptors or interaction partners to interfere with kinesin-1 motility on microtubules. Further, to investigate the interference of tau isoforms with kinesin-1 motility, recombinant human 2N3R tau-GFP and 2N4R tau-mScarlet was purified from insect cells and added to the modified kinesin-1-dependent microtubule gliding assay, either individually or combined at different ratios. In addition, the binding of these tau variants to microtubules was assessed. The kinesin-1-dependent microtubule gliding assays was modified to operate sensitively and robustly in the presence of β-glycerophosphate (to inhibit endogenous phosphatases in whole cell lysates), and methylcellulose (to prevent microtubule detachment from kinesin-1 motors due to presence of β-glycerophosphate). Under these conditions, neither recombinant human FUS-GFP nor endogenous FUS-GFP variants in lysates of spinal motor neurons bound to microtubules or interfered with kinesin-1 motility. In contrast, both tau isoforms used in the present study bound to microtubules and impaired kinesin-1 motility, while 2N4R tau-mScarlet was a much more potent inhibitor of microtubule gliding and displayed a 20-fold stronger binding affinity to microtubules compared to 2N3R tau-GFP. Interestingly, increasing ratios of 4R:3R tau isoforms impaired kinesin-1-dependent microtubule gliding. In addition, the presence of 2N4R tau-mScarlet strongly prevented 2N3R tau-GFP from binding to microtubules. This study provides evidence that neither wildtype FUS nor the FUS-P525L variant directly interfere with axonal transport by interacting with kinesin-1 motors or microtubules. Further, the present data suggests that neither FUS variant impedes kinesin-1 motility on microtubules by interacting with endogenous adaptor proteins present in cell lysates of iPSC-derived spinal motor neurons. Therefore, it is proposed that axonal transport defects are not directly caused by interaction of cytoplasmic mislocalized FUS with the motors or microtubules, but rather arise as a consequence of other pathological events triggered by mutant FUS variants. In particular, this study demonstrates that an increased ratio of 4R:3R tau isoforms is sufficient to impair kinesin-1 motility on microtubules due to increased decoration of microtubules with 4R tau isoforms, preventing 3R tau isoforms from binding to microtubules. This strongly suggests that an increased ratio of 4R:3R tau isoforms, since FUS no longer regulates splicing of tau pre-mRNA upon its cytoplasmic mislocalization, may be sufficient to cause or contribute to the axonal transport defects observed early in FUS-ALS pathology. / Neurodegenerative Erkrankungen sind in den letzten Jahrzehnten mit zunehmender Tendenz zu einer der häufigsten Todesursachen weltweit geworden. Amyotrophe Lateralsklerose (ALS) ist die häufigste neurodegenerative Erkrankung, die spezifisch spinale (untere) und kortikale (obere) Motoneuronen im Rückenmark bzw. im Hirnstamm betrifft. Es handelt sich in der Regel um eine spät einsetzende Krankheit (das mittlere Erkrankungsalter in Deutschland beträgt 61 Jahre) und führt innerhalb von 2-5 Jahren nach Auftreten der Symptome zum Tod aufgrund von Atemversagen. Bisher gibt es keine Heilung für ALS und es wurden nur zwei Medikamente für die Behandlung zugelassen, die die Lebensdauer um bis zu sechs Monate verlängern oder das Fortschreiten der Krankheit bei einer Subpopulation von Patienten verlangsamen. Ungefähr 90% der ALS-Fälle sind sporadisch, während ungefähr 10% familiär sind und daher durch Mutationen in bestimmten Genen verursacht werden, darunter fused in sarcoma (FUS), einem DNA- und RNA-bindenden Protein. Mutationen in FUS machen etwa 5% der familiären Fälle aus und treten überwiegend in der Kernlokalisierungssequenz (NLS) auf, wie beispielsweise die FUS-P525L Mutation. Neuronen, die diese Mutante exprimieren, zeigen eine starke zytoplasmatische Fehllokalisierung von FUS und damit einen Verlust seiner Funktionen im Zellkern. Neben anderen pathologischen Ereignissen wurden in mehreren FUS-ALS Modellsystemen Defekte im Mikrotubuli-basierenden axonalen Transport früh im Krankheitsverlauf beobachtet, was auf seine Rolle als eines der Hauptmerkmale dieser Krankheit hindeutet. Der Mechanismus, wie der Transport innerhalb dieser Neuronen beeinträchtigt wird, ist jedoch bis heute unbekannt. Ziel dieser Studie ist es, zwei mögliche Mechanismen zu untersuchen, wie das mutierte FUS-P525L Protein den axonalen Transport entlang von Mikrotubuli beeinflusst. Zunächst wurde analysiert, ob FUS direkt mit Mikrotubuli oder Motorproteinen interagiert und ob zytoplasmatische fehllokalisierte FUS-P525L Protein diese Interaktion verändert. Ferner kann zytoplasmatische fehllokalisiertes FUS-P525L seine reguläre Rolle beim Spleißen von Prä-mRNAs nicht mehr erfüllen, darunter die mRNA, die für das mit Mikrotubuli-assoziierte Protein Tau kodiert. Dies führt zu einem erhöhten Verhältnis von translatierten Tau-Isoformen, die vier Mikrotubuli-Bindestellen (4R) enthalten, zu solchen mit drei Bindestellen (3R). Es ist bekannt, dass 4R-Tau-Isoformen eine stärkere Bindungsaffinität zu Mikrotubuli im Vergleich zu 3R-Tau-Isoformen aufweisen und daher den Transport stärker beeinträchtigen können, indem sie als Hindernis für Motorproteine agieren. In dieser Studie wurde daher untersucht, ob eine Erhöhung des Verhältnisses von 4R:3R-Tau-Isoform ausreicht, um den Mikrotubuli-basierenden Transport zu beeinträchtigen. Der axonale Transport wurde in vitro unter Verwendung eines Kinesin-1-gestuerten Mikrotubuli Motilitätsassay rekonstruiert, bei welchem Mikrotubuli von darunterliegenden oberflächenimmobilisierte Kinesin-1 Motorproteinen transportiert werden, also über die Oberfläche gleiten. Der Assay wurde modifiziert und optimiert, um in Gegenwart komplexer Lösungen wie Ganzzelllysaten sensitiv und robust zu funktionieren, und die Gleitgeschwindigkeit der Mikrotubuli wurde als Maß für die Motilität der darunterliegenden Motoren analysiert. Um die direkte Wechselwirkung von FUS-Varianten mit Kinesin-1 Motorproteinen oder Mikrotubuli zu bestimmen, wurde dem Assay rekombinantes menschliches Wildtyp-FUS-GFP und FUS-P525L-GFP hinzugegeben. Zusätzlich wurden ALS-patientenspezifische, induzierte pluripotente Stammzellen (iPSCs), welche dieselben FUS-Varianten exprimieren, zu spinalen Motoneuronen differenziert und ihre Zelllysate in diesem Assay angewendet, um zu bestimmen, ob FUS-Varianten endogene Adapter oder Interaktionspartner für die Interaction mit Kinesin-1 oder Mikrotubuli benötigen. Um den Einfluss von Tau-Isoformen auf die Kinesin-1 Motilität zu untersuchen, wurde rekombinantes menschliches 2N3R Tau-GFP und 2N4R Tau-mScarlet aus Insektenzellen aufgereinigt und dem modifizierten Kinesin-1-gesteuerten Mikrotubuli Motilitätsassay entweder einzeln oder in unterschiedlichen Verhältnissen kombiniert hinzugegeben. Zusätzlich wurde die Bindung dieser Tau-Varianten an Mikrotubuli analysiert. Der Kinesin-1-gesteuerte Mikrotubuli Motilitätsassay wurden so modifiziert, dass er in Gegenwart von β-Glycerophosphat (zur Hemmung endogener Phosphatasen in Ganzzelllysaten) und Methylcellulose (zur Verhinderung der Ablösung von Mikrotubuli von Kinesin-1 Motoren aufgrund von β-Glycerophosphat) empfindlich und robust funktioniert. Unter diesen Bedingungen zeigten weder rekombinantes menschliches FUS-GFP noch endogene FUS-GFP-Varianten in Lysaten von spinalen Motoneuronen eine Wechselwirkung mit Mikrotubuli und beeinträchtigten auch nicht die Kinesin-1 Motilität. Im Gegensatz dazu banden beide in der vorliegenden Studie verwendeten Tau-Isoformen an Mikrotubuli und beeinträchtigten die Kinesin-1-Motilität, wobei 2N4R Tau-mScarlet das Gleiten von Mikrotubuli viel stärkerer beeinträchtigte und eine 20-fach stärkere Bindungsaffinität zu Mikrotubuli im Vergleich zu 2N3R Tau-GFP zeigte. Ferner beeinträchtigten steigende Verhältnisse von 4R:3R Tau-Isoformen über Kinesin-1 gleitende Mikrotubuli, während die Präsenz von 2N4R Tau-mScarlet die Bindung von 2N3R Tau-GFP an Mikrotubuli stark verminderte. Diese Studie liefert Hinweise darauf, dass weder Wildtyp-FUS noch die FUS P525L-Variante den axonalen Transport direkt beeinflussen, da sie nicht mit Kinesin-1 Motorproteinen oder Mikrotubuli interagieren. Ferner legen die vorliegenden Daten nahe, dass keine der FUS-Varianten die Kinesin-1 Motilität auf Mikrotubuli durch Wechselwirkung mit endogenen Adapterproteinen behindert, die in Zelllysaten von iPSC-differenzierte spinalen Motoneuronen vorhanden sind. Dies legt nahe, dass axonale Transportdefekte nicht durch direkte Wechselwirkung von zytoplasmatisch fehllokalisiertem FUS Protein mit Motorproteinen oder Mikrotubuli verursacht werden, sondern als Folge anderer pathologischer Ereignisse auftreten, die durch mutierte FUS-Varianten entstehen. Insbesondere zeigt diese Studie, dass ein erhöhtes Verhältnis von 4R:3R Tau-Isoformen ausreicht, um die Kinesin-1 Motilität auf Mikrotubuli zu behindern. Dies geschieht vermutlich aufgrund der erhöhten Bindung von 4R Tau-Isoformen an Mikrotubuli, weil dadurch die Bindung von 3R Tau-Isoformen an Mikrotubuli verhindert wird. Dies deutet stark darauf hin, dass ein erhöhtes Verhältnis von 4R:3R Tau-Isoformen, verursacht durch die fehlende regulatorische Beteiligung von FUS am Spleißen von Tau-Prä-mRNA aufgrund der zytoplasmatischen Fehllokalisation von FUS, wahrscheinlich zu den axonalen Transportdefekten beiträgt, die früh in der FUS-ALS-Pathologie beobachtet wurden.
|
3 |
Examining FYCO1 as a modulator of autophagy for alpha-synuclein aggregate clearance in hiPSC derived neuronsBeer, Judith 21 February 2024 (has links)
Parkinson’s disease (PD) is the second most common neurodegenerative disorder worldwide affecting 1 - 2 % of the population older than 65. Patients develop characteristic motoric dysfunctions alongside early-onset non-motor symptoms including sleeping disorders, anxiety or depression and late-stage cognitive deficits such as dementia. To date, dopamine-replacement therapies are the gold standard for treating PD patients, improving motoric disorders by compensating for the loss of dopaminergic neurons in the substantia nigra, however no curative therapies to prevent disease progression are yet available. The pathomechanism underlying PD is complex, and the interplay of factors causing the disease is not entirely understood. The formation of α-synuclein protein aggregates, being one of the hallmarks associated with PD, is regarded as a major contributor to neuronal death and the spreading of PD pathology throughout different brain regions as the disease progresses. In the past, deficits in cellular protein clearance machinery have been affiliated with the accumulation of α-synuclein aggregates in PD. In particular, impairements in the macroautophagy-lysosomal pathway (here referred to as autophagy), which is involved in the degradation of large cytosolic components, were found to promote α-synuclein aggregation. In contrast, autophagic stimulation has been shown to benefit α-synuclein degradation and rescue PD phenotypes in cell and rodent models. In this study, I examined the role of FYCO1 in modulating neuronal autophagic processes for α-synuclein aggregate clearance in hiPSC-derived neurons. FYCO1 is an interaction partner of the central autophagic regulator RAB7 but was mostly unnoticed since it was not found detrimental to cellular homeostasis under basal conditions. Still, previous work of our group has identified FYCO1 to rescue PD phenotypes in model systems such as HEK cells and Drosophila, due to improved α-synuclein clearance following FYCO1 overexpression. Mechanistically, FYCO1 is involved in autophagosome-lysosome fusion events by binding to autophagic vesicles, which is required for autophagosome maturation and final degradation. In addition, FYCO1 affiliates autophagic vesicles with the cellular transport machinery via kinesin motor proteins. While fusion promotion can be assigned to an enhancing effect on autophagic clearance, FYCO1-induced anterograde transport promotion is opposite to the retrograde trafficking route of autophagic vesicles for maturation, which is of special importance in neuronal axons. Here, I illuminated FYCO1 effects on both axonal vesicle transport processes and somal vesicle pools to evaluate its ability to promote autophagy-related degradation in neurons. To this end, I established a lentiviral transduction-based model in hiPSC-derived neurons to express FYCO1 in the presence of either a fluorescently labelled marker for autophagic vesicles (LC3-TFL) or in the presence of α-synuclein. In neuronal axons, FYCO1 overexpression impaired retrograde autophagic transport resulting in less movement, implying an inhibitory effect on axonal autophagy. In contrast, FYCO1 enhanced autophagic processes in neuronal somata by upregulating LC3 levels, promoting the collection of α-synuclein in autophagic vesicle clusters and increasing the colocalisation of autophagosomes with lysosomal markers, pointing to the advance in autophagosome maturation. I could not fully resolve, whether α-synuclein degradation was promoted by this induction, as α-synuclein clearance was not indicated yet in the time course of three weeks. Still, studying mutant forms of FYCO1 revealed deficits in autophagosome maturation, which were not represented with wild-type FYCO1. In particular, the autophagosome-interaction domain was essential for autophagosome-lysosome fusion and additionally seemed to be relevant for autophagosomes entering axonal transport, while mutations in the kinesin binding domain caused autophagosome acidification impairments. The most pronounced effect of FYCO1 overexpression in neurons was the modulation of lysosomal vesicles. Besides increasing lysosomal localisation to autophagic vesicles, FYCO1 promoted retrograde trafficking of axonal lysosomal vesicles, by a so far unresolved mechanism. As increasing transport of lysosomes toward the neuronal soma can be connected to the upregulation of autophagy, I hypothesise FYCO1 to be a mediator in autophagy induction signalling. Nevertheless, such an effect needs to be verified in future studies. Conclusively, with this work, I contributed to the understanding of FYCO1’s role in enhancing neuronal autophagic processes but further studies in more advanced PD models are required to evaluate whether this could contribute to an increased clearance of α-synuclein aggregates.
|
4 |
Developing assays to characterize the effects of LRRK2 G2019S on axonal lysosomesBhatia, Priyanka 20 February 2024 (has links)
A striking feature of Parkinson's disease (PD) is that the distal axonal terminals of neurons degenerate prior to the soma, a process referred to as 'dying-back'. Another hallmark of the disease is the pathological accumulation of abnormal protein aggregates in soma and axons. Lysosomes, a critical component of the protein quality control machinery, have thus been thought to be altered in PD. LRRK2 G2019S, a gain-of-kinase-function mutation, is one of PD's most common known causative mutations, and LRRK2-specific small molecule inhibitors have been developed as possible therapeutics. However, LRRK2 G2019S is incompletely penetrant, and its role in axonal degeneration is unclear. LRRK2 phosphorylates a subset of Rab GTPases, including Rab10. Since Rab GTPases are mediators of organelle trafficking, we speculated that LRRK2 G2019S affects the transport of organelles, such as lysosomes, thereby contributing to early PD pathogenesis. Using neural progenitor cell-derived neurons from two LRRK2 G2019S-PD patients; we developed a model of axonal trafficking of lysosomes to characterize the impact of mutant LRRK2 on lysosomal trafficking. In comparison to their isogenic gene-corrected controls, we observed a subtle reduction in mutant axonal lysosomal speed, which could indicate that mutant LRRK2 mildly disrupts retrograde lysosomal transport. We also observed that this trafficking phenotype was only partially rescued by LRRK2 kinase inhibitors, which could indicate the importance of other factors regulating axonal transport. Consistent with this idea, we found that mutant LRRK2 was associated with increased co-localization of phosphorylated Rab10 on a small subset of distal axonal lysosomes. Furthermore, the over-expression of Rab10 only mildly affected lysosomal trafficking in axons. Interestingly, damaging the lysosomal membrane increased LRRK2-dependent Rab10 phosphorylation, leading us to speculate that membrane damage in the axon might induce LRRK2 activity. Since lysosomes have been shown to mediate plasma membrane repair, we speculated that membrane damage might exacerbate LRRK2-dependent phenotypes in distal axons. Axotomy was used to test this idea, and we observed an inconsistent delay in the regrowth of mutant axons after axotomy. Moreover, we identified an association between mutant LRRK2 and the transient increase in lysosomes at the injury site, indicating that LRRK2 G2019S might potentially affect damage-prone distal axons. Since the LRRK2 G2019S-associated phenotypes observed in our assays were relatively mild in one isogenic pair, we were curious about the clinical and genetic phenotypes of the patients from whom the somatic cells for neural progenitor cell generation were sourced. Interestingly, we observed that clinical features of PD, including age-of-onset, motor symptoms, cognitive impairment, and the level of cerebrospinal fluid biomarkers, were heterogeneous between the two patients. Additionally, genetic analysis of specific PD risk-associated loci in MAPT and SNCA revealed that one patient was more at risk of developing PD than the other, indicating influence from genetic factors in addition to LRRK2 G2019S. These factors might affect the axonal phenotypes observed in our assays. Overall, we have developed assays to investigate the effects of LRRK2 G2019S on axonal lysosomes. These assays can potentially be a useful tool to better understand early pathogenesis in heterogeneous PD patients and test targeted therapeutics that can be successful over an eclectic cohort of PD patients, all of whom are diagnosed based on deteriorating motor symptoms.:TABLE OF CONTENTS I
LIST OF FIGURES IV
LIST OF TABLES VI
ABBREVIATIONS VII
1 INTRODUCTION 1
1.1 Neurodegenerative diseases 1
1.2 Parkinson’s disease 2
1.2.1 General Features 2
1.2.2 Phenomenon of “dying back” in PD 6
1.2.3 Contribution of axonal architecture and function to “dying back” 7
1.2.4 Etiology of PD 10
1.2.4.1 Environmental factors 10
1.2.4.2 Genetic factors linked to axonal function 11
1.3 Lysosomes 12
1.3.1 Composition and biogenesis of lysosomes 13
1.3.2 Lysosomes as digestive centers 15
1.3.3 Lysosomes as secretory organelles 18
1.3.4 Lysosomes in PD 20
1.3.4.1 Genetic PD factors linked to lysosomal function 21
1.4 Leucine-rich repeat kinase 2 (LRRK2) 22
1.4.1 LRRK2 domain organization and function 22
1.4.2 Clinical features of PD patients with LRRK2 mutations (LRRK2-PD) 24
1.4.3 LRRK2 animal models 24
1.4.4 LRRK2 induced pluripotent stem cell (iPSC)-based models 25
1.4.5 Animal and iPSC-based models demonstrate a role for LRRK2 in the endo-lysosomal system 27
1.4.6 LRRK2 kinase inhibitors 30
2 AIMS OF THE THESIS 32
3 MATERIALS AND METHODS 33
3.1 Materials 33
3.1.1 Chemicals 33
3.1.2 Purchased kits 34
3.1.3 Plasmids 34
3.1.4 Antibodies 35
3.1.5 Dyes 36
3.1.6 Primers and oligonucleotides 36
3.1.7 Cell culture media and reagents 37
3.1.8 Small molecules 38
3.1.9 Compounds 38
3.1.10 Cell culture media 39
3.1.11 Human Neural Progenitor Cell (NPC) lines 40
3.2 Methods 41
3.2.1 Ethics statement 41
3.2.2 Licenses 41
3.2.3 Information about iPSC and NPC line generation 41
3.2.4 Preparation of cell culture coated plates 41
3.2.5 Maintenance of NPCs 42
3.2.6 Differentiation of NPCs to neurons 42
3.2.7 Preparation of microfluidic chambers 43
3.2.8 Seeding neurons as single cells 44
3.2.9 HEK293T cell culture 45
3.2.10 Treatment of neurons with compounds 45
3.2.11 Genomic DNA isolation 46
3.2.12 Polymerase-Chain Reaction (PCR) 46
3.2.13 Agarose gel electrophoresis 46
3.2.14 Plasmid DNA isolation 46
3.2.15 Lentiviral vector production 47
3.2.16 Lentiviral infection of human neurons 48
3.2.17 Protein isolation and quantification 48
3.2.18 Capillary electrophoresis 49
3.2.19 Axotomy 49
3.2.20 Immunostaining 50
3.2.21 Live cell imaging 51
3.2.22 Quantification of axonal trafficking using kymographs 52
3.2.23 Quantification of axonal trafficking using an object based method 53
3.2.24 Apotome imaging and quantification 54
3.2.25 Confocal imaging and quantification 54
3.2.26 Clinical and biomarker data collection 55
4 RESULTS 57
4.1 Establishing an axonal lysosomal trafficking assay 57
4.1.1 NPCs from LRRK2 G2019S patients and their respective isogenic controls differentiate into neurons 57
4.1.2 Axons can be spatially separated from soma and dendrites 60
4.1.3 Setting up the axonal trafficking assay 62
4.2 Axonal lysosomal trafficking assay detects LRRK2 G2019S associated changes in lysosome movement 65
4.3 Axonal lysosomal trafficking assay detects partial rescue by a small molecule LRRK2 inhibitor 71
4.4 LRRK2 G2019S is associated with an increase in the proportion of lysosomes co-localizing with phosphorylated Rab10 76
4.5 Rab10 over-expression mildly affects lysosomal trafficking in axons 78
4.6 Lysosomal membrane damage increases LRRK2-mediated Rab10 phosphorylation 81
4.7 LRRK2 G2019S is not associated with consistent effects on long-term axonal regrowth after axotomy 82
4.8 LRRK2 G2019S is associated with transient accumulation of lysosomes at the injury site after axotomy 86
4.9 Assessment of clinical, biomarker and genetic data from the LRRK2 G2019S patient donors 88
5 DISCUSSION 92
6 APPENDIX 101
7 SUMMARY 104
8 ZUSSAMENFASSUNG 106
9 BIBLIOGRAPHY 108
10 ACKNOWLEDGEMENTS 136
11 DECLARATIONS 138
|
Page generated in 0.0798 seconds