@phdthesis{Saal2017, author = {Saal, Lena}, title = {Whole transcriptome profiling of compartmentalized motoneurons}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-140006}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2017}, abstract = {Spinal muscular atrophy and amyotrophic lateral sclerosis are the two most common devastating motoneuron diseases. The mechanisms leading to motoneuron degeneration are not resolved so far, although different hypotheses have been built on existing data. One possible mechanism is disturbed axonal transport of RNAs in the affected motoneurons. The underlying question of this study was therefore to characterize changes in transcript levels of distinct RNAs in cell culture models of spinal muscular atrophy and amyotrophic lateral sclerosis, especially in the axonal compartment of primary motoneurons. To investigate this in detail we first established compartmentalized cultures of Primary mouse motoneurons. Subsequently, total RNA of both compartments was extracted separately and either linearly amplified and subjected to microarray profiling or whole transcriptome amplification followed by RNA-Sequencing was performed. To make the whole transcriptome amplification method suitable for compartmentalized cultures, we adapted a double-random priming strategy. First, we applied this method for initial optimization onto serial dilutions of spinal cord RNA and later on to the compartmentalized motoneurons. Analysis of the data obtained from wildtype cultures already revealed interesting results. First, the RNA composition of axons turned out to be highly similar to the somatodendritic compartment. Second, axons seem to be particularly enriched for transcripts related to protein synthesis and energy production. In a next step we repeated the experiments by using knockdown cultures. The proteins depleted hereby are Smn, Tdp-43 and hnRNP R. Another experiment was performed by knocking down the non-coding RNA 7SK, the main interacting RNA of hnRNP R. Depletion of Smn led to a vast number of deregulated transcripts in the axonal and somatodendritic compartment. Transcripts downregulated in the axons upon Smn depletion were especially enriched for GOterms related to RNA processing and encode proteins located in neuron projections including axons and growth cones. Strinkingly, among the upregulated transcripts in the somatodendritic compartment we mainly found MHC class I transcripts suggesting a potential neuroprotective role. In contrast, although knockdown of Tdp-43 also revealed a large number of downregulated transcripts in the axonal compartment, these transcripts were mainly associated with functions in transcriptional regulation and RNA splicing. For the hnRNP R knockdown our results were again different. Here, we observed downregulated transcripts in the axonal compartment mainly associated with regulation of synaptic transmission and nerve impulses. Interestingly, a comparison between deregulated transcripts in the axonal compartment of both hnRNP R and 7SK knockdown presented a significant overlap of several transcripts suggesting some common mechanism for both knockdowns. Thus, our data indicate that a loss of disease-associated proteins involved in axonal RNA transport causes distinct transcriptome alterations in motor axons.}, subject = {Axon}, language = {en} } @phdthesis{Wetzel2013, author = {Wetzel, Andrea}, title = {The role of TrkB and NaV1.9 in activity-dependent axon growth in motoneurons}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-92877}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2013}, abstract = {W{\"a}hrend der Entwicklung des Nervensystems lassen sich bei Motoneuronen aktivit{\"a}tsabh{\"a}ngige Kalziumstr{\"o}me eobachten, die das Axonwachstum regulieren. Diese Form der neuronalen Spontanaktivit{\"a}t sowie das Auswachsen von Axonen sind bei Motoneuronen, die aus Tiermodellen der Spinalen Muskelatrophie isoliert werden, gest{\"o}rt. Experimente aus unserer Arbeitsgruppe haben gezeigt, dass spontane Erregbarkeit und aktivit{\"a}tsabh{\"a}ngiges Axonwachstum von kultivierten Motoneuronen auch unter Verwendung von Toxinen beeintr{\"a}chtigt sind, welche die Aktivit{\"a}t von spannungsabh{\"a}ngigen Natriumkan{\"a}len blockieren. In diesen Versuchen war die Wirkung von Saxitoxin effizienter als die Wirkung von Tetrodotoxin. Wir identifizierten den Saxitoxin-sensitiven/Tetrodotoxin-insensitiven spannungsabh{\"a}ngigen Natriumkanal NaV1.9 als Trigger f{\"u}r das {\"O}ffnen spannungsabh{\"a}ngiger Kalziumkan{\"a}le. Die Expression von NaV1.9 in Motoneuronen konnte {\"u}ber quantitative RT-PCR nachgewiesen werden und antik{\"o}rperf{\"a}rbungen offenbarten eine Anreicherung des Kanals im axonalen Wachstumskegel sowie an Ranvier'schen Schn{\"u}rringen von isolierten Nervenfasern wildtypischer M{\"a}use. Motoneurone von NaV1.9 knock-out M{\"a}usen zeigen reduzierte Spontanaktivit{\"a}t und eine Reduktion des Axonwachstums, welche durch NaV1.9 {\"U}berexpression normalisiert werden kann. In Motoneuronen von Smn-defizienten M{\"a}usen konnte keine Abweichung der NaV1.9 Proteinverteilung nachgewiesen werden. K{\"u}rzlich wurden Patienten identifiziert, die eine missense-Mutation im NaV1.9 kodierenden SCN11A Gen tragen. Diese Patienten k{\"o}nnen keinerlei Schmerz empfinden und leiden zudem an Muskelschw{\"a}che in Kombination mit einer verz{\"o}gerten motorischen Entwicklung. Im Rahmen dieser Doktorarbeit konnten molekularbiologische Untersuchungen an M{\"a}usen, welche die Mutation im orthologen Scn11a Gen tragen, zur Aufkl{\"a}rung des Krankheitsmechanismus beitragen. Die Kooperationsstudie zeigte, dass eine gesteigerte Funktion von NaV1.9 diese spezifische Kanalerkrankung ausl{\"o}st, was die Wichtigkeit von NaV1.9 in menschlichen Motoneuronen unterstreicht. Eine fr{\"u}here Studie beschrieb an hippocampalen Neuronen, dass die Rezeptortyrosinkinase tropomyosin receptor kinase B (TrkB) den NaV1.9 Kanal {\"o}ffnen kann. Im Wachstumskegel von Motoneuronen ist TrkB nachweisbar und folglich in r{\"a}umlicher N{\"a}he zu NaV1.9 zu finden. Um zu pr{\"u}fen, ob TrkB in die spontane Erregbarkeit von Motoneuronen involviert ist, wurden TrkB knock-out M{\"a}use untersucht. Isolierte Motoneurone von TrkB knock-out M{\"a}usen weisen eine Reduktion der Spontanaktivit{\"a}t und eine Verringerung des Axonwachstums auf. Ob TrkB und NaV1.9 hierbei funktionell gekoppelt sind, ist Gegenstand k{\"u}nftiger Forschung.}, subject = {Motoneuron}, language = {en} } @phdthesis{Sivadasan2016, author = {Sivadasan, Rajeeve}, title = {The role of RNA binding proteins in motoneuron diseases}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-141907}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2016}, abstract = {Motoneuron diseases form a heterogeneous group of pathologies characterized by the progressive degeneration of motoneurons. More and more genetic factors associated with motoneuron diseases encode proteins that have a function in RNA metabolism, suggesting that disturbed RNA metabolism could be a common underlying problem in several, perhaps all, forms of motoneuron diseases. Recent results suggest that SMN interacts with hnRNP R and TDP-43 in neuronal processes, which are not part of the classical SMN complex. This point to an additional function of SMN, which could contribute to the high vulnerability of spinal motoneurons in spinal muscular atrophy (SMA) and amyotrophic lateral sclerosis (ALS). The current study elucidates functional links between SMN, the causative factor of SMA (spinal muscular atrophy), hnRNP R, and TDP-43, a genetic factor in ALS (amyotrophic lateral sclerosis). In order to characterize the functional interaction of SMN with hnRNP R and TDP-43, we produced recombinant proteins and investigated their interaction by co-immunoprecipitation. These proteins bind directly to each other, indicating that no other co-factors are needed for this interaction. SMN potentiates the ability of hnRNP R and TDP-43 to bind to ß-actin mRNA. Depletion of SMN alters the subcellular distribution of hnRNP R in motoneurons both in SMN-knockdown motoneurons and SMA mutant mouse (delta7 SMA). These data point to functions of SMN beyond snRNP assembly which could be crucial for recruitment and transport of RNA particles into axons and axon terminals, a mechanism which may contribute to SMA pathogenesis and ALS. ALS and FTLD (frontotemporal lobar degeneration) are linked by several lines of evidence with respect to clinical and pathological characteristics. Both sporadic and familial forms are a feature of the ALS-FTLD spectrum, with numerous genes having been associated with these pathological conditions. Both diseases are characterized by the pathological cellular aggregation of proteins. Interestingly, some of these proteins such as TDP-43 and FUS have also common relations not only with ALS-FTLD but also with SMA. Intronic hexanucleotide expansions in C9ORF72 are common in ALS and FTLD but it is unknown whether loss of function, toxicity by the expanded RNA or dipeptides from non ATG-initiated translation is responsible for the pathophysiology. This study tries to characterize the cellular function of C9ORF72 protein. To address this, lentiviral based knockdown and overexpression of C9ORF72 was used in isolated mouse motoneurons. The results clearly show that survival of these motoneurons was not affected by altered C9ORF72 levels, whereas adverse effects on axon growth and growth cone size became apparent after C9ORF72 suppression. Determining the protein interactome revealed several proteins in complexes with C9ORF72. Interestingly, C9ORF72 is present in a complex with cofilin and other actin binding proteins that modulate actin dynamics. These interactions were confirmed both by co-precipitation analyses and in particular by functional studies showing altered actin dynamics in motoneurons with reduced levels of C9ORF72. Importantly, the phosphorylation of cofilin is enhanced in C9ORF72 depleted motoneurons and patient derived lymphoblastoid cells with reduced C9ORF72 levels. These findings indicate that C9ORF72 regulates axonal actin dynamics and the loss of this function could contribute to disease pathomechanisms in ALS and FTLD.}, subject = {Motoneuron}, language = {en} } @phdthesis{Pei2000, author = {Pei, Geng}, title = {The Role of Raf-mediated Signalling Pathways for Motoneuron}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-1846}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2000}, abstract = {The transmission of proliferative and developmental signals from activated cell-surface receptors to initiation of cellular responses in the nucleus is synergically controlled by the coordinated action of a diverse set of intracellular signalling proteins. The Ras/Raf/MEK/MAPK signalling pathway has been shown to control the expression of genes which are crucial for the physiological regulation of cell proliferation, differentiation and apoptosis. Within this signalling cascade, the Raf protein family of serine/threonine kinases serves as a central intermediate which connects to many of other signal transduction pathways. To elucidate the signalling functions of the different Raf kinases in motoneurons during development, the expression, distribution and subcellular localization of Rafs in the spinal cord and the facial nucleus in brainstem of mice at various embryonic and postnatal stages were investigated. Moreover, we have investigated the intracellular redistribution of Raf molecules in isolated motoneurons from 13 or 14 day old mouse embryos, after addition or withdrawal of neurotrophic factors to induce Raf kinases activation in vitro. Furthermore, in order to investigate the potential anti-apoptotic function of Raf kinases on motoneurons, we isolated motoneurons from B-raf-/- and c-raf-1-/- mouse embryos and analysed the survival and differentiation effects of neurotrophic factors in motoneurons lacking B-Raf and c-Raf-1. We provide evidence here that all three Raf kinases are expressed in mouse spinal motoneurons. Their expression increases during the period of naturally occurring cell death of motoneurons. In sections of embryonic and postnatal spinal cord, motoneurons express exclusively B-Raf and c-Raf-1, but not A-Raf, and subcellularly Raf kinases are obviously colocalized with mitochondria. In isolated motoneurons, most of the B-Raf or c-Raf-1 immunoreactivity is located in the perinuclear space but also in the nucleus, especially after activation by addition of CNTF and BDNF in vitro. We found that c-Raf-1 translocation from the cytosol into the nucleus of motoneurons after its activation by neurotrophic factors is a distinct event. As a central finding of our study, we observed that the viability of isolated motoneurons from B-raf but not c-raf-1 knockout mice is lost even in the presence of CNTF and other neurotrophic factors. This indicates that B-Raf but not c-Raf-1, which is still present in B-raf deficient motoneurons, plays a crucial role in mediating the survival effect of neurotrophic factors during development. In order to prove that B-Raf is an essential player in this scenario, we have re-expressed B-Raf in mutant sensory and motor neurons by transfection. The motoneurons and the sensory neurons from B-raf knockout mouse which were transfected with exogenous B-raf gene revealed the same viability in the presence of neurotrophic factors as primary neurons from wild-type mice. Our results suggest that Raf kinases have important signalling functions in motoneurons in mouse CNS. In vitro, activation causes redistribution of Raf protein kinases, particularly for c-Raf-1, from motoneuronal cytoplasm into the nucleus. This redistribution of c-Raf-1, however, is not necessary for the survival effect of neurotrophic factors, given that B-raf-/- motor and sensory neurons can not survive despite the presence of c-Raf-1. We hypothesize that c-Raf-1 nuclear translocation may play a direct role in transcriptional regulation as a consequence of neurotrophic factor induced phosphorylation and activation of c-Raf-1 in motoneurons. Moreover, the identification of target genes for nuclear translocated c-Raf-1 and of specific cellular functions initiated by this mechanism awaits its characterization.}, subject = {Maus}, language = {en} } @phdthesis{Rathod2012, author = {Rathod, Reenaben Jagdishbhai}, title = {Study of local protein synthesis in growth cones of embryonic mouse motor neurons}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-72045}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2012}, abstract = {In cultured motoneurons of a mouse model for the motoneuron disease spinal muscular atrophy (SMA), reduced levels of the protein SMN (survival of motoneurons) cause defects in axonal growth. This correlates with reduced β-actin mRNA and protein in growth cones, indicating that anterograde transport and local translation of β-actin mRNA are crucial for motoneuron function. However, direct evidence that indeed local translation is a physiological phenomenon in growth cones of motoneurons was missing. Here, a lentiviral GFP-based reporter construct was established to monitor local protein synthesis of β-actin mRNA. Time-lapse imaging of fluorescence recovery after photobleaching (FRAP) in living motoneurons revealed that β-actin is locally translated in the growth cones of embryonic motoneurons. Interestingly, local translation of the β-actin reporter construct was differentially regulated by different laminin isoforms, indicating that laminins provide extracellular cues for the regulation of local translation in growth cones. Notably, local translation of β-actin mRNA was deregulated when motoneurons of a mouse model for type I SMA (Smn-/-; SMN2) were analyzed. In situ hybridization revealed reduced levels of β-actin mRNA in the axons of Smn-/-; SMN2 motoneurons. The distribution of the β-actin mRNA was not modified by different laminin isoforms as revealed by in situ hybridization against the mRNA of the eGFP encoding element of the β-actin reporter. In case of the mRNA of α-actin and γ-actin isoforms, the endogenous mRNA did not localize to the axons and the localization pattern was not affected by the SMN levels expressed in the cell. Taken together our findings suggest that regulation of local translation of β-actin in growth cones of motoneurons critically depends on laminin signaling and the amount of SMN protein. Embryonic stem cell (ESC)-derived motoneurons are an excellent in vitro system to sort out biochemical and cellular pathways which are defective in neurodegenerative diseases like SMA. Here, a protocol for the differentiation and antibody-mediated enrichment of ESC-derived motoneurons is presented, which was optimized during the course of this study. Notably, this study contributes the production and purification of highly active recombinant sonic hedgehog (Shh), which was needed for the efficient differentiation of mouse ESCs to motoneurons. ESC-derived motoneurons will now offer high amounts of cellular material to allow the biochemical identification of disease-relevant molecular components involved in regulated local protein synthesis in axons and growth cones of motoneurons.}, subject = {Motoneuron}, language = {en} } @phdthesis{Subramanian2011, author = {Subramanian, Narayan}, title = {Role of NaV1.9 in activity dependent axon growth in embryonic cultured motoneurons}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-57536}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2011}, abstract = {Spontaneous neural activity has been shown to regulate crucial events in neurite growth including axonal branching and path finding. In animal models of spinal muscular atrophy (SMA) cultured embryonic mouse motoneurons show distinct defect in axon elongation and neural activity. This defect is governed by abnormal clustering of Ca2+ channels in the axonal regions and the protruding growth cone area. The mechanisms that regulate the opening of calcium channels in developing motoneurons are not yet clear. The question was addressed by blocking neural activity in embryonic cultured motoneurons by pharmacological inhibition of voltage-gated sodium channels (VGSC) by saxitoxin (STX) and tetrodotoxin (TTX). Low dosages of STX resulted in significant reduction of axon growth and neural activity in cultured motoneurons. This pharmacological treatment did not affect survival of motoneurons in comparison to control motoneurons that was grown in the presence of survival neurotrophic factors BDNF and CNTF. It was also found that STX was 10 times more potent than TTX a common inhibitor of VGSC with a reduced activity on the TTX-insensitive sodium channels NaV1.5, NaV1.8 and NaV1.9. Reverse Transcriptase-PCR experiments revealed the presence of NaV1.9 as the likely candidate that begins to express from embryonic stage sixteen in the mouse spinal cord. Immunolabelling experiments showed that the channel is expressed in the axonal compartments and axonal growth cones in cultured motoneurons. Suppression of NaV1.9 in cultured motoneurons by lentivirus mediated short hairpin-RNA (shRNA) resulted in shorter axon length in comparison with uninfected and scrambled constructs. Further, embryonic motoneurons cultured from NaV1.9 knockout mice also showed a significant reduction in neural activity and axon growth. The findings of this work highlight the role of NaV1.9 as an important contender in regulating activity dependent axon growth in embryonic cultured motoneurons. NaV1.9 could therefore be considered as a prospective molecule that could play an important role in regulating axon growth in motoneuron disease models like spinal muscular atrophy (SMA).}, subject = {Axon}, language = {en} } @phdthesis{ThangarajSelvaraj2013, author = {Thangaraj Selvaraj, Bhuvaneish}, title = {Role of CNTF-STAT3 signaling for microtubule dynamics inaxon growth and maintenance: Implications in motoneuron diseases}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-76889}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2013}, abstract = {Neurotrophic factor signaling modulates differentiation, axon growth and maintenance, synaptic plasticity and regeneration of neurons after injury. Ciliary neurotrophic factor (CNTF), a Schwann cell derived neurotrophic factor, has an exclusive role in axon maintenance, sprouting and synaptic preservation. CNTF, but not GDNF, has been shown to alleviate motoneuron degeneration in pmn mutant mice carrying a missense mutation in Tbce gene, a model for Amyotrophic Lateral Sclerosis (ALS). This current study elucidates the distinct signaling mechanism by which CNTF rescues the axonal degeneration in pmn mutant mice. ...}, subject = {Ciliary neurotrophic factor}, language = {en} } @phdthesis{Schmitt2017, author = {Schmitt, Dominique}, title = {Initial characterization of mouse Syap1 in the nervous system: Search for interaction partners, effects of gene knockdown and knockout, and tissue distribution with focus on the adult brain}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-147319}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2017}, abstract = {The synapse-associated protein of 47 kDa (Sap47) in Drosophila melanogaster is the founding member of a phylogenetically conserved protein family of hitherto unknown molecular function. Sap47 is localized throughout the entire neuropil of adult and larval brains and closely associated with glutamatergic presynaptic vesicles of larval motoneurons. Flies lacking the protein are viable and fertile and do not exhibit gross structural or marked behavioral deficiencies indicating that Sap47 is dispensable for basic synaptic function, or that its function is compensated by other related proteins. Syap1 - the mammalian homologue of Sap47 - was reported to play an essential role in Akt1 phosphorylation in various non-neuronal cells by promoting the association of mTORC2 with Akt1 which is critical for the downstream signaling cascade for adipogenesis. The function of Syap1 in the vertebrate nervous system, however, is unknown so far. The present study provides a first description of the subcellular localization of mouse Syap1 in cultured motoneurons as well as in selected structures of the adult mouse nervous system and reports initial functional experiments. Preceding all descriptive experiments, commercially available Syap1 antibodies were tested for their specificity and suitability for this study. One antibody raised against the human protein was found to recognize specifically both the human and murine Syap1 protein, providing an indispensable tool for biochemical, immunocytochemical and immunohistochemical studies. In the course of this work, a Syap1 knockout mouse was established and investigated. These mice are viable and fertile and do not show obvious changes in morphology or phenotype. As observed for Sap47 in flies, Syap1 is widely distributed in the synaptic neuropil, particularly in regions rich in glutamatergic synapses but it was also detected at perinuclear Golgi-associated sites in certain groups of neuronal somata. In motoneurons the protein is especially observed in similar perinuclear structures, partially overlapping with Golgi markers and in axons, dendrites and axonal growth cones. Biochemical and immunohistochemical analyses showed widespread Syap1 expression in the central nervous system with regionally distinct distribution patterns in cerebellum, hippocampus or olfactory bulb. Besides its expression in neurons, Syap1 is also detected in non-neuronal tissue e.g. liver, kidney and muscle tissue. In contrast, non-neuronal cells in the brain lack the typical perinuclear accumulation. First functional studies with cultured primary motoneurons on developmental, structural and functional aspects reveal no influence of Syap1 depletion on survival and morphological features such as axon length or dendritic length. Contrary to expectations, in neuronal tissues or cultured motoneurons a reduction of Akt phosphorylation at Ser473 or Thr308 was not detected after Syap1 knockdown or knockout.}, subject = {Synapse}, language = {en} } @phdthesis{Massih2024, author = {Massih, Bita}, title = {Human stem cell-based models to analyze the pathophysiology of motor neuron diseases}, publisher = {Frontiers in Cell and Developmental Biology}, doi = {10.25972/OPUS-34637}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-346374}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2024}, abstract = {Motor neuron diseases (MNDs) encompass a variety of clinically and genetically heterogeneous disorders, which lead to the degeneration of motor neurons (MNs) and impaired motor functions. MNs coordinate and control movement by transmitting their signal to a target muscle cell. The synaptic endings of the MN axon and the contact site of the muscle cell thereby form the presynaptic and postsynaptic structures of the neuromuscular junction (NMJ). In MNDs, synaptic dysfunction and synapse elimination precede MN loss suggesting that the NMJ is an early target in the pathophysiological cascade leading to MN death. In this study, we established new experimental strategies to analyze human MNDs by patient derived induced pluripotent stem cells (iPSCs) and investigated pathophysiological mechanisms in two different MNDs. To study human MNDs, specialized cell culture systems that enable the connection of MNs to their target muscle cells are required to allow the formation of NMJs. In the first part of this study, we established and validated a human neuromuscular co-culture system consisting of iPSC derived MNs and 3D skeletal muscle tissue derived from myoblasts. We generated 3D muscle tissue by culturing primary myoblasts in a defined extracellular matrix in self-microfabricated silicone dishes that support the 3D tissue formation. Subsequently, iPSCs from healthy donors and iPSCs from patients with the progressive MND Amyotrophic Lateral Sclerosis (ALS) were differentiated into MNs and used for 3D neuromuscular co-cultures. Using a combination of immunohistochemistry, calcium imaging, and pharmacological stimulations, we characterized and confirmed the functionality of the 3D muscle tissue and the 3D neuromuscular co-cultures. Finally, we applied this system as an in vitro model to study the pathophysiology of ALS and found a decrease in neuromuscular coupling, muscle contraction, and axonal outgrowth in co-cultures with MNs harboring ALS-linked superoxide dismutase 1 (SOD1) mutation. In summary, this co-culture system presents a human model for MNDs that can recapitulate aspects of ALS pathophysiology. In the second part of this study, we identified an impaired unconventional protein secretion (UPS) of Sod1 as pathological mechanisms in Pleckstrin homology domain-containing family G member 5 (Plekhg5)-associated MND. Sod1 is a leaderless cytosolic protein which is secreted in an autophagy-dependent manner. We found that Plekhg5 depletion in primary MNs and NSC34 cells leads to an impaired secretion of wildtype Sod1, indicating that Plekhg5 drives the UPS of Sod1 in vitro. By interfering with different steps during the biogenesis of autophagosomes, we could show that Plekhg5-regulated Sod1 secretion is determined by autophagy. To analyze our findings in a clinically more relevant model we utilized human iPSC MNs from healthy donors and ALS patients with SOD1 mutations. We observed reduced SOD1 secretion in ALS MNs which coincides with reduced protein expression of PLEKHG5 compared to healthy and isogenic control MNs. To confirm this correlation, we depleted PLEKHG5 in control MNs and found reduced extracellular SOD1 levels, implying that SOD1 secretion depends on PLEKHG5. In summary, we found that Plekh5 regulates the UPS of Sod1 in mouse and human MNs and that Sod1 secretion occurs in an autophagy dependent manner. Our data shows an unreported mechanistic link between two MND-associated proteins.}, subject = {Tissue Engineering}, language = {en} } @phdthesis{Deng2023, author = {Deng, Chunchu}, title = {Dynamic remodeling of endoplasmic reticulum and ribosomes in axon terminals of wildtype and Spinal Muscular Atrophy motoneurons}, doi = {10.25972/OPUS-26495}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-264954}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2023}, abstract = {In highly polarized neurons, endoplasmic reticulum (ER) forms a dynamic and continuous network in axons that plays important roles in lipid synthesis, Ca2+ homeostasis and the maintenance of synapses. However, the mechanisms underlying the regulation of axonal ER dynamics and its function in regulation of local translation still remain elusive. In the course of my thesis, I investigated the fast dynamic movements of ER and ribosomes in the growth cone of wildtype motoneurons as well as motoneurons from a mouse model of Spinal Muscular Atrophy (SMA), in response to Brain-derived neurotrophic factor (BDNF) stimulation. Live cell imaging data show that ER extends into axonal growth cone filopodia along actin filaments and disruption of actin cytoskeleton by cytochalasin D treatment impairs the dynamic movement of ER in the axonal filopodia. In contrast to filopodia, ER movements in the growth cone core seem to depend on coordinated actions of the actin and microtubule cytoskeleton. Myosin VI is especially required for ER movements into filopodia and drebrin A mediates actin/microtubule coordinated ER dynamics. Furthermore, we found that BDNF/TrkB signaling induces assembly of 80S ribosomes in growth cones on a time scale of seconds. Activated ribosomes relocate to the presynaptic ER and undergo local translation. These findings describe the dynamic interaction between ER and ribosomes during local translation and identify a novel potential function for the presynaptic ER in intra-axonal synthesis of transmembrane proteins such as the α-1β subunit of N-type Ca2+ channels in motoneurons. In addition, we demonstrate that in Smn-deficient motoneurons, ER dynamic movements are impaired in axonal growth cones that seems to be due to impaired actin cytoskeleton. Interestingly, ribosomes fail to undergo rapid structural changes in Smn-deficient growth cones and do not associate to ER in response to BDNF. Thus, aberrant ER dynamics and ribosome response to extracellular stimuli could affect axonal growth and presynaptic function and maintenance, thereby contributing to the pathology of SMA.}, subject = {Motoneuron}, language = {en} } @phdthesis{Moradi2017, author = {Moradi, Mehri}, title = {Differential roles of α-, β- and γ-actin isoforms in regulation of cytoskeletal dynamics and stability during axon elongation and collateral branch formation in motoneurons}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-147453}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2017}, abstract = {In highly polarized cells like neurons, cytoskeleton dynamics play a crucial role in establishing neuronal connections during development and are required for adult plasticity. Actin turnover is particularly important for neurite growth, axon path finding, branching and synaptogenesis. Motoneurons establish several thousand branches that innervate neuromuscular synapses (NMJs). Axonal branching and terminal arborization are fundamental events during the establishment of synapses in motor endplates. Branching process is triggered by the assembly of actin filaments along the axon shaft giving rise to filopodia formation. The unique contribution of the three actin isoforms, α-, β- and γ-actin, in filopodia stability and dynamics during this process is not well characterized. Here, we performed high resolution in situ hybridization and qRT-PCR and showed that in primary mouse motoneurons α-, β- and γ-actin isoforms are expressed and their transcripts are translocated into axons. Using FRAP experiments, we showed that transcripts for α-, β- and γ-actin become locally translated in axonal growth cones and translation hot spots of the axonal branch points. Using live cell imaging, we showed that shRNA depletion of α-actin reduces dynamics of axonal filopodia which correlates with reduced number of collateral branches and impairs axon elongation. Depletion of β-actin correlates with reduced dynamics of growth cone filopoida, disturbs axon elongation and impairs presynaptic differentiation. Also, depletion of γ-actin impairs axonal growth and decreases axonal filopodia dynamics. These findings implicate that actin isoforms accomplish unique functions during development of motor axons. Depletions of β- and γ-actin lead to compensatory upregulation of other two isoforms. Consistent with this, total actin levels remain unaltered and F-actin polymerization capacity is preserved. After the knockdown of either α- or γ-actin, the levels of β-actin increase in the G-actin pool indicating that polymerization and stability of β-actin filaments depend on α- or γ-actin. This study provides evidence both for unique and overlapping function of actin isoforms in motoneuron growth and differentiation. In the soma of developing motoneurons, actin isoforms act redundantly and thus could compensate for each other's loss. In the axon, α-, β- and γ-actin accomplish specific functions, i.e. β-actin regulates axon elongation and plasticity and α- and γ-actin regulate axonal branching. Furthermore, we show that both axonal transport and local translation of α-, β- and γ-actin isoforms are impaired in Smn knockout motoneurons, indicating a role for Smn protein in RNA granule assembly and local translation of these actin isoforms in primary mouse motoneurons.}, subject = {Motoneuron}, language = {en} } @phdthesis{Hochgraefe2009, author = {Hochgr{\"a}fe, Katja}, title = {Cre-loxP based mouse models to study prionpathogenesis in the motor nervous system}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-45967}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2009}, abstract = {Prion diseases such as scrapie in sheep, bovine spongiform encephalopathy (BSE) in cattle or Creutzfeldt-Jakob disease (CJD) in humans are fatal neurodegenerative disorders characterized by brain lesions and the accumulation of a disease-associated protein, designated PrPSc. How prions proceed to damage neurons and whether all or only subsets of neurons have to be affected for the onset of the clinical disease is still elusive. The manifestation of clinical prion disease is characterized by motor dysfunctions, dementia and death. Furthermore loss of motor neurons (MN) in the spinal cord is a constant finding in different mouse models of prion disease, suggesting that MN are vulnerable cells for triggering the onset of clinical symptoms. To determine whether the protection of MN against prion induced dysfunctions is an approach for holding the disease at the sub-clinical level, we established a novel conditional model for Cre-mediated expression of a dominant-negative PrP mutant (PrPQ167R) in the cells of interest. Dominant-negative PrP mutants provide protection of prion induced dysfunctions by inhibiting prion replication. Transgenic mice were generated carrying a floxed LacZ marker gene followed by the coding sequence of PrPQ167R under control of the human ubiquitin C promoter. Two Cre strains have been used to direct PrPQ167R expression either to a subset of MN of the spinal cord (Hb9-Cre) or to various neuronal cell populations of the spinal cord and brain (NF-L-Cre). Transgenic mice were infected with mouse-adapted prions via different inoculation routes (intranerval, intracerebral and intraperitoneal) and monitored for effects on incubation time and pathology. Tg floxed LacZ-PrPQ167R/NF-L-Cre mice showed about 15\% prolonged survival upon intraperitoneal low dose prion infection, whereas survival of Tg floxed LacZ-PrPQ167R/Hb9-Cre mice was comparable to control littermates. The results suggest that the protection of spinal MN prolongs the incubation period but is not sufficient to completely inhibit clinical prion disease. In a second approach, Cre was transferred into the hind limb muscles of transgenic mice via a double-stranded adeno-associated virus vector (dsAAV2-Cre). The goal of this strategy was to target a broader cell population and thus to enhance expression levels of protective PrPQ167R in the spinal cord of Tg floxed-LacZ-PrPQ167R mice. After intramuscular (i.m.) application of dsAAV2-Cre, exhibiting a physical titer of 5x1010 GP/ml, recombinant transgenic DNA was detected only in the muscle tissue, pointing out that functional Cre-recombinase was expressed at the side of virus application. However, dsAAV2-Cre did neither induce recombination of transgenic DNA in the spinal cord or brain nor expression of dominant-negative PrPQ167R. In conclusion the dsAAV2-Cre vectors system needs further improvement to achieve efficient transport from muscle tissue to the central nervous system (CNS). 105 7 SUMMARY The lymphoreticular system (LRS) is an early site of prion replication. In splenic tissue prion infectivity is associated with follicular dendritic cells (FDC) as well as with Band T-lymphocytes. However, it is still unknown if those cell types are able to replicate the infectious agent or if other PrP-expressing cell types are engaged. To investigate if neurons and in particular MN are involved, transgenic mice carrying one allele of floxed Prnp (lox2+=\&\#56256;\&\#56320;) and either one allele of Hb9-Cre or NF-L-Cre were generated on a Prnp0=0 background. Therefore a conditional PrP knockout was established in a subset of MN of the spinal cord (Hb9-Cre) or in various neuronal populations of the spinal cord and brain (NF-L-Cre). Transgenic mice were inoculated with prions to study the accumulation of PrPSc and prion infectivity in spleen and spinal cord at an early time point after infection. The findings show that PrPSc accumulation in mice with MN-specific PrP depletion (lox2+=\&\#56256;\&\#56320;/ Hb9-Cre) was comparable to control littermates, while pan-neuronal PrP deficient mice (lox2+=\&\#56256;\&\#56320;/NF-L-Cre) were not able to accumulate PrPSc in splenic tissue until 50 days post inoculation. Moreover spleens of lox2+=\&\#56256;\&\#56320;/NF-L-Cre mice exhibited a clearly reduced prion infectivity titer, suggesting that accumulation of prions in the spleen is dependent on PrP expression in the nervous tissue.}, subject = {Prionkrankheit}, language = {en} } @phdthesis{Viswanathan2022, author = {Viswanathan, Aravindan}, title = {Biochemical and structural characterisation of modules within the SMN complex}, doi = {10.25972/OPUS-19474}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-194749}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2022}, abstract = {Cellular proteome profiling revealed that most biomolecules do not exist in isolation, but rather are incorporated into modular complexes. These assembled complexes are usually very large, consisting of 10 subunits on an average and include either proteins alone, or proteins and nucleic acids. Consequently, such macromolecular assemblies rather than individual biopolymers perform the vast majority of cellular activities. The faithful assembly of such molecular assemblies is often aided by trans-acting factors in vivo, to preclude aggregation of complex components and/or non-cognate interactions. A paradigm for an assisted assembly of a macromolecular machine is the formation of the common Sm/LSm core of spliceosomal and histone-mRNA processing U snRNPs. The key assembly factors united in the Protein Arginine Methyltransferase 5 (PRMT5) and the Survival Motor Neuron (SMN) complexes orchestrate the assembly of the Sm/LSm core on the U snRNAs. Assembly is initiated by the PRMT5-complex subunit pICln, which pre-arranges the Sm/LSm proteins into spatial positions occupied in the mature U snRNPs. The SMN complex subsequently binds these Sm/LSm units, displaces pICln and catalyses the Sm ring closure on the Sm-site of the U snRNA. The SMN complex consists of the eponoymous SMN protein linked in a modular network of interactions with eight other proteins, termed Gemins 2-8 and Unrip. Despite functional and structural characterisation of individual protein components and/or sub-complexes of this assembly machinery, coherent understanding of the structural framework of the core SMN complex remained elusive. The current work, employing a combined approach of biochemical and structural studies, aimed to contribute to the understanding of how distinct modules within the SMN complex coalecse to form the macromolecular SMN complex. A novel atomic resolution (1.5 {\AA}) structure of the human Gemin8:7:6 sub-complex, illustrates how the peripheral Gemin7:6 module is tethered to the SMN complex via Gemin8's C-terminus. In this model, Gemin7 engages with both Gemin6 and Gemin8 via the N- and C-termini of its Sm-fold like domain. This highly conserved interaction mode is reflected in the pronounced sequence conservation and identical biochemical behaviour of similar sub-complexes from divergent species, namely S. pombe and C. elegans. Despite lacking significant sequence similarity to the Sm proteins, the dimeric Gemin7:6 complex share structural resemblance to the Sm heteromers. The hypothesis that the dimeric Gemin7:6 functions as a Sm-surrogate during Sm core assembly could not be confirmed in this work. The functional relevance of the structural mimicry of the dimeric Gemin7:6 sub-complex with the Sm heterodimers therefore still remains unclear. Reduced levels of functional SMN protein is the cause of the devastating neurodegenerative disease, Spinal Muscular Atrophy (SMA). The C-terminal YG-zipper motif of SMN is a major hot-spot for most SMA patient mutations. In this work, adding to the existing inventory of the human and fission yeast YG-box models, a novel 2.2 {\AA} crystal structure of the nematode SMN's YG-box domain adopting the glycine zipper motif has been reported. Furthermore, it could be assessed that SMA patient mutations mapping to this YG-box domain greatly influences SMN's self-association competency, a property reflected in both the human and nematode YG-box biochemical handles. The shared molecular architecture and biochemical behaviour of the nematode SMN YG-box domain with its human and fission yeast counterparts, reiterates the pronounced conservation of this oligomerisation motif across divergent organisms. Apart from serving as a multimerization domain, SMN's YG-box also acts as interaction platform for Gemin8. A systematic investigation of SMA causing missense mutations uncovered that Gemin8's incorporation into the SMN complex is influenced by the presence of certain SMA patient mutations, albeit independent of SMN's oligomerisation status. Consequently, loss of Gemin8 association in the presence of SMA patient mutations would also affect the incorporation of Gemin7:6 sub-complex. Gemin8, therefore sculpts the heteromeric SMN complex by bridging the Gemin7:6 and SMN:Gemin2 sub-units, a modular feature shared in both the human and nematode SMN complexes. These findings provide an important foundation and a prospective structural framework for elucidating the core architecture of the SMN complex in the ongoing Cryo-EM studies.}, subject = {Proteom}, language = {en} }