257 results on '"J. Kahle"'
Search Results
2. Sirtuin-1 sensitive lysine-136 acetylation drives phase separation and pathological aggregation of TDP-43
- Author
-
Jorge Garcia Morato, Friederike Hans, Felix von Zweydorf, Regina Feederle, Simon J. Elsässer, Angelos A. Skodras, Christian Johannes Gloeckner, Emanuele Buratti, Manuela Neumann, and Philipp J. Kahle
- Subjects
Science - Abstract
TDP-43 is a nucleic acid binding protein, whose insoluble aggregates are neuropathological hallmarks of specific subsets of patients with amyotrophic lateral sclerosis and frontotemporal lobar degeneration. Post-translational modifications and acetylation of TDP-43 impact its interaction with RNA, its localization in the cells, and are linked to disease. Using antibodies generated against TDP-43 lysine acetylation sites, sirtuin-1 was found to potently deacetylate amber suppressed [acK136]TDP-43 and reduce its aggregation propensity. Thus, distinct lysine acetylations modulate nuclear import, RNA binding as well as phase separation and aggregation of TDP-43, suggesting regulatory mechanisms for TDP-43 pathogenesis.
- Published
- 2022
- Full Text
- View/download PDF
3. Microglial inclusions and neurofilament light chain release follow neuronal α-synuclein lesions in long-term brain slice cultures
- Author
-
Melanie Barth, Mehtap Bacioglu, Niklas Schwarz, Renata Novotny, Janine Brandes, Marc Welzer, Sonia Mazzitelli, Lisa M. Häsler, Manuel Schweighauser, Thomas V. Wuttke, Deborah Kronenberg-Versteeg, Karina Fog, Malene Ambjørn, Ania Alik, Ronald Melki, Philipp J. Kahle, Derya R. Shimshek, Henner Koch, Mathias Jucker, and Gaye Tanriöver
- Subjects
Alpha-synuclein ,Microglia ,Neurofilament light chain ,Slice culture ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract 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 (αS) inclusions were induced in long-term murine and human slice cultures by seeded aggregation. An αS seed-recognizing human antibody was tested for blocking seeding and/or spreading of the αS lesions. Release of neurofilament light chain (NfL) into the culture medium was assessed. Results To study initial stages of α-synucleinopathies, we induced αS inclusions in murine hippocampal slice cultures by seeded aggregation. Induction of αS inclusions in neurons was apparent as early as 1week post-seeding, followed by the occurrence of microglial inclusions in vicinity of the neuronal lesions at 2–3 weeks. The amount of αS inclusions was dependent on the type of αS seed and on the culture’s genetic background (wildtype vs A53T-αS genotype). Formation of αS 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 αS seeds resulted in spreading of αS inclusions to neuronally connected hippocampal subregions, and seeding and spreading could be inhibited by an αS 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 αS lesions was achieved at 1week post-seeding in combination with viral A53T-αS expressions. Conclusion The successful translation of these brain cultures from mouse to human with the first reported induction of human αS 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.
- Published
- 2021
- Full Text
- View/download PDF
4. Prominent microglial inclusions in transgenic mouse models of α-synucleinopathy that are distinct from neuronal lesions
- Author
-
Gaye Tanriöver, Mehtap Bacioglu, Manuel Schweighauser, Jasmin Mahler, Bettina M. Wegenast-Braun, Angelos Skodras, Ulrike Obermüller, Melanie Barth, Deborah Kronenberg-Versteeg, K. Peter R. Nilsson, Derya R. Shimshek, Philipp J. Kahle, Yvonne S. Eisele, and Mathias Jucker
- Subjects
Synuclein ,Microglia ,Inclusion ,Prion-like ,Amyloid ,Conformation ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Alpha-synucleinopathies are a group of progressive neurodegenerative disorders, characterized by intracellular deposits of aggregated α-synuclein (αS). The clinical heterogeneity of these diseases is thought to be attributed to conformers (or strains) of αS but the contribution of inclusions in various cell types is unclear. The aim of the present work was to study αS conformers among different transgenic (TG) mouse models of α-synucleinopathies. To this end, four different TG mouse models were studied (Prnp-h[A53T]αS; Thy1-h[A53T]αS; Thy1-h[A30P]αS; Thy1-mαS) that overexpress human or murine αS and differed in their age-of-symptom onset and subsequent disease progression. Postmortem analysis of end-stage brains revealed robust neuronal αS pathology as evidenced by accumulation of αS serine 129 (p-αS) phosphorylation in the brainstem of all four TG mouse lines. Overall appearance of the pathology was similar and only modest differences were observed among additionally affected brain regions. To study αS 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 αS 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 αS, but were largely p-αS-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 αS aggregation. Although nature and significance of microglial inclusions for human α-synucleinopathies remain to be clarified, the previously overlooked abundance of microglial inclusions in TG mouse models of α-synucleinopathy bears importance for mechanistic and preclinical-translational studies.
- Published
- 2020
- Full Text
- View/download PDF
5. LAG3 is not expressed in human and murine neurons and does not modulate α‐synucleinopathies
- Author
-
Marc Emmenegger, Elena De Cecco, Marian Hruska‐Plochan, Timo Eninger, Matthias M Schneider, Melanie Barth, Elena Tantardini, Pierre de Rossi, Mehtap Bacioglu, Rebekah G Langston, Alice Kaganovich, Nora Bengoa‐Vergniory, Andrès Gonzalez‐Guerra, Merve Avar, Daniel Heinzer, Regina Reimann, Lisa M Häsler, Therese W Herling, Naunehal S Matharu, Natalie Landeck, Kelvin Luk, Ronald Melki, Philipp J Kahle, Simone Hornemann, Tuomas P J Knowles, Mark R Cookson, Magdalini Polymenidou, Mathias Jucker, and Adriano Aguzzi
- Subjects
LAG3 ,neurodegeneration ,prionoids ,α‐synuclein ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract While the initial pathology of Parkinson’s disease and other α‐synucleinopathies is often confined to circumscribed brain regions, it can spread and progressively affect adjacent and distant brain locales. This process may be controlled by cellular receptors of α‐synuclein fibrils, one of which was proposed to be the LAG3 immune checkpoint molecule. Here, we analysed the expression pattern of LAG3 in human and mouse brains. Using a variety of methods and model systems, we found no evidence for LAG3 expression by neurons. While we confirmed that LAG3 interacts with α‐synuclein fibrils, the specificity of this interaction appears limited. Moreover, overexpression of LAG3 in cultured human neural cells did not cause any worsening of α‐synuclein pathology ex vivo. The overall survival of A53T α‐synuclein transgenic mice was unaffected by LAG3 depletion, and the seeded induction of α‐synuclein lesions in hippocampal slice cultures was unaffected by LAG3 knockout. These data suggest that the proposed role of LAG3 in the spreading of α‐synucleinopathies is not universally valid.
- Published
- 2021
- Full Text
- View/download PDF
6. Graphical prior elicitation in univariate models.
- Author
-
Christopher J. Casement and David J. Kahle
- Published
- 2018
- Full Text
- View/download PDF
7. Human Dopaminergic Neurons Lacking PINK1 Exhibit Disrupted Dopamine Metabolism Related to Vitamin B6 Co-Factors
- Author
-
Christine Bus, Laimdota Zizmare, Marita Feldkaemper, Sven Geisler, Maria Zarani, Anna Schaedler, Franziska Klose, Jakob Admard, Craig J. Mageean, Giuseppe Arena, Petra Fallier-Becker, Aslihan Ugun-Klusek, Klaudia K. Maruszczak, Konstantina Kapolou, Benjamin Schmid, Doron Rapaport, Marius Ueffing, Nicolas Casadei, Rejko Krüger, Thomas Gasser, Daniela M. Vogt Weisenhorn, Philipp J. Kahle, Christoph Trautwein, Christian J. Gloeckner, and Julia C. Fitzgerald
- Subjects
Molecular Biology ,Molecular Neuroscience ,Stem Cells Research ,Omics ,Science - Abstract
Summary: PINK1 loss-of-function mutations cause early onset Parkinson disease. PINK1-Parkin mediated mitophagy has been well studied, but the relevance of the endogenous process in the brain is debated.Here, the absence of PINK1 in human dopaminergic neurons inhibits ionophore-induced mitophagy and reduces mitochondrial membrane potential. Compensatory, mitochondrial renewal maintains mitochondrial morphology and protects the respiratory chain. This is paralleled by metabolic changes, including inhibition of the TCA cycle enzyme mAconitase, accumulation of NAD+, and metabolite depletion. Loss of PINK1 disrupts dopamine metabolism by critically affecting its synthesis and uptake. The mechanism involves steering of key amino acids toward energy production rather than neurotransmitter metabolism and involves cofactors related to the vitamin B6 salvage pathway identified using unbiased multi-omics approaches.We propose that reduction of mitochondrial membrane potential that cannot be controlled by PINK1 signaling initiates metabolic compensation that has neurometabolic consequences relevant to Parkinson disease.
- Published
- 2020
- Full Text
- View/download PDF
8. Parkin-dependent mitophagy occurs via proteasome-dependent steps sequentially targeting separate mitochondrial sub-compartments for autophagy
- Author
-
Anna Lechado-Terradas, Sandra Schepers, Katharina I. Zittlau, Karan Sharma, Orkun Ok, Julia C. Fitzgerald, Stefan Geimer, Benedikt Westermann, Boris Macek, and Philipp J. Kahle
- Published
- 2022
- Full Text
- View/download PDF
9. Failure of diet-induced transcriptional adaptations in alpha-synuclein transgenic mice
- Author
-
Alexander Kilzheimer, Thomas Hentrich, Carola Rotermund, Philipp J Kahle, and Julia M Schulze-Hentrich
- Subjects
Gene Expression Profiling ,metabolism [Parkinson Disease] ,Mice, Transgenic ,General Medicine ,Mice, Inbred C57BL ,Mice ,genetics [Parkinson Disease] ,ddc:570 ,genetics [alpha-Synuclein] ,alpha-Synuclein ,Genetics ,Animals ,metabolism [alpha-Synuclein] ,adverse effects [Diet, High-Fat] ,Molecular Biology ,Genetics (clinical) - Abstract
Nutritional influences have been discussed as potential modulators of Parkinson’s disease (PD) pathology through various epidemiological and physiological studies. In animal models, a high-fat diet (HFD) with greater intake of lipid-derived calories leads to accelerated disease onset and progression. The underlying molecular mechanisms of HFD-induced aggravated pathology, however, remain largely unclear. In this study, we aimed to further illuminate the effects of a fat-enriched diet in PD by examining the brainstem and hippocampal transcriptome of alpha-synuclein transgenic mice exposed to a life-long HFD. Investigating individual transcript isoforms, differential gene expression and co-expression clusters, we observed that transcriptional differences between wild-type (WT) and transgenic animals intensified in both regions under HFD. Both brainstem and hippocampus displayed strikingly similar transcriptomic perturbation patterns. Interestingly, expression differences resulted mainly from responses in WT animals to HFD, while these genes remained largely unchanged or were even slightly oppositely regulated by diet in transgenic animals. Genes and co-expressed gene groups exhibiting this dysregulation were linked to metabolic and mitochondrial pathways. Our findings propose the failure of metabolic adaptions as the potential explanation for accelerated disease unfolding under exposure to HFD. From the identified clusters of co-expressed genes, several candidates lend themselves to further functional investigations.
- Published
- 2022
- Full Text
- View/download PDF
10. Confidence intervals for the ratio of two Poisson rates under one-way differential misclassification using double sampling.
- Author
-
David J. Kahle, Philip D. Young, Brandi A. Greer, and Dean M. Young
- Published
- 2016
- Full Text
- View/download PDF
11. Chronic–Progressive Dopaminergic Deficiency Does Not Induce Midbrain Neurogenesis
- Author
-
Mareike Fauser, Francisco Pan-Montojo, Christian Richter, Philipp J. Kahle, Sigrid C. Schwarz, Johannes Schwarz, Alexander Storch, and Andreas Hermann
- Subjects
adult neurogenesis ,periventricular regions ,non-neurogenic regions ,Parkinson´s disease ,dopaminergic neurodegeneration ,transgenic animal model ,Cytology ,QH573-671 - Abstract
Background: Consecutive adult neurogenesis is a well-known phenomenon in the ventricular–subventricular zone of the lateral wall of the lateral ventricles (V–SVZ) and has been controversially discussed in so-called “non-neurogenic” 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 chronic–progressive dopaminergic neurodegeneration on both consecutive adult neurogenesis in the PVRs of the V–SVZ and mid/hindbrain aNSCs in two mechanistically different transgenic animal models of Parkinson´s disease (PD). Methods: We used Thy1-m[A30P]h α synuclein mice and Leu9′Ser hypersensitive α4* nAChR mice to assess the influence of midbrain dopaminergic neuronal loss on neurogenic activity in the PVRs of the V–SVZ, the aqueduct and the fourth ventricle. Results: In both animal models, overall proliferative activity in the V–SVZ was not altered, though the proportion of B2/activated B1 cells on all proliferating cells was reduced in the V–SVZ in Leu9′Ser hypersensitive α4* 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.
- Published
- 2021
- Full Text
- View/download PDF
12. Environmental Enrichment Prevents Transcriptional Disturbances Induced by Alpha-Synuclein Overexpression
- Author
-
Zinah Wassouf, Thomas Hentrich, Sebastian Samer, Carola Rotermund, Philipp J. Kahle, Ingrid Ehrlich, Olaf Riess, Nicolas Casadei, and Julia M. Schulze-Hentrich
- Subjects
alpha-synuclein ,enriched environment ,Parkinson's disease ,gene-environment interaction ,immediate early genes ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Onset and progression of neurodegenerative disorders, including synucleinopathies such as Parkinson's disease, have been associated with various environmental factors. A highly compelling association from a therapeutic point of view has been found between a physically active lifestyle and a significantly reduced risk for Parkinson's disease. Mimicking such conditions in animal models by promoting physical activity, social interactions, and novel surroundings yields in a so-called enriched environment known to enhance adult neurogenesis, increase synaptic plasticity, and decelerate neuronal loss. Yet, the genes that connect beneficial environmental cues to the genome and delay disease-related symptoms have remained largely unclear. To identify such mediator genes, we used a 2 × 2 factorial design opposing genotype and environment. Specifically, we compared wildtype to transgenic mice overexpressing human SNCA, a key gene in synucleinopathies encoding alpha-synuclein, and housed them in a standard and enriched environment from weaning to 12 months of age before profiling their hippocampal transcriptome using RNA-sequencing. Under standard environmental conditions, differentially expressed genes were overrepresented for calcium ion binding, membrane, synapse, and other Gene Ontology terms previously linked to alpha-synuclein biology. Upregulated genes were significantly enriched for genes attributed to astrocytes, microglia, and oligodendrocytes. These disturbances in gene activity were accompanied by reduced levels of several presynaptic proteins and the immediate early genes EGR1 and NURR1. Intriguingly, housing transgenic animals in the enriched environment prevented most of these perturbations in gene activity. In addition, a sustained activation specifically in transgenic animals housed in enriched conditions was observed for several immediate early genes including Egr1, Nr4a2/Nurr1, Arc, and Homer1a. These findings suggest a compensatory mechanism through an enriched environment-activated immediate early gene network that prevented most disturbances induced by alpha-synuclein overexpression. This regulatory framework might harbor attractive targets for novel therapeutic approaches that mimic beneficial environmental stimuli.
- Published
- 2018
- Full Text
- View/download PDF
13. Intracranial administration of alpha-synuclein fibrils in A30P-synuclein transgenic mice causes robust synucleinopathy and microglial induction
- Author
-
Philipp J. Kahle, Renee C Gentzel, Lei Ma, Jacob Marcus, Sean M. Smith, Dawn Toolan, Joel B. Schachter, and Sarah Jinn
- Subjects
Genetically modified mouse ,Aging ,Alpha-Synuclein ,Parkinson's disease ,Synucleinopathies ,administration & dosage [alpha-Synuclein] ,Gene Expression ,Mice, Transgenic ,adverse effects [alpha-Synuclein] ,A30P ,Biology ,chemistry.chemical_compound ,genetics [Parkinson Disease] ,medicine ,Animals ,Transgenic mice ,ddc:610 ,metabolism [alpha-Synuclein] ,Alpha-synuclein ,Parkinson's Disease ,Microglia ,Tyrosine hydroxylase ,Dementia with Lewy bodies ,General Neuroscience ,pathology [Microglia] ,genetics [Synucleinopathies] ,Parkinson Disease ,etiology [Synucleinopathies] ,medicine.disease ,pathology [Parkinson Disease] ,Cell biology ,Disease Models, Animal ,medicine.anatomical_structure ,pathology [Synucleinopathies] ,chemistry ,alpha-Synuclein ,genetics [alpha-Synuclein] ,Synuclein ,etiology [Parkinson Disease] ,Neurology (clinical) ,Geriatrics and Gerontology ,Developmental Biology - Abstract
Synucleinopathies are neurodegenerative disorders involving pathological alpha-synuclein (αSyn) protein, including dementia with Lewy bodies, multiple system atrophy and Parkinson's disease (PD). Current in vivo models of synucleinopathy include transgenic mice overexpressing αSyn variants and methods based on administration of aggregated, exogenous αSyn. Combining these techniques offers the ability to study consequences of introducing pathological αSyn into primed neuronal environments likely to develop synucleinopathy. Herein, we characterize the impacts pre-formed fibrils (PFFs) of recombinant, human αSyn have in mice overexpressing human A30P αSyn, a mutation associated with autosomal dominant PD. A30P mouse brain contains detergent insoluble αSyn biochemically similar to PD brain, and these mice develop Lewy-like synucleinopathy with age. Administration of PFFs in A30P mice resulted in regionally-specific accumulations of phosphorylated synuclein, microglial induction and a motor phenotype that differed from PFF-induced effects in wildtype mice. Surprisingly, PFF-induced losses of tyrosine hydroxylase were similar in A30P and wildtype mice. Thus, the PFF-A30P model recapitulates key aspects of synucleinopathy with induction of microglia, creating an appropriate system for evaluating neurodegenerative therapeutics.
- Published
- 2021
- Full Text
- View/download PDF
14. Nuclear import factor transportin and arginine methyltransferase 1 modify FUS neurotoxicity in Drosophila
- Author
-
Sandra Jäckel, Anna K. Summerer, Catherine M. Thömmes, Xia Pan, Aaron Voigt, Jörg B. Schulz, Tobias M. Rasse, Dorothee Dormann, Christian Haass, and Philipp J. Kahle
- Subjects
FUS ,Caz ,Transportin ,PRMT ,ALS ,FTLD ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Inclusions containing Fused in Sarcoma (FUS) are found in familial and sporadic cases of the incurable progressive motor neuron disease amyotrophic lateral sclerosis and in a common form of dementia, frontotemporal dementia. Most disease-associated mutations are located in the C-terminal proline–tyrosine nuclear localization sequence (PY-NLS) of FUS and impair its nuclear import. It has been shown in cell culture that the nuclear import of FUS is mediated by transportin, which binds the PY-NLS and the last arginine/glycine/glycine-rich (RGG) domain of FUS. Methylation of this last RGG domain by protein arginine methyltransferases (PRMTs) weakens transportin binding and therefore impairs nuclear translocation of FUS. To investigate the requirements for the nuclear import of FUS in an in vivo model, we generated different transgenic Drosophila lines expressing human FUS wild type (hFUS wt) and two disease-related variants P525L and R495X, in which the NLS is mutated or completely absent, respectively. To rule out effects caused by heterologous hFUS expression, we analysed the corresponding variants for the Drosophila FUS orthologue Cabeza (Caz wt, P398L, Q349X). Expression of these variants in eyes and motor neurons confirmed the PY-NLS-dependent nuclear localization of FUS/Caz and caused neurodegenerative effects. Surprisingly, FUS/Caz toxicity was correlated to the degree of its nuclear localization in this overexpression model. High levels of nuclear FUS/Caz became insoluble and reduced the endogenous Caz levels, confirming FUS autoregulation in Drosophila. RNAi-mediated knockdown of the two transportin orthologues interfered with the nuclear import of FUS/Caz and also enhanced the eye phenotype. Finally, we screened the Drosophila PRMT proteins (DART1-9) and found that knockdown of Dart1 led to a reduction in methylation of hFUS P525L and aggravated its phenotype.These findings show that the molecular mechanisms controlling the nuclear import of FUS/Caz and FUS autoregulation are conserved between humans and Drosophila. In addition to the well-known neurodegenerative effects of FUS loss-of function, our data suggest toxic potential of overexpressed FUS in the nucleus and of insoluble FUS.
- Published
- 2015
- Full Text
- View/download PDF
15. Basic Bayesian Model Checking
- Author
-
John W. Seaman, David J. Kahle, and James D. Stamey
- Published
- 2022
- Full Text
- View/download PDF
16. An Overview of Bayesian Computation
- Author
-
David J. Kahle, John W. Seaman, and James D. Stamey
- Published
- 2022
- Full Text
- View/download PDF
17. Immunotherapy targeting α-synuclein protofibrils reduced pathology in (Thy-1)-h[A30P] α-synuclein mice
- Author
-
Veronica Lindström, Therese Fagerqvist, Eva Nordström, Fredrik Eriksson, Anna Lord, Stina Tucker, Jessica Andersson, Malin Johannesson, Heinrich Schell, Philipp J. Kahle, Christer Möller, Pär Gellerfors, Joakim Bergström, Lars Lannfelt, and Martin Ingelsson
- Subjects
Protofibril-selective antibody ,Immunization ,α-Synuclein transgenic mice ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Several lines of evidence suggest that accumulation of aggregated alpha-synuclein (α-synuclein) in the central nervous system (CNS) is an early pathogenic event in Parkinson's disease and other Lewy body disorders. In recent years, animal studies have indicated immunotherapy with antibodies directed against α-synuclein as a promising novel treatment strategy. Since large α-synuclein oligomers, or protofibrils, have been demonstrated to possess pronounced cytotoxic properties, such species should be particularly attractive as therapeutic targets. In support of this, (Thy-1)-h[A30P] α-synuclein transgenic mice with motor dysfunction symptoms were found to display increased levels of α-synuclein protofibrils in the CNS. An α-synuclein protofibril-selective monoclonal antibody (mAb47) was evaluated in this α-synuclein transgenic mouse model. As measured by ELISA, 14 month old mice treated for 14 weeks with weekly intraperitoneal injections of mAb47 displayed significantly lower levels of both soluble and membrane-associated protofibrils in the spinal cord. Besides the lower levels of pathogenic α-synuclein demonstrated, a reduction of motor dysfunction in transgenic mice upon peripheral administration of mAb47 was indicated. Thus, immunotherapy with antibodies targeting toxic α-synuclein species holds promise as a future disease-modifying treatment in Parkinson's disease and related disorders.
- Published
- 2014
- Full Text
- View/download PDF
18. Signatures of glial activity can be detected in the CSF proteome
- Author
-
Timo Eninger, Stephan A. Müller, Mehtap Bacioglu, Manuel Schweighauser, Marius Lambert, Luis F. Maia, Jonas J. Neher, Sarah M. Hornfeck, Ulrike Obermüller, Gernot Kleinberger, Christian Haass, Philipp J. Kahle, Matthias Staufenbiel, Lingyan Ping, Duc M. Duong, Allan I. Levey, Nicholas T. Seyfried, Stefan F. Lichtenthaler, Mathias Jucker, and Stephan A. Kaeser
- Subjects
Multidisciplinary ,Proteome ,Gene Expression Profiling ,metabolism [Parkinson Disease] ,Parkinson Disease ,tau Proteins ,metabolism [Proteome] ,cerebrospinal fluid [Alzheimer Disease] ,metabolism [Cerebrospinal Fluid] ,Mice ,cerebrospinal fluid [Biomarkers] ,Alzheimer Disease ,cerebrospinal fluid [Parkinson Disease] ,Animals ,Humans ,ddc:500 ,Single-Cell Analysis ,metabolism [Neuroglia] ,Neuroglia ,metabolism [Alzheimer Disease] ,Biomarkers ,Cerebrospinal Fluid - Abstract
Single-cell transcriptomics has revealed specific glial activation states associated with the pathogenesis of neurodegenerative diseases, such as Alzheimer’s and Parkinson’s disease. While these findings may eventually lead to new therapeutic opportunities, little is known about how these glial responses are reflected by biomarker changes in bodily fluids. Such knowledge, however, appears crucial for patient stratification, as well as monitoring disease progression and treatment responses in clinical trials. Here, we took advantage of well-described mouse models of β-amyloidosis and α-synucleinopathy to explore cerebrospinal fluid (CSF) proteome changes related to their respective proteopathic lesions. Nontargeted liquid chromatography-mass spectrometry revealed that the majority of proteins that undergo age-related changes in CSF of either mouse model were linked to microglia and astrocytes. Specifically, we identified a panel of more than 20 glial-derived proteins that were increased in CSF of aged β-amyloid precursor protein- and α-synuclein-transgenic mice and largely overlap with previously described disease-associated glial genes identified by single-cell transcriptomics. Our results also show that enhanced shedding is responsible for the increase of several of the identified glial CSF proteins as exemplified for TREM2. Notably, the vast majority of these proteins can also be quantified in human CSF and reveal changes in Alzheimer’s disease cohorts. The finding that cellular transcriptome changes translate into corresponding changes of CSF proteins is of clinical relevance, supporting efforts to identify fluid biomarkers that reflect the various functional states of glial responses in cerebral proteopathies, such as Alzheimer’s and Parkinson’s disease.
- Published
- 2022
- Full Text
- View/download PDF
19. Multiple distinct pathways lead to hyperubiquitylated insoluble TDP-43 protein independent of its translocation into stress granules
- Author
-
Hanna Glasebach, Friederike Hans, and Philipp J. Kahle
- Subjects
Indoles ,drug effects [Solubility] ,Protein aggregation ,Biochemistry ,Ubiquitin ,genetics [Heat-Shock Proteins] ,GSK-3 ,genetics [Oxidative Stress] ,Sorbitol ,genetics [Ubiquitination] ,genetics [Frontotemporal Dementia] ,Heat-Shock Proteins ,biology ,Kinase ,Chemistry ,drug effects [Osmotic Pressure] ,pharmacology [Acetylcysteine] ,Molecular Bases of Disease ,Cell biology ,DNA-Binding Proteins ,drug effects [Mutation] ,Protein Transport ,genetics [Amyotrophic Lateral Sclerosis] ,Frontotemporal Dementia ,genetics [Protein Transport] ,Signal transduction ,analogs & derivatives [Adenine] ,Signal Transduction ,drug effects [Signal Transduction] ,genetics [DNA-Binding Proteins] ,pharmacology [Indoles] ,Protein Aggregation, Pathological ,Stress granule ,Osmotic Pressure ,mental disorders ,pharmacology [Adenine] ,Humans ,genetics [Protein Aggregation, Pathological] ,genetics [Glycogen Synthase Kinase 3 beta] ,ddc:610 ,Protein kinase A ,pathology [Amyotrophic Lateral Sclerosis] ,Molecular Biology ,Glycogen Synthase Kinase 3 beta ,Adenine ,Endoplasmic reticulum ,pharmacology [Sorbitol] ,Amyotrophic Lateral Sclerosis ,Ubiquitination ,nutritional and metabolic diseases ,Cell Biology ,chemistry [DNA-Binding Proteins] ,Acetylcysteine ,nervous system diseases ,Oxidative Stress ,HEK293 Cells ,Solubility ,Mutation ,pathology [Frontotemporal Dementia] ,biology.protein ,chemistry [Heat-Shock Proteins] - Abstract
Insoluble, hyperubiquitylated TAR DNA-binding protein of 43 kDa (TDP-43) in the central nervous system characterizes frontotemporal dementia and ALS in many individuals with these neurodegenerative diseases. The causes for neuropathological TDP-43 aggregation are unknown, but it has been suggested that stress granule (SG) formation is important in this process. Indeed, in human embryonic kidney HEK293E cells, various SG-forming conditions induced very strong TDP-43 ubiquitylation, insolubility, and reduced splicing activity. Osmotic stress–induced SG formation and TDP-43 ubiquitylation occurred rapidly and coincided with colocalization of TDP-43 and SG markers. Washout experiments confirmed the rapid dissolution of SGs, accompanied by normalization of TDP-43 ubiquitylation and solubility. Surprisingly, interference with the SG process using a protein kinase R–like endoplasmic reticulum kinase inhibitor (GSK2606414) or the translation blocker emetine did not prevent TDP-43 ubiquitylation and insolubility. Thus, parallel pathways may lead to pathological TDP-43 modifications independent of SG formation. Using a panel of kinase inhibitors targeting signaling pathways of the osmotic shock inducer sorbitol, we could largely rule out the stress-activated and extracellular signal–regulated protein kinase modules and glycogen synthase kinase 3β. For arsenite, but not for sorbitol, quenching oxidative stress with N-acetylcysteine did suppress both SG formation and TDP-43 ubiquitylation and insolubility. Thus, sodium arsenite appears to promote SG formation and TDP-43 modifications via oxidative stress, but sorbitol stimulates TDP-43 ubiquitylation and insolubility via a novel pathway(s) independent of SG formation. In conclusion, pathological TDP-43 modifications can be mediated via multiple distinct pathways for which SGs are not essential.
- Published
- 2020
- Full Text
- View/download PDF
20. A Novel Wnt5a Variant in Context of Maternal Diabetes Leading to a Severe Phenotypic Presentation
- Author
-
Prapti Singh, Juliette J. Kahle, Alexander N. Wein, Kayla Quirin, Yang Cao, and Jorge L. Granadillo
- Published
- 2022
- Full Text
- View/download PDF
21. Temporal Analysis of Protein Ubiquitylation and Phosphorylation During Parkin-Dependent Mitophagy
- Author
-
Katharina I. Zittlau, Anna Lechado-Terradas, Nicolas Nalpas, Sven Geisler, Philipp J. Kahle, and Boris Macek
- Subjects
quantitative proteomics ,Ubiquitin-Protein Ligases ,Mitophagy ,Ubiquitination ,Biochemistry ,Analytical Chemistry ,mitochondria ,metabolism [Ubiquitin-Protein Ligases] ,mitophagy ,ubiquitin ,Humans ,ddc:610 ,parkin ,Phosphorylation ,Molecular Biology ,HeLa Cells - Abstract
Mitophagy, the selective degradation of mitochondria by autophagy, affects defective mitochondria following damage or stress. At the onset of mitophagy, parkin ubiquitylates proteins on the mitochondrial outer membrane. While the role of parkin at the onset of mitophagy is well understood, less is known about its activity during later stages in the process. Here, we used HeLa cells expressing catalytically active or inactive parkin to perform temporal analysis of the proteome, ubiquitylome, and phosphoproteome during 18 h after induction of mitophagy by mitochondrial uncoupler carbonyl cyanide m-chlorophenyl hydrazine. Abundance profiles of proteins downregulated in parkin-dependent manner revealed a stepwise and "outside-in" directed degradation of mitochondrial subcompartments. While ubiquitylation of mitochondrial outer membrane proteins was enriched among early parkin-dependent targets, numerous mitochondrial inner membrane, matrix, and cytosolic proteins were also found ubiquitylated at later stages of mitophagy. Phosphoproteome analysis revealed a possible crosstalk between phosphorylation and ubiquitylation during mitophagy on key parkin targets, such as voltage-dependent anion channel 2.
- Published
- 2022
- Full Text
- View/download PDF
22. Olfactory neuron-specific expression of A30P alpha-synuclein exacerbates dopamine deficiency and hyperactivity in a novel conditional model of early Parkinson's disease stages
- Author
-
Silke Nuber, Elisabeth Petrasch-Parwez, Oscar Arias-Carrión, Leanie Koch, Zacharias Kohl, Jacqueline Schneider, Carsten Calaminus, Rolf Dermietzel, Anna Samarina, Jana Boy, Huu P. Nguyen, Peter Teismann, Thirumalaisamy Palanichamy Velavan, Philipp J. Kahle, Stephan von Hörsten, Markus Fendt, Rejko Krüger, and Olaf Riess
- Subjects
Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Mutations in the N-terminus of the gene encoding α-synuclein (α-syn) are linked to autosomal dominantly inherited Parkinson's disease (PD). The vast majority of PD patients develop neuropsychiatric symptoms preceding motor impairments. During this premotor stage, synucleinopathy is first detectable in the olfactory bulb (OB) and brain stem nuclei; however its impact on interconnected brain regions and related symptoms is still less far understood. Using a novel conditional transgenic mouse model, displaying region-specific expression of human mutant α-syn, we evaluated effect and reversibility of olfactory synucleinopathy. Our data showed that induction of mutant A30P α-syn expression increased transgenic deposition into somatodendritic compartment of dopaminergic neurons, without generating fibrillar inclusions. We found reversibly reduced levels of dopamine and metabolites in the OB, suggesting an impact of A30P α-syn on olfactory neurotransmitter content. We further showed that mutant A30P expression led to neurodegenerative changes on an ultrastructural level and a behaviorally hyperactive response correlated with novelty, odor processing and stress associated with an increased dopaminergic tone in midbrain regions. Our present data indicate that mutant (A30P) α-syn is directly implicated in reduction of dopamine signaling in OB interneurons, which mediates further alterations in brain regions without transgenic expression leading functionally to a hyperactive response. These modulations of neurotransmission may underlie in part some of the early neuropsychiatric symptoms in PD preceding dysfunction of the nigrostriatal dopaminergic system.
- Published
- 2011
- Full Text
- View/download PDF
23. Regulation of mitochondrial cargo-selective autophagy by posttranslational modifications
- Author
-
Milana Fraiberg, Boris Macek, Zvulun Elazar, Philipp J. Kahle, Anna Lechado Terradas, and Katharina Zittlau
- Subjects
Mitochondrial fission factor ,PTM, post-translational modification ,Mitochondrial Turnover ,MIM, mitochondrial inner membrane ,NGLY1, N-glycanase 1 ,LIR, LC3-interacting region ,BNIP, Bcl-2 19 kDa protein-interacting protein ,Biochemistry ,PD, Parkinson's disease ,Mitochondrial Dynamics ,DNM1L ,FUNDC1, FUN14 domain-containing protein 1 ,USP, ubiquitin-specific protease ,VPS, vacuolar sorting protein ,Mitophagy ,MFF, mitochondrial fission factor ,TFEB, transcription factor EB ,MFN, mitofusin ,PARL, presenilin-associated rhomboid-like protease ,HK2, hexokinase 2 ,OMA1, overlapping with the mitochondrial matrix ATPase associated with diverse cellular activities (m-AAA) protease 1 ,Chemistry ,phosphorylation ,SUMO, small ubiquitin-related modifier ,DFCP1, double FYVE domain-containing protein 1 ,NBR1, next to breast cancer type 1 susceptibility gene 1 protein ,DRP1, dynamin-related protein 1 ,Far, factor arrest ,RAB, Ras-associated binding protein ,MIRO, mitochondrial Rho GTPase ,GCN5L1, general control of amino acid synthesis protein 5-like 1 ,Cell biology ,BAK, Bcl-2 homologous antagonist/killer ,Mitochondria ,mitochondria ,OPA1, optic atrophy protein 1 ,ddc:540 ,ARIH1, ariadne-1 homolog in humans ,SIRT, sirtuin ,Mitochondrial fission ,genetics [Mitochondrial Proteins] ,Signal Transduction ,FIS1 ,autophagy ,WIPI, WD40 repeat protein interacting with phosphoinositides ,Bcl, B cell lymphoma ,TBK1, tumor necrosis factor receptor-associated factor family member associated NF-κB activator-binding kinase 1 ,NF-κB, nuclear factor-κB ,PINK1 ,PINK1, phosphatase and tensin homolog (PTEN)-induced kinase 1 ,mTOR, mammalian target of rapamycin ,metabolism [Mitochondrial Proteins] ,CCCP, carbonyl cyanide m-chlorophenyl hydrazone ,RING, really interesting new gene ,Mitochondrial Proteins ,SNARE, soluble N-ethylmaleimide-sensitive factor-attachment protein receptor ,ΔΨm, mitochondrial membrane potential ,LC3, microtubule-associated protein light chain 3 ,FKBP8, FK506-binding protein 8 ,GABARAP, γ-amino-butyric acid receptor-associated protein ,MOM, mitochondrial outer membrane ,Animals ,HOPS, homotypic fusion and protein sorting ,ubiquitylation ,ATG, autophagy-related protein ,FIS1, mitochondrial fission 1 protein ,Molecular Biology ,gp78, glycoprotein 78 ,CLEAR, coordinated lysosomal expression and regulation ,MGRN1, mahogunin RING finger protein 1 ,ULK1, uncoordinated protein 51-like kinase 1 ,protein kinase PINK1 ,JBC Reviews ,BH3, Bcl-2 homology 3 domain ,Ubiquitination ,AMBRA1, activating molecule in beclin-1 regulated autophagy protein 1 ,VDAC, voltage-dependent anion selective channel ,Cell Biology ,metabolism [Mitochondria] ,HUWE1, homologous to the E6-AP carboxyl terminus (HECT), ubiquitin-associated and WWE domain-containing protein 1 ,FIP200, focal adhesion kinase-interacting protein of 200 kDa ,BAD, Bcl-2 associated agonist of cell death ,DUB, deubiquitylating enzyme ,Mcl-1, induced myeloid leukemia cell differentiation protein ,AMPK, AMP-activated protein kinase ,PI3P, phosphatidylinositol 3-phosphate ,MUL1 ,genetics [Mitochondria] ,TOM, translocase of the outer membrane ,mTORC1, mTOR complex 1 ,Ubl, ubiquitin-like domain ,MTS, mitochondrial targeting sequence ,MITOL, mitochondrial ubiquitin ligase ,ubiquitin ligase parkin ,MUL1, mitochondrial ubiquitin ligase 1 ,NDP52, nuclear dot protein of 52 kDa - Abstract
Mitochondria are important organelles in eukaryotes. Turnover and quality control of mitochondria are regulated at the transcriptional and posttranslational level by several cellular mechanisms. Removal of defective mitochondrial proteins is mediated by mitochondria resident proteases or by proteasomal degradation of individual proteins. Clearance of bulk mitochondria occurs via a selective form of autophagy termed mitophagy. In yeast and some developing metazoan cells (e.g., oocytes and reticulocytes), mitochondria are largely removed by ubiquitin-independent mechanisms. In such cases, the regulation of mitophagy is mediated via phosphorylation of mitochondria-anchored autophagy receptors. On the other hand, ubiquitin-dependent recruitment of cytosolic autophagy receptors occurs in situations of cellular stress or disease, where dysfunctional mitochondria would cause oxidative damage. In mammalian cells, a well-studied ubiquitin-dependent mitophagy pathway induced by mitochondrial depolarization is regulated by the mitochondrial protein kinase PINK1, which upon activation recruits the ubiquitin ligase parkin. Here, we review mechanisms of mitophagy with an emphasis on posttranslational modifications that regulate various mitophagy pathways. We describe the autophagy components involved with particular emphasis on posttranslational modifications. We detail the phosphorylations mediated by PINK1 and parkin-mediated ubiquitylations of mitochondrial proteins that can be modulated by deubiquitylating enzymes. We also discuss the role of accessory factors regulating mitochondrial fission/fusion and the interplay with pro- and antiapoptotic Bcl-2 family members. Comprehensive knowledge of the processes of mitophagy is essential for the understanding of vital mitochondrial turnover in health and disease.
- Published
- 2021
- Full Text
- View/download PDF
24. Author Reply to Peer Reviews of LAG3 is not expressed in human and murine neurons and does not modulate α-synucleinopathies
- Author
-
Adriano Aguzzi, Mathias Jucker, Magdalini Polymenidou, Mark R. Cookson, Tuomas P. J. Knowles, Simone Hornemann, Philipp J. Kahle, Ronald Melki, Kelvin Luk, Natalie Landeck, Naunehal S. Matharu, Therese W. Herling, Lisa M. Häsler, Regina Reimann, Daniel Heinzer, Merve Avar, Andrès Gonzalez-Guerra, Nora Bengoa-Vergniory, Alice Kaganovich, Rebekah G. Langston, Mehtap Bacioglu, Pierre de Rossi, Elena Tantardini, Melanie Barth, Matthias M. Schneider, Timo Eninger, Marian Hruska-Plochan, Elena De Cecco, and Marc Emmenegger
- Published
- 2021
- Full Text
- View/download PDF
25. LAG3 is not expressed in human and murine neurons and does not modulate α-synucleinopathies
- Author
-
Pierre De Rossi, Therese W. Herling, Marian Hruska-Plochan, Nora Bengoa-Vergniory, Ronald Melki, Natalie Landeck, Mathias Jucker, Simone Hornemann, Philipp J. Kahle, Rebekah G. Langston, Andrés González-Guerra, Merve Avar, Melanie Barth, Timo Eninger, Lisa M. Häsler, Kelvin C. Luk, Mehtap Bacioglu, Daniel Heinzer, Elena Tantardini, Marc Emmenegger, Tuomas P. J. Knowles, Magdalini Polymenidou, Elena De Cecco, Alice Kaganovich, Matthias Schneider, Naunehal S. Matharu, Mark R. Cookson, Adriano Aguzzi, and Regina Reimann
- Subjects
Genetically modified mouse ,Synucleinopathies ,0303 health sciences ,LAG3 ,Hippocampal slice ,Biology ,Fibril ,Immune checkpoint ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,Receptor ,030217 neurology & neurosurgery ,Ex vivo ,030304 developmental biology - Abstract
While the initial pathology of Parkinson’s disease and other α-synucleinopathies is often confined to circumscribed brain regions, it can spread and progressively affect adjacent and distant brain locales. This process may be controlled by cellular receptors of α-synuclein fibrils, one of which was proposed to be the LAG3 immune checkpoint molecule. Here, we analyzed the expression pattern of LAG3 in human and mouse brains. Using a variety of methods and model systems, we found no evidence for LAG3 expression by neurons. While we confirmed that LAG3 interacts with α-synuclein fibrils, the specificity of this interaction appears limited. Moreover, overexpression of LAG3 in cultured human neural cells did not cause any worsening of α-synuclein pathology ex vivo. The overall survival of A53T α-synuclein transgenic mice was unaffected by LAG3 depletion and the seeded induction of α-synuclein lesions in hippocampal slice cultures was unaffected by LAG3 knockout. These data suggest that the proposed role of LAG3 in the spreading of α-synucleinopathies is not universally valid.
- Published
- 2021
- Full Text
- View/download PDF
26. Chronic–Progressive Dopaminergic Deficiency Does Not Induce Midbrain Neurogenesis
- Author
-
Sigrid C. Schwarz, Mareike Fauser, Johannes Schwarz, Francisco Pan-Montojo, Andreas Hermann, Philipp J. Kahle, Christian Richter, and Alexander Storch
- Subjects
0301 basic medicine ,Dopamine ,Neurogenesis ,adult neurogenesis ,periventricular regions ,non-neurogenic regions ,Parkinson´s disease ,dopaminergic neurodegeneration ,transgenic animal model ,Hindbrain ,Biology ,Receptors, Nicotinic ,physiology [Mesencephalon] ,Article ,Midbrain ,03 medical and health sciences ,Lateral ventricles ,deficiency [Dopamine] ,0302 clinical medicine ,Mesencephalon ,Dopaminergic Cell ,Lateral Ventricles ,ddc:570 ,medicine ,Animals ,Humans ,metabolism [alpha-Synuclein] ,lcsh:QH301-705.5 ,Cell Proliferation ,physiology [Lateral Ventricles] ,Dopaminergic ,Neurodegeneration ,General Medicine ,medicine.disease ,Neural stem cell ,Rhombencephalon ,Mice, Inbred C57BL ,030104 developmental biology ,nervous system ,lcsh:Biology (General) ,metabolism [Receptors, Nicotinic] ,physiology [Rhombencephalon] ,alpha-Synuclein ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Background: Consecutive adult neurogenesis is a well-known phenomenon in the ventricular–subventricular zone of the lateral wall of the lateral ventricles (V–SVZ) and has been controversially discussed in so-called “non-neurogenic” 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 chronic–progressive dopaminergic neurodegeneration on both consecutive adult neurogenesis in the PVRs of the V–SVZ and mid/hindbrain aNSCs in two mechanistically different transgenic animal models of Parkinson´s disease (PD). Methods: We used Thy1-m[A30P]h α synuclein mice and Leu9′Ser hypersensitive α4* nAChR mice to assess the influence of midbrain dopaminergic neuronal loss on neurogenic activity in the PVRs of the V–SVZ, the aqueduct and the fourth ventricle. Results: In both animal models, overall proliferative activity in the V–SVZ was not altered, though the proportion of B2/activated B1 cells on all proliferating cells was reduced in the V–SVZ in Leu9′Ser hypersensitive α4* 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.
- Published
- 2021
- Full Text
- View/download PDF
27. Survival-Promoting Activity of Inhibitors of Cyclin-Dependent Kinases on Primary Neurons Correlates with Inhibition of c-Jun Kinase-1
- Author
-
Marc A. Markus, Philipp J. Kahle, Andrea Winkler, Sonja Horstmann, Johanna M.H. Anneser, and Gian Domenico Borasio
- Subjects
Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Cyclin-dependent kinases and mitogen-activated protein kinases have been implicated in the regulation of cellular survival and apoptosis. We tested the effect of two mitogen-activated/cyclin-dependent kinase inhibitors, olomoucine and butyrolactone I, on thein vitrosurvival of chick embryonic neurons. Sensory, sympathetic, and ciliary neurons, when prepared at their respective time point of programmed cell death, could be rescued from apoptosis by both inhibitors in a dose-dependent fashion. In contrast, dividing sympathetic precursors underwent apoptosis when treated with olomoucine, but not butyrolactone I, at the same range of concentration. With similar potency, olomoucine and butyrolactone I inhibited immunocomplex c-Jun kinase activity. Both substances inhibited neurite outgrowth in a dose-dependent manner; developmentally younger neurons were more sensitive to this effect than older ones. These results suggest that certain mitogen-activated/cyclin-dependent kinases associated with cell division in neuronal precursors (i) may become essential components of the apoptotic machinery by the time neurons reach their phase of naturally occurring cell death and (ii) may be necessary for neurite outgrowth during development.
- Published
- 1997
- Full Text
- View/download PDF
28. Ataxin1L is a regulator of HSC function highlighting the utility of cross-tissue comparisons for gene discovery.
- Author
-
Juliette J Kahle, George P Souroullas, Peng Yu, Fabian Zohren, Yoontae Lee, Chad A Shaw, Huda Y Zoghbi, and Margaret A Goodell
- Subjects
Genetics ,QH426-470 - Abstract
Hematopoietic stem cells (HSCs) are rare quiescent cells that continuously replenish the cellular components of the peripheral blood. Observing that the ataxia-associated gene Ataxin-1-like (Atxn1L) was highly expressed in HSCs, we examined its role in HSC function through in vitro and in vivo assays. Mice lacking Atxn1L had greater numbers of HSCs that regenerated the blood more quickly than their wild-type counterparts. Molecular analyses indicated Atxn1L null HSCs had gene expression changes that regulate a program consistent with their higher level of proliferation, suggesting that Atxn1L is a novel regulator of HSC quiescence. To determine if additional brain-associated genes were candidates for hematologic regulation, we examined genes encoding proteins from autism- and ataxia-associated protein-protein interaction networks for their representation in hematopoietic cell populations. The interactomes were found to be highly enriched for proteins encoded by genes specifically expressed in HSCs relative to their differentiated progeny. Our data suggest a heretofore unappreciated similarity between regulatory modules in the brain and HSCs, offering a new strategy for novel gene discovery in both systems.
- Published
- 2013
- Full Text
- View/download PDF
29. FAS-dependent cell death in α-synuclein transgenic oligodendrocyte models of multiple system atrophy.
- Author
-
Christine L Kragh, Gwenaëlle Fillon, Amanda Gysbers, Hanne D Hansen, Manuela Neumann, Christiane Richter-Landsberg, Christian Haass, Bernard Zalc, Catherine Lubetzki, Wei-Ping Gai, Glenda M Halliday, Philipp J Kahle, and Poul H Jensen
- Subjects
Medicine ,Science - Abstract
Multiple system atrophy is a parkinsonian neurodegenerative disorder. It is cytopathologically characterized by accumulation of the protein p25α in cell bodies of oligodendrocytes followed by accumulation of aggregated α-synuclein in so-called glial cytoplasmic inclusions. p25α is a stimulator of α-synuclein aggregation, and coexpression of α-synuclein and p25α in the oligodendroglial OLN-t40-AS cell line causes α-synuclein aggregate-dependent toxicity. In this study, we investigated whether the FAS system is involved in α-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 α-synuclein and p25α relies on stimulation of the death domain receptor FAS and caspase-8 activation. Using primary oligodendrocytes derived from PLP-α-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 α-synuclein dependent degeneration and thus represent a potential target for protective intervention.
- Published
- 2013
- Full Text
- View/download PDF
30. Medin amyloid forms age‐associated aggregates in the brain vasculature and may contribute to cerebral β‐amyloidosis
- Author
-
Angelos Skodras, Anna Julia Koppelmann, Jillian Madine, Katleen Wild, Lisa M. Haesler, Felix von Zweydorf, Karoline Degenhardt, Rusheka Maxwell, Philipp J. Kahle, Domenico Del Turco, Ulrike Obermüller, Thomas Deller, Mathias Jucker, Christian Johannes Gloeckner, Jonas J. Neher, Hannah A. Davies, Jessica Wagner, and Carola Rotermund
- Subjects
Pathology ,medicine.medical_specialty ,Brain vasculature ,Amyloid ,Epidemiology ,business.industry ,Health Policy ,Amyloidosis ,medicine.disease ,Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,medicine ,Neurology (clinical) ,Geriatrics and Gerontology ,business - Published
- 2020
- Full Text
- View/download PDF
31. Nonneutralizing Factor VIII Antibodies as a Marker for Inhibitor Development in a Longitudinal Cohort of Hemophilia A Patients
- Author
-
Diana Stichel, Christoph Königs, Aleksander Orlowski, Christine Heller, Thomas Klingebiel, Sonja Neimanis, Dirk Schwabe, Stephan Schultze-Strasser, J. Kahle, and Anja Schmidt
- Subjects
Oncology ,medicine.medical_specialty ,biology ,business.industry ,Internal medicine ,biology.protein ,medicine ,Antibody ,Longitudinal cohort ,business - Published
- 2020
- Full Text
- View/download PDF
32. Sirtuin-1 Sensitive Lysine-136 Acetylation Drives Phase Separation and Pathological Aggregation of TDP-43
- Author
-
Jorge Garcia Morato, Manuela Neumann, Christian Johannes Gloeckner, Angelos Skodras, Friederike Hans, Simon J Elsaesser, Felix von Zweydorf, Emanuele Buratti, Regina Feederle, and Philipp J. Kahle
- Subjects
Lysine ,Protein Aggregation, Pathological ,metabolism [Protein Aggregation, Pathological] ,Sirtuin 1 ,Ubiquitin ,mental disorders ,Humans ,biology ,Chemistry ,metabolism [Amyotrophic Lateral Sclerosis] ,metabolism [Sirtuin 1] ,Amyotrophic Lateral Sclerosis ,nutritional and metabolic diseases ,RNA ,Acetylation ,metabolism [Lysine] ,nervous system diseases ,Cell biology ,DNA-Binding Proteins ,genetics [Sirtuin 1] ,metabolism [RNA] ,RNA splicing ,Sirtuin ,biology.protein ,ddc:500 ,metabolism [DNA-Binding Proteins] ,Nuclear transport ,Protein Processing, Post-Translational - Abstract
The trans-activation response DNA-binding protein TDP-43 regulates RNA processing and forms neuropathological aggregates in patients with amyotrophic lateral sclerosis and frontotemporal lobar degeneration. Investigating TDP-43 post-translational modifications, we discovered that K84 acetylation reduced nuclear import whereas K136 acetylation impaired RNA binding and splicing capabilities of TDP-43. Such failure of RNA interaction triggered TDP-43 phase separation mediated by the C-terminal low complexity domain, leading to the formation of insoluble aggregates with pathologically phosphorylated and ubiquitinated TDP-43. Confirming the results from site-directed mutagenesis, we succeeded to introduce authentic acetyl-lysine at the identified sites via amber suppression. [AcK84]TDP-43 showed cytoplasmic mislocalization and the aggregation propensity of [acK136]TDP-43 was confirmed. With newly developed antibodies, we found that the nuclear sirtuin SIRT1 can potently deacetylate [acK136]TDP-43. Moreover, SIRT1 reduced the aggregation propensity of [acK136]TDP-43. Thus, distinct lysine acetylations modulate nuclear import, RNA binding and phase separation of TDP-43, suggesting novel regulatory mechanisms for TDP-43 pathogenesis.
- Published
- 2020
- Full Text
- View/download PDF
33. Medin aggregation causes cerebrovascular dysfunction in aging wild-type mice
- Author
-
Philipp J. Kahle, Rusheka Maxwell, Katleen Wild, Regina Feederle, Jasmin K Hefendehl, Jonas J. Neher, Felix von Zweydorf, Domenico Del Turco, Hannah A. Davies, Karoline Degenhardt, Anna Julia Koppelmann, Thomas Deller, Mathias Jucker, Tammaryn Lashley, Christian Johannes Gloeckner, Jessica Wagner, Michael Candlish, Carola Rotermund, Jillian Madine, and Angelos Skodras
- Subjects
metabolism [Milk Proteins] ,Male ,genetics [Amyloid] ,Aging ,Brain vasculature ,metabolism [Vascular Diseases] ,Mice ,MFGE8 protein, human ,Aorta ,Aged, 80 and over ,education.field_of_study ,metabolism [Antigens, Surface] ,Multidisciplinary ,physiology [Brain Chemistry] ,Biological Sciences ,Milk Proteins ,Medin ,Cerebrovascular Circulation ,Antigens, Surface ,Mfg-e8 ,Cerebrovascular Dysfunction ,Amyloid ,Mfge8 protein, mouse ,Female ,ddc:500 ,cerebrovascular dysfunction ,medicine.medical_specialty ,Population ,pathology [Aorta] ,MFG-E8 ,physiology [Cerebrovascular Circulation] ,genetics [Milk Proteins] ,genetics [Antigens, Surface] ,medicine.artery ,Internal medicine ,medicine ,Animals ,Humans ,metabolism [Aging] ,Vascular Diseases ,Healthy aging ,education ,metabolism [Amyloid] ,Brain Chemistry ,business.industry ,Upper body ,aging ,Wild type mice ,metabolism [Aorta] ,Mice, Inbred C57BL ,Endocrinology ,business ,pathology [Vascular Diseases] - Abstract
Significance Vascular dysfunction, as it develops either during normal aging or vascular disease, remains a major medical problem. The amyloid Medin, which is derived from its precursor protein MFG-E8 (through unknown mechanisms), forms insoluble aggregates in the vasculature of virtually anybody over 50 years of age, and it has been hypothesized that Medin aggregation could contribute to age-associated vascular decline; however, mechanistic analyses have so far been lacking. Our data now demonstrate that reminiscent of humans, mice also develop Medin deposits in an age-dependent manner. Importantly, mice that genetically lack Medin show reduced vascular dysfunction in the aged brain. Therefore, the prevention of Medin accumulation should be investigated as a novel therapeutic approach to preserve vascular health in the aging population.
- Published
- 2020
- Full Text
- View/download PDF
34. Prominent microglial inclusions in transgenic mouse models of alpha-synucleinopathy that are distinct from neuronal lesions
- Author
-
Ulrike Obermüller, Mathias Jucker, Deborah Kronenberg-Versteeg, Mehtap Bacioglu, Melanie Barth, Angelos Skodras, Jasmin Mahler, Gaye Tanriöver, Philipp J. Kahle, Yvonne S. Eisele, Manuel Schweighauser, Derya R. Shimshek, Bettina M. Wegenast-Braun, K. Peter R. Nilsson, Jucker, Mathias [0000-0001-9045-1072], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Synucleinopathies ,Protein Conformation ,Synuclein ,Microglia ,Inclusion ,Prion-like ,Amyloid ,Conformation ,Parkinsons disease ,lcsh:RC346-429 ,Mice ,0302 clinical medicine ,pathology [Inclusion Bodies] ,pathology [Neurons] ,metabolism [alpha-Synuclein] ,Inclusion Bodies ,Neurons ,Chemistry ,pathology [Microglia] ,genetics [Synucleinopathies] ,medicine.anatomical_structure ,genetics [alpha-Synuclein] ,alpha-Synuclein ,Phosphorylation ,Intracellular ,Neurovetenskaper ,Genetically modified mouse ,Cell type ,Transgene ,Mice, Transgenic ,Protein Aggregation, Pathological ,Pathology and Forensic Medicine ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,pathology [Protein Aggregation, Pathological] ,metabolism [Protein Aggregation, Pathological] ,medicine ,genetics [Protein Aggregation, Pathological] ,Animals ,Humans ,ddc:610 ,lcsh:Neurology. Diseases of the nervous system ,Research ,Neurosciences ,Molecular biology ,Disease Models, Animal ,030104 developmental biology ,pathology [Synucleinopathies] ,chemistry [alpha-Synuclein] ,Parkinson’s disease ,Neurology (clinical) ,030217 neurology & neurosurgery - Abstract
Alpha-synucleinopathies are a group of progressive neurodegenerative disorders, characterized by intracellular deposits of aggregated alpha-synuclein (alpha S). The clinical heterogeneity of these diseases is thought to be attributed to conformers (or strains) of alpha S but the contribution of inclusions in various cell types is unclear. The aim of the present work was to study alpha S conformers among different transgenic (TG) mouse models of alpha-synucleinopathies. To this end, four different TG mouse models were studied (Prnp-h[A53T]alpha S; Thy1-h[A53T]alpha S; Thy1-h[A30P]alpha S; Thy1-m alpha S) that overexpress human or murine alpha S and differed in their age-of-symptom onset and subsequent disease progression. Postmortem analysis of end-stage brains revealed robust neuronal alpha S pathology as evidenced by accumulation of alpha S serine 129 (p-alpha S) phosphorylation in the brainstem of all four TG mouse lines. Overall appearance of the pathology was similar and only modest differences were observed among additionally affected brain regions. To study alpha 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 alpha 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 alpha S, but were largely p-alpha 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 alpha S aggregation. Although nature and significance of microglial inclusions for human alpha-synucleinopathies remain to be clarified, the previously overlooked abundance of microglial inclusions in TG mouse models of alpha-synucleinopathy bears importance for mechanistic and preclinical-translational studies. Funding Agencies|ECEuropean Commission Joint Research CentreEuropean Community (EC); EU/EFPIA/Innovative Medicines Initiative 2 Joint Undertaking (IMPRiND) [116060]; Alexander von Humboldt-FoundationAlexander von Humboldt Foundation; German Academic Exchange Service (DAAD)Deutscher Akademischer Austausch Dienst (DAAD); Swedish Research CouncilSwedish Research Council [2016-00748]
- Published
- 2020
- Full Text
- View/download PDF
35. Impaired c-Fos and polo-like kinase 2 induction in the limbic system of fear-conditioned α-synuclein transgenic mice.
- Author
-
Heinrich Schell, Cindy Boden, André Maia Chagas, and Philipp J Kahle
- Subjects
Medicine ,Science - Abstract
α-Synuclein (αSYN) is genetically and neuropathologically linked to a spectrum of neurodegenerative diseases including Parkinson's disease, dementia with Lewy bodies, and related disorders. Cognitive impairment is recapitulated in several αSYN transgenic mouse lines. However, the mechanisms of dysfunction in affected neurons are largely unknown. Here we measured neuronal activity induced gene products in the limbic system of αSYN transgenic mice upon fear conditioning (FC). Induction of the synaptic plasticity marker c-Fos was significantly reduced in the amygdala and hippocampus of (Thy1)-h[A30P]αSYN transgenic mice in an age-dependent manner. Similarly, the neuronal activity inducible polo-like kinase 2 (Plk2) that can phosphorylate αSYN at the pathological site serine-129 was up-regulated in both brain regions upon FC. Plk2 inductions were also significantly impaired in aged (Thy1)-h[A30P]αSYN transgenic mice, both in the amygdala and hippocampus. Plk2 inductions in the amygdala after FC were paralleled by a small but significant increase in the number of neuronal cell bodies immunopositive for serine-129 phosphorylated αSYN in young but not aged (Thy1)-h[A30P]αSYN transgenic mice. In addition, we observed in the aged hippocampus a distinct type of apparently unmodified transgenic αSYN profiles resembling synaptic accumulations of αSYN. Thus, the cognitive decline observed in aged αSYN transgenic mice might be due to impairment of neurotransmission and synaptic plasticity in the limbic system by distinct αSYN species.
- Published
- 2012
- Full Text
- View/download PDF
36. Elimination of factor VIII‐specific B cells by immunotoxins composed of a single factor VIII domain fused to Pseudomonas exotoxin A
- Author
-
Kerstin Brettschneider, J. Kahle, Anja Schmidt, Dirk Schwabe, Christoph Königs, Diana Stichel, and Aleksander Orlowski
- Subjects
0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,Virulence Factors ,Recombinant Fusion Proteins ,Bacterial Toxins ,Exotoxins ,Serum Albumin, Human ,030204 cardiovascular system & hematology ,Hemophilia A ,Immune tolerance ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Protein Domains ,Immunotoxin ,hemic and lymphatic diseases ,Escherichia coli ,Immune Tolerance ,Animals ,Humans ,Pseudomonas exotoxin ,C2 domain ,ADP Ribose Transferases ,Mice, Knockout ,B-Lymphocytes ,Factor VIII ,biology ,Chemistry ,Immunotoxins ,Antibodies, Monoclonal ,Hematology ,Antibodies, Neutralizing ,Fusion protein ,Molecular biology ,030104 developmental biology ,Monoclonal ,biology.protein ,Antibody ,Spleen ,Ex vivo ,Protein Binding - Abstract
Essentials There is still a need for novel therapeutic approaches for hemophilia A patients with inhibitors. A factor VIII domain was used as the targeting moiety for elimination of FVIII-specific B cells. The immunodominant C2 domain was fused to exotoxin A from Pseudomonas aeruginosa (hC2-ETA). Murine C2 domain-specific B cells were selectively and efficiently eliminated by hC2-ETA ex vivo. SUMMARY: Background Today, the most serious complication for patients with hemophilia A undergoing factor VIII (FVIII) replacement therapy is the development of neutralizing antibodies (inhibitors). Although inhibitors can be eradicated by application of high doses of FVIII, the immune tolerance induction therapy fails in up to 30% of patients. Hence, there is still an urgent need for novel therapeutic approaches for patients with persisting inhibitors. Objectives In the present study, the potential use of immunotoxins containing exotoxin A (ETA) from Pseudomonas aeruginosa for selective elimination of FVIII-specific B cells was explored. Methods The immunodominant C2 domain of human FVIII was used as a targeting moiety instead of the full-length FVIII protein and the resulting human C2 domain-ETA fusion protein (hC2-ETA) was produced in Escherichia coli. Results Binding studies with monoclonal C2 domain-specific antibodies confirmed the conformational integrity of the C2 domain in hC2-ETA. The functionality of hC2-ETA was tested ex vivo by incubation of splenocytes from inhibitor-positive FVIII knockout mice with hC2-ETA and controls. FVIII-specific memory B cells from splenocytes were differentiated by FVIII stimulation in antibody-secreting cells (ASC) and detected by an enzyme-linked immunospot assay. Although the controls showed no effect, incubation of splenocytes with hC2-ETA reduced the number of C2-specific ASC in a dose-dependent fashion, indicating specific and efficient elimination of C2-specific memory B cells. Conclusions Overall, the results of the study support the fact that FVIII domain immunotoxins might be a potential new tool for the elimination of FVIII-specific B cells in patients with hemophilia A and persisting inhibitors.
- Published
- 2018
- Full Text
- View/download PDF
37. Identification and characterization of ubiquitinylation sites in TAR DNA-binding protein of 43 kDa (TDP-43)
- Author
-
Marita Eckert, Christian Johannes Gloeckner, Philipp J. Kahle, Friederike Hans, and Felix von Zweydorf
- Subjects
0301 basic medicine ,Active Transport, Cell Nucleus ,genetics [DNA-Binding Proteins] ,Context (language use) ,Biochemistry ,Serine ,03 medical and health sciences ,metabolism [Ubiquitin] ,mental disorders ,Humans ,NLS ,Protein phosphorylation ,metabolism [Cell Nucleus] ,Phosphorylation ,Site-directed mutagenesis ,Molecular Biology ,chemistry [Lysine] ,Cell Nucleus ,Ubiquitin ,Chemistry ,Lysine ,TDP-43 protein, human ,Mutagenesis ,nutritional and metabolic diseases ,Molecular Bases of Disease ,Cell Biology ,chemistry [DNA-Binding Proteins] ,nervous system diseases ,Cell biology ,DNA-Binding Proteins ,HEK293 Cells ,030104 developmental biology ,Solubility ,ddc:540 ,Mutation ,Mutagenesis, Site-Directed ,metabolism [DNA-Binding Proteins] ,Protein Processing, Post-Translational ,Nuclear localization sequence - Abstract
TAR DNA-binding protein of 43 kDa (TDP-43) forms pathological aggregates in neurodegenerative diseases, particularly in certain forms of frontotemporal dementia and amyotrophic lateral sclerosis. Pathological modifications of TDP-43 include proteolytic fragmentation, phosphorylation, and ubiquitinylation. A pathognomonic TDP-43 C-terminal fragment (CTF) spanning amino acids 193-414 contains only four lysine residues that could be potentially ubiquitinylated. Here, serial mutagenesis of these four lysines to arginine revealed that not a single residue is responsible for the ubiquitinylation of mCherry-tagged CTF. Removal of all four lysines was necessary to suppress ubiquitinylation. Interestingly, Lys-408 substitution enhanced the pathological phosphorylation of the immediately adjacent serine residues 409/410 in the context of mCherry-CTF. Thus, Lys-408 ubiquitinylation appears to hinder Ser-409/410 phosphorylation in TDP-43 CTF. However, we did not observe the same effect for full-length TDP-43. We extended the mutagenesis study to full-length TDP-43 and performed MS. Ubiquitinylated lysine residues were identified in the nuclear localization sequence (NLS; Lys-84 and Lys-95) and RNA-binding region (mostly Lys-160, Lys-181, and Lys-263). Mutagenesis of Lys-84 confirmed its importance as the major determinant for nuclear import, whereas Lys-95 mutagenesis did not significantly affect TDP-43's nucleo-cytoplasmic distribution, solubility, aggregation, and RNA-processing activities. Nevertheless, the K95A mutant had significantly reduced Ser-409/410 phosphorylation, emphasizing the suspected interplay between TDP-43 ubiquitinylation and phosphorylation. Collectively, our analysis of TDP-43 ubiquitinylation sites indicates that the NLS residues Lys-84 and Lys-95 have more prominent roles in TDP-43 function than the more C-terminal lysines and suggests a link between specific ubiquitinylation events and pathological TDP-43 phosphorylation.
- Published
- 2018
- Full Text
- View/download PDF
38. Monitoring α‐synuclein multimerization in vivo
- Author
-
Joerg B. Schulz, Philipp J. Kahle, Istvan Katona, Yasmine Wasser, Anand Goswami, Tiago F. Outeiro, Vibha Prasad, Aaron Voigt, and Friederike Hans
- Subjects
Male ,0301 basic medicine ,Amyloid ,Parkinson's disease ,animal diseases ,Protein aggregation ,environment and public health ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,Bimolecular fluorescence complementation ,0302 clinical medicine ,In vivo ,ddc:570 ,metabolism [Drosophila melanogaster] ,Serine ,Genetics ,medicine ,chemistry [Amyloid] ,Animals ,metabolism [Reactive Oxygen Species] ,metabolism [alpha-Synuclein] ,Phosphorylation ,Molecular Biology ,biology ,Chemistry ,Rotenone ,biology.organism_classification ,medicine.disease ,nervous system diseases ,Cell biology ,Disease Models, Animal ,Drosophila melanogaster ,030104 developmental biology ,Proteostasis ,chemistry [alpha-Synuclein] ,nervous system ,genetics [alpha-Synuclein] ,alpha-Synuclein ,health occupations ,growth & development [Drosophila melanogaster] ,Protein Multimerization ,Reactive Oxygen Species ,030217 neurology & neurosurgery ,Biotechnology - Abstract
The pathophysiology of Parkinson's disease is characterized by the abnormal accumulation of α-synuclein (α-Syn), eventually resulting in the formation of Lewy bodies and neurites in surviving neurons in the brain. Although α-Syn aggregation has been extensively studied in vitro, there is limited in vivo knowledge on α-Syn aggregation. Here, we used the powerful genetics of Drosophila melanogaster and developed an in vivo assay to monitor α-Syn accumulation by using a bimolecular fluorescence complementation assay. We found that both genetic and pharmacologic manipulations affected α-Syn accumulation. Interestingly, we also found that alterations in the cellular protein degradation mechanisms strongly influenced α-Syn accumulation. Administration of compounds identified as risk factors for Parkinson's disease, such as rotenone or heavy metal ions, had only mild or even no impact on α-Syn accumulation in vivo. Finally, we show that increasing phosphorylation of α-Syn at serine 129 enhances the accumulation and toxicity of α-Syn. Altogether, our study establishes a novel model to study α-Syn accumulation and illustrates the complexity of manipulating proteostasis in vivo.-Prasad, V., Wasser, Y., Hans, F., Goswami, A., Katona, I., Outeiro, T. F., Kahle, P. J., Schulz, J. B., Voigt, A. Monitoring α-synuclein multimerization in vivo.
- Published
- 2018
- Full Text
- View/download PDF
39. Neuropathology in mice expressing mouse alpha-synuclein.
- Author
-
Claus Rieker, Kumlesh K Dev, Katja Lehnhoff, Samuel Barbieri, Iwona Ksiazek, Sabine Kauffmann, Simone Danner, Heinrich Schell, Cindy Boden, Markus A Ruegg, Philipp J Kahle, Herman van der Putten, and Derya R Shimshek
- Subjects
Medicine ,Science - Abstract
α-Synuclein (αSN) 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 (mαSN), 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 mαSN in central neurons at levels reaching up to six-fold compared to endogenous mαSN. Unlike transgenic mice expressing human wildtype or mutant forms of αSN, these mαSN transgenic mice showed pronounced ubiquitin immunopathology in spinal cord and brainstem. Isoelectric separation of mαSN species revealed multiple isoforms including two Ser129-phosphorylated species in the most severely affected brain regions. Neuronal Ser129-phosphorylated αSN occurred 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 αSN 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 mαSN transgenic mice, was similar to what we have reported for mice expressing human αSN wildtype or mutant forms. In hippocampal neurons, the mαSN 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 αSN, mαSN transgenic mice displayed only end-stage phenotypic alterations that manifested alongside with neuropathology. Altogether these findings show that increased levels of wildtype mαSN 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.
- Published
- 2011
- Full Text
- View/download PDF
40. Reduced basal autophagy and impaired mitochondrial dynamics due to loss of Parkinson's disease-associated protein DJ-1.
- Author
-
Guido Krebiehl, Sabine Ruckerbauer, Lena F Burbulla, Nicole Kieper, Brigitte Maurer, Jens Waak, Hartwig Wolburg, Zemfira Gizatullina, Frank N Gellerich, Dirk Woitalla, Olaf Riess, Philipp J Kahle, Tassula Proikas-Cezanne, and Rejko Krüger
- Subjects
Medicine ,Science - Abstract
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.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.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.
- Published
- 2010
- Full Text
- View/download PDF
41. TDP-43-mediated neuron loss in vivo requires RNA-binding activity.
- Author
-
Aaron Voigt, David Herholz, Fabienne C Fiesel, Kavita Kaur, Daniel Müller, Peter Karsten, Stephanie S Weber, Philipp J Kahle, Till Marquardt, and Jörg B Schulz
- Subjects
Medicine ,Science - Abstract
Alteration and/or mutations of the ribonucleoprotein TDP-43 have been firmly linked to human neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). The relative impacts of TDP-43 alteration, mutation, or inherent protein function on neural integrity, however, remain less clear--a situation confounded by conflicting reports based on transient and/or random-insertion transgenic expression. We therefore performed a stringent comparative investigation of impacts of these TDP-43 modifications on neural integrity in vivo. To achieve this, we systematically screened ALS/FTLD-associated and synthetic TDP-43 isoforms via same-site gene insertion and neural expression in Drosophila; followed by transposon-based motor neuron-specific transgenesis in a chick vertebrate system. Using this bi-systemic approach we uncovered a requirement of inherent TDP-43 RNA-binding function--but not ALS/FTLD-linked mutation, mislocalization, or truncation--for TDP-43-mediated neurotoxicity in vivo.
- Published
- 2010
- Full Text
- View/download PDF
42. Parkin deficiency delays motor decline and disease manifestation in a mouse model of synucleinopathy.
- Author
-
Margot Fournier, Jérémie Vitte, Jérôme Garrigue, Dominique Langui, Jean-Philippe Dullin, Françoise Saurini, Naïma Hanoun, Fernando Perez-Diaz, Fabien Cornilleau, Chantal Joubert, Héctor Ardila-Osorio, Sabine Traver, René Duchateau, Cécile Goujet-Zalc, Katerina Paleologou, Hilal A Lashuel, Christian Haass, Charles Duyckaerts, Charles Cohen-Salmon, Philipp J Kahle, Michel Hamon, Alexis Brice, and Olga Corti
- Subjects
Medicine ,Science - Abstract
In synucleinopathies, including Parkinson's disease, partially ubiquitylated alpha-synuclein species phosphorylated on serine 129 (P(S129)-alpha-synuclein) accumulate abnormally. Parkin, an ubiquitin-protein ligase that is dysfunctional in autosomal recessive parkinsonism, protects against alpha-synuclein-mediated toxicity in various models.We analyzed the effects of Parkin deficiency in a mouse model of synucleinopathy to explore the possibility that Parkin and alpha-synuclein act in the same biochemical pathway. Whether or not Parkin was present, these mice developed an age-dependent neurodegenerative disorder preceded by a progressive decline in performance in tasks predictive of sensorimotor dysfunction. The symptoms were accompanied by the deposition of P(S129)-alpha-synuclein but not P(S87)-alpha-synuclein in neuronal cell bodies and neuritic processes throughout the brainstem and the spinal cord; activation of caspase 9 was observed in 5% of the P(S129)-alpha-synuclein-positive neurons. As in Lewy bodies, ubiquitin-immunoreactivity, albeit less abundant, was invariably co-localized with P(S129)-alpha-synuclein. During late disease stages, the disease-specific neuropathological features revealed by ubiquitin- and P(S129)-alpha-synuclein-specific antibodies were similar in mice with or without Parkin. However, the proportion of P(S129)-alpha-synuclein-immunoreactive neuronal cell bodies and neurites co-stained for ubiquitin was lower in the absence than in the presence of Parkin, suggesting less advanced synucleinopathy. Moreover, sensorimotor impairment and manifestation of the neurodegenerative phenotype due to overproduction of human alpha-synuclein were significantly delayed in Parkin-deficient mice.These findings raise the possibility that effective compensatory mechanisms modulate the phenotypic expression of disease in parkin-related parkinsonism.
- Published
- 2009
- Full Text
- View/download PDF
43. Algebraic stability modes in rotational shear flow
- Author
-
T Gebler, D Plümacher, J Kahle, and M Oberlack
- Subjects
Fluid Flow and Transfer Processes ,Mechanical Engineering ,General Physics and Astronomy - Abstract
We investigate the two-dimensional (2D) stability of rotational shear flows in an unbounded domain. The eigenvalue problem is formulated by using a novel algebraic mode decomposition distinct from the normal modes with temporal evolution exp ( ω t ) . Based on the work of Nold and Oberlack (2013 Phys. Fluids 25 104101), we show how these new modes can be constructed from the symmetries of the linearized stability equation. For the azimuthal base flow velocity V ( r ) = r − 1 an additional symmetry exists, such that a mode with algebraic temporal evolution t s is found. s refers to an eigenvalue for the algebraic growth or decay of the kinetic energy of the perturbations. An eigenvalue problem for the viscous and inviscid stability using algebraic modes is formulated on an infinite domain with r → ∞ . An asymptotic analysis of the eigenfunctions shows that the flow is linearly stable under 2D perturbations. We find stable modes with the algebraic mode ansatz, which can not be obtained by a normal mode analysis. The stability results are in line with Rayleigh’s inflection point theorem.
- Published
- 2021
- Full Text
- View/download PDF
44. LRRK2 functions as a scaffolding kinase of ASK1-mediated neuronal cell death
- Author
-
Jung-Soon Mo, Eun-Jung Ann, Ji-Seon Ahn, Hye-Jin Lee, Philipp J. Kahle, Hee-Sae Park, Thomas Gasser, Guang-Hui Liu, Mi-Yeon Kim, Young Chul Lee, Sergiy M. Yarmoluk, Ji-Hye Yoon, Juan Carlos Izpisua Belmonte, Eun-Hye Jo, and Wongi Seol
- Subjects
0301 basic medicine ,MAP Kinase Kinase 3 ,Induced Pluripotent Stem Cells ,metabolism [Leucine-Rich Repeat Serine-Threonine Protein Kinase-2] ,Apoptosis ,Biology ,Mitogen-activated protein kinase kinase ,genetics [Signal Transduction] ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,MAP Kinase Kinase Kinase 5 ,metabolism [MAP Kinase Kinase 3] ,p38 Mitogen-Activated Protein Kinases ,MAP2K7 ,03 medical and health sciences ,0302 clinical medicine ,metabolism [MAP Kinase Kinase Kinase 5] ,genetics [Parkinson Disease] ,ddc:570 ,Humans ,pathology [Neurons] ,ASK1 ,Phosphorylation ,genetics [Apoptosis] ,Molecular Biology ,Neurons ,genetics [MAP Kinase Kinase Kinase 5] ,MAP kinase kinase kinase ,Cyclin-dependent kinase 4 ,genetics [p38 Mitogen-Activated Protein Kinases] ,Cyclin-dependent kinase 5 ,Cyclin-dependent kinase 2 ,Parkinson Disease ,Cell Biology ,pathology [Parkinson Disease] ,nervous system diseases ,Cell biology ,metabolism [Induced Pluripotent Stem Cells] ,030104 developmental biology ,metabolism [Neurons] ,Cancer research ,biology.protein ,genetics [Leucine-Rich Repeat Serine-Threonine Protein Kinase-2] ,Cyclin-dependent kinase 9 ,030217 neurology & neurosurgery ,Signal Transduction ,genetics [MAP Kinase Kinase 3] - Abstract
Leucine-rich repeat kinase 2 (LRRK2), a multi-domain protein, is a key causative factor in Parkinson's disease (PD). Identification of novel substrates and the molecular mechanisms underlying the effects of LRRK2 are essential for understanding the pathogenesis of PD. In this study, we showed that LRRK2 played an important role in neuronal cell death by directly phosphorylating and activating apoptosis signal-regulating kinase 1 (ASK1). LRRK2 phosphorylated ASK1 at Thr832 that is adjacent to Thr845, which serves as an autophosphorylation site. Moreover, results of binding and kinase assays showed that LRRK2 acted as a scaffolding protein by interacting with each components of the ASK1-MKK3/6-p38 MAPK pathway through its specific domains and increasing the proximity to downstream targets. Furthermore, LRRK2-induced apoptosis was suppressed by ASK1 inhibition in neuronal stem cells derived from patients with PD. These results clearly indicate that LRRK2 acts as an upstream kinase in the ASK1 pathway and plays an important role in the pathogenesis of PD.
- Published
- 2017
- Full Text
- View/download PDF
45. PINK1 Regulates Dopamine and Lipids at Mitochondria to Maintain Synapses and Neuronal Function
- Author
-
T. Gasser, Betül Uysal, Garcia A, Nicolas Casadei, Anna Schaedler, Katharina Zittlau, Britta Brügger, Philipp J. Kahle, Sven Geisler, Petra Fallier-Becker, Aslihan Ugun-Klusek, Marita Feldkaemper, Gerard J.M. Martens, L. Schwarz, Boris Macek, Christine Bus, Vogt Weisenhorn Dm, Jos F. Brouwers, Henner Koch, Wolfgang Wurst, Julia C. Fitzgerald, Rejko Krüger, Schmidt B, Klaudia K. Maruszczak, Hector Flores-Romero, Doron Rapaport, and Zarani M
- Subjects
Tyrosine hydroxylase ,Chemistry ,Dopamine ,Mitophagy ,Dopaminergic ,medicine ,Phosphorylation ,PINK1 ,Oxidative phosphorylation ,Mitochondrion ,medicine.drug ,Cell biology - Abstract
Mitochondrial dysfunction contributes to the pathogenesis of Parkinson’s disease but it is not clear why inherent mitochondrial defects lead specifically to the death of dopaminergic neurons of the mid brain. PINK1 is mitochondrial kinase andPINK1mutations cause early onset Parkinson’s disease.We found that in neuronal progenitors, PINK1 regulates mitochondrial morphology, mitochondrial contact to the endoplasmic reticulum (ER) and the phosphorylation of Miro1. A compensatory metabolic shift towards lipid synthesis provides mitochondria with the components needed for membrane renewal and oxidative phosphorylation, maintaining the mitochondrial network once mature.Cholesterol is increased by loss of PINK1, promoting overall membrane rigidity. This alters the distribution of phosphorylated DAT at synapses and impairs dopamine uptake. PINK1 is required for the phosphorylation of tyrosine hydroxylase at Ser19, dopamine and calcium homeostasis and dopaminergic pacemaking.We suggest a novel mechanism for PINK1 pathogenicity in Parkinson’s disease in addition to but not exclusive of mitophagy. We also provide a basis for potential therapeutics by showing that low doses of the cholesterol depleting drug ß-cyclodextrin reverse PINK1-specific phenotypes.
- Published
- 2019
- Full Text
- View/download PDF
46. mpoly: Multivariate Polynomials in R.
- Author
-
David J. Kahle
- Published
- 2013
- Full Text
- View/download PDF
47. ggmap: Spatial Visualization with ggplot2.
- Author
-
David J. Kahle and Hadley Wickham
- Published
- 2013
- Full Text
- View/download PDF
48. MicroRNA Biomarkers of Toxicity in Biological Matrices
- Author
-
Alison H. Harrill, Charles E. Wood, Brian N. Chorley, Shaun D. McCullough, and Juliette J. Kahle
- Subjects
0301 basic medicine ,Biology ,Toxicology ,Bioinformatics ,Protein markers ,MicroRNAs ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,chemistry ,030220 oncology & carcinogenesis ,Toxicity Tests ,microRNA ,Toxicity ,Animals ,Humans ,Biomarker (medicine) ,Liquid biopsy ,Xenobiotic ,Biomarkers - Abstract
Biomarker measurements that reliably correlate with tissue injury and that can be measured within accessible biofluids offer benefits in terms of cost, time, and convenience when assessing chemical and drug-induced toxicity in model systems or human cohorts. MicroRNAs (miRNAs) have emerged in recent years as a promising new class of biomarker for monitoring toxicity. Recent enthusiasm for miRNA biomarker research has been fueled by evidence that certain miRNAs are cell-type specific and are released during injury, thus raising the possibility of using biofluid-based miRNAs as a "liquid biopsy" that may be obtained by sampling extracellular fluids. As biomarkers, miRNAs demonstrate improved stability as compared with many protein markers and sequences are largely conserved across species, simplifying analytical techniques. Recent efforts have sought to identify miRNAs that are released into accessible biofluids following xenobiotic exposure, using compounds that target specific organs. Whereas still early in the discovery phase, miRNA biomarkers will have an increasingly important role in the assessment of adverse effects of both environmental chemicals and pharmaceutical drugs. Here, we review the current findings of biofluid-based miRNAs, as well as highlight technical challenges in assessing toxicologic pathology using these biomarkers.
- Published
- 2016
- Full Text
- View/download PDF
49. The Design, Deployment, and Evaluation of the CORAL Pre-Exascale Systems
- Author
-
Bill Hanson, Chris Marroquin, Martin Ohmacht, Sarp Oral, Tom Gooding, Feiyi Wang, Adam Moody, Mallikarjun Shankar, Junqi Yin, Ben Casses, Gene Davison, Sudharshan S. Vazhkudai, David Appelhans, Arthur S. Bland, Ian Karlin, Verónica G. Vergara Larrea, Al Geist, James H. Rogers, Py C. Watson, Chris Chambreau, Bronis R. de Supinski, Robert S. Blackmore, Fernando Pizzano, Matthew L. Leininger, Elsa Gonsiorowski, J. Kahle, Lance D. Weems, Drew Schmidt, Bryan S. Rosenburg, Leopold Grinberg, Scott Atchley, Bob Walkup, Ramesh Pankajakshan, Wayne Joubert, Don Maxwell, James C. Sexton, Dustin Leverman, Adam Bertsch, Matthew A Ezell, Bill Hartner, Christopher Zimmer, George Chochia, and Robin Goldstone
- Subjects
File system ,geography ,TOP500 ,Speedup ,Summit ,geography.geographical_feature_category ,Computer science ,020206 networking & telecommunications ,02 engineering and technology ,computer.software_genre ,01 natural sciences ,Titan (supercomputer) ,Software deployment ,0103 physical sciences ,0202 electrical engineering, electronic engineering, information engineering ,Operating system ,Benchmark (computing) ,IBM ,010306 general physics ,computer - Abstract
CORAL, the Collaboration of Oak Ridge, Argonne and Livermore, is fielding two similar IBM systems, Summit and Sierra, with NVIDIA GPUs that will replace the existing Titan and Sequoia systems. Summit and Sierra are currently ranked No. 1 and No. 3, respectively on the Top500 list. We discuss the design and key differences of the systems. Our evaluation of the systems highlights the following. Applications that fit in HBM see the most benefit and may prefer more GPUs; however, for some applications, the CPU-GPU bandwidth is more important than the number of GPUs. The node-local burst buffer scales linearly, and can achieve a 4X improvement over the parallel file system for large jobs; smaller jobs, however, may benefit from writing directly to the PFS. Finally, several CPU, network and memory bound analytics and GPU-bound deep learning codes achieve up to a 11X and 79X speedup/node, respectively over Titan.
- Published
- 2018
- Full Text
- View/download PDF
50. Modification of Ozone-Induced Changes in Lung Function by Moderate Recent Life Stress
- Author
-
Juliette J. Kahle, Radhika Dhingra, David Diaz-Sanchez, and Martin Case
- Subjects
chemistry.chemical_compound ,Ozone ,chemistry ,General Earth and Planetary Sciences ,Response heterogeneity ,Physiology ,sense organs ,Biology ,Life stress ,Lung function ,General Environmental Science - Abstract
While changes in lung function (LF) due to ozone (OZ) exposure are well-documented in healthy populations, potential sources of response heterogeneity, like stress, remain uncharacterized. Though e...
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.